9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Targeting of the Nasal Mucosa by Japanese Encephalitis Virus for Non-Vector-Borne Transmission

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          JEV, a main cause of severe viral encephalitis in humans, has a complex ecology composed of a mosquito-waterbird cycle and a cycle involving pigs, which amplifies virus transmission to mosquitoes, leading to increased human cases. JEV can be transmitted between pigs by contact in the absence of arthropod vectors. Moreover, virus or viral RNA is found in oronasal secretions and the nasal epithelium. Using nasal mucosa tissue explants and three-dimensional porcine nasal epithelial cells cultures and macrophages as ex vivo and in vitro models, we determined that the nasal epithelium could be a route of entry as well as exit for the virus. Infection of nasal epithelial cells resulted in apical and basolateral virus shedding and release of monocyte recruiting chemokines and therefore infection and replication in macrophages, which is favored by epithelial-cell-derived cytokines. The results are relevant to understand the mechanism of non-vector-borne direct transmission of JEV.

          ABSTRACT

          The mosquito-borne Japanese encephalitis virus (JEV) causes severe central nervous system diseases and cycles between Culex mosquitoes and different vertebrates. For JEV and some other flaviviruses, oronasal transmission is described, but the mode of infection is unknown. Using nasal mucosal tissue explants and primary porcine nasal epithelial cells (NEC) at the air-liquid interface (ALI) and macrophages as ex vivo and in vitro models, we determined that the nasal epithelium could represent the route of entry and exit for JEV in pigs. Porcine NEC at the ALI exposed to with JEV resulted in apical and basolateral virus shedding and release of monocyte recruiting chemokines, indicating infection and replication in macrophages. Moreover, macrophages stimulated by alarmins, including interleukin-25, interleukin-33, and thymic stromal lymphopoietin, were more permissive to the JEV infection. Altogether, our data are important to understand the mechanism of non-vector-borne direct transmission of Japanese encephalitis virus in pigs.

          IMPORTANCE JEV, a main cause of severe viral encephalitis in humans, has a complex ecology composed of a mosquito-waterbird cycle and a cycle involving pigs, which amplifies virus transmission to mosquitoes, leading to increased human cases. JEV can be transmitted between pigs by contact in the absence of arthropod vectors. Moreover, virus or viral RNA is found in oronasal secretions and the nasal epithelium. Using nasal mucosa tissue explants and three-dimensional porcine nasal epithelial cells cultures and macrophages as ex vivo and in vitro models, we determined that the nasal epithelium could be a route of entry as well as exit for the virus. Infection of nasal epithelial cells resulted in apical and basolateral virus shedding and release of monocyte recruiting chemokines and therefore infection and replication in macrophages, which is favored by epithelial-cell-derived cytokines. The results are relevant to understand the mechanism of non-vector-borne direct transmission of JEV.

          Related collections

          Most cited references68

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Experimental Infection of North American Birds with the New York 1999 Strain of West Nile Virus

          To evaluate transmission dynamics, we exposed 25 bird species to West Nile virus (WNV) by infectious mosquito bite. We monitored viremia titers, clinical outcome, WNV shedding (cloacal and oral), seroconversion, virus persistence in organs, and susceptibility to oral and contact transmission. Passeriform and charadriiform birds were more reservoir competent (a derivation of viremia data) than other species tested. The five most competent species were passerines: Blue Jay (Cyanocitta cristata), Common Grackle (Quiscalus quiscula), House Finch (Carpodacus mexicanus), American Crow (Corvus brachyrhynchos), and House Sparrow (Passer domesticus). Death occurred in eight species. Cloacal shedding of WNV was observed in 17 of 24 species, and oral shedding in 12 of 14 species. We observed contact transmission among four species and oral in five species. Persistent WNV infections were found in tissues of 16 surviving birds. Our observations shed light on transmission ecology of WNV and will benefit surveillance and control programs.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Re(de)fining the dendritic cell lineage.

            Dendritic cells (DCs) are essential mediators of innate and adaptive immune responses. Study of these critical cells has been complicated by their similarity to other hematopoietic lineages, particularly monocytes and macrophages. Progress has been made in three critical areas of DC biology: the characterization of lineage-restricted progenitors in the bone marrow, the identification of cytokines and transcription factors required during differentiation, and the development of genetic tools for the visualization and depletion of DCs in vivo. Collectively, these advances have clarified the nature of the DC lineage and have provided novel insights into their function during health and disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Type I and Type III Interferons Drive Redundant Amplification Loops to Induce a Transcriptional Signature in Influenza-Infected Airway Epithelia

              Introduction The type I interferon family is a group of cytokines encoded by a single IFNβ gene and a tandem cluster of multiple IFNα genes that were first characterized for their ability to interfere with influenza virus replication [1] and are now recognized as powerful inducers of the host response to viral infections. IFN induction by influenza A virus (IAV) depends on recognition of viral components by either cytoplasmic receptors or the Toll-like receptor (TLR) system, depending on the infected cell type. While plasmacytoid dendritic cells (pDC) use TLR7 to recognize influenza virus, in fibroblasts and conventional DCs IFNβ induction requires recognition of RNA viral genomes by the cytoplasmic RNA helicase retinoic acid-induced gene I (RIG-I) [2], [3]. Upon RNA binding, RIG-I interacts with the mitochondrial adaptor protein MAVS (also known as IPS-1, CARDIF and VISA) and initiates a signalling cascade that culminates in the activation of the transcriptional factors AP-1, NF-κB and IRF3, and the expression of IFNβ and IFNα4 in mouse (or IFNα1 in humans) [4]–[7]. Once secreted, IFNβ and IFNα4 acts in both a paracrine and autocrine way through binding to the ubiquitously expressed heterodimeric IFNα/β receptor (IFNAR1/2) to induce activation of the receptor–associated tyrosine kinases JAK1 and Tyk2 and subsequent phosphorylation of the transcriptional factors STAT1 and STAT2 [8]. Activated STATs then form transcription factor complexes, including STAT1 homodimers and a STAT1/STAT2/IRF9 heterotrimer known as ISGF3 [9], and mediate the induction of hundreds of IFN-stimulated genes (ISGs), whose expression determines the establishment of an antiviral state inside the cell. Recently, a novel group of IFNs was described and named type III IFNs. This new IFN family has three members: IFNλ1 (a pseudogene in mouse), IFNλ2 and IFNλ3, alternatively named IL-29, IL-28A and IL-28B respectively [10], [11]. IFNλ induction depends on the same triggers and signalling pathways that regulate type I IFN expression [12], [13], with the RIG-I/MAVS/TBK1/IRF3 axis being particularly relevant in mouse embryonic fibroblasts (MEFs) upon viral infection [14]. IL-28/29 act through a distinct receptor complex consisting of IL-28Rα, specific for type III IFNs, and the IL-10Rβ chain, which is also part of the receptors for IL-10, IL-22 and IL-26. Despite activating different receptors, both type I and III IFNs activate the JAK-STAT signalling pathway, that leads to the formation of the ISGF3 complex [15] and the induction of ISGs. The most important determinant of the different biological activities of IFNα/β and IFNλ is the distribution of their receptors. While the receptor for type I IFNs is expressed on all cells, IL-28Rα is found primarily on epithelial cells of both the respiratory and the gastrointestinal tract [16]–[18]. This finding suggests that type III IFNs may act in a cell-type restricted manner and may selectively contribute to the innate immunity of mucosal surfaces, potential entry sites for many pathogenic viruses. The airway epithelium is a pseudostratified, columnar epithelium consisting of ciliated, basal and secretory goblet cells, that lies at the interface between the host and the environment and provides the first line of defence against inhaled microorganisms. Airway epithelial cells represent the target of many respiratory viruses, including Influenza virus, Adenovirus, Rhinovirus and RSV [19]. As epithelial cells express both cell surface and endosomal pattern recognition receptors (PRRs) and intracellular viral sensors, they can promptly detect invading microbes and react by producing cytokines, chemokines and antimicrobial peptides, thus initiating inflammatory and immune responses [20], [21]. While the PRRs and downstream signals required for influenza ISG induction have been mapped out in detail in other cell types, it is less clear which recognition systems are in action in airway epithelia. Moreover, while the importance of type III IFNs in epithelial responses has been documented, it is unclear whether the signatures induced by interferon type I or III overlap and which, if any, ISG subsets are selectively induced by one or the other. To address these issues, we established cultures of primary differentiated murine tracheal epithelial cells (MTEC) and, using a genetic approach together with microarray analysis, investigated the mechanisms that lead to IFN induction in response to influenza A infection. We also assessed the relative contribution of type I and III IFNs to the establishment of an antiviral state by comparing the pattern of influenza-induced gene expression in the absence of either IFNα/β signalling, IFNλ signalling or both. These studies help define the biology and nature of the antiviral state induced by different IFNs in primary cells and determine whether and which genes are still induced by influenza infection in the complete absence of both IFN type I and III signalling. Results In MTEC, the induction of interferons requires MAVS and virus replication Primary MTEC were grown to confluence and exposed to air for 14 days, leading to formation of a fully differentiated, polarized epithelium containing ciliated and secretory goblet and Clara cells (Fig. 1A), a cellular composition which closely matches that of airway epithelia in vivo. These differentiated cultures were then infected with IAV, at a multiplicity of infection (moi) of 0.3. Intracellular FACS analysis of the infected cultures showed that approximately 10% of MTEC were expressing viral nucleoprotein (NP) 24 hours post infection (hpi) (Fig. S1). Total RNA was isolated from five replicate cultures 24 hpi and analyzed using microarrays. We first investigated which type of IFN was induced in response to the infection. To this purpose, after normalizing the signal intensity of each probe in each sample to the median intensity of that probe in the control group, and filtering for genes that were expressed above background level, we searched for the probe sets which represent IFNs in the data set. As shown in Fig. 1B, both type I (IFNβ, α4 and α5) and type III IFNs (IL-28A/B) were significantly induced in the infected samples, with IL-28A/B being the most strongly upregulated. The induction of all other IFNα genes did not reach statistical significance. Type II IFN (IFNγ) did not pass the initial filtering for genes expressed above background and was not induced upon infection (not shown). To identify the pattern recognition receptors responsible for IFN induction in this experimental model, we infected MTEC cultures from wild-type and knock-out mice and analyzed the expression of both IFNβ and IL-28 by quantitative RT-PCR and ELISA. While in some immune cells TLR7 is the major PRR mediating IFN induction by influenza virus [22], we found that the absence of TLR7 or its downstream adaptor MyD88 had no impact on the induction of IFNs in influenza-infected epithelia. Similarly, the adaptor TRIF used by TLR3 and TLR4 is not required for IFN induction in epithelia. In contrast, MTEC deficient in MAVS were unable to produce IFNs in response to influenza infection, suggesting involvement of the RIG-I pathway (Fig. 1C, 1D). We next sought to determine whether virus replication is required for the induction of IFNs. Treatment of PR8 at 65°C inactivates the viral polymerase and prevents viral replication, without abrogating virus attachment to the cells (Fig. S2A, S2B). This treatment abolished the virus' ability to induce IFNs (Fig. 1C, 1D) indicating that IAV virus access to the cytoplasm and subsequent replication are required to initiate an IFN response in MTEC. 10.1371/journal.ppat.1003773.g001 Figure 1 In primary MTEC, type I and III IFNs are induced upon influenza virus A infection, in a MAVS and replication dependent way. (A) Mouse tracheal epithelial cells were costained for ZO-1 (green) and β tubulin IV (red), or Mucin 5A (red) and Clara cell secretory protein (CCSP) (green). Images were acquired at ×20 magnification. (B) Total RNA from mock infected and PR8 infected (moi = 0.3) MTEC was analysed using Affymetrix Mouse Genome 430 2.0 microarrays. The signal intensity of each probe was first normalized on the median intensity of that probe across the control group and then represented as log2 fold change relative to the controls. Asterisks indicated statistically significant differences (unpaired t test; **, p 85% of immune cells in these chimeras have wt phenotype. When these chimeras were infected with the PR8 strain, high susceptibility and mortality was found only in the group lacking IFN receptors on stromal cells and this correlated with higher viral titers (Fig. 6A, B). These results suggest that the ability to respond to IFNs in infected airway epithelia is crucial for successful elimination of the virus from infected animals. 10.1371/journal.ppat.1003773.g006 Figure 6 Lack of type I and III IFN signalling in the stromal compartment increases susceptibility to IAV infection. Chimeric mice were infected i.n. with 105 TCID50 PR8, (A) Weight loss and mortality were measured. Graphs show mean ± SEM and are representative of 2 independent experiments (n = 6). (B) Viral replication was assessed by qRT-PCR on total lung RNA at 4 days post infection. *** p<0.001, ** p<0.01 by 2-way ANOVA with Bonferroni post tests (weight loss), Log-rank (Mantel-Cox) Test (survival) or unpaired t test (RT-PCR quantification). A residual production of IFNs is responsible for ISG induction in MAVS−/− epithelia Infected MAVS−/− and IRF3−/−IRF7−/− epithelia show ISG induction in the absence of any detectable IFN upregulation, while IFNAR1−/−IL-28Rα−/− cells, which produce but cannot respond to IFNs, did not express ISGs in response to infection. Although type I IFN genes are tightly regulated in response to viral infection, many tissues constitutively secrete low amounts of type I IFN even in the absence of infection (reviewed in [28]). It has been proposed that these constitutive levels of IFNs are required to maintain basal expression of IFN-inducible signalling intermediates (STAT1/2, IRF7/9/5) and to modulate the relative expression of STAT proteins, therefore “priming” cells for future responses. For these reasons, we sought to determine the basal level of different IFN-signalling intermediates at steady state and upon infection in IFNAR1−/−IL-28Rα−/− cells. Wild-type and double knock-out cells were infected with influenza A and the level of different STAT and IRF molecules analyzed by qPCR. Some of these molecules (IRF7, IRF9, STAT1, STAT2) are known ISGs and were upregulated in wild-type but not in IFNAR1−/−IL-28Rα−/− epithelia at 24 hours post infection. However, the levels of all these transcripts measured relatively to HPRT at steady state were comparable in the two genotypes (Fig. 7A, 7B) and in MAVS−/− and IRF3−/−IRF7−/− cells (not shown). 10.1371/journal.ppat.1003773.g007 Figure 7 Basal expression of signalling intermediates is similar in wild-type and IFNAR1−/−IL-28Rα−/− double knock-out MTEC. (A) Relative expression of IRF3, IRF1, IRF7 and IRF9 was determined in both mock infected and PR8 infected MTEC by qRT-PCR. The transcripts quantities are expressed as ratio to HPRT levels +/− SEM. (B) The same RNA samples were analyzed for STAT1, STAT2, STAT3, STAT4 and STAT6 expression. To test directly whether residual IFN production is responsible for ISG induction in MAVS and IRF3/7 deficient epithelia, we compared ISG induction in infected MAVS deficient epithelia in the presence or absence of an antibody cocktail blocking both IFNαβ and IFNλ signalling. As shown in Figure 8A, antibody-treated epithelium had further reduced ISG expression compared to the untreated one. To confirm independently the requirement of autocrine signalling by soluble factors for ISG induction, we infected wild-type epithelia in the presence or absence of brefeldin A (BFA). BFA treatment left IFN gene induction unaffected but abolished ISG induction, indicating that soluble factors which include IFNs are required for ISG induction (Fig. 8B). 10.1371/journal.ppat.1003773.g008 Figure 8 Residual IFN production is responsible for ISG induction in MAVS−/− epithelia. (A) qRT-PCR analysis of Rsad2, Oasl2 and STAT1 in MAVS−/− epithelia infected with PR8 in the presence or absence of blocking anti-IFNAR and neutralizing anti-IL28A/B antibodies. (B) Wild-type epithelia were PR8-infected in the presence or absence of brefeldin A (2.5 µg/ml). Expression of the indicated genes was determined by qRT-PCR at 24 hpi. All transcripts were normalized to HPRT levels and then expressed as fold induction relative to the mean of mock infected controls, +/− SEM. Asterisks indicate differences that are statistically significant (unpaired t test; *, P<0.05; **, P<0.01). Collectively, these results indicate that the different responses to infection in MAVS−/− and IRF3−/−IRF7−/− compared to IFNAR1−/−IL-28Rα−/− cells can not be ascribed to a lack of priming in the latter, but are most likely due to a residual production of IFN in MAVS−/− and IRF3−/−IRF7−/− cells, that, in the context of infection, is sufficient to ensure ISG induction. Discussion Here, we delineate the influenza-triggered pathways leading to the induction of an antiviral state in primary airway epithelia, the first and most important target tissue of the virus in an infected organism. We show that TLR7 or other TLRs relying on the adaptor molecules MyD88 and TRIF are not involved in the induction of interferons, while the RIG-I/MAVS pathway is crucial for this process. We also show that between the two transcription factors IRF3 and IRF7 implied in IFN induction, IRF3 is of less importance than IRF7, but complete abolition of influenza-triggered IFN expression is seen only in the absence of both molecules. Most importantly, we show that, upon influenza infection, IFN type I and III independently mediate parallel amplification loops leading to the induction of a completely overlapping set of ISGs, and that this induction is abolished only when none of the two amplification loops are active. The general paradigm for type I IFN induction involves recruitment of transcription factors that are activated by phosphorylation in response to signalling cascades stimulated during viral infection. The IFNβ promoter contains four positive regulatory domains (PRDI-IV), which are occupied by different transcription factors. PRDI and III are binding sites for IRF3 (early during infection, due to its constitutive expression) and IRF7 (with delayed kinetics, due to its inducible expression through an IFN-dependent positive feedback loop), while PRDIV and PRDII bind the ATF-2/c-Jun AP-1 and the p50/RelA NF-κB complexes respectively (reviewed in [8]). This initial model of a positive feedback loop, in which IRF3 is primarily responsible for the early induction of IFNβ while IRF7 is required later in the response [29], was subsequently modified when a study performed on IRF7−/− MEFs revealed that both the early and the late production of type I IFN induced by VSV or EMCV is abolished in the absence of IRF7 [30]. Our results are in line with these observations and identify IRF7 as the major regulator of both type I and type III IFN responses in epithelial cells. Indeed, our data in fig. 6A suggests that the expression level of IRF7 was higher than that of IRF3 at steady state, which would support the notion that IRF7 could act directly downstream of viral recognition to induce IFNs at the earliest stage of infection. Moreover, our data indicates that, even later during an infection, the expression of IFNβ1 and IL-28A/B can be sustained independently of the IFN-driven positive amplification loop (Fig. 5B). IFNλ is preferentially induced by influenza A virus, both in vivo and in vitro [31]. We extend these findings to primary airway epithelia and demonstrate that epithelial-derived IFN type I or type III are sufficient to fuel their respective amplification loop and that no extrinsic IFNs from other cells, for instance immune cells, is required to induce an epithelial IFN signature. The importance of type III IFNs in epithelial responses has been well documented. Several studies have shown that IFNλ protects the epithelium of lung, intestine and vagina from viral infections and that IFNAR1−/−IL-28Rα−/− double deficient mice are more susceptible to viral infection than each single knock-out strain [17], [18], [32]. Using chimeric mice with a wild type immune system and either wild type or IFNAR1−/−IL-28Rα−/− double deficient stroma, we show here that IFN unresponsiveness in the stromal cell compartment is sufficient to render mice more susceptible to influenza infection. Previous studies have shown that the promoters of many ISGs have a simple structure and can be easily turned on directly by IRF proteins independently of interferon. In these studies, alternative pathways of direct ISG induction were suggested that rely on either IRF3 [23], IRF7 [25], IRF1 [26] or peroxisomal MAVS/IRF1/IRF3 [33]. More recently, the cytosolic exonuclease Trex1 has also been identified as a negative regulator of a novel pathway involving STING, TBK1, IRF3 and IRF7 that can lead to interferon-independent activation of ISGs [34]. The finding that a subset of ISGs could still be induced in both MAVS- and IRF3/IRF7-deficient epithelia suggested to us that IFN-independent ISG induction may take place here. However, the nearly complete absence of ISG induction in IFNAR1−/−IL-28Rα−/− epithelia led us to conclude that, at least in our experimental model, no other mechanism can efficiently replace the IFN loop for the induction of ISGs. Constitutive low-level signalling of IFNβ (IFN “priming”) has been suggested to help preserve IFN responsiveness but also to allow IFN-independent ISG induction [28], by maintaining the expression of STATs and other signalling intermediates. It could be argued that, unlike IFNAR1−/−IL-28Rα−/− epithelia, MAVS−/− and IRF3−/−IRF7−/− cells still possess this sub-threshold signalling which helps maintain STATs and IRFs at sufficient levels to allow for direct ISG induction upon viral trigger, even in the absence of IFNs. We did however not detect steady-state differences in the expression of a range of IRF and STAT molecules between genotypes and therefore have no evidence that differences in IFN priming contribute to the phenomenon described here. Previous studies that assessed IFN independent ISG induction have mostly relied on IFNAR deficient cells to confirm IFN independence. Here we show that care must be taken to evaluate IFN independence. As IFN type III can stand in for IFN type I in inducing an IFN signature, the analysis of each single receptor knock-out epithelium would have wrongly suggested complete independence of ISG induction from the IFN system. Moreover, while the IFN signature was completely abolished in influenza infected IFNAR1−/−IL-28Rα−/− cells, the addition of neutralizing antibodies against secreted type I and type III IFNs, used in combination on wild-type epithelia or for the complementary IFN on IFN receptor single knock-out epithelia, had little effect on ISG induction (not shown), indicating that even minute concentrations of IFN were still able to induce ISG expression in responsive cells. In vivo studies with single-knock-out mice clearly showed that ISG induction by type III IFN translates into less powerful protection against influenza A virus than ISG induction by type I IFN [32]. At present it is unclear whether slight differences in the kinetics of virus-triggered induction of type I and type III IFN may account for this observation. An alternative explanation is that lung macrophages which are productively infected by most influenza A virus strains and which do not respond well to type III IFN may quickly amplify the incoming virus in the respiratory tract of IFNAR1-deficient mice and thus overwhelm the type III-mediated protection of epithelial cells in such mice. Through the induction of ISGs, the IFN system has potent effects not only to directly combat virus, but also on cell physiology and survival and on the immune response. Therefore, it is considered a very tightly controlled system to avoid excessive inflammation, cell death and tissue damage. On this background, we were surprised to find that in MAVS and IRF3/7 deficient epithelia, IFNs at levels below ELISA or qPCR detection threshold still lead to only slightly reduced ISG upregulation. ISG induction disappeared in MAVS deficient epithelia when IFN type I and III signalling was blocked by an antibody cocktail, indicating that a residual IFN production drives ISG induction in these cells. In apparent contrast to these results, when exogenous IFN was titrated onto epithelial cultures, the minimum amount of IFN required to induce ISG expression was clearly detectable by the ELISA assay (not shown). Possible reasons for this discrepancy could be differences in the bioactivity of endogenous versus recombinant IFNs and differential ability by the ELISAs to detect endogenous or recombinant IFNs. Moreover, biological explanations include local concentrations of endogenous IFNs that may be much higher than those measured in the total supernatant, and the possibility that autocrine IFNs bind to their receptor already in intracellular vesicles and are therefore not measured by ELISA. Overall these data suggest that presence or absence, rather than absolute amounts of IFN, determine the response. The biological sense of such a binary switch could be to respond robustly even to small perturbations of the steady state, which may gain the host precious time when infected by fast-replicating viruses. In the uninfected state, absolute IFN shut-down would be required to avoid chronic “Flu-like symptoms”, a scenario that is in contradiction to the proposed priming effect of sub-threshold IFN levels. One hypothesis for how the vast differences in IFN levels are translated into a largely unaltered IFN signature in MAVS−/− and IRF3−/−IRF7−/− epithelia is that Influenza A infection may render cells much more IFN-sensitive by unknown pathways. To test this hypothesis, we titrated exogenous IFN on uninfected or infected wt or MAVS-deficient epithelia and measured ISG induction. No increased IFN responsiveness was found in infected versus uninfected epithelia, suggesting that there is no synergy between these two signals (not shown). An alternative hypothesis is that IFN protein that is prestored [35] and therefore not measurable by qPCR (not transcriptionally controlled) or by ELISA (too low sensitivity) could be released locally and mediates the observed ISG induction. In conclusion, we show here that airway epithelia rely on two parallel, redundant amplification loops to induce an IFN signature in response to influenza A infection. Only a small fraction of genes, mostly non ISGs, are induced by the virus in the absence of both IFN systems, and no ISG appears to rely specifically on one IFN system only. This complete redundancy may guarantee induction of antiviral responses even if one or the other IFN system is blocked, for instance by specific virally encoded antagonists. In contrast to the two redundant IFN loops in epithelia, the majority of immune cells respond only to IFN type I, thus potentially allowing for differential control of the epithelial antiviral state and the induction of immune responses: while high levels of IFN type I would activate both epithelia and immune cells, high levels of type III would specifically activate epithelial responses but leave immune responses unaffected, which may help limit immune-mediated pathology in the lung and at other mucosal surfaces. Materials and Methods Ethics statement All animal breeding was approved by the local ethical committee of the NIMR and is part of a project approved by the UK Home Office (licence number 80/2236). Breeding was conducted according to local guidelines and UK Home Office regulations under the Animals Scientific Procedures Act 1986 (ASPA). Viruses Influenza A virus strain A/PR/8/34 (H1N1) was grown in day 10 embryonated chicken eggs, and titrated on MDCK by 50% tissue culture infective dose (TCID50), according to the Spearman-Karber method. Mice All cells used in this study were derived from mice on the C57BL/6 background. MAVS−/− mice [36] and tracheae from TLR7−/− mice [37] were kindly provided by Dr. C. Reis e Sousa; MyD88−/− [38], IFNAR1−/− [39] and TRIF−/− mice [40] were kindly provided by Dr. A. O'Garra. These and C57BL/6 wt mice were bred in-house under SPF conditions. Tracheae from Irf3−/− [29]; Irf7−/− [29]; Irf3−/−Irf7−/− cells were also used. IL-28Rα−/− [12], IFNAR1−/− [39] and IL-28Rα−/−IFNAR1−/− cells were obtained from mice on a congenic B6.A2G-Mx1 background carrying an intact Mx1 gene [41]. To generate chimeric mice, naïve B6.A2G-Mx1 and B6.A2G-Mx1 IL-28Rα−/−IFNAR1−/− recipient mice were lethally irradiated with 1000 rad and reconstituted with donor B6.A2G-Mx1 BM cells (7×106) by intravenous injection. Chimeric mice were maintained for 7 weeks and chimerism assessed by IFNAR1 (MAR1-5A3 antibody) expression on Gr1+, CD19+, CD4+ and CD8+ cells in the blood (Fig. S6). Chimeric mice were then infected intranasally with 105 TCID50 of Influenza A/PR/8/34 in 30 µl PBS after anesthesia. Primary mouse tracheal epithelial cell culture (MTEC) Isolation and culture of primary MTEC were performed as previously described [42]. Briefly, cells isolated by enzymatic treatment were seeded onto 0.4 µm pore size clear polyester membrane (Corning) coated with a collagen solution. At confluence, media was removed from the upper chamber to establish an air- liquid interface (ALI). Fully differentiated, 10–14 days-old post ALI cultures were routinely used for experiments. In some experiments, cultures were infected in the presence of brefeldin A (2.5 µg/ml) or neutralizing anti-IL28A/B, anti-IL28B (R&D Systems) and blocking anti-IFNAR1 (MAR1-5A3) antibodies, at a final concentration of 10 µg/ml each. Immunofluorescence microscopy Differentiated, ALI day 14 cultures were fixed in 4% paraformaldehyde and permeabilized with 0.1% Triton X-100. Cells were then incubated with the indicated primary antibodies for 1 hour at room temperature, washed, incubated with fluorochrome-conjugated secondary antibody, washed and finally mounted. Image acquisition and processing information: (i) microscope: Olympus IX70; (II) magnification: 20×; (III) imaging medium: Vectashield with DAPI (Vector labs); (IV) fluorochromes: Alexa Fluor 488, Alexa Fluor 568. (V) acquisition software: Softworx. Images were processed with Image J. Virus infection The apical surface of MTEC cultures was washed extensively to remove accumulated mucins before inoculation with IAV (moi = 0.3). After incubation at 37°C for 1 h, the virus inoculum was removed and the cultures were incubated in complete growth medium for 24 hours. Aliquots of the supernatants were collected at different time points and titrated by ELISA as described below. Cells were then lysed to extract RNA or for detection of viral protein by Western blotting. RNA extraction and real time-PCR RNA was isolated from MTEC cultures by directly lysing the cells in the transwells, using the Qiagen RNeasy mini kit, according to the manufacturer's instructions. One microgram total RNA was reverse transcribed using the ThermoScript RT-PCR System kit (Invitrogen). The cDNA served as template for the amplification of genes of interest and the housekeeping gene (Hprt1) by real-time PCR, using TaqMan Gene Expression Assays (Applied Biosystems), universal PCR Master Mix (Applied Biosystems) and the ABI-PRISM 7900 sequence detection system (Applied Biosystems). The fold increase in mRNA expression was determined using the ΔΔCt method relatively to the values in mock treated samples, after normalization to Hprt1 gene expression. Microarray data analysis Total RNA harvested from MTEC cultures was hybridized using Affymetrix Mouse Genome 430 2.0 microarrays. The raw intensities values for each entity were preprocessed by RMA normalization against the median intensity in mock infected samples. Using GeneSpring 11.5, all transcripts were filtered based on signal values, to select the ones whose level of expression was in the 100–20th percentile, in at least 50% of samples. Student's t test (infected versus mock infected) or 2-way ANOVA (parameters: treatment and genotype) were performed to identify gene significantly differentially expressed relative to controls (≥4-fold change; p<0.01, Benjamini-Hochberg multiple test correction). Ingenuity Pathway Analysis (IPA) was used to select, annotate and visualize gene by function and pathway. ISGs were identified with the Interferome database (www.interferome.org/). Microarray data can be accessed at GEO under accession number GSE43710 for the superseries. Quantification of protein by ELISA Cell culture supernatants were harvested from the apical compartments of mock or IAV infected samples. IL-28A/B was measured using the IL-28A/B ELISA Duo kit (R&D Systems), IFNβ with the Verikine IFNβ ELISA kit (PBL Interferon Source). Supporting Information Figure S1 Flow cytometric measurement of influenza infection of epithelial cells. Fully differentiated MTEC cultures were infected with A/PR/8/34 (H1N1) (moi = 0.3). At 24 hours post infection, cell were washed, trypsinized to get single cell suspension and then fixed in 4% paraformaldehyde. After permeabilization, cells were stained with anti-NP/M-FITC antibodies (Imagen Oxoid) and then analysed by flow cytometry. (TIF) Click here for additional data file. Figure S2 Heat inactivation blocks virus replication but still permits virus binding to cells. (A) Fully differentiated MTEC cultures were infected with either PR8 or PR8 (HI) (heat inactivated at 65°C) (moi = 3). At the indicated time point, cells were washed and lysed in 1% Triton X-100. Cell extracts were then run on a polyacrylamide gel, transferred to nitrocellulose membrane and probed with specific antibodies plus horseradish peroxidase-conjugate secondary antibody (Biorad). The early appearance of influenza Matrix protein (M) testifies binding of the virions to the target cells; the later positiveness for influenza NS is indicative of viral replication. (B) MTEC cultures were infected with PR8 at a moi = 0.3. 36 hours post infection supernatants from the apical side were collected and titrated on MDCK. Virus titres were expressed as tissue culture infectious dose 50 (TCID50). (TIF) Click here for additional data file. Figure S3 Transcriptional response to influenza infection in the absence of MAVS. Total RNA from mock and PR8 infected wild-type and MAVS−/− cells was analysed using Affymetrix Mouse Genome 430 2.0 microarrays, at 24 hpi. The raw intensities values for each entity were processed by RMA normalization against the median intensity in mock infected samples. All transcripts were filtered based on signal values, to select the ones whose level of expression was in the 100–20th percentile, in at least 50% of samples. Supervised analysis was performed using statistical filtering (≥4-fold change relative to mock infected wild-type in at least one treatment group; 2-way ANOVA, p<0.01, Benjamini-Hochberg multiple test correction). (A) K-means clustering of the differentially expressed genes. (B) Heat map of the upregulated genes shown in (A). The range of fold changes is expressed in a log2 scale. (TIF) Click here for additional data file. Figure S4 Identification of genes induced independently of IFN type I or type III. Total RNA from mock and PR8 infected cells was analysed using Affymetrix Mouse Genome 430 2.0 microarrays at 24 hpi. Supervised analysis was performed as in figure 5A, using statistical filtering (≥4-fold change relative to mock infected wt; 2-way ANOVA, p<0.01, Benjamini-Hochberg multiple test correction). The upregulated genes shown in figure 5A were further partitioned by K-means clustering to generate two clusters of which the one shown in (A) includes the genes that were upregulated upon infection in all genotypes. (B) shows the same cluster as a heat map. The cluster in (C) includes the 110 genes that were upregulated only in wild-type and single knock-out epithelia. 58 genes of the latter list were recognized as ISG by the INTERFEROME database. The range of fold changes is expressed in a log2 scale. (TIF) Click here for additional data file. Figure S5 Assessment of genes differentially expressed in the absence of either IFN type I or type III. Total RNA from mock and PR8 infected cells was analysed using Affymetrix Mouse Genome 430 2.0 microarrays at 24 hpi. Supervised analysis was performed as in figure 5A, using statistical filtering (≥4-fold change relative to mock infected wt; 2-way ANOVA, p<0.01, Benjamini-Hochberg multiple test correction). The upregulated genes described in figure 5A were further filtered (≥4-fold change in expression for corresponding time points for the following pairs of condition: IFNAR1−/− versus wild-type; IL-28Rα−/− versus wild-type; IL-28Rα−/− versus IFNAR1−/−) to generate the heat map shown here. (TIF) Click here for additional data file. Figure S6 Confirmation of successful bone marrow grafting in chimera experiments. Seven weeks after BM graft, mice were bled from the tail vein. Blood was collected in heparin and stained with the following antibodies: CD19 (6D5), Gr-1 (1A8), CD4 (RM4-5), CD8 (53-6.7) and IFNAR1 (MAR1-5A3). Samples were read by FACSCanto (BD Bioscience) and analyzed by FlowJo 9.6.2. Cells from wild-type C57BL/6 (solid grey), IFNAR1−/− (black line) and chimeric mice (coloured lines) are shown. (TIF) Click here for additional data file.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                J Virol
                J. Virol
                jvi
                jvi
                JVI
                Journal of Virology
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                0022-538X
                1098-5514
                3 October 2018
                27 November 2018
                15 December 2018
                27 November 2018
                : 92
                : 24
                : e01091-18
                Affiliations
                [a ]Institute of Virology and Immunology, Mittelhäusern, Switzerland
                [b ]Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
                [c ]Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
                University of Texas Southwestern Medical Center
                Author notes
                Address correspondence to Artur Summerfield, Artur.summerfield@ 123456ivi.admin.ch .

                R.O.B. and P.M. contributed equally to this article.

                Citation García-Nicolás O, Braun RO, Milona P, Lewandowska M, Dijkman R, Alves MP, Summerfield A. 2018. Targeting of the nasal mucosa by Japanese encephalitis virus for non-vector-borne transmission. J Virol 92:e01091-18. https://doi.org/10.1128/JVI.01091-18.

                Author information
                https://orcid.org/0000-0003-0320-2743
                Article
                JVI01091-18
                10.1128/JVI.01091-18
                6258954
                30282716
                05a2c0c7-85fd-4a3a-82df-b1311d683268
                Copyright © 2018 García-Nicolás et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 16 July 2018
                : 28 September 2018
                Page count
                Figures: 10, Tables: 1, Equations: 0, References: 79, Pages: 16, Words: 9943
                Funding
                Funded by: EC | Seventh Framework Programme (FP7), https://doi.org/10.13039/100011102;
                Award ID: 278433-PREDEMICS
                Award Recipient : Award Recipient :
                Categories
                Pathogenesis and Immunity
                Custom metadata
                December 2018

                Microbiology & Virology
                japanese encephalitis virus,direct contact transmission,macrophages,nasal epithelial cells,pig

                Comments

                Comment on this article