key: cord-0719511-oua5vylw authors: Bedford, James G.; Infusini, Giuseppe; Dagley, Laura F.; Villalon-Letelier, Fernando; Zheng, Ming Z. M.; Bennett-Wood, Vicki; Reading, Patrick C.; Wakim, Linda M. title: Airway Exosomes Released During Influenza Virus Infection Serve as a Key Component of the Antiviral Innate Immune Response date: 2020-05-12 journal: Front Immunol DOI: 10.3389/fimmu.2020.00887 sha: 4d124b37e288a897cd92c9513f7f6915858b1d0b doc_id: 719511 cord_uid: oua5vylw Exosomes are extracellular vesicles secreted by cells that have an important biological function in intercellular communication by transferring biologically active proteins, lipids, and RNAs to neighboring or distant cells. While a role for exosomes in antimicrobial defense has recently emerged, currently very little is known regarding the nature and functional relevance of exosomes generated in vivo, particularly during an active viral infection. Here, we characterized exosomes released into the airways during influenza virus infection. We show that these vesicles dynamically change in protein composition over the course of infection, increasing expression of host proteins with known anti-influenza activity, and viral proteins with the potential to trigger host immune responses. We show that exosomes released into the airways during influenza virus infection trigger pulmonary inflammation and carry viral antigen that can be utilized by antigen presenting cells to drive the induction of a cellular immune response. Moreover, we show that attachment factors for influenza virus, namely α2,3 and α2,6-linked sialic acids, are present on the surface of airway exosomes and these vesicles have the ability to neutralize influenza virus, thereby preventing the virus from binding and entering target cells. These data reveal a novel role for airway exosomes in the antiviral innate immune defense against influenza virus infection. Exosomes are small vesicles (30-100 nm in diameter) of endocytic origin that are released from cells into the extracellular environment during normal and pathological conditions (1) . They are formed by the inward budding of late endosomal membranes that give rise to intracellular multivesicular bodies (MVBs) which fuse with the plasma membrane releasing the intraluminal exosomes into the extracellular space. They are secreted by virtually all cell types and are present in bodily fluids such as blood, urine, saliva, breast milk, bronchial, and nasal lavage (2-6). Although the protein composition of exosomes reflects that of the parent cell, exosomes are generally rich in tetraspanins (CD9, CD63, CD81), heat shock proteins and Rab proteins. Exosomes are an important tool for intercellular communication through the transfer of biologically active proteins, lipids, and RNAs (7, 8) . Only recently has a role for exosomes in viral pathogenesis and antimicrobial defense emerged. In vitro studies have demonstrated that exosomes play a dual role, promoting pathogen transmission, and exacerbation of infection in some instances but contributing to host defense and control of infection in others. For example, exosomes released from cells infected with an array of RNA viruses including human immunodeficiency virus, hepatitis C virus, human Tcell lymphotropic virus and dengue virus, carry viral proteins and RNA and these vesicles can facilitate virion-independent transfer of replication-competent virus between cells (1, 9) . In this context, exosomes facilitate the spread of the virus. However, exosomes can also limit the spread of virus through a variety of mechanisms. Exosomes isolated from certain cell lines can express an array of interferon (IFN)-induced antiviral proteins which interfere with viral replication and enhance the ability of uninfected cells to resist infection (10) . Exosomes serve as a method for the intercellular transfer of these proteins, conferring broad-spectrum viral resistance. Exosomes generated in the presence of type I IFN and expressing an array of antiviral molecules could render cells resistant to in vitro infection by hepatitis B virus (11) or Dengue virus (12) . Furthermore, exosomes recovered from human respiratory epithelial cell lines have also been reported to bind and neutralize human influenza virus (13) . To date, nearly all data supporting the antiviral activity of exosomes has been derived from in vitro studies where exosomes were derived from cultured cell lines. Currently, very little is known regarding the characteristics of exosomes generated in vivo, how they might change during infection and whether they contribute to the antiviral immune response. Here we profiled exosomes released into the airways of mice over the course of an influenza virus infection. Proteomic analysis of these vesicles revealed temporal changes in protein composition, with exosomes gaining host proteins with known anti-influenza activity, as well as viral proteins with the potential to trigger host immune responses. This altered protein composition afforded these vesicles new capabilities allowing them to trigger pulmonary inflammation and serve as a source of viral antigen that could be utilized for the induction of a cellular immune response. We also show that attachment factors for influenza virus, namely α2,3 and α2,6-linked sialic acids, are present on the surface of airway exosomes and these vesicles have the ability to neutralize influenza virus, thereby preventing the virus from binding and entering target cells. These data highlight a variety of biological functions by which exosomes released into the airways during an influenza virus infection assist in the antiviral innate immune response. To gain insight into the nature of exosomes generated in vivo, we optimized protocols to isolate exosomes from murine bronchial alveolar lavage fluid (BALf) and profiled the protein composition of these vesicles at different time-points over the course of influenza virus infection using mass spectrometry. Mice infected with influenza virus via the intranasal route were euthanized at various times post-infection and BALf was collected, filtered (0.2 µm) and exosomes were purified by ultra-centrifugation (14) followed by further enrichment by immune-absorption onto magnetic beads coated with antibodies against the tetraspanin CD9, a common exosomal marker (15) (Supplementary Figure 1) . The preparation resulted in the isolation of highly pure exosome preparations. To confirm exosome preparations were free from influenza virion contamination we stained the vesicles with anti-CD9 goldlabeled particles and visualized the samples using electron microscopy. We failed to identify any influenza virions, which can be easily discriminated from exosomes based on morphology, in our purified exosome preparations ( Figure 1A) . Moreover, when we cultured purified airway exosomes recovered from mice on day 2 post influenza virus infection on MDCKs, a canine kidney cell line that is highly permissive to influenza virus infection, we did not observe any overt infection, as measured by an increase in influenza virus nucleoprotein (NP) staining, implying that there were no infectious virions in our exosome preparation or the level present was below our limit of detection ( Figure 1B) . We next performed semiquantitative proteomics analysis on exosome preparations which resulted in the identification of 2,688 proteins. To confirm the exosome preparations were pure we firstly checked our dataset for a panel of classic exosome markers and found that these were present and highly abundant in our samples, whereas protein markers for other cellular compartments (i.e., ER or Golgi) were rare (Supplementary Figure 2 and Supplementary Table 1) . Principle components analysis of the exosome samples revealed striking differences in the protein composition of these vesicles over the course of the influenza virus infection ( Figure 1C) . Moreover, the effect was enduring, as even at day 20 post influenza infection the protein composition of airway exosomes failed to return to the baseline composition of exosomes recovered from naïve animals. To identify proteins elevated in the exosomes in response to influenza virus infection we refined our analysis to proteins that increased in abundance over the acute phase (day 3-7) of infection (Figures 1C,D and Supplementary Tables 2, 3) . Interestingly, many antiviral proteins with known anti-influenza activity, including members of the IFIT and IFITM families, were present at high levels in exosomes recovered from mice during the acute phase of infection ( Figure 1F ). In addition, expression levels of proteins of the mucin family were also markedly enhanced on exosomes during the acute phase of infection. In previous studies, exosomes recovered from respiratory epithelial cell lines were shown to express sialylated mucins and these vesicles could bind and neutralize influenza virus (13) . In addition, four influenza virus proteins (HA, NS1, NP, and M1) were identified in airway exosomes ( Figure 1G ). Together, these data confirm the presence of host proteins with known anti-influenza activity, and viral proteins with the potential to trigger host immune responses, in exosomes derived from the airways of influenza virusinfected mice. We tested the capacity of exosomes recovered from the BALf of influenza virus-infected mice to trigger pulmonary inflammation. To this end, we intranasally transferred into the airways of naïve mice an equal amount of exosomes recovered from the BALf of either naïve mice, mice infected 2 days prior with influenza virus, or mice given an inflammatory agent (poly I:C). Four days following exosome delivery we assessed the level of inflammation by measuring cytokine levels in the BALf and immune cell infiltration into the lung tissue. As a positive control for pulmonary inflammation, we also included a cohort of mice directly infected with influenza virus. The delivery of exosomes recovered from the airways of influenza virus-infected mice resulted in the production of IL-6, MCP-1 and TNF, which was a very similar inflammatory profile to that observed following direct infection with influenza virus (Figures 2A-C) . Widening our analysis to include assessment of a more extensive panel of inflammatory cytokines revealed that exosomes recovered from the airways of influenza virusinfected mice also resulted in the production of both type I and type II interferon (Supplementary Figure 3) . In contrast, the intranasal delivery of exosomes recovered from naïve or poly I:C treated mice did not evoke the release of any cytokines (Figures 2A-C) , implying that the PAMPs or potential DAMPs loaded into the exosomes generated during virus infection were essential to trigger inflammation. Exosomes derived from influenza virus infected mice did contain viral RNA which may serve as the PAMP triggering the observed inflammatory response (Supplementary Figure 4) . Consistent with the capacity to trigger the release of inflammatory cytokines, the intranasal delivery of exosomes recovered from the BALf of influenza virus-infected mice, but not naïve mice also resulted in the recruitment of neutrophils while the number of CD8 + and CD4 + T cells remained stable irrespective of the treatment (Figures 2D-F) . Thus, exosomes released into the airways during an influenza virus infection are inflammatory, causing the release of cytokines/chemokines and resulting in the recruitment of innate immune cells. Previous studies have reported a role for exosomes in antigen presentation demonstrating that they can deliver immunologically relevant proteins/peptides to antigen presenting cells which in turn can utilize these antigens to Frontiers in Immunology | www.frontiersin.org initiate a T cell response. As exosomes released into the airways during influenza virus infection carried a number of viral proteins (Figure 1H ), as well as major histocompatibility class I and II complexes (Figure 3A) , we next assessed whether these vesicles could initiate CD8 + and CD4 + T cell activation. To analyze antigen-specific, MHC-restricted T cell stimulation by exosomes, we infected mice with a recombinant influenza virus engineered to express the CD8 (SIINFEKL, x31-OVA 1 ) (16) epitope from the model antigen Ovalbumin (OVA). Then, on days 1-4 post-infection, exosomes recovered from the BALf were cultured with carboxyfluorescein diacetate succinimidyl ester (CFSE) labeled OVA-specific OT-I.CD8 + T cells with or without dendritic cells. Irrespective of the time point at which they were recovered, exosomes failed to drive OT-I.CD8 + T cell proliferation if dendritic cells were not added to the cultures (Figures 3B,C) . In contrast, CD8 + T cell proliferation was observed when exosomes and dendritic cells were present in the cultures, implying that airway exosomes can act as a source of antigen but cannot present CD8 + T cell epitopes directly. Dendritic cells fed exosomes recovered from the BALf at day 2-3 p.i. were capable of driving the most robust T cell division, which is consistent with our proteomic analysis demonstrating the viral antigen load in the exosomes peaked