Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Virol ; 97(11): e0090623, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37843369

ABSTRACT

IMPORTANCE: It is well known that influenza A viruses (IAV) initiate host cell infection by binding to sialic acid, a sugar molecule present at the ends of various sugar chains called glycoconjugates. These sugar chains can vary in chain length, structure, and composition. However, it remains unknown if IAV strains preferentially bind to sialic acid on specific glycoconjugate type(s) for host cell infection. Here, we utilized CRISPR gene editing to abolish sialic acid on different glycoconjugate types in human lung cells, and evaluated human versus avian IAV infections. Our studies show that both human and avian IAV strains can infect human lung cells by utilizing any of the three major sialic acid-containing glycoconjugate types, specifically N-glycans, O-glycans, and glycolipids. Interestingly, simultaneous elimination of sialic acid on all three major glycoconjugate types in human lung cells dramatically decreased human IAV infection, yet had little effect on avian IAV infection. These studies show that avian IAV strains effectively utilize other less prevalent glycoconjugates for infection, whereas human IAV strains rely on a limited repertoire of glycoconjugate types. The remarkable ability of avian IAV strains to utilize diverse glycoconjugate types may allow for easy transmission into new host species.


Subject(s)
Influenza A virus , Influenza, Human , Lung , Receptors, Cell Surface , Animals , Humans , Carrier Proteins/metabolism , Glycoconjugates/metabolism , Influenza A virus/metabolism , Lung/virology , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Sugars/metabolism , Influenza in Birds/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism
2.
PLoS Pathog ; 17(5): e1009517, 2021 05.
Article in English | MEDLINE | ID: mdl-33970958

ABSTRACT

It is well documented that influenza A viruses selectively package 8 distinct viral ribonucleoprotein complexes (vRNPs) into each virion; however, the role of host factors in genome assembly is not completely understood. To evaluate the significance of cellular factors in genome assembly, we generated a reporter virus carrying a tetracysteine tag in the NP gene (NP-Tc virus) and assessed the dynamics of vRNP localization with cellular components by fluorescence microscopy. At early time points, vRNP complexes were preferentially exported to the MTOC; subsequently, vRNPs associated on vesicles positive for cellular factor Rab11a and formed distinct vRNP bundles that trafficked to the plasma membrane on microtubule networks. In Rab11a deficient cells, however, vRNP bundles were smaller in the cytoplasm with less co-localization between different vRNP segments. Furthermore, Rab11a deficiency increased the production of non-infectious particles with higher RNA copy number to PFU ratios, indicative of defects in specific genome assembly. These results indicate that Rab11a+ vesicles serve as hubs for the congregation of vRNP complexes and enable specific genome assembly through vRNP:vRNP interactions, revealing the importance of Rab11a as a critical host factor for influenza A virus genome assembly.


Subject(s)
Genome, Viral , Influenza A virus/genetics , Influenza, Human/virology , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , Virus Assembly , rab GTP-Binding Proteins/metabolism , A549 Cells , HEK293 Cells , Humans , Influenza A virus/isolation & purification , Influenza, Human/genetics , Ribonucleoproteins/genetics , Viral Proteins/genetics , Virus Replication , rab GTP-Binding Proteins/genetics
3.
PLoS Pathog ; 16(9): e1008842, 2020 09.
Article in English | MEDLINE | ID: mdl-32898178

ABSTRACT

Signaling through retinoic acid inducible gene I (RIG-I) like receptors (RLRs) is tightly regulated, with activation occurring upon sensing of viral nucleic acids, and suppression mediated by negative regulators. Under homeostatic conditions aberrant activation of melanoma differentiation-associated protein-5 (MDA5) is prevented through editing of endogenous dsRNA by RNA editing enzyme Adenosine Deaminase Acting on RNA (ADAR1). In addition, ADAR1 is postulated to play pro-viral and antiviral roles during viral infections that are dependent or independent of RNA editing activity. Here, we investigated the importance of ADAR1 isoforms in modulating influenza A virus (IAV) replication and revealed the opposing roles for ADAR1 isoforms, with the nuclear p110 isoform restricting versus the cytoplasmic p150 isoform promoting IAV replication. Importantly, we demonstrate that p150 is critical for preventing sustained RIG-I signaling, as p150 deficient cells showed increased IFN-ß expression and apoptosis during IAV infection, independent of RNA editing activity. Taken together, the p150 isoform of ADAR1 is important for preventing sustained RIG-I induced IFN-ß expression and apoptosis during viral infection.


Subject(s)
Adenosine Deaminase/metabolism , Apoptosis , DEAD Box Protein 58/metabolism , Influenza A virus/physiology , Influenza, Human/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Virus Replication , A549 Cells , Adenosine Deaminase/genetics , DEAD Box Protein 58/genetics , HEK293 Cells , Humans , Influenza, Human/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , RNA-Binding Proteins/genetics , Receptors, Immunologic
4.
Life Sci Alliance ; 3(8)2020 08.
Article in English | MEDLINE | ID: mdl-32611549

ABSTRACT

HA plays a critical role in influenza infection and, thus HA is a potential target for antivirals. Recently, our laboratories have described a novel fusion inhibitor, termed CBS1117, with EC50 ∼3 µM against group 1 HA. In this work, we characterize the binding properties of CBS1117 to avian H5 HA by x-ray crystallography, NMR, and mutagenesis. The x-ray structure of the complex shows that the compound binds near the HA fusion peptide, a region that plays a critical role in HA-mediated fusion. NMR studies demonstrate binding of CBS1117 to H5 HA in solution and show extensive hydrophobic contacts between the compound and HA surface. Mutagenesis studies further support the location of the compound binding site proximal to the HA fusion peptide and identify additional amino acids that are important to compound binding. Together, this work gives new insights into the CBS1117 mechanism of action and can be exploited to further optimize this compound and better understand the group specific activity of small-molecule inhibitors of HA-mediated entry.


Subject(s)
Antiviral Agents/chemistry , Hemagglutinins/ultrastructure , Animals , Antiviral Agents/pharmacology , Binding Sites/drug effects , Birds/virology , Crystallography, X-Ray/methods , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinins/metabolism , Humans , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/drug effects , Influenza in Birds/metabolism , Influenza, Human/metabolism , Models, Molecular , Orthomyxoviridae Infections , Virus Internalization/drug effects
5.
J Virol ; 94(9)2020 04 16.
Article in English | MEDLINE | ID: mdl-32075925

ABSTRACT

Seasonal influenza virus infections cause mild illness in healthy adults, as timely viral clearance is mediated by the functions of cytotoxic T cells. However, avian H5N1 influenza virus infections can result in prolonged and fatal illness across all age groups, which has been attributed to the overt and uncontrolled activation of host immune responses. Here, we investigate how excessive innate immune responses to H5N1 impair subsequent adaptive T cell responses in the lungs. Using recombinant H1N1 and H5N1 strains sharing 6 internal genes, we demonstrate that H5N1 (2:6) infection in mice causes higher stimulation and increased migration of lung dendritic cells to the draining lymph nodes, resulting in greater numbers of virus-specific T cells in the lungs. Despite robust T cell responses in the lungs, H5N1 (2:6)-infected mice showed inefficient and delayed viral clearance compared with H1N1-infected mice. In addition, we observed higher levels of inhibitory signals, including increased PD-1 and interleukin-10 (IL-10) expression by cytotoxic T cells in H5N1 (2:6)-infected mice, suggesting that delayed viral clearance of H5N1 (2:6) was due to the suppression of T cell functions in vivo Importantly, H5N1 (2:6)-infected mice displayed decreased numbers of tissue-resident memory T cells compared with H1N1-infected mice; however, despite the decreased number of tissue-resident memory T cells, H5N1 (2:6) was protected against a heterologous challenge from H3N2 virus (X31). Taken together, our study provides mechanistic insight for the prolonged viral replication and protracted illness observed in H5N1-infected patients.IMPORTANCE Influenza viruses cause upper respiratory tract infections in humans. In healthy adults, seasonal influenza virus infections result in mild disease. Occasionally, influenza viruses endemic in domestic birds can cause severe and fatal disease even in healthy individuals. In avian influenza virus-infected patients, the host immune system is activated in an uncontrolled manner and is unable to control infection in a timely fashion. In this study, we investigated why the immune system fails to effectively control a modified form of avian influenza virus. Our studies show that T cell functions important for clearing virally infected cells are impaired by higher negative regulatory signals during modified avian influenza virus infection. In addition, memory T cell numbers were decreased in modified avian influenza virus-infected mice. Our studies provide a possible mechanism for the severe and prolonged disease associated with avian influenza virus infections in humans.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza, Human/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Birds , Humans , Immunity, Innate/immunology , Immunologic Memory/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A virus/physiology , Influenza, Human/metabolism , Lung/virology , Mice , Orthomyxoviridae Infections/virology , Virus Replication/genetics
6.
Cell Rep ; 23(2): 596-607, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29642015

ABSTRACT

The emergence of influenza A viruses (IAVs) from zoonotic reservoirs poses a great threat to human health. As seasonal vaccines are ineffective against zoonotic strains, and newly transmitted viruses can quickly acquire drug resistance, there remains a need for host-directed therapeutics against IAVs. Here, we performed a genome-scale CRISPR/Cas9 knockout screen in human lung epithelial cells with a human isolate of an avian H5N1 strain. Several genes involved in sialic acid biosynthesis and related glycosylation pathways were highly enriched post-H5N1 selection, including SLC35A1, a sialic acid transporter essential for IAV receptor expression and thus viral entry. Importantly, we have identified capicua (CIC) as a negative regulator of cell-intrinsic immunity, as loss of CIC resulted in heightened antiviral responses and restricted replication of multiple viruses. Therefore, our study demonstrates that the CRISPR/Cas9 system can be utilized for the discovery of host factors critical for the replication of intracellular pathogens.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Knockout Techniques/methods , Influenza A Virus, H5N1 Subtype/physiology , A549 Cells , Gene Library , Genome, Human , Humans , Influenza A Virus, H5N1 Subtype/genetics , Lentivirus/genetics , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Virus Internalization , Virus Replication
7.
PLoS Pathog ; 13(3): e1006270, 2017 03.
Article in English | MEDLINE | ID: mdl-28282445

ABSTRACT

The cellular and molecular mechanisms underpinning the unusually high virulence of highly pathogenic avian influenza H5N1 viruses in mammalian species remains unknown. Here, we investigated if the cell tropism of H5N1 virus is a determinant of enhanced virulence in mammalian species. We engineered H5N1 viruses with restricted cell tropism through the exploitation of cell type-specific microRNA expression by incorporating microRNA target sites into the viral genome. Restriction of H5N1 replication in endothelial cells via miR-126 ameliorated disease symptoms, prevented systemic viral spread and limited mortality, despite showing similar levels of peak viral replication in the lungs as compared to control virus-infected mice. Similarly, restriction of H5N1 replication in endothelial cells resulted in ameliorated disease symptoms and decreased viral spread in ferrets. Our studies demonstrate that H5N1 infection of endothelial cells results in excessive production of cytokines and reduces endothelial barrier integrity in the lungs, which culminates in vascular leakage and viral pneumonia. Importantly, our studies suggest a need for a combinational therapy that targets viral components, suppresses host immune responses, and improves endothelial barrier integrity for the treatment of highly pathogenic H5N1 virus infections.


Subject(s)
Endothelial Cells/virology , Influenza A Virus, H5N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Orthomyxoviridae Infections/pathology , Viral Tropism/physiology , Animals , Blotting, Western , Disease Models, Animal , Female , Ferrets , Flow Cytometry , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Virulence
8.
PLoS Pathog ; 12(7): e1005754, 2016 07.
Article in English | MEDLINE | ID: mdl-27438481

ABSTRACT

Retinoic acid inducible gene-I (RIG-I) is an innate RNA sensor that recognizes the influenza A virus (IAV) RNA genome and activates antiviral host responses. Here, we demonstrate that RIG-I signaling plays a crucial role in restricting IAV tropism and regulating host immune responses. Mice deficient in the RIG-I-MAVS pathway show defects in migratory dendritic cell (DC) activation, viral antigen presentation, and priming of CD8+ and CD4+ T cell responses during IAV infection. These defects result in decreased frequency of polyfunctional effector T cells and lowered protection against heterologous IAV challenge. In addition, our data show that RIG-I activation is essential for protecting epithelial cells and hematopoietic cells from IAV infection. These diverse effects of RIG-I signaling are likely imparted by the actions of type I interferon (IFN), as addition of exogenous type I IFN is sufficient to overcome the defects in antigen presentation by RIG-I deficient BMDC. Moreover, the in vivo T cell defects in RIG-I deficient mice can be overcome by the activation of MDA5 -MAVS via poly I:C treatment. Taken together, these findings demonstrate that RIG-I signaling through MAVS is critical for determining the quality of polyfunctional T cell responses against IAV and for providing protection against subsequent infection from heterologous or novel pandemic IAV strains.


Subject(s)
Membrane Proteins/immunology , Nerve Tissue Proteins/immunology , Orthomyxoviridae Infections/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Coculture Techniques , Dendritic Cells/immunology , Disease Models, Animal , Flow Cytometry , Influenza A virus/immunology , Interferon Type I/biosynthesis , Interferon Type I/immunology , Lymphocyte Activation/immunology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Polymerase Chain Reaction , Receptors, Cell Surface , T-Lymphocytes/metabolism
9.
Curr Protoc Microbiol ; Chapter 15: 15G.4.1-15G.4.16, 2013.
Article in English | MEDLINE | ID: mdl-23686828

ABSTRACT

The incorporation of a fluorescent reporter gene into a replication-competent influenza A virus (IAV) has made it possible to trace IAV infection in vivo. This protocol describes the process of inserting a green fluorescent protein (GFP) reporter into the IAV genome using the established reverse genetics system. The strategy begins with the reorganization of segment eight of the IAV genome, during which the open reading frames of nonstructural protein 1 (NS1) and the nuclear export protein (NEP) are separated to allow for GFP fusion to the NS1 protein. The NS1, GFP, and NEP open reading frames (ORF) are then cloned into the IAV rescue system backbone. Upon construction of the GFP-encoding segment eight rescue plasmid, recombinant NS1-GFP influenza virus can be rescued via co-transfection with the remaining seven rescue plasmids. The generated NS1-GFP IAV can subsequently be used to visualize infected cells, both in vitro and in vivo.


Subject(s)
Artificial Gene Fusion , Genes, Reporter , Green Fluorescent Proteins/metabolism , Influenza A virus/genetics , Reverse Genetics/methods , Staining and Labeling/methods , Green Fluorescent Proteins/genetics , Influenza A virus/growth & development , Viral Nonstructural Proteins/genetics , Virology/methods
10.
Cell Rep ; 3(1): 23-9, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23333274

ABSTRACT

Influenza A virus is unique as an RNA virus in that it replicates in the nucleus and undergoes splicing. With only ten major proteins, the virus must gain nuclear access, replicate, assemble progeny virions in the cytoplasm, and then egress. In an effort to elucidate the coordination of these events, we manipulated the transcript levels from the bicistronic nonstructural segment that encodes the spliced virus product responsible for genomic nuclear export. We find that utilization of an erroneous splice site ensures the slow accumulation of the viral nuclear export protein (NEP) while generating excessive levels of an antagonist that inhibits the cellular response to infection. Modulation of this simple transcriptional event results in improperly timed export and loss of virus infection. Together, these data demonstrate that coordination of the influenza A virus life cycle is set by a "molecular timer" that operates on the inefficient splicing of a virus transcript.


Subject(s)
Influenza A virus/genetics , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , RNA Splicing/genetics , Active Transport, Cell Nucleus , Animals , Gene Silencing , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Small Interfering/metabolism , Ribonucleoproteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication
11.
J Virol ; 86(24): 13475-85, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035211

ABSTRACT

Influenza A virus (IAV) is an unremitting virus that results in significant morbidity and mortality worldwide. Key to the viral life cycle is the RNA-dependent RNA polymerase (RdRp), a heterotrimeric complex responsible for both transcription and replication of the segmented genome. Here, we demonstrate that the viral polymerase utilizes a small RNA enhancer to regulate enzymatic activity and maintain stoichiometric balance of the viral genome. We demonstrate that IAV synthesizes small viral RNAs (svRNAs) that interact with the viral RdRp in order to promote genome replication in a segment-specific manner. svRNAs localize to the nucleus, the site of IAV replication, are synthesized from the positive-sense genomic intermediate, and interact within a novel RNA binding channel of the polymerase PA subunit. Synthetic svRNAs promote polymerase activity in vitro, while loss of svRNA inhibits viral RNA synthesis in a segment-specific manner. Taking these observations together, we mechanistically define svRNA as a small regulatory enhancer RNA, which functions to promote genome replication and maintain segment balance through allosteric modulation of polymerase activity.


Subject(s)
Influenza A virus/enzymology , RNA-Dependent RNA Polymerase/metabolism , Blotting, Western , Cell Line , Genome, Viral , Humans , Immunoprecipitation , Influenza A virus/genetics
12.
Proc Natl Acad Sci U S A ; 107(25): 11525-30, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20534471

ABSTRACT

The discovery of regulatory small RNAs continues to reshape paradigms in both molecular biology and virology. Here we describe examples of influenza A virus-derived small viral RNAs (svRNAs). svRNAs are 22-27 nt in length and correspond to the 5' end of each of the viral genomic RNA (vRNA) segments. Expression of svRNA correlates with the accumulation of vRNA and a bias in RNA-dependent RNA polymerase (RdRp) activity from transcription toward genome replication. Synthesis of svRNA requires the RdRp, nucleoprotein and the nuclear export protein NS2. In addition, svRNA is detectable during replication of various influenza A virus subtypes across multiple host species and associates physically with the RdRp. We demonstrate that depletion of svRNA has a minimal impact on mRNA and complementary vRNA (cRNA) but results in a dramatic loss of vRNA in a segment-specific manner. We propose that svRNA triggers the viral switch from transcription to replication through interactions with the viral polymerase machinery. Taken together, the discovery of svRNA redefines the mechanistic switch of influenza virus transcription/replication and provides a potential target for broad-range, anti-influenza virus-based therapeutics.


Subject(s)
DNA Replication , Influenza A virus/genetics , RNA, Small Interfering/genetics , Transcription, Genetic , Active Transport, Cell Nucleus , Cell Line, Tumor , DNA-Directed RNA Polymerases/metabolism , Humans , MicroRNAs/genetics , Models, Genetic , Oligonucleotides/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Sequence Analysis, DNA
13.
Proc Natl Acad Sci U S A ; 107(25): 11519-24, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20534531

ABSTRACT

MicroRNAs (miRNAs) are short noncoding RNAs that exert posttranscriptional gene silencing and regulate gene expression. In addition to the hundreds of conserved cellular miRNAs that have been identified, miRNAs of viral origin have been isolated and found to modulate both the viral life cycle and the cellular transcriptome. Thus far, detection of virus-derived miRNAs has been largely limited to DNA viruses, suggesting that RNA viruses may be unable to exploit this aspect of transcriptional regulation. Lack of RNA virus-produced miRNAs has been attributed to the replicative constraints that would incur following RNase III processing of a genomic hairpin. To ascertain whether the generation of viral miRNAs is limited to DNA viruses, we investigated whether influenza virus could be designed to deliver functional miRNAs without affecting replication. Here, we describe a modified influenza A virus that expresses cellular microRNA-124 (miR-124). Insertion of the miR-124 hairpin into an intron of the nuclear export protein transcript resulted in endogenous processing and functional miR-124. We demonstrate that a viral RNA genome incorporating a hairpin does not result in segment instability or miRNA-mediated genomic targeting, thereby permitting the virus to produce a miRNA without having a negative impact on viral replication. This work demonstrates that RNA viruses can produce functional miRNAs and suggests that this level of transcriptional regulation may extend beyond DNA viruses.


Subject(s)
Influenza A virus/genetics , MicroRNAs/genetics , RNA Processing, Post-Transcriptional , RNA, Viral , Animals , Dogs , Gene Expression Regulation, Viral , Genetic Engineering/methods , Genetic Vectors , Humans , Mice , MicroRNAs/metabolism , Models, Genetic , Molecular Sequence Data , Plasmids/metabolism , RNA Interference , RNA, Viral/metabolism
14.
Nat Biotechnol ; 27(6): 572-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19483680

ABSTRACT

Influenza A virus leads to yearly epidemics and sporadic pandemics. Present prophylactic strategies focus on egg-grown, live, attenuated influenza vaccines (LAIVs), in which attenuation is generated by conferring temperature sensitivity onto the virus. Here we describe an alternative approach to attenuating influenza A virus based on microRNA-mediated gene silencing. By incorporating nonavian microRNA response elements (MREs) into the open-reading frame of the viral nucleoprotein, we generate reassortant LAIVs for H1N1 and H5N1 that are attenuated in mice but not in eggs. MRE-based LAIVs show a greater than two-log reduction in mortality compared with control viruses lacking MREs and elicit a diverse antibody response. This approach might be combined with existing LAIVs to increase attenuation and improve vaccine safety.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines , MicroRNAs , RNA Interference , Vaccines, Attenuated , Animals , Body Weight , Cell Line , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Mice , MicroRNAs/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , RNA, Viral/genetics , Response Elements/genetics , Species Specificity , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...