Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Trends Immunol ; 41(1): 3-6, 2020 01.
Article in English | MEDLINE | ID: mdl-31791719

ABSTRACT

A recent article by Sung et al. identified the CLEC2 platelet receptor as an important factor of lethal dengue virus infection. Formation of neutrophil extracellular traps via crosstalk with CLEC5A and TLR2 neutrophils were ascribed a causative role in DENV infection. This provides new insights for the development of candidate broad-spectrum therapies against hemorrhagic virus infections.


Subject(s)
Dengue Virus , Extracellular Traps , Extracellular Vesicles , Infections , Blood Platelets , Humans , Lectins, C-Type , Receptors, Cell Surface , Toll-Like Receptor 2
2.
Vet Pathol ; 59(4): 661-672, 2022 07.
Article in English | MEDLINE | ID: mdl-35001763

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulted in an ongoing pandemic with millions of deaths worldwide. Infection of humans can be asymptomatic or result in fever, fatigue, dry cough, dyspnea, and acute respiratory distress syndrome with multiorgan failure in severe cases. The pathogenesis of COVID-19 is not fully understood, and various models employing different species are currently applied. Ferrets can be infected with SARS-CoV-2 and efficiently transmit the virus to contact animals. In contrast to hamsters, ferrets usually show mild disease and viral replication restricted to the upper airways. Most reports have used the intranasal inoculation route, while the intratracheal infection model is not well characterized. Herein, we present clinical, virological, and pathological data from young ferrets intratracheally inoculated with SARS-CoV-2. Infected animals showed no significant clinical signs, and had transient infection with peak viral RNA loads at 4 days postinfection, mild to moderate rhinitis, and pulmonary endothelialitis/vasculitis. Viral antigen was exclusively found in the respiratory epithelium of the nasal cavity, indicating a particular tropism for cells in this location. Viral antigen was associated with epithelial damage and influx of inflammatory cells, including activated neutrophils releasing neutrophil extracellular traps. Scanning electron microscopy of the nasal respiratory mucosa revealed loss of cilia, shedding, and rupture of epithelial cells. The currently established ferret SARS-CoV-2 infection models are comparatively discussed with SARS-CoV-2 pathogenesis in mink, and the advantages and disadvantages of both species as research models for zoonotic betacoronaviruses are highlighted.


Subject(s)
COVID-19 , Rodent Diseases , Animals , Antigens, Viral , COVID-19/veterinary , Cricetinae , Disease Models, Animal , Ferrets , Respiratory Mucosa , SARS-CoV-2
3.
Int J Mol Sci ; 23(9)2022 May 04.
Article in English | MEDLINE | ID: mdl-35563514

ABSTRACT

Similar to many other respiratory viruses, SARS-CoV-2 targets the ciliated cells of the respiratory epithelium and compromises mucociliary clearance, thereby facilitating spread to the lungs and paving the way for secondary infections. A detailed understanding of mechanism involved in ciliary loss and subsequent regeneration is crucial to assess the possible long-term consequences of COVID-19. The aim of this study was to characterize the sequence of histological and ultrastructural changes observed in the ciliated epithelium during and after SARS-CoV-2 infection in the golden Syrian hamster model. We show that acute infection induces a severe, transient loss of cilia, which is, at least in part, caused by cilia internalization. Internalized cilia colocalize with membrane invaginations, facilitating virus entry into the cell. Infection also results in a progressive decline in cells expressing the regulator of ciliogenesis FOXJ1, which persists beyond virus clearance and the termination of inflammatory changes. Ciliary loss triggers the mobilization of p73+ and CK14+ basal cells, which ceases after regeneration of the cilia. Although ciliation is restored after two weeks despite the lack of FOXJ1, an increased frequency of cilia with ultrastructural alterations indicative of secondary ciliary dyskinesia is observed. In summary, the work provides new insights into SARS-CoV-2 pathogenesis and expands our understanding of virally induced damage to defense mechanisms in the conducting airways.


Subject(s)
COVID-19 , Animals , Cilia/metabolism , Cricetinae , Epithelium , Homeostasis , Mesocricetus , Respiratory Mucosa/metabolism , SARS-CoV-2
4.
J Virol ; 93(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30429338

ABSTRACT

The impact of the Zika virus (ZIKV) epidemic highlights the need for vaccines that reduce or prevent infection and reliably prevent teratogenic complications. The live-attenuated measles virus (MV) vaccine strains are a promising vaccine platform, since they induce robust humoral and cellular immune responses against additional antigens and have an excellent safety record. To explore its potential to protect against ZIKV, we compared a recombinant Schwarz strain MV that encodes ZIKV prM and soluble E proteins (MV-Zika-sE) with a prototypic alum-adjuvanted whole inactivated ZIKV particle vaccine. Analysis of MV-Zika-sE-infected cells confirmed antigen expression, and the virus replicated with vaccine strain characteristics. Immunized IFNAR-/--CD46Ge mice developed E protein-specific and neutralizing antibodies, and ZIKV E-specific cellular immune responses were observed by gamma interferon (IFN-γ) enzyme-linked immunospot (ELISpot) and in vitro T cell proliferation assays. To analyze protective efficacy, vaccinated female mice were challenged with ZIKV after allogeneic mating. In MV-Zika-sE-vaccinated mice, weight gain was similar to that in uninfected mice, while no plasma viremia was detectable in the majority of the animals. In contrast, infected control animals gained less weight and experienced about 100-fold higher viremia over at least 3 days. Moreover, vaccination with MV-Zika-sE reduced the ZIKV load in different organs and the placentas and prevented infection of the fetus. Consequently, no fetal growth retardation, anemia, or death due to ZIKV infection was seen in MV-Zika-sE-vaccinated dams. In contrast, the inactivated ZIKV vaccine had little to no effect in our studies. Therefore, the MV-derived ZIKV vaccine is a promising candidate for further preclinical and clinical development.IMPORTANCE Zika virus (ZIKV) is a mosquito-borne flavivirus that causes a variety of neurological complications, including congenital birth defects. Despite the urgent need, no ZIKV vaccine has yet been licensed. Recombinant vaccine strain-derived measles viruses (MV) constitute a promising vector platform to induce immunity against foreign pathogens by expressing antigens from additional transcription units while at the same time possessing a remarkable safety profile. This concept has already been validated against different pathogens, including at least 3 other flaviviruses, and our data show that vaccination with MV expressing soluble ZIKV E protein significantly diminishes infection and prevents fetal loss or damage in an allogeneic mouse pregnancy model. It can thus be regarded as a promising emergency vaccine candidate with the potential for inclusion in routine vaccination settings in areas of endemicity to prevent teratogenic effects of circulating ZIKV during pregnancy, comparable to standard rubella virus vaccination.


Subject(s)
Disease Models, Animal , Measles Vaccine/administration & dosage , Measles virus/immunology , Viral Envelope Proteins/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Antibodies, Viral/blood , Female , Genome, Viral , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Measles Vaccine/immunology , Membrane Cofactor Protein/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pregnancy , Receptor, Interferon alpha-beta/physiology , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Zika Virus/genetics , Zika Virus Infection/immunology , Zika Virus Infection/virology
5.
Am J Pathol ; 187(4): 831-840, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28189564

ABSTRACT

The acute respiratory distress syndrome (ARDS) is the leading cause of death in influenza A virus (IAV)-infected patients. Hereby, the cellular importin-α7 gene plays a major role. It promotes viral replication in the lung, thereby increasing the risk for the development of pneumonia complicated by ARDS. Herein, we analyzed whether the recently emerged H7N9 avian IAV has already adapted to human importin-α7 use, which is associated with high-level virus replication in the mammalian lung. Using a cell-based viral polymerase activity assay, we could detect a decreased H7N9 IAV polymerase activity when importin-α7 was silenced by siRNA. Moreover, virus replication was diminished in the murine cells lacking the importin-α7 gene. Consistently, importin-α7 knockout mice presented reduced pulmonary virus titers and lung lesions as well as enhanced survival rates compared to wild-type mice. In summary, our results show that H7N9 IAV have acquired distinct features of adaptation to human host factors that enable enhanced virulence in mammals. In particular, adaptation to human importin-α7 mediates elevated virus replication in the mammalian lung, which might have contributed to ARDS observed in H7N9-infected patients.


Subject(s)
Influenza A Virus, H7N9 Subtype/physiology , Mammals/virology , Respiratory System/metabolism , Respiratory System/virology , Virus Replication , alpha Karyopherins/metabolism , Animals , Chemokines/metabolism , Cytokines/metabolism , DNA-Directed DNA Polymerase/metabolism , Gene Deletion , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Influenza A Virus, H7N9 Subtype/pathogenicity , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Virulence , alpha Karyopherins/genetics
6.
PLoS Pathog ; 11(5): e1004924, 2015 May.
Article in English | MEDLINE | ID: mdl-26024522

ABSTRACT

Influenza A virus (IAV) defective RNAs are generated as byproducts of error-prone viral RNA replication. They are commonly derived from the larger segments of the viral genome and harbor deletions of various sizes resulting in the generation of replication incompatible viral particles. Furthermore, small subgenomic RNAs are known to be strong inducers of pattern recognition receptor RIG-I-dependent type I interferon (IFN) responses. The present study identifies a novel IAV-induced defective RNA derived from the PB2 segment of A/Thailand/1(KAN-1)/2004 (H5N1). It encodes a 10 kDa protein (PB2∆) sharing the N-terminal amino acid sequence of the parental PB2 protein followed by frame shift after internal deletion. PB2∆ induces the expression of IFNß and IFN-stimulated genes by direct interaction with the cellular adapter protein MAVS, thereby reducing viral replication of IFN-sensitive viruses such as IAV or vesicular stomatitis virus. This induction of IFN is completely independent of the defective RNA itself that usually serves as pathogen-associated pattern and thus does not require the cytoplasmic sensor RIG-I. These data suggest that not only defective RNAs, but also some defective RNA-encoded proteins can act immunostimulatory. In this particular case, the KAN-1-induced defective RNA-encoded protein PB2∆ enhances the overwhelming immune response characteristic for highly pathogenic H5N1 viruses, leading to a more severe phenotype in vivo.


Subject(s)
Influenza A virus/physiology , Interferon Type I/metabolism , Orthomyxoviridae Infections/metabolism , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Hemagglutination Tests , Immunoprecipitation , Interferon Type I/genetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , RNA, Messenger/genetics , RNA-Dependent RNA Polymerase/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured , Viral Proteins/genetics , Virus Replication
7.
J Gen Virol ; 97(1): 39-48, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26560088

ABSTRACT

Avian influenza viruses of subtype H9N2 that are found worldwide are occasionally transmitted to humans and pigs. Furthermore, by co-circulating with other influenza subtypes, they can generate new viruses with the potential to also cause zoonotic infections, as observed in 1997 with H5N1 or more recently with H7N9 and H10N8 viruses. Comparative analysis of the adaptive mutations in polymerases of different viruses indicates that their impact on the phylogenetically related H9N2 and H7N9 polymerases is higher than on the non-related H7N7 and H1N1pdm09 polymerases. Analysis of polymerase reassortants composed of subunits of different viruses demonstrated that the efficient enhancement of polymerase activity by H9N2-PB2 does not depend on PA and PB1. These observations suggest that the PB2 subunit of the H9N2 polymerase has a high adaptive potential and may therefore be an important pandemic risk factor.


Subject(s)
Influenza A Virus, H9N2 Subtype/enzymology , Influenza in Birds/virology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Adaptation, Biological , Animals , Birds , Female , Humans , Influenza A Virus, H9N2 Subtype/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , RNA-Dependent RNA Polymerase/genetics , Reassortant Viruses/enzymology , Reassortant Viruses/genetics , Swine , Viral Proteins/genetics , Virulence Factors/genetics
8.
J Virol ; 89(17): 9010-20, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26085167

ABSTRACT

Viral drug resistance is believed to be less likely to occur if compounds are directed against cellular rather than viral proteins. In this study, we analyzed the feasibility of a crucial viral replication factor, namely, importin-α7, as a cellular drug target to combat pandemic influenza viruses. Surprisingly, only five viral lung-to-lung passages were required to achieve 100% lethality in importin-α7⁻/⁻ mice that otherwise are resistant. Viral escape from importin-α7 requirement was mediated by five mutations in the viral ribonucleoprotein complex and the surface glycoproteins. Moreover, the importin-α7⁻/⁻ mouse-adapted strain became even more virulent for wild-type mice than the parental strain. These studies show that targeting host proteins may still result in viral escape by alternative pathways, eventually giving rise to even more virulent virus strains. Thus, therapeutic intervention strategies should consider a multitarget approach to reduce viral drug resistance. IMPORTANCE Here, we investigated the long-standing hypothesis based on in vitro studies that viral drug resistance occurrence is less likely if compounds are directed against cellular rather than viral proteins. Here, we challenged this hypothesis by analyzing, in an in vivo animal model, the feasibility of targeting the cellular factor importin-α7, which is crucial for human influenza virus replication and pathogenesis, as an efficient antiviral strategy against pandemic influenza viruses. In summary, our studies suggest that resistance against cellular factors is possible in vivo, and the emergence of even more virulent viral escape variants calls for particular caution. Thus, therapeutic intervention strategies should consider a multitarget approach using compounds against viral as well as cellular factors to reduce the risk of viral drug resistance and potentially increased virulence.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/drug therapy , Virulence Factors/genetics , alpha Karyopherins/genetics , Animals , Antiviral Agents/pharmacology , Cell Line , Dogs , Drug Resistance, Viral/genetics , HEK293 Cells , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/virology , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering , Virus Replication/genetics
9.
J Infect Dis ; 212 Suppl 2: S316-21, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26185094

ABSTRACT

Ebola virus (EBOV) protein 24 antagonizes the host interferon (IFN) response by hijacking select nuclear importin-α isoforms. Thereby, it blocks STAT1-mediated IFN-α/ß and IFN-γ synthesis. However, owing to the lack of importin-α knockout animal models in the past, their role in EBOV pathogenesis remained largely unknown. Here, we demonstrate that importin-α7 is involved in the formation of EBOV inclusion bodies and replication. However, deletion of the gene encoding importin-α7 was not sufficient to increase survival rates among mice infected with EBOV.


Subject(s)
Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/virology , Inclusion Bodies, Viral/physiology , Virulence/physiology , alpha Karyopherins/metabolism , Animals , Cell Line , Chlorocebus aethiops , DNA Replication/genetics , Ebolavirus/genetics , Ebolavirus/metabolism , Mice , Mice, Inbred C57BL , Vero Cells , Viral Proteins/metabolism , Virulence/genetics , Virus Replication/genetics
10.
J Virol ; 88(16): 8735-42, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899203

ABSTRACT

UNLABELLED: Mutation D701N in the PB2 protein is known to play a prominent role in the adaptation of avian influenza A viruses to mammalian hosts. In contrast, little is known about the nearby mutations S714I and S714R, which have been observed in some avian influenza viruses highly pathogenic for mammals. We have generated recombinant H5N1 viruses with PB2 displaying the avian signature 701D or the mammalian signature 701N and serine, isoleucine, and arginine at position 714 and compared them for polymerase activity and virus growth in avian and mammalian cells, as well as for pathogenicity in mice. Mutation D701N led to an increase in polymerase activity and replication efficiency in mammalian cells and in mouse pathogenicity, and this increase was significantly enhanced when mutation D701N was combined with mutation S714R. Stimulation by mutation S714I was less distinct. These observations indicate that PB2 mutation S714R, in combination with the mammalian signature at position 701, has the potential to promote the adaptation of an H5N1 virus to a mammalian host. IMPORTANCE: Influenza A/H5N1 viruses are avian pathogens that have pandemic potential, since they are spread over large parts of Asia, Africa, and Europe and are occasionally transmitted to humans. It is therefore of high scientific interest to understand the mechanisms that determine the host specificity and pathogenicity of these viruses. It is well known that the PB2 subunit of the viral polymerase is an important host range determinant and that PB2 mutation D701N plays an important role in virus adaptation to mammalian cells. In the present study, we show that mutation S714R is also involved in adaptation and that it cooperates with D701N in exposing a nuclear localization signal that mediates importin-α binding and entry of PB2 into the nucleus, where virus replication and transcription take place.


Subject(s)
Adaptation, Physiological/genetics , Influenza A Virus, H5N1 Subtype/genetics , Mammals/virology , Mutation/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Animals , Dogs , HEK293 Cells , Humans , Influenza, Human/genetics , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mammals/genetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology
11.
J Virol ; 88(14): 8166-79, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24829333

ABSTRACT

Influenza A viruses recruit components of the nuclear import pathway to enter the host cell nucleus and promote viral replication. Here, we analyzed the role of the nuclear import factor importin-α7 in H1N1 influenza virus pulmonary tropism by using various ex vivo imaging techniques (magnetic resonance imaging, confocal laser scanning microscopy, and correlative light-electron microscopy). We infected importin-α7 gene-deficient (α7(-/-)) mice with a recombinant H1N1 influenza virus and compared the in vivo viral kinetics with those in wild-type (WT) mice. In WT mice, influenza virus replication in the bronchial and alveolar epithelium already occurred a few days after infection. Accordingly, extensive mononuclear infiltration and alveolar destruction were present in the lungs of infected WT mice, followed by 100% lethality. Conversely, in α7(-/-) mice, virus replication was restricted mostly to the bronchial epithelium with marginal alveolar infection, resulting in significantly reduced lung damage and enhanced animal survival. To investigate the host immune response during alveolar virus replication, we studied the role of primary macrophages in virus propagation and clearance. The ability of macrophages to support or clear the virus infection, as well as the host cellular immune responses, did not significantly differ between WT and α7(-/-) mice. However, cytokine and chemokine responses were generally elevated in WT mice, likely reflective of increased viral replication in the lung. In summary, these data show that a cellular factor, importin-α7, is required for enhanced virus replication in the alveolar epithelium, resulting in elevated cytokine and chemokine levels, extensive mononuclear infiltration, and thus, severe pneumonia and enhanced virulence in mice. Importance: Influenza A viruses are respiratory pathogens that may cause pneumonia in humans. Viral infection and replication in the alveoli of the respiratory tract are believed to be crucial for the development of the acute respiratory distress syndrome associated with fatal outcomes in influenza virus-infected patients. Here, we report the requirement of a cellular factor, importin-α7, for efficient virus replication in the alveolar epithelium of mice. Using complementary ex vivo imaging approaches, we show that influenza virus replication is restricted to the bronchial epithelium, followed by enhanced survival in importin-α7-deficient mice. In contrast, the presence of this gene results in enhanced virus replication in the alveoli, elevated cytokine and chemokine responses, mononuclear infiltration, alveolar destruction, and 100% lethality in wild-type mice. Taken together, our results show that importin-α7 is particularly required for virus replication in the alveolar epithelium in association with severe pneumonia and death in mice.


Subject(s)
Epithelial Cells/virology , Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/physiology , Lung/pathology , Viral Tropism , Virus Replication , alpha Karyopherins/metabolism , Animals , Cytokines/metabolism , Lung/virology , Magnetic Resonance Imaging , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Respiratory Mucosa/virology , Survival Analysis , alpha Karyopherins/deficiency
12.
J Virol ; 88(9): 4744-51, 2014 May.
Article in English | MEDLINE | ID: mdl-24522916

ABSTRACT

UNLABELLED: Cleavage of the hemagglutinin (HA) by host proteases is essential for the infectivity of influenza viruses. Here, we analyzed the role of the serine protease TMPRSS2, which activates HA in the human respiratory tract, in pathogenesis in a mouse model. Replication of the human H7N9 isolate A/Anhui/1/13 and of human H1N1 and H3N2 viruses was compared in TMPRSS2 knockout (TMPRSS2(-/-)) and wild-type (WT) mice. Knockout of TMPRSS2 expression inhibited H7N9 influenza virus replication in explants of murine tracheas, bronchi, and lungs. H1N1 virus replication was also strongly suppressed in airway explants of TMPRSS2(-/-) mice, while H3N2 virus replication was only marginally affected. H7N9 and H1N1 viruses were apathogenic in TMPRSS2(-/-) mice, whereas WT mice developed severe disease with mortality rates of 100% and 20%, respectively. In contrast, all H3N2 infected TMPRSS2(-/-) and WT mice succumbed to lethal infection. Cleavage analysis showed that H7 and H1 are efficiently activated by TMPRSS2, whereas H3 is less susceptible to the protease. Our data demonstrate that TMPRSS2 is a host factor that is essential for pneumotropism and pathogenicity of H7N9 and H1N1 influenza virus in mice. In contrast, replication of H3N2 virus appears to depend on another, not yet identified protease, supporting the concept that human influenza viruses differ in protease specificity. IMPORTANCE: Cleavage of the hemagglutinin (HA) by host proteases is essential for the infectivity of influenza virus, but little is known about its relevance for pathogenesis in mammals. Here, we show that knockout mice that do not express the HA-activating protease TMPRSS2 are resistant to pulmonary disease with lethal outcome when infected with influenza A viruses of subtypes H7N9 and H1N1, whereas they are not protected from lethal H3N2 virus infection. These findings demonstrate that human influenza viruses differ in protease specificity, and that expression of the appropriate protease in respiratory tissues is essential for pneumotropism and pathogenicity. Our observations also demonstrate that HA-activating proteases and in particular TMPRSS2 are promising targets for influenza therapy.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H7N9 Subtype/physiology , Influenza A Virus, H7N9 Subtype/pathogenicity , Lung/virology , Serine Endopeptidases/metabolism , Viral Tropism , Animal Structures/virology , Animals , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Mice , Mice, Knockout , Organ Culture Techniques , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Serine Endopeptidases/genetics , Survival Analysis , Trachea/virology , Virulence
13.
J Virol ; 88(16): 9379-90, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24920808

ABSTRACT

UNLABELLED: Viruses take advantage of host posttranslational modifications for their own benefit. It was recently reported that influenza A virus proteins interact extensively with the host sumoylation system. Thereby, several viral proteins, including NS1 and M1, are sumoylated to facilitate viral replication. However, to what extent sumoylation is exploited by influenza A virus is not fully understood. In this study, we found that influenza A virus nucleoprotein (NP) is a bona fide target of sumoylation in both NP-transfected cells and virus-infected cells. We further found that NP is sumoylated at the two most N-terminal residues, lysines 4 and 7, and that sumoylation at lysine 7 of NP is highly conserved across different influenza A virus subtypes and strains, including the recently emerged human H7N9 virus. While NP stability and polymerase activity are little affected by sumoylation, the NP sumoylation-defective WSN-NPK4,7R virus exhibited early cytoplasmic localization of NP. The growth of the WSN-NPK4,7R virus was highly attenuated compared to that of the wild-type WSN virus, and the lysine residue at position 7 is indispensable for the virus's survival, as illustrated by the rapid emergence of revertant viruses. Thus, sumoylation of influenza A virus NP is essential for intracellular trafficking of NP and for virus growth, illustrating sumoylation as a crucial strategy extensively exploited by influenza A virus for survival in its host. IMPORTANCE: Host posttranslational modifications are heavily targeted by viruses for their own benefit. We and others previously reported that influenza A virus interacts extensively with the host sumoylation system. However, the functional outcomes of viral sumoylation are not fully understood. Here we found that influenza A virus nucleoprotein (NP), an essential component for virus replication, is a new target of SUMO. This is the first study to find that NP from different influenza A viruses, including recently emerged H7N9, is sumoylated at conserved lysine 7. Our data further illustrated that sumoylation of influenza A virus NP is essential for intracellular trafficking of NP and virus growth, indicating that influenza A virus relies deeply on sumoylation to survive in host cells. Strategies to downregulate viral sumoylation could thus be a potential antiviral treatment.


Subject(s)
Influenza A Virus, H7N9 Subtype/metabolism , Influenza A virus/metabolism , Protein Transport/genetics , RNA-Binding Proteins/metabolism , Sumoylation/genetics , Viral Core Proteins/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Dogs , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza A virus/genetics , Lysine/genetics , Lysine/metabolism , Madin Darby Canine Kidney Cells , Nucleocapsid Proteins , RNA-Binding Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Viral Core Proteins/genetics , Viral Proteins/genetics , Virus Replication/genetics
14.
J Virol ; 88(8): 4595-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478425

ABSTRACT

We determined the pattern of attachment of the avian-origin H7N9 influenza viruses A/Anhui/1/2013 and A/Shanghai/1/2013 to the respiratory tract in ferrets, macaques, mice, pigs, and guinea pigs and compared it to that in humans. The H7N9 attachment pattern in macaques, mice, and to a lesser extent pigs and guinea pigs resembled that in humans more closely than the attachment pattern in ferrets. This information contributes to our knowledge of the different animal models for influenza.


Subject(s)
Disease Models, Animal , Influenza A Virus, H7N9 Subtype/physiology , Influenza, Human/virology , Respiratory System/virology , Virus Attachment , Animals , China , Female , Ferrets , Guinea Pigs , Humans , Influenza A Virus, H7N9 Subtype/genetics , Macaca , Male , Mice , Swine
15.
Curr Top Microbiol Immunol ; 385: 35-60, 2014.
Article in English | MEDLINE | ID: mdl-25033751

ABSTRACT

Viral pathogenesis involves numerous interactions between viral and cellular factors. In recent years, the influenza virus polymerase complex has emerged as a major determinant of interspecies transmission and pathogenicity. The viral RNA-dependent RNA polymerase, in concert with the nucleoprotein, mediates transcription and replication of the viral RNA genome in the nucleus of the infected cell. The activity by which the viral polymerase complex performs these processes in mammalian cells is considered to be a major contributor to viral pathogenicity in mammals. In this chapter, we summarise our current understanding on the pathogenicity determinants in the viral polymerase complex and highlight some of its cellular interaction partners. We particularly discuss the role of importin-α isoforms in host adaptation and pathogenesis as well as the role of the viral polymerase in regulating cellular responses to viral infection.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Influenza, Human/virology , Orthomyxoviridae/enzymology , Orthomyxoviridae/pathogenicity , Viral Proteins/metabolism , Animals , DNA-Directed RNA Polymerases/genetics , Humans , Orthomyxoviridae/genetics , Viral Proteins/genetics
16.
Bioessays ; 35(1): 23-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23239226

ABSTRACT

After viral entry into the cell, the nuclear envelope poses a major cellular barrier that needs to be overcome upon adaptation of highly pathogenic avian influenza viruses (HPAIV) to the new host. To ensure efficient viral transcription and replication in the nucleus of the host cell, the viral polymerase complex of avian influenza viruses needs to switch from recognition of avian to mammalian components of the nuclear import machinery. Recent evidence suggests that influenza viruses have evolved different mechanisms to utilize importin-α isoforms as components of this machinery, bridging pre- and post-nuclear import on both sides of the nuclear envelope.


Subject(s)
Cell Nucleus/metabolism , Cell Nucleus/virology , DNA-Directed RNA Polymerases/metabolism , Influenza A virus/physiology , Nuclear Envelope/metabolism , Virus Replication , Active Transport, Cell Nucleus , Animals , Birds , Humans , Influenza in Birds , Influenza, Human , Nuclear Envelope/virology , Protein Isoforms , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , alpha Karyopherins/metabolism
17.
J Infect Dis ; 209 Suppl 3: S93-9, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24966196

ABSTRACT

Sex-specific endocrine and immune responses are widely recognized to account for differential disease outcomes between females and males. Surprisingly, sex-specific risk assessments for influenza, a viral pathogen that affects human populations worldwide through seasonal epidemics and irregular occurring pandemics, are sparse and-if available-ambiguous. To date, this precludes proposing an unequivocal sex-dependent susceptibility to influenza. However, one undisputable observation recurrently confirmed during influenza seasons of the last decades is the significantly increased risk for pregnant women. This increased risk is likely attributable to the contradictory demands for the maternal immune system to adapt to pregnancy and to simultaneously mount an immune response to clear the influenza virus infection. Here, we review published evidence on the potential association between sex on influenza risk and propose that future epidemiologic studies should carefully dissect surveillance data for sex-specific effects. Moreover, we propose potential mechanisms involved in enhanced risk for severe influenza during pregnancy that could be studied to identify causal pathways.


Subject(s)
Influenza, Human/immunology , Disease Susceptibility/immunology , Female , Humans , Influenza, Human/virology , Male , Pregnancy , Pregnancy Complications, Infectious/immunology , Sex Factors
18.
PLoS Pathog ; 8(1): e1002488, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22275867

ABSTRACT

Influenza A viruses may cross species barriers and transmit to humans with the potential to cause pandemics. Interplay of human- (PB2 627K) and avian-like (PB2 627E) influenza polymerase complexes with unknown host factors have been postulated to play a key role in interspecies transmission. Here, we have identified human importin-α isoforms (α1 and α7) as positive regulators of human- but not avian-like polymerase activity. Human-like polymerase activity correlated with efficient recruitment of α1 and α7 to viral ribonucleoprotein complexes (vRNPs) without affecting subcellular localization. We also observed that human-like influenza virus growth was impaired in α1 and α7 downregulated human lung cells. Mice lacking α7 were less susceptible to human- but not avian-like influenza virus infection. Thus, α1 and α7 are positive regulators of human-like polymerase activity and pathogenicity beyond their role in nuclear transport.


Subject(s)
Influenza A virus/enzymology , Influenza, Human/transmission , Orthomyxoviridae Infections/transmission , Viral Proteins/genetics , alpha Karyopherins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Transformed , Down-Regulation , Host-Pathogen Interactions , Humans , Influenza A virus/growth & development , Influenza A virus/pathogenicity , Influenza, Human/virology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Orthomyxoviridae Infections/virology , Protein Binding , Protein Isoforms , Viral Proteins/metabolism , Virus Replication , alpha Karyopherins/genetics
19.
EBioMedicine ; 105: 105191, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38865747

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19) frequently leads to neurological complications after recovery from acute infection, with higher prevalence in women. However, mechanisms by which SARS-CoV-2 disrupts brain function remain unclear and treatment strategies are lacking. We previously demonstrated neuroinflammation in the olfactory bulb of intranasally infected hamsters, followed by alpha-synuclein and tau accumulation in cortex, thus mirroring pathogenesis of neurodegenerative diseases such as Parkinson's or Alzheimer's disease. METHODS: To uncover the sex-specific spatiotemporal profiles of neuroinflammation and neuronal dysfunction following intranasal SARS-CoV-2 infection, we quantified microglia cell density, alpha-synuclein immunoreactivity and inhibitory interneurons in cortical regions, limbic system and basal ganglia at acute and late post-recovery time points. FINDINGS: Unexpectedly, microglia cell density and alpha-synuclein immunoreactivity decreased at 6 days post-infection, then rebounded to overt accumulation at 21 days post-infection. This biphasic response was most pronounced in amygdala and striatum, regions affected early in Parkinson's disease. Several brain regions showed altered densities of parvalbumin and calretinin interneurons which are involved in cognition and motor control. Of note, females appeared more affected. INTERPRETATION: Our results demonstrate that SARS-CoV-2 profoundly disrupts brain homeostasis without neuroinvasion, via neuroinflammatory and protein regulation mechanisms that persist beyond viral clearance. The regional patterns and sex differences are in line with neurological deficits observed after SARS-CoV-2 infection. FUNDING: Federal Ministry of Health, Germany (BMG; ZMV I 1-2520COR501 to G.G.), Federal Ministry of Education and Research, Germany (BMBF; 03COV06B to G.G.), Ministry of Science and Culture of Lower Saxony in Germany (14-76403-184, to G.G. and F.R.).

20.
Cell Microbiol ; 14(5): 774-89, 2012 May.
Article in English | MEDLINE | ID: mdl-22289149

ABSTRACT

The mechanism of budding of influenza A virus revealed important deviation from the consensus mechanism of budding of retroviruses and of a growing number of negative-strand RNA viruses. This study is focused on the role of the influenza A virus matrix protein M1 in virus release. We found that a mutation of the proline residue at position 16 of the matrix protein induces inhibition of virus detachment from cells. Depletion of the M1-binding protein RACK1 also impairs virus release and RACK1 binding requires the proline residue at position 16 of M1. The impaired M1-RACK1 interaction does not affect the plasma membrane binding of M1; in contrast, RACK1 is recruited to detergent-resistant membranes in a M1-proline-16-dependent manner. The proline-16 mutation in M1 and depletion of RACK1 impairs the pinching-off of the budding virus particles. These findings reveal the active role of the viral matrix protein in the release of influenza A virus particles that involves a cross-talk with a RACK1-mediated pathway.


Subject(s)
GTP-Binding Proteins/metabolism , Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/physiology , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Viral Matrix Proteins/metabolism , Virus Release , Cell Line , Humans , Influenza A Virus, H1N1 Subtype/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Proline/genetics , Proline/metabolism , Protein Interaction Mapping , Receptors for Activated C Kinase , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL