Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
PLoS Pathog ; 17(9): e1009566, 2021 09.
Article in English | MEDLINE | ID: mdl-34555124

ABSTRACT

The hemagglutinin (HA) of A/H3N2 pandemic influenza viruses (IAVs) of 1968 differed from its inferred avian precursor by eight amino acid substitutions. To determine their phenotypic effects, we studied recombinant variants of A/Hong Kong/1/1968 virus containing either human-type or avian-type amino acids in the corresponding positions of HA. The precursor HA displayed receptor binding profile and high conformational stability typical for duck IAVs. Substitutions Q226L and G228S, in addition to their known effects on receptor specificity and replication, marginally decreased HA stability. Substitutions R62I, D63N, D81N and N193S reduced HA binding avidity. Substitutions R62I, D81N and A144G promoted viral replication in human airway epithelial cultures. Analysis of HA sequences revealed that substitutions D63N and D81N accompanied by the addition of N-glycans represent common markers of avian H3 HA adaptation to mammals. Our results advance understanding of genotypic and phenotypic changes in IAV HA required for avian-to-human adaptation and pandemic emergence.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza in Birds/genetics , Influenza, Human/genetics , Viral Zoonoses/genetics , Animals , Ducks , Humans , Pandemics
2.
Life Sci Alliance ; 3(9)2020 09.
Article in English | MEDLINE | ID: mdl-32703818

ABSTRACT

The novel emerged SARS-CoV-2 has rapidly spread around the world causing acute infection of the respiratory tract (COVID-19) that can result in severe disease and lethality. For SARS-CoV-2 to enter cells, its surface glycoprotein spike (S) must be cleaved at two different sites by host cell proteases, which therefore represent potential drug targets. In the present study, we show that S can be cleaved by the proprotein convertase furin at the S1/S2 site and the transmembrane serine protease 2 (TMPRSS2) at the S2' site. We demonstrate that TMPRSS2 is essential for activation of SARS-CoV-2 S in Calu-3 human airway epithelial cells through antisense-mediated knockdown of TMPRSS2 expression. Furthermore, SARS-CoV-2 replication was also strongly inhibited by the synthetic furin inhibitor MI-1851 in human airway cells. In contrast, inhibition of endosomal cathepsins by E64d did not affect virus replication. Combining various TMPRSS2 inhibitors with furin inhibitor MI-1851 produced more potent antiviral activity against SARS-CoV-2 than an equimolar amount of any single serine protease inhibitor. Therefore, this approach has considerable therapeutic potential for treatment of COVID-19.


Subject(s)
Alveolar Epithelial Cells/virology , Betacoronavirus/physiology , Furin/genetics , Serine Endopeptidases/genetics , Spike Glycoprotein, Coronavirus/metabolism , Alveolar Epithelial Cells/cytology , Animals , Binding Sites , Cell Line , Chlorocebus aethiops , HEK293 Cells , Humans , Proteolysis , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Vero Cells , Virus Internalization , Virus Replication
3.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31597765

ABSTRACT

Previous studies revealed that certain avian influenza A viruses (IAVs), including zoonotic H5N1 and H7N9 IAVs, infect cultured human lung microvascular endothelial cells (HULEC) more efficiently than other IAVs and that tropism to HULEC is determined by viral hemagglutinin (HA). To characterize mechanisms of HA-mediated endotheliotropism, we used 2:6 recombinant IAVs harboring HAs from distinctive avian and human viruses and found that efficient infection of HULEC correlated with low conformational stability of the HA. We next studied effects on viral infectivity of single-point amino acid substitutions in the HA of 2:6 recombinant virus A/Vietnam/1203/2004-PR8 (H5N1). Substitutions H8Q, H103Y, T315I, and K582I (K58I in the HA2 subunit), which increased stability of the HA, markedly reduced viral infectivity for HULEC, whereas substitutions K189N and K218Q, which altered typical H5N1 virus-like receptor specificity and reduced binding avidity of the HA, led to only marginal reduction of infectivity. None of these substitutions affected virus infection in MDCK cells. We confirmed the previous observation of elevated basal expression of IFITM3 protein in HULEC and found that endosomal acidification is less efficient in HULEC than in MDCK cells. In accord with these findings, counteraction of IFITM3-mediated restriction by amphotericin B and reduction of endosomal pH by moderate acidification of the extracellular medium enhanced infectivity of viruses with stable HA for HULEC without significant effect on infectivity for MDCK cells. Collectively, our results indicate that relatively high pH optimum of fusion of the HA of zoonotic H5N1 and H7N9 IAVs allows them to overcome antiviral effects of inefficient endosomal acidification and IFITM3 in human endothelial cells.IMPORTANCE Receptor specificity of the HA of IAVs is known to be a critical determinant of viral cell tropism. Here, we show that fusion properties of the HA may also play a key role in the tropism. Thus, we demonstrate that IAVs having a relatively low pH optimum of fusion cannot efficiently infect human endothelial cells owing to their relatively high endosomal pH and increased expression of fusion-inhibiting IFITM3 protein. These restrictions can be overcome by IAVs with elevated pH of fusion, such as zoonotic H5N1 and H7N9. Our results illustrate that the infectivity of IAVs depends on an interplay between HA conformational stability, endosomal acidification and IFITM3 expression in target cells, and the extracellular pH. Given significant variation of levels of HA stability among animal, human, and zoonotic IAVs, our findings prompt further studies on the fusion-dependent tropism of IAVs to different cell types in humans and its role in viral host range and pathogenicity.


Subject(s)
Endosomes/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H7N9 Subtype/genetics , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , Reassortant Viruses/genetics , Amino Acid Substitution , Animals , Dogs , Endosomes/virology , Endothelial Cells/metabolism , Endothelial Cells/virology , Gene Expression Regulation , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Host-Pathogen Interactions/genetics , Humans , Hydrogen-Ion Concentration , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/pathogenicity , Lung/metabolism , Lung/virology , Madin Darby Canine Kidney Cells , Membrane Proteins/metabolism , Models, Molecular , Mutation , Protein Binding , Protein Conformation , RNA-Binding Proteins/metabolism , Reassortant Viruses/metabolism , Reassortant Viruses/pathogenicity , Structure-Activity Relationship , Viral Tropism/genetics , Virus Replication
4.
Emerg Microbes Infect ; 8(1): 1324-1336, 2019.
Article in English | MEDLINE | ID: mdl-31503518

ABSTRACT

Avian influenza A viruses (AIV) of the H7 subtype continue to evolve posing a pandemic threat. However, molecular markers of H7N7 AIV pathogenicity and transmission in mammals remain poorly understood. In this study, we performed a systematic in vitro and in vivo analysis by comparing an H7N7 highly pathogenic AIV and its ferret adapted variant. Passaging an H7N7 AIV in ferrets led to six mutations in genes encoding the viral polymerase complex and the viral surface proteins. Here, we show that mutations in the H7 hemagglutinin gene cause increased pathogenicity in mice. Contact transmission between guinea pigs required additional mutations in the gene encoding the polymerase subunit PB1. Thus, particular vigilance is required with respect to HA and PB1 mutations as predictive molecular markers to assess the pandemic risk posed by emerging H7 avian influenza viruses.


Subject(s)
Disease Transmission, Infectious , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H7N7 Subtype/pathogenicity , Mutant Proteins/genetics , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Viral Proteins/genetics , Animals , Disease Models, Animal , Ferrets , Guinea Pigs , Influenza A Virus, H7N7 Subtype/genetics , Orthomyxoviridae Infections/pathology , Serial Passage , Virulence Factors/genetics
5.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31391268

ABSTRACT

Cleavage of influenza virus hemagglutinin (HA) by host cell proteases is essential for virus infectivity and spread. We previously demonstrated in vitro that the transmembrane protease TMPRSS2 cleaves influenza A virus (IAV) and influenza B virus (IBV) HA possessing a monobasic cleavage site. Subsequent studies revealed that TMPRSS2 is crucial for the activation and pathogenesis of H1N1pdm and H7N9 IAV in mice. In contrast, activation of H3N2 IAV and IBV was found to be independent of TMPRSS2 expression and supported by an as-yet-undetermined protease(s). Here, we investigated the role of TMPRSS2 in proteolytic activation of IAV and IBV in three human airway cell culture systems: primary human bronchial epithelial cells (HBEC), primary type II alveolar epithelial cells (AECII), and Calu-3 cells. Knockdown of TMPRSS2 expression was performed using a previously described antisense peptide-conjugated phosphorodiamidate morpholino oligomer, T-ex5, that interferes with splicing of TMPRSS2 pre-mRNA, resulting in the expression of enzymatically inactive TMPRSS2. T-ex5 treatment produced efficient knockdown of active TMPRSS2 in all three airway cell culture models and prevented proteolytic activation and multiplication of H7N9 IAV in Calu-3 cells and H1N1pdm, H7N9, and H3N2 IAV in HBEC and AECII. T-ex5 treatment also inhibited the activation and spread of IBV in AECII but did not affect IBV activation in HBEC and Calu-3 cells. This study identifies TMPRSS2 as the major HA-activating protease of IAV in human airway cells and IBV in type II pneumocytes and as a potential target for the development of novel drugs to treat influenza infections.IMPORTANCE Influenza A viruses (IAV) and influenza B viruses (IBV) cause significant morbidity and mortality during seasonal outbreaks. Cleavage of the viral surface glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. Inhibition of relevant proteases provides a promising therapeutic approach that may avoid the development of drug resistance. HA of most influenza viruses is cleaved at a monobasic cleavage site, and a number of proteases have been shown to cleave HA in vitro This study demonstrates that the transmembrane protease TMPRSS2 is the major HA-activating protease of IAV in primary human bronchial cells and of both IAV and IBV in primary human type II pneumocytes. It further reveals that human and murine airway cells can differ in their HA-cleaving protease repertoires. Our data will help drive the development of potent and selective protease inhibitors as novel drugs for influenza treatment.


Subject(s)
Influenza A virus/physiology , Influenza B virus/physiology , Influenza, Human/virology , Serine Endopeptidases/metabolism , Animals , Bronchi/cytology , Cells, Cultured , Epithelial Cells/virology , Gene Knockdown Techniques , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Host-Pathogen Interactions , Humans , Influenza, Human/enzymology , Influenza, Human/metabolism , Mice , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Pulmonary Alveoli/cytology , Serine Endopeptidases/genetics , Up-Regulation , Virus Replication
7.
Cell Rep ; 26(2): 438-446.e5, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30625326

ABSTRACT

Yellow fever virus (YFV), a deadly human pathogen, is the prototype of the genus Flavivirus. Recently, YFV re-emerged in Africa and Brazil, leading to hundreds of deaths, with some cases imported to China. Prophylactic or therapeutic countermeasures are urgently needed. Previously, several human monoclonal antibodies against YFV were screened out by phage display. Here, we find that one of them, 5A, exhibits high neutralizing potency and good protection. Crystallographic analysis of the YFV envelope (E) protein in its pre- and post-fusion states shows conformations similar to those observed in other E proteins of flaviviruses. Furthermore, the structures of 5A in complex with the E protein in both states are resolved, revealing an invariant recognition site. Structural analysis and functional data suggest that 5A has high neutralization potency because it interferes with virus entry by preventing both virus attachment and fusion. These findings will be instrumental for immunogen or inhibitor design.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Molecular Docking Simulation , Viral Envelope Proteins/immunology , Yellow Fever/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/therapeutic use , Antibody Affinity , Chlorocebus aethiops , Cricetinae , Cricetulus , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Vero Cells , Viral Envelope Proteins/chemistry , Yellow Fever/prevention & control , Yellow fever virus/immunology
8.
Virology ; 522: 37-45, 2018 09.
Article in English | MEDLINE | ID: mdl-30014856

ABSTRACT

Ducks, gulls and shorebirds represent the major hosts of influenza A viruses (IAVs) in nature, but distinctions of IAVs in different birds are not well defined. Here we characterized the receptor specificity of gull IAVs with HA subtypes H4, H6, H14, H13 and H16 using synthetic sialylglycopolymers. In contrast to duck IAVs, gull IAVs efficiently bound to fucosylated receptors and often preferred sulfated and non-sulfated receptors with Galß1-4GlcNAc cores over the counterparts with Galß1-3GlcNAc cores. Unlike all other IAVs of aquatic birds, H16 IAVs showed efficient binding to Neu5Acα2-6Gal-containing receptors and bound poorly to Neu5Acα2-3Galß1-3-terminated (duck-type) receptors. Analysis of HA crystal structures and amino acid sequences suggested that the amino acid at position 222 is an important determinant of the receptor specificity of IAVs and that transmission of duck viruses to gulls and shorebirds is commonly accompanied by substitutions at this position.


Subject(s)
Charadriiformes/virology , Influenza A virus/isolation & purification , Influenza A virus/physiology , Influenza in Birds/virology , Oligosaccharides/metabolism , Receptors, Virus/metabolism , Virus Attachment , Amino Acid Sequence , Animals , Binding Sites , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Models, Molecular , Oligosaccharides/chemistry , Protein Conformation , Receptors, Virus/chemistry
9.
Viruses ; 10(4)2018 03 31.
Article in English | MEDLINE | ID: mdl-29614716

ABSTRACT

Wild ducks are known to be able to carry avian influenza viruses over long distances and infect domestic ducks, which in their turn infect domestic chickens. Therefore, prevention of virus transmission between ducks and chickens is important to control the spread of avian influenza. Here we used a low pathogenic wild aquatic bird virus A/duck/Moscow/4182/2010 (H5N3) for prevention of highly pathogenic avian influenza virus (HPAIV) transmission between ducks and chickens. We first confirmed that the ducks orally infected with H5N1 HPAIV A/chicken/Kurgan/3/2005 excreted the virus in feces. All chickens that were in contact with the infected ducks became sick, excreted the virus, and died. However, the ducks orally inoculated with 104 50% tissue culture infective doses of A/duck/Moscow/4182/2010 and challenged 14 to 90 days later with H5N1 HPAIV did not excrete the challenge virus. All contact chickens survived and did not excrete the virus. Our results suggest that low pathogenic virus of wild aquatic birds can be used for prevention of transmission of H5N1 viruses between ducks and chickens.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/immunology , Poultry Diseases/prevention & control , Poultry Diseases/transmission , Vaccines, Live, Unattenuated/immunology , Virus Shedding/immunology , Administration, Oral , Animals , Animals, Domestic , Chickens , Ducks , Feces/virology , Immunization , Influenza A Virus, H5N1 Subtype/immunology , Influenza A virus/classification , Influenza A virus/pathogenicity , Influenza Vaccines/administration & dosage , Poultry Diseases/mortality , Vaccines, Live, Unattenuated/administration & dosage
10.
EBioMedicine ; 28: 143-150, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29337134

ABSTRACT

BACKGROUND: It is a continuous matter of discussion whether immune activation by vaccination in general and Influenza vaccination in particular increases the risk for clinical deterioration of autoimmune diseases. This prospective study investigated the serological and clinical course of autoimmune Myasthenia gravis (MG) after a seasonal influenza vaccination. METHODS: This randomized, placebo-controlled, double-blind study enrolled MG patients with antibodies against acetylcholine-receptors (AChR-ab). They were allocated to receive seasonal influenza vaccine or placebo. The primary endpoint was the relative change of AChR-ab-titer over 12weeks. A relative increase of 20% was set as non-inferiority margin. Secondary endpoints were clinical changes in the modified Quantitative Myasthenia Gravis Score (QMG), increase of anti-influenza-ELISA-antibodies, and changes of treatment. The study is registered with Clinicaltrialsregister.eu, EudraCT number 2006-004374-27. FINDINGS: 62 patients were included. Mean±standard deviation (median) in the vaccine and placebo group were AChR-ab-titer changes of -6.0%±23.3% (-4.0%) and -2.8%±22.0% (-0.5%) and QMG score changes of -0.08±0.27 (0.17) and 0.11±0.31 (0.00), respectively. The difference between groups (Hodges-Lehmann estimate with 95% CI) was - for the AChR-ab-titer change 4·0% [-13.3%, 4.5%] (p=0.28 for testing a difference, p<0.0001 for testing non-inferiority) and for the QMG change 0·00 [-0.17, 0.00] (p=0.79 for testing a difference). The occurrence of 74 adverse events (AE) was comparable between groups. The most common AE was flu-like symptoms. One serious AE (hospitalisation following gastrointestinal haemorrhage) in the verum group was not related to the vaccine. INTERPRETATION: Influenza vaccination in MG is safe. Uprating the potential risk of a severe course of MG exacerbation during influenza infection compared to the 95% CI differences for the endpoints, vaccination is principally indicated in this patient population.


Subject(s)
Antibodies, Viral/immunology , Disease Progression , Influenza, Human/immunology , Myasthenia Gravis/immunology , Myasthenia Gravis/virology , Receptors, Cholinergic/immunology , Vaccination , Adult , Aged , Double-Blind Method , Female , Humans , Male , Middle Aged , Vaccination/adverse effects
11.
Methods Mol Biol ; 1628: 3-14, 2017.
Article in English | MEDLINE | ID: mdl-28573607

ABSTRACT

Since the discovery of Marburg virus 50 years ago, filoviruses have reemerged in the human population more than 40 times. Already the first episode was as dramatic as most of the subsequent ones, but none of them was as devastating as the West-African Ebola virus outbreak in 2013-2015. Although progress toward a better understanding of the viruses is impressive, there is clearly a need to improve and strengthen the measures to detect and control these deadly infections.


Subject(s)
Disease Outbreaks/history , Hemorrhagic Fever, Ebola/history , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/epidemiology , History, 20th Century , History, 21st Century , Humans , Marburgvirus/pathogenicity
12.
J Virol ; 91(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28356532

ABSTRACT

The replication and pathogenicity of influenza A viruses (IAVs) critically depend on their ability to tolerate the antiviral interferon (IFN) response. To determine a potential role for the IAV hemagglutinin (HA) in viral sensitivity to IFN, we studied the restriction of IAV infection in IFN-ß-treated human epithelial cells by using 2:6 recombinant IAVs that shared six gene segments of A/Puerto Rico/8/1934 virus (PR8) and contained HAs and neuraminidases of representative avian, human, and zoonotic H5N1 and H7N9 viruses. In A549 and Calu-3 cells, viruses displaying a higher pH optimum of HA-mediated membrane fusion, H5N1-PR8 and H7N9-PR8, were less sensitive to the IFN-induced antiviral state than their counterparts with HAs from duck and human viruses, which fused at a lower pH. The association between a high pH optimum of fusion and reduced IFN sensitivity was confirmed by using HA point mutants of A/Hong Kong/1/1968-PR8 that differed solely by their fusion properties. Furthermore, similar effects of the viral fusion pH on IFN sensitivity were observed in experiments with (i) primary human type II alveolar epithelial cells and differentiated cultures of human airway epithelial cells, (ii) nonrecombinant zoonotic and pandemic IAVs, and (iii) preparations of IFN-α and IFN-λ1. A higher pH of membrane fusion and reduced sensitivity to IFN correlated with lower restriction of the viruses in MDCK cells stably expressing the IFN-inducible transmembrane proteins IFITM2 and IFITM3, which are known to inhibit viral fusion. Our results reveal that the pH optimum of HA-driven membrane fusion of IAVs is a determinant of their sensitivity to IFN and IFITM proteins.IMPORTANCE The IFN system constitutes an important innate defense against viral infection. Substantial information is available on how IAVs avoid detection by sensors of the IFN system and disable IFN signaling pathways. Much less is known about the ability of IAVs to tolerate the antiviral activity of IFN-induced cellular proteins. The IFN-induced proteins of the IFITM family block IAV entry into target cells and can restrict viral spread and pathogenicity. Here we show for the first time that the sensitivity of IAVs to the IFN-induced antiviral state and IFITM2 and IFITM3 proteins depends on the pH value at which the viral HA undergoes a conformational transition and mediates membrane fusion. Our data imply that the high pH optimum of membrane fusion typical of zoonotic IAVs of gallinaceous poultry, such as H5N1 and H7N9, may contribute to their enhanced virulence in humans.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype/physiology , Influenza A Virus, H7N9 Subtype/physiology , Interferons/immunology , Membrane Fusion , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , A549 Cells , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Line , Dogs , Ducks , Epithelial Cells/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Hydrogen-Ion Concentration , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H7N9 Subtype/chemistry , Influenza A Virus, H7N9 Subtype/genetics , Interferon-beta/immunology , Madin Darby Canine Kidney Cells , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , Virus Internalization , Virus Replication
13.
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
14.
J Virol ; 90(3): 1569-77, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26608319

ABSTRACT

UNLABELLED: The H1N1 Eurasian avian-like swine (EAsw) influenza viruses originated from an avian H1N1 virus. To characterize potential changes in the membrane fusion activity of the hemagglutinin (HA) during avian-to-swine adaptation of the virus, we studied EAsw viruses isolated in the first years of their circulation in pigs and closely related contemporary H1N1 viruses of wild aquatic birds. Compared to the avian viruses, the swine viruses were less sensitive to neutralization by lysosomotropic agent NH4Cl in MDCK cells, had a higher pH optimum of hemolytic activity, and were less stable at acidic pH. Eight amino acid substitutions in the HA were found to separate the EAsw viruses from their putative avian precursor; four substitutions-T492S, N722D, R752K, and S1132F-were located in the structural regions of the HA2 subunit known to play a role in acid-induced conformational transition of the HA. We also studied low-pH-induced syncytium formation by cell-expressed HA proteins and found that the HAs of the 1918, 1957, 1968, and 2009 pandemic viruses required a lower pH for fusion induction than did the HA of a representative EAsw virus. Our data show that transmission of an avian H1N1 virus to pigs was accompanied by changes in conformational stability and fusion promotion activity of the HA. We conclude that distinctive host-determined fusion characteristics of the HA may represent a barrier for avian-to-swine and swine-to-human transmission of influenza viruses. IMPORTANCE: Continuing cases of human infections with zoonotic influenza viruses highlight the necessity to understand which viral properties contribute to interspecies transmission. Efficient binding of the HA to cellular receptors in a new host species is known to be essential for the transmission. Less is known about required adaptive changes in the membrane fusion activity of the HA. Here we show that adaptation of an avian influenza virus to pigs in Europe in 1980s was accompanied by mutations in the HA, which decreased its conformational stability and increased pH optimum of membrane fusion activity. This finding represents the first formal evidence of alteration of the HA fusion activity/stability during interspecies transmission of influenza viruses under natural settings.


Subject(s)
Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/physiology , Virus Internalization/drug effects , Adaptation, Biological , Animals , Birds , Cell Fusion , Cell Line , Evolution, Molecular , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Hydrogen-Ion Concentration , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza in Birds/virology , Mutation, Missense , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Protein Conformation/drug effects , Protein Stability/drug effects , Swine , Swine Diseases/virology
15.
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
16.
J Virol ; 89(23): 12211-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26378170

ABSTRACT

Hemagglutinin (HA) of H3N2/1968 pandemic influenza viruses differs from the putative avian precursor by seven amino acid substitutions. Substitutions Q226L and G228S are known to be essential for adaptation of avian HA to mammals. We found that introduction of avian-virus-like amino acids at five other HA positions (positions 62, 81, 92, 144, and 193) of A/Hong Kong/1/1968 virus decreased viral replication in human cells and transmission in pigs. Thus, substitutions at some of these positions facilitated emergence of the pandemic virus.


Subject(s)
Amino Acid Substitution/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/epidemiology , Influenza, Human/genetics , Models, Molecular , Pandemics/history , Cluster Analysis , Computational Biology , Evolution, Molecular , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , History, 20th Century , Humans , Models, Genetic , Phylogeny
18.
Eur J Cell Biol ; 94(7-9): 368-74, 2015.
Article in English | MEDLINE | ID: mdl-26074198

ABSTRACT

The segmented genome of influenza viruses is translocated into the nucleus to initiate transcription and replication. The gene segments are present as viral ribonucleoprotein (vRNP) particles composed of RNA, multiple copies of the nucleoprotein (NP), and the polymerase subunits PB1, PB2 and PA. The PB2 subunit and each NP monomer contain a nuclear localisation signal (NLS) that binds to importin-α. To throw light on the role of the NLSs of NP and PB2 in nuclear transport, we have analysed the effect of mutation D701N, responsible for the exposure of the NLS domain of PB2, on the intracellular localisation of vRNPs. We show that exposure of PB2 NLS significantly enhances the amount of vRNPs present in the nucleus. These observations suggest that entry of vRNPs into the nucleus depends on controlled interplay of the NLSs of PB2 and NP with the nuclear import machinery.


Subject(s)
Active Transport, Cell Nucleus/genetics , Influenza A Virus, H7N7 Subtype/genetics , Influenza A Virus, H9N2 Subtype/genetics , RNA-Dependent RNA Polymerase/genetics , Ribonucleoproteins/metabolism , Viral Proteins/genetics , Active Transport, Cell Nucleus/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , HEK293 Cells , Humans , Mutation/genetics , Nuclear Localization Signals/genetics , Protein Transport/genetics , Protein Transport/physiology , RNA, Viral/genetics , Virus Replication/genetics
19.
J Infect Dis ; 212 Suppl 2: S322-8, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26092855

ABSTRACT

The surface glycoprotein (GP) is responsible for Ebola virus (EBOV) attachment and membrane fusion during virus entry. Surface expression of highly glycosylated GP causes marked cytotoxicity via masking of a wide range of cellular surface molecules, including integrins. Considerable amounts of surface GP are shed from virus-infected cells in a soluble truncated form by tumor necrosis factor α-converting enzyme. In this study, the role of GP shedding was investigated using a reverse genetics approach by comparing recombinant viruses possessing amino acid substitutions at the GP shedding site. Virus with an L635V substitution showed a substantial decrease in shedding, whereas a D637V substitution resulted in a striking increase in the release of shed GP. Variations in shedding efficacy correlated with observed differences in the amounts of shed GP in the medium, GP present in virus-infected cells, and GP present on virions. An increase in shedding appeared to be associated with a reduction in viral cytotoxicity, and, vice versa, the virus that shed less was more cytotoxic. An increase in shedding also resulted in a reduction in viral infectivity, whereas a decrease in shedding efficacy enhanced viral growth characteristics in vitro. Differences in shedding efficacy and, as a result, differences in the amount of mature GP available for incorporation into budding virions did not equate to differences in overall release of viral particles. Likewise, data suggest that the resulting differences in the amount of mature GP on the cell surface led to variations in the GP content of released particles and, as a consequence, in infectivity. In conclusion, fine-tuning of the levels of EBOV GP expressed at the surface of virus-infected cells via GP shedding plays an important role in EBOV replication by orchestrating the balance between optimal virion GP content and cytotoxicity caused by GP.


Subject(s)
Ebolavirus/metabolism , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Membrane Glycoproteins/metabolism , Amino Acid Substitution/genetics , Animals , Cell Line , Chlorocebus aethiops , Ebolavirus/genetics , Membrane Glycoproteins/genetics , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/genetics , Virion/metabolism , Virion/pathogenicity , Virulence/genetics , Virus Internalization , Virus Replication/genetics
20.
Antiviral Res ; 120: 89-100, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26022200

ABSTRACT

Antiviral medication is used for the treatment of severe influenza infections, of which the neuraminidase inhibitors (NAIs) are the most effective drugs, approved so far. Here, we investigated the antiviral efficacy of the peptidomimetic furin inhibitor MI-701 in combination with oseltamivir carboxylate and ribavirin against the infection of highly pathogenic avian influenza viruses (HPAIV) that are activated by the host protease furin. Cell cultures infected with the strains A/Thailand/1(KAN-1)/2004 (H5N1) and A/FPV/Rostock/1934 (H7N1) were treated with each agent alone, or in double and triple combinations. MI-701 alone achieved a concentration-dependent reduction of virus propagation. Double treatment of MI-701 with oseltamivir carboxylate and triple combination with ribavirin showed synergistic inhibition and a pronounced delay of virus propagation. MI-701 resistant mutants were not observed. Emergence of NA mutation H275Y conferring high oseltamivir resistance was significantly delayed in the presence of MI-701. Our data indicate that combination with a potent furin inhibitor significantly enhances the therapeutic efficacy of conventional antivirals drugs against HPAIV infection.


Subject(s)
Antiviral Agents/metabolism , Furin/antagonists & inhibitors , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/drug effects , Oseltamivir/metabolism , Peptidomimetics/metabolism , Ribavirin/metabolism , Animals , Dogs , Drug Resistance, Viral , Drug Synergism , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H7N1 Subtype/growth & development , Madin Darby Canine Kidney Cells , Microbial Sensitivity Tests , Mutant Proteins/genetics , Mutation, Missense , Neuraminidase/genetics , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL