Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Virology ; 589: 109926, 2024 01.
Article in English | MEDLINE | ID: mdl-37952465

ABSTRACT

H9N2 subtype avian influenza virus (AIV) can transmit by direct as well as airborne contacts. It has been widespread in poultry and continued to contribute to zoonotic spillover events by providing its six internal genes for the reassortment of novel influenza viruses (eg, H7N9) that infect poultry and humans. Compared to H7N9, H9N2 virus displays an efficient airborne transmissibility in poultry, but the mechanisms of transmission difference have been insufficiently studied. The Hemagglutinin (HA) and viral polymerase acidic protein (PA) have been implicated in the airborne transmission of influenza A viruses. Accordingly, we generated the reassortant viruses of circulating airborne transmissible H9N2 and non-airborne transmissible H7N9 viruses carrying HA and/or PA gene. The introduction of the PA gene from H7N9 into the genome of H9N2 virus resulted in a reduction in airborne transmission among chickens, while the isolated introduction of the HA gene segment completely eliminated airborne transmission among chickens. We further showed that introduction of HA gene of non-transmissible H7N9 did not influence the HA/NA balance of H9N2 virus, but increased the threshold for membrane fusion and decreased the acid stability. Thus, our results indicate that HA protein plays a key role in replication, stability, and airborne transmission of the H9N2 subtype AIV.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Influenza, Human , Humans , Animals , Chickens , Hemagglutinins , Influenza A Virus, H7N9 Subtype/genetics , Respiratory Aerosols and Droplets , Poultry , Viral Proteins/genetics , Viral Proteins/metabolism , Reassortant Viruses/genetics , Reassortant Viruses/metabolism , Phylogeny
2.
J Virol ; 96(13): e0053122, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35727032

ABSTRACT

Segmented RNA viruses are a taxonomically diverse group that can infect plant, wildlife, livestock and human hosts. A shared feature of these viruses is the ability to exchange genome segments during coinfection of a host by a process termed "reassortment." Reassortment enables rapid evolutionary change, but where transmission involves a biological arthropod vector, this change is constrained by the selection pressures imposed by the requirement for replication in two evolutionarily distant hosts. In this study, we use an in vivo, host-arbovirus-vector model to investigate the impact of reassortment on two phenotypic traits, virus infection rate in the vector and virulence in the host. Bluetongue virus (BTV) (Reoviridae) is the causative agent of bluetongue (BT), an economically important disease of domestic and wild ruminants and deer. The genome of BTV comprises 10 linear segments of dsRNA, and the virus is transmitted between ruminants by Culicoides biting midges (Diptera: Ceratopogonidae). Five strains of BTV representing three serotypes (BTV-1, BTV-4, and BTV-8) were isolated from naturally infected ruminants in Europe and ancestral/reassortant lineage status assigned through full genome sequencing. Each strain was then assessed in parallel for the ability to replicate in vector Culicoides and to cause BT in sheep. Our results demonstrate that two reassortment strains, which themselves became established in the field, had obtained high replication ability in C. sonorensis from one of the ancestral virus strains, which allowed inferences of the genome segments conferring this phenotypic trait. IMPORTANCE Reassortment between virus strains can lead to major shifts in the transmission parameters and virulence of segmented RNA viruses, with consequences for spread, persistence, and impact. The ability of these pathogens to adapt rapidly to their environment through this mechanism presents a major challenge in defining the conditions under which emergence can occur. Utilizing a representative mammalian host-insect vector infection and transmission model, we provide direct evidence of this phenomenon in closely related ancestral and reassortant strains of BTV. Our results demonstrate that efficient infection of Culicoides observed for one of three ancestral BTV strains was also evident in two reassortant strains that had subsequently emerged in the same ecosystem.


Subject(s)
Arthropod Vectors , Bluetongue virus , Bluetongue , Ceratopogonidae , Sheep Diseases , Animals , Arthropod Vectors/virology , Bluetongue/transmission , Bluetongue/virology , Bluetongue virus/classification , Bluetongue virus/genetics , Bluetongue virus/pathogenicity , Ceratopogonidae/virology , Deer , Phenotype , Reassortant Viruses/metabolism , Sheep , Sheep Diseases/transmission , Sheep Diseases/virology , Virus Replication
3.
Bioessays ; 43(3): e2000240, 2021 03.
Article in English | MEDLINE | ID: mdl-33200842

ABSTRACT

Severe acute respiratory syndrome-coronavirus (SARS-CoV)-2's origin is still controversial. Genomic analyses show SARS-CoV-2 likely to be chimeric, most of its sequence closest to bat CoV RaTG13, whereas its receptor binding domain (RBD) is almost identical to that of a pangolin CoV. Chimeric viruses can arise via natural recombination or human intervention. The furin cleavage site in the spike protein of SARS-CoV-2 confers to the virus the ability to cross species and tissue barriers, but was previously unseen in other SARS-like CoVs. Might genetic manipulations have been performed in order to evaluate pangolins as possible intermediate hosts for bat-derived CoVs that were originally unable to bind to human receptors? Both cleavage site and specific RBD could result from site-directed mutagenesis, a procedure that does not leave a trace. Considering the devastating impact of SARS-CoV-2 and importance of preventing future pandemics, researchers have a responsibility to carry out a thorough analysis of all possible SARS-CoV-2 origins.


Subject(s)
COVID-19/transmission , Genetic Engineering/ethics , Mutagenesis, Site-Directed/methods , Reassortant Viruses/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , Base Sequence , COVID-19/pathology , COVID-19/virology , China , Chiroptera/virology , Eutheria/virology , Furin/metabolism , Humans , Protein Binding , Reassortant Viruses/metabolism , Reassortant Viruses/pathogenicity , Receptors, Virus/genetics , Receptors, Virus/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Sequence Alignment , Spike Glycoprotein, Coronavirus/metabolism
4.
Virology ; 549: 77-84, 2020 10.
Article in English | MEDLINE | ID: mdl-32853849

ABSTRACT

Human infections with highly pathogenic avian influenza (HPAI) H7N9 virus were detected in late 2016. We examined the drug resistance profile of 30 HPAI H7N9 isolates from Mainland of China (2016-2019). Altogether, 23% (7/30) carried neuraminidase inhibitors (NAIs) - resistance mutations, and 13% (4/30) displayed reduced susceptibility to NAIs in neuraminidase (NA) inhibition test. An HPAI H7N9 reassortment virus we prepared was passaged with NAIs for 10 passages. Passage with zanamivir induced an E119G substitution in NA, whereas passage with oseltamivir induced R292K and E119V substitutions that simulated that seen in oseltamivir -treated HPAI H7N9 cases, indicating that the high frequency of resistant strains in the HPAI H7N9 isolates is related to NAIs use. In presence of NAIs, R238I, A146E, G151E and G234T substitutions were found in HA1 region of HA. No amino acid mutations were found in the internal genes of the recombinant virus.


Subject(s)
Drug Resistance, Viral/genetics , Influenza A Virus, H7N9 Subtype/genetics , Mutation , Neuraminidase/genetics , Reassortant Viruses/genetics , Viral Proteins/genetics , Amino Acid Substitution , Animals , Antiviral Agents/pharmacology , Birds/virology , Enzyme Inhibitors/pharmacology , Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza in Birds/pathology , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza, Human/pathology , Influenza, Human/transmission , Influenza, Human/virology , Microbial Sensitivity Tests , Models, Molecular , Neuraminidase/metabolism , Oseltamivir/pharmacology , Protein Conformation , Reassortant Viruses/drug effects , Reassortant Viruses/metabolism , Reassortant Viruses/pathogenicity , Viral Proteins/metabolism , Zanamivir/pharmacology
5.
J Virol ; 94(13)2020 06 16.
Article in English | MEDLINE | ID: mdl-32321810

ABSTRACT

The Birnaviridae family, responsible for major economic losses to poultry and aquaculture, is composed of nonenveloped viruses with a segmented double-stranded RNA (dsRNA) genome that replicate in discrete cytoplasmic virus factories (VFs). Reassortment is common; however, the underlying mechanism remains unknown given that VFs may act as a barrier to genome mixing. In order to provide new information on VF trafficking during dsRNA virus coinfection, we rescued two recombinant infectious bursal disease viruses (IBDVs) of strain PBG98 containing either a split GFP11 or a tetracysteine (TC) tag fused to the VP1 polymerase (PBG98-VP1-GFP11 and PBG98-VP1-TC). DF-1 cells transfected with GFP1-10 prior to PBG98-VP1-GFP11 infection or stained with a biarsenical derivative of the red fluorophore resorufin (ReAsH) following PBG98-VP1-TC infection, had green or red foci in the cytoplasm, respectively, that colocalized with VP3 and dsRNA, consistent with VFs. The average number of VFs decreased from a mean of 60 to 5 per cell between 10 and 24 h postinfection (hpi) (P < 0.0001), while the average area increased from 1.24 to 45.01 µm2 (P < 0.0001), and live cell imaging revealed that the VFs were highly dynamic structures that coalesced in the cytoplasm. Small VFs moved faster than large (average 0.57 µm/s at 16 hpi compared to 0.22 µm/s at 22 hpi), and VF coalescence was dependent on an intact microtubule network and actin cytoskeleton. During coinfection with PBG98-VP1-GFP11 and PBG98-VP1-TC viruses, discrete VFs initially formed from each input virus that subsequently coalesced 10 to 16 hpi, and we speculate that Birnaviridae reassortment requires VF coalescence.IMPORTANCE Reassortment is common in viruses with segmented double-stranded RNA (dsRNA) genomes. However, these viruses typically replicate within discrete cytoplasmic virus factories (VFs) that may represent a barrier to genome mixing. We generated the first replication competent tagged reporter birnaviruses, infectious bursal disease viruses (IBDVs) containing a split GFP11 or tetracysteine (TC) tag and used the viruses to track the location and movement of IBDV VFs, in order to better understand the intracellular dynamics of VFs during a coinfection. Discrete VFs initially formed from each virus that subsequently coalesced from 10 h postinfection. We hypothesize that VF coalescence is required for the reassortment of the Birnaviridae This study provides new information that adds to our understanding of dsRNA virus VF trafficking.


Subject(s)
Infectious bursal disease virus/genetics , Reassortant Viruses/genetics , Virus Replication/genetics , Animals , Cell Line , Coinfection/metabolism , Cytoplasm , RNA Viruses/genetics , Reassortant Viruses/metabolism , Viral Structural Proteins/genetics
6.
Transbound Emerg Dis ; 67(4): 1677-1687, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32266788

ABSTRACT

The first documented avian influenza virus subtype H16N3 was isolated in 1975 and is currently detectable in many countries worldwide. However, the prevalence, biological characteristics and threat to humans of the avian influenza virus H16N3 subtype in China remain poorly understood. We performed avian influenza surveillance in major wild bird gatherings across the country from 2017 to 2019, resulting in the isolation of two H16N3 subtype influenza viruses. Phylogenetic analysis showed these viruses belong to the Eurasian lineage, and both viruses presented the characteristics of inter-species reassortment. In addition, the two viruses exhibited limited growth capacity in MDCK and A549 cells. Receptor-binding assays indicated that the two H16N3 viruses presented dual receptor-binding profiles, being able to bind to both human and avian-type receptors, where GBHG/NX/2/2018(H16N3) preferentially bound the avian-type receptor, while GBHG/NX/1/2018(H16N3) showed greater binding to the human-type receptor, even the mice virulence data showed the negative results. Segments from other species have been introduced into the H16N3 avian influenza virus, which may alter its pathogenicity and host tropism, potentially posing a threat to animal and human health in the future. Consequently, it is necessary to increase monitoring of the emergence and spread of avian influenza subtype H16N3 in wild birds.


Subject(s)
Birds/virology , Influenza A virus/isolation & purification , Influenza in Birds/virology , Reassortant Viruses/isolation & purification , Animals , Animals, Wild/virology , China/epidemiology , Feces/virology , Female , Genes, Viral , Genome, Viral , Humans , Influenza A virus/metabolism , Influenza in Birds/epidemiology , Mice , Mice, Inbred BALB C , Phylogeny , RNA, Viral , Reassortant Viruses/metabolism , Receptors, Virus/metabolism , Viral Plaque Assay
7.
Sci Adv ; 6(10): eaaz1590, 2020 03.
Article in English | MEDLINE | ID: mdl-32181359

ABSTRACT

Paramyxoviruses are negative-polarity RNA viruses of major clinical importance. The dynamic interaction of the RNA-dependent RNA polymerase (RdRP) complex with the encapsidated RNA genome is mechanistically and structurally poorly understood. Having generated recombinant measles (MeV) and canine distemper (CDV) viruses with truncated nucleocapsid (N) protein showing defects in replication kinetics, we have applied a viral evolution approach to the problem. Passaging of recombinants resulted in long-range compensatory mutations that restored RdRP bioactivity in minigenome assays and efficient replication of engineered viruses. Compensatory mutations clustered at an electronically compatible acidic loop in N-core and a basic face of the phosphoprotein X domain (P-XD). Co-affinity precipitations, biolayer interferometry, and molecular docking revealed an electrostatic-driven transiently forming interface between these domains. The compensatory mutations reduced electrostatic compatibility of these microdomains and lowered coprecipitation efficiency, consistent with a molecular checkpoint function that regulates paramyxovirus polymerase mobility through modulation of conformational stability of the P-XD assembly.


Subject(s)
Distemper Virus, Canine/genetics , Measles virus/genetics , Nucleocapsid Proteins/chemistry , Phosphoproteins/chemistry , RNA-Dependent RNA Polymerase/chemistry , Reassortant Viruses/genetics , Virus Replication/genetics , Animals , Binding Sites , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Cricetulus , Distemper Virus, Canine/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Measles virus/metabolism , Molecular Docking Simulation , Mutation , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Reassortant Viruses/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity , Vero Cells
8.
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
9.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31597771

ABSTRACT

Efficient human-to-human transmission is a prerequisite for a novel influenza virus to cause an influenza pandemic; however, the genetic determinants of influenza virus transmission are still not fully understood. In this study, we compared the respiratory droplet transmissibilities of four H7N9 viruses that are genetic closely related and found that these viruses have dissimilar transmissibilities in guinea pigs: A/Anhui/1/2013 (AH/1) transmitted efficiently, whereas the other three viruses did not transmit. The three nontransmissible viruses have one to eight amino acid differences compared with the AH/1 virus. To investigate which of these amino acids is important for transmission, we used reverse genetics to generate a series of reassortants and mutants in the AH/1 background and tested their transmissibility in guinea pigs. We found that the neuraminidase (NA) of the nontransmissible virus A/chicken/Shanghai/S1053/2013 had low enzymatic activity that impaired the transmission of AH/1 virus, and three amino acid mutations-V292I and K627E in PB2 and D156E in M1-independently abolished the transmission of the AH/1 virus. We further found that an NA reassortant and three single-amino-acid mutants replicated less efficiently than the AH/1 virus in A549 cells and that the amino acid at position 156 of M1 affected the morphology of H7N9 viruses. Our study identifies key amino acids in PB2 and M1 that play important roles in H7N9 influenza virus transmission and provides new insights into the transmissibility of influenza virus.IMPORTANCE Efficient transmission is a prerequisite for a novel influenza virus to cause an influenza pandemic; however, the genetic determinants of influenza virus transmission remain poorly understood. H7N9 influenza viruses, which emerged in 2013 in China, have caused over 1,560 human infection cases, showing clear pandemic potential. Previous studies have shown that the H7N9 viruses differ in their transmissibility in animal models. In this study, we found two amino acids in PB2 (292V and 627K) and one in M1 (156D) that are extremely important for H7N9 virus transmission. Of note, PB2 292V and M1 156D appear in most H7N9 viruses, and the PB2 627K mutation could easily occur when the H7N9 virus replicates in humans. Our study thus identifies new amino acids that are important for influenza virus transmission and suggests that just a few key amino acid changes can render the H7N9 virus transmissible in mammals.


Subject(s)
Influenza A Virus, H7N9 Subtype/genetics , Neuraminidase/genetics , Orthomyxoviridae Infections/transmission , RNA-Dependent RNA Polymerase/genetics , Reassortant Viruses/genetics , Viral Matrix Proteins/genetics , Viral Proteins/genetics , A549 Cells , Amino Acid Substitution , Animals , Gene Expression , Guinea Pigs , Humans , Influenza A Virus, H7N9 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/pathogenicity , Mutation , Neuraminidase/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , RNA-Dependent RNA Polymerase/metabolism , Reassortant Viruses/metabolism , Reassortant Viruses/pathogenicity , Reverse Genetics , Structure-Activity Relationship , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism , Virus Replication
10.
J Gen Virol ; 100(7): 1079-1092, 2019 07.
Article in English | MEDLINE | ID: mdl-31169484

ABSTRACT

Candidate vaccine viruses (CVVs) for seasonal influenza A virus are made by reassortment of the antigenic virus with an egg-adapted strain, typically A/Puerto Rico/8/34 (PR8). Many 2009 A(H1N1) pandemic (pdm09) high-growth reassortants (HGRs) selected this way contain pdm09 segment 2 in addition to the antigenic genes. To investigate this, we made CVV mimics by reverse genetics (RG) that were either 6 : 2 or 5 : 3 reassortants between PR8 and two pdm09 strains, A/California/7/2009 (Cal7) and A/England/195/2009, differing in the source of segment 2. The 5 : 3 viruses replicated better in MDCK-SIAT1 cells than the 6 : 2 viruses, but the 6 : 2 CVVs gave higher haemagglutinin (HA) antigen yields from eggs. This unexpected phenomenon reflected temperature sensitivity conferred by pdm09 segment 2, as the egg HA yields of the 5 : 3 viruses improved substantially when viruses were grown at 35 °C compared with 37.5 °C, whereas the 6 : 2 virus yields did not. However, the authentic 5 : 3 pdm09 HGRs, X-179A and X-181, were not markedly temperature sensitive despite their PB1 sequences being identical to that of Cal7, suggesting compensatory mutations elsewhere in the genome. Sequence comparisons of the PR8-derived backbone genes identified polymorphisms in PB2, NP, NS1 and NS2. Of these, PB2 N701D affected the temperature dependence of viral transcription and, furthermore, improved and drastically reduced the temperature sensitivity of the HA yield from the 5 : 3 CVV mimic. We conclude that the HA yield of pdm09 CVVs can be affected by an epistatic interaction between PR8 PB2 and pdm09 PB1, but that this can be minimized by ensuring that the backbones used for vaccine manufacture in eggs contain PB2 701D.


Subject(s)
Epistasis, Genetic , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/growth & development , Influenza, Human/virology , Viral Proteins/genetics , Animals , Chick Embryo , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Influenza Vaccines/genetics , Influenza Vaccines/metabolism , Reassortant Viruses/genetics , Reassortant Viruses/growth & development , Reassortant Viruses/metabolism , Temperature , Viral Proteins/metabolism
11.
Virology ; 523: 22-26, 2018 10.
Article in English | MEDLINE | ID: mdl-30075357

ABSTRACT

Live bird market surveillance for avian influenza viruses in Cambodia in 2015 has led to the detection of two 7:1 reassortant influenza A(H5N1) clade 2.3.2.1c viruses. These reassortant strains, designated A/duck/Cambodia/Z564W35M1/2015 and A/chicken/Cambodia/Z850W49M1/2015, both contained a single gene (PB1 and matrix gene, respectively) from concurrently circulating A(H9N2) influenza viruses. All other viral genes from both isolates clustered with A(H5N1) clade 2.3.2.1 viruses. Continued and prolonged co-circulation of influenza A(H5N1) and A(H9N2) viruses in Cambodian live bird markets may present a risk for the emergence of novel influenza reassortant viruses with negative agricultural and/or public health implications.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H9N2 Subtype/genetics , Influenza in Birds/epidemiology , Poultry Diseases/epidemiology , Reassortant Viruses/genetics , Animals , Cambodia/epidemiology , Chickens , Ducks , Epidemiological Monitoring , Gene Expression , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A Virus, H9N2 Subtype/classification , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza A Virus, H9N2 Subtype/metabolism , Influenza in Birds/transmission , Influenza in Birds/virology , Phylogeny , Poultry Diseases/transmission , Poultry Diseases/virology , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Reassortant Viruses/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Virus Res ; 253: 20-27, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29859234

ABSTRACT

The reassortment of two highly pathogenic avian influenza (HPAI) H5N1 and H7N9 viruses presents a potential challenge to human health. The hemagglutinins (HAs) and neuraminidases (NAs) of these simultaneously circulating avian influenza viruses were evaluated using the pseudoparticle (pp) system. Native and mismatched virus pps were generated to investigate their biological characteristics. The HAs and NAs of the two viruses reassorted successfully to generate infectious viral particles. H7 was demonstrated to have the ability to reassort with NA from the H5N1 viruses, resulting in the generation of virions that were highly infectious to bronchial epithelial cells. Although the Anhui H5+Anhui N9 combination showed an moderate infectivity to the four cell lines, it was most sensitive to oseltamivir. The H7 in the pps was found to be predominantly HA0. Further, H5 in the pps primarily presented as HA1, owing to the particular mechanisms underlying its maturation. All NAs predominantly existed in monomer form. In our study, HAs/NAs, in all combinations, were functional and able to perform their corresponding function in the viral life cycle. Our data suggest that HAs/NAs from the (HPAI) H5N1 and H7N9 viruses are capable of assembly into infectious virions, posing a threat topublic health.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/metabolism , Influenza, Human/virology , Neuraminidase/metabolism , Reassortant Viruses/metabolism , Virion/metabolism , Animals , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/enzymology , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza in Birds/virology , Neuraminidase/genetics , Poultry Diseases/virology , Reassortant Viruses/enzymology , Reassortant Viruses/genetics , Recombination, Genetic , Virion/enzymology , Virion/genetics , Virion/pathogenicity , Virulence
13.
Viral Immunol ; 31(6): 457-469, 2018.
Article in English | MEDLINE | ID: mdl-29870311

ABSTRACT

Influenza virus infections can be complicated by bacterial superinfections, which are medically relevant because of a complex interaction between the host, the virus, and the bacteria. Studies to date have implicated several influenza virus genes, varied host immune responses, and bacterial virulence factors, however, the host-pathogen interactions that predict survival versus lethal outcomes remain undefined. Previous work by our group showed that certain influenza viruses could yield a survival phenotype (A/swine/Texas/4199-2/98-H3N2, TX98), whereas others were associated with a lethal phenotype (A/Puerto Rico/8/34-H1N1, PR8). Based on this observation, we developed the hypothesis that individual influenza virus genes could contribute to a superinfection, and that the host response after influenza virus infection could influence superinfection severity. The present study analyzes individual influenza virus gene contributions to superinfection severity using reassortant viruses created using TX98 and PR8 viral genes. Host and pathogen interactions, relevant to survival and lethal phenotypes, were studied with a focus on pathogen clearance, host cellular infiltrates, and cytokine levels after infection. Specifically, we found that the hemagglutinin gene expressed by an influenza virus can contribute to the severity of a secondary bacterial infection, likely through modulation of host proinflammatory responses. Altogether, these results advance our understanding of molecular mechanisms underlying influenza virus-bacteria superinfections and identify viral and corresponding host factors that may contribute to morbidity and mortality.


Subject(s)
Alphainfluenzavirus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza, Human/immunology , Reassortant Viruses/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Superinfection/immunology , Animals , Disease Models, Animal , Female , Host-Pathogen Interactions/immunology , Humans , Influenza, Human/diagnosis , Influenza, Human/mortality , Influenza, Human/virology , Alphainfluenzavirus/metabolism , Mice, Inbred BALB C , Reassortant Viruses/metabolism , Severity of Illness Index , Streptococcal Infections/microbiology , Streptococcal Infections/mortality , Superinfection/microbiology , Superinfection/mortality , Virulence Factors/immunology
14.
Arch Virol ; 163(9): 2471-2476, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29786119

ABSTRACT

Recombination occurs frequently between enteroviruses (EVs) which are classified within the same species of the Picornaviridae family. Here, using viral metagenomics, the genomes of two recombinant EV-Gs (strains EVG 01/NC_CHI/2014 and EVG 02/NC_CHI/2014) found in the feces of pigs from a swine farm in China are described. The two strains are characterized by distinct insertion of a papain-like protease gene from toroviruses classified within the Coronaviridae family. According to recent reports the site of the torovirus protease insertion was located at the 2C/3A junction region in EVG 02/NC_CHI/2014. For the other variant EVG 01/NC_CHI/2014, the inserted protease sequence replaced the entire viral capsid protein region up to the VP1/2A junction. These two EV-G strains were highly prevalent in the same pig farm with all animals shedding the full-length genome (EVG 02/NC_CHI/2014) while 65% also shed the capsid deletion mutant (EVG 01/NC_CHI/2014). A helper-defective virus relationship between the two co-circulating EV-G recombinants is hypothesized.


Subject(s)
Enterovirus Infections/veterinary , Enteroviruses, Porcine/genetics , Genome, Viral , Reassortant Viruses/genetics , Swine Diseases/epidemiology , Torovirus Infections/veterinary , Torovirus/genetics , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , China/epidemiology , Endopeptidases/genetics , Endopeptidases/metabolism , Enterovirus Infections/epidemiology , Enterovirus Infections/virology , Enteroviruses, Porcine/classification , Enteroviruses, Porcine/metabolism , Farms , Feces/virology , Gene Deletion , Genetic Variation , Metagenomics/methods , Phylogeny , Prevalence , Reassortant Viruses/classification , Reassortant Viruses/metabolism , Recombination, Genetic , Swine , Swine Diseases/virology , Torovirus/classification , Torovirus/metabolism , Torovirus Infections/epidemiology , Torovirus Infections/virology , Viral Proteins/genetics , Viral Proteins/metabolism
15.
Sci Rep ; 7(1): 11886, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28928365

ABSTRACT

Swine play a key role in the ecology and transmission of influenza A viruses (IAVs) between species. However, the epidemiology and diversity of swine IAVs is not completely understood. In this cohort study, we sampled on a weekly basis 132 3-week old pigs for 15 weeks. We found two overlapping epidemic events of infection in which most pigs (98.4%) tested PCR positive for IAVs. The prevalence rate of infection ranged between 0 and 86% per week and the incidence density ranged between 0 and 71 cases per 100 pigs-week. Three distinct influenza viral groups (VGs) replicating as a "swarm" of viruses were identified (swine H1-gamma, H1-beta, and H3-cluster-IV IAVs) and co-circulated at different proportions over time suggesting differential allele fitness. Furthermore, using deep genome sequencing 13 distinct viral genome constellations were differentiated. Moreover, 78% of the pigs had recurrent infections with IAVs closely related to each other or IAVs clearly distinct. Our results demonstrated the molecular complexity of swine IAVs during natural infection of pigs in which novel strains of IAVs with zoonotic and pandemic potential can emerge. These are key findings to design better health interventions to reduce the transmission of swine IAVs and minimize the public health risk.


Subject(s)
Epidemics , Genome, Viral , Influenza A virus/genetics , Orthomyxoviridae Infections/genetics , Reassortant Viruses/genetics , Swine Diseases/genetics , Animals , Influenza A virus/metabolism , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Reassortant Viruses/metabolism , Swine , Swine Diseases/epidemiology , Swine Diseases/metabolism , Swine Diseases/virology
16.
Sci Rep ; 7(1): 10232, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28860626

ABSTRACT

The outbreak of a pandemic influenza H1N1 in 2009 required the rapid generation of high-yielding vaccines against the A/California/7/2009 virus, which were achieved by either addition or deletion of a glycosylation site in the influenza proteins hemagglutinin and neuraminidase. In this report, we have systematically evaluated the glycan composition, structural distribution and topology of glycosylation for two high-yield candidate reassortant vaccines (NIBRG-121xp and NYMC-X181A) by combining various enzymatic digestions with high performance liquid chromatography and multiple-stage mass spectrometry. Proteomic data analyses of the full-length protein sequences determined 9 N-glycosylation sites of hemagglutinin, and defined 6 N-glycosylation sites and the glycan structures of low abundance neuraminidase, which were occupied by high-mannose, hybrid and complex-type N-glycans. A total of ~300 glycopeptides were analyzed and manually validated by tandem mass spectrometry. The specific N-glycan structure and topological location of these N-glycans are highly correlated to the spatial protein structure and the residential ligand binding. Interestingly, sulfation, fucosylation and bisecting N-acetylglucosamine of N-glycans were also reliably identified at the specific glycosylation sites of the two influenza proteins that may serve a crucial role in regulating the protein structure and increasing the protein abundance of the influenza virus reassortants.


Subject(s)
Hemagglutinins/chemistry , Influenza A Virus, H1N1 Subtype/metabolism , Neuraminidase/chemistry , Proteomics/methods , Reassortant Viruses/metabolism , Viral Proteins/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid , Glycosylation , Hemagglutinins/analysis , Hemagglutinins/genetics , Humans , Influenza A Virus, H1N1 Subtype/chemistry , Influenza Vaccines/chemistry , Influenza Vaccines/metabolism , Models, Molecular , Neuraminidase/analysis , Polysaccharides/analysis , Polysaccharides/chemistry , Protein Conformation , Reassortant Viruses/chemistry , Tandem Mass Spectrometry , Viral Proteins/analysis
17.
Influenza Other Respir Viruses ; 11(3): 298-303, 2017 05.
Article in English | MEDLINE | ID: mdl-28245096

ABSTRACT

This report describes a triple-reassortant influenza A virus with a HA that resembles H3 of human seasonal influenza from 2004 to 2005, N2 from influenza A virus already established in swine, and the internal gene cassette from A(H1N1)pdm09 has spread in Danish pig herds. The virus has been detected in several Danish pig herds during the last 2-3 years and may possess a challenge for human as well as animal health.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A virus/genetics , Influenza, Human/virology , Neuraminidase/genetics , Orthomyxoviridae Infections/veterinary , Reassortant Viruses/genetics , Swine Diseases/virology , Viral Proteins/genetics , Animals , Denmark , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A virus/classification , Influenza A virus/metabolism , Neuraminidase/metabolism , Orthomyxoviridae Infections/virology , Phylogeny , Reassortant Viruses/classification , Reassortant Viruses/immunology , Reassortant Viruses/metabolism , Recombination, Genetic , Seasons , Swine , Viral Proteins/metabolism
18.
Antimicrob Agents Chemother ; 60(11): 6679-6691, 2016 11.
Article in English | MEDLINE | ID: mdl-27572398

ABSTRACT

T-705 (favipiravir) is a new antiviral agent in advanced clinical development for influenza therapy. It is supposed to act as an alternative substrate for the viral polymerase, causing inhibition of viral RNA synthesis or virus mutagenesis. These mechanisms were also proposed for ribavirin, an established and broad antiviral drug that shares structural similarity with T-705. We here performed a comparative analysis of the effects of T-705 and ribavirin on influenza virus and host cell functions. Influenza virus-infected cell cultures were exposed to T-705 or ribavirin during single or serial virus passaging. The effects on viral RNA synthesis and infectious virus yield were determined and mutations appearing in the viral genome were detected by whole-genome virus sequencing. In addition, the cellular nucleotide pools as well as direct inhibition of the viral polymerase enzyme were quantified. We demonstrate that the anti-influenza virus effect of ribavirin is based on IMP dehydrogenase inhibition, which results in fast and profound GTP depletion and an imbalance in the nucleotide pools. In contrast, T-705 acts as a potent and GTP-competitive inhibitor of the viral polymerase. In infected cells, viral RNA synthesis is completely inhibited by T-705 or ribavirin at ≥50 µM, whereas exposure to lower drug concentrations induces formation of noninfectious particles and accumulation of random point mutations in the viral genome. This mutagenic effect is 2-fold higher for T-705 than for ribavirin. Hence, T-705 and ribavirin both act as purine pseudobases but profoundly differ with regard to the mechanism behind their antiviral and mutagenic effects on influenza virus.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Gene Expression Regulation, Viral , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Pyrazines/pharmacology , Reassortant Viruses/drug effects , Ribavirin/pharmacology , A549 Cells , Amides/chemistry , Animals , Antiviral Agents/chemistry , Chick Embryo , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Dogs , Humans , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/metabolism , Madin Darby Canine Kidney Cells , Mutation/drug effects , Pyrazines/chemistry , RNA, Viral/antagonists & inhibitors , RNA, Viral/biosynthesis , Reassortant Viruses/genetics , Reassortant Viruses/growth & development , Reassortant Viruses/metabolism , Ribavirin/chemistry , Sequence Analysis, RNA , Structure-Activity Relationship , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
19.
PLoS One ; 11(7): e0160040, 2016.
Article in English | MEDLINE | ID: mdl-27454606

ABSTRACT

In 2013, the first case of human infection with an avian influenza A virus (H7N9) was reported in China, and the human infection with this virus has continued as of 2016. At the request of the WHO, we have successfully developed candidate reassortant vaccine virus using A/Anhui/1/2013 and the high egg-growth master virus A/PR/8/1934. Recent plans regarding influenza vaccine production include using cell-cultured systems in Japan and several other countries. However, egg-based vaccine viruses are not always suitable for cell-cultured vaccine production due to potential issues with growth, protein yield and antigenic stability. Therefore, in this study, we have developed a high-growth master virus (hg-PR8) adapted to qualified NIID-MDCK cells that are competent for vaccine production. The virus hg-PR8 was obtained after 20 serial passages of A/Puerto Rico/8/1934 (PR8) in NIID-MDCK cells. The viral titer of hg-PR8 was 108.6 plaque-forming units per milliliter (PFU/mL). Seven amino acid substitutions were identified in the amino acid sequences of PB2, PB1, PA, NA, M and NS of hg-PR8 compared to the sequence of the original PR8 (org-PR8) strain. The growth capacities of the reassortant viruses, which possess heterologous internal genes from hg-PR8 or org-PR8, indicated that the amino acid changes in PB2 and NS2 similarly affected growth capacity in NIID-MDCK cells. To assess the suitability of hg-PR8 as a master virus, we generated 6:2 reassortant viruses possessing the HA and NA segments from A/Anhui/1/2013 (H7N9) and the remaining segments from hg-PR8. The virus titers of the reassortant strains were 107-108 PFU/mL. The antigenicity of the viruses was stable during ten passages of the viruses in NIID-MDCK cells. In comparison with the egg-based reassortant vaccine viruses with identical HA and NA segments, the hg-PR8-based viruses showed 1.5- to 2-fold higher protein yields in NIID-MDCK cells.


Subject(s)
Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Reassortant Viruses/immunology , Adaptation, Biological , Amino Acid Substitution , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , Cell Line , Cells, Cultured , Dogs , Genes, Viral , Glycosylation , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/metabolism , Influenza, Human/immunology , Influenza, Human/prevention & control , Mutation , Reassortant Viruses/genetics , Reassortant Viruses/metabolism , Virus Replication/genetics
20.
Infect Genet Evol ; 41: 279-288, 2016 07.
Article in English | MEDLINE | ID: mdl-27101787

ABSTRACT

H9N2 influenza A viruses are on the list of potentially pandemic subtypes. Therefore, it is important to understand how genomic reassortment and genetic polymorphisms affect phenotypes of H9N2 viruses circulating in the wild bird reservoir. A comparative genetic analysis of North American H9N2 isolates of wild bird origin identified a naturally occurring reassortant virus containing gene segments derived from both North American and Eurasian lineage ancestors. The PB2 segment of this virus encodes 10 amino acid changes that distinguish it from other H9 strains circulating in North America. G590S, one of the 10 amino acid substitutions observed, was present in ~12% of H9 viruses worldwide. This mutation combined with R591 has been reported as a marker of pathogenicity for human pandemic 2009 H1N1 viruses. Screening by polymerase reporter assay of all the natural polymorphisms at these two positions identified G590/K591 and S590/K591 as the most active, with the highest polymerase activity recorded for the SK polymorphism. Rescued viruses containing these two polymorphic combinations replicated more efficiently in MDCK cells and they were the only ones tested that were capable of establishing productive infection in NHBE cells. A global analysis of all PB2 sequences identified the K591 signature in six viral HA/NA subtypes isolated from several hosts in seven geographic locations. Interestingly, introducing the K591 mutation into the PB2 of a human-adapted H3N2 virus did not affect its polymerase activity. Our findings demonstrate that a single point mutation in the PB2 of a low pathogenic H9N2 isolate could have a significant effect on viral phenotype and increase its propensity to infect mammals. However, this effect is not universal, warranting caution in interpreting point mutations without considering protein sequence context.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/genetics , Point Mutation , Reassortant Viruses/genetics , Viral Proteins/genetics , Amino Acid Substitution , Animals , Birds/virology , Bronchi/pathology , Bronchi/virology , Dogs , Epithelial Cells/pathology , Epithelial Cells/virology , Gene Expression , HEK293 Cells , Host Specificity , Humans , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/metabolism , Influenza A Virus, H9N2 Subtype/classification , Influenza A Virus, H9N2 Subtype/metabolism , Lysine/metabolism , Madin Darby Canine Kidney Cells , Phylogeny , Polymorphism, Genetic , Reassortant Viruses/metabolism , Viral Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...