Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Eur J Clin Microbiol Infect Dis ; 42(6): 741-746, 2023 Jun.
Article En | MEDLINE | ID: mdl-37084118

Consensus PCR assays that can be used to sensitively detect several herpesvirus (HV) species across the different subfamilies were developed in this study. Primers containing degenerate bases were designed to amplify regions of the DNA polymerase (DPOL) gene of alpha- and gamma-HVs, and the glycoprotein B (gB) gene of beta-HVs in a singleplex, non-nested touchdown PCR format. The singleplex touchdown consensus PCR (STC-PCR) was used to amplify the DNA of eight human and 24 animal HVs. The assay was able to detect the lowest DNA dilution of 10-5 for alpha-HVs and 10-3 for beta- and gamma-HVs. In comparison, lowest detection limits of 10-5, 10-3, and 10-2 were obtained for alpha-, beta-, and gamma-HVs respectively when a nested PCR was used. The findings in this study suggest that the STC-PCR assays can be employed for the molecular surveys and clinical detection of novel and known HVs.


DNA, Viral , Herpesviridae , Animals , Humans , Consensus , DNA, Viral/genetics , DNA, Viral/analysis , Herpesviridae/genetics , DNA Primers/genetics , Polymerase Chain Reaction
2.
PLoS Pathog ; 18(5): e1010150, 2022 05.
Article En | MEDLINE | ID: mdl-35536868

Most of our understanding of the ecology and evolution of avian influenza A virus (AIV) in wild birds is derived from studies conducted in the northern hemisphere on waterfowl, with a substantial bias towards dabbling ducks. However, relevant environmental conditions and patterns of avian migration and reproduction are substantially different in the southern hemisphere. Through the sequencing and analysis of 333 unique AIV genomes collected from wild birds collected over 15 years we show that Australia is a global sink for AIV diversity and not integrally linked with the Eurasian gene pool. Rather, AIV are infrequently introduced to Australia, followed by decades of isolated circulation and eventual extinction. The number of co-circulating viral lineages varies per subtype. AIV haemagglutinin (HA) subtypes that are rarely identified at duck-centric study sites (H8-12) had more detected introductions and contemporary co-circulating lineages in Australia. Combined with a lack of duck migration beyond the Australian-Papuan region, these findings suggest introductions by long-distance migratory shorebirds. In addition, on the available data we found no evidence of directional or consistent patterns in virus movement across the Australian continent. This feature corresponds to patterns of bird movement, whereby waterfowl have nomadic and erratic rainfall-dependant distributions rather than consistent intra-continental migratory routes. Finally, we detected high levels of virus gene segment reassortment, with a high diversity of AIV genome constellations across years and locations. These data, in addition to those from other studies in Africa and South America, clearly show that patterns of AIV dynamics in the Southern Hemisphere are distinct from those in the temperate north.


Influenza A virus , Influenza in Birds , Animals , Animals, Wild , Australia/epidemiology , Birds , Ducks , Genetic Variation , Influenza A virus/genetics , Influenza in Birds/epidemiology , Phylogeny
3.
HardwareX ; 8: e00124, 2020 Oct.
Article En | MEDLINE | ID: mdl-35498236

Bioengineering techniques for producing fibers from biomaterials is a growing requirement in medical device technology research and development environments. Scale-up and control of diameter, shape, and length of fibrous proteins and elastomeric polymers are essential to produce defined and consistent materials for experimentation and clinical use. Here, we developed a novel wet spinning fiber extruder and spooler system engineered to draw precipitated fibers several meters in length across five spools. By controlling both the extrusion and spooling rate, the diameter of the fiber can be controlled on the order of 10-1000 µm. Using this system, we extruded and spooled precipitated Type-1 Collagen fibers up to 7.5 m in length on a single spool with a controllable diameter range of 30-50 µm. Furthermore, this device facilitated bundling of fibers directly on the spool in order to create 1-12 cm long fiber bundles for experimentation. This system may be used in the laboratory to scale up biomaterial fiber production to produce degradable scaffolds made from synthetic or natural materials for a range of biomedical applications.

4.
J Virol ; 93(11)2019 06 01.
Article En | MEDLINE | ID: mdl-30894472

Wild birds harbor a huge diversity of avian avulaviruses (formerly avian paramyxoviruses). Antarctic penguin species have been screened for avian avulaviruses since the 1980s and, as such, are known hosts of these viruses. In this study, we screened three penguin species from the South Shetland Islands and the Antarctic Peninsula for avian avulaviruses. We show that Adelie penguins (Pygoscelis adeliae) are hosts for four different avian avulavirus species, the recently described avian avulaviruses 17 to 19 and avian avulavirus 10-like, never before isolated in Antarctica. A total of 24 viruses were isolated and sequenced; avian avulavirus 17 was the most common, and phylogenetic analysis demonstrated patterns of occurrence, with different genetic clusters corresponding to penguin age and location. Following infection in specific-pathogen-free (SPF) chickens, all four avian avulavirus species were shed from the oral cavity for up to 7 days postinfection. There was limited shedding from the cloaca in a proportion of infected chickens, and all but one bird seroconverted by day 21. No clinical signs were observed. Taken together, we propose that penguin species, including Antarctic penguins, may be the central reservoir for a diversity of avian avulavirus species and that these viruses have the potential to infect other avian hosts.IMPORTANCE Approximately 99% of all viruses are still to be described, and in our changing world, any one of these unknown viruses could potentially expand their host range and cause epidemic disease in wildlife, agricultural animals, or humans. Avian avulavirus 1 causes outbreaks in wild birds and poultry and is thus well described. However, for many avulavirus species, only a single specimen has been described, and their viral ecology and epidemiology are unknown. Through the detection of avian avulaviruses in penguins from Antarctica, we have been able to expand upon our understanding of three avian avulavirus species (avian avulaviruses 17 to 19) and report a potentially novel avulavirus species. Importantly, we show that penguins appear to play a key role in the epidemiology of avian avulaviruses, and we encourage additional sampling of this avian group.


Avulavirus/genetics , Disease Reservoirs/virology , Spheniscidae/virology , Animals , Antarctic Regions , Avulavirus/pathogenicity , Base Sequence , Chickens/genetics , Host Specificity , Phylogeny , Spheniscidae/metabolism
5.
Euro Surveill ; 20(45)2015.
Article En | MEDLINE | ID: mdl-26608955

Surveillance of circulating influenza strains for antiviral susceptibility is important to ensure patient treatment guidelines remain appropriate. Influenza A(H3N2) and A(H1N1)pdm09 virus isolates containing mutations at the Q136 residue of the neuraminidase (NA) that conferred reduced susceptibility to the NA inhibitor (NAI) zanamivir were detected during antiviral susceptibility monitoring. Interestingly, the mutations were not detectable in the viruses from respective clinical specimens, only in the cultured isolates. We showed that variant viruses containing the Q136K and Q136R NA mutations were preferentially selected in Madin-Darby canine kidney epithelial (MDCK) cells, but were less well supported in MDCK-SIAT1 cells and embryonated eggs. The effect of Q136K, Q136R, Q136H and Q136L substitutions in NA subtypes N1 and N2 on NAI susceptibility and in vitro viral fitness was assessed. This study highlights the challenges that cell culture derived mutations can pose to the NAI susceptibility analysis and interpretation and reaffirms the need to sequence viruses from respective clinical specimens to avoid misdiagnosis. However, we also demonstrate that NA mutations at residue Q136 can confer reduced zanamivir, peramivir or laninamivir susceptibility, and therefore close monitoring of viruses for mutations at this site from patients being treated with these antivirals is important.


Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Neuraminidase/genetics , Zanamivir/pharmacology , Animals , Dogs , Enzyme Inhibitors/pharmacology , Female , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza, Human/epidemiology , Madin Darby Canine Kidney Cells/drug effects , Microbial Sensitivity Tests , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/genetics
6.
J Theor Biol ; 382: 259-71, 2015 Oct 07.
Article En | MEDLINE | ID: mdl-26188087

Through accumulation of genetic mutations in the neuraminidase gene, the influenza virus can become resistant to antiviral drugs such as oseltamivir. Quantifying the fitness of emergent drug-resistant influenza viruses, relative to contemporary circulating viruses, provides valuable information to complement existing efforts in the surveillance of drug-resistance. We have previously developed a co-infection based method for the assessment of the relative in vivo fitness of two competing viruses. We have also introduced a model of within-host co-infection dynamics that enables relative within-host fitness to be quantified in these competitive-mixtures experiments. The model assumed that fitness differences between co-infecting strains were mediated by strain-dependent viral production rates from infected epithelial cells. Here we extend the model to enable a more complete exploration of biological processes that may differ between virus pairs and hence generate fitness differences. We use the extended model to re-analyse data from competitive-mixtures experiments that investigated the fitness of oseltamivir-resistant (OR) H1N1 pandemic 2009 ("H1N1pdm09") viruses that emerged during a community outbreak in Australia in 2011. Results are consistent with those of our previous analysis, suggesting that the within-host replication fitness of these OR viruses is not compromised relative to that of related oseltamivir-susceptible (OS) strains, and that potentially permissive mutations in the neuraminidase gene (V241I and N369K) significantly enhance the fitness of H1N1pdm09 OR viruses. These results are consistent regardless of the hypothesised biological cause of fitness difference.


Host-Pathogen Interactions/physiology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/virology , Virus Replication/physiology , Amino Acid Substitution , Disease Outbreaks , Genes, Viral , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/epidemiology , Models, Biological , Mutation/genetics , Neuraminidase/genetics
7.
J Antimicrob Chemother ; 70(7): 2004-12, 2015 Jul.
Article En | MEDLINE | ID: mdl-25786478

OBJECTIVES: The burden of disease due to influenza B is often underestimated. Clinical studies have shown that oseltamivir, a widely used neuraminidase inhibitor (NAI) antiviral drug, may have reduced effectiveness against influenza B viruses. Therefore, it is important to study the effect of neuraminidase mutations in influenza B viruses that may further reduce NAI susceptibility, and to determine whether these mutations have the same effect in the two lineages of influenza B viruses that are currently circulating (B/Yamagata-like and B/Victoria-like). METHODS: We characterized the effect of 16 amino acid substitutions across five framework residues and four monomeric interface residues on the susceptibility to four different NAIs (oseltamivir, zanamivir, peramivir and laninamivir). RESULTS: Framework residue mutations E117A and E117G conferred highly reduced inhibition to three of the four NAIs, but substantially reduced neuraminidase activity, whereas other framework mutations retained a greater level of NA activity. Mutations E105K, P139S and G140R of the monomeric interface were also found to cause highly reduced inhibition, but, interestingly, their effect was substantially greater in a B/Victoria-like neuraminidase than in a B/Yamagata-like neuraminidase, with some susceptibility values being up to 1000-fold different between lineages. CONCLUSIONS: The frequency and the effect of key neuraminidase mutations on neuraminidase activity and NAI susceptibility can differ substantially between the two influenza B lineages. Therefore, future surveillance, analysis and interpretation of influenza B virus NAI susceptibility should consider the B lineage of the neuraminidase in the same manner as already occurs for different influenza A neuraminidase subtypes.


Antiviral Agents/pharmacology , Drug Resistance, Viral , Influenza B virus/drug effects , Influenza B virus/enzymology , Mutation, Missense , Neuraminidase/genetics , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Neuraminidase/chemistry , Protein Conformation
8.
J Virol Methods ; 205: 38-52, 2014 09 01.
Article En | MEDLINE | ID: mdl-24797460

The ferret is an excellent model for many human infectious diseases including influenza, SARS-CoV, henipavirus and pneumococcal infections. The ferret is also used to study cystic fibrosis and various cancers, as well as reproductive biology and physiology. However, the range of reagents available to measure the ferret immune response is very limited. To address this deficiency, high-throughput real time RT-PCR TaqMan assays were developed to measure the expression of fifteen immune mediators associated with the innate and adaptive immune responses (IFNα, IFNß, IFNγ, IL1α, IL1ß, IL2, IL4, IL6, IL8, IL10, IL12p40, IL17, Granzyme A, MCP1, TNFα), as well as four endogenous housekeeping genes (ATF4, HPRT, GAPDH, L32). These assays have been optimized to maximize reaction efficiency, reduce the amount of sample required (down to 1ng RNA per real time RT-PCR reaction) and to select the most appropriate housekeeping genes. Using these assays, the expression of each of the tested genes could be detected in ferret lymph node cells stimulated with mitogens or infected with influenza virus in vitro. These new tools will allow a more comprehensive analysis of the ferret immune responses following infection or in other disease states.


Adaptive Immunity , Cytokines/genetics , Immunity, Innate , Orthomyxoviridae/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Disease Models, Animal , Female , Ferrets , Genes, Essential/genetics , Humans , Leukocytes/immunology , Male , Mitogens/immunology , Real-Time Polymerase Chain Reaction
9.
PLoS Pathog ; 10(4): e1004065, 2014 Apr.
Article En | MEDLINE | ID: mdl-24699865

Oseltamivir is relied upon worldwide as the drug of choice for the treatment of human influenza infection. Surveillance for oseltamivir resistance is routinely performed to ensure the ongoing efficacy of oseltamivir against circulating viruses. Since the emergence of the pandemic 2009 A(H1N1) influenza virus (A(H1N1)pdm09), the proportion of A(H1N1)pdm09 viruses that are oseltamivir resistant (OR) has generally been low. However, a cluster of OR A(H1N1)pdm09 viruses, encoding the neuraminidase (NA) H275Y oseltamivir resistance mutation, was detected in Australia in 2011 amongst community patients that had not been treated with oseltamivir. Here we combine a competitive mixtures ferret model of influenza infection with a mathematical model to assess the fitness, both within and between hosts, of recent OR A(H1N1)pdm09 viruses. In conjunction with data from in vitro analyses of NA expression and activity we demonstrate that contemporary A(H1N1)pdm09 viruses are now more capable of acquiring H275Y without compromising their fitness, than earlier A(H1N1)pdm09 viruses circulating in 2009. Furthermore, using reverse engineered viruses we demonstrate that a pair of permissive secondary NA mutations, V241I and N369K, confers robust fitness on recent H275Y A(H1N1)pdm09 viruses, which correlated with enhanced surface expression and enzymatic activity of the A(H1N1)pdm09 NA protein. These permissive mutations first emerged in 2010 and are now present in almost all circulating A(H1N1)pdm09 viruses. Our findings suggest that recent A(H1N1)pdm09 viruses are now more permissive to the acquisition of H275Y than earlier A(H1N1)pdm09 viruses, increasing the risk that OR A(H1N1)pdm09 will emerge and spread worldwide.


Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Genetic Fitness , Influenza A Virus, H1N1 Subtype/genetics , Mutation, Missense , Neuraminidase/genetics , Oseltamivir/pharmacology , Viral Proteins/genetics , Amino Acid Substitution , Animals , Dogs , Ferrets , Humans , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Influenza, Human/genetics , Madin Darby Canine Kidney Cells , Neuraminidase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors
10.
J Virol ; 87(24): 13706-18, 2013 Dec.
Article En | MEDLINE | ID: mdl-24109221

Yearly vaccination with the trivalent inactivated influenza vaccine (TIV) is recommended, since current vaccines induce little cross neutralization to divergent influenza strains. Whether the TIV can induce antibody-dependent cellular cytotoxicity (ADCC) responses that can cross-recognize divergent influenza virus strains is unknown. We immunized 6 influenza-naive pigtail macaques twice with the 2011-2012 season TIV and then challenged the macaques, along with 12 control macaques, serially with H1N1 and H3N2 viruses. We measured ADCC responses in plasma to a panel of H1 and H3 hemagglutinin (HA) proteins and influenza virus-specific CD8 T cell (CTL) responses using a sensitive major histocompatibility complex (MHC) tetramer reagent. The TIV was weakly immunogenic and, although binding antibodies were detected by enzyme-linked immunosorbent assay (ELISA), did not induce detectable influenza virus-specific ADCC or CTL responses. The H1N1 challenge elicited robust ADCC to both homologous and heterologous H1 HA proteins, but not influenza virus HA proteins from different subtypes (H2 to H7). There was no anamnestic influenza virus-specific ADCC or CTL response in vaccinated animals. The subsequent H3N2 challenge did not induce or boost ADCC either to H1 HA proteins or to divergent H3 proteins but did boost CTL responses. ADCC or CTL responses were not induced by TIV vaccination in influenza-naive macaques. There was a marked difference in the ability of infection compared to that of vaccination to induce cross-reactive ADCC and CTL responses. Improved vaccination strategies are needed to induce broad-based ADCC immunity to influenza.


Antibodies, Viral/immunology , Antibody-Dependent Cell Cytotoxicity , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Animals , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/genetics , Influenza B virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/prevention & control , Influenza, Human/virology , Macaca , Male , Vaccination
11.
Influenza Other Respir Viruses ; 7(6): 904-8, 2013 Nov.
Article En | MEDLINE | ID: mdl-23551973

A minor viral population of oseltamivir-resistant A(H3N2) viruses (E119V neuraminidase mutation) was selected and maintained in a continually infected immunocompromised child following initial oseltamivir treatment. A subsequent course of oseltamivir given 7 weeks later rapidly selected for the E119V variant resulting in a near-pure population of the resistant virus. The study highlights the challenges of oseltamivir treatment of immunocompromised patients that are continually shedding virus and demonstrates the ability of the E119V oseltamivir-resistant virus to be maintained for prolonged periods even in the absence of drug-selective pressure.


Antiviral Agents/therapeutic use , Drug Resistance, Viral , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza, Human/drug therapy , Influenza, Human/virology , Oseltamivir/therapeutic use , Antiviral Agents/pharmacology , Child, Preschool , Humans , Immunocompromised Host , Influenza A Virus, H3N2 Subtype/genetics , Male , Mutation, Missense , Oseltamivir/pharmacology , Selection, Genetic
13.
PLoS One ; 7(2): e30960, 2012.
Article En | MEDLINE | ID: mdl-22363523

Highly pathogenic H5N1 avian influenza viruses have caused major disease outbreaks in domestic and free-living birds with transmission to humans resulting in 59% mortality amongst 564 cases. The mutation of the amino acid at position 627 of the viral polymerase basic-2 protein (PB2) from glutamic acid (E) in avian isolates to lysine (K) in human isolates is frequently found, but it is not known if this change affects the fitness and pathogenicity of the virus in birds. We show here that horizontal transmission of A/Vietnam/1203/2004 H5N1 (VN/1203) virus in chickens and ducks was not affected by the change of K to E at PB2-627. All chickens died between 21 to 48 hours post infection (pi), while 70% of the ducks survived infection. Virus replication was detected in chickens within 12 hours pi and reached peak titers in spleen, lung and brain between 18 to 24 hours for both viruses. Viral antigen in chickens was predominantly in the endothelium, while in ducks it was present in multiple cell types, including neurons, myocardium, skeletal muscle and connective tissues. Virus replicated to a high titer in chicken thrombocytes and caused upregulation of TLR3 and several cell adhesion molecules, which may explain the rapid virus dissemination and location of viral antigen in endothelium. Virus replication in ducks reached peak values between 2 and 4 days pi in spleen, lung and brain tissues and in contrast to infection in chickens, thrombocytes were not involved. In addition, infection of chickens with low pathogenic VN/1203 caused neuropathology, with E at position PB2-627 causing significantly higher infection rates than K, indicating that it enhances virulence in chickens.


Amino Acids/metabolism , Chickens/virology , Ducks/virology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/virology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Animals , Antigens, Viral/immunology , Binding Sites , Blood Platelets/virology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza in Birds/pathology , Influenza in Birds/transmission , RNA-Dependent RNA Polymerase/chemistry , Structure-Activity Relationship , Up-Regulation/genetics , Viral Proteins/chemistry , Virulence , Virus Shedding/physiology
...