Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Immunol ; 14: 1129765, 2023.
Article in English | MEDLINE | ID: mdl-36926342

ABSTRACT

Introduction: External Quality Assessment (EQA) schemes are designed to provide a snapshot of laboratory proficiency, identifying issues and providing feedback to improve laboratory performance and inter-laboratory agreement in testing. Currently there are no international EQA schemes for seasonal influenza serology testing. Here we present a feasibility study for conducting an EQA scheme for influenza serology methods. Methods: We invited participant laboratories from industry, contract research organizations (CROs), academia and public health institutions who regularly conduct hemagglutination inhibition (HAI) and microneutralization (MN) assays and have an interest in serology standardization. In total 16 laboratories returned data including 19 data sets for HAI assays and 9 data sets for MN assays. Results: Within run analysis demonstrated good laboratory performance for HAI, with intrinsically higher levels of intra-assay variation for MN assays. Between run analysis showed laboratory and strain specific issues, particularly with B strains for HAI, whilst MN testing was consistently good across labs and strains. Inter-laboratory variability was higher for MN assays than HAI, however both assays showed a significant reduction in inter-laboratory variation when a human sera pool is used as a standard for normalization. Discussion: This study has received positive feedback from participants, highlighting the benefit such an EQA scheme would have on improving laboratory performance, reducing inter laboratory variation and raising awareness of both harmonized protocol use and the benefit of biological standards for seasonal influenza serology testing.


Subject(s)
Influenza, Human , Humans , Hemagglutination , Laboratories , Feasibility Studies , Seasons
2.
Avian Pathol ; 52(1): 36-50, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36205531

ABSTRACT

Newcastle disease (ND) is caused by virulent forms of avian paramyxovirus-1 (APMV-1) and is an economically important disease of poultry world-wide. Pigeon paramyxovirus 1 (PPMV-1), a sub-group of APMV-1 is endemic in Columbiformes and can cause infections of poultry. An outbreak of ND in partridges in Scotland, UK, in 2006 (APMV-1/partridge/UK(Scotland)/7575/06) was identified as a class II, genotype VI.2.1.1.2.1, more commonly associated with PPMV-1. It has been hypothesized that game birds may be a route of transmission into commercial poultry settings due to the semi-feral rearing system, which potentially brings them into contact with both wild-birds and poultry species. Therefore, the pathogenesis and transmission of APMV-1/partridge/UK(Scotland)/7575/06 in game birds and chickens was investigated, and compared to a contemporary PPMV-1 isolate, PPMV-1/pigeon/UK/015874/15. Viral shedding and seroconversion profiles demonstrated that pheasants were susceptible to infection with APMV-1/partridge/UK(Scotland)/7575/06 with limited clinical signs observed although they were able to excrete and transmit virus. In contrast, partridges and pheasants showed limited infection with PPMV-1/pigeon/UK/015874/15, causing mild clinical disease. Chickens, however, were productively infected and were able to transmit virus in the absence of clinical signs. From the data, it can be deduced that whilst game birds may play a role in the transmission and epidemiology of genotype VI.2 APMV-1 viruses, the asymptomatic nature of circulation within these species precludes evaluation of natural infection by clinical surveillance. It therefore remains a possibility that genotype VI.2 APMV-1 infection in game birds has the potential for asymptomatic circulation and remains a potential threat to avian production systems.RESEARCH HIGHLIGHTS Demonstration of infection of game birds with Pigeon paramyxovirus-1 (PPMV-1).There are differing dynamics of infection between different game bird species.Differing dynamics of infection between different PPMV-1 isolates and genotypes in game birds and chickens.


Subject(s)
Chickens , Newcastle Disease , Animals , Phylogeny , Newcastle disease virus , Poultry , Quail , Genotype
3.
Nat Med ; 28(5): 1031-1041, 2022 05.
Article in English | MEDLINE | ID: mdl-35361992

ABSTRACT

Since its emergence in 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused hundreds of millions of cases and continues to circulate globally. To establish a novel SARS-CoV-2 human challenge model that enables controlled investigation of pathogenesis, correlates of protection and efficacy testing of forthcoming interventions, 36 volunteers aged 18-29 years without evidence of previous infection or vaccination were inoculated with 10 TCID50 of a wild-type virus (SARS-CoV-2/human/GBR/484861/2020) intranasally in an open-label, non-randomized study (ClinicalTrials.gov identifier NCT04865237 ; funder, UK Vaccine Taskforce). After inoculation, participants were housed in a high-containment quarantine unit, with 24-hour close medical monitoring and full access to higher-level clinical care. The study's primary objective was to identify an inoculum dose that induced well-tolerated infection in more than 50% of participants, with secondary objectives to assess virus and symptom kinetics during infection. All pre-specified primary and secondary objectives were met. Two participants were excluded from the per-protocol analysis owing to seroconversion between screening and inoculation, identified post hoc. Eighteen (~53%) participants became infected, with viral load (VL) rising steeply and peaking at ~5 days after inoculation. Virus was first detected in the throat but rose to significantly higher levels in the nose, peaking at ~8.87 log10 copies per milliliter (median, 95% confidence interval (8.41, 9.53)). Viable virus was recoverable from the nose up to ~10 days after inoculation, on average. There were no serious adverse events. Mild-to-moderate symptoms were reported by 16 (89%) infected participants, beginning 2-4 days after inoculation, whereas two (11%) participants remained asymptomatic (no reportable symptoms). Anosmia or dysosmia developed more slowly in 15 (83%) participants. No quantitative correlation was noted between VL and symptoms, with high VLs present even in asymptomatic infection. All infected individuals developed serum spike-specific IgG and neutralizing antibodies. Results from lateral flow tests were strongly associated with viable virus, and modeling showed that twice-weekly rapid antigen tests could diagnose infection before 70-80% of viable virus had been generated. Thus, with detailed characterization and safety analysis of this first SARS-CoV-2 human challenge study in young adults, viral kinetics over the course of primary infection with SARS-CoV-2 were established, with implications for public health recommendations and strategies to affect SARS-CoV-2 transmission. Future studies will identify the immune factors associated with protection in those participants who did not develop infection or symptoms and define the effect of prior immunity and viral variation on clinical outcome.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Viral , Humans , Kinetics , Treatment Outcome , Viral Load , Young Adult
4.
Emerg Infect Dis ; 26(2): 273-281, 2020 02.
Article in English | MEDLINE | ID: mdl-31961298

ABSTRACT

Influenza A(H1N1)pdm09 (pH1N1) virus has become established in swine in the United Kingdom and currently co-circulates with previously enzootic swine influenza A virus (IAV) strains, including avian-like H1N1 and human-like H1N2 viruses. During 2010, a swine influenza A reassortant virus, H1N2r, which caused mild clinical disease in pigs in the United Kingdom, was isolated. This reassortant virus has a novel gene constellation, incorporating the internal gene cassette of pH1N1-origin viruses and hemagglutinin and neuraminidase genes of swine IAV H1N2 origin. We investigated the pathogenesis and infection dynamics of the H1N2r isolate in pigs (the natural host) and in ferrets, which represent a human model of infection. Clinical and virologic parameters were mild in both species and both intraspecies and interspecies transmission was observed when initiated from either infected pigs or infected ferrets. This novel reassortant virus has zoonotic and reverse zoonotic potential, but no apparent increased virulence or transmissibility, in comparison to pH1N1 viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N2 Subtype/genetics , Influenza, Human/virology , Swine Diseases/epidemiology , Animals , Ferrets , Genes, Viral , Humans , Male , Reassortant Viruses/genetics , Swine , Swine Diseases/transmission , Swine Diseases/virology , United Kingdom/epidemiology , Zoonoses
5.
J Virol Methods ; 265: 9-14, 2019 03.
Article in English | MEDLINE | ID: mdl-30579921

ABSTRACT

Newcastle disease is a devastating disease of poultry caused by Newcastle disease virus (NDV), a virulent form of avian avulavirus 1 (AAvV-1). A rapid, sensitive and specific means for the detection of NDV is fundamental for the control of this notifiable transboundary virus. Although several real-time RT-PCR assays exist for the detection of AAvV-1, diagnostic sensitivity and specificities can be sub-optimal. In this study, we describe a modification to an existing AAvV-1 l-gene RT-PCR screening assay, where the original probe set was replaced with minor groove binding (MGB) probes, to create the MGB l-gene assay. The diagnostic sensitivity and specificity of this assay was evaluated against a broad panel of both Class I and Class II AAvV-1 viruses of diverse and representative lineages/genotypes in both clinical samples and amplified viruses, and compared with a number of previously published real-time RT-PCR screening assays for AAvV-1. The MGB l-gene assay outperformed all other assays in this assessment, with enhanced sensitivity and specificity, detecting isolates from a broad range of virus lineages/genotypes (including contemporaneously-circulating strains). The assay has also proved its value for screening original clinical samples for the presence of AAvV-1, thus providing an improved screening assay for routine detection of this notifiable disease agent.


Subject(s)
Avulavirus Infections/veterinary , Avulavirus/isolation & purification , Bird Diseases/diagnosis , Bird Diseases/virology , DNA Primers/genetics , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Avulavirus/classification , Avulavirus/genetics , Avulavirus Infections/diagnosis , Avulavirus Infections/virology , Birds , Genotype , Sensitivity and Specificity
6.
Vet Res ; 45: 118, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25431115

ABSTRACT

Highly pathogenic avian influenza (HPAI) H5N1 viruses cause severe infection in chickens at near complete mortality, but corresponding infection in ducks is typically mild or asymptomatic. To understand the underlying molecular differences in host response, primary chicken and duck lung cells, infected with two HPAI H5N1 viruses and a low pathogenicity avian influenza (LPAI) H2N3 virus, were subjected to RNA expression profiling. Chicken cells but not duck cells showed highly elevated immune and pro-inflammatory responses following HPAI virus infection. HPAI H5N1 virus challenge studies in chickens and ducks corroborated the in vitro findings. To try to determine the underlying mechanisms, we investigated the role of signal transducer and activator of transcription-3 (STAT-3) in mediating pro-inflammatory response to HPAIV infection in chicken and duck cells. We found that STAT-3 expression was down-regulated in chickens but was up-regulated or unaffected in ducks in vitro and in vivo following H5N1 virus infection. Low basal STAT-3 expression in chicken cells was completely inhibited by H5N1 virus infection. By contrast, constitutively active STAT-3 detected in duck cells was unaffected by H5N1 virus infection. Transient constitutively-active STAT-3 transfection in chicken cells significantly reduced pro-inflammatory response to H5N1 virus infection; on the other hand, chemical inhibition of STAT-3 activation in duck cells increased pro-inflammatory gene expression following H5N1 virus infection. Collectively, we propose that elevated pro-inflammatory response in chickens is a major pathogenicity factor of HPAI H5N1 virus infection, mediated in part by the inhibition of STAT-3.


Subject(s)
Avian Proteins/genetics , Chickens , Ducks , Influenza A Virus, H5N1 Subtype/physiology , Influenza in Birds/immunology , Poultry Diseases/immunology , Animals , Avian Proteins/metabolism , Gene Expression , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/genetics , Influenza in Birds/virology , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis/veterinary , Poultry Diseases/genetics , Poultry Diseases/virology , Sequence Analysis, DNA/veterinary
7.
Virus Res ; 178(2): 383-91, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24050997

ABSTRACT

Pigs are thought to play a role in the adaptation of avian influenza (AI) viruses to mammalian hosts. To better understand this mechanism and to identify key mutations two highly pathogenic AI (HPAI) viruses (H5N1 and H7N7) were grown in pig cells, To mimic the pressure of an immune response, these viruses were grown in the presence of antiserum to the homologous virus or porcine IFN-γ. Mutations were identified in both viruses grown in vitro in the presence and absence of antisera or IFN-γ and included the PB2 mutations, E627K or 627E,D701N, described previously as requirements for the adaptation of AI viruses to mammalian species. Additional mutations were also identified in PB1, HA, NP and M genes for viruses passaged in the presence of immune pressure. The infectivity of these viruses was then assessed using ex vivo pig bronchi and lung organ cultures. For lung explants, higher levels of virus were detected in organ cultures infected with H5N1 HPAI viruses passaged in pig cell lines regardless of the presence or absence of homologous antisera or IFN-γ when compared with the wild-type parental viruses. No infection was observed for any of the H7N7 HPAI viruses. These results suggest that the mutations identified in H5N1 HPAI viruses may provide a replication or infection advantage in pigs in vivo and that pigs may continue to play an important role in the ecology of influenza A viruses including those of avian origin.


Subject(s)
Influenza A Virus, H5N1 Subtype/pathogenicity , Respiratory System/virology , Adaptation, Biological , Animals , DNA Mutational Analysis , Influenza A Virus, H7N7 Subtype/pathogenicity , Mutation, Missense , Organ Culture Techniques , Serial Passage , Swine , Viral Load , Viral Proteins/genetics
8.
Sci Rep ; 3: 2478, 2013.
Article in English | MEDLINE | ID: mdl-23963354

ABSTRACT

Infection of chickens with low pathogenicity avian influenza (LPAI) virus results in mild clinical signs while infection with highly pathogenic avian influenza (HPAI) viruses causes death of the birds within 36-48 hours. Since natural killer (NK) cells have been shown to play an important role in influenza-specific immunity, we hypothesise that NK cells are involved in this difference in pathogenicity. To investigate this, the role of chicken NK-cells in LPAI virus infection was studied. Next activation of lung NK cells upon HPAI virus infection was analysed. Infection with a H9N2 LPAI virus resulted in the presence of viral RNA in the lungs which coincided with enhanced activation of lung NK cells. The presence of H5N1 viruses, measured by detection of viral RNA, did not induce activation of lung NK cells. This suggests that decreased NK-cell activation may be one of the mechanisms associated with the enhanced pathogenicity of H5N1 viruses.


Subject(s)
Chickens/immunology , Chickens/virology , Influenza in Birds/immunology , Influenza in Birds/virology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Orthomyxoviridae/pathogenicity , Animals , Influenza in Birds/pathology
9.
Vet Microbiol ; 162(2-4): 944-948, 2013 Mar 23.
Article in English | MEDLINE | ID: mdl-23266109

ABSTRACT

To simulate a field situation in which pigs are in close contact with poultry and thus provide a potential mixing vessel for avian, swine and human influenza viruses, uninfected pigs were placed in contact with Pekin ducks or chickens infected with a H5N1 highly pathogenic avian influenza (HPAI) virus. To sustain prolonged exposure, newly inoculated birds were added at regular intervals. Although influenza virus was detected in birds and environmental samples, 14 days exposure to infected birds failed to produce evidence of infection in the pigs. The ability of pigs to generate reassortant viruses with these particular virus variants (H5N1 clade 2.2.1) may therefore be limited.


Subject(s)
Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/transmission , Influenza in Birds/virology , Orthomyxoviridae Infections/veterinary , Swine Diseases/transmission , Swine Diseases/virology , Animals , Chickens , Ducks , Influenza A Virus, H5N1 Subtype/classification , Influenza in Birds/pathology , Orthomyxoviridae Infections/transmission , Swine , Turkeys
10.
Influenza Other Respir Viruses ; 7(3): 393-402, 2013 May.
Article in English | MEDLINE | ID: mdl-22716314

ABSTRACT

BACKGROUND: Pigs are thought to act as intermediate hosts in the ecology of influenza viruses of both avian and human origin. The recent development of procedures for pig ex vivo respiratory organ explants has provided new tools for the assessment of influenza virus infection in pigs. OBJECTIVES: To use pig ex vivo organ explants to assess the susceptibility of pigs to infection with contemporary viruses, for which there is evidence of human infection and that are thought to pose the greatest threat to pig and human populations. METHODS: Pig tracheal, bronchi and lung ex vivo organ explants were infected with both highly pathogenic and low pathogenic avian influenza (AI) virus and the pandemic H1N1 [A(H1N1)pdm/09] virus. Successful infection of explants was detected using a positive-sense RNA real-time RT-PCR assay and anti-nucleoprotein immunohistochemistry. The distribution of cell-surface α2-3- and α2-6-linked sialic acid receptors, the avian- and mammalian influenza A virus-preferred host receptors, respectively, was also characterised for the ex vivo organ cultures and uninfected pig material following necropsy. RESULTS: The α2-3 and α2-6 sialic acid receptor staining on tracheal, bronchi and lung organ explant sections showed similar distributions to those seen for pig tissue following necropsy. While the pig ex vivo organ cultures were susceptible to nearly all viruses tested, lower levels of virus were detected in trachea and bronchi after infection. CONCLUSION: These results confirm that pigs are susceptible to contemporary viruses that may threaten both veterinary and human health and contribute to the ecology of influenza A viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/veterinary , Respiratory System/virology , Swine Diseases/virology , Animals , Female , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/physiology , Male , Organ Culture Techniques , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Pandemics , Swine , Swine Diseases/epidemiology , Virulence
11.
Avian Dis ; 54(1 Suppl): 194-200, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20521632

ABSTRACT

Since 2005 there have been five incursions into Great Britain of highly pathogenic avian influenza (HPAI) viruses of subtype H5N1 related to the ongoing global epizootic. The first incursion occurred in October 2005 in birds held in quarantine after importation from Taiwan. Two incursions related to wild birds: one involved a single dead whooper swan found in March 2006 in the sea off the east coast of Scotland, and the other involved 10 mute swans and a Canada goose found dead over the period extending from late December 2007 to late February 2008 on or close to a swannery on the south coast of England. The other two outbreaks occurred in commercial poultry in January 2007 and November 2007, both in the county of Suffolk. The first of these poultry outbreaks occurred on a large turkey farm, and there was no further spread. The second outbreak occurred on a free-range farm rearing turkeys, ducks, and geese and spread to birds on a second turkey farm that was culled as a dangerous contact. Viruses isolated from these five outbreaks were confirmed to be Asian H5N1 HPAI viruses; the quarantine outbreak was attributed to a clade 2.3 virus and the other four to clade 2.2 viruses. This article describes the outbreaks, their control, and the possible origins of the responsible viruses.


Subject(s)
Disease Outbreaks/veterinary , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza in Birds/epidemiology , Animals , Birds , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/virology , Phylogeny , Time Factors , United Kingdom/epidemiology
12.
Avian Dis ; 54(1 Suppl): 399-404, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20521669

ABSTRACT

Surveillance of wild birds for avian influenza viruses has been compulsory in the European Union (EU) since 2005, primarily as a means of detecting H5N1 highly pathogenic avian influenza (HPAI) virus and of monitoring the circulation of low pathogenicity avian influenza (LPAI) virus H5 and H7 strains. In 2007, 79,392 wild birds were tested throughout the EU. H5N1 HPAI was detected in 329 birds from four Member States (MS); affected birds were almost entirely of the orders Podicipediformes (grebes) and Anseriformes (waterfowl) during the summer months. LPAI was detected in 1485 wild birds among 21 MS. A total of 1250 birds were positive for influenza A but were not discriminated any further; LPAI H5 was detected in 105 birds, exclusively of the order Anseriformes. LPAI H7 was detected in seven birds. LPAI of other subtypes was found in 123 birds. Epidemiologic evidence and phylogenetic analysis of H5N1 viruses indicate that H5N1 did not appear to persist in the EU from 2006 but was reintroduced, probably from the Middle East.


Subject(s)
Animals, Wild , Birds , European Union , Influenza A Virus, H5N1 Subtype , Influenza in Birds/epidemiology , Animal Migration , Animals , Europe/epidemiology , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/virology , Phylogeny , Population Surveillance , Time Factors
13.
Influenza Other Respir Viruses ; 4(1): 17-25, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20021503

ABSTRACT

BACKGROUND: Highly pathogenic avian influenza (HPAI) H5N1 viruses have recently displayed increased virulence for wild waterfowl. OBJECTIVES: To study the effect of host age on the shedding and tissue dissemination of a HPAI H5N1 virus in infected Pekin ducks. METHODS: Pekin ducks in two age-matched groups (n = 18), 8 and 12 weeks old (wo) were each infected with 10(6) EID(50)/0.1 ml of HPAI A/turkey/Turkey/1/05 (H5N1, clade 2.2). Each day for 5 days, birds were monitored clinically, and cloacal and oropharyngeal swabs collected, before three birds from each group were selected randomly for post-mortem examination. Tissue samples were collected for examination by real-time RT-PCR, histopathology and immunohistochemistry (IHC). RESULTS: Severe clinical signs, including incoordination and torticollis were observed in the 8 wo group resulting in 100% mortality by 4 dpi. Mild clinical signs were observed in the 12 wo group with no mortality. Real-time RT-PCR and IHC results demonstrated the systemic spread of H5N1 virus in birds of both age groups. Higher levels of virus shedding were detected in oropharyngeal swabs than in cloacal swabs, with similar levels of shedding detected in both age groups. Variations in level and temporal dissemination of virus within tissues of older ducks, and the presence of the virus in brain and heart were observed, which coincided with the appearance of clinical signs preceding death in younger birds. CONCLUSIONS: These results are consistent with reports of natural infections of wild waterfowl and poultry possibly indicating an age-related association with dissemination and clinical outcome in ducks following infection with H5N1 HPAI virus.


Subject(s)
Ducks/virology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/virology , Age Factors , Animals , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza in Birds/pathology , Virulence , Virus Shedding
14.
Philos Trans R Soc Lond B Biol Sci ; 364(1530): 2739-47, 2009 Sep 27.
Article in English | MEDLINE | ID: mdl-19687042

ABSTRACT

The emergence and spread of H5N1 avian influenza viruses from Asia through to Europe and Africa pose a significant animal disease problem and have raised concerns that the virus may pose a pandemic threat to humans. The epizootological factors that have influenced the wide distribution of the virus are complex, and the variety of viruses currently circulating reflects these factors. Sequence analysis of the virus genes sheds light on the H5N1 virus evolution during its emergence and spread, but the degree of virus variation at the level of an individual infected bird has been described in only a few studies. Here, we describe some results of a study in which turkeys, ducks and chickens were infected with either one of two H5N1 or one of three H7N1 viruses, and the degree of sequence variation within an individual infected avian host was examined. We developed 'deep amplicon' sequence analysis for this work, and the methods and results provide a background framework for application to disease outbreaks in the field.


Subject(s)
Disease Outbreaks/veterinary , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/virology , Poultry , Zoonoses/virology , Animals , Genetic Variation , Influenza in Birds/epidemiology , RNA, Viral/chemistry , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Alignment , Sequence Analysis, DNA , Specific Pathogen-Free Organisms , Zoonoses/epidemiology
15.
Influenza Other Respir Viruses ; 3(2): 63-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19496843

ABSTRACT

OBJECTIVES: The HPAI H5N2 strain that caused an outbreak in ostriches of the Eastern Cape Province, South Africa in 2004 was characterized. DESIGN: Haemagglutination inhibition (HI) and agar gel immunodiffusion (AGID) were performed on sera from ostrich farms in the outbreak region, and intravenous pathogenicity (IVPI) tests, reverse-transcriptase-polymerase-chain reaction (RT-PCR), nucleic acid sequencing and phylogenetic comparisons were performed on the HPAI H5N2 virus isolated during the outbreak. RESULTS: The deduced amino acid sequence at the HA0 cleavage site determined by RT-PCR and nucleotide sequencing was PQREKRRKKRGLF and thus the virus fell within the definition of a highly pathogenic virus, but in an IVPI test in chickens on the virus isolated from the index case and a value of 0.63 was recorded, which is below the criterion for highly pathogenic viruses in this in vivo test. After a further passage in embryonated eggs a second IVPI was carried out and an elevated value of 1.19 was obtained. Cloacal swabs were taken from the initial IVPI birds, inoculated into embryonated chickens eggs and a third IVPI was then performed on the resulting haemagglutinating, infective allantoic fluid. An index of 2.73 was recorded. CONCLUSIONS: HI tests appeared to be the more sensitive test compared to AGID when testing for antibodies to avian influenza in sera. An ostrich-derived virus with a virulent HA0 cleavage site was not initially virulent in chickens but after passage in the latter the virulence increased. Phylogenetic analyses demonstrated the link between AI viruses carried by wild ducks and those infecting ostriches.


Subject(s)
Influenza A Virus, H5N2 Subtype/classification , Influenza A Virus, H5N2 Subtype/isolation & purification , Influenza in Birds/virology , Struthioniformes , Animals , Birds , Disease Outbreaks , Genotype , Influenza A Virus, H5N2 Subtype/genetics , Influenza A Virus, H5N2 Subtype/pathogenicity , Molecular Sequence Data , Phylogeny , South Africa , Struthioniformes/virology , Virulence
16.
Avian Pathol ; 37(6): 619-27, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19023759

ABSTRACT

Asian H5N1 (hereafter referred to as panzootic H5N1) highly pathogenic avian influenza (HPAI) virus has caused large numbers of deaths in both poultry and wild-bird populations. Recent isolates of this virus have been reported to cause disease and death in commercial ducks, which has not been seen with other HPAI viruses. However, little is known about either the dissemination of this H5N1 within the organs or the cause of death in infected ducks. Nineteen 4-week-old Pekin ducks were infected with 10(6.7) median egg infectious doses of HPAI A/turkey/Turkey/1/05 (H5N1, clade 2.2) in 0.1ml via the intranasal and intraocular routes. Cloacal and oropharyngeal swabs were taken daily before three animals were selected randomly and killed humanely for postmortem examination, when samples of tissues were taken for real-time reverse transcriptase-polymerase chain reaction, histopathological examination and immunohistochemistry. Clinical signs were first observed 4 days post infection (d.p.i.) and included depression, reluctance to feed, in-coordination and torticollis resulting in the death of all the birds remaining on 5d.p.i. Higher levels of virus shedding were detected from oropharyngeal swabs than from cloacal swabs. Real-time reverse transcriptase-polymerase chain reaction and immunohistochemistry identified peak levels of virus at 2d.p.i. in several organs. In the spleen, lung, kidney, caecal tonsils, breast muscle and thigh muscle the levels were greatly reduced at 3d.p.i. However, the highest viral loads were detected in the heart and brain from 3d.p.i. and coincided with the appearance of clinical signs and death. Our experimental results demonstrate the systemic spread of this HPAI H5N1 virus in Pekin ducks, and the localization of virus in the brain and heart tissue preceding death.


Subject(s)
Ducks , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/virology , Animals , Brain/virology , Heart/virology , Immunohistochemistry , Kidney/virology , Liver/virology , Lung/virology , Lymphoid Tissue/virology , Muscle, Skeletal/virology , Respiratory System/virology
17.
Avian Pathol ; 36(5): 347-50, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17899457

ABSTRACT

Four avian influenza viruses have been recognized that have genetic coding for highly pathogenic avian influenza viruses, but do not show virulence for chickens. The two different mechanisms that prevent this potential being expressed have been determined for A/chicken/Pennsylvania/1/83 (H5N2) and A/goose/Guandong/2/96 (H5N1), but neither of these applies to A/turkey/England/87-92BFC/91 (H5N1) or A/chicken/Texas/298313/04 (H5N2).


Subject(s)
Chickens/virology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N2 Subtype/pathogenicity , Influenza in Birds/virology , Animals , Virulence
18.
Emerg Infect Dis ; 13(4): 611-3, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17553279

ABSTRACT

Genetic analysis of highly pathogenic avian influenza (H5N1) viruses from poultry and hooded vultures in Burkina Faso shows that these viruses belong to 1 of 3 sublineages initially found in Nigeria and later in other African countries. Hooded vultures could potentially be vectors or sentinels of influenza subtype H5N1, as are cats and swans elsewhere.


Subject(s)
Animals, Wild/virology , Falconiformes/virology , Influenza in Birds/virology , Poultry/virology , Animals , Burkina Faso/epidemiology , Chickens/virology , Disease Vectors , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza in Birds/epidemiology , Phylogeny , Poultry Diseases/epidemiology , Poultry Diseases/virology , Sentinel Surveillance
19.
Avian Dis ; 51(1 Suppl): 393-5, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17494592

ABSTRACT

We previously described the use of an established reverse genetics system for the generation of recombinant human influenza A viruses from cloned cDNAs. Here, we have assembled a set of plasmids to allow recovery of the avian H5N1 influenza virus A/Turkey/England/50-92/91 entirely from cDNA. This system enables us to introduce mutations or truncations into the cDNAs to create mutant viruses altered specifically in a chosen gene. These mutant viruses can then be used in future pathogenesis studies in chickens and in studies to understand the host range restrictions of avian influenza viruses in humans.


Subject(s)
Genetic Techniques , Influenza A Virus, H5N1 Subtype/genetics , Animals , Cell Line , Cricetinae , DNA, Complementary , DNA, Viral , DNA-Directed RNA Polymerases/metabolism , Dogs , Humans
20.
Avian Pathol ; 36(3): 245-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17497339

ABSTRACT

The results of pathological, virological and polymerase chain reaction examinations carried out on 35 mute swans (Cygnus olor) that succumbed to a highly pathogenic avian influenza virus (H5N1) infection during an outbreak in Southern Hungary are reported. The most frequently observed macroscopic lesions included: haemorrhages under the epicardium, in the proventricular and duodenal mucosa and pancreas; focal necrosis in the pancreas; myocardial degeneration; acute mucous enteritis; congestion of the spleen and lung, and the accumulation of sero-mucinous exudate in the body cavity. Histopathological lesions comprised: lymphocytic meningo-encephalomyelitis accompanied by gliosis and occasional perivascular haemorrhages; multi-focal myocardial necrosis with lympho-histiocytic infiltration; pancreatitis with focal necrosis; acute desquamative mucous enteritis; lung congestion and oedema; oedema of the tracheal mucosa and, in young birds, the atrophy of the bursa of Fabricius as a result of lymphocyte depletion and apoptosis. The observed lesions and the moderate to good body conditions were compatible with findings in acute highly pathogenic avian influenza infections of other bird species reported in the literature. Skin lesions and lesions typical for infections caused by strains of lower pathogenicity (low pathogenic avian influenza virus) such as emaciation or fibrinous changes in the reproductive and respiratory organs, sinuses and airsacs were not observed. The H5N1 subtype avian influenza virus was isolated in embryonated fowl eggs from all cases and it was identified by classical and molecular virological methods.


Subject(s)
Anseriformes/virology , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/pathology , Influenza in Birds/virology , Animals , Disease Outbreaks/veterinary , Hungary/epidemiology , Influenza in Birds/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...