Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
J Virol ; 96(17): e0072322, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35975999

RESUMEN

The production of type I interferon (IFN) is the hallmark of the innate immune response. Most, if not all, mammalian viruses have a way to circumvent this response. Fundamental knowledge on viral evasion of innate immune responses may facilitate the design of novel antiviral therapies. To investigate how human metapneumovirus (HMPV) interacts with the innate immune response, recombinant viruses lacking G, short hydrophobic (SH), or M2-2 protein expression were assessed for IFN induction in A549 cells. HMPV lacking G or SH protein expression induced similarly low levels of IFN, compared to the wild-type virus, whereas HMPV lacking M2-2 expression induced significantly more IFN than the wild-type virus. However, sequence analysis of the genomes of M2-2 mutant viruses revealed large numbers of mutations throughout the genome. Over 70% of these nucleotide substitutions were A-to-G and T-to-C mutations, consistent with the properties of the adenosine deaminase acting on RNA (ADAR) protein family. Knockdown of ADAR1 by CRISPR interference confirmed the role of ADAR1 in the editing of M2-2 deletion mutant virus genomes. More importantly, Northern blot analyses revealed the presence of defective interfering RNAs (DIs) in M2-2 mutant viruses and not in the wild-type virus or G and SH deletion mutant viruses. DIs are known to be potent inducers of the IFN response. The presence of DIs in M2-2 mutant virus stocks and hypermutated virus genomes interfere with studies on HMPV and the innate immune response and should be addressed in future studies. IMPORTANCE Understanding the interaction between viruses and the innate immune response is one of the barriers to the design of antiviral therapies. Here, we investigated the role of the G, SH, and M2-2 proteins of HMPV as type I IFN antagonists. In contrast to other studies, no IFN-antagonistic functions could be observed for the G and SH proteins. HMPV with a deletion of the M2-2 protein did induce type I IFN production upon infection of airway epithelial cells. However, during generation of virus stocks, these viruses rapidly accumulated DIs, which are strong activators of the type I IFN response. Additionally, the genomes of these viruses were hypermutated, which was prevented by generating stocks in ADAR knockdown cells, confirming a role for ADAR in hypermutation of HMPV genomes or DIs. These data indicate that a role of the HMPV M2-2 protein as a bona fide IFN antagonist remains elusive.


Asunto(s)
Inmunidad Innata , Interferón Tipo I , Metapneumovirus , Proteínas Virales , Células A549 , Adenosina Desaminasa , Antivirales/metabolismo , Humanos , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/inmunología , Metapneumovirus/genética , Metapneumovirus/metabolismo , Proteínas de Unión al ARN , Proteínas Virales/genética , Proteínas Virales/metabolismo
2.
J Virol ; 93(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30700609

RESUMEN

Influenza A/H2N2 viruses caused a pandemic in 1957 and continued to circulate in humans until 1968. The antigenic evolution of A/H2N2 viruses over time and the amino acid substitutions responsible for this antigenic evolution are not known. Here, the antigenic diversity of a representative set of human A/H2N2 viruses isolated between 1957 and 1968 was characterized. The antigenic change of influenza A/H2N2 viruses during the 12 years that this virus circulated was modest. Two amino acid substitutions, T128D and N139K, located in the head domain of the H2 hemagglutinin (HA) molecule, were identified as important determinants of antigenic change during A/H2N2 virus evolution. The rate of A/H2N2 virus antigenic evolution during the 12-year period after introduction in humans was half that of A/H3N2 viruses, despite similar rates of genetic change.IMPORTANCE While influenza A viruses of subtype H2N2 were at the origin of the Asian influenza pandemic, little is known about the antigenic changes that occurred during the twelve years of circulation in humans, the role of preexisting immunity, and the evolutionary rates of the virus. In this study, the antigenic map derived from hemagglutination inhibition (HI) titers of cell-cultured virus isolates and ferret postinfection sera displayed a directional evolution of viruses away from earlier isolates. Furthermore, individual mutations in close proximity to the receptor-binding site of the HA molecule determined the antigenic reactivity, confirming that individual amino acid substitutions in A/H2N2 viruses can confer major antigenic changes. This study adds to our understanding of virus evolution with respect to antigenic variability, rates of virus evolution, and potential escape mutants of A/H2N2.


Asunto(s)
Sustitución de Aminoácidos , Antígenos Virales , Evolución Molecular , Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H2N2 del Virus de la Influenza A , Gripe Humana , Pandemias , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Perros , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H2N2 del Virus de la Influenza A/genética , Subtipo H2N2 del Virus de la Influenza A/inmunología , Gripe Humana/epidemiología , Gripe Humana/genética , Gripe Humana/inmunología , Células de Riñón Canino Madin Darby , Mutación Missense , Factores de Tiempo
3.
RNA Biol ; 14(11): 1606-1616, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28662365

RESUMEN

RNA structures are increasingly recognized to be of importance during influenza A virus replication. Here, we investigated a predicted conserved hairpin in the M gene segment (nt 967-994) within the region of the vRNA 5' packaging signal. The existence of this RNA structure and its possible role in virus replication was investigated using a compensatory mutagenesis approach. Mutations were introduced in the hairpin stem, based on natural variation. Virus replication properties were studied for the mutant viruses with disrupted and restored RNA structures. Viruses with structure-disrupting mutations had lower virus titers and a significantly reduced median plaque size when compared with the wild-type (WT) virus, while viruses with structure restoring-mutations replicated comparable to WT. Moreover, virus replication was also reduced when mutations were introduced in the hairpin loop, suggesting its involvement in RNA interactions. Northern blot and FACS experiments were performed to study differences in RNA levels as well as production of M1 and M2 proteins, expressed via alternative splicing. Stem-disruptive mutants caused lower vRNA and M2 mRNA levels and reduced M2 protein production at early time-points. When the RNA structure was restored, vRNA, M2 mRNA and M2 protein levels were increased, demonstrating a compensatory effect. Thus, this study provides evidence for functional importance of the predicted M RNA structure and suggests its role in splicing regulation.


Asunto(s)
Virus de la Influenza A/genética , ARN Mensajero/química , ARN Viral/química , Proteínas de la Matriz Viral/química , Replicación Viral , Empalme Alternativo , Animales , Emparejamiento Base , Secuencia Conservada , Perros , Células HEK293 , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/metabolismo , Secuencias Invertidas Repetidas , Células de Riñón Canino Madin Darby , Mutagénesis , Conformación de Ácido Nucleico , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Relación Estructura-Actividad , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus
4.
Antimicrob Agents Chemother ; 60(8): 4620-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27185803

RESUMEN

The clinical impact of infections with respiratory viruses belonging to the family Paramyxoviridae argues for the development of antiviral therapies with broad-spectrum activity. Favipiravir (T-705) has demonstrated potent antiviral activity against multiple RNA virus families and is presently in clinical evaluation for the treatment of influenza. Here we demonstrate in vitro activity of T-705 against the paramyxoviruses human metapneumovirus (HMPV), respiratory syncytial virus, human parainfluenza virus, measles virus, Newcastle disease virus, and avian metapneumovirus. In addition, we demonstrate activity against HMPV in hamsters. T-705 treatment inhibited replication of all paramyxoviruses tested in vitro, with 90% effective concentration (EC90) values of 8 to 40 µM. Treatment of HMPV-challenged hamsters with T-705 at 200 mg/kg of body weight/day resulted in 100% protection from infection of the lungs. In all treated and challenged animals, viral RNA remained detectable in the respiratory tract. The observation that T-705 treatment had a significant effect on infectious viral titers, with a limited effect on viral genome titers, is in agreement with its proposed mode of action of viral mutagenesis. However, next-generation sequencing of viral genomes isolated from treated and challenged hamsters did not reveal (hyper)mutation. Polymerase activity assays revealed a specific effect of T-705 on the activity of the HMPV polymerase. With the reported antiviral activity of T-705 against a broad range of RNA virus families, this small molecule is a promising broad-range antiviral drug candidate for limiting the viral burden of paramyxoviruses and for evaluation for treatment of infections with (re)emerging viruses, such as the henipaviruses.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Metapneumovirus/efectos de los fármacos , Infecciones por Paramyxoviridae/tratamiento farmacológico , Pirazinas/farmacología , Animales , Anticuerpos Antivirales/inmunología , Chlorocebus aethiops , Cricetinae , Células HEK293 , Humanos , Pulmón/virología , Mesocricetus , Virus Sincitiales Respiratorios/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
5.
Curr Top Microbiol Immunol ; 385: 137-55, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25048542

RESUMEN

Influenza A viruses cause yearly epidemics and occasional pandemics. In addition, zoonotic influenza A viruses sporadically infect humans and may cause severe respiratory disease and fatalities. Fortunately, most of these viruses do not have the ability to be efficiently spread among humans via aerosols or respiratory droplets (airborne transmission) and to subsequently cause a pandemic. However, adaptation of these zoonotic viruses to humans by mutation or reassortment with human influenza A viruses may result in airborne transmissible viruses with pandemic potential. Although our knowledge of factors that affect mammalian adaptation and transmissibility of influenza viruses is still limited, we are beginning to understand some of the biological traits that drive airborne transmission of influenza viruses among mammals. Increased understanding of the determinants and mechanisms of airborne transmission may aid in assessing the risks posed by avian influenza viruses to human health, and preparedness for such risks. This chapter summarizes recent discoveries on the genetic and phenotypic traits required for avian influenza viruses to become airborne transmissible between mammals.


Asunto(s)
Virus de la Influenza A/fisiología , Gripe Aviar/virología , Gripe Humana/transmisión , Mamíferos/virología , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , Enfermedades de los Porcinos/transmisión , Enfermedades de los Porcinos/virología , Animales , Aves , Humanos , Virus de la Influenza A/genética , Gripe Aviar/transmisión , Gripe Humana/virología , Porcinos
6.
Euro Surveill ; 20(12)2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25846491

RESUMEN

Highly pathogenic avian influenza (HPAI) A(H5N8) viruses that emerged in poultry in east Asia since 2010 spread to Europe and North America by late 2014. Despite detections in migrating birds, the role of free-living wild birds in the global dispersal of H5N8 virus is unclear. Here, wild bird sampling activities in response to the H5N8 virus outbreaks in poultry in the Netherlands are summarised along with a review on ring recoveries. HPAI H5N8 virus was detected exclusively in two samples from ducks of the Eurasian wigeon species, among 4,018 birds sampled within a three months period from mid-November 2014. The H5N8 viruses isolated from wild birds in the Netherlands were genetically closely related to and had the same gene constellation as H5N8 viruses detected elsewhere in Europe, in Asia and in North America, suggesting a common origin. Ring recoveries of migratory duck species from which H5N8 viruses have been isolated overall provide evidence for indirect migratory connections between East Asia and Western Europe and between East Asia and North America. This study is useful for better understanding the role of wild birds in the global epidemiology of H5N8 viruses. The need for sampling large numbers of wild birds for the detection of H5N8 virus and H5N8-virus-specific antibodies in a variety of species globally is highlighted, with specific emphasis in north-eastern Europe, Russia and northern China.


Asunto(s)
Animales Salvajes/virología , Aves/virología , Brotes de Enfermedades/veterinaria , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Migración Animal , Animales , Virus de la Influenza A/clasificación , Países Bajos/epidemiología , Filogenia , ARN Viral/genética , Vigilancia de Guardia , Análisis de Secuencia de ADN
7.
Eur J Clin Microbiol Infect Dis ; 33(4): 479-90, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24078062

RESUMEN

Influenza A viruses cause yearly seasonal epidemics and occasional global pandemics in humans. In the last century, four human influenza A virus pandemics have occurred. Occasionally, influenza A viruses that circulate in other species cross the species barrier and infect humans. Virus reassortment (i.e. mixing of gene segments of multiple viruses) and the accumulation of mutations contribute to the emergence of new influenza A virus variants. Fortunately, most of these variants do not have the ability to spread among humans and subsequently cause a pandemic. In this review, we focus on the threat of animal influenza A viruses which have shown the ability to infect humans. In addition, genetic factors which could alter the virulence of influenza A viruses are discussed. The identification and characterisation of these factors may provide insights into genetic traits which change virulence and help us to understand which genetic determinants are of importance for the pandemic potential of animal influenza A viruses.


Asunto(s)
Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Humanos , Virulencia
8.
J Infect ; 88(3): 106133, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38432583

RESUMEN

OBJECTIVES: To study the effect of mycophenolate mofetil (MMF) on various vaccination responses in kidney transplant recipients. METHODS: In a randomized controlled trial (EudraCT nr.: 2014-001372-66), low immunologically risk kidney transplant recipients were randomized to TAC/MMF or TAC-monotherapy (TACmono), six months post-transplantation. One year after transplantation, in a pre-specified sub-study, recipients were vaccinated against pneumococcus, tetanus and influenza. Blood was sampled before and 21 days after vaccination. Adequate vaccination responses were defined by international criteria. A post-hoc analysis was conducted on SARS-CoV-2 vaccination responses within the same cohort. RESULTS: Seventy-one recipients received pneumococcal and tetanus vaccines (TAC/MMF: n = 37, TACmono: n = 34), with 29 also vaccinated against influenza. When vaccinated, recipients were 60 (54-66) years old, with median eGFR of 54 (44-67) ml/min, tacrolimus trough levels 6.1 (5.4-7.0) ug/L in both groups and TAC/MMF daily MMF dose of 1000 (500-2000) mg. Adequate vaccination responses were: pneumococcal (TAC/MMF 43%, TACmono 74%, p = 0.016), tetanus (TAC/MMF 35%, TACmono 82%, p < 0.0001) and influenza (TAC/MMF 20%, TACmono 71%, p = 0.0092). Only 7% of TAC/MMF responded adequately to all three compared to 36% of TACmono (p = 0.080). Additionally, 40% of TAC/MMF responded inadequately to all three, whereas all TACmono patients responded adequately to at least one vaccination (p = 0.041). Lower SARS-CoV-2 vaccination antibody responses correlated with lower pneumococcal antibody vaccination responses (correlation coefficient: 0.41, p = 0.040). CONCLUSIONS: MMF on top of tacrolimus severely hampers antibody responses to a broad range of vaccinations.


Asunto(s)
Gripe Humana , Trasplante de Riñón , Tétanos , Humanos , Persona de Mediana Edad , Anciano , Ácido Micofenólico/uso terapéutico , Tacrolimus/uso terapéutico , Inmunosupresores/uso terapéutico , Gripe Humana/tratamiento farmacológico , Formación de Anticuerpos , Vacunas contra la COVID-19 , Tétanos/prevención & control , Tétanos/tratamiento farmacológico
9.
Vet Pathol ; 50(3): 548-59, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23242805

RESUMEN

Many subtypes of low-pathogenicity avian influenza (LPAI) virus circulate in wild bird reservoirs, but their prevalence may vary among species. We aimed to compare by real-time reverse-transcriptase polymerase chain reaction, virus isolation, histology, and immunohistochemistry the distribution and pathogenicity of 2 such subtypes of markedly different origins in Mallard ducks (Anas platyrhynchos): H2N3 isolated from a Mallard duck and H13N6 isolated from a Ring-billed Gull (Larus delawarensis). Following intratracheal and intraesophageal inoculation, neither virus caused detectable clinical signs, although H2N3 virus infection was associated with a significantly decreased body weight gain during the period of virus shedding. Both viruses replicated in the lungs and air sacs until approximately day 3 after inoculation and were associated with a locally extensive interstitial, exudative, and proliferative pneumonia. Subtype H2N3, but not subtype H13N6, went on to infect the epithelia of the intestinal mucosa and cloacal bursa, where it replicated without causing lesions until approximately day 5 after inoculation. Larger quantities of subtype H2N3 virus were detected in cloacal swabs than in pharyngeal swabs. The possible clinical significance of LPAI virus-associated pulmonary lesions and intestinal tract infection in ducks deserves further evaluation.


Asunto(s)
Charadriiformes/virología , Patos/virología , Virus de la Influenza A/fisiología , Gripe Aviar/virología , Replicación Viral , Sacos Aéreos/virología , Animales , Cloaca/patología , Cloaca/virología , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Inmunohistoquímica/veterinaria , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/patogenicidad , Gripe Aviar/patología , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Pulmón/patología , Pulmón/virología , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Esparcimiento de Virus
10.
Euro Surveill ; 18(16): 20460, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23611030

RESUMEN

The recently identified human infections with avian influenza A(H7N9) viruses in China raise important questions regarding possible source and risk to humans. Sequence comparison with an influenza A(H7N7) outbreak in the Netherlands in 2003 and an A(H7N1) epidemic in Italy in 1999­2000 suggests that widespread circulation of A(H7N9) viruses must have occurred in China. The emergence of human adaptation marker PB2 E627K in human A(H7N9) cases parallels that of the fatal A(H7N7) human case in the Netherlands.


Asunto(s)
Brotes de Enfermedades , Virus de la Influenza A/genética , Gripe Aviar/virología , Gripe Humana/epidemiología , Gripe Humana/virología , Análisis de Secuencia/métodos , Animales , China/epidemiología , Humanos , Subtipo H7N1 del Virus de la Influenza A/genética , Subtipo H7N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H7N7 del Virus de la Influenza A/genética , Subtipo H7N7 del Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Gripe Humana/diagnóstico , Italia/epidemiología , Países Bajos/epidemiología , Aves de Corral
11.
J Gen Virol ; 93(Pt 8): 1645-1648, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22535774

RESUMEN

Exchange of gene segments between mammalian and avian influenza A viruses may lead to the emergence of potential pandemic influenza viruses. Since co-infection of single cells with two viruses is a prerequisite for reassortment to take place, we assessed frequencies of double-infection in vitro using influenza A/H5N1 and A/H1N1 viruses expressing the reporter genes eGFP or mCherry. Double-infected A549 and Madin-Darby canine kidney cells were detected by confocal microscopy and flow cytometry.


Asunto(s)
Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Proteínas Luminiscentes/genética , Animales , Línea Celular , Línea Celular Tumoral , Perros , Citometría de Flujo , Regulación Viral de la Expresión Génica , Genes Virales , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Virus Reordenados/genética , Virus Reordenados/metabolismo , Proteína Fluorescente Roja
12.
J Virol ; 85(6): 2695-702, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21228239

RESUMEN

Infection with seasonal influenza viruses induces a certain extent of protective immunity against potentially pandemic viruses of novel subtypes, also known as heterosubtypic immunity. Here we demonstrate that infection with a recent influenza A/H3N2 virus strain induces robust protection in ferrets against infection with a highly pathogenic avian influenza virus of the H5N1 subtype. Prior H3N2 virus infection reduced H5N1 virus replication in the upper respiratory tract, as well as clinical signs, mortality, and histopathological changes associated with virus replication in the brain. This protective immunity correlated with the induction of T cells that cross-reacted with H5N1 viral antigen. We also demonstrated that prior vaccination against influenza A/H3N2 virus reduced the induction of heterosubtypic immunity otherwise induced by infection with the influenza A/H3N2 virus. The implications of these findings are discussed in the context of vaccination strategies and vaccine development aiming at the induction of immunity to pandemic influenza.


Asunto(s)
Protección Cruzada , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Encéfalo/patología , Encéfalo/virología , Modelos Animales de Enfermedad , Femenino , Hurones , Histocitoquímica , Vacunas contra la Influenza/administración & dosificación , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/virología , Sistema Respiratorio/patología , Sistema Respiratorio/virología , Análisis de Supervivencia , Linfocitos T/inmunología
13.
J Virol ; 85(22): 12057-61, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21917970

RESUMEN

The 2009 H1N1 influenza pandemic provided an opportunity to study human virus-specific T cell responses after infection with a novel influenza virus against which limited humoral immunity existed in the population. Here we describe the magnitude, kinetics, and nature of the virus-specific T cell response using intracellular gamma interferon (IFN-γ) staining and fluorochrome-labeled major histocompatibility complex (MHC) class I-peptide complexes. We demonstrate that influenza virus-infected patients develop recall T cell responses that peak within 1 week postinfection and that contract rapidly. In particular, effector cell frequencies declined rapidly postinfection in favor of relatively larger proportions of central memory cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Adolescente , Adulto , Linfocitos T CD8-positivos/química , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interferón gamma/biosíntesis , Masculino , Persona de Mediana Edad , Coloración y Etiquetado , Factores de Tiempo , Adulto Joven
14.
J Virol ; 85(23): 12742-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21937642

RESUMEN

Equine influenza virus is a major respiratory pathogen in horses, and outbreaks of disease often lead to substantial disruption to and economic losses for equestrian industries. The hemagglutinin (HA) protein is of key importance in the control of equine influenza because HA is the primary target of the protective immune response and the main component of currently licensed influenza vaccines. However, the influenza virus HA protein changes over time, a process called antigenic drift, and vaccine strains must be updated to remain effective. Antigenic drift is assessed primarily by the hemagglutination inhibition (HI) assay. We have generated HI assay data for equine influenza A (H3N8) viruses isolated between 1968 and 2007 and have used antigenic cartography to quantify antigenic differences among the isolates. The antigenic evolution of equine influenza viruses during this period was clustered: from 1968 to 1988, all isolates formed a single antigenic cluster, which then split into two cocirculating clusters in 1989, and then a third cocirculating cluster appeared in 2003. Viruses from all three clusters were isolated in 2007. In one of the three clusters, we show evidence of antigenic drift away from the vaccine strain over time. We determined that a single amino acid substitution was likely responsible for the antigenic differences among clusters.


Asunto(s)
Evolución Molecular , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H3N8 del Virus de la Influenza A/genética , Subtipo H3N8 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/virología , Sustitución de Aminoácidos , Animales , Antígenos Virales/clasificación , Antígenos Virales/inmunología , Western Blotting , Células Cultivadas , Perros , Pruebas de Inhibición de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Hemaglutininas/inmunología , Hemaglutininas/metabolismo , Caballos , Subtipo H3N8 del Virus de la Influenza A/aislamiento & purificación , Riñón/citología , Riñón/metabolismo , Riñón/virología , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Filogenia , ARN Mensajero/genética , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
15.
Nature ; 442(7098): 37, 2006 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-16823443

RESUMEN

As the avian influenza virus H5N1 swept from Asia across Russia to Europe, Nigeria was the first country in Africa to report the emergence of this highly pathogenic virus. Here we analyse H5N1 sequences in poultry from two different farms in Lagos state and find that three H5N1 lineages were independently introduced through routes that coincide with the flight paths of migratory birds, although independent trade imports cannot be excluded.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/transmisión , Gripe Aviar/virología , Aves de Corral/virología , Agricultura , Migración Animal , Animales , Asia/epidemiología , Europa (Continente)/epidemiología , Vuelo Animal , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H5N1 del Virus de la Influenza A/fisiología , Gripe Aviar/epidemiología , Nigeria/epidemiología , Filogenia , Aves de Corral/fisiología , Federación de Rusia/epidemiología
16.
Euro Surveill ; 17(40): 20290, 2012 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23078800
17.
J Infect Dis ; 204(5): 777-82, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21844304

RESUMEN

A retrospective nationwide study on the use of intravenous (IV) zanamivir in patients receiving intensive care who were pretreated with oseltamivir in the Netherlands was performed. In 6 of 13 patients with a sustained reduction of the viral load, the median time to start IV zanamivir was 9 days (range, 4-11 days) compared with 14 days (range, 6-21 days) in 7 patients without viral load reduction (P = .052). Viral load response did not influence mortality. We conclude that IV zanamivir as late add-on therapy has limited effectiveness. The effect of an immediate start with IV zanamivir monotherapy or in combination with other drugs need to be evaluated.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/tratamiento farmacológico , Zanamivir/uso terapéutico , Adolescente , Adulto , Preescolar , Enfermedad Crítica , Quimioterapia Combinada , Humanos , Lactante , Infusiones Intravenosas , Persona de Mediana Edad , Países Bajos , Oseltamivir/uso terapéutico , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento , Carga Viral , Zanamivir/administración & dosificación
18.
Vet Microbiol ; 269: 109437, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35472508

RESUMEN

Newcastle Disease virus (NDV) has shown promise as an oncolytic virus for treatment of a wide range of tumours. NDV with a multi-basic cleavage site (MBCS) in the fusion (F) protein (NDV F3aa) has increased oncolytic efficacy in several tumour models, but also increased virulence in chickens compared to non-virulent NDV F0, raising potential environmental safety issues. Previously, we generated a variant of NDV F3aa with a disrupted V protein gene and a substitution of phenylalanine to serine at position 117 of the F protein (NDV F3aa-S-STOPV). Compared to NDV F3aa this virus had decreased virulence in embryonated chicken eggs. In this study, the virulence of the virus was evaluated upon inoculation of six-week-old chickens through a natural infection route and by determination of the intracerebral pathogenicity index (ICPI). Based on these data NDV F3aa-S-STOPV classified as a non-virulent virus. Although NDV F3aa was classified as a virulent virus based on the ICPI, the virus was also less pathogenic than NDV F0 upon inoculation of six-week-old chickens. These data indicate that NDV with a MBCS is not necessarily pathogenic in chickens. In addition, these data show that F3aa-S-STOPV is safe to use in viro-immunotherapies without posing a threat for chickens upon accidental exposure.


Asunto(s)
Enfermedad de Newcastle , Enfermedades de las Aves de Corral , Animales , Pollos , Virus de la Enfermedad de Newcastle/genética , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Virulencia/genética
19.
Sci Rep ; 12(1): 11729, 2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35821511

RESUMEN

Highly pathogenic avian influenza (HPAI) viruses of the A/Goose/Guangdong/1/1996 lineage (GsGd), which threaten the health of poultry, wildlife and humans, are spreading across Asia, Europe, Africa and North America but are currently absent from South America and Oceania. In December 2021, H5N1 HPAI viruses were detected in poultry and a free-living gull in St. John's, Newfoundland and Labrador, Canada. Our phylogenetic analysis showed that these viruses were most closely related to HPAI GsGd viruses circulating in northwestern Europe in spring 2021. Our analysis of wild bird migration suggested that these viruses may have been carried across the Atlantic via Iceland, Greenland/Arctic or pelagic routes. The here documented incursion of HPAI GsGd viruses into North America raises concern for further virus spread across the Americas by wild bird migration.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Animales , Animales Salvajes , Europa (Continente)/epidemiología , Gansos , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , América del Norte/epidemiología , Filogenia , Aves de Corral
20.
J Virol ; 84(22): 11841-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20826698

RESUMEN

All lyssaviruses cause fatal encephalitis in mammals. There is sufficient antigenic variation within the genus to cause variable vaccine efficacy, but this variation is difficult to characterize quantitatively: sequence analysis cannot yet provide detailed antigenic information, and antigenic neutralization data have been refractory to high-resolution robust interpretation. Here, we address these issues by using state-of-the-art antigenic analyses to generate a high-resolution antigenic map of a global panel of 25 lyssaviruses. We compared the calculated antigenic distances with viral glycoprotein ectodomain sequence data. Although 67% of antigenic variation was predictable from the glycoprotein amino acid sequence, there are in some cases substantial differences between genetic and antigenic distances, thus highlighting the risk of inferring antigenic relationships solely from sequence data at this time. These differences included epidemiologically important antigenic differences between vaccine strains and wild-type rabies viruses. Further, we quantitatively assessed the antigenic relationships measured by using rabbit, mouse, and human sera, validating the use of nonhuman experimental animals as a model for determining antigenic variation in humans. The use of passive immune globulin is a crucial component of rabies postexposure prophylaxis, and here we also show that it is possible to predict the reactivity of immune globulin against divergent lyssaviruses.


Asunto(s)
Antígenos Virales/inmunología , Lyssavirus/inmunología , Infecciones por Rhabdoviridae/virología , Proteínas Virales/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/inmunología , Variación Antigénica , Antígenos Virales/química , Antígenos Virales/genética , Línea Celular , Cricetinae , Humanos , Lyssavirus/química , Lyssavirus/clasificación , Lyssavirus/genética , Ratones , Datos de Secuencia Molecular , Pruebas de Neutralización , Conejos , Infecciones por Rhabdoviridae/inmunología , Homología de Secuencia de Aminoácido , Proteínas Virales/química , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda