Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Microbiol ; 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849624

RESUMEN

Influenza virus infection is initiated by the attachment of the viral haemagglutinin (HA) protein to sialic acid receptors on the host cell surface. Most virus particles enter cells through clathrin-mediated endocytosis (CME). However, it is unclear how viral binding signals are transmitted through the plasma membrane triggering CME. Here we found that metabotropic glutamate receptor subtype 2 (mGluR2) and potassium calcium-activated channel subfamily M alpha 1 (KCa1.1) are involved in the initiation and completion of CME of influenza virus using an siRNA screen approach. Influenza virus HA directly interacted with mGluR2 and used it as an endocytic receptor to initiate CME. mGluR2 interacted and activated KCa1.1, leading to polymerization of F-actin, maturation of clathrin-coated pits and completion of the CME of influenza virus. Importantly, mGluR2-knockout mice were significantly more resistant to different influenza subtypes than the wild type. Therefore, blocking HA and mGluR2 interaction could be a promising host-directed antiviral strategy.

3.
Emerg Microbes Infect ; 13(1): 2284301, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37966272

RESUMEN

Due to the fact that many avian influenza viruses that kill chickens are not lethal to ducks, farmers are reluctant to use avian influenza inactivated vaccines on ducks. Large numbers of unvaccinated ducks play an important role in the transmission of avian influenza viruses from wild birds to domestic poultry, creating a substantial challenge to vaccination strategies for avian influenza control. To solve this problem, we constructed a recombinant duck enteritis virus (DEV), rDEV-dH5/H7, using a live attenuated DEV vaccine strain (vDEV) as a vector. rDEV-dH5/H7 carries the hemagglutinin gene of two H5 viruses [GZ/S4184/17 (H5N6) (clade 2.3.4.4 h) and LN/SD007/17 (H5N1) (clade 2.3.2.1d)] and an H7 virus [GX/SD098/17 (H7N9)]. These three hemagglutinin genes were stably inherited in rDEV-dH5/H7 and expressed in rDEV-dH5/H7-infected cells. Animal studies revealed that rDEV-dH5/H7 and vDEV induced similar neutralizing antibody responses and protection against lethal DEV challenge. Importantly, rDEV-dH5/H7 induced strong and long-lasting hemagglutinin inhibition antibodies against different H5 and H7 viruses and provided complete protection against challenges with homologous and heterologous highly pathogenic H5 and H7 influenza viruses in ducks. Our study shows that rDEV-dH5/H7 could serve as an ideal live attenuated vaccine to protect ducks against infection with lethal DEV and highly pathogenic avian influenza viruses.


Asunto(s)
Enteritis , Subtipo H5N1 del Virus de la Influenza A , Subtipo H7N9 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Aviar , Animales , Patos , Hemaglutininas , Pollos , Subtipo H5N1 del Virus de la Influenza A/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H7N9 del Virus de la Influenza A/genética , Vacunas contra la Influenza/genética , Vectores Genéticos
4.
Front Immunol ; 14: 1086297, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36875062

RESUMEN

The conserved protective epitopes of hemagglutinin (HA) are essential to the design of a universal influenza vaccine and new targeted therapeutic agents. Over the last 15 years, numerous broadly neutralizing antibodies (bnAbs) targeting the HA of influenza A viruses have been isolated from B lymphocytes of human donors and mouse models, and their binding epitopes identified. This work has brought new perspectives for identifying conserved protective epitopes of HA. In this review, we succinctly analyzed and summarized the antigenic epitopes and functions of more than 70 kinds of bnAb. The highly conserved protective epitopes are concentrated on five regions of HA: the hydrophobic groove, the receptor-binding site, the occluded epitope region of the HA monomers interface, the fusion peptide region, and the vestigial esterase subdomain. Our analysis clarifies the distribution of the conserved protective epitope regions on HA and provides distinct targets for the design of novel vaccines and therapeutics to combat influenza A virus infection.


Asunto(s)
Hemaglutininas , Virus de la Influenza A , Humanos , Animales , Ratones , Epítopos , Dominios Proteicos , Linfocitos B , Anticuerpos ampliamente neutralizantes
5.
Transbound Emerg Dis ; 69(4): 2156-2172, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34192815

RESUMEN

H6 avian influenza virus (AIV) is one of the most prevalent AIV subtypes in the world. Our previous studies have demonstrated that H6 AIVs isolated from live poultry markets pose a potential threat to human health. In recent years, increasing number of H6 AIVs has been constantly isolated from poultry farms. In order to understand the biological characteristics of H6 AIVs in the context of farms, here, we analyzed the phylogenetic relationships, antigenicity, replication in mice and receptor binding properties of H6 AIVs isolated from farms in China between 2014 and 2018. Phylogenetic analysis showed that 19 different genotypes were formed among 20 representative H6 viruses. Notably, the internal genes of these H6 viruses exhibited complicated relationships with different subtypes of AIVs worldwide, indicating that these viruses are the products of complex and frequent reassortment events. Antigenic analysis revealed that 13 viruses tested were divided into three antigenic groups. 10 viruses examined could all replicate in the respiratory organs of infected mice without prior adaptation. Receptor binding analysis demonstrated that some of the H6 AIVs bound to both α-2, 3-linked glycans (avian-type receptor) and α-2, 6-linked glycans (human-type receptor), thereby posing a potential threat to human health. Together, these findings revealed the prevalence, complicated genetic evolution, diverse antigenicity, and dual receptor binding specificity of H6 AIVs in the settings of poultry farms, which emphasize the importance to continuously monitor the evolution and biological properties of H6 AIVs in nature.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Enfermedades de los Roedores , Animales , China/epidemiología , Granjas , Humanos , Gripe Aviar/epidemiología , Ratones , Filogenia , Aves de Corral
6.
mBio ; 12(4): e0101721, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34281396

RESUMEN

Virus-host interactions are complicated processes, and multiple cellular proteins promote or inhibit viral replication through different mechanisms. Recent progress has implicated circular RNAs (circRNAs) in cancer biology and progression; however, the role of circRNAs in viral infection remains largely unclear. Here, we detected 11,620 circRNAs in A549 cells and found that 411 of them were differentially expressed in influenza virus-infected A549 cells. We characterized a novel intronic circRNA, AIVR, that was upregulated in influenza virus-infected A549 cells and found that silencing of AIVR significantly promoted influenza virus replication in A549 cells. We further found that AIVR predominantly localizes in the cytoplasm and works as a microRNA (miRNA) sponge. One of the miRNAs absorbed by AIVR binds the mRNA of CREBBP, which is an important component of the large nucleoprotein complex interferon beta (IFN-ß) enhanceosome that accelerates IFN-ß production. AIVR overexpression significantly increased the mRNA and protein levels of IFN-ß in the influenza virus-infected A549 cells. Therefore, the upregulation of AIVR is a cellular antiviral strategy, with AIVR exerting its antiviral effect by absorbing miRNA and promoting the expression of CREBBP to facilitate IFN-ß production. Our study provides new insights into the roles of circRNAs in the cellular innate antiviral response. IMPORTANCE Circular RNAs (circRNAs) are new members of the long noncoding RNA families and have been identified in a variety of organisms, including plants, animals, and humans. Accumulating data indicate that circRNAs perform multiple functions in a variety of cellular processes associated with human diseases, such as Alzheimer's disease and cancer; however, the roles of circRNAs in virus infection have been largely uninvestigated. In this study, we investigated the cellular circRNA response upon influenza virus infection and found that 411 circRNAs were differentially expressed in the virus-infected cells. We identified a novel human intronic circRNA (we named AIVR) that antagonizes influenza virus replication. Upregulated circRNA AIVR absorbs an miRNA that binds the mRNA of CREBBP, leading to an increase in the cellular expression of CREBBP and then accelerating IFN-ß production. This study advances the understanding of the roles of circRNAs in the cellular innate antiviral response.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Expresión Génica , Interacciones Microbiota-Huesped/genética , Interferón beta/inmunología , MicroARNs/metabolismo , ARN Circular/genética , Replicación Viral/genética , Células A549 , Proteína de Unión a CREB/genética , Células HEK293 , Interacciones Microbiota-Huesped/inmunología , Humanos , Interferón beta/genética , MicroARNs/genética
7.
Emerg Microbes Infect ; 9(1): 1793-1803, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32686602

RESUMEN

Lethal infection of wild birds with different subtypes of H5 viruses continuously occur. To investigate the genetic evolution and pathogenicity of H5 viruses in wild birds, we performed a detailed genetic and biologic analysis of 27 viruses, including H5N1, H5N2, H5N6, and H5N8 subtypes, that were responsible for avian influenza outbreaks in wild birds in China over the past decade. We found that these 27 viruses, bearing different clades/subclades of HA, were complicated reassortants and formed 12 different genotypes. Ten of the viruses tested were highly pathogenic in chickens, but showed distinct pathotypes in ducks and mice. Five of these 10 viruses, which were all from clade2.3.4.4, could bind human-type receptors. Our findings reveal the diversity of the genetic and biologic properties of H5 viruses circulating in wild birds and highlight the need to carefully monitor and evaluate the risks these viruses pose to animal and public health.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H5N2 del Virus de la Influenza A/aislamiento & purificación , Subtipo H5N8 del Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/epidemiología , Animales , Animales Salvajes/virología , Embrión de Pollo , Pollos/virología , China/epidemiología , Patos/virología , Evolución Molecular , Femenino , Genotipo , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/genética , Subtipo H5N8 del Virus de la Influenza A/genética , Ratones , Ratones Endogámicos BALB C , Virus Reordenados/genética
8.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31694949

RESUMEN

Influenza A virus (IAV) coopts numerous host factors to complete its replication cycle. Here, we identify free fatty acid receptor 2 (FFAR2) as a cofactor for IAV entry into host cells. We found that downregulation of FFAR2 or Ffar2 expression significantly reduced the replication of IAV in A549 or RAW 264.7 cells. The treatment of A549 cells with small interfering RNA (siRNA) targeting FFAR2 or the FFAR2 pathway agonists 2-(4-chlorophenyl)-3-methyl-N-(thiazol-2-yl)butanamide (4-CMTB) and compound 58 (Cmp58) [(S)-2-(4-chlorophenyl)-3,3-dimethyl-N-(5-phenylthiazol-2-yl)butanamide] dramatically inhibited the nuclear accumulation of viral nucleoprotein (NP) at early time points postinfection, indicating that FFAR2 functions in the early stage of the IAV replication cycle. FFAR2 downregulation had no effect on the expression of sialic acid (SA) receptors on the cell membrane, the attachment of IAV to the SA receptors, or the activity of the viral ribonucleoprotein (vRNP) complex. Rather, the amount of internalized IAVs was significantly reduced in FFAR2-knocked-down or 4-CMTB- or Cmp58-treated A549 cells. Further studies showed that FFAR2 associated with ß-arrestin1 and that ß-arrestin1 interacted with the ß2-subunit of the AP-2 complex (AP2B1), the essential adaptor of the clathrin-mediated endocytosis pathway. Notably, siRNA knockdown of either ß-arrestin1 or AP2B1 dramatically impaired IAV replication, and AP2B1 knockdown or treatment with Barbadin, an inhibitor targeting the ß-arrestin1/AP2B1 complex, remarkably decreased the amount of internalized IAVs. Moreover, we found that FFAR2 interacted with three G protein-coupled receptor (GPCR) kinases (i.e., GRK2, GRK5, and GRK6) whose downregulation inhibited IAV replication. Together, our findings demonstrate that the FFAR2 signaling cascade is important for the efficient endocytosis of IAV into host cells.IMPORTANCE To complete its replication cycle, IAV hijacks the host endocytosis machinery to invade cells. However, the underlying mechanisms of how IAV is internalized into host cells remain poorly understood, emphasizing the need to elucidate the role of host factors in IAV entry into cells. In this study, we identified FFAR2 as an important host factor for the efficient replication of both low-pathogenic and highly pathogenic IAV. We revealed that FFAR2 facilitates the internalization of IAV into target cells during the early stage of infection. Upon further characterization of the role of FFAR2-associated proteins in virus replication, we found that the FFAR2-ß-arrestin1-AP2B1 signaling cascade is important for the efficient endocytosis of IAV. Our findings thus further our understanding of the biological details of IAV entry into host cells and establish FFAR2 as a potential target for antiviral drug development.


Asunto(s)
Endocitosis , Virus de la Influenza A/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Internalización del Virus , Células A549 , Subunidades beta de Complejo de Proteína Adaptadora/genética , Subunidades beta de Complejo de Proteína Adaptadora/metabolismo , Animales , Perros , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Ratones , Células RAW 264.7 , Receptores Acoplados a Proteínas G/genética , Replicación Viral/fisiología , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo
9.
Vaccine ; 37(40): 5925-5929, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31471151

RESUMEN

Ducks play a key role in the maintenance and spread of avian influenza viruses (AIVs) in nature, and control of AIVs in ducks has important implications for AIV eradication from poultry. We previously constructed a recombinant duck enteritis virus (DEV), rDEVus78HA, that expresses the HA gene of an H5N1 AIV and showed that rDEVus78HA immunization provides complete protection against both DEV and H5N1 AIV challenge in specific-pathogen-free ducks. In this study, we performed a 60-week clinical trial and found that this rDEVus78HA vaccine can function as a bivalent vaccine in farmed ducks against lethal challenge with DEV and H5N1 virus. Moreover, we found that rDEVus78HA-vaccinated ducks were efficiently protected against challenges with recently isolated heterologous H5N6 and H5N8 viruses. Our results demonstrate that rDEVus78HA could be extremely valuable for the control of DEV and H5 AIVs in ducks.


Asunto(s)
Patos/inmunología , Enteritis/inmunología , Vectores Genéticos/inmunología , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Enfermedades de las Aves de Corral/inmunología , Animales , Patos/virología , Enteritis/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Vacunación/métodos , Vacunas Sintéticas/inmunología
10.
mBio ; 10(3)2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31213560

RESUMEN

Avian influenza viruses (AIVs) must acquire mammalian-adaptive mutations before they can efficiently replicate in and transmit among humans. The PB2 E627K mutation is known to play a prominent role in the mammalian adaptation of AIVs. The H7N9 AIVs that emerged in 2013 in China easily acquired the PB2 E627K mutation upon replication in humans. Here, we generate a series of reassortant or mutant H7N9 AIVs and test them in mice. We show that the low polymerase activity attributed to the viral PA protein is the intrinsic driving force behind the emergence of PB2 E627K during H7N9 AIV replication in mice. Four residues in the N-terminal region of PA are critical in mediating the PB2 E627K acquisition. Notably, due to the identity of viral PA protein, the polymerase activity and growth of H7N9 AIV are highly sensitive to changes in expression levels of human ANP32A protein. Furthermore, the impaired viral polymerase activity of H7N9 AIV caused by the depletion of ANP32A led to reduced virus replication in Anp32a-/- mice, abolishing the acquisition of the PB2 E627K mutation and instead driving the virus to acquire the alternative PB2 D701N mutation. Taken together, our findings show that the emergence of the PB2 E627K mutation of H7N9 AIV is driven by the intrinsic low polymerase activity conferred by the viral PA protein, which also involves the engagement of mammalian ANP32A.IMPORTANCE The emergence of the PB2 E627K substitution is critical in the mammalian adaptation and pathogenesis of AIV. H7N9 AIVs that emerged in 2013 possess a prominent ability in gaining the PB2 E627K mutation in humans. Here, we demonstrate that the acquisition of the H7N9 PB2 E627K mutation is driven by the low polymerase activity conferred by the viral PA protein in human cells, and four PA residues are collectively involved in this process. Notably, the H7N9 PA protein leads to significant dependence of viral polymerase function on human ANP32A protein, and Anp32a knockout abolishes PB2 E627K acquisition in mice. These findings reveal that viral PA and host ANP32A are crucial for the emergence of PB2 E627K during adaptation of H7N9 AIVs to humans.


Asunto(s)
ADN Polimerasa Dirigida por ADN/metabolismo , Subtipo H7N9 del Virus de la Influenza A/genética , ARN Polimerasa Dependiente del ARN/genética , Virus Reordenados/genética , Proteínas Virales/genética , Animales , Pollos , China , Femenino , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Subtipo H7N9 del Virus de la Influenza A/enzimología , Gripe Aviar/virología , Gripe Humana/virología , Ratones , Mutación , Proteínas Nucleares/metabolismo , Infecciones por Orthomyxoviridae/virología , Proteínas de Unión al ARN/metabolismo , Replicación Viral
11.
Vet Microbiol ; 232: 146-150, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31030839

RESUMEN

Newcastle disease virus (NDV) is a major threat to poultry worldwide. Virulent Newcastle disease virus infection can cause 100% morbidity and mortality in chickens. Vaccination is the most effective way to prevent and control NDV outbreaks in poultry. Previously, we demonstrated that a duck enteritis virus (DEV) vaccine strain is a promising vector to generate recombinant vaccines in chickens. Here, we constructed two recombinant DEVs expressing the F protein (rDEV-F) or HN protein (rDEV-HN) of NDV. We then evaluated the protective efficacy of these recombinant DEVs in specific-pathogen-free chickens. rDEV-F induced 100% protection of chickens from lethal NDV challenge after a single dose of 104 TCID50, whereas rDEV-HN did not induce effective protection. rDEV-F may therefore serve as a promising vaccine candidate for chickens. This is the first report of a DEV-vectored vaccine providing robust protection against lethal NDV infection in chickens.


Asunto(s)
Mardivirus/genética , Enfermedad de Newcastle/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales , Pollos/inmunología , Pollos/virología , Patos/virología , Proteína HN/genética , Proteína HN/inmunología , Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/inmunología , Enfermedades de las Aves de Corral/virología , Organismos Libres de Patógenos Específicos , Vacunación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas Virales de Fusión/genética , Vacunas Virales/administración & dosificación
12.
Cell Host Microbe ; 24(4): 558-568.e7, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30269969

RESUMEN

H7N9 low pathogenic influenza viruses emerged in China in 2013 and mutated to highly pathogenic strains in 2017, resulting in human infections and disease in chickens. To control spread, a bivalent H5/H7 inactivated vaccine was introduced in poultry in September 2017. To monitor virus evolution and vaccine efficacy, we collected 53,884 poultry samples across China from February 2017 to January 2018. We isolated 252 H7N9 low pathogenic viruses, 69 H7N9 highly pathogenic viruses, and one H7N2 highly pathogenic virus, of which two low pathogenic and 14 highly pathogenic strains were collected after vaccine introduction. Genetic analysis of highly pathogenic strains revealed nine genotypes, one of which is predominant and widespread and contains strains exhibiting high virulence in mice. Additionally, some H7N9 and H7N2 viruses carrying duck virus genes are lethal in ducks. Thus, although vaccination reduced H7N9 infections, the increased virulence and expanded host range to ducks pose new challenges.


Asunto(s)
Enfermedades Transmisibles Emergentes/virología , Evolución Molecular , Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Animales , Pollos , China , Enfermedades Transmisibles Emergentes/sangre , Enfermedades Transmisibles Emergentes/inmunología , Enfermedades Transmisibles Emergentes/mortalidad , Patos , Femenino , Humanos , Subtipo H7N9 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/sangre , Gripe Aviar/inmunología , Gripe Aviar/mortalidad , Gripe Humana/sangre , Gripe Humana/inmunología , Gripe Humana/mortalidad , Ratones , Ratones Endogámicos BALB C , Organismos Libres de Patógenos Específicos , Vacunas de Productos Inactivados/inmunología , Virulencia/genética
13.
PLoS Pathog ; 14(1): e1006851, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29352288

RESUMEN

Transcription and replication of the influenza A virus (IAV) genome occur in the nucleus of infected cells and are carried out by the viral ribonucleoprotein complex (vRNP). As a major component of the vRNP complex, the viral nucleoprotein (NP) mediates the nuclear import of the vRNP complex via its nuclear localization signals (NLSs). Clearly, an effective way for the host to antagonize IAV infection would be by targeting vRNP nuclear import. Here, we identified phospholipid scramblase 1 (PLSCR1) as a binding partner of NP by using a yeast two-hybrid (Y2H) screen. The interaction between NP and PLSCR1 in mammalian cells was demonstrated by using co-immunoprecipitation and pull-down assays. We found that the stable overexpression of PLSCR1 suppressed the nuclear import of NP, hindered the virus life cycle, and significantly inhibited the replication of various influenza subtypes. In contrast, siRNA knockdown or CRISPR/Cas9 knockout of PLSCR1 increased virus propagation. Further analysis indicated that the inhibitory effect of PLSCR1 on the nuclear import of NP was not caused by affecting the phosphorylation status of NP or by stimulating the interferon (IFN) pathways. Instead, PLSCR1 was found to form a trimeric complex with NP and members of the importin α family, which inhibited the incorporation of importin ß, a key mediator of the classical nuclear import pathway, into the complex, thus impairing the nuclear import of NP and suppressing virus replication. Our results demonstrate that PLSCR1 negatively regulates virus replication by interacting with NP in the cytoplasm and preventing its nuclear import.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas del Núcleo Viral/metabolismo , Replicación Viral , Células A549 , Transporte Activo de Núcleo Celular , Animales , Células Cultivadas , Perros , Regulación hacia Abajo , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Proteínas de la Nucleocápside , Unión Proteica , Transporte de Proteínas
14.
Cell Res ; 27(12): 1409-1421, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29151586

RESUMEN

Certain low pathogenic avian influenza viruses can mutate to highly pathogenic viruses when they circulate in domestic poultry, at which point they can cause devastating poultry diseases and severe economic damage. The H7N9 influenza viruses that emerged in 2013 in China had caused severe human infections and deaths. However, these viruses were nonlethal in poultry. It is unknown whether the H7N9 viruses can acquire additional mutations during their circulation in nature and become lethal to poultry and more dangerous for humans. Here, we evaluated the evolution of H7N9 viruses isolated from avian species between 2013 and 2017 in China and found 23 different genotypes, 7 of which were detected only in ducks and were genetically distinct from the other 16 genotypes that evolved from the 2013 H7N9 viruses. Importantly, some H7N9 viruses obtained an insertion of four amino acids in their hemagglutinin (HA) cleavage site and were lethal in chickens. The index strain was not lethal in mice or ferrets, but readily obtained the 627K or 701N mutation in its PB2 segment upon replication in ferrets, causing it to become highly lethal in mice and ferrets and to be transmitted efficiently in ferrets by respiratory droplet. H7N9 viruses bearing the HA insertion and PB2 627K mutation have been detected in humans in China. Our study indicates that the new H7N9 mutants are lethal to chickens and pose an increased threat to human health, and thus highlights the need to control and eradicate the H7N9 viruses to prevent a possible pandemic.


Asunto(s)
Pollos/virología , Subtipo H7N9 del Virus de la Influenza A/genética , Mutación , Virulencia/genética , Animales , China , Humanos
15.
J Virol ; 90(21): 9797-9805, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27558424

RESUMEN

The H5N1 avian influenza viruses emerged in Southeast Asia in the late 20th century and have evolved into multiple phylogenetic clades based on their hemagglutinin (HA)-encoding genes. The clade 7.2 viruses were first detected in chickens in northern China in 2006, and vaccines specifically targeted to the clade were developed and have been used in poultry in China since 2006. During routine surveillance and disease diagnosis, we isolated seven H5 viruses between 2011 and 2014 that bear the clade 7.2 HA genes. Here, we performed extensive studies to understand how the clade 7.2 H5 viruses have evolved in chickens in China. Full genome sequence analysis revealed that the seven viruses formed two subtypes (four H5N1 viruses and three H5N2 viruses) and four genotypes by deriving genes from other influenza viruses. All of the viruses had antigenically drifted from the clade 7.2 viruses that were isolated in 2006. Pathogenicity studies of four viruses, one from each genotype, revealed that all of the viruses are highly pathogenic in chickens, but none of them could replicate in ducks. The four viruses exclusively bound to avian-type receptors and replicated only in the turbinates and/or lungs of mice; none of them were lethal to mice at a dosage of 106 50% egg infective doses (EID50). Our study indicates that although the clade 7.2 viruses have not been eradicated from poultry through vaccination, they have not become more dangerous to other animals (e.g., ducks and mice) and humans. IMPORTANCE: Animal influenza viruses can acquire the ability to infect and kill humans. The H5N1 viruses have been a concern in recent decades because of their clear pandemic potential. We sorted H5N1 influenza viruses into different phylogenetic clades based on their HA genes. The clade 7.2 viruses were detected in chickens in several provinces of northern China in 2006. Vaccines for these viruses were subsequently developed and have been used ever since to control infection of poultry. Here, we analyzed the genetic and biologic properties of seven clade 7.2 viruses that were isolated from chickens between 2011 and 2014. We found that after nearly 9 years of circulation in chickens, the clade 7.2 viruses still exclusively bind to avian-type receptors and are of low pathogenicity to mice, suggesting that these H5 viruses pose a low risk to human public health.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Gripe Aviar/virología , Animales , Pollos , China/epidemiología , Patos/virología , Genoma Viral/genética , Genotipo , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Filogenia , Aves de Corral , Vacunación/métodos
16.
Avian Dis ; 60(1 Suppl): 238-40, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27309061

RESUMEN

An inactivated H5N1 avian influenza (AI) vaccine (Re-6) that bears the HA and NA genes from a clade 2.3.2.1 H5N1 virus, A/duck/Guangdong/S1322/10 (DK/GD/S1322/10), has been used in domestic poultry in China and other Southeast Asian countries to control clade 2.3.2.1 H5N1viruses since 2012. The efficacy of this vaccine against H5N1 viruses isolated in recent years has not been reported. In this study, we evaluated the protection efficacy of the Re-6 vaccine in chickens against challenge with four clade 2.3.2.1 H5N1 viruses, one clade 2.3.4.4 H5N1 virus, and one clade 7.2 H5N1 virus; these viruses were isolated in mainland China, Hong Kong, and the Democratic People's Republic of Korea between 2011 and 2015. The vaccinated chickens were completely protected (no disease signs, virus shedding, or death) from the challenge with the four clade 2.3.2.1 H5N1 viruses. In the clade 7.2 virus-challenged group, all of the vaccinated chickens remained healthy and survived for the entire 2-wk observation period; virus shedding was only detected from 1 of 10 chickens on day 3 postchallenge. In the clade 2.3.4.4 virus-challenged group, 8 of the 10 vaccinated chickens remained healthy and survived the 2-wk observation period; however, virus shedding was detected from 8 of 10 chickens on day 5 postchallenge. These results indicate that the Re-6 vaccine provides solid protection against clade 2.3.2.1, good protection against clade 7.2, and poor protection against clade 2.3.4.4.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Animales , Pollos , China , República Popular Democrática de Corea , Patos , Subtipo H5N1 del Virus de la Influenza A/clasificación , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/fisiología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Gripe Aviar/inmunología , Gripe Aviar/virología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Esparcimiento de Virus
17.
Avian Dis ; 60(1 Suppl): 253-5, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27309064

RESUMEN

The Goose/Guangdong-lineage H5 viruses have evolved into diverse clades and subclades based on their hemagglutinin (HA) gene during their circulation in wild birds and poultry. Since late 2013, the clade 2.3.4.4 viruses have become widespread in poultry and wild bird populations around the world. Different subtypes of the clade 2.3.4.4 H5 viruses, including H5N1, H5N2, H5N6, and H5N8, have caused vast disease outbreaks in poultry in Asia, Europe, and North America. In this study, we developed a new H5N1 inactivated vaccine by using a seed virus (designated as Re-8) that contains the HA and NA genes from a clade 2.3.4.4 virus, A/chicken/Guizhou/4/13(H5N1) (CK/GZ/4/13), and its six internal genes from the high-growth A/Puerto Rico/8/1934 (H1N1) virus. We evaluated the protective efficacy of this vaccine in chickens challenged with one H5N1 clade 2.3.2.1b virus and six different subtypes of clade 2.3.4.4 viruses, including H5N1, H5N2, H5N6, and H5N8 strains. In the clade 2.3.2.1b virus DK/GX/S1017/13-challenged groups, half of the vaccinated chickens shed virus through the oropharynx and two birds (20%) died during the observation period. All of the control chickens shed viruses and died within 6 days of infection with challenge virus. All of the vaccinated chickens remained healthy following challenge with the six clade 2.3.4.4 viruses, and virus shedding was not detected from any of these birds; however, all of the control birds shed viruses and died within 4 days of challenge with the clade 2.3.4.4 viruses. Our results indicate that the Re-8 vaccine provides protection against different subtypes of clade 2.3.4.4 H5 viruses.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Animales , Pollos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/genética , Subtipo H5N8 del Virus de la Influenza A/genética , Subtipo H5N8 del Virus de la Influenza A/inmunología , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Gripe Aviar/inmunología , Gripe Aviar/virología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Vacunación , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/genética , Vacunas de Productos Inactivados/inmunología
18.
Sci Rep ; 5: 11233, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26058711

RESUMEN

The continued spread of the newly emerged H7N9 viruses among poultry in China, together with the emergence of drug-resistant variants and the possibility of human-to-human transmission, has spurred attempts to develop an effective vaccine. An MF59-adjuvant H7N9 inactivated vaccine is reported to be well-tolerated and immunogenic in humans; however a study in ferrets indicated that while a single dose of the inactivated H7N9 vaccine reduced disease severity, it did not prevent virus replication and transmission. In this study, we used reverse genetics to produce a cold-adapted, live attenuated H7N9 vaccine (H7N9/AAca) that contains wild-type HA and NA genes from AH/1, and the backbone of the cold-adapted influenza H2N2 A/Ann Arbor/6/60 virus (AAca). H7N9/AAca was attenuated in mice and ferrets, and induced robust neutralizing antibody responses in rhesus mice, ferrets, and guinea pigs immunized once or twice intranasally. The animals immunized twice were completely protected from H7N9 virus challenge. Importantly, the animals vaccinated once were fully protected from transmission when exposed to or in contact with the H7N9 virus-inoculated animals. These results demonstrate that a cold-adapted H7N9 vaccine can prevent H7N9 virus transmission; they provide a compelling argument for further testing of this vaccine in human trials.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/fisiología , Vacunas Atenuadas/inmunología , Replicación Viral/inmunología , Animales , Subtipo H7N9 del Virus de la Influenza A/inmunología
19.
J Vet Med Sci ; 77(4): 413-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25650059

RESUMEN

Avian influenza (AI) is an infectious disease caused by avian influenza viruses (AIVs) which belong to the influenza virus A group. AI causes tremendous economic losses in poultry industry and pose great threatens to human health. Active serologic surveillance is necessary to prevent and control the spread of AI. In this study, a protein microarray using nucleoprotein (NP) of H5N1 AIV expressed in insect cells was developed to detect antibodies against AIV NP protein. The protein microarray was used to test Newcastle disease virus (NDV), infectious bursal disease virus (IBDV), AIV positive and negative sera. The results indicated that the protein microarray could hybridize specifically with antibodies against AIV with strong signals and without cross-hybridization. Moreover, 76 field serum samples were detected by microarray, enzyme-linked immunosorbent assay (ELISA) and hemagglutination inhibition test (HI). The positive rate was 92.1% (70/76), 93.4% (71/76) and 89.4% (68/76) by protein microarray, ELISA and HI test, respectively. Compared with ELISA, the microarray showed 100% (20/20) agreement ratio in chicken and 98.2% (55/56) in ornamental bird. In conclusion, this method provides an alternative serological diagnosis for influenza antibody screening and will provide a basis for the development of protein microarrays that can be used to respectively detect antibodies of different AIV subtypes and other pathogens.


Asunto(s)
Anticuerpos Antivirales/sangre , Virus de la Influenza A/inmunología , Gripe Aviar/diagnóstico , Nucleoproteínas/inmunología , Nucleoproteínas/metabolismo , Análisis por Matrices de Proteínas/veterinaria , Animales , Antígenos Virales , Línea Celular , Pollos , Clonación Molecular , Ensayo de Inmunoadsorción Enzimática , Regulación Viral de la Expresión Génica , Gripe Aviar/virología , Mariposas Nocturnas , Sensibilidad y Especificidad
20.
Biomed Res Int ; 2014: 703930, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25215289

RESUMEN

In 2010, a novel Tembusu virus (TMUV) that caused a severe decrease in the egg production of ducks was isolated in southeast China. Given the novelty of this duck pathogen, little information is available regarding its pathogenesis. Here, we systematically investigated the replication kinetics of TMUV PTD2010 in adult male and female ducks. We found that PTD2010 was detectable in most of the parenchymatous organs as well as the oviduct and intestinal tract from days 1 to 7 after inoculation. Viral titers were maintained at high levels for at least 9 days in the spleen, kidney, bursa of Fabricius, brain, and ovary. No virus was detected in any of these organs or tissues at 18 days after inoculation. PTD2010, thus, causes systemic infections in male and female ducks; its replication kinetics show similar patterns in most organs, with the exception of the ovaries and testes.


Asunto(s)
Patos/virología , Flavivirus , Enfermedades de las Aves de Corral/virología , Tropismo Viral , Animales , Femenino , Intestinos/patología , Masculino , Ovario/patología , Enfermedades de las Aves de Corral/patología , Testículo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...