Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Pathog ; 18(4): e1010446, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35377920

RESUMEN

Host defense systems employ posttranslational modifications to protect against invading pathogens. Here, we found that protein inhibitor of activated STAT 1 (PIAS1) interacts with the nucleoprotein (NP), polymerase basic protein 1 (PB1), and polymerase basic protein 2 (PB2) of influenza A virus (IAV). Lentiviral-mediated stable overexpression of PIAS1 dramatically suppressed the replication of IAV, whereas siRNA knockdown or CRISPR/Cas9 knockout of PIAS1 expression significantly increased virus growth. The expression of PIAS1 was significantly induced upon IAV infection in both cell culture and mice, and PIAS1 was involved in the overall increase in cellular SUMOylation induced by IAV infection. We found that PIAS1 inhibited the activity of the viral RNP complex, whereas the C351S or W372A mutant of PIAS1, which lacks the SUMO E3 ligase activity, lost the ability to suppress the activity of the viral RNP complex. Notably, the SUMO E3 ligase activity of PIAS1 catalyzed robust SUMOylation of PB2, but had no role in PB1 SUMOylation and a minimal role in NP SUMOylation. Moreover, PIAS1-mediated SUMOylation remarkably reduced the stability of IAV PB2. When tested in vivo, we found that the downregulation of Pias1 expression in mice enhanced the growth and virulence of IAV. Together, our findings define PIAS1 as a restriction factor for the replication and pathogenesis of IAV.


Asunto(s)
Virus de la Influenza A , Proteínas Inhibidoras de STAT Activados , Sumoilación , Replicación Viral , Animales , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Ratones , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Virulencia
2.
J Virol ; 96(4): e0163021, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34908445

RESUMEN

The matrix protein (M1) of influenza A virus plays an important role in replication, assembly, and budding. A previous study found that aspartic acid (D) at position 30 and alanine (A) at position 215 of M1 contribute to the high pathogenicity of H5N1 viruses in mice, and double mutations of D to asparagine (N) at position 30 (D30N) and A to threonine (T) at position 215 (A215T) in M1 dramatically attenuate H5N1 viruses in mice. However, the underlying mechanisms by which these M1 mutations attenuate the virulence of H5N1 viruses are unknown. Here, we found that the amino acid mutation A215T eliminates the SUMOylation of M1 by reducing its interaction with the host SUMO1 protein, significantly reducing the stability of M1, slowing the export of the M1-vRNP complex from the nucleus to the cytoplasm, and reducing viral replication in MDCK cells. We further found that the D30N mutation in M1 alters the shape of progeny viruses from filamentous to spherical virions. Our findings reveal an essential role for M1 215A SUMOylation and M1 30D-related filamentous morphology in the pathogenesis of avian influenza viruses, which could be targeted in novel antiviral drug designs. IMPORTANCE Identification of the pathogenic mechanism of highly pathogenic avian influenza viruses in mammals is helpful to develop novel anti-influenza virus strategies. Two amino acid mutations (D30N and A215T) in M1 were found to collectively attenuate H5N1 influenza viruses in mice, but the underlying mechanism remained unknown. This study found that the A215T mutation significantly decreases the SUMOylation of M1, which in turn attenuates the replication of H5N1 virus in mammalian cells. The D30N mutation in M1 was found to change the virion shape from filamentous to spherical. These findings are important for understanding the molecular mechanism of virulence of highly pathogenic avian influenza viruses in mammals.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/virología , Proteínas de la Matriz Viral/metabolismo , Replicación Viral , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , Perros , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/ultraestructura , Células de Riñón Canino Madin Darby , Ratones , Mutación , Infecciones por Orthomyxoviridae/metabolismo , Estabilidad Proteica , Ribonucleoproteínas/metabolismo , Sumoilación , Proteínas de la Matriz Viral/genética , Virión/ultraestructura , Virulencia/genética , Replicación Viral/genética
3.
PLoS Pathog ; 17(12): e1010141, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34871331

RESUMEN

Influenza virus infection is dependent on host cellular factors, and identification of these factors and their underlying mechanisms can provide important information for the development of strategies to inhibit viral infection. Here, we used a highly pathogenic H5N1 influenza virus to perform a genome-wide CRISPR/Cas9 gene knockout screen in human lung epithelial cells (A549 cells), and found that knockout of transmembrane protein immunoglobulin superfamily DCC subclass member 4 (IGDCC4) significantly reduced the replication of the virus in A549 cells. Further studies showed that IGDCC4 interacted with the viral hemagglutinin protein and facilitated virus internalization into host cells. Animal infection studies showed that replication of H5N1 virus in the nasal turbinates, lungs, and kidneys of IGDCC4-knockout mice was significantly lower than that in the corresponding organs of wild-type mice. Half of the IGDCC4-knockout mice survived a lethal H5N1 virus challenge, whereas all of the wild-type mice died within 11 days of infection. Our study identifies a novel host factor that promotes influenza virus infection by facilitating internalization and provides insights that will support the development of antiviral therapies.


Asunto(s)
Receptor DCC/metabolismo , Endocitosis/fisiología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/virología , Internalización del Virus , Células A549 , Animales , Sistemas CRISPR-Cas , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Noqueados
4.
PLoS Pathog ; 17(4): e1009561, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33905456

RESUMEN

The H7N9 avian influenza virus (AIV) that emerged in China have caused five waves of human infection. Further human cases have been successfully prevented since September 2017 through the use of an H7N9 vaccine in poultry. However, the H7N9 AIV has not been eradicated from poultry in China, and its evolution remains largely unexplored. In this study, we isolated 19 H7N9 AIVs during surveillance and diagnosis from February 2018 to December 2019, and genetic analysis showed that these viruses have formed two different genotypes. Animal studies indicated that the H7N9 viruses are highly lethal to chicken, cause mild infection in ducks, but have distinct pathotypes in mice. The viruses bound to avian-type receptors with high affinity, but gradually lost their ability to bind to human-type receptors. Importantly, we found that H7N9 AIVs isolated in 2019 were antigenically different from the H7N9 vaccine strain that was used for H7N9 influenza control in poultry, and that replication of these viruses cannot, therefore, be completely prevented in vaccinated chickens. We further revealed that two amino acid mutations at positions 135 and 160 in the HA protein added two glycosylation sites and facilitated the escape of the H7N9 viruses from the vaccine-induced immunity. Our study provides important insights into H7N9 virus evolution and control.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/aislamiento & purificación , Vacunas contra la Influenza/uso terapéutico , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/virología , Animales , Animales de Zoológico/virología , Pollos/virología , China/epidemiología , Patos/virología , Control de Infecciones/métodos , Subtipo H7N9 del Virus de la Influenza A/clasificación , Subtipo H7N9 del Virus de la Influenza A/fisiología , Gripe Aviar/epidemiología , Gripe Aviar/virología , Ratones , Filogenia , Vigilancia de la Población , Aves de Corral , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/prevención & control
5.
PLoS Pathog ; 17(2): e1009336, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33571308

RESUMEN

Posttranslational modifications, such as SUMOylation, play specific roles in the life cycle of invading pathogens. However, the effect of SUMOylation on the adaptation, pathogenesis, and transmission of influenza A virus (IAV) remains largely unknown. Here, we found that a conserved lysine residue at position 612 (K612) of the polymerase basic protein 1 (PB1) of IAV is a bona fide SUMOylation site. SUMOylation of PB1 at K612 had no effect on the stability or cellular localization of PB1, but was critical for viral ribonucleoprotein (vRNP) complex activity and virus replication in vitro. When tested in vivo, we found that the virulence of SUMOylation-defective PB1/K612R mutant IAVs was highly attenuated in mice. Moreover, the airborne transmission of a 2009 pandemic H1N1 PB1/K612R mutant virus was impaired in ferrets, resulting in reversion to wild-type PB1 K612. Mechanistically, SUMOylation at K612 was essential for PB1 to act as the enzymatic core of the viral polymerase by preserving its ability to bind viral RNA. Our study reveals an essential role for PB1 K612 SUMOylation in the pathogenesis and transmission of IAVs, which can be targeted for the design of anti-influenza therapies.


Asunto(s)
Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/transmisión , ARN Viral/metabolismo , Sumoilación , Proteínas Virales/metabolismo , Replicación Viral , Animales , Perros , Femenino , Hurones , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , ARN Viral/genética , Proteínas Virales/química , Proteínas Virales/genética , Acoplamiento Viral
6.
J Med Virol ; 95(2): e28476, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36609855

RESUMEN

The H10 subtypes of avian influenza viruses pose a continual threat to the poultry industry and human health. The sporadic spillover of H10 subtypes viruses from poultry to humans is represented by the H10N8 human cases in 2013 and the recent H10N3 human infection in 2021. However, the genesis and characteristics of the recent reassortment H10N3 viruses have not been systemically investigated. In this study, we characterized 20 H10N3 viruses isolated in live poultry markets during routine nationwide surveillance in China from 2014 to 2021. The viruses in the recent reassortant genotype acquired their hemagglutinin (HA) and neuraminidase (NA) genes from the duck H10 viruses and H7N3 viruses, respectively, whereas the internal genes were derived from chicken H9N2 viruses as early as 2019. Receptor-binding analysis indicated that two of the tested H10N3 viruses had a higher affinity for human-type receptors than for avian-type receptors, highlighting the potential risk of avian-to-human transmission. Animal studies showed that only viruses belonging to the recent reassortant genotype were pathogenic in mice; two tested viruses transmitted via direct contact and one virus transmitted by respiratory droplets in guinea pigs, though with limited efficiency. These findings emphasize the need for enhanced surveillance of H10N3 viruses.


Asunto(s)
Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Humanos , Animales , Cobayas , Ratones , Subtipo H9N2 del Virus de la Influenza A/genética , Subtipo H7N3 del Virus de la Influenza A , Aves de Corral , Pollos , China/epidemiología , Filogenia , Virus Reordenados/genética
7.
Virol J ; 20(1): 261, 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-37957729

RESUMEN

BACKGROUND: Avian influenza (AI) is a disease caused by the avian influenza virus (AIV). These viruses spread naturally among wild aquatic birds worldwide and infect domestic poultry, other birds, and other animal species. Currently, real-time reverse transcription polymerase chain reaction (rRT-PCR) is mainly used to detect the presence of pathogens and has good sensitivity and specificity. However, the diagnosis requires sophisticated instruments under laboratory conditions, which significantly limits point-of-care testing (POCT). Rapid, reliable, non-lab-equipment-reliant, sensitive, and specific diagnostic tests are urgently needed for rapid clinical detection and diagnosis. Our study aimed to develop a reverse transcription recombinase polymerase amplification (RT-RPA)/CRISPR method which improves on these limitations. METHODS: The Cas12a protein was purified by affinity chromatography with Ni-agarose resin and observed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Specific CRISPR RNA (crRNA) and primers targeting the M and NP genes of the AIV were designed and screened. By combining RT-RPA with the Cas12a/crRNA trans-cleavage system, a detection system that uses fluorescence readouts under blue light or lateral flow strips was established. Sensitivity assays were performed using a tenfold dilution series of plasmids and RNA of the M and NP genes as templates. The specificity of this method was determined using H1-H16 subtype AIVs and other avian pathogens, such as newcastle disease virus (NDV), infectious bursal disease virus (IBDV), and infectious bronchitis virus (IBV). RESULTS: The results showed that the method was able to detect AIV and that the detection limit can reach 6.7 copies/µL and 12 copies/µL for the M and NP gene, respectively. In addition, this assay showed no cross-reactivity with other avian-derived RNA viruses such as NDV, IBDV, and IBV. Moreover, the detection system presented 97.5% consistency and agreement with rRT-PCR and virus isolation for detecting samples from poultry. This portable and accurate method has great potential for AIV detection in the field. CONCLUSION: An RT-RPA/CRISPR method was developed for rapid, sensitive detection of AIV. The new system presents a good potential as an accurate, user-friendly, and inexpensive platform for point-of-care testing applications.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Animales , Gripe Aviar/diagnóstico , Sistemas CRISPR-Cas , Aves , Aves de Corral , Sensibilidad y Especificidad , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Virus de la Enfermedad de Newcastle/genética , ARN
8.
Macromol Rapid Commun ; 44(13): e2300092, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37029933

RESUMEN

Poly(amic acid) (PAA) materials as the precursor of polyimide generally show remarkably poor mechanical properties, thus limiting their application as the engineering plastics. In this study, it is demonstrated that the mechanical properties of PAA materials can be improved significantly for tens of folds with breaking strength >50 MPa, Young's modulus >400 MPa, and elongation at break >300% by incorporation of 20% (mol%) poly(propylene glycol) (PPO) soft segments. The optimization for suitable hard-soft composition with 20% PPO and the existence of various hydrogen bonds with different binding energies can dissipate energies efficiently, which simultaneously improve the material strength and toughness. In addition, PAA82 films exhibit excellent tolerance toward cyclic stretch, and have the capability to resist various harsh conditions including solar radiation testing (1 sun), heat (85 °C), alkalinity (pH 10), and acidity (pH 4) over one month. Noted that PAA82 films can be laminated with Kapton films, which show excellent resistance to ultrahigh (200 °C) and ultralow temperature (-196 °C). The laminated film also exhibits bulletproof property with a thickness of 6 mm. The strategy via modulation of hard-soft compositions and hydrogen bonds in PAA materials shows great potentials to improve the mechanical properties of polymeric materials.


Asunto(s)
Plásticos , Polímeros , Enlace de Hidrógeno , Polímeros/química , Temperatura , Calor
9.
Euro Surveill ; 28(41)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37824247

RESUMEN

BackgroundTwo human cases of avian influenza A (H3N8) virus infection were reported in China in 2022.AimTo characterise H3N8 viruses circulating in China in September 2021-May 2022.MethodsWe sampled poultry and poultry-related environments in 25 Chinese provinces. After isolating H3N8 viruses, whole genome sequences were obtained for molecular and phylogenetic analyses. The specificity of H3N8 viruses towards human or avian receptors was assessed in vitro. Their ability to replicate in chicken and mice, and to transmit between guinea pigs was also investigated.ResultsIn total, 98 H3N8 avian influenza virus isolates were retrieved from 38,639 samples; genetic analysis of 31 representative isolates revealed 17 genotypes. Viruses belonging to 10 of these genotypes had six internal genes originating from influenza A (H9N2) viruses. These reassorted viruses could be found in live poultry markets and comprised the strains responsible for the two human infections. A subset of nine H3N8 viruses (including six reassorted) that replicated efficiently in mice bound to both avian-type and human-type receptors in vitro. Three reassorted viruses were shed by chickens for up to 9 days, replicating efficiently in their upper respiratory tract. Five reassorted viruses tested on guinea pigs were transmissible among these by respiratory droplets.ConclusionAvian H3N8 viruses with H9N2 virus internal genes, causing two human infections, occurred in live poultry markets in China. The low pathogenicity of H3N8 viruses in poultry allows their continuous circulation with potential for reassortment. Careful monitoring of spill-over infections in humans is important to strengthen early-warning systems and maintain influenza pandemic preparedness.


Asunto(s)
Subtipo H3N8 del Virus de la Influenza A , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Enfermedades de las Aves de Corral , Animales , Humanos , Ratones , Cobayas , Gripe Humana/epidemiología , Aves de Corral , Gripe Aviar/epidemiología , Subtipo H9N2 del Virus de la Influenza A/genética , Filogenia , Pollos , China/epidemiología , Enfermedades de las Aves de Corral/epidemiología
11.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31666373

RESUMEN

The low-pathogenic H7N9 influenza viruses that emerged in 2013 acquired an insertion of four amino acids in their hemagglutinin cleavage site and thereby became highly pathogenic to chickens in 2017. Previous studies indicated that these highly pathogenic H7N9 viruses are virulent in chickens but have distinct pathotypes in mice. A/chicken/Guangdong/SD098/2017 (CK/SD098) is avirulent, with a 50% mouse lethal dose (MLD50) of >7.5 log10 50% egg infectious dose (EID50), whereas A/chicken/Hunan/S1220/2017 (CK/S1220) is virulent in mice, with an MLD50 of 3.2 log10 EID50 In this study, we explored the genetic determinants that contribute to the difference in virulence between these two H7N9 viruses by generating a series of reassortants and mutants in the CK/S1220 virus background and testing their virulence in mice. We found that the reassortant CK/1220-SD098-NP, carrying the nucleoprotein (NP) of CK/SD098, was avirulent in mice, with an MLD50 of >107.5 EID50 The NPs of these two viruses differ by two amino acids, at positions 286 and 437. We further demonstrated that the amino acid mutations A286V and T437M of NP independently slowed the process of NP import to and export from the nucleus and thus jointly impaired the viral life cycle and attenuated the virulence of these H7N9 viruses in mice. Our study identified new virulence determinants in NP and provided novel targets for the development of live attenuated vaccines and antiviral drugs against influenza viruses.IMPORTANCE The H7N9 influenza viruses that emerged in China in 2013 have caused over 1,500 human infections, with a mortality rate of nearly 40%. The viruses were initially low pathogenic but became highly pathogenic in chickens at the beginning of 2017 and caused severe disease outbreaks in poultry. Several studies suggested that the highly pathogenic H7N9 viruses have increased virulence in mammals; however, the genetic basis of the virulence of H7N9 viruses in mammals is not fully understood. Here, we found that two amino acids, 286A and 437T, in NP are prerequisites for the virulence of H7N9 viruses in mice and the mutations A286V and T437M collectively eliminate the virulence of H7N9 viruses in mice. Our study further demonstrated that the virulence of influenza viruses is a polygenic trait, and the newly identified virulence-related residues in NP may provide new targets for attenuated influenza vaccine and antiviral drug development.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/metabolismo , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Mutación Missense , Infecciones por Orthomyxoviridae/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas del Núcleo Viral/metabolismo , Sustitución de Aminoácidos , Animales , Pollos , Perros , Células HEK293 , Humanos , Subtipo H7N9 del Virus de la Influenza A/genética , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Proteínas de la Nucleocápside , Infecciones por Orthomyxoviridae/genética , Proteínas de Unión al ARN/genética , Vacunas Atenuadas/genética , Vacunas Atenuadas/metabolismo , Proteínas del Núcleo Viral/genética
12.
Cryobiology ; 101: 105-114, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33989617

RESUMEN

Germplasm preservation of livestock or endangered animals and expansion of germline stem cells are important. The purpose of this study is to investigate whether supplementation of trehalose to the freezing medium (FM) reduces tissular damage and improves the quality of testicular cells in the cryopreserved bovine testicular tissues. We herein established an optimized protocol for the cryopreservation of bovine testicular tissues, and the isolation as well as culture of bovine germ cells containing spermatogonial stem cells (SSCs) from these tissues. The results showed that FM containing 10% dimethyl sulfoxide (Me2SO/DMSO), 10% knockout serum replacement (KSR) and 20% trehalose (FM5) combined with the uncontrolled slow freezing (USF) procedures has the optimized cryoprotective effect on bovine testicular tissues. The FM5 + USF protocol reduced the cell apoptosis, maintained high cell viability, supported the structural integrity and seminiferous epithelial cohesion similar to that in the fresh tissues. Viable germ cells containing SSCs were effectively isolated from these tissues and they maintained germline marker expressions in the co-testicular cells and co-mouse embryonic fibroblasts (MEF) feeder culture systems respectively, during the short-term culture. Additionally, upregulated transcriptions of spermatogenic differentiation marker C-KIT and meiotic marker SYCP3 were detected in these cells after retinoic acid-induced differentiation. Together, FM5 + USF is suitable for the cryopreservation of bovine testicular tissues, with benefits of reducing the apoptosis, maintaining the cell viability, supporting the testicular structure integrity, and sustaining the survival and differentiation potential of bovine germ cells containing SSCs.


Asunto(s)
Criopreservación , Trehalosa , Animales , Bovinos , Supervivencia Celular , Criopreservación/métodos , Crioprotectores/farmacología , Dimetilsulfóxido , Fibroblastos , Masculino , Ratones , Espermatogonias , Testículo , Trehalosa/farmacología
13.
Emerg Infect Dis ; 26(12): 2956-2960, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33030424

RESUMEN

In January 2020, the subclade 2.3.4.4h of highly pathogenic avian influenza (H5N6) virus infected migratory whooper swans and mute swans in Xinjiang, western China. The virus is lethal to chickens and ducks but has low pathogenicity in mice. Antigenically, this subclade is similar to the H5N1 vaccine seed virus Re-11.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Animales , Pollos , China/epidemiología , Brotes de Enfermedades , Patos , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/epidemiología , Ratones , Filogenia
14.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597771

RESUMEN

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


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/genética , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/transmisión , ARN Polimerasa Dependiente del ARN/genética , Virus Reordenados/genética , Proteínas de la Matriz Viral/genética , Proteínas Virales/genética , Células A549 , Sustitución de Aminoácidos , Animales , Expresión Génica , Cobayas , Humanos , Subtipo H7N9 del Virus de la Influenza A/metabolismo , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Mutación , Neuraminidasa/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , ARN Polimerasa Dependiente del ARN/metabolismo , Virus Reordenados/metabolismo , Virus Reordenados/patogenicidad , Genética Inversa , Relación Estructura-Actividad , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/metabolismo , Replicación Viral
15.
J Virol ; 90(4): 1872-9, 2016 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-26656683

RESUMEN

UNLABELLED: We isolated two H5N1 viruses, A/duck/Hunan/S4020/2008 (DK/08) and A/chicken/Guangxi/S2039/2009 (CK/09), from live-bird markets during routine surveillance and found that these two viruses are genetically similar but differ in their replication and virulence in mice. The CK/09 virus is lethal for mice with a 50% mouse lethal dose (MLD50) of 1.6 log10 50% egg infectious doses (EID50), whereas the DK/08 virus is nonpathogenic for mice with an MLD50 value of 6.2 log10 EID50. We explored the genetic basis of the virulence difference of these two viruses by generating a series of reassortant viruses and mutants in the lethal virus CK/09 background and evaluating their virulence in mice. We found that the PB1 gene of the DK/08 virus dramatically attenuated the virulence of the CK/09 virus and that the amino acid at position 622 in PB1 made an important contribution. We further demonstrated that the mutation of glycine (G) to aspartic acid (D) at position 622 in PB1 partially impaired the binding of PB1 to viral RNA, thereby dramatically decreasing the polymerase activity and attenuating H5N1 virus virulence in mice. Our results identify a novel virulence-related marker of H5N1 influenza viruses and provide a new target for live attenuated vaccine development. IMPORTANCE: H5N1 avian influenza viruses have caused the deaths of nearly 60% of the humans that they have infected since 1997 and clearly represent a threat to public health. A thorough understanding of the genetic basis of virulence determinants will provide important insights for antiviral drug and live attenuated vaccine development. Several virulence-related markers in the PB2, PA, M1, and NS1 proteins of H5N1 viruses have been identified. In this study, we isolated two H5N1 avian influenza viruses that are genetically similar but differ in their virulence in mice, and we identified a new virulence-related marker in the PB1 gene. We found that the mutation of glycine (G) to aspartic acid (D) at position 622 in PB1 partially impairs the binding of PB1 to viral RNA, thereby attenuating H5N1 virus virulence in mice. This newly identified virulence-related marker could be applied to the development of live attenuated vaccines against H5N1 influenza.


Asunto(s)
Glicina/metabolismo , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/patología , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Femenino , Glicina/genética , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/fisiología , Dosificación Letal Mediana , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Virus Reordenados/fisiología , Genética Inversa , Análisis de Supervivencia , Proteínas Virales/genética , Virulencia , Factores de Virulencia/genética , Replicación Viral
16.
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
17.
J Virol ; 90(3): 1455-69, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26581996

RESUMEN

UNLABELLED: H4 avian influenza virus (AIV) is one of the most prevalent influenza virus subtypes in the world. However, whether H4 AIVs pose a threat to public health remains largely unclear. Here, we analyzed the phylogenetic relationships, receptor binding properties, replication, and transmissibility in mammals of H4 AIVs isolated from live poultry markets in China between 2009 and 2012. Genomic sequence analysis of 36 representative H4 viruses revealed 32 different genotypes, indicating that these viruses are undergoing complex and frequent reassortment events. All 32 viruses tested could replicate in the respiratory organs of infected mice without prior adaptation. Receptor binding analysis demonstrated that the H4 AIVs bound to α-2,6-linked glycans, although they retained the binding preference for α-2,3-linked glycans. When we tested the direct-contact transmission of 10 H4 viruses in guinea pigs, we found that three viruses did not transmit to any of the contact animals, one virus transmitted to one of three contact animals, and six viruses transmitted to all three contact animals. When we further tested the respiratory droplet transmissibility of four of the viruses that transmitted efficiently via direct contact, we found that three of them could transmit to one or two of the five exposed animals. Our study demonstrates that the current circulating H4 AIVs can infect, replicate in, and transmit to mammalian hosts, thereby posing a potential threat to human health. These findings emphasize the continual need for enhanced surveillance of H4 AIVs. IMPORTANCE: Numerous surveillance studies have documented the wide distribution of H4 AIVs throughout the world, yet the biological properties of H4 viruses have not been well studied. In this study, we found that multiple genotypes of H4 viruses are cocirculating in the live poultry markets of China and that H4 viruses can replicate in mice, possess human-type receptor binding specificity, and transmit between guinea pigs via direct contact. Strikingly, some H4 strains also can transmit via respiratory droplet, albeit with limited efficiency. These results clearly show the potential threat posed by H4 viruses to public health.


Asunto(s)
Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/virología , Infecciones por Orthomyxoviridae/veterinaria , Aves de Corral/virología , Acoplamiento Viral , Replicación Viral , Animales , China , Análisis por Conglomerados , Femenino , Genoma Viral , Cobayas , Virus de la Influenza A/genética , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/transmisión , Filogenia , ARN Viral/genética , Receptores Virales/análisis , Análisis de Secuencia de ADN , Homología de Secuencia
18.
J Virol ; 89(12): 6506-10, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25855738

RESUMEN

We analyzed eight H10N8 viruses isolated from ducks and chickens in live poultry markets from 2009 to 2013 in China. These viruses showed distinct genetic diversity and formed five genotypes: the four duck isolates formed four different genotypes, whereas the four chicken viruses belong to a single genotype. The viruses bound to both human- and avian-type receptors, and four of the viruses caused 12.7% to 22.5% body weight loss in mice.


Asunto(s)
Pollos/virología , Patos/virología , Variación Genética , Subtipo H10N8 del Virus de la Influenza A/fisiología , Acoplamiento Viral , Replicación Viral , Animales , Peso Corporal , China , Modelos Animales de Enfermedad , Genotipo , Humanos , Subtipo H10N8 del Virus de la Influenza A/genética , Subtipo H10N8 del Virus de la Influenza A/aislamiento & purificación , Ratones , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , ARN Viral/genética , Análisis de Secuencia de ADN , Virulencia
19.
PLoS Pathog ; 10(11): e1004508, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25411973

RESUMEN

H9N2 subtype influenza viruses have been detected in different species of wild birds and domestic poultry in many countries for several decades. Because these viruses are of low pathogenicity in poultry, their eradication is not a priority for animal disease control in many countries, which has allowed them to continue to evolve and spread. Here, we characterized the genetic variation, receptor-binding specificity, replication capability, and transmission in mammals of a series of H9N2 influenza viruses that were detected in live poultry markets in southern China between 2009 and 2013. Thirty-five viruses represented 17 genotypes on the basis of genomic diversity, and one specific "internal-gene-combination" predominated among the H9N2 viruses. This gene combination was also present in the H7N9 and H10N8 viruses that have infected humans in China. All of the 35 viruses preferentially bound to the human-like receptor, although two also retained the ability to bind to the avian-like receptor. Six of nine viruses tested were transmissible in ferrets by respiratory droplet; two were highly transmissible. Some H9N2 viruses readily acquired the 627K or 701N mutation in their PB2 gene upon infection of ferrets, further enhancing their virulence and transmission in mammals. Our study indicates that the widespread dissemination of H9N2 viruses poses a threat to human health not only because of the potential of these viruses to cause an influenza pandemic, but also because they can function as "vehicles" to deliver different subtypes of influenza viruses from avian species to humans.


Asunto(s)
Variación Genética , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Animales , Secuencia de Bases , Pollos , China , Perros , Hurones , Humanos , Subtipo H9N2 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/aislamiento & purificación , Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Gripe Aviar/genética , Gripe Aviar/transmisión , Gripe Humana/genética , Gripe Humana/transmisión , Células de Riñón Canino Madin Darby , Datos de Secuencia Molecular , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/transmisión , Enfermedades de las Aves de Corral/virología
20.
J Clin Microbiol ; 53(12): 3884-7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26378283

RESUMEN

We developed hemagglutinin- and neuraminidase-specific one-step reverse transcription-loop-mediated isothermal amplification assays for detecting the H10N8 virus. The detection limit of the assays was 10 copies of H10N8 virus, and the assays did not amplify nonspecific RNA. The assays can detect H10N8 virus from chicken samples with high sensitivity and specificity, and they can serve as an effective tool for detecting and monitoring H10N8 virus in live poultry markets.


Asunto(s)
Subtipo H10N8 del Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/diagnóstico , Técnicas de Amplificación de Ácido Nucleico/métodos , Transcripción Reversa , Animales , Pollos , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H10N8 del Virus de la Influenza A/genética , Gripe Aviar/virología , Neuraminidasa/genética , Sensibilidad y Especificidad , Temperatura , Factores de Tiempo , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA