Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Virol ; 93(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30814289

RESUMEN

Infection by enteroviruses can cause severe neurological complications in humans. The interactions between the enteroviral and host proteins may facilitate the virus replication and be involved in the pathogenicity of infected individuals. It has been shown that human enteroviruses possess various mechanisms to suppress host innate immune responses in infected cells. Previous studies showed that infection by enterovirus 71 (EV71) causes the degradation of MDA5, which is a critical cytoplasmic pathogen sensor in the recognition of picornaviruses for initiating transcription of type I interferons. In the present study, we demonstrated that the RNA-dependent RNA polymerase (RdRP; also denoted 3Dpol) encoded by EV71 interacts with the caspase activation and recruitment domains (CARDs) of MDA5 and plays a role in the inhibition of MDA5-mediated beta interferon (IFN-ß) promoter activation and mRNA expression. In addition, we found that the 3Dpol protein encoded by coxsackievirus B3 also interacted with MDA5 and downregulated the antiviral signaling initiated by MDA5. These findings indicate that enteroviral RdRP may function as an antagonist against the host antiviral innate immune response.IMPORTANCE Infection by enteroviruses causes severe neurological complications in humans. Human enteroviruses possess various mechanisms to suppress the host type I interferon (IFN) response in infected cells to establish viral replication. In the present study, we found that the enteroviral 3Dpol protein (or RdRP), which is a viral RNA-dependent RNA polymerase for replicating viral RNA, plays a role in the inhibition of MDA5-mediated beta interferon (IFN-ß) promoter activation. We further demonstrated that enteroviral 3Dpol protein interacts with the caspase activation and recruitment domains (CARDs) of MDA5. These findings indicate that enteroviral RdRP functions as an antagonist against the host antiviral response.


Asunto(s)
Enterovirus Humano A/metabolismo , Helicasa Inducida por Interferón IFIH1/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Dominio de Reclutamiento y Activación de Caspasas/genética , Dominio de Reclutamiento y Activación de Caspasas/fisiología , Enterovirus/genética , Enterovirus/metabolismo , Enterovirus Humano A/genética , Enterovirus Humano B/metabolismo , Infecciones por Enterovirus/virología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Interferón beta/metabolismo , Interferones/metabolismo , Interferones/fisiología , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , Transducción de Señal , Replicación Viral
2.
J Proteome Res ; 17(4): 1474-1484, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29558158

RESUMEN

Influenza A virus infections can result in severe respiratory diseases. The H7N9 subtype of avian influenza A virus has been transmitted to humans and caused severe disease and death. Nonstructural protein 1 (NS1) of influenza A virus is a virulence determinant during viral infection. To elucidate the functions of the NS1 encoded by influenza A H7N9 virus (H7N9 NS1), interaction partners of H7N9 NS1 in human cells were identified with immunoprecipitation followed by SDS-PAGE coupled with liquid chromatography-tandem mass spectrometry (GeLC-MS/MS). We identified 36 cellular proteins as the interacting partners of the H7N9 NS1, and they are involved in RNA processing, mRNA splicing via spliceosome, and the mRNA surveillance pathway. Two of the interacting partners, cleavage and polyadenylation specificity factor subunit 2 (CPSF2) and CPSF7, were confirmed to interact with H7N9 NS1 using coimmunoprecipitation and immunoblotting based on the previous finding that the two proteins are involved in pre-mRNA polyadenylation machinery. Furthermore, we illustrate that overexpression of H7N9 NS1, as well as infection by the influenza A H7N9 virus, interfered with pre-mRNA polyadenylation in host cells. This study comprehensively profiled the interactome of H7N9 NS1 in host cells, and the results demonstrate a novel endotype for H7N9 NS1 in inhibiting host mRNA maturation.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/química , ARN Mensajero/antagonistas & inhibidores , Proteínas no Estructurales Virales/farmacología , Animales , Factor de Especificidad de Desdoblamiento y Poliadenilación , Interacciones Microbiota-Huesped , Humanos , Immunoblotting , Inmunoprecipitación , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Unión Proteica , Factores de Escisión y Poliadenilación de ARNm
3.
J Virol ; 90(7): 3789-93, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26764009

RESUMEN

A panel of influenza A viruses expressing chimeric hemagglutinins (cHA) with intragroup or intergroup head/stalk combinations was generated. Viruses were characterized for growth kinetics and preservation of stalk epitopes. With a few notable exceptions, cHA viruses behaved similarly to wild-type viruses and maintained stalk epitopes, which indicated their potential as vaccine candidates to induce stalk-specific antibodies.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/biosíntesis , Virus de la Influenza A/genética , Proteínas Recombinantes/biosíntesis , Línea Celular , Epítopos/genética , Expresión Génica , Inestabilidad Genómica , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/fisiología , Vacunas contra la Influenza/genética , Pruebas de Neutralización , Proteínas Recombinantes/genética , Ensayo de Placa Viral , Replicación Viral
4.
Proc Natl Acad Sci U S A ; 110(50): 20248-53, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24277853

RESUMEN

The molecular basis for the diversity across influenza strains is poorly understood. To gain insight into this question, we mutagenized the viral genome and sequenced recoverable viruses. Only two small regions in the genome were enriched for insertions, the hemagglutinin head and the immune-modulatory nonstructural protein 1. These proteins play a major role in host adaptation, and thus need to be able to evolve rapidly. We propose a model in which certain influenza A virus proteins (or protein domains) exist as highly plastic scaffolds, which will readily accept mutations yet retain their functionality. This model implies that the ability to rapidly acquire mutations is an inherent aspect of influenza HA and nonstructural protein 1 proteins; further, this may explain why rapid antigenic drift and a broad host range is observed with influenza A virus and not with some other RNA viruses.


Asunto(s)
Adaptación Biológica/genética , Evolución Molecular , Genoma Viral/genética , Virus de la Influenza A/genética , Modelos Genéticos , Mutagénesis/genética , Proteínas no Estructurales Virales/genética , Animales , Secuencia de Bases , Cartilla de ADN/genética , Perros , Genes Virales/genética , Células HEK293 , Hemaglutininas/genética , Humanos , Células de Riñón Canino Madin Darby , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
5.
J Infect Dis ; 208(11): 1898-905, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23901080

RESUMEN

BACKGROUND: Reassortment within polymerase genes causes changes in the pathogenicity of influenza A viruses. We previously reported that the 2009 pH1N1 PA enhanced the pathogenicity of seasonal H1N1. We examined the effects of the PA gene from the HPAI H5N1 following its introduction into currently circulating seasonal influenza viruses. METHODS: To evaluate the role of H5N1 PA in altering the virulence of seasonal influenza viruses, we generated a recombinant seasonal H3N2 (3446) that expressed the H5N1 PA protein (VPA) and evaluated the RNP activity, growth kinetics, and pathogenicity of the reassortant virus in mice. RESULTS: Compared with the wild-type 3446 virus, the substitution of the H5N1 PA gene into the 3446 virus (VPA/3446) resulted in increased RNP activity and an increased replication rate in A549 cells. The recombinant VPA/3446 virus also caused more severe pneumonia in Casp 1(-/-) mice than in IL1ß(-/-) and wild-type B6 mice. CONCLUSIONS: Although the PA from H5N1 is incidentally compatible with a seasonal H3N2 backbone, the H5N1 PA affected the virulence of seasonal H3N2, particularly in inflammasome-related innate immunity deficient mice. These findings highlight the importance of monitoring PA reassortment in seasonal flu, and confirm the role of the Caspase-1 gene in influenza pathogenesis.


Asunto(s)
Caspasa 1/metabolismo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/virología , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Perros , Ingeniería Genética , Humanos , Inmunidad Innata , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , ARN Polimerasa Dependiente del ARN/metabolismo , Virus Reordenados , Organismos Libres de Patógenos Específicos , Proteínas Virales/metabolismo , Virulencia , Replicación Viral
6.
J Infect Dis ; 204(6): 864-72, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21849283

RESUMEN

BACKGROUND: The 2009 influenza A pandemic virus (H1N1(pdm)) may reassort with old seasonal influenza A virus (H1N1141) in humans and potentially change their pathogenicity. METHODS AND RESULTS: This study focuses on the reassortment of ribonucleoproteins (RNPs) among H1N1(pdm) and seasonal influenza A viruses. A single RNP gene reassortment altered reporter gene expression levels driven by polymerase complex in transfection system. The growth rates of recombinant viruses with different RNP recombinations were changed in A549 cells. Mice were infected with recombinant viruses containing single RNP gene reassortment, and pathogenicity was examined. The results demonstrated that the median lethal dose (LD50) of the PB2141/PB1141/PA(pdm)/NP141 recombinant virus was lower than that of the seasonal H1N1 virus. Viral titers of this reassorted virus in the lung and spleen were significantly higher than that in seasonal H1N1 virus-challenged mice. CONCLUSIONS: Although the changes of RNP activity did not exactly reflect to mice virulence, we consistently observed that the PA gene of H1N1(pdm) results in increased polymerase activity, better replication in mice, and lower LD50. Our findings suggest that monitoring of gene reassortment for the 2009 pandemic influenza and seasonal human viruses is also important, which would help to constrain the potential emergence of a more virulent influenza A variant.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Proteínas de Unión al ARN/genética , Virus Reordenados/patogenicidad , Proteínas del Núcleo Viral/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/virología , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas de la Nucleocápside , Infecciones por Orthomyxoviridae/mortalidad , Virus Reordenados/genética , Enfermedades de los Roedores/patología , Enfermedades de los Roedores/virología , Análisis de Supervivencia , Virulencia
7.
J Virol ; 84(19): 10051-62, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660199

RESUMEN

PB1-F2 is a viral protein that is encoded by the PB1 gene of influenza A virus by alternative translation. It varies in length and sequence context among different strains. The present study examines the functions of PB1-F2 proteins derived from various human and avian viruses. While H1N1 PB1-F2 was found to target mitochondria and enhance apoptosis, H5N1 PB1-F2, surprisingly, did not localize specifically to mitochondria and displayed no ability to enhance apoptosis. Introducing Leu into positions 69 (Q69L) and 75 (H75L) in the C terminus of H5N1 PB1-F2 drove 40.7% of the protein to localize to mitochondria compared with the level of mitochondrial localization of wild-type H5N1 PB1-F2, suggesting that a Leu-rich sequence in the C terminus is important for targeting of mitochondria. However, H5N1 PB1-F2 contributes to viral RNP activity, which is responsible for viral RNA replication. Lastly, although the swine-origin influenza virus (S-OIV) contained a truncated form of PB1-F2 (12 amino acids [aa]), potential mutation in the future may enable it to contain a full-length product. Therefore, the functions of this putative S-OIV PB1-F2 (87 aa) were also investigated. Although this PB1-F2 from the mutated S-OIV shares only 54% amino acid sequence identity with that of seasonal H1N1 virus, it also increased viral RNP activity. The plaque size and growth curve of the viruses with and without S-OIV PB1-F2 differed greatly. The PB1-F2 protein has various lengths, amino acid sequences, cellular localizations, and functions in different strains, which result in strain-specific pathogenicity. Such genetic and functional diversities make it flexible and adaptable in maintaining the optimal replication efficiency and virulence for various strains of influenza A virus.


Asunto(s)
Virus de la Influenza A/fisiología , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis , Aves/virología , Línea Celular , Femenino , Genes Virales , Variación Genética , Células HeLa , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/fisiología , Subtipo H7N7 del Virus de la Influenza A/genética , Subtipo H7N7 del Virus de la Influenza A/patogenicidad , Subtipo H7N7 del Virus de la Influenza A/fisiología , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Porcinos/virología , Transfección , Proteínas Virales/química , Proteínas Virales/genética , Virulencia/fisiología , Replicación Viral/fisiología
8.
Viruses ; 13(10)2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34696339

RESUMEN

Influenza A virus nonstructural protein 1 (NS1) plays an important role in evading host innate immunity. NS1 inhibits interferon (IFN) responses via multiple mechanisms, including sequestering dsRNA and suppressing retinoic acid-inducible gene I (RIG-I) signaling by interacting with RIG-I and tripartite motif-containing protein 25 (TRIM25). In the current study, we demonstrated the mitochondrial localization of NS1 at the early stage of influenza virus infection. Since NS1 does not contain mitochondria-targeting signals, we suspected that there is an association between the NS1 and mitochondrial proteins. This hypothesis was tested by demonstrating the interaction of NS1 with mitochondrial antiviral-signaling protein (MAVS) in a RIG-I-independent manner. Importantly, the association with MAVS facilitated the mitochondrial localization of NS1 and thereby significantly impeded MAVS-mediated Type I IFN production.


Asunto(s)
Mitocondrias/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Células A549 , Proteína 58 DEAD Box , Células HEK293 , Humanos , Inmunidad Innata/inmunología , Virus de la Influenza A/genética , Gripe Humana/inmunología , Interferones/metabolismo , Dominios Proteicos , Receptores Inmunológicos , Transducción de Señal , Factores de Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteínas no Estructurales Virales/genética
9.
Mol Ther Nucleic Acids ; 17: 10-23, 2019 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-31173947

RESUMEN

The role of microRNA (miRNA) in influenza A virus (IAV) host species specificity is not well understood as yet. Here, we show that a host miRNA, miR-1290, is induced through the extracellular signal-regulated kinase (ERK) pathway upon IAV infection and is associated with increased viral titers in human cells and ferret animal models. miR-1290 was observed to target and reduce expression of the host vimentin gene. Vimentin binds with the PB2 subunit of influenza A virus ribonucleoprotein (vRNP), and knockdown of vimentin expression significantly increased vRNP nuclear retention and viral polymerase activity. Interestingly, miR-1290 was not detected in either chicken cells or mouse animal models, and the 3' UTR of the chicken vimentin gene contains no binding site for miR-1290. These findings point to a host species-specific mechanism by which IAV upregulates miR-1290 to disrupt vimentin expression and retain vRNP in the nucleus, thereby enhancing viral polymerase activity and viral replication.

10.
Trends Microbiol ; 25(7): 546-561, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28242053

RESUMEN

Internal ribosome entry sites (IRESs) can be found in the mRNA of many viruses as well as in cellular genes involved in the stress response, cell cycle, and apoptosis. IRES-mediated translation can occur when dominant cap-dependent translation is inhibited, and viruses can take advantage of this to subvert host translation machinery. In this review, we focus on the four major types of IRES identified in RNA viruses, and outline their distinct structural properties and requirements of translational factors. We further discuss auxiliary host factors known as IRES trans-acting factors (ITAFs), which are involved in the modulation of optimal IRES activity. Currently known strategies employed by viruses to harness ITAFs and regulate IRES activity are also highlighted.


Asunto(s)
Regulación de la Expresión Génica , Sitios Internos de Entrada al Ribosoma , Virus ARN/genética , Interacciones Huésped-Patógeno/genética , Humanos , Picornaviridae/genética , Picornaviridae/metabolismo , Biosíntesis de Proteínas , Virus ARN/metabolismo
11.
Microbes Infect ; 19(12): 570-579, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28939355

RESUMEN

Prevention and treatment measures against influenza virus infection remain limited, and alternative host protection strategies are badly needed. In this review, we discuss the regulatory role of intestinal microbiota in influenza infections, and present the latest evidence for strategies seeking to harness gut microbiota for the management of influenza infections.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunomodulación/inmunología , Gripe Humana/prevención & control , Intestinos/microbiología , Humanos , Gripe Humana/virología
12.
PLoS One ; 12(11): e0188214, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29145464

RESUMEN

The viral ribonucleoprotein (vRNP) of influenza A virus is formed by virion RNA (vRNA), viral polymerase complex, and nucleoprotein (NP). The NP plays an important role in facilitating the replication and stabilization of viral RNA. To explore host factors that may be involved in the regulation of viral replication through interactions with NP, we conducted an immunoprecipitation experiment followed by mass spectrometry to identify NP-associated cellular proteins. Here, we demonstrate that NP can interact and colocalize with heterogeneous nuclear ribonucleoprotein (hnRNP) A2/B1 in mammalian cells and that the interaction may occur via direct binding to the glycine-rich domain (GRD) of hnRNP A2/B1. In addition, two residues in the tail loop of NP, F412 and R422, are required for the interaction of hnRNP A2/B1. Because the knockdown of hnRNP A2/B1 expression reduces viral RNP activity, hnRNP A2/B1 may act as a positive regulator in viral RNA synthesis of influenza A virus. More importantly, the findings in this research demonstrate that host proteins can regulate the replication of influenza A virus by interacting with NP.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Virus de la Influenza A/fisiología , Nucleoproteínas/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Animales , Línea Celular , Perros , Técnicas de Silenciamiento del Gen , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Virus de la Influenza A/genética , Unión Proteica , ARN Viral/genética
13.
mBio ; 8(3)2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28611246

RESUMEN

Avian influenza A viruses generally do not replicate efficiently in human cells, but substitution of glutamic acid (Glu, E) for lysine (Lys, K) at residue 627 of avian influenza virus polymerase basic protein 2 (PB2) can serve to overcome host restriction and facilitate human infectivity. Although PB2 residue 627 is regarded as a species-specific signature of influenza A viruses, host restriction factors associated with PB2627E have yet to be fully investigated. We conducted immunoprecipitation, followed by differential proteomic analysis, to identify proteins associating with PB2627K (human signature) and PB2627E (avian signature) of influenza A/WSN/1933(H1N1) virus, and the results indicated that Tu elongation factor, mitochondrial (TUFM), had a higher binding affinity for PB2627E than PB2627K in transfected human cells. Stronger binding of TUFM to avian-signature PB2590G/591Q and PB2627E in the 2009 swine-origin pandemic H1N1 and 2013 avian-origin H7N9 influenza A viruses was similarly observed. Viruses carrying avian-signature PB2627E demonstrated increased replication in TUFM-deficient cells, but viral replication decreased in cells overexpressing TUFM. Interestingly, the presence of TUFM specifically inhibited the replication of PB2627E viruses, but not PB2627K viruses. In addition, enhanced levels of interaction between TUFM and PB2627E were noted in the mitochondrial fraction of infected cells. Furthermore, TUFM-dependent autophagy was reduced in TUFM-deficient cells infected with PB2627E virus; however, autophagy remained consistent in PB2627K virus-infected cells. The results suggest that TUFM acts as a host restriction factor that impedes avian-signature influenza A virus replication in human cells in a manner that correlates with autophagy.IMPORTANCE An understanding of the mechanisms that influenza A viruses utilize to shift host tropism and the identification of host restriction factors that can limit infection are both critical to the prevention and control of emerging viruses that cross species barriers to target new hosts. Using a proteomic approach, we revealed a novel role for TUFM as a host restriction factor that exerts an inhibitory effect on avian-signature PB2627E influenza virus propagation in human cells. We further found that increased TUFM-dependent autophagy correlates with the inhibitory effect on avian-signature influenza virus replication and may serve as a key intrinsic mechanism to restrict avian influenza virus infection in humans. These findings provide new insight regarding the TUFM mitochondrial protein and may have important implications for the development of novel antiviral strategies.


Asunto(s)
Autofagia , Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/fisiología , Proteínas Mitocondriales/metabolismo , Factor Tu de Elongación Peptídica/metabolismo , Replicación Viral , Células A549 , Animales , Humanos , Inmunoprecipitación , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/fisiología , Gripe Humana/virología , Mitocondrias/química , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Factor Tu de Elongación Peptídica/deficiencia , Factor Tu de Elongación Peptídica/genética , Unión Proteica , Proteómica
14.
Nat Commun ; 8(1): 846, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-29018261

RESUMEN

The aim of candidate universal influenza vaccines is to provide broad protection against influenza A and B viruses. Studies have demonstrated that broadly reactive antibodies require Fc-Fc gamma receptor interactions for optimal protection; however, the innate effector cells responsible for mediating this protection remain largely unknown. Here, we examine the roles of alveolar macrophages, natural killer cells, and neutrophils in antibody-mediated protection. We demonstrate that alveolar macrophages play a dominant role in conferring protection provided by both broadly neutralizing and non-neutralizing antibodies in mice. Our data also reveal the potential mechanisms by which alveolar macrophages mediate protection in vivo, namely antibody-induced inflammation and antibody-dependent cellular phagocytosis. This study highlights the importance of innate effector cells in establishing a broad-spectrum antiviral state, as well as providing a better understanding of how multiple arms of the immune system cooperate to achieve an optimal antiviral response following influenza virus infection or immunization.Broadly reactive antibodies that recognize influenza A virus HA can be protective, but the mechanism is not completely understood. Here, He et al. show that the inflammatory response and phagocytosis mediated by the interaction between protective antibodies and macrophages are essential for protection.


Asunto(s)
Anticuerpos Neutralizantes/fisiología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Activación de Macrófagos , Macrófagos Alveolares/fisiología , Células A549 , Animales , Perros , Femenino , Células HEK293 , Hemaglutininas/inmunología , Humanos , Células Asesinas Naturales/fisiología , Células de Riñón Canino Madin Darby , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/fisiología , Infecciones por Orthomyxoviridae/inmunología , Fagocitosis , Receptores de IgG/metabolismo
15.
PLoS One ; 9(12): e115016, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25506939

RESUMEN

The influenza A virus contains 8 segmented genomic RNAs and was considered to encode 10 viral proteins until investigators identified the 11th viral protein, PB1-F2, which uses an alternative reading frame of the PB1 gene. The recently identified PB1-N40, PA-N155 and PA-N182 influenza A proteins have shown the potential for using a leaking ribosomal scanning mechanism to generate novel open reading frames (ORFs). These novel ORFs provide examples of the manner in which the influenza A virus expands its coding capacity by using overlapping reading frames. In this study, we performed a computational search, based on a ribosome scanning mechanism, on all influenza A coding sequences to identify possible forward-reading ORFs that could be translated into novel viral proteins. We specified that the translated products had a prevalence ≥5% to eliminate sporadic ORFs. A total of 1,982 ORFs were thus identified and presented in terms of their locations, lengths and Kozak sequence strengths. We further provided an abridged list of ORFs by requiring every candidate an upstream start codon (within the upstream third of the primary transcript), a strong Kozak consensus sequence and high prevalence (≥95% and ≥50% for in-frame and alternative-frame ORFs, respectively). The PB1-F2, PB1-N40, PA-N155 and PA-N182 proteins all fulfilled our filtering criteria. Subject to these three stringent settings, we additionally named 16 novel ORFs for all influenza A genomes except for HA and NA, for which 43 HA and 11 NA ORFs from their respective subtypes were also recognized.


Asunto(s)
Virus de la Influenza A/genética , Sistemas de Lectura Abierta , Proteínas Virales/genética , Mapeo Cromosómico , Codón Iniciador , Genómica
16.
Virology ; 447(1-2): 32-44, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24210097

RESUMEN

Neuraminidase (NA) is a homotetramer viral surface glycoprotein that is essential for virus release during influenza virus infections. Previous studies have not explored why influenza NA forms a tetramer when the bacterial monomer NA already exhibits excellent NA enzymatic activity levels. In this study, we focused on 28 highly conserved residues among all NA subtypes, identifying 21 of 28 positions as crucial residues for viral survival by using reverse genetics. Maintaining NA enzymatic activity levels is critical and numerous conserved residues were located at the oligomerization interface; however, these mutations did not affect NA enzymatic activity levels or NA cellular localization, but rather affected the stability of NA oligomerization, suggesting that the oligomerization of NA is essential for viral viability. An increased understanding of the biological functions of NA, in particular NA oligomerization, could facilitate an alternative design for antivirals to combat influenza virus infections.


Asunto(s)
Virus de la Influenza A/fisiología , Viabilidad Microbiana , Neuraminidasa/genética , Neuraminidasa/metabolismo , Multimerización de Proteína , Proteínas Virales/genética , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Análisis Mutacional de ADN , Humanos , Virus de la Influenza A/genética , Modelos Moleculares , Datos de Secuencia Molecular , Neuraminidasa/química , Conformación Proteica , Genética Inversa , Alineación de Secuencia , Ensayo de Placa Viral , Proteínas Virales/química
17.
J Ethnopharmacol ; 134(3): 614-23, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21232589

RESUMEN

AIMS OF THE STUDY: Ko-Ken Tang (KKT, aka kakkon-to), a conventional Chinese herbal medicine, has been used for the treatment of the common cold, fever and influenza virus infection. However, the underlying mechanism of its activity against influenza virus infection remains elusive. In this study, the antiviral effect and its underlying mechanism was evaluated, including the investigation of anti-influenza virus activity of KKT on MDCK cells and corresponding mechanism related to phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway and its consecutive viral RNP nuclear export. MATERIALS AND METHODS: The antiviral activity of non-toxic concentration of KKT was examined against various strains of influenza virus and enterovirus 71 by neutralization assay. PI3K/Akt signaling activated by influenza virus was inspected in A549 cells by western blot. Inhibition of influenza polymerase activity by KKT was measured with plasmid-based reverse genetics using primer extension assay and luciferase reporter assay. Inhibition of viral vRNP nuclear export was demonstrated by laser confocal microscopy and interspecies heterokaryon assay. RESULTS: KKT inhibits influenza virus replication but not entry, and it exhibits a broad spectrum inhibitory activity against human influenza A viruses and enterovirus 71. KKT does not inhibit viral polymerase activity but directly blocks the virus-induced phosphatidylinositol 3-kinase/Akt signaling pathway, which in turns causes retention of viral nucleoprotein in the nucleus, thereby interfering with virus propagation. The inhibition by KKT of the nuclear export of viral protein was further confirmed by heterokaryon assay. CONCLUSIONS: The results obtained in this study give scientific support to KKT for the treatment of influenza virus infection. KKT could be of potential use in the management of seasonal pandemic influenza virus infection in addition to other clinically available drugs.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza B/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ribonucleoproteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Secuencia de Bases , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Cartilla de ADN , Perros , Virus de la Influenza A/metabolismo , Virus de la Influenza B/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Transporte de Proteínas
18.
J Med Chem ; 53(4): 1519-33, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-20092255

RESUMEN

By using a cell-based high throughput screening campaign, a novel angelicin derivative 6a was identified to inhibit influenza A (H1N1) virus induced cytopathic effect in Madin-Darby canine kidney cell culture in low micromolar range. Detailed structure-activity relationship studies of 6a revealed that the angelicin scaffold is essential for activity in pharmacophore B, while meta-substituted phenyl/2-thiophene rings are optimal in pharmacophore A and C. The optimized lead 4-methyl-9-phenyl-8-(thiophene-2-carbonyl)-furo[2,3-h]chromen-2-one (8g, IC(50) = 70 nM) showed 64-fold enhanced activity compared to the high throughput screening (HTS) hit 6a. Also, 8g was found effective in case of influenza A (H3N2) and influenza B virus strains similar to approved anti-influenza drug zanamivir (4). Preliminary mechanistic studies suggest that these compounds act as anti-influenza agents by inhibiting ribonucleoprotein (RNP) complex associated activity and have the potential to be developed further, which could form the basis for developing additional defense against influenza pandemics.


Asunto(s)
Antivirales/síntesis química , Furocumarinas/síntesis química , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza B/efectos de los fármacos , Animales , Antivirales/química , Antivirales/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Perros , Descubrimiento de Drogas , Furocumarinas/química , Furocumarinas/farmacología , Humanos , Virus de la Influenza A/fisiología , Virus de la Influenza B/fisiología , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
19.
Emerg Infect Dis ; 12(9): 1353-60, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17073083

RESUMEN

Position-specific entropy profiles created from scanning 306 human and 95 avian influenza A viral genomes showed that 228 of 4591 amino acid residues yielded significant differences between these 2 viruses. We subsequently used 15,785 protein sequences from the National Center for Biotechnology Information (NCBI) to assess the robustness of these signatures and obtained 52 "species-associated" positions. Specific mutations on those points may enable an avian influenza virus to become a human virus. Many of these signatures are found in NP, PA, and PB2 genes (viral ribonucleoproteins [RNPs]) and are mostly located in the functional domains related to RNP-RNP interactions that are important for viral replication. Upon inspecting 21 human-isolated avian influenza viral genomes from NCBI, we found 19 that exhibited > or =1 species-associated residue changes; 7 of them contained > or =2 substitutions. Histograms based on pairwise sequence comparison showed that NP disjointed most between human and avian influenza viruses, followed by PA and PB2.


Asunto(s)
Secuencia de Aminoácidos , Genoma Viral , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Gripe Aviar/virología , Gripe Humana/virología , Animales , Aves/virología , Humanos , Subtipo H5N1 del Virus de la Influenza A/clasificación , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H7N7 del Virus de la Influenza A/clasificación , Subtipo H7N7 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/clasificación , Subtipo H9N2 del Virus de la Influenza A/genética , Datos de Secuencia Molecular , Mutación , Análisis de Secuencia de ADN , Especificidad de la Especie , Proteínas Virales/genética
20.
J Clin Microbiol ; 43(4): 1651-61, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15814980

RESUMEN

A laboratory-based surveillance network of 11 clinical virological laboratories for influenza viruses was established in Taiwan under the coordination of the Center for Disease Control and Prevention (CDC), Taiwan. From October 2000 to March 2004, 3,244 influenza viruses were isolated, including 1,969 influenza A and 1,275 influenza B viruses. The influenza infections usually occurred frequently in winter in the northern hemisphere. However, the influenza seasonality in Taiwan was not clear during the four seasons under investigation. For example, the influenza A viruses peaked during the winters of 2001, 2002, and 2003. However, some isolated peaks were also found in the summer and fall (June to November) of 2001 and 2002. An unusual peak of influenza B also occurred in the summer of 2002 (June to August). Phylogenetic analysis shows that influenza A isolates from the same year were often grouped together. However, influenza B isolates from the year 2002 clustered into different groups, and the data indicate that both B/Victoria/2/87-like and B/Yamagata/16/88-like lineages of influenza B viruses were cocirculating. Sequence comparison of epidemic strains versus vaccine strains shows that many vaccine-like Taiwanese strains were circulating at least 2 years before the vaccine strains were introduced. No clear seasonality of influenza reports in Taiwan occurred in contrast to other more continental regions.


Asunto(s)
Virus de la Influenza A/genética , Virus de la Influenza B/genética , Gripe Humana/epidemiología , Laboratorios , Epidemiología Molecular , Vigilancia de Guardia , Animales , Perros , Humanos , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza B/clasificación , Virus de la Influenza B/aislamiento & purificación , Gripe Humana/virología , Datos de Secuencia Molecular , Filogenia , Análisis de Secuencia de ADN , Taiwán/epidemiología , Virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA