Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Virol ; 91(20)2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28768857

RESUMEN

While the RNA-dependent RNA polymerase L protein of rabies virus (RABV), a member of the genus Lyssavirus of the family Rhabdoviridae, has potential to be a therapeutic target for rabies, the molecular functions of this protein have remained largely unknown. In this study, to obtain a novel experimental tool for molecular function analysis of the RABV L protein, we established by using a reverse genetics approach an L gene-deficient RABV (Nishi-ΔL/Nluc), which infects, propagates, and correspondingly produces NanoLuc luciferase in cultured neuroblastoma cells transfected to express the L protein. trans-Complementation with wild-type L protein, but not that with a functionally defective L protein mutant, efficiently supported luciferase production by Nishi-ΔL/Nluc, confirming its potential for function analysis of the L protein. Based on the findings obtained from comprehensive genetic analyses of L genes from various RABV and other lyssavirus species, we examined the functional importance of a highly conserved L protein region at positions 1914 to 1933 by a trans-complementation assay with Nishi-ΔL/Nluc and a series of L protein mutants. The results revealed that the amino acid sequence at positions 1929 to 1933 (NPYNE) is functionally important, and this was supported by other findings that this sequence is critical for binding of the L protein with its essential cofactor, P protein, and thus also for L protein's RNA polymerase activity. Our findings provide useful information for the development of an anti-RABV drug targeting the L-P protein interaction.IMPORTANCE To the best of our knowledge, this is the first report on the establishment of an L gene-deficient, reporter gene-expressing virus in all species of the order Mononegavirales, also highlighting its applicability to a trans-complementation assay, which is useful for molecular function analyses of their L proteins. Moreover, this study revealed for the first time that the NPYNE sequence at positions 1929 to 1933 in the RABV L protein is important for L protein's interaction with the P protein, consistent with and extending the results of a previous study showing that the P protein-binding domain in the L protein is located in its C-terminal region, at positions 1562 to 2127. This study indicates that the NPYNE sequence is a promising target for the development of an inhibitor of viral RNA synthesis, which has high potential as a therapeutic drug for rabies.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Genes Virales , Virus de la Rabia/enzimología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Animales , Línea Celular , ARN Polimerasas Dirigidas por ADN/química , Prueba de Complementación Genética , Luciferasas/biosíntesis , Luciferasas/genética , Lyssavirus/genética , Mutación , Fosfoproteínas/metabolismo , ARN Viral/genética , Virus de la Rabia/genética , Genética Inversa , Rhabdoviridae/genética , Proteínas Virales/química , Replicación Viral
2.
J Virol ; 90(20): 9394-405, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27512058

RESUMEN

UNLABELLED: Rho GTPases are involved in a variety of cellular activities and are regulated by guanine nucleotide exchange factors and GTPase-activating proteins (GAPs). We found that the activation of Rho GTPases by lysophosphatidic acid promotes the growth of human parainfluenza virus type 2 (hPIV-2). Furthermore, hPIV-2 infection causes activation of RhoA, a Rho GTPase. We hypothesized that Graf1 (also known as ARHGAP26), a GAP, regulates hPIV-2 growth by controlling RhoA signaling. Immunofluorescence analysis showed that hPIV-2 infection altered Graf1 localization from a homogenous distribution within the cytoplasm to granules. Graf1 colocalized with hPIV-2 P, NP, and L proteins. Graf1 interacts with P and V proteins via their N-terminal common region, and the C-terminal Src homology 3 domain-containing region of Graf1 is important for these interactions. In HEK293 cells constitutively expressing Graf1, hPIV-2 growth was inhibited, and RhoA activation was not observed during hPIV-2 infection. In contrast, Graf1 knockdown restored hPIV-2 growth and RhoA activation. Overexpression of hPIV-2 P and V proteins enhanced hPIV-2-induced RhoA activation. These results collectively suggested that hPIV-2 P and V proteins enhanced hPIV-2 growth by binding to Graf1 and that Graf1 inhibits hPIV-2 growth through RhoA inactivation. IMPORTANCE: Robust growth of hPIV-2 requires Rho activation. hPIV-2 infection causes RhoA activation, which is suppressed by Graf1. Graf1 colocalizes with viral RNP (vRNP) in hPIV-2-infected cells. We found that Graf1 interacts with hPIV-2 P and V proteins. We also identified regions in these proteins which are important for this interaction. hPIV-2 P and V proteins enhanced the hPIV-2 growth via binding to Graf1, while Graf1 inhibited hPIV-2 growth through RhoA inactivation.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Virus de la Parainfluenza 2 Humana/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Citoplasma/metabolismo , Citoplasma/virología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Células HeLa , Humanos , Células Vero , Dominios Homologos src/fisiología
3.
J Virol ; 90(18): 8226-37, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27384657

RESUMEN

UNLABELLED: Rabies virus (RABV) P gene mRNA encodes five in-frame start codons, resulting in expression of full-length P protein (P1) and N-terminally truncated P proteins (tPs), designated P2, P3, P4, and P5. Despite the fact that some tPs are known as interferon (IFN) antagonists, the importance of tPs in the pathogenesis of RABV is still unclear. In this study, to examine whether tPs contribute to pathogenesis, we exploited a reverse genetics approach to generate CE(NiP)ΔP2-5, a mutant of pathogenic CE(NiP) in which the P gene was mutated by replacing all of the start codons (AUG) for tPs with AUA. We confirmed that while CE(NiP) expresses detectable levels of P2 and P3, CE(NiP)ΔP2-5 has an impaired ability to express these tPs. After intramuscular inoculation, CE(NiP)ΔP2-5 caused significantly lower morbidity and mortality rates in mice than did CE(NiP), indicating that tPs play a critical role in RABV neuroinvasiveness. Further examinations revealed that this less neuroinvasive phenotype of CE(NiP)ΔP2-5 correlates with its impaired ability to replicate in muscle cells, indicative of the importance of tPs in viral replication in muscle cells. We also demonstrated that CE(NiP)ΔP2-5 infection induced a higher level of Ifn-ß gene expression in muscle cells than did CE(NiP) infection, consistent with the results of an IFN-ß promoter reporter assay suggesting that all tPs function to antagonize IFN induction in muscle cells. Taken together, our findings strongly suggest that tPs promote viral replication in muscle cells through their IFN antagonist activities and thereby support infection of peripheral nerves. IMPORTANCE: Despite the fact that previous studies have demonstrated that P2 and P3 of RABV have IFN antagonist activities, the actual importance of tPs in pathogenesis has remained unclear. Here, we provide the first evidence that tPs contribute to the pathogenesis of RABV, especially its neuroinvasiveness. Our results also show the mechanism underlying the neuroinvasiveness driven by tPs, highlighting the importance of their IFN antagonist activities, which support viral replication in muscle cells.


Asunto(s)
Factores Inmunológicos/metabolismo , Interferón beta/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Isoformas de Proteínas/metabolismo , Virus de la Rabia/patogenicidad , Rabia/patología , Proteínas Estructurales Virales/metabolismo , Animales , Encéfalo/virología , Línea Celular , Cricetinae , Factores Inmunológicos/genética , Inyecciones Intramusculares , Ratones , Chaperonas Moleculares , Músculos/virología , Fosfoproteínas/genética , Isoformas de Proteínas/genética , Rabia/virología , Virus de la Rabia/genética , Genética Inversa , Análisis de Supervivencia , Proteínas Estructurales Virales/genética , Virulencia , Replicación Viral
4.
J Virol ; 90(17): 7640-6, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27279623

RESUMEN

UNLABELLED: Receptor destruction has been considered one of the mechanisms of homologous Sendai virus (SeV) interference. However, direct evidence of receptor destruction upon virus infection and its relevance to interference is missing. To investigate a precise mechanism of homologous interference, we established SeV persistently infected cells. The persistently infected cells inhibited superinfection by homologous SeV but supported replication of human parainfluenza virus 2 (hPIV2) and influenza A virus (IAV). We confirmed that SeV particles could not attach to or penetrate the infected cells and that the hemagglutinin-neuraminidase (HN) protein of SeV was involved in the interference. Lectin blot assays showed that the α2,3-linked sialic acids were specifically reduced in the SeV-infected cells, but the level of α2,6-linked sialic acids had not changed. As infection with IAV removed both α2,3- and α2,6-linked sialic acids, especially α2,3-linked sialic acids, IAV-infected cells inhibited superinfection of SeV. These results provide concrete evidence that destruction of the specific SeV receptor, α2,3-linked sialic acids, is relevant to homologous interference by SeV. IMPORTANCE: Viral interference is a classically observed phenomenon, but the precise mechanism is not clear. Using SeV interference, we provide concrete evidence that reduction of the α2,3-linked sialic acid receptor by the HN of SeV is closely related with viral interference. Since SeV infection resulted in decrease of only α2,3-linked sialic acids, IAV, which also utilized α2,6-linked sialic acids to initiate infection, superinfected the SeV-infected cells. In contrast, SeV could not superinfect the IAV-infected cells because both α2,3- and α2,6-linked sialic acids were removed. These results indicate that receptor destruction critically contributes to viral interference.


Asunto(s)
Proteína HN/metabolismo , Receptores Virales/metabolismo , Virus Sendai/enzimología , Virus Sendai/fisiología , Interferencia Viral , Animales , Línea Celular , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Parainfluenza 2 Humana/crecimiento & desarrollo , Ácidos Siálicos/metabolismo
5.
J Gen Virol ; 97(7): 1520-1530, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27072881

RESUMEN

Gene expression of paramyxoviruses is regulated by genome-encoded cis-acting elements; however, whether all the required elements for viral growth have been identified is not clear. Using a mini-replicon system, it has been shown that human parainfluenza virus type 2 (hPIV2) polymerase can recognize the promoter elements of parainfluenza virus type 5 (PIV5), but reporter activity is lower in this case. We constructed a series of luciferase-encoding chimeric PIV2/5 mini-genomes that are basically hPIV2, but whose leader (le), mRNA start signal and trailer sequence are partially replaced with those of PIV5. Studies of the chimeric PIV2/5 mini-replicons demonstrated that replacement of hPIV2 le with PIV5 le results in remarkably weak luciferase expression. Further mutagenesis identified the responsible region as positions 25-30 of the PIV5 le. Using recombinant hPIV2, the impact of this region on viral life cycles was assessed. Insertion of the mutation at this region facilitated viral growth, genomic replication and mRNA transcription at the early stage of infection, which elicited severe cell damage. In contrast, at the late infection stage it caused a reduction in viral transcription. Here, we identify a novel cis-acting element in the internal region of an le sequence that is involved in the regulation of polymerase, and which contributes to maintaining a balance between viral growth and cytotoxicity.


Asunto(s)
Regiones no Traducidas 5'/genética , Quimera/genética , Virus de la Parainfluenza 2 Humana/genética , Virus de la Parainfluenza 5/genética , Regiones Promotoras Genéticas/genética , ARN Polimerasa Dependiente del ARN/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Secuencia de Bases , Línea Celular Tumoral , Genoma Viral/genética , Células HeLa , Humanos , Virus de la Parainfluenza 2 Humana/crecimiento & desarrollo , Virus de la Parainfluenza 5/crecimiento & desarrollo , Infecciones por Paramyxoviridae/virología , ARN Viral/genética , Replicón/genética , Transcripción Genética/genética , Replicación Viral/genética
6.
J Virol ; 90(1): 444-56, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491155

RESUMEN

UNLABELLED: Over the past 2 decades, several novel influenza virus proteins have been identified that modulate viral infections in vitro and/or in vivo. The PB2 segment, which is one of the longest influenza A virus segments, is known to encode only one viral protein, PB2. In the present study, we used reverse transcription-PCR (RT-PCR) targeting viral mRNAs transcribed from the PB2 segment to look for novel viral proteins encoded by spliced mRNAs. We identified a new viral protein, PB2-S1, encoded by a novel spliced mRNA in which the region corresponding to nucleotides 1513 to 1894 of the PB2 mRNA is deleted. PB2-S1 was detected in virus-infected cells and in cells transfected with a protein expression plasmid encoding PB2. PB2-S1 localized to mitochondria, inhibited the RIG-I-dependent interferon signaling pathway, and interfered with viral polymerase activity (dependent on its PB1-binding capability). The nucleotide sequences around the splicing donor and acceptor sites for PB2-S1 were highly conserved among pre-2009 human H1N1 viruses but not among human H1N1pdm and H3N2 viruses. PB2-S1-deficient viruses, however, showed growth kinetics in MDCK cells and virulence in mice similar to those of wild-type virus. The biological significance of PB2-S1 to the replication and pathogenicity of seasonal H1N1 influenza A viruses warrants further investigation. IMPORTANCE: Transcriptome analysis of cells infected with influenza A virus has improved our understanding of the host response to viral infection, because such analysis yields considerable information about both in vitro and in vivo viral infections. However, little attention has been paid to transcriptomes derived from the viral genome. Here we focused on the splicing of mRNA expressed from the PB2 segment and identified a spliced viral mRNA encoding a novel viral protein. This result suggests that other, as yet unidentified viral proteins encoded by spliced mRNAs could be expressed in virus-infected cells. A viral transcriptome including the viral spliceosome should be evaluated to gain new insights into influenza virus infection.


Asunto(s)
Perfilación de la Expresión Génica , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Isoformas de Proteínas/genética , Empalme del ARN , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/antagonistas & inhibidores , Perros , Células Epiteliales/virología , Femenino , Expresión Génica , Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Mitocondrias/química , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Isoformas de Proteínas/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Receptores Inmunológicos , Proteínas Virales/metabolismo , Virulencia
7.
Med Microbiol Immunol ; 205(3): 209-18, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26582554

RESUMEN

It has been reported that dual or multiple viruses can coinfect epithelial cells of the respiratory tract. However, little has been reported on in vitro interactions of coinfected viruses. To explore how coinfection of different viruses affects their biological property, we examined growth of influenza A virus (IAV) and human parainfluenza virus type 2 (hPIV2) during coinfection of Vero cells. We found that IAV growth was enhanced by coinfection with hPIV2. The enhanced growth of IAV was not reproduced by coinfection with an hPIV2 mutant with reduced cell fusion activity, or by ectopic expression of the V protein of hPIV2. In contrast, induction of cell fusion by ectopic expression of the hPIV2 HN and F proteins augments IAV growth. hPIV2 coinfection supported IAV growth in cells originated from the respiratory epithelium. The enhancement correlated closely with cell fusion ability of hPIV2 in those cells. These results indicate that cell fusion induced by hPIV2 infection is beneficial to IAV replication and that enhanced viral replication by coinfection with different viruses can modify their pathological consequences.


Asunto(s)
Virus de la Influenza A/crecimiento & desarrollo , Interacciones Microbianas , Virus de la Parainfluenza 2 Humana/crecimiento & desarrollo , Animales , Fusión Celular , Chlorocebus aethiops , Células Epiteliales/virología , Proteína HN/genética , Proteína HN/metabolismo , Virus de la Parainfluenza 2 Humana/genética , Células Vero , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Cultivo de Virus
8.
Microbiol Immunol ; 59(11): 676-83, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26446904

RESUMEN

Gene expression of nonsegmented negative-strand RNA viruses (nsNSVs) such as parainfluenza viruses requires the RNA synthesis activity of their polymerase L protein; however, the detailed mechanism of this process is poorly understood. In this study, a parainfluenza minireplicon assay expressing secretory Gaussia luciferase (Gluc) was established to analyze large protein (L) activity. Measurement of Gluc expression in the culture medium of cells transfected with the minigenome and viral polymerase components enabled quick and concise calculation of L activity. By comparing the amino acid sequences in conserved region III (CRIII), a putative polymerase-active domain of the L protein, two strictly conserved aspartates were identified in all families of nsNSV. A series of L mutants from human parainfluenza virus type 2 and parainfluenza virus type 5 showed that these aspartates are necessary for reporter gene expression. It was also confirmed that these aspartates are important for the production of viral mRNA and antigenome cRNA, but not for a polymerase-complex formation. These findings suggest that these two aspartates are key players in the nucleotidyl transfer reaction using two metal ions.


Asunto(s)
Ácido Aspártico/genética , Copépodos/enzimología , Luciferasas/metabolismo , Virus de la Parainfluenza 2 Humana/enzimología , Virus de la Parainfluenza 2 Humana/metabolismo , Transfección/métodos , Proteínas Virales/genética , Proteínas Virales/fisiología , Replicación Viral/fisiología , Animales , Células Cultivadas , Secuencia Conservada , Humanos
9.
Cell Host Microbe ; 16(6): 795-805, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25464832

RESUMEN

Host factors required for viral replication are ideal drug targets because they are less likely than viral proteins to mutate under drug-mediated selective pressure. Although genome-wide screens have identified host proteins involved in influenza virus replication, limited mechanistic understanding of how these factors affect influenza has hindered potential drug development. We conducted a systematic analysis to identify and validate host factors that associate with influenza virus proteins and affect viral replication. After identifying over 1,000 host factors that coimmunoprecipitate with specific viral proteins, we generated a network of virus-host protein interactions based on the stage of the viral life cycle affected upon host factor downregulation. Using compounds that inhibit these host factors, we validated several proteins, notably Golgi-specific brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF1) and JAK1, as potential antiviral drug targets. Thus, virus-host interactome screens are powerful strategies to identify targetable host factors and guide antiviral drug development.


Asunto(s)
Antivirales/farmacología , Gripe Humana/metabolismo , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/metabolismo , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas Virales/metabolismo , Evaluación Preclínica de Medicamentos , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Gripe Humana/tratamiento farmacológico , Gripe Humana/genética , Gripe Humana/virología , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Orthomyxoviridae/genética , Unión Proteica/efectos de los fármacos , Proteínas Virales/genética
10.
J Virol ; 87(21): 11316-22, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23926345

RESUMEN

The influenza A virus genome comprises eight single-stranded negative-sense RNA segments (vRNAs). All eight vRNAs are selectively packaged into each progeny virion via so-called segment-specific genome-packaging signal sequences that are located in the noncoding and terminal coding regions of both the 3' and the 5' ends of the vRNAs. However, it remains unclear how these signals ensure that eight different vRNAs are packaged. Here, by using a reverse genetics system, we demonstrated that, in the absence of the other seven vRNAs, a recombinant NP vRNA bearing only a reporter gene flanked by the noncoding NP regions was incorporated into virus-like particles (VLPs) as efficiently as a recombinant NP vRNA bearing the reporter gene flanked by the complete NP packaging signals (i.e., the noncoding sequences and the terminal coding regions). Viruses that comprised a recombinant NP vRNA whose packaging signal was disrupted, and the remaining seven authentic vRNAs, did not undergo multiple cycles of replication; however, a recombinant NP vRNA with only the noncoding regions was readily incorporated into VLPs, suggesting that the packaging signal as currently defined is not necessarily essential for the packaging of the vRNA in which it resides; rather, it is required for the packaging of the full set of vRNAs. We propose that the 3' and 5' noncoding regions of each vRNA bear a virion incorporation signal for that vRNA and that the terminal coding regions serve as a bundling signal that ensures the incorporation of the complete set of eight vRNAs into the virion.


Asunto(s)
Virus de la Influenza A/fisiología , ARN Viral/genética , ARN Viral/metabolismo , Ensamble de Virus , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Animales , Línea Celular , Genes Reporteros , Genoma Viral , Humanos , Genética Inversa
11.
J Virol ; 87(9): 4861-71, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23408626

RESUMEN

The influenza A virus NS1 protein affects virulence through several mechanisms, including the host's innate immune response and various signaling pathways. Highly pathogenic avian influenza (HPAI) viruses of the H5N1 subtype continue to evolve through reassortment and mutations. Our recent phylogenetic analysis identified a group of HPAI H5N1 viruses with two characteristic mutations in NS1: the avian virus-type PDZ domain-binding motif ESEV (which affects virulence) was replaced with ESKV, and NS1-138F (which is highly conserved among all influenza A viruses and may affect the activation of the phosphatidylinositol 3-kinase [PI3K]/Akt signaling pathway) was replaced with NS1-138Y. Here, we show that an HPAI H5N1 virus (A/duck/Hunan/69/2004) encoding NS1-ESKV and NS1-138Y was confined to the respiratory tract of infected mice, whereas a mutant encoding NS1-ESEV and NS1-138F caused systemic infection and killed mice more efficiently. Mutation of either one of these sites had small effects on virulence. In addition, we found that the amino acid at NS1-138 affected not only the induction of the PI3K/Akt pathway but also the interaction of NS1 with cellular PDZ domain proteins. Similarly, the mutation in the PDZ domain-binding motif of NS1 altered its binding to cellular PDZ domain proteins and affected Akt phosphorylation. These findings suggest a functional interplay between the mutations at NS1-138 and NS1-229 that results in a synergistic effect on influenza virulence.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Femenino , Células HEK293 , Humanos , Subtipo H5N1 del Virus de la Influenza A/química , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Humana/enzimología , Gripe Humana/genética , Ratones , Ratones Endogámicos BALB C , Dominios PDZ , Unión Proteica , Estructura Terciaria de Proteína , Proteínas no Estructurales Virales/genética , Virulencia
12.
Sci Rep ; 3: 1106, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23346363

RESUMEN

Although two classes of antivirals, NA inhibitors and M2 ion channel blockers, are licensed for influenza treatment, dual resistant mutants, including highly pathogenic H5N1 viruses, have appeared. Alternative treatment options are, therefore, needed. Influenza A viral RNA (vRNA) transcription/replication is a promising target for antiviral development, since it is essential for virus replication. Accordingly, an efficient and reliable method to identify vRNA transcription/replication inhibitors is desirable. Here, we developed a cell-based screening system by establishing a cell line that stably expresses influenza viral ribonucleoprotein complex (vRNP). Compound library screening using this cell line allowed us to identify a compound that inhibits vRNA transcription/replication by using reporter protein expression from virus-like RNA as a readout and virus replication in vitro. vRNP-expressing cells have potential as a simple and convenient high-throughput screening (HTS) system, and, thus, are promising to identify vRNA transcription/replication inhibitors for various RNA viruses, especially for primary screens.


Asunto(s)
Antivirales/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , ARN Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Perros , Evaluación Preclínica de Medicamentos/métodos , Vectores Genéticos/genética , Células HEK293 , Humanos , Virus de la Influenza A/genética , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Células de Riñón Canino Madin Darby , Virus ARN/efectos de los fármacos , Virus ARN/genética , ARN Viral/genética , Partículas Ribonucleoproteicas en Bóveda/efectos de los fármacos , Partículas Ribonucleoproteicas en Bóveda/genética , Proteínas Virales/genética , Replicación Viral/genética
13.
J Virol ; 87(4): 2226-33, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221570

RESUMEN

A novel swine-origin H1N1 influenza virus [A(H1N1)pdm09 virus] caused the 2009 influenza pandemic. Most patients exhibited mild symptoms similar to seasonal influenza, but some experienced severe clinical signs and, in the worst cases, died. Such differences in symptoms are generally associated with preexisting medical conditions, but recent reports indicate the possible involvement of viral factors in clinical severity. To better understand the mechanism of pathogenicity of the A(H1N1)pdm09 virus, here, we compared five viruses that are genetically similar but were isolated from patients with either severe or mild symptoms. In a mouse model, A/Norway/3487/2009 (Norway3487) virus exhibited greater pathogenicity than did A/Osaka/164/2009 (Osaka164) virus. By exploiting reassortant viruses between these two viruses, we found that viruses possessing the hemagglutinin (HA) gene of Norway3487 in the genetic background of Osaka164 were more pathogenic in mice than other reassortant viruses, indicating a role for HA in the high virulence of Norway3487 virus. Intriguingly, a virus possessing HA, NA, and NS derived from Norway3487 exhibited greater pathogenicity in mice in concert with PB2 and PB1 derived from Osaka164 than did the parental Norway3487 virus. These findings demonstrate that reassortment between A(H1N1)pdm09 viruses can lead to increased pathogenicity and highlight the need for continued surveillance of A(H1N1)pdm09 viruses.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Virus Reordenados/genética , Virus Reordenados/aislamiento & purificación , Virus Reordenados/patogenicidad , Proteínas Virales/genética , Virulencia , Factores de Virulencia/genética
15.
J Virol ; 86(18): 10200-2, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22740412

RESUMEN

By using two reporter protein-encoding virus-like RNAs derived from identical viral RNA (vRNA) segments, we assessed their incorporation efficiency into single progeny virions. Most plaques formed by the recombinant viruses that were generated in cells positive for both reporter genes expressed only one or the other protein. These results suggest that two virus-like RNAs encoding different reporter proteins compete for incorporation into virions, and individual influenza virions incorporate single, but not multiple, copies of homologous vRNA segments.


Asunto(s)
Genes Virales , Virus de la Influenza A/genética , Animales , Línea Celular , Perros , Genes Reporteros , Humanos , Virus de la Influenza A/fisiología , Neuraminidasa/genética , Plásmidos/genética , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , Proteínas Recombinantes/genética , Transfección , Proteínas Virales/genética , Virión/genética , Virión/fisiología , Ensamble de Virus/genética , Ensamble de Virus/fisiología
16.
Proc Natl Acad Sci U S A ; 109(12): 4615-20, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22393008

RESUMEN

The identification of host factors involved in virus replication is important to understand virus life cycles better. Accordingly, we sought host factors that interact with the influenza viral nonstructural protein 2 by using coimmunoprecipitation followed by mass spectrometry. Among proteins associating with nonstructural protein 2, we focused on the ß subunit of the F1Fo-ATPase, which received a high probability score in our mass spectrometry analysis. The siRNA-mediated down-regulation of the ß subunit of the F1Fo-ATPase reduced influenza virion formation and virus growth in cell culture. We further found that efficient influenza virion formation requires the ATPase activity of F1Fo-ATPase and that plasma membrane-associated, but not mitochondrial, F1Fo-ATPase is important for influenza virion formation and budding. Hence, our data identify plasma membrane-associated F1Fo-ATPase as a critical host factor for efficient influenza virus replication.


Asunto(s)
Membrana Celular/metabolismo , Orthomyxoviridae/metabolismo , ATPasas de Translocación de Protón/química , Proteínas no Estructurales Virales/química , Liberación del Virus , Células HEK293 , Humanos , Gripe Humana/virología , Espectrometría de Masas/métodos , Modelos Biológicos , Modelos Estadísticos , Plásmidos/metabolismo , Proteómica/métodos , ATPasas de Translocación de Protón/metabolismo , ARN Interferente Pequeño/metabolismo , Replicación Viral
17.
J Virol ; 85(16): 8449-52, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21680507

RESUMEN

Reassortment is important for influenza virus evolution and the generation of novel viruses with pandemic potential; however, the factors influencing reassortment are still poorly understood. Here, using reverse genetics and a replicon assay, we demonstrated that a mixed polymerase complex containing a pandemic (H1N1) 2009 influenza virus PB2 on a seasonal H1N1 virus background has reduced polymerase activity, leading to impaired virus viability. Adaptation of viruses containing the mixed polymerase complex resulted in compensatory mutations in PB1. Taken together, our results identify the cooperation between PB2 and PB1 as an important restricting factor for reassortment of influenza viruses.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H1N1 del Virus de la Influenza A/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Virus Reordenados/genética , Proteínas Virales/metabolismo , Humanos , Gripe Humana/virología , ARN Polimerasa Dependiente del ARN/genética , Virus Reordenados/enzimología , Ribonucleoproteínas/genética , Proteínas Virales/genética
18.
J Virol Methods ; 173(1): 1-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21185869

RESUMEN

Real-time RT-PCR is used to quantify individual influenza viral RNAs. However, conventional real-time RT-PCR, using strand-specific primers, has been shown to produce not only the anticipated strand-specific products, but also substantial amounts of non-strand-specific products, indicating lack of specificity. Therefore, in this study, a novel strand-specific real-time RT-PCR method was established to quantify the three types of influenza viral RNA (vRNA, cRNA, and mRNA) separately. This method is based on reverse transcription using tagged primers to add a 'tag' sequence at the 5' end and the hot-start method. Real-time PCR using the 'tag' portion as the forward primer and a segment-specific reverse primer ensured the specificity for quantifying the three types of RNA. Using this method, specific target RNA was detected at 100-100,000-folds higher level than other types of RNA. This method was also used to evaluate the vRNA, cRNA, and mRNA levels of segments 5 and 6 in MDCK cells infected with influenza A virus at different time point post-infections. The cRNA level was 1/10 to 1/100 lower than that of the vRNA and mRNA. Moreover, different dynamics of vRNA, cRNA, and mRNA synthesis were observed; the copy number of the vRNA gradually increased throughout the infection, the cRNA increased and then plateaued, while the mRNA increased and then decreased. This novel method thus provides data critical for understanding the influenza virus life cycle, including transcription, replication, and genome incorporation into virions.


Asunto(s)
Virus de la Influenza A/genética , ARN Complementario/aislamiento & purificación , ARN Mensajero/aislamiento & purificación , ARN Viral/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Virología/métodos , Animales , Línea Celular , Cartilla de ADN/genética , Perros , ARN Complementario/genética , ARN Mensajero/genética , ARN Viral/genética , Sensibilidad y Especificidad
19.
Nature ; 467(7317): 859-62, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20944748

RESUMEN

Herpes simplex virus-1 (HSV-1), the prototype of the α-herpesvirus family, causes life-long infections in humans. Although generally associated with various mucocutaneous diseases, HSV-1 is also involved in lethal encephalitis. HSV-1 entry into host cells requires cellular receptors for both envelope glycoproteins B (gB) and D (gD). However, the gB receptors responsible for its broad host range in vitro and infection of critical targets in vivo remain unknown. Here we show that non-muscle myosin heavy chain IIA (NMHC-IIA), a subunit of non-muscle myosin IIA (NM-IIA), functions as an HSV-1 entry receptor by interacting with gB. A cell line that is relatively resistant to HSV-1 infection became highly susceptible to infection by this virus when NMHC-IIA was overexpressed. Antibody to NMHC-IIA blocked HSV-1 infection in naturally permissive target cells. Furthermore, knockdown of NMHC-IIA in the permissive cells inhibited HSV-1 infection as well as cell-cell fusion when gB, gD, gH and gL were coexpressed. Cell-surface expression of NMHC-IIA was markedly and rapidly induced during the initiation of HSV-1 entry. A specific inhibitor of myosin light chain kinase, which regulates NM-IIA by phosphorylation, reduced the redistribution of NMHC-IIA as well as HSV-1 infection in cell culture and in a murine model for herpes stromal keratitis. NMHC-IIA is ubiquitously expressed in various human tissues and cell types and, therefore, is implicated as a functional gB receptor that mediates broad HSV-1 infectivity both in vitro and in vivo. The identification of NMHC-IIA as an HSV-1 entry receptor and the involvement of NM-IIA regulation in HSV-1 infection provide an insight into HSV-1 entry and identify new targets for antiviral drug development.


Asunto(s)
Herpesvirus Humano 1/fisiología , Miosina Tipo IIA no Muscular/metabolismo , Receptores Virales/metabolismo , Adsorción , Animales , Azepinas/farmacología , Células CHO , Fusión Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HL-60 , Herpes Simple/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/metabolismo , Humanos , Ratones , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Naftalenos/farmacología , Miosina Tipo IIA no Muscular/deficiencia , Miosina Tipo IIA no Muscular/genética , Temperatura , Regulación hacia Arriba , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos
20.
PLoS Pathog ; 6(9): e1001106, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20862325

RESUMEN

Highly pathogenic H5N1 influenza A viruses have spread across Asia, Europe, and Africa. More than 500 cases of H5N1 virus infection in humans, with a high lethality rate, have been reported. To understand the molecular basis for the high virulence of H5N1 viruses in mammals, we tested the virulence in ferrets of several H5N1 viruses isolated from humans and found A/Vietnam/UT3062/04 (UT3062) to be the most virulent and A/Vietnam/UT3028/03 (UT3028) to be avirulent in this animal model. We then generated a series of reassortant viruses between the two viruses and assessed their virulence in ferrets. All of the viruses that possessed both the UT3062 hemagglutinin (HA) and nonstructural protein (NS) genes were highly virulent. By contrast, all those possessing the UT3028 HA or NS genes were attenuated in ferrets. These results demonstrate that the HA and NS genes are responsible for the difference in virulence in ferrets between the two viruses. Amino acid differences were identified at position 134 of HA, at positions 200 and 205 of NS1, and at positions 47 and 51 of NS2. We found that the residue at position 134 of HA alters the receptor-binding property of the virus, as measured by viral elution from erythrocytes. Further, both of the residues at positions 200 and 205 of NS1 contributed to enhanced type I interferon (IFN) antagonistic activity. These findings further our understanding of the determinants of pathogenicity of H5N1 viruses in mammals.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Infecciones por Orthomyxoviridae/virología , Proteínas no Estructurales Virales/genética , Virulencia/fisiología , Animales , Células Cultivadas , Perros , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Gripe Humana/genética , Gripe Humana/patología , Interferones/metabolismo , Riñón/citología , Riñón/metabolismo , Riñón/virología , Pulmón/citología , Pulmón/metabolismo , Pulmón/virología , Masculino , Visón , Mutación/genética , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Proteínas no Estructurales Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...