Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Vet Res ; 55(1): 36, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38520035

RESUMEN

Equine influenza virus (EIV) remains a threat to horses, despite the availability of vaccines. Strategies to monitor the virus and prevent potential vaccine failure revolve around serological assays, RT-qPCR amplification, and sequencing the viral hemagglutinin (HA) and neuraminidase (NA) genes. These approaches overlook the contribution of other viral proteins in driving virulence. This study assesses the potential of long-read nanopore sequencing for fast and precise sequencing of circulating equine influenza viruses. Therefore, two French Florida Clade 1 strains, including the one circulating in winter 2018-2019 exhibiting more pronounced pathogenicity than usual, as well as the two currently OIE-recommended vaccine strains, were sequenced. Our results demonstrated the reliability of this sequencing method in generating accurate sequences. Sequence analysis of HA revealed a subtle antigenic drift in the French EIV strains, with specific substitutions, such as T163I in A/equine/Paris/1/2018 and the N188T mutation in post-2015 strains; both substitutions were in antigenic site B. Antigenic site E exhibited modifications in post-2018 strains, with the N63D substitution. Segment 2 sequencing also revealed that the A/equine/Paris/1/2018 strain encodes a longer variant of the PB1-F2 protein when compared to other Florida clade 1 strains (90 amino acids long versus 81 amino acids long). Further biological and biochemistry assays demonstrated that this PB1-F2 variant has enhanced abilities to abolish the mitochondrial membrane potential ΔΨm and permeabilize synthetic membranes. Altogether, our results highlight the interest in rapidly characterizing the complete genome of circulating strains with next-generation sequencing technologies to adapt vaccines and identify specific virulence markers of EIV.


Asunto(s)
Enfermedades de los Caballos , Subtipo H3N8 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Vacunas , Animales , Aminoácidos/genética , Genómica , Caballos , Subtipo H3N8 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/veterinaria , Reproducibilidad de los Resultados , Análisis de Secuencia/veterinaria , Factores de Virulencia
2.
Front Microbiol ; 13: 915202, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36386681

RESUMEN

The ongoing COVID-19 pandemic continues to pose a need for new and efficient therapeutic strategies. We explored antisense therapy using oligonucleotides targeting the severe acute respiratory syndrome coronavirus (SARS-CoV-2) genome. We predicted in silico four antisense oligonucleotides (ASO gapmers with 100% PTO linkages and LNA modifications at their 5' and 3'ends) targeting viral regions ORF1a, ORF1b, N and the 5'UTR of the SARS-CoV-2 genome. Efficiency of ASOs was tested by transfection in human ACE2-expressing HEK-293T cells and monkey VeroE6/TMPRSS2 cells infected with SARS-CoV-2. The ORF1b-targeting ASO was the most efficient, with a 71% reduction in the number of viral genome copies. N- and 5'UTR-targeting ASOs also significantly reduced viral replication by 55 and 63%, respectively, compared to non-related control ASO (ASO-C). Viral titration revealed a significant decrease in SARS-CoV-2 multiplication both in culture media and in cells. These results show that anti-ORF1b ASO can specifically reduce SARS-CoV-2 genome replication in vitro in two different cell infection models. The present study presents proof-of concept of antisense oligonucleotide technology as a promising therapeutic strategy for COVID-19.

3.
PLoS Pathog ; 18(9): e1010799, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36067253

RESUMEN

The binding of the SARS-CoV-2 spike to angiotensin-converting enzyme 2 (ACE2) promotes virus entry into the cell. Targeting this interaction represents a promising strategy to generate antivirals. By screening a phage-display library of biosynthetic protein sequences build on a rigid alpha-helicoidal HEAT-like scaffold (named αReps), we selected candidates recognizing the spike receptor binding domain (RBD). Two of them (F9 and C2) bind the RBD with affinities in the nM range, displaying neutralisation activity in vitro and recognizing distinct sites, F9 overlapping the ACE2 binding motif. The F9-C2 fusion protein and a trivalent αRep form (C2-foldon) display 0.1 nM affinities and EC50 of 8-18 nM for neutralization of SARS-CoV-2. In hamsters, F9-C2 instillation in the nasal cavity before or during infections effectively reduced the replication of a SARS-CoV-2 strain harbouring the D614G mutation in the nasal epithelium. Furthermore, F9-C2 and/or C2-foldon effectively neutralized SARS-CoV-2 variants (including delta and omicron variants) with EC50 values ranging from 13 to 32 nM. With their high stability and their high potency against SARS-CoV-2 variants, αReps provide a promising tool for SARS-CoV-2 therapeutics to target the nasal cavity and mitigate virus dissemination in the proximal environment.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Tratamiento Farmacológico de COVID-19 , Proteínas Recombinantes de Fusión , Glicoproteína de la Espiga del Coronavirus , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/química , Antivirales/farmacología , Humanos , Peptidil-Dipeptidasa A/metabolismo , Unión Proteica , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
4.
Viruses ; 14(3)2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35336925

RESUMEN

Influenza virus transcription is catalyzed by the viral RNA-polymerase (FluPol) through a cap-snatching activity. The snatching of the cap of cellular mRNA by FluPol is preceded by its binding to the flexible C-terminal domain (CTD) of the RPB1 subunit of RNA-polymerase II (Pol II). To better understand how FluPol brings the 3'-end of the genomic RNAs in close proximity to the host-derived primer, we hypothesized that FluPol may recognize additional Pol II subunits/domains to ensure cap-snatching. Using binary complementation assays between the Pol II and influenza A FluPol subunits and their structural domains, we revealed an interaction between the N-third domain of PB2 and RPB4. This interaction was confirmed by a co-immunoprecipitation assay and was found to occur with the homologous domains of influenza B and C FluPols. The N-half domain of RPB4 was found to be critical in this interaction. Punctual mutants generated at conserved positions between influenza A, B, and C FluPols in the N-third domain of PB2 exhibited strong transcriptional activity defects. These results suggest that FluPol interacts with several domains of Pol II (the CTD to bind Pol II), initiating host transcription and a second transcription on RPB4 to locate FluPol at the proximity of the 5'-end of nascent host mRNA.


Asunto(s)
Gripe Humana , Orthomyxoviridae , Humanos , Orthomyxoviridae/genética , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Viral/genética , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/genética , Transcripción Viral , Replicación Viral
5.
Proteins ; 90(3): 848-857, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34779026

RESUMEN

We introduce multiple interface string alignment (MISA), a visualization tool to display coherently various sequence and structure based statistics at protein-protein interfaces (SSE elements, buried surface area, ΔASA , B factor values, etc). The amino acids supporting these annotations are obtained from Voronoi interface models. The benefit of MISA is to collate annotated sequences of (homologous) chains found in different biological contexts, that is, bound with different partners or unbound. The aggregated views MISA/SSE, MISA/BSA, MISA/ΔASA, and so forth, make it trivial to identify commonalities and differences between chains, to infer key interface residues, and to understand where conformational changes occur upon binding. As such, they should prove of key relevance for knowledge-based annotations of protein databases such as the Protein Data Bank. Illustrations are provided on the receptor binding domain of coronaviruses, in complex with their cognate partner or (neutralizing) antibodies. MISA computed with a minimal number of structures complement and enrich findings previously reported. The corresponding package is available from the Structural Bioinformatics Library (http://sbl.inria.frand https://sbl.inria.fr/doc/Multiple_interface_string_alignment-user-manual.html).


Asunto(s)
Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/química , Secuencia de Aminoácidos , Biología Computacional , Bases de Datos de Proteínas , Modelos Moleculares , Unión Proteica , Conformación Proteica , Análisis de Secuencia de Proteína , Interfaz Usuario-Computador
6.
J Mol Biol ; 432(10): 3353-3359, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32222384

RESUMEN

Here, we describe the crystal structures of two distinct isoforms of ligand-free human karyopherin RanBP5 and investigate its global propensity to interact with influenza A virus polymerase. Our results confirm the general architecture and mechanism of the IMB3 karyopherin-ß subfamily whilst also highlighting differences with the yeast orthologue Kap121p. Moreover, our results provide insight into the structural flexibility of ß-importins in the unbound state. Based on docking of a nuclear localisation sequence, point mutations were designed, which suppress influenza PA-PB1 subcomplex binding to RanBP5 in a binary protein complementation assay.


Asunto(s)
Núcleo Celular/metabolismo , Virus de la Influenza A/enzimología , Mutación Puntual , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo , beta Carioferinas/química , Sitios de Unión , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Transporte de Proteínas , beta Carioferinas/genética
7.
J Gen Virol ; 100(2): 133-134, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30484763

RESUMEN

Picobirnaviridae is a family of viruses with bi-segmented (rarely unsegmented) dsRNA genomes comprising about 4.4 kbp in total, with small, non-enveloped spherical virions. The family includes one genus (Picobirnavirus) grouping three genetic clusters with high sequence variability, two defined by viruses infecting vertebrates and a third with viruses found in invertebrates. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the taxonomy of Picobirnaviridae, which is available at www.ictv.global/report/picobirnaviridae.


Asunto(s)
Picobirnavirus/clasificación , Picobirnavirus/genética , Enfermedades de los Animales/virología , Animales , Invertebrados/virología , Picobirnavirus/aislamiento & purificación , Infecciones por Virus ARN/veterinaria , Vertebrados/virología
8.
J Gen Virol ; 100(1): 5-6, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30484762

RESUMEN

Birnaviridae is a family of viruses with bi-segmented dsRNA genomes totalling about 6 kbp forming icosahedral, non-enveloped virions. The family includes four genera, members of three of which (Aquabirnavirus, Avibirnavirus and Blosnavirus) infect vertebrates (excluding mammals), whereas members of the fourth genus (Entomobirnavirus) infect insects. Each genus includes 1-3 species. Infectious pancreatic necrosis virus of salmonids and infectious bursal disease virus of poultry are two economically important birnaviruses. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the taxonomy of Birnaviridae, which is available at www.ictv.global/report/birnaviridae.


Asunto(s)
Birnaviridae/clasificación , ARN Viral/genética , Virión/ultraestructura , Animales , Birnaviridae/genética , Birnaviridae/aislamiento & purificación , Birnaviridae/ultraestructura , Insectos/virología , Vertebrados/virología
9.
Int J Mol Sci ; 18(8)2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28749409

RESUMEN

Highly pathogenic influenza A viruses (IAV) infections represent a serious threat to humans due to their considerable morbidity and mortality capacities. A good understanding of the molecular mechanisms responsible for the acute lung injury observed during this kind of infection is essential to design adapted therapies. In the current study, using an unbiased transcriptomic approach, we compared the host-responses of mice infected with two different subtypes of IAV: H1N1 vs. H5N1. The host-response comparison demonstrated a clear difference between the transcriptomic profiles of H1N1- and H5N1-infected mice despite identical survival kinetics and similar viral replications. The ontological analysis of the two transcriptomes showed two probable causes of death: induction of an immunopathological state of the lung for the H1N1 strain vs. development of respiratory dysfunction in the case of the H5N1 IAV. Finally, a clear signature responsible for lung edema was specifically associated with the H5N1 infection. We propose a potential mechanism of edema development based on predictive bioinformatics tools.


Asunto(s)
Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Animales , Análisis por Conglomerados , Epistasis Genética , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Modelos Biológicos , Infecciones por Orthomyxoviridae/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Carga Viral
10.
J Gen Virol ; 98(6): 1196-1208, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28613140

RESUMEN

PB1-F2 is a viral protein encoded by influenza A viruses (IAVs). PB1-F2 is implicated in virulence by triggering immune cell apoptosis and enhancing inflammation. To obtain an insight into the molecular mechanisms of PB1-F2-mediated virulence, we used the yeast two-hybrid approach to find new PB1-F2 cellular interactors. This allowed us to identify calcium-binding and coiled-coil domain 2 (CALCOCO2, also known as NDP52) as a binding partner of PB1-F2. Binding of PB1-F2 to CALCOCO2 was confirmed by pull-down. Surface plasmon resonance binding experiments enabled us to estimate the dissociation constant (Kd) of the two partners to be around 20 nM. Using bioinformatics tools, we designed a CALCOCO2 interaction map based on previous knowledge and showed a strong connection between this protein and the type I interferon production pathways and the I-κB kinase/NF-κB signalling pathway. NF-κB reporter assays in which CALCOCO2, MAVS and PB1-F2 were co-expressed showed a cooperation of these three proteins to increase the inflammatory response. By contrast, PB1-F2 inhibits the TBK1-dependent activation of an ISRE reporter plasmid. We also demonstrated that the signal transducer TRAF6 is implicated in the enhancement of NF-κB activity mediated by PB1-F2/CALCOCO2 binding. Altogether, this report provides evidence of an interaction link between PB1-F2 and human proteins, and allows a better understanding of the involvement of PB1-F2 in the pathologic process mediated by IAV.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Proteínas Nucleares/metabolismo , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Biología Computacional , Humanos , Cinética , Unión Proteica , Mapeo de Interacción de Proteínas , Resonancia por Plasmón de Superficie , Técnicas del Sistema de Dos Híbridos
11.
PLoS One ; 12(4): e0176355, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28441462

RESUMEN

Avian Influenza virus (AIV) is a major concern for the global poultry industry. Since 2012, several countries have reported AIV outbreaks among domestic poultry. These outbreaks had tremendous impact on poultry production and socio-economic repercussion on farmers. In addition, the constant emergence of highly pathogenic AIV also poses a significant risk to human health. In this study, we used a chicken lung epithelial cell line (CLEC213) to gain a better understanding of the molecular consequences of low pathogenic AIV infection in their natural host. Using a transcriptome profiling approach based on microarrays, we identified a cluster of mitochondrial genes highly induced during the infection. Interestingly, most of the regulated genes are encoded by the mitochondrial genome and are involved in the oxidative phosphorylation metabolic pathway. The biological consequences of this transcriptomic induction result in a 2.5- to 4-fold increase of the ATP concentration within the infected cells. PB1-F2, a viral protein that targets the mitochondria was not found associated to the boost of activity of the respiratory chain. We next explored the possibility that ATP may act as a host-derived danger signal (through production of extracellular ATP) or as a boost to increase AIV replication. We observed that, despite the activation of the P2X7 purinergic receptor pathway, a 1mM ATP addition in the cell culture medium had no effect on the virus replication in our epithelial cell model. Finally, we found that oligomycin, a drug that inhibits the oxidative phosphorylation process, drastically reduced the AIV replication in CLEC213 cells, without apparent cellular toxicity. Collectively, our results suggest that AIV is able to boost the metabolic capacities of its avian host in order to provide the important energy needs required to produce progeny virus.


Asunto(s)
Transporte de Electrón/genética , Células Epiteliales/virología , Gripe Aviar/metabolismo , Pulmón/virología , Mitocondrias/metabolismo , Animales , Línea Celular , Pollos , Transporte de Electrón/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Virus de la Influenza A , Gripe Aviar/virología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Mitocondrias/genética , Oligomicinas/farmacología , Fosforilación Oxidativa/efectos de los fármacos , Transcriptoma , Replicación Viral/efectos de los fármacos
12.
PLoS One ; 11(10): e0165361, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27798704

RESUMEN

The influenza A virus (IAV) PB1-F2 protein is a virulence factor contributing to the pathogenesis observed during IAV infections in mammals. In this study, using a mouse model, we compared the host response associated with PB1-F2 with an early transcriptomic signature that was previously associated with neutrophils and consecutively fatal IAV infections. This allowed us to show that PB1-F2 is partly involved in neutrophil-related mechanisms leading to death. Using neutropenic mice, we confirmed that the harmful effect of PB1-F2 is due to an excessive inflammation mediated by an increased neutrophil mobilization. We identified the downstream effects of this PB1-F2-exacerbated neutrophil recruitment. PB1-F2 had no impact on the lymphocyte recruitment in the airways at day 8 pi. However, functional genomics analysis and flow cytometry in broncho-alveolar lavages at 4 days pi revealed that PB1-F2 induced a NK cells deficiency. Thus, our results identify PB1-F2 as an important immune disruptive factor during the IAV infection.


Asunto(s)
Células Asesinas Naturales/inmunología , Infiltración Neutrófila , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Proteínas Virales/metabolismo , Animales , Femenino , Inflamación/patología , Luciferasas/metabolismo , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Neutropenia/complicaciones , Neutropenia/patología , Neutrófilos/patología , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/mortalidad , Linfocitos T/patología , Transcriptoma/genética
13.
J Biol Chem ; 291(17): 9060-72, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26896002

RESUMEN

PB1-F2 is a virulence factor of influenza A virus (IAV) whose functions remain misunderstood. The different roles of PB1-F2 may be linked to its structural polymorphism and to its propensity to assemble into oligomers and amyloid fibers in the vicinity of the membrane of IAV-infected cells. Here, we monitored the impact of PB1-F2 on the biochemical composition and protein structures of human epithelial pulmonary cells (A549) and monocytic cells (U937) upon IAV infection using synchrotron Fourier-transform infrared (FTIR) and deep UV (DUV) microscopies at the single-cell level. Cells were infected with a wild-type IAV and its PB1-F2 knock-out mutant for analyses at different times post-infection. IR spectra were recorded in each condition and processed to evaluate the change in the component band of the spectra corresponding to the amide I (secondary structure) and the CH stretching region (membrane). The IR spectra analysis revealed that expression of PB1-F2 in U937 cells, but not in A549 cells, results in the presence of a specific ß-aggregate signature. Furthermore, the lipid membrane composition of U937 cells expressing PB1-F2 was also altered in a cell type-dependent manner. Using DUV microscopy and taking advantage of the high content of tryptophan residues in the sequence of PB1-F2 (5/90 aa), we showed that the increase of the autofluorescent signal recorded in monocytic cells could be correlated with the IR detection of ß-aggregates. Altogether, our results constitute an important step forward in the understanding of the cell type-dependent function of PB1-F2.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/metabolismo , Agregado de Proteínas , Proteínas Virales/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/virología , Células HeLa , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/genética , Microscopía Fluorescente , Células U937 , Proteínas Virales/genética
14.
J Virol ; 90(7): 3684-93, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26792748

RESUMEN

UNLABELLED: The influenza virus RNA-dependent RNA polymerase, which is composed of three subunits, PB1, PB2, and PA, catalyzes genome replication and transcription within the cell nucleus. The PA linker (residues 197 to 256) can be altered by nucleotide substitutions to engineer temperature-sensitive (ts), attenuated mutants that display a defect in the transport of the PA-PB1 complex to the nucleus at a restrictive temperature. In this study, we investigated the ability of the PA linker to tolerate deletion mutations for further in vitro and in vivo characterization. Four viable mutants with single-codon deletions were generated; all of them exhibited a ts phenotype that was associated with the reduced efficiency of replication/transcription of a pseudoviral reporter RNA in a minireplicon assay. Using fluorescently tagged PB1, we observed that the deletion mutants did not efficiently recruit PB1 to reach the nucleus at a restrictive temperature (39.5°C). Mouse infections showed that the four mutants were attenuated and induced antibodies that were able to protect mice from challenge with a lethal homologous wild-type virus. Serial in vitro passages of two deletion mutants at 39.5°C and 37°C did not allow the restoration of a wild-type phenotype among virus progeny. Thus, our results identify codons that can be deleted in the PA gene to engineer genetically stable ts mutants that could be used to design novel attenuated vaccines. IMPORTANCE: In order to generate genetically stable live influenza A virus vaccines, we constructed viruses with single-codon deletions in a discrete domain of the RNA polymerase PA gene. The four rescued viruses exhibited a temperature-sensitive phenotype that we found was associated with a defect in the transport of the PA-PB1 dimer to the nucleus, where viral replication occurs. These ts deletion mutants were shown to be attenuated and to be able to produce antibodies in mice and to protect them from a lethal challenge. Assays to select revertants that were able to grow efficiently at a restrictive temperature failed, showing that these deletion mutants are genetically more stable than conventional substitution mutants. These results are of interest for the design of genetically stable live influenza virus vaccines.


Asunto(s)
Codón , Virus de la Influenza A/fisiología , Proteínas Mutantes/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Eliminación de Secuencia , Proteínas Virales/metabolismo , Replicación Viral , Animales , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Femenino , Inestabilidad Genómica , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Ratones Endogámicos BALB C , Viabilidad Microbiana , Proteínas Mutantes/genética , Infecciones por Orthomyxoviridae/virología , ARN Polimerasa Dependiente del ARN/genética , Temperatura , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Proteínas Virales/genética
15.
J Biol Chem ; 291(2): 739-51, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26601953

RESUMEN

PB1-F2 is a small accessory protein encoded by an alternative open reading frame in PB1 segments of most influenza A virus. PB1-F2 is involved in virulence by inducing mitochondria-mediated immune cells apoptosis, increasing inflammation, and enhancing predisposition to secondary bacterial infections. Using biophysical approaches we characterized membrane disruptive activity of the full-length PB1-F2 (90 amino acids), its N-terminal domain (52 amino acids), expressed by currently circulating H1N1 viruses, and its C-terminal domain (38 amino acids). Both full-length and N-terminal domain of PB1-F2 are soluble at pH values ≤6, whereas the C-terminal fragment was found soluble only at pH ≤ 3. All three peptides are intrinsically disordered. At pH ≥ 7, the C-terminal part of PB1-F2 spontaneously switches to amyloid oligomers, whereas full-length and the N-terminal domain of PB1-F2 aggregate to amorphous structures. When incubated with anionic liposomes at pH 5, full-length and the C-terminal part of PB1-F2 assemble into amyloid structures and disrupt membrane at nanomolar concentrations. PB1-F2 and its C-terminal exhibit no significant antimicrobial activity. When added in the culture medium of mammalian cells, PB1-F2 amorphous aggregates show no cytotoxicity, whereas PB1-F2 pre-assembled into amyloid oligomers or fragmented nanoscaled fibrils was highly cytotoxic. Furthermore, the formation of PB1-F2 amyloid oligomers in infected cells was directly reflected by membrane disruption and cell death as observed in U937 and A549 cells. Altogether our results demonstrate that membrane-lytic activity of PB1-F2 is closely linked to supramolecular organization of the protein.


Asunto(s)
Amiloide/toxicidad , Liposomas/metabolismo , Proteínas Virales/toxicidad , Antiinfecciosos/farmacología , Bacillus subtilis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Escherichia coli/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/ultraestructura , Liposomas/ultraestructura , Pruebas de Sensibilidad Microbiana , Permeabilidad , Agregado de Proteínas/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Virales/química
16.
J Virol ; 89(12): 6376-90, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25855727

RESUMEN

UNLABELLED: The influenza virus RNA-dependent RNA polymerase catalyzes genome replication and transcription within the cell nucleus. Efficient nuclear import and assembly of the polymerase subunits PB1, PB2, and PA are critical steps in the virus life cycle. We investigated the structure and function of the PA linker (residues 197 to 256), located between its N-terminal endonuclease domain and its C-terminal structured domain that binds PB1, the polymerase core. Circular dichroism experiments revealed that the PA linker by itself is structurally disordered. A large series of PA linker mutants exhibited a temperature-sensitive (ts) phenotype (reduced viral growth at 39.5°C versus 37°C/33°C), suggesting an alteration of folding kinetic parameters. The ts phenotype was associated with a reduced efficiency of replication/transcription of a pseudoviral reporter RNA in a minireplicon assay. Using a fluorescent-tagged PB1, we observed that ts and lethal PA mutants did not efficiently recruit PB1 to reach the nucleus at 39.5°C. A protein complementation assay using PA mutants, PB1, and ß-importin IPO5 tagged with fragments of the Gaussia princeps luciferase showed that increasing the temperature negatively modulated the PA-PB1 and the PA-PB1-IPO5 interactions or complex stability. The selection of revertant viruses allowed the identification of different types of compensatory mutations located in one or the other of the three polymerase subunits. Two ts mutants were shown to be attenuated and able to induce antibodies in mice. Taken together, our results identify a PA domain critical for PB1-PA nuclear import and that is a "hot spot" to engineer ts mutants that could be used to design novel attenuated vaccines. IMPORTANCE: By targeting a discrete domain of the PA polymerase subunit of influenza virus, we were able to identify a series of 9 amino acid positions that are appropriate to engineer temperature-sensitive (ts) mutants. This is the first time that a large number of ts mutations were engineered in such a short domain, demonstrating that rational design of ts mutants can be achieved. We were able to associate this phenotype with a defect of transport of the PA-PB1 complex into the nucleus. Reversion substitutions restored the ability of the complex to move to the nucleus. Two of these ts mutants were shown to be attenuated and able to produce antibodies in mice. These results are of high interest for the design of novel attenuated vaccines and to develop new antiviral drugs.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H1N1 del Virus de la Influenza A/efectos de la radiación , Mutación , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/efectos de la radiación , Transporte Activo de Núcleo Celular , Animales , Dicroismo Circular , Femenino , Prueba de Complementación Genética , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica/efectos de la radiación , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , ARN Polimerasa Dependiente del ARN/química , Temperatura , Proteínas Virales/química
17.
Expert Opin Drug Discov ; 10(4): 345-71, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25792362

RESUMEN

INTRODUCTION: Influenza viruses are a threat to human health. There are presently only two methods for treating influenza: vaccines, which require yearly updates, and two classes of antivirals that suffer with the problem of resistance by current human influenza viruses; this is especially the case with amantadine and rimantadine. Consequently, there is an urgent need for the development of new antivirals with new mechanisms of action. AREAS COVERED: In this review, the authors focus on viral protein domains, their associated activity and their inhibition by small molecules defined by a structure-based design with a special emphasis on the ribonucleoprotein complex and its inhibitors. Several new classes of antiviral candidates targeting viral replication through individual domains of the polymerase and the nucleoprotein (NP) have been developed through structure-based design. EXPERT OPINION: To date, the antivirals targeting neuraminidase are by far the most developed and potent. Antiviral candidates targeting the NP and polymerase domains are in the pipeline but their pharmacokinetics needs further studies. The recently published structures of the polymerase expand the possibilities for development of new antivirals. Combination therapies targeting conserved viral targets and new cellular proteins or exploiting drug promiscuity hold promises to fight against the emergence of resistance.


Asunto(s)
Antivirales/uso terapéutico , Diseño de Fármacos , Gripe Humana/tratamiento farmacológico , Animales , Antivirales/farmacocinética , Antivirales/farmacología , Farmacorresistencia Viral , Humanos , Gripe Humana/virología , Ribonucleoproteínas/antagonistas & inhibidores , Ribonucleoproteínas/metabolismo , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
18.
PLoS One ; 9(6): e100679, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24959667

RESUMEN

Highly pathogenic avian influenza virus (HPAIV) is a permanent threat due to its capacity to cross species barriers and generate severe infections and high mortality in humans. Recent findings have highlighted the potential role of PB1-F2, a small accessory influenza protein, in the pathogenesis process mediated by HPAIV in mammals. In this study, using a recombinant H5N1 HPAIV (wt) and its PB1-F2-deleted mutant (ΔF2), we studied the effects of PB1-F2 in a chicken model. Unexpectedly, when using low inoculation dose we observed that the wt-infected chickens had a higher survival rate than the ΔF2-infected chickens, a feature that contrasts with what is usually observed in mammals. High inoculation dose had similar mortality rate for both viruses, and comparison of the bio-distribution of the two viruses indicated that the expression of PB1-F2 allows a better spreading of the virus within chicken embryos. Transcriptomic profiles of lungs and blood cells were characterized at two days post-infection in chickens inoculated with the wild type (wt) or the ΔF2 mutant viruses. In lungs, the expression of PB1-F2 during the infection induced pathways related to calcium signaling and repressed a large panel of immunological functions. In blood cells, PB1-F2 was associated with a gene signature specific for mitochondrial dysfunction and down-modulated leucocytes activation. Finally we compared the effect of PB1-F2 in lungs of chickens and mice. We identified that gene signature associated to tissue damages is a PB1-F2 feature shared by the two species; by contrast, the early inhibition of immune response mediated by PB1-F2 observed in chickens is not seen in mice. In summary, our data suggest that PB1-F2 expression deeply affect the immune response in chickens in a way that may attenuate pathogenicity at low infection dose, a feature differing from what was previously observed in mammal species.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Proteínas Virales/genética , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Embrión de Pollo , Pollos , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Aviar/genética , Gripe Aviar/inmunología , Gripe Aviar/mortalidad , Pulmón/patología , Pulmón/virología , Ratones , Mutación , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Proteínas Virales/inmunología , Virulencia/genética , Replicación Viral , Esparcimiento de Virus
19.
Biosens Bioelectron ; 59: 6-13, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24686222

RESUMEN

Influenza virus represents a major concern of human health and animal production. PB1-F2 is a small proapoptotic protein supposed to contribute to the virulence of influenza A virus (IAV). However, the molecular mechanism of action of PB1-F2 is still unclear.PB1-F2 expression and behavior during the viral cycle is difficult to follow with classical biochemical methods. In this work we have developed an electrochemical biosensor based on immuno-detection system for quantification of PB1-F2 protein in infected cell. The electrochemical immunosensor was based on conducting copolypyrrole integrating ferrocenyl group as redox marker for enhancing signal detection. A specific anti-PB1-F2 monoclonal antibody was immobilized on the copolypyrrole layer via biotin-streptavidin system. We demonstrate that this electrochemical system sensitively detect purified recombinant PB1-F2 over a wide range of concentrations from 5 nM to 1.5 µM. The high sensor sensitivity allowed the detection of PB1-F2 in lysates of infected cells confirming that PB1-F2 is expressed in early stages of viral cycle. The immunosensor developed shows enhanced performances for the evaluation of PB1-F2 protein concentration in biological samples and could be applied for studying of PB1-F2 during influenza virus infection.


Asunto(s)
Técnicas Electroquímicas/métodos , Virus de la Influenza A/aislamiento & purificación , Gripe Humana/diagnóstico , Proteínas Virales/análisis , Técnicas Biosensibles/métodos , Línea Celular , Células Epiteliales/virología , Compuestos Ferrosos/química , Humanos , Inmunoensayo/métodos , Gripe Humana/virología , Metalocenos , Modelos Moleculares , Polímeros/química , Pirroles/química , Sensibilidad y Especificidad
20.
Future Microbiol ; 8(12): 1537-45, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24266354

RESUMEN

The nucleoprotein (NP) of influenza virus covers the viral RNA entirely and it is this NP-RNA complex that is the template for transcription and replication by the viral polymerase. Purified NP forms a dynamic equilibrium between monomers and small oligomers, but only the monomers can oligomerize onto RNA. Therefore, drugs that stabilize the monomers or that induce abnormal oligomerization may have an antiviral effect, as would drugs that interfere with RNA binding. Crystal structures have been produced for monomeric and dimeric mutants, and for trimers and tetramers; high-resolution electron microscopy structures have also been calculated for the viral NP-RNA complex. We explain how these structures and the dynamic oligomerization equilibrium of NP can be and have been used for anti-influenza drug development.


Asunto(s)
Antivirales/farmacología , Virus de la Influenza A/metabolismo , Gripe Humana/virología , Nucleoproteínas/química , Nucleoproteínas/metabolismo , ARN Viral/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Animales , Humanos , Virus de la Influenza A/química , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/genética , Gripe Humana/tratamiento farmacológico , Nucleoproteínas/antagonistas & inhibidores , Nucleoproteínas/genética , ARN Viral/genética , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA