Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Biotechnol ; 31(9): 844-847, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23934176

RESUMEN

Recent gain-of-function studies in influenza A virus H5N1 strains revealed that as few as three-amino-acid changes in the hemagglutinin protein confer the capacity for viral transmission between ferrets. As transmission between ferrets is considered a surrogate indicator of transmissibility between humans, these studies raised concerns about the risks of gain-of-function influenza A virus research. Here we present an approach to strengthen the biosafety of gain-of-function influenza experiments. We exploit species-specific endogenous small RNAs to restrict influenza A virus tropism. In particular, we found that the microRNA miR-192 was expressed in primary human respiratory tract epithelial cells as well as in mouse lungs but absent from the ferret respiratory tract. Incorporation of miR-192 target sites into influenza A virus did not prevent influenza replication and transmissibility in ferrets, but did attenuate influenza pathogenicity in mice. This molecular biocontainment approach should be applicable beyond influenza A virus to minimize the risk of experiments involving other pathogenic viruses.


Asunto(s)
Investigación Biomédica , Subtipo H5N1 del Virus de la Influenza A , MicroARNs , Virología , Animales , Investigación Biomédica/métodos , Investigación Biomédica/normas , Peso Corporal , Hurones , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Infecciones por Orthomyxoviridae/virología , Gestión de Riesgos , Análisis de Supervivencia , Tropismo Viral/genética , Virología/métodos , Virología/normas , Replicación Viral/genética
2.
Cell Rep ; 3(1): 23-9, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23333274

RESUMEN

Influenza A virus is unique as an RNA virus in that it replicates in the nucleus and undergoes splicing. With only ten major proteins, the virus must gain nuclear access, replicate, assemble progeny virions in the cytoplasm, and then egress. In an effort to elucidate the coordination of these events, we manipulated the transcript levels from the bicistronic nonstructural segment that encodes the spliced virus product responsible for genomic nuclear export. We find that utilization of an erroneous splice site ensures the slow accumulation of the viral nuclear export protein (NEP) while generating excessive levels of an antagonist that inhibits the cellular response to infection. Modulation of this simple transcriptional event results in improperly timed export and loss of virus infection. Together, these data demonstrate that coordination of the influenza A virus life cycle is set by a "molecular timer" that operates on the inefficient splicing of a virus transcript.


Asunto(s)
Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/virología , Empalme del ARN/genética , Transporte Activo de Núcleo Celular , Animales , Silenciador del Gen , Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , ARN Interferente Pequeño/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
3.
Proc Natl Acad Sci U S A ; 109(30): 12117-22, 2012 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-22778433

RESUMEN

A coordinated innate and adaptive immune response, orchestrated by antigen presenting cells (APCs), is required for effective clearance of influenza A virus (IAV). Although IAV primarily infects epithelial cells of the upper respiratory tract, APCs are also susceptible. To determine if virus transcription in these cells is required to generate protective innate and adaptive immune responses, we engineered IAV to be selectively attenuated in cells of hematopoietic origin. Incorporation of hematopoietic-specific miR-142 target sites into the nucleoprotein of IAV effectively silenced virus transcription in APCs, but had no significant impact in lung epithelial cells. Here we demonstrate that inhibiting IAV replication in APCs in vivo did not alter clearance, or the generation of IAV-specific CD8 T cells, suggesting that cross-presentation is sufficient for cytotoxic T lymphocyte activation. In contrast, loss of in vivo virus infection, selectively in APCs, resulted in a significant reduction of retinoic acid-inducible gene I-dependent type I IFN (IFN-I). These data implicate the formation of virus replication intermediates in APCs as the predominant trigger of IFN-I in vivo. Taking these data together, this research describes a unique platform to study the host response to IAV and provides insights into the mechanism of antigen presentation and the induction of IFN-I.


Asunto(s)
Inmunidad Adaptativa/inmunología , Células Presentadoras de Antígenos/virología , Regulación Viral de la Expresión Génica/inmunología , Inmunidad Innata/inmunología , Virus de la Influenza A/inmunología , Replicación Viral/inmunología , Animales , Secuencia de Bases , Northern Blotting , Western Blotting , Línea Celular , Clonación Molecular , Cartilla de ADN/genética , Perros , Fibroblastos , Citometría de Flujo , Regulación Viral de la Expresión Génica/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Interferón Tipo I/metabolismo , Macrófagos , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Nucleoproteínas/genética , Nucleoproteínas/metabolismo
4.
Proc Natl Acad Sci U S A ; 107(25): 11519-24, 2010 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-20534531

RESUMEN

MicroRNAs (miRNAs) are short noncoding RNAs that exert posttranscriptional gene silencing and regulate gene expression. In addition to the hundreds of conserved cellular miRNAs that have been identified, miRNAs of viral origin have been isolated and found to modulate both the viral life cycle and the cellular transcriptome. Thus far, detection of virus-derived miRNAs has been largely limited to DNA viruses, suggesting that RNA viruses may be unable to exploit this aspect of transcriptional regulation. Lack of RNA virus-produced miRNAs has been attributed to the replicative constraints that would incur following RNase III processing of a genomic hairpin. To ascertain whether the generation of viral miRNAs is limited to DNA viruses, we investigated whether influenza virus could be designed to deliver functional miRNAs without affecting replication. Here, we describe a modified influenza A virus that expresses cellular microRNA-124 (miR-124). Insertion of the miR-124 hairpin into an intron of the nuclear export protein transcript resulted in endogenous processing and functional miR-124. We demonstrate that a viral RNA genome incorporating a hairpin does not result in segment instability or miRNA-mediated genomic targeting, thereby permitting the virus to produce a miRNA without having a negative impact on viral replication. This work demonstrates that RNA viruses can produce functional miRNAs and suggests that this level of transcriptional regulation may extend beyond DNA viruses.


Asunto(s)
Virus de la Influenza A/genética , MicroARNs/genética , Procesamiento Postranscripcional del ARN , ARN Viral , Animales , Perros , Regulación Viral de la Expresión Génica , Ingeniería Genética/métodos , Vectores Genéticos , Humanos , Ratones , MicroARNs/metabolismo , Modelos Genéticos , Datos de Secuencia Molecular , Plásmidos/metabolismo , Interferencia de ARN , ARN Viral/metabolismo
5.
Nat Biotechnol ; 27(6): 572-6, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19483680

RESUMEN

Influenza A virus leads to yearly epidemics and sporadic pandemics. Present prophylactic strategies focus on egg-grown, live, attenuated influenza vaccines (LAIVs), in which attenuation is generated by conferring temperature sensitivity onto the virus. Here we describe an alternative approach to attenuating influenza A virus based on microRNA-mediated gene silencing. By incorporating nonavian microRNA response elements (MREs) into the open-reading frame of the viral nucleoprotein, we generate reassortant LAIVs for H1N1 and H5N1 that are attenuated in mice but not in eggs. MRE-based LAIVs show a greater than two-log reduction in mortality compared with control viruses lacking MREs and elicit a diverse antibody response. This approach might be combined with existing LAIVs to increase attenuation and improve vaccine safety.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Vacunas contra la Influenza , MicroARNs , Interferencia de ARN , Vacunas Atenuadas , Animales , Peso Corporal , Línea Celular , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Ratones , MicroARNs/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , ARN Viral/genética , Elementos de Respuesta/genética , Especificidad de la Especie , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
6.
Mol Ther ; 16(11): 1883-90, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18714310

RESUMEN

Naturally occurring strains of Newcastle disease virus (NDV) are currently being investigated in multiple clinical trials for oncolytic cancer therapy in the United States and abroad. We have previously reported, for the first time, the development of recombinant NDVs designed for enhanced cancer therapeutic efficacy. Specifically, we have shown that NDV engineered to express interleukin-2 (IL-2) generates a robust therapeutic response associated with increased tumor-specific T-cell infiltration after intratumoral administration in mice. We have now demonstrated that this therapeutic response is dependent on T cells and we have investigated the potential to focus the NDV-induced immune response toward a tumor-associated antigen (TAA) to enhance the inherent therapeutic efficacy of NDV further. We found that intratumoral treatments of tumor-bearing mice with recombinant NDV expressing a model TAA elicited an enhanced tumor-specific response, resulting in a significant increase in the number of complete tumor regressions compared with control NDV. Additionally, coadministration of NDV expressing a model TAA with NDV expressing IL-2 enhanced the TAA-directed response and led to more complete tumor regressions. Our results show that TAA-directed immunotherapy by oncolytic recombinant NDV alone or in combination with IL-2 results in an enhanced therapeutic efficacy and warrant consideration in the development of cancer therapies based on the use of oncolytic NDV.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Neoplasias Experimentales/terapia , Virus de la Enfermedad de Newcastle/genética , Virus Oncolíticos/genética , Animales , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Epítopos de Linfocito T , Femenino , Vectores Genéticos , Antígenos de Histocompatibilidad Clase II/biosíntesis , Interleucina-2/genética , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Virus de la Enfermedad de Newcastle/metabolismo , Virus Oncolíticos/metabolismo , Trasplante Heterólogo , beta-Galactosidasa/biosíntesis
7.
Cancer Res ; 67(17): 8285-92, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17804743

RESUMEN

Naturally occurring strains of Newcastle disease virus (NDV) have shown oncolytic therapeutic efficacy in preclinical studies and are currently in clinical trials. Here, we have evaluated the possibility to enhance the cancer therapeutic potential of NDV by means of reverse genetics. Mice bearing s.c. implanted CT26 tumors were treated with intratumoral (i.t.) injections of a recombinant NDV modified to contain a highly fusogenic F protein. These treated mice exhibited significant reduction in tumor development compared with mice treated with the unmodified virus. Furthermore, mice in a CT26 metastatic tumor model treated with an i.v. injection of the genetically engineered NDV exhibited prolonged survival compared with wild-type control virus. In addition, we examined whether the oncolytic properties of NDV could be improved by expression of immunostimulatory molecules. In this regard, we engineered several NDVs to express granulocyte macrophage colony-stimulating factor, IFN-gamma, interleukin 2 (IL-2), or tumor necrosis factor alpha, and evaluated their therapeutic potential in an immunocompetent colon carcinoma tumor model. Mice bearing s.c. CT26 tumors treated with i.t. injections of recombinant NDV expressing IL-2 showed dramatic reductions in tumor growth, with a majority of the mice undergoing complete and long-lasting remission. Our data show the use of reverse genetics to develop enhanced recombinant NDV vectors as effective therapeutic agents for cancer treatment.


Asunto(s)
Carcinoma/terapia , Neoplasias del Colon/terapia , Ingeniería Genética/métodos , Virus de la Enfermedad de Newcastle/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Carga Tumoral , Células Tumorales Cultivadas
8.
Science ; 315(5816): 1274-8, 2007 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-17332413

RESUMEN

IKKepsilon is an IKK (inhibitor of nuclear factor kappaBkinase)-related kinase implicated in virus induction of interferon-beta (IFNbeta). We report that, although mice lacking IKKepsilon produce normal amounts of IFNbeta, they are hypersusceptible to viral infection because of a defect in the IFN signaling pathway. Specifically, a subset of type I IFN-stimulated genes are not activated in the absence of IKKepsilon because the interferon-stimulated gene factor 3 complex (ISGF3) does not bind to promoter elements of the affected genes. We demonstrate that IKKepsilon is activated by IFNbeta and that IKKepsilon directly phosphorylates signal transducer and activator of transcription 1 (STAT1), a component of ISGF3. We conclude that IKKepsilon plays a critical role in the IFN-inducible antiviral transcriptional response.


Asunto(s)
Regulación de la Expresión Génica , Quinasa I-kappa B/metabolismo , Subtipo H1N1 del Virus de la Influenza A , Interferón beta/inmunología , Infecciones por Orthomyxoviridae/inmunología , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Células Cultivadas , Dimerización , Quinasa I-kappa B/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón beta/metabolismo , Pulmón/patología , Pulmón/virología , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Fosforilación , Regiones Promotoras Genéticas , Proteínas de Unión al ARN , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , Transcripción Genética , Carga Viral , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA