Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Retrovirology ; 21(1): 13, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898526

RESUMEN

Retroviruses exploit host proteins to assemble and release virions from infected cells. Previously, most studies focused on interacting partners of retroviral Gag proteins that localize to the cytoplasm or plasma membrane. Given that several full-length Gag proteins have been found in the nucleus, identifying the Gag-nuclear interactome has high potential for novel findings involving previously unknown host processes. Here we systematically compared nuclear factors identified in published HIV-1 proteomic studies and performed our own mass spectrometry analysis using affinity-tagged HIV-1 and RSV Gag proteins mixed with nuclear extracts. We identified 57 nuclear proteins in common between HIV-1 and RSV Gag, and a set of nuclear proteins present in our analysis and ≥ 1 of the published HIV-1 datasets. Many proteins were associated with nuclear processes which could have functional consequences for viral replication, including transcription initiation/elongation/termination, RNA processing, splicing, and chromatin remodeling. Examples include facilitating chromatin remodeling to expose the integrated provirus, promoting expression of viral genes, repressing the transcription of antagonistic cellular genes, preventing splicing of viral RNA, altering splicing of cellular RNAs, or influencing viral or host RNA folding or RNA nuclear export. Many proteins in our pulldowns common to RSV and HIV-1 Gag are critical for transcription, including PolR2B, the second largest subunit of RNA polymerase II (RNAPII), and LEO1, a PAF1C complex member that regulates transcriptional elongation, supporting the possibility that Gag influences the host transcription profile to aid the virus. Through the interaction of RSV and HIV-1 Gag with splicing-related proteins CBLL1, HNRNPH3, TRA2B, PTBP1 and U2AF1, we speculate that Gag could enhance unspliced viral RNA production for translation and packaging. To validate one putative hit, we demonstrated an interaction of RSV Gag with Mediator complex member Med26, required for RNA polymerase II-mediated transcription. Although 57 host proteins interacted with both Gag proteins, unique host proteins belonging to each interactome dataset were identified. These results provide a strong premise for future functional studies to investigate roles for these nuclear host factors that may have shared functions in the biology of both retroviruses, as well as functions specific to RSV and HIV-1, given their distinctive hosts and molecular pathology.


Asunto(s)
Productos del Gen gag , VIH-1 , Humanos , VIH-1/fisiología , VIH-1/genética , Productos del Gen gag/metabolismo , Productos del Gen gag/genética , Núcleo Celular/metabolismo , Núcleo Celular/virología , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Virus del Sarcoma de Rous/fisiología , Virus del Sarcoma de Rous/genética , Proteómica , Interacciones Huésped-Patógeno , Replicación Viral , Interacciones Microbiota-Huesped , Espectrometría de Masas
2.
J Biol Chem ; 299(6): 104730, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37084813

RESUMEN

Integration of retroviral DNA into the host genome involves the formation of integrase (IN)-DNA complexes termed intasomes. Further characterization of these complexes is needed to understand their assembly process. Here, we report the single-particle cryo-EM structure of the Rous sarcoma virus (RSV) strand transfer complex (STC) intasome produced with IN and a preassembled viral/target DNA substrate at 3.36 Å resolution. The conserved intasome core region consisting of IN subunits contributing active sites interacting with viral/target DNA has a resolution of 3 Å. Our structure demonstrated the flexibility of the distal IN subunits relative to the IN subunits in the conserved intasome core, similar to results previously shown with the RSV octameric cleaved synaptic complex intasome produced with IN and viral DNA only. An extensive analysis of higher resolution STC structure helped in the identification of nucleoprotein interactions important for intasome assembly. Using structure-function studies, we determined the mechanisms of several IN-DNA interactions critical for assembly of both RSV intasomes. We determined the role of IN residues R244, Y246, and S124 in cleaved synaptic complex and STC intasome assemblies and their catalytic activities, demonstrating differential effects. Taken together, these studies advance our understanding of different RSV intasome structures and molecular determinants involved in their assembly.


Asunto(s)
Integrasas , Virus del Sarcoma de Rous , Integración Viral , ADN Viral/química , ADN Viral/ultraestructura , Integrasas/química , Integrasas/ultraestructura , Virus del Sarcoma de Rous/genética , Virus del Sarcoma de Rous/química , Microscopía por Crioelectrón
3.
Infect Genet Evol ; 96: 105139, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34798320

RESUMEN

A small non-coding, evolutionarily conserved regulatory RNA molecule known as microRNA (miRNA) regulates various cellular activities and pathways. MicroRNAs remain evolutionarily conserved in different species of same taxa. They are present in all organisms including viruses. Viral miRNAs are small, less conserved and less stable and have higher negative minimal folding free energy than miRNAs of different organisms. The size of viral precursor miRNA is approximately 60-119 nucleotides in length. The structure of the mature miRNA sequences is predicted by using higher negative MFE (ΔG) value. Rous sarcoma Virus (RSV), named after its inventor Peyton Rous, has been known for causing tumors in the chicken for which it is known as an oncogenic retrovirus. Using specific criteria we have predicted 5 potential miRNAs in RSV which targeted 8 tumor suppressor genes in Gallus gallus. This study aims to predict the potential miRNAs, secondary structures and their targets for better understanding of the regulatory network of Rous sarcoma virus miRNA in forming sarcoma.


Asunto(s)
Pollos , Genes Supresores de Tumor/fisiología , MicroARNs/genética , Enfermedades de las Aves de Corral/virología , ARN Viral/genética , Virus del Sarcoma de Rous/genética , Sarcoma Aviar/virología , Animales
4.
J Virol ; 95(20): e0064821, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34319154

RESUMEN

During retroviral replication, unspliced viral genomic RNA (gRNA) must escape the nucleus for translation into viral proteins and packaging into virions. "Complex" retroviruses, such as human immunodeficiency virus (HIV), use cis-acting elements on the unspliced gRNA in conjunction with trans-acting viral proteins to facilitate this escape. "Simple" retroviruses, such as Mason-Pfizer monkey virus (MPMV) and murine leukemia virus (MLV), exclusively use cis-acting elements on the gRNA in conjunction with host nuclear export proteins for nuclear escape. Uniquely, the simple retrovirus Rous sarcoma virus (RSV) has a Gag structural protein that cycles through the nucleus prior to plasma membrane binding. This trafficking has been implicated in facilitating gRNA nuclear export and is thought to be a required mechanism. Previously described mutants that abolish nuclear cycling displayed enhanced plasma membrane binding, enhanced virion release, and a significant loss in genome incorporation resulting in loss of infectivity. Here, we describe a nuclear cycling-deficient RSV Gag mutant that has similar plasma membrane binding and genome incorporation to wild-type (WT) virus and surprisingly is replication competent, albeit with a slower rate of spread than observed in WT virus. This mutant suggests that RSV Gag nuclear cycling is not strictly required for RSV replication. IMPORTANCE While mechanisms for retroviral Gag assembly at the plasma membrane are beginning to be characterized, characterization of intermediate trafficking locales remain elusive. This is in part due to the difficulty of tracking individual proteins from translation to plasma membrane binding. Rous sarcoma virus (RSV) Gag nuclear cycling is a unique phenotype that may provide comparative insight to viral trafficking evolution and may present a model intermediate to cis- and trans-acting mechanisms for gRNA export.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Productos del Gen gag/genética , Virus del Sarcoma de Rous/genética , Transporte Activo de Núcleo Celular/genética , Animales , Línea Celular , Núcleo Celular/virología , Productos del Gen gag/metabolismo , Genoma Viral/genética , Humanos , Ratones , ARN Viral/genética , Retroviridae/genética , Virus del Sarcoma de Rous/metabolismo , Virión/metabolismo , Ensamble de Virus
5.
Viruses ; 13(5)2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-34068261

RESUMEN

Retroviruses are unique in that they package their RNA genomes as non-covalently linked dimers. Failure to dimerize their genomes results in decreased infectivity and reduced packaging of genomic RNA into virus particles. Two models of retrovirus genome dimerization have been characterized: in murine leukemia virus (MLV), genomic RNA dimerization occurs co-transcriptionally in the nucleus, resulting in the preferential formation of genome homodimers; whereas in human immunodeficiency virus (HIV-1), genomic RNA dimerization occurs in the cytoplasm and at the plasma membrane, with a random distribution of heterodimers and homodimers. Although in vitro studies have identified the genomic RNA sequences that facilitate dimerization in Rous sarcoma virus (RSV), in vivo characterization of the location and preferences of genome dimerization has not been performed. In this study, we utilized three single molecule RNA imaging approaches to visualize genome dimers of RSV in cultured quail fibroblasts. The formation of genomic RNA heterodimers within cells was dependent on the presence of the dimerization initiation site (DIS) sequence in the L3 stem. Subcellular localization analysis revealed that heterodimers were present the nucleus, cytoplasm, and at the plasma membrane, indicating that genome dimers can form in the nucleus. Furthermore, single virion analysis revealed that RSV preferentially packages genome homodimers into virus particles. Therefore, the mechanism of RSV genomic RNA dimer formation appears more similar to MLV than HIV-1.


Asunto(s)
Genoma Viral , Hibridación Fluorescente in Situ , Imagen Molecular , ARN Viral , Virus del Sarcoma de Rous/genética , Membrana Celular , Núcleo Celular , Células Cultivadas , Citoplasma , Dimerización , Humanos , Hibridación Fluorescente in Situ/métodos , Imagen Molecular/métodos , ARN Viral/química , ARN Viral/genética
6.
Commun Biol ; 4(1): 330, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712691

RESUMEN

Despite conserved catalytic integration mechanisms, retroviral intasomes composed of integrase (IN) and viral DNA possess diverse structures with variable numbers of IN subunits. To investigate intasome assembly mechanisms, we employed the Rous sarcoma virus (RSV) IN dimer that assembles a precursor tetrameric structure in transit to the mature octameric intasome. We determined the structure of RSV octameric intasome stabilized by a HIV-1 IN strand transfer inhibitor using single particle cryo-electron microscopy. The structure revealed significant flexibility of the two non-catalytic distal IN dimers along with previously unrecognized movement of the conserved intasome core, suggesting ordered conformational transitions between intermediates that may be important to capture the target DNA. Single amino acid substitutions within the IN C-terminal domain affected intasome assembly and function in vitro and infectivity of pseudotyped RSV virions. Unexpectedly, 17 C-terminal amino acids of IN were dispensable for virus infection despite regulating the transition of the tetrameric intasome to the octameric form in vitro. We speculate that this region may regulate the binding of highly flexible distal IN dimers to the intasome core to form the octameric complex. Our studies reveal key steps in the assembly of RSV intasomes.


Asunto(s)
Microscopía por Crioelectrón , ADN Viral/ultraestructura , Integrasas/ultraestructura , Virus del Sarcoma de Rous/ultraestructura , Imagen Individual de Molécula , Integración Viral , ADN Viral/metabolismo , Integrasa de VIH/ultraestructura , Inhibidores de Integrasa/farmacología , Integrasas/metabolismo , Simulación del Acoplamiento Molecular , Conformación Proteica , Multimerización de Proteína , Virus del Sarcoma de Rous/efectos de los fármacos , Virus del Sarcoma de Rous/enzimología , Virus del Sarcoma de Rous/genética , Integración Viral/efectos de los fármacos , Replicación Viral
7.
J Biotechnol ; 323: 92-97, 2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-32771428

RESUMEN

Silkworms have been used as a host for the production of recombinant proteins in a baculovirus expression system using Bombyx mori nucleopolyhedrovirus (BmNPV). To coexpress several recombinant proteins, a silkworm must be coinfected with several recombinant BmNPVs, which requires a difficult DNA manipulation procedure. In this study, we constructed recombinant BmNPVs containing three expression cassettes, Rous sarcoma virus (RSV) Gag protein, surface antigen 1 of Neospora caninum (NcSAG1) and SAG1-related sequence 2 of N. caninum (NcSRS2), by Gibson assembly and the Bac-to-Bac system, designated BmNPV/SAG-SRS-Gag and BmNPV/SAG-Gag-SRS. BmNPV/SAG-SRS-Gag was expressed in silkworms and characterized. NcSAG1 and NcSRS2 were purified with RSV Gag proteins using sucrose density gradient centrifugation and affinity chromatography. RSV Gag formed virus-like particles (RSV-LPs) at a diameter of 20-30 nm based on transmission electron microscopy (TEM). Immuno-TEM analysis showed that both NcSAG1 and NcSRS2 were displayed on the surface of the RSV-LPs. These results indicate that RSV-LPs displaying two different kinds of proteins were produced in the hemolymph of silkworm larvae by the single polycistronic strategy. This expression platform is efficient for generating multiantigen-displaying VLPs and facilitates the development of vaccines against infectious diseases.


Asunto(s)
Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Bombyx/genética , Nucleopoliedrovirus/genética , Proteínas Recombinantes , Animales , Antígenos de Superficie , Hemolinfa , Larva , Neospora , Proteínas Protozoarias/genética , Virus del Sarcoma de Rous/genética , Vacunas Sintéticas , Virión
8.
Viruses ; 12(5)2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32455905

RESUMEN

Retroviruses package their full-length, dimeric genomic RNA (gRNA) via specific interactions between the Gag polyprotein and a "Ψ" packaging signal located in the gRNA 5'-UTR. Rous sarcoma virus (RSV) gRNA has a contiguous, well-defined Ψ element, that directs the packaging of heterologous RNAs efficiently. The simplicity of RSV Ψ makes it an informative model to examine the mechanism of retroviral gRNA packaging, which is incompletely understood. Little is known about the structure of dimerization initiation sites or specific Gag interaction sites of RSV gRNA. Using selective 2'-hydroxyl acylation analyzed by primer extension (SHAPE), we probed the secondary structure of the entire RSV 5'-leader RNA for the first time. We identified a putative bipartite dimerization initiation signal (DIS), and mutation of both sites was required to significantly reduce dimerization in vitro. These mutations failed to reduce viral replication, suggesting that in vitro dimerization results do not strictly correlate with in vivo infectivity, possibly due to additional RNA interactions that maintain the dimers in cells. UV crosslinking-coupled SHAPE (XL-SHAPE) was next used to determine Gag-induced RNA conformational changes, revealing G218 as a critical Gag contact site. Overall, our results suggest that disruption of either of the DIS sequences does not reduce virus replication and reveal specific sites of Gag-RNA interactions.


Asunto(s)
Genoma Viral , ARN Viral/genética , Virus del Sarcoma de Rous/genética , Animales , Dimerización , Productos del Gen gag/metabolismo , Genómica , Conformación de Ácido Nucleico , ARN Viral/química , Sarcoma Aviar/virología , Análisis de Secuencia de ARN , Ensamble de Virus , Replicación Viral
9.
mBio ; 11(2)2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32265329

RESUMEN

Packaging of genomic RNA (gRNA) by retroviruses is essential for infectivity, yet the subcellular site of the initial interaction between the Gag polyprotein and gRNA remains poorly defined. Because retroviral particles are released from the plasma membrane, it was previously thought that Gag proteins initially bound to gRNA in the cytoplasm or at the plasma membrane. However, the Gag protein of the avian retrovirus Rous sarcoma virus (RSV) undergoes active nuclear trafficking, which is required for efficient gRNA encapsidation (L. Z. Scheifele, R. A. Garbitt, J. D. Rhoads, and L. J. Parent, Proc Natl Acad Sci U S A 99:3944-3949, 2002, https://doi.org/10.1073/pnas.062652199; R. Garbitt-Hirst, S. P. Kenney, and L. J. Parent, J Virol 83:6790-6797, 2009, https://doi.org/10.1128/JVI.00101-09). These results raise the intriguing possibility that the primary contact between Gag and gRNA might occur in the nucleus. To examine this possibility, we created a RSV proviral construct that includes 24 tandem repeats of MS2 RNA stem-loops, making it possible to track RSV viral RNA (vRNA) in live cells in which a fluorophore-conjugated MS2 coat protein is coexpressed. Using confocal microscopy, we observed that both wild-type Gag and a nuclear export mutant (Gag.L219A) colocalized with vRNA in the nucleus. In live-cell time-lapse images, the wild-type Gag protein trafficked together with vRNA as a single ribonucleoprotein (RNP) complex in the nucleoplasm near the nuclear periphery, appearing to traverse the nuclear envelope into the cytoplasm. Furthermore, biophysical imaging methods suggest that Gag and the unspliced vRNA physically interact in the nucleus. Taken together, these data suggest that RSV Gag binds unspliced vRNA to export it from the nucleus, possibly for packaging into virions as the viral genome.IMPORTANCE Retroviruses cause severe diseases in animals and humans, including cancer and acquired immunodeficiency syndromes. To propagate infection, retroviruses assemble new virus particles that contain viral proteins and unspliced vRNA to use as gRNA. Despite the critical requirement for gRNA packaging, the molecular mechanisms governing the identification and selection of gRNA by the Gag protein remain poorly understood. In this report, we demonstrate that the Rous sarcoma virus (RSV) Gag protein colocalizes with unspliced vRNA in the nucleus in the interchromatin space. Using live-cell confocal imaging, RSV Gag and unspliced vRNA were observed to move together from inside the nucleus across the nuclear envelope, suggesting that the Gag-gRNA complex initially forms in the nucleus and undergoes nuclear export into the cytoplasm as a viral ribonucleoprotein (vRNP) complex.


Asunto(s)
Núcleo Celular/virología , Productos del Gen gag/metabolismo , Genoma Viral , ARN Viral/metabolismo , Virus del Sarcoma de Rous/genética , Ensamble de Virus , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Línea Celular Transformada , Núcleo Celular/metabolismo , Fibroblastos/virología , Microscopía Confocal , Codorniz , ARN Viral/análisis , Virus del Sarcoma de Rous/metabolismo , Imagen de Lapso de Tiempo
10.
Methods Mol Biol ; 1742: 227-235, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29330804

RESUMEN

Hypoxia has long been recognized as a driving force of tumor progression and therapeutic resistance, and the transcription factor HIF-1α is believed to play a crucial role in these processes. Here we describe an efficient RCAS/Nes-TVA system that allows for in vivo manipulation of HIF-1α expression in the mouse neural progenitor cells. Simple production of the recombinant avian virus RCAS enables quick delivery of gene of interest through injection into the neural progenitors of transgenic mice expressing the viral cognate receptor TVA under the nestin promoter. By crossing with various commercially available genetically engineered mouse strains, a repertoire of mouse models can be created to study gene-specific effects on glioma genesis. This chapter provides details of plasmid construction, viral production, and intracranial delivery of transgenes, a methodology that can be easily adapted to a specific purpose.


Asunto(s)
Carcinogénesis/genética , Glioma/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células-Madre Neurales/citología , Virus del Sarcoma de Rous/genética , Animales , Proteínas Aviares/genética , Hipoxia de la Célula , Línea Celular , Pollos , Vectores Genéticos , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Nestina/genética , Células-Madre Neurales/patología , Receptores Virales/genética
11.
Viruses ; 9(8)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28763028

RESUMEN

All retroviruses use their full-length primary transcript as the major mRNA for Group-specific antigen (Gag) capsid proteins. This results in a long 3' untranslated region (UTR) downstream of the termination codon. In the case of Rous sarcoma virus (RSV), there is a 7 kb 3'UTR downstream of the gag terminator, containing the pol, env, and src genes. mRNAs containing long 3'UTRs, like those with premature termination codons, are frequently recognized by the cellular nonsense-mediated mRNA decay (NMD) machinery and targeted for degradation. To prevent this, RSV has evolved an RNA stability element (RSE) in the RNA immediately downstream of the gag termination codon. This 400-nt RNA sequence stabilizes premature termination codons (PTCs) in gag. It also stabilizes globin mRNAs with long 3'UTRs, when placed downstream of the termination codon. It is not clear how the RSE stabilizes the mRNA and prevents decay. We show here that the presence of RSE inhibits deadenylation severely. In addition, the RSE also impairs decapping (DCP2) and 5'-3' exonucleolytic (XRN1) function in knockdown experiments in human cells.


Asunto(s)
Regiones no Traducidas 3' , Estabilidad del ARN , ARN Mensajero/genética , ARN Viral/genética , Virus del Sarcoma de Rous/genética , Codón de Terminación , Técnicas de Silenciamiento del Gen , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Humanos , Degradación de ARNm Mediada por Codón sin Sentido , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Virus del Sarcoma de Rous/metabolismo
12.
Proc Natl Acad Sci U S A ; 114(26): E5148-E5157, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28607078

RESUMEN

The extent of virus transmission among individuals and species is generally determined by the presence of specific membrane-embedded virus receptors required for virus entry. Interaction of the viral envelope glycoprotein (Env) with a specific cellular receptor is the first and crucial step in determining host specificity. Using a well-established retroviral model-avian Rous sarcoma virus (RSV)-we analyzed changes in an RSV variant that had repeatedly been able to infect rodents. By envelope gene (env) sequencing, we identified eight mutations that do not match the already described mutations influencing the host range. Two of these mutations-one at the beginning (D32G) of the surface Env subunit (SU) and the other at the end of the fusion peptide region (L378S)-were found to be of critical importance, ensuring transmission to rodent, human, and chicken cells lacking the appropriate receptor. Furthermore, we carried out assays to examine the virus entry mechanism and concluded that these two mutations cause conformational changes in the Env variant and that these changes lead to an activated, or primed, state of Env (normally induced after Env interaction with the receptor). In summary, our results indicate that retroviral host range extension is caused by spontaneous Env activation, which circumvents the need for original cell receptor. This activation is, in turn, caused by mutations in various env regions.


Asunto(s)
Productos del Gen env , Vectores Genéticos , Mutación Missense , Virus del Sarcoma de Rous , Transducción Genética , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Pollos , Productos del Gen env/genética , Productos del Gen env/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Ratas , Virus del Sarcoma de Rous/genética , Virus del Sarcoma de Rous/metabolismo
13.
Mol Ther ; 25(5): 1187-1198, 2017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28365028

RESUMEN

Conventional plasmid vectors are incapable of achieving sustained levels of transgene expression in vivo even in quiescent mammalian tissues because the transgene expression cassette is silenced. Transcriptional silencing results from the presence of the bacterial plasmid backbone or virtually any DNA sequence of >1 kb in length placed outside of the expression cassette. Here, we show that transcriptional silencing can be substantially forestalled by increasing the An/Tn sequence composition in the plasmid bacterial backbone. Increasing numbers of An/Tn sequences increased sustained transcription of both backbone sequences and adjacent expression cassettes. In order to recapitulate these expression profiles in compact and portable plasmid DNA backbones, we engineered the standard kanamycin or ampicillin antibiotic resistance genes, optimizing the number of An/Tn sequence without altering the encoded amino acids. The resulting vector backbones yield sustained transgene expression from mouse liver, providing generic DNA vectors capable of sustained transgene expression without additional genes or mammalian regulatory elements.


Asunto(s)
Dependovirus/genética , Farmacorresistencia Microbiana/genética , Ingeniería Genética/métodos , Vectores Genéticos/química , Plásmidos/química , alfa 1-Antitripsina/genética , Ampicilina/farmacología , Animales , Antibacterianos/farmacología , Dependovirus/metabolismo , Femenino , Silenciador del Gen , Vectores Genéticos/metabolismo , Humanos , Kanamicina/farmacología , Hígado/metabolismo , Hígado/virología , Ratones , Ratones Endogámicos C57BL , Motivos de Nucleótidos , Plásmidos/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Virus del Sarcoma de Rous/genética , Virus del Sarcoma de Rous/metabolismo , Transcripción Genética/efectos de los fármacos , Transgenes , alfa 1-Antitripsina/metabolismo
14.
Virology ; 498: 181-191, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27596537

RESUMEN

The weak polyadenylation site (PAS) of Rous sarcoma virus (RSV) is activated by the juxtaposition of SR protein binding sites within the spatially separate negative regulator of splicing (NRS) element and the env RNA splicing enhancer (Env enhancer), which are far upstream of the PAS. Juxtaposition occurs by formation of the NRS - 3' ss splicing regulatory complex and is thought to provide a threshold of SR proteins that facilitate long-range stimulation of the PAS. We provide evidence for the threshold model by showing that greater than three synthetic SR protein binding sites are needed to substitute for the Env enhancer, that either the NRS or Env enhancer alone promotes polyadenylation when the distance to the PAS is decreased, and that SR protein binding sites promote binding of the polyadenylation factor cleavage factor I (CFIm) to the weak PAS. These observations may be relevant for cellular PASs.


Asunto(s)
Poliadenilación , ARN Mensajero , ARN Viral , Proteínas de Unión al ARN/metabolismo , Virus del Sarcoma de Rous/genética , Virus del Sarcoma de Rous/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Animales , Sitios de Unión , Línea Celular , Orden Génico , Sistemas de Lectura Abierta , Poli A , Unión Proteica
15.
Proc Natl Acad Sci U S A ; 113(15): 3927-31, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27035994

RESUMEN

This article summarizes the essential steps in understanding the chicken Rous sarcoma virus (RSV) genome association with a nonpermissive rodent host cell genome. This insight was made possible by in-depth study of RSV-transformed rat XC cells, which were called virogenic because they indefinitely carry virus genetic information in the absence of any infectious virus production. However, the virus was rescued by association of XC cells with chicken fibroblasts, allowing cell fusion between both partners. This and additional studies led to the interpretation that the RSV genome gets integrated into the host cell genome as a provirus. Study of additional rodent virogenic cell lines provided evidence that the transcript of oncogene v-src can be transmitted to other retroviruses and produce cell transformation by itself. As discussed in the text, two main questions related to nonpermissiveness to retrovirus infection remain to be solved. The first is changes in the retrovirus envelope gene allowing virus entry into a nonpermissive cell. The second is the nature of the permissive cell functions required by the nonpermissive cell to ensure infectious virus production. Both lines of investigation are being pursued.


Asunto(s)
Fusión Celular , Pollos/virología , Proteína Oncogénica pp60(v-src)/genética , Provirus/crecimiento & desarrollo , Virus del Sarcoma de Rous/crecimiento & desarrollo , Virus del Sarcoma de Rous/genética , Animales , Línea Celular , Transformación Celular Viral , Productos del Gen env/genética , Genoma Viral/genética , Provirus/genética , Ratas
16.
J Virol ; 90(12): 5700-5714, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27053549

RESUMEN

UNLABELLED: Extensive studies of orthoretroviral capsids have shown that many regions of the CA protein play unique roles at different points in the virus life cycle. The N-terminal domain (NTD) flexible-loop (FL) region is one such example: exposed on the outer capsid surface, it has been implicated in Gag-mediated particle assembly, capsid maturation, and early replication events. We have now defined the contributions of charged residues in the FL region of the Rous sarcoma virus (RSV) CA to particle assembly. Effects of mutations on assembly were assessed in vivo and in vitro and analyzed in light of new RSV Gag lattice models. Virus replication was strongly dependent on the preservation of charge at a few critical positions in Gag-Gag interfaces. In particular, a cluster of charges at the beginning of FL contributes to an extensive electrostatic network that is important for robust Gag assembly and subsequent capsid maturation. Second-site suppressor analysis suggests that one of these charged residues, D87, has distal influence on interhexamer interactions involving helix α7. Overall, the tolerance of FL to most mutations is consistent with current models of Gag lattice structures. However, the results support the interpretation that virus evolution has achieved a charge distribution across the capsid surface that (i) permits the packing of NTD domains in the outer layer of the Gag shell, (ii) directs the maturational rearrangements of the NTDs that yield a functional core structure, and (iii) supports capsid function during the early stages of virus infection. IMPORTANCE: The production of infectious retrovirus particles is a complex process, a choreography of protein and nucleic acid that occurs in two distinct stages: formation and release from the cell of an immature particle followed by an extracellular maturation phase during which the virion proteins and nucleic acids undergo major rearrangements that activate the infectious potential of the virion. This study examines the contributions of charged amino acids on the surface of the Rous sarcoma virus capsid protein in the assembly of appropriately formed immature particles and the maturational transitions that create a functional virion. The results provide important biological evidence in support of recent structural models of the RSV immature virions and further suggest that immature particle assembly and virion maturation are controlled by an extensive network of electrostatic interactions and long-range communication across the capsid surface.


Asunto(s)
Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Productos del Gen gag/química , Virus del Sarcoma de Rous/química , Virus del Sarcoma de Rous/fisiología , Ensamble de Virus , Secuencia de Aminoácidos , Cápside/metabolismo , Proteínas de la Cápside/genética , Microscopía por Crioelectrón , Productos del Gen gag/genética , Microscopía Electrónica , Modelos Moleculares , Mutación , Virus del Sarcoma de Rous/genética , Virus del Sarcoma de Rous/ultraestructura , Electricidad Estática , Virión/metabolismo , Virión/ultraestructura
17.
Nature ; 530(7590): 362-6, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26887497

RESUMEN

Integration of the reverse-transcribed viral DNA into the host genome is an essential step in the life cycle of retroviruses. Retrovirus integrase catalyses insertions of both ends of the linear viral DNA into a host chromosome. Integrase from HIV-1 and closely related retroviruses share the three-domain organization, consisting of a catalytic core domain flanked by amino- and carboxy-terminal domains essential for the concerted integration reaction. Although structures of the tetrameric integrase-DNA complexes have been reported for integrase from prototype foamy virus featuring an additional DNA-binding domain and longer interdomain linkers, the architecture of a canonical three-domain integrase bound to DNA remained elusive. Here we report a crystal structure of the three-domain integrase from Rous sarcoma virus in complex with viral and target DNAs. The structure shows an octameric assembly of integrase, in which a pair of integrase dimers engage viral DNA ends for catalysis while another pair of non-catalytic integrase dimers bridge between the two viral DNA molecules and help capture target DNA. The individual domains of the eight integrase molecules play varying roles to hold the complex together, making an extensive network of protein-DNA and protein-protein contacts that show both conserved and distinct features compared with those observed for prototype foamy virus integrase. Our work highlights the diversity of retrovirus intasome assembly and provides insights into the mechanisms of integration by HIV-1 and related retroviruses.


Asunto(s)
ADN Viral/química , Integrasas/química , Virus del Sarcoma de Rous/química , Virus del Sarcoma de Rous/enzimología , Dominio Catalítico , Cristalografía por Rayos X , ADN Viral/metabolismo , VIH-1/enzimología , VIH-1/metabolismo , Integrasas/metabolismo , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Virus del Sarcoma de Rous/genética , Virus del Sarcoma de Rous/metabolismo , Spumavirus/enzimología , Integración Viral
18.
Gene Ther ; 23(1): 67-77, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26204498

RESUMEN

Hepatocellular carcinoma develops in cirrhotic liver. The nitric oxide (NO) synthase type III (NOS-3) overexpression induces cell death in hepatoblastoma cells. The study developed gene therapy designed to specifically overexpress NOS-3 in cultured hepatoma cells, and in tumors derived from orthotopically implanted tumor cells in fibrotic livers. Liver fibrosis was induced by CCl4 administration in mice. The first-generation adenoviruses were designed to overexpress NOS-3 or green fluorescent protein, and luciferase complementary DNA under the regulation of murine alpha-fetoprotein (AFP) and Rous Sarcoma Virus (RSV) promoters, respectively. Both adenovirus and Hepa 1-6 cells were used for in vitro and in vivo experiments. Adenoviruses were administered through the tail vein 2 weeks after orthotopic tumor cell implantation. AFP-NOS-3/RSV-luciferase increased oxidative-related DNA damage, p53, CD95/CD95L expression and caspase-8, -9 and -3 activities in cultured Hepa 1-6 cells. The increased expression of CD95/CD95L and caspase-8 activity was abolished by Nω-nitro-l-arginine methyl ester hydrochloride, p53 and CD95 small interfering RNA. AFP-NOS-3/RSV-luciferase adenovirus increased cell death markers, and reduced cell proliferation of established tumors in fibrotic livers. The increase of oxidative/nitrosative stress induced by NOS-3 overexpression induced DNA damage, p53, CD95/CD95L expression and cell death in hepatocellular carcinoma cells. The effectiveness of the gene therapy has been demonstrated in vitro and in vivo.


Asunto(s)
Carcinoma Hepatocelular/terapia , Regulación Neoplásica de la Expresión Génica , Terapia Genética/métodos , Neoplasias Hepáticas/terapia , Óxido Nítrico Sintasa de Tipo III/genética , Adenoviridae/genética , Animales , Carcinoma Hepatocelular/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Línea Celular Tumoral , Proliferación Celular , Daño del ADN , ADN Complementario/genética , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Vectores Genéticos , Hígado/citología , Hígado/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/terapia , Neoplasias Hepáticas/genética , Ratones , NG-Nitroarginina Metil Éster/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Virus del Sarcoma de Rous/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo
19.
J Virol ; 90(5): 2473-85, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26676779

RESUMEN

UNLABELLED: The principles underlying membrane binding and assembly of retroviral Gag proteins into a lattice are understood. However, little is known about how these processes are related. Using purified Rous sarcoma virus Gag and Gag truncations, we studied the interrelation of Gag-Gag interaction and Gag-membrane interaction. Both by liposome binding and by surface plasmon resonance on a supported bilayer, Gag bound to membranes much more tightly than did matrix (MA), the isolated membrane binding domain. In principle, this difference could be explained either by protein-protein interactions leading to cooperativity in membrane binding or by the simultaneous interaction of the N-terminal MA and the C-terminal nucleocapsid (NC) of Gag with the bilayer, since both are highly basic. However, we found that NC was not required for strong membrane binding. Instead, the spacer peptide assembly domain (SPA), a putative 24-residue helical sequence comprising the 12-residue SP segment of Gag and overlapping the capsid (CA) C terminus and the NC N terminus, was required. SPA is known to be critical for proper assembly of the immature Gag lattice. A single amino acid mutation in SPA that abrogates assembly in vitro dramatically reduced binding of Gag to liposomes. In vivo, plasma membrane localization was dependent on SPA. Disulfide cross-linking based on ectopic Cys residues showed that the contacts between Gag proteins on the membrane are similar to the known contacts in virus-like particles. Taken together, we interpret these results to mean that Gag membrane interaction is cooperative in that it depends on the ability of Gag to multimerize. IMPORTANCE: The retroviral structural protein Gag has three major domains. The N-terminal MA domain interacts directly with the plasma membrane (PM) of cells. The central CA domain, together with immediately adjoining sequences, facilitates the assembly of thousands of Gag molecules into a lattice. The C-terminal NC domain interacts with the genome, resulting in packaging of viral RNA. For assembly in vitro with purified Gag, in the absence of membranes, binding of NC to nucleic acid somehow facilitates further Gag-Gag interactions that lead to formation of the Gag lattice. The contributions of MA-mediated membrane binding to virus particle assembly are not well understood. Here, we report that in the absence of nucleic acid, membranes provide a platform that facilitates Gag-Gag interactions. This study demonstrates that the binding of Gag, but not of MA, to membranes is cooperative and identifies SPA as a major factor that controls this cooperativity.


Asunto(s)
Productos del Gen gag/metabolismo , Membrana Dobles de Lípidos/metabolismo , Multimerización de Proteína , Virus del Sarcoma de Rous/fisiología , Análisis Mutacional de ADN , Productos del Gen gag/genética , Unión Proteica , Estructura Terciaria de Proteína , Virus del Sarcoma de Rous/genética
20.
Pharm Res ; 32(11): 3699-707, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26047779

RESUMEN

PURPOSE: Virus-like particles (VLPs) have been used as drug carriers for drug delivery systems. In this study, hCC49 single chain fragment variable (scFv)-displaying Rous sarcoma virus-like particles (RSV VLPs) were produced in silkworm larvae to be a specific carrier of an anti-cancer drug. METHOD: RSV VLPs displaying hCC49 scFv were created by the fusion of the transmembrane and cytoplasmic domains of hemagglutinin from influenza A (H1N1) virus and produced in silkworm larvae. The display of hCC49 scFv on the surface of RSV VLPs was confirmed by enzyme-linked immunosorbent assay using tumor-associated glycoprotein-72 (TAG-72), fluorescent microscopy, and immunoelectron microscopy. Fluorescein isothiocyanate (FITC) or doxorubicin (DOX) was incorporated into hCC49 scFv-displaying RSV VLPs by electroporation and specific targeting of these VLPs was investigated by fluorescent microscopy and cytotoxicity assay using LS174T cells. RESULTS: FITC was delivered to LS174T human colon adenocarcinoma cells by hCC49 scFv-displaying RSV VLPs, but not by RSV VLPs. This indicated that hCC49 scFv allowed FITC-loaded RSV VLPs to be delivered to LS174T cells. DOX, which is an anti-cancer drug with intrinsic red fluorescence, was also loaded into hCC49 scFv-displaying RSV VLPs by electroporation; the DOX-loaded hCC49 scFv-displaying RSV VLPs killed LS174T cells via the specific delivery of DOX that was mediated by hCC49 scFv. HEK293 cells were alive even though in the presence of DOX-loaded hCC49 scFv-displaying RSV VLPs. CONCLUSION: These results showed that hCC49 scFv-displaying RSV VLPs from silkworm larvae offered specific drug delivery to colon carcinoma cells in vitro. This scFv-displaying enveloped VLP system could be applied to drug and gene delivery to other target cells.


Asunto(s)
Anticuerpos Antineoplásicos/genética , Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Virus del Sarcoma de Rous/genética , Anticuerpos de Cadena Única/genética , Virión/genética , Animales , Bombyx/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/patología , Portadores de Fármacos , Productos del Gen gag/metabolismo , Células HEK293 , Humanos , Larva/genética , Virus del Sarcoma de Rous/metabolismo , Vacunas de Partículas Similares a Virus/genética , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...