Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nucleic Acids Res ; 48(14): 8178-8187, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32619241

RESUMEN

The application of gene-editing technology is currently limited by the lack of safe and efficient methods to deliver RNA-guided endonucleases to target cells. We engineered lentivirus-based nanoparticles to co-package the U6-sgRNA template and the CRISPR-associated protein 9 (Cas9) fused with a virion-targeted protein Vpr (Vpr.Prot.Cas9), for simultaneous delivery to cells. Equal spatiotemporal control of the vpr.prot.cas9 and gag/pol gene expression (the presence of Rev responsive element, RRE) greatly enhanced the encapsidation of the fusion protein and resulted in the production of highly efficient lentivector nanoparticles. Transduction of the unconcentrated, Vpr.Prot.Cas9-containing vectors led to >98% disruption of the EGFP gene in reporter HEK293-EGFP cells with minimal cytotoxicity. Furthermore, we detected indels in the targeted endogenous loci at frequencies of up to 100% in cell lines derived from lymphocytes and monocytes and up to 15% in primary CD4+ T cells by high-throughput sequencing. This approach may provide a platform for the efficient, dose-controlled and tissue-specific delivery of genome editing enzymes to cells and it may be suitable for simultaneous endogenous gene disruption and a transgene delivery.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas , Edición Génica/métodos , Elementos de Respuesta , Proteína 9 Asociada a CRISPR/genética , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Lentivirus/genética , Nanopartículas/química , Células THP-1 , Transducción Genética/métodos
2.
PLoS Pathog ; 15(2): e1007533, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30768644

RESUMEN

Retroviruses have evolved multiple means to counteract host restriction factors such as single-stranded DNA-specific deoxycytidine deaminases (APOBEC3s, A3s). These include exclusion of A3s from virions by an A3-unreactive nucleocapsid or expression of an A3-neutralizing protein (Vif, Bet). However, a number of retroviruses package A3s and do not encode apparent vif- or bet-like genes, yet they replicate in the presence of A3s. The mode by which they overcome deleterious restriction remains largely unknown. Here we show that the prototypic betaretrovirus, mouse mammary tumor virus (MMTV), packages similar amounts of A3s as HIV-1ΔVif, yet its proviruses carry a significantly lower level of A3-mediated deamination events than the lentivirus. The G-to-A mutation rate increases when the kinetics of reverse transcription is reduced by introducing a mutation (F120L) to the DNA polymerase domain of the MMTV reverse transcriptase (RT). A similar A3-sensitizing effect was observed when the exposure time of single-stranded DNA intermediates to A3s during reverse transcription was lengthened by reducing the dNTP concentration or by adding suboptimal concentrations of an RT inhibitor to infected cells. Thus, the MMTV RT has evolved to impede access of A3s to transiently exposed minus DNA strands during reverse transcription, thereby alleviating inhibition by A3 family members. A similar mechanism may be used by other retroviruses and retrotransposons to reduce deleterious effects of A3 proteins.


Asunto(s)
Citidina Desaminasa/genética , Citosina Desaminasa/genética , Virus del Tumor Mamario del Ratón/genética , Desaminasas APOBEC , Desaminasa APOBEC-3G/genética , Desaminasa APOBEC-3G/metabolismo , Animales , Línea Celular , Citidina Desaminasa/metabolismo , Citosina Desaminasa/metabolismo , ADN , ADN de Cadena Simple , Células HEK293 , Células HeLa , Humanos , Virus del Tumor Mamario del Ratón/crecimiento & desarrollo , Virus del Tumor Mamario del Ratón/patogenicidad , Ratones , Mutación/genética , Nucleocápside , Polimerizacion , Unión Proteica , Retroviridae , Transcripción Reversa/genética , Virión
3.
J Gen Virol ; 98(9): 2362-2367, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28809145

RESUMEN

Infectivity of the mouse mammary tumour virus (MMTV) is inhibited by mouse APOBEC3 (mA3) which is efficiently packaged into virions. As the inhibition is only partial, the virus can replicate in tissues expressing mA3 and complete its replication cycle. Here, we have examined the sensitivity of MMTV to inhibition by a human orthologue of mA3, A3G. We report that the virus containing A3G is only moderately susceptible to inhibition by the human factor. Whereas the vif-deficient HIV-1 vector produced in human epithelial cells expressing endogenous levels of A3G was efficiently inhibited, an MMTV vector remained fully infectious. Greater A3G expression levels were necessary to restrict infectivity of MMTV, but only when the factor retained its deaminase activity. Furthermore, the spreading kinetic of a replication competent MMTV was only moderately accelerated in cells with downmodulated A3G expression. These data suggest that MMTV has evolved a mechanism to neutralize antiviral activity of APOBEC3 proteins.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , Virus del Tumor Mamario del Ratón/fisiología , Infecciones por Retroviridae/veterinaria , Enfermedades de los Roedores/enzimología , Desaminasa APOBEC-3G/genética , Animales , Humanos , Virus del Tumor Mamario del Ratón/genética , Ratones , Infecciones por Retroviridae/enzimología , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/virología , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/virología , Ensamble de Virus , Replicación Viral
4.
Retrovirology ; 12: 43, 2015 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-25980759

RESUMEN

BACKGROUND: Mouse mammary tumour virus (MMTV) is a betaretrovirus that infects rodent cells and uses mouse tranferrin receptor 1 (TfR1) for cell entry. Several MMTV strains have been shown to productively infect, in addition to murine cells, various heterologous cell lines including those of human origin, albeit less efficiently than murine cells. Furthermore, there have been reports that the continued passage of MMTV in heterologous cell lines gives rise to novel variants that are able to infect naive non-murine cells with higher efficiency than the parental virus. RESULTS: We show that MMTV(C3H), like other MMTV strains, that had undergone a number of replication cycles in non-murine cells displayed an increased replication kinetic, as compared to parental virus, when applied on naive human cells. Sequence analysis of several replication kinetic variants and the parental virus, together with calculation of the ratio of non-synonymous to synonymous mutations at individual codons, revealed that several regions within the viral genome were under strong positive selection pressure during viral replication in human cells. The mutation responsible, at least in part, for the phenotypic change was subsequently mapped to the segment of env encoding the receptor binding site (F40HGFR44). Introduction of the identified mutation, leading to single amino acid substitution (G42E), into egfp-containing recombinant MMTV virions enhanced their ability to bind to and infect human cells. Interestingly, neither the replication kinetic mutant nor the parental virus required human TfR1 for infection. Knock-out of TFR1 gene from the human genome did not decrease the susceptibility of Hs578T cells to virus infection. Furthermore, the expression of human TfR1, in contrast to mouse TfR1, did not enhance the susceptibility of MMTV-resistant Chinese hamster ovary cells. Thus, human TfR1 is dispensable for infection and another cell surface molecule mediates the MMTV entry into human cells. CONCLUSION: Taken together, our data explain the mechanism enabling MMTV to form 'host-range variants' in non-murine cells that has been known for a long time, the basis of which remained obscure. Our findings may expand our understanding of how viruses gain capability to cross species-specific barriers to infect new hosts.


Asunto(s)
Especificidad del Huésped , Virus del Tumor Mamario del Ratón/fisiología , Proteínas Mutantes/metabolismo , Mutación Missense , Receptores de Transferrina/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Sustitución de Aminoácidos , Animales , Línea Celular , Humanos , Proteínas Mutantes/genética , Pase Seriado , Proteínas del Envoltorio Viral/genética , Replicación Viral
5.
J Gen Virol ; 96(Pt 3): 650-662, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25491421

RESUMEN

Mouse mammary tumour virus (MMTV) is a member of the genus Betaretrovirus, infects rodent cells and uses mouse tranferrin receptor 1 for cell entry. Several MMTV strains have been shown to productively infect, in addition to murine cells, various heterologous cell lines including those of human origin, albeit less efficiently than murine cells. Here, we analysed whether MMTV from C3H mice [MMTV(C3H)], reported previously to be incapable of infecting human cells, could productively infect human cells. Using a recently described high-titre MMTV-based vector carrying MMTV(C3H) envelope protein (Env), we successfully transduced cells of human origin. Furthermore, WT MMTV(C3H) was able to infect human cells, albeit less efficiently than mouse cells. The established infection was, however, sufficient to enable virus spread to every cell in culture. The infectivity of WT MMTV(C3H) and MMTV-based vectors carrying MMTV(C3H)Env was blocked by heat inactivation, an inhibitor of reverse transcription (3'-azido-3'-deoxythymidine) and pre-incubation with neutralizing anti-MMTV antibodies that did not neutralize vectors pseudotyped with amphotropic murine leukemia virus Env, providing evidence for an authentic, receptor-mediated and reverse transcriptase-dependent infection process. Persistently infected human Hs578T cells produced infectious virions capable of infecting naïve human breast cells in culture, the infectivity of which could also be blocked by neutralizing anti-MMTV antibodies, demonstrating that virus particles released by the persistently infected Hs578T cells were related antigenically to the virus produced from murine cells. Taken together, our results show that MMTV(C3H), like MMTV(GR) and MMTV(RIII), is able not only to infect but also to replicate in cultured human breast cells.


Asunto(s)
Glándulas Mamarias Humanas/virología , Virus del Tumor Mamario del Ratón/fisiología , Infecciones Tumorales por Virus/virología , Animales , ADN Viral/genética , Regulación Viral de la Expresión Génica/fisiología , Vectores Genéticos , Humanos , Virus del Tumor Mamario del Ratón/clasificación , Ratones , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
6.
Retrovirology ; 11: 34, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24779422

RESUMEN

BACKGROUND: Mouse mammary tumor virus (MMTV) is a complex, milk-born betaretrovirus, which preferentially infects dendritic cells (DC) in the gastrointestinal tract and then spreads to T and B lymphocytes and finally to the mammary gland. It is not clear how the prototypic betaretrovirus infects mucosal DCs and naïve lymphocytes as these cells are considered to be non-proliferative. Studies of MMTV biology have been hampered by the difficulty of obtaining sufficient virus/vector titers after transfection of a molecular clone in cultured cells. To surmount this barrier we developed a novel MMTV-based vector system with a split genome design containing potent posttranscriptional regulatory functions. RESULTS: Using this system, vector particles were produced to markedly greater titers (>1000-fold) than those obtained previously. The titers (>106 transduction units /ml) were comparable to those achieved with lentiviral or gammaretroviral vectors. Importantly, the vector transduced the enhanced green fluorescence protein gene into the chromosomes of non-dividing cells, such as cells arrested at the G2/M phase of the cell cycle and unstimulated hematopoietic progenitor cells, at an efficiency similar to that obtained with the HIV-1-based vector. In contrast to HIV-1, MMTV transductions were not affected by knocking down the expression of a factor involved in nuclear import of the HIV-1 pre-integration complexes, TNPO3. In contrast to HIV-1, the MMTV-based vector did not preferentially integrate in transcription units. Additionally, no preference for integration near transcription start sites, the regions preferentially targeted by gammaretroviral vectors, was observed. The vector derived from MMTV exhibits a random integration pattern. CONCLUSIONS: Overall, the betaretroviral vector system should facilitate molecular virology studies of the prototypic betaretrovirus as well as studies attempting to elucidate fundamental cellular processes such as nuclear import pathways. Random integration in cycling and non-cycling cells may be applicable in unbiased gene delivery.


Asunto(s)
Núcleo Celular/genética , Vectores Genéticos/genética , Virus del Tumor Mamario del Ratón/genética , Integración Viral/genética , beta Carioferinas/genética , Animales , Línea Celular , Línea Celular Tumoral , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Expresión Génica , Proteínas Fluorescentes Verdes , Células HEK293 , VIH-1/genética , Células HeLa , Humanos , Ratones , Procesamiento Postranscripcional del ARN/genética , Retroviridae/genética , Sitio de Iniciación de la Transcripción/fisiología , Transfección/métodos
7.
J Gen Virol ; 93(Pt 2): 308-318, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22113011

RESUMEN

Mouse mammary tumor virus (MMTV) is a complex betaretrovirus, which utilizes a Rev-like auxiliary protein Rem to export the unspliced viral RNA from the nucleus. MMTV env mRNA appears to be exported via a distinct, Rem-independent, mechanism. Here, we analysed the effect of an extensively folded region coinciding with the 5' leader sequence on env gene expression. We found that the presence of the 5' leader stimulates expression of the envelope protein. Enhanced Env production was accompanied by increased cytoplasmic levels of env mRNA. The 5' leader promotes nucleocytoplasmic translocation and increases stability of env mRNA. The region responsible for this effect was mapped to the distal part of the 5' leader. Furthermore, the 5' leader inserted in the sense orientation into a heterologous luciferase expression construct increased luciferase activity.


Asunto(s)
Regiones no Traducidas 5' , Virus del Tumor Mamario del Ratón/fisiología , Proteínas del Envoltorio Viral/biosíntesis , Replicación Viral , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Genes Reporteros , Humanos , Virus del Tumor Mamario del Ratón/genética , Modelos Moleculares , Conformación de Ácido Nucleico , Estabilidad del ARN , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas del Envoltorio Viral/genética
8.
Viruses ; 14(5)2022 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-35632628

RESUMEN

A human betaretrovirus (HBRV) has been linked with the autoimmune liver disease, primary biliary cholangitis (PBC), and various cancers, including breast cancer and lymphoma. HBRV is closely related to the mouse mammary tumor virus, and represents the only exogenous betaretrovirus characterized in humans to date. Evidence of infection in patients with PBC has been demonstrated through the identification of proviral integration sites in lymphoid tissue, the major reservoir of infection, as well as biliary epithelium, which is the site of the disease process. Accordingly, we tested the hypothesis that patients with PBC harbor a transmissible betaretrovirus by co-cultivation of PBC patients' lymph node homogenates with the HS578T breast cancer line. Because of the low level of HBRV replication, betaretrovirus producing cells were subcloned to optimize viral isolation and production. Evidence of infection was provided by electron microscopy, RT-PCR, in situ hybridization, cloning of the HBRV proviral genome and demonstration of more than 3400 integration sites. Further evidence of viral transmissibility was demonstrated by infection of biliary epithelial cells. While HBRV did not show a preference for integration proximal to specific genomic features, analyses of common insertion sites revealed evidence of integration proximal to cancer associated genes. These studies demonstrate the isolation of HBRV with features similar to mouse mammary tumor virus and confirm that patients with PBC display evidence of a transmissible viral infection.


Asunto(s)
Betaretrovirus , Neoplasias de la Mama , Cirrosis Hepática Biliar , Animales , Femenino , Humanos , Cirrosis Hepática Biliar/etiología , Virus del Tumor Mamario del Ratón/genética , Ratones , Provirus/genética
9.
Nucleic Acids Res ; 36(19): 6284-94, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18835854

RESUMEN

Mouse mammary tumor virus (MMTV) has previously been shown to encode a functional homolog of the human immunodeficiency virus-1 (HIV-1) nuclear export protein Rev, termed Rem. Here, we show that deletion of the rem gene from a MMTV molecular clone interfered with the nucleo-cytoplasmic transport of genomic length viral mRNA and resulted in a loss of viral capsid (Gag) protein production. Interestingly, nuclear export of single-spliced env mRNA was only moderately affected, suggesting that this transcript is, at least to some extent, transported via a distinct, Rem-independent export mechanism. To identify and characterize a cis-acting RNA element required for Rem responsiveness (RmRE), extensive computational and functional analyses were performed. By these means a region of 490 nt corresponding to positions nt 8517-nt 9006 in the MMTV reference strain was identified as RmRE. Deletion of this fragment, which spans the env-U3 junction region, abolished Gag expression. Furthermore, insertion of this sequence into a heterologous HIV-1-based reporter construct restored, in the presence of Rem, HIV-1 Gag expression to levels determined for the Rev/RRE export system. These results clearly demonstrate that the identified region, whose geometry resembles that of other retroviral-responsive elements, is capable to functionally substitute, in the presence of Rem, for Rev/RRE and thus provide unequivocal evidence that MMTV is a complex retrovirus.


Asunto(s)
Virus del Tumor Mamario del Ratón/genética , ARN Viral/química , Proteínas de Unión al ARN/metabolismo , Secuencias Reguladoras de Ácido Ribonucleico , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Animales , Sitios de Unión , Gatos , Línea Celular , Núcleo Celular/metabolismo , Biología Computacional , Genes Reporteros , VIH-1/genética , ARN Viral/metabolismo , Eliminación de Secuencia , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
10.
J Virol ; 82(3): 1360-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18032509

RESUMEN

Based on integration site preferences, retroviruses can be placed into three groups. Viruses that comprise the first group, murine leukemia virus and foamy virus, integrate preferentially near transcription start sites. The second group, notably human immunodeficiency virus and simian immunodeficiency virus, preferentially targets transcription units. Avian sarcoma-leukosis virus (ASLV) and human T-cell leukemia virus (HTLV), forming the third group, show little preference for any genomic feature. We have previously shown that some human cells sustain mouse mammary tumor virus (MMTV) infection; therefore, we infected a susceptible human breast cell line, Hs578T, and, without introducing a species-specific bias, compared the MMTV integration profile to those of other retroviruses. Additionally, we infected a mouse cell line, NMuMG, and thus we could compare MMTV integration site selection in human and mouse cells. In total, we examined 468 unique MMTV integration sites. Irrespective of whether human or mouse cells were infected, no integration bias favoring transcription start sites was detected, a profile that is reminiscent of that of ASLV and HTLV. However, in contrast to ASLV and HTLV, not even a modest tendency in favor of integration within genes was observed. Similarly, repetitive sequences and genes that are frequently tagged by MMTV in mammary tumors were not preferentially targeted in cell culture either in mouse or in human cells; hence, we conclude that MMTV displays the most random dispersion of integration sites among retroviruses determined so far.


Asunto(s)
Virus del Tumor Mamario del Ratón/fisiología , Integración Viral/fisiología , Animales , Línea Celular Tumoral , Humanos , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Integración Viral/genética
11.
Retrovirology ; 4: 73, 2007 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-17931409

RESUMEN

BACKGROUND: The role of mouse mammary tumor virus (MMTV) as a causative agent in human breast carcinogenesis has recently been the subject of renewed interest. The proposed model is based on the detection of MMTV sequences in human breast cancer but not in healthy breast tissue. One of the main drawbacks to this model, however, was that until now human cells had not been demonstrated to sustain productive MMTV infection. RESULTS: Here, we show for the first time the rapid spread of mouse mammary tumor virus, MMTV(GR), in cultured human mammary cells (Hs578T), ultimately leading to the infection of every cell in culture. The replication of the virus was monitored by quantitative PCR, quantitative RT-PCR and immunofluorescence imaging. The infected human cells expressed, upon cultivation with dexamethasone, MMTV structural proteins and released spiked B-type virions, the infectivity of which could be neutralized by anti-MMTV antibody. Replication of the virus was efficiently blocked by an inhibitor of reverse transcription, 3'-azido-3'-deoxythymidine. The human origin of the infected cells was confirmed by determining a number of integration sites hosting the provirus, which were unequivocally identified as human sequences. CONCLUSION: Taken together, our results show that human cells can support replication of mouse mammary tumor virus.


Asunto(s)
Neoplasias de la Mama/virología , Virus del Tumor Mamario del Ratón/efectos de los fármacos , Animales , Línea Celular , ADN Viral/análisis , ADN Viral/genética , Humanos , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/fisiología , Reacción en Cadena de la Polimerasa/métodos , Infecciones Tumorales por Virus/inmunología , Replicación Viral/efectos de los fármacos , Zidovudina/farmacología
12.
Cancer Res ; 65(15): 6651-9, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16061645

RESUMEN

Mouse mammary tumor virus (MMTV) has long been speculated to be involved in human breast cancer and more recently in human primary biliary cirrhosis. Despite complete proviral sequences markedly homologous to MMTV being identified in human breast cancer tissue, no convincing evidence has been presented to date that MMTV can infect human cells. Using both wild-type and a genetically marked virus (MMTV-EGFP), we show here the successful infection of a number of different human cells by MMTV. Furthermore, infection of human cells is shown to be almost as efficient as the infection of murine mammary epithelial cells. Sequencing of PCR products from integrated proviruses reveals that reverse transcription and integration of the viral genome has occurred as expected. Furthermore, sequencing of two independent MMTV proviral integration sites reveal them to be present only in the human and not in the mouse genome. Infection requires an intact MMTV envelope protein and is blocked either by heat inactivation of the virus or by specific neutralizing anti-MMTV serum, ruling out a nonspecific mechanism of viral transfer. Thus, MMTV can infect human cells and this finding provides a possible explanation for the detection by others of MMTV sequences in human breast cancer patients.


Asunto(s)
Virus del Tumor Mamario del Ratón/patogenicidad , Animales , Secuencia de Bases , Neoplasias de la Mama/virología , Gatos , Línea Celular , Productos del Gen env/genética , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Riñón/citología , Riñón/virología , Virus del Tumor Mamario del Ratón/genética , Ratones , Datos de Secuencia Molecular , Provirus/genética , Provirus/patogenicidad , Transfección , Virión/genética , Virión/patogenicidad , Inactivación de Virus , Integración Viral
13.
Sci Rep ; 7: 44459, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28290554

RESUMEN

A multitude of viral factors - either inhibiting the induction of the IFN-system or its effectors - have been described to date. However, little is known about the role of structural components of the incoming virus particle in protecting against IFN-induced antiviral factors during or immediately after entry. In this study, we take advantage of the previously reported property of Classical swine fever virus (family Flaviviridae, genus Pestivirus) to tolerate a deletion of the core protein if a compensatory mutation is present in the NS3-helicase-domain (Vp447∆c). In contrast to the parental virus (Vp447), which causes a hemorrhagic-fever-like disease in pigs, Vp447∆c is avirulent in vivo. In comparison to Vp447, growth of Vp447∆c in primary porcine cells and IFN-treated porcine cell lines was reduced >20-fold. Also, primary porcine endothelial cells and IFN-pretreated porcine cell lines were 8-24 times less susceptible to Vp447∆c. This reduction of susceptibility could be partially reversed by loading Vp447∆c particles with different levels of core protein. In contrast, expression of core protein in the recipient cell did not have any beneficial effect. Therefore, a protective effect of core protein in the incoming virus particle against the products of IFN-stimulated genes could be demonstrated.


Asunto(s)
Virus de la Fiebre Porcina Clásica/genética , Infecciones por Pestivirus/genética , Pestivirus/genética , Proteínas del Núcleo Viral/genética , Animales , Virus de la Fiebre Porcina Clásica/patogenicidad , Virus ADN/genética , Pestivirus/patogenicidad , Infecciones por Pestivirus/virología , Porcinos/virología , Replicación Viral/genética
14.
Vet Microbiol ; 117(2-4): 276-83, 2006 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-16879934

RESUMEN

Astroviral infection has been described as one of the causes of porcine diarrhoeal disease. Here we describe the detection of astrovirus-like particles by electron microscopy in a diarrhoeal specimen. Furthermore, a cytopathic virus was isolated and propagated in an established porcine kidney cell line, PK-15. Reverse transcription and PCR performed with astrovirus-specific primers amplified a product with the expected size. Sequencing of the PCR product revealed that the virus observed by electron microscopy and propagated in the porcine cell line is an astrovirus, showing 86% identity at the nucleotide level with the only known porcine astrovirus, PAstV. Phylogenetic analysis clustered the novel isolate, Sb4685, together with PAstV in a broad clade comprising mammalian astroviruses.


Asunto(s)
Infecciones por Astroviridae/veterinaria , Astroviridae/aislamiento & purificación , Animales , Astroviridae/clasificación , Astroviridae/ultraestructura , Secuencia de Bases , Línea Celular , Análisis por Conglomerados , Diarrea/veterinaria , Diarrea/virología , Amplificación de Genes , Microscopía Electrónica/veterinaria , Peso Molecular , Filogenia , ARN Viral/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Porcinos
15.
PLoS One ; 11(11): e0164639, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27828961

RESUMEN

European honey bees are highly important in crop pollination, increasing the value of global agricultural production by billions of dollars. Current knowledge about virulence and pathogenicity of Deformed wing virus (DWV), a major factor in honey bee colony mortality, is limited. With this study, we close the gap between field research and laboratory investigations by establishing a complete in vitro model for DWV pathogenesis. Infectious DWV was rescued from a molecular clone of a DWV-A genome that induces DWV symptoms such as crippled wings and discoloration. The expression of DWV proteins, production of infectious virus progeny, and DWV host cell tropism could be confirmed using newly generated anti-DWV monoclonal antibodies. The recombinant RNA fulfills Koch's postulates circumventing the need of virus isolation and propagation of pure virus cultures. In conclusion, we describe the development and application of a reverse genetics system for the study of DWV pathogenesis.


Asunto(s)
Virus de Insectos/genética , Picornaviridae/genética , Virus ARN/genética , ARN Viral/genética , Animales , Anticuerpos Monoclonales/inmunología , Secuencia de Bases , Abejas/virología , Western Blotting , Proteínas de la Cápside/inmunología , Genoma Viral/genética , Interacciones Huésped-Patógeno , Inmunohistoquímica , Virus de Insectos/metabolismo , Virus de Insectos/fisiología , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Filogenia , Picornaviridae/clasificación , Picornaviridae/metabolismo , Poliproteínas/genética , Poliproteínas/metabolismo , Pupa/virología , Virus ARN/metabolismo , Virus ARN/ultraestructura , ARN Viral/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Homología de Secuencia de Ácido Nucleico , Proteínas Virales/metabolismo , Alas de Animales/virología
16.
Vet Microbiol ; 107(3-4): 171-8, 2005 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-15863276

RESUMEN

Porcine reproductive and respiratory syndrome virus is a virus with a relatively high level of sequence variation, especially between the European and the American-type strains. This high degree of sequence variations will influence the accuracy of results obtained by molecular diagnostic methods, especially if mutations are located in the primer or probe binding regions of PCR or real-time PCR based assays. In order to provide the basis for the development of an accurate, region-tailored assay we have analysed samples obtained from several Austrian pig herds using a series of RT-PCR assays. Subsequent sequencing of the PCR products from ORF5 and phylogenetic analysis revealed the occurrence of both the European and the American-type of porcine reproductive and respiratory syndrome virus in Austria. Since vaccination with the American-type vaccine is not authorized in Austria the import of vaccinated animals is the most likely explanation for the occurrence of these strains. The study highlights the importance of a detailed study on the sequence variations occurring in the region of interest before the development of a reliable PCR-based assay.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina/diagnóstico , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Animales , Austria , Secuencia de Bases , Variación Genética , Datos de Secuencia Molecular , Sistemas de Lectura Abierta/genética , Filogenia , Virus del Síndrome Respiratorio y Reproductivo Porcino/clasificación , ARN Viral/química , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Alineación de Secuencia , Porcinos
17.
PLoS One ; 10(6): e0131515, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121257

RESUMEN

The Gag protein of the mouse mammary tumor virus (MMTV) is the chief determinant of subcellular targeting. Electron microscopy studies show that MMTV Gag forms capsids within the cytoplasm and assembles as immature particles with MMTV RNA and the Y box binding protein-1, required for centrosome maturation. Other betaretroviruses, such as Mason-Pfizer monkey retrovirus (M-PMV), assemble adjacent to the pericentriolar region because of a cytoplasmic targeting and retention signal in the Matrix protein. Previous studies suggest that the MMTV Matrix protein may also harbor a similar cytoplasmic targeting and retention signal. Herein, we show that a substantial fraction of MMTV Gag localizes to the pericentriolar region. This was observed in HEK293T, HeLa human cell lines and the mouse derived NMuMG mammary gland cells. Moreover, MMTV capsids were observed adjacent to centrioles when expressed from plasmids encoding either MMTV Gag alone, Gag-Pro-Pol or full-length virus. We found that the cytoplasmic targeting and retention signal in the MMTV Matrix protein was sufficient for pericentriolar targeting, whereas mutation of the glutamine to alanine at position 56 (D56/A) resulted in plasma membrane localization, similar to previous observations from mutational studies of M-PMV Gag. Furthermore, transmission electron microscopy studies showed that MMTV capsids accumulate around centrioles suggesting that, similar to M-PMV, the pericentriolar region may be a site for MMTV assembly. Together, the data imply that MMTV Gag targets the pericentriolar region as a result of the MMTV cytoplasmic targeting and retention signal, possibly aided by the Y box protein-1 required for the assembly of centrosomal microtubules.


Asunto(s)
Centriolos/metabolismo , Productos del Gen gag/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Regiones no Traducidas 5'/genética , Secuencia de Aminoácidos , Animales , Cápside/ultraestructura , Productos del Gen gag/química , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Secuencias Reguladoras de Ácidos Nucleicos/genética
18.
Virology ; 337(1): 1-6, 2005 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15914215

RESUMEN

We have identified a novel, multiple spliced, subgenomic mRNA species in MMTV producing cells of different origin containing an open reading frame encoding a 39-kDa Rev-like protein, Rem (regulator of expression of MMTV). An EGFP-Rem fusion protein is shown to be predominantly in the nucleolus. Further leptomycin B inhibits the nuclear export of nonspliced MMTV transcripts, implicating Rem in nuclear export by the Crm1 pathway in MMTV. Rem is thus reminiscent of the Rec protein from the related endogenous human retrovirus, HERV-K.


Asunto(s)
Productos del Gen rev/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Regulación Viral de la Expresión Génica , Productos del Gen rev/genética , Virus del Tumor Mamario del Ratón/genética , Ratones , Empalme del ARN , Proteínas Virales/genética
19.
J Gen Virol ; 81(Pt 10): 2497-2502, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10993939

RESUMEN

The major envelope glycoprotein genes (ORF5) of seven Czech isolates of porcine reproductive and respiratory syndrome virus (PRRSV) were amplified and their nucleotide sequences were determined. ORF5 displayed nucleotide and amino acid identities of 87.5-100% and 87. 6-100%, respectively, among the isolates. In a phylogenetic tree, all European isolates were grouped in a genotype distinct from that of reference American strains (VR-2332, IAF-Klop). Among the European isolates, two different clades were identified. Two Czech isolates (V-501 and V-503) and Italian strain PRRSV 2156 fell into one clade. The remaining European strains comprised the second clade. Surprisingly, two separately clustered strains (V-501 and V-516) were isolated from the same herd. Additionally, the possible effect of in vitro cultivation on the nucleotide sequence was analysed. Nine point mutations in the ORF5 region resulted from 152 in vitro passages of the V-502 isolate in MARC-145 cells.


Asunto(s)
Glicoproteínas de Membrana/genética , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Proteínas del Envoltorio Viral/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , República Checa , ADN Viral/química , Datos de Secuencia Molecular , Mutación Puntual , Alineación de Secuencia , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA