Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 247
Filtrar
1.
J Virol ; 94(12)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32238588

RESUMEN

Tetherin/BST-2 is an antiviral protein that blocks the release of enveloped viral particles by linking them to the membrane of producing cells. At first, BST-2 genes were described only in humans and other mammals. Recent work identified BST-2 orthologs in nonmammalian vertebrates, including birds. Here, we identify the BST-2 sequence in domestic chicken (Gallus gallus) for the first time and demonstrate its activity against avian sarcoma and leukosis virus (ASLV). We generated a BST-2 knockout in chicken cells and showed that BST-2 is a major determinant of an interferon-induced block of ASLV release. Ectopic expression of chicken BST-2 blocks the release of ASLV in chicken cells and of human immunodeficiency virus type 1 (HIV-1) in human cells. Using metabolic labeling and pulse-chase analysis of HIV-1 Gag proteins, we verified that chicken BST-2 blocks the virus at the release stage. Furthermore, we describe BST-2 orthologs in multiple avian species from 12 avian orders. Previously, some of these species were reported to lack BST-2, highlighting the difficulty of identifying sequences of this extremely variable gene. We analyzed BST-2 genes in the avian orders Galliformes and Passeriformes and showed that they evolve under positive selection. This indicates that avian BST-2 is involved in host-virus evolutionary arms races and suggests that BST-2 antagonists exist in some avian viruses. In summary, we show that chicken BST-2 has the potential to act as a restriction factor against ASLV. Characterizing the interaction of avian BST-2 with avian viruses is important in understanding innate antiviral defenses in birds.IMPORTANCE Birds are important hosts of viruses that have the potential to cause zoonotic infections in humans. However, only a few antiviral genes (called viral restriction factors) have been described in birds, mostly because birds lack counterparts of highly studied mammalian restriction factors. Tetherin/BST-2 is a restriction factor, originally described in humans, that blocks the release of newly formed virus particles from infected cells. Recent work identified BST-2 in nonmammalian vertebrate species, including birds. Here, we report the BST-2 sequence in domestic chicken and describe its antiviral activity against a prototypical avian retrovirus, avian sarcoma and leukosis virus (ASLV). We also identify BST-2 genes in multiple avian species and show that they evolve rapidly in birds, which is an important indication of their relevance for antiviral defense. Analysis of avian BST-2 genes will shed light on defense mechanisms against avian viral pathogens.


Asunto(s)
Proteínas Aviares/inmunología , Virus del Sarcoma Aviar/inmunología , Antígeno 2 del Estroma de la Médula Ósea/inmunología , Evolución Molecular , Galliformes/inmunología , Sarcoma Aviar/inmunología , Secuencia de Aminoácidos , Animales , Proteínas Aviares/genética , Virus del Sarcoma Aviar/genética , Virus del Sarcoma Aviar/patogenicidad , Antígeno 2 del Estroma de la Médula Ósea/genética , Línea Celular , Fibroblastos/inmunología , Fibroblastos/virología , Galliformes/genética , Galliformes/virología , Regulación de la Expresión Génica , Células HEK293 , VIH-1/genética , VIH-1/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Passeriformes/genética , Passeriformes/inmunología , Passeriformes/virología , Sarcoma Aviar/genética , Sarcoma Aviar/virología , Selección Genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Liberación del Virus , Replicación Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
2.
PLoS One ; 12(3): e0174557, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28358926

RESUMEN

Previously rodent preclinical research in gliomas frequently involved implantation of cell lines such as C6 and 9L into the rat brain. More recently, mouse models have taken over, the genetic manipulability of the mouse allowing the creation of genetically accurate models outweighed the disadvantage of its smaller brain size that limited time allowed for tumor progression. Here we illustrate a method that allows glioma formation in the rat using the replication competent avian-like sarcoma (RCAS) virus / tumor virus receptor-A (tv-a) transgenic system of post-natal cell type-specific gene transfer. The RCAS/tv-a model has emerged as a particularly versatile and accurate modeling technology by enabling spatial, temporal, and cell type-specific control of individual gene transformations and providing de novo formed glial tumors with distinct molecular subtypes mirroring human GBM. Nestin promoter-driven tv-a (Ntv-a) transgenic Sprague-Dawley rat founder lines were created and RCAS PDGFA and p53 shRNA constructs were used to initiate intracranial brain tumor formation. Tumor formation and progression were confirmed and visualized by magnetic resonance imaging (MRI) and spectroscopy. The tumors were analyzed using histopathological and immunofluorescent techniques. All experimental animals developed large, heterogeneous brain tumors that closely resembled human GBM. Median survival was 92 days from tumor initiation and 62 days from the first point of tumor visualization on MRI. Each tumor-bearing animal showed time dependent evidence of malignant progression to high-grade glioma by MRI and neurological examination. Post-mortem tumor analysis demonstrated the presence of several key characteristics of human GBM, including high levels of tumor cell proliferation, pseudopalisading necrosis, microvascular proliferation, invasion of tumor cells into surrounding tissues, peri-tumoral reactive astrogliosis, lymphocyte infiltration, presence of numerous tumor-associated microglia- and bone marrow-derived macrophages, and the formation of stem-like cell niches within the tumor. This transgenic rat model may enable detailed interspecies comparisons of fundamental cancer pathways and clinically relevant experimental imaging procedures and interventions that are limited by the smaller size of the mouse brain.


Asunto(s)
Encéfalo/diagnóstico por imagen , Glioma/genética , Nestina/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Proteína p53 Supresora de Tumor/genética , Animales , Virus del Sarcoma Aviar/genética , Virus del Sarcoma Aviar/patogenicidad , Encéfalo/patología , Encéfalo/virología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Ingeniería Genética , Glioma/diagnóstico por imagen , Glioma/patología , Glioma/virología , Humanos , Macrófagos/patología , Imagen por Resonancia Magnética , Ratones , Ratas , Ratas Transgénicas
3.
Cell Mol Life Sci ; 72(23): 4671-80, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26109426

RESUMEN

Human induced pluripotent stem cells (hiPSC) differentiate into multiple cell types. Selective cell targeting is often needed for analyzing gene function by overexpressing proteins in a distinct population of hiPSC-derived cell types and for monitoring cell fate in response to stimuli. However, to date, this has not been possible, as commonly used viruses enter the hiPSC via ubiquitously expressed receptors. Here, we report for the first time the application of a heterologous avian receptor, the tumor virus receptor A (TVA), to selectively transduce TVA(+) cells in a mixed cell population. Expression of the TVA surface receptor via genetic engineering renders cells susceptible for infection by avian leucosis virus (ALV). We generated hiPSC lines with this stably integrated, ectopic TVA receptor gene that expressed the receptor while retaining pluripotency. The undifferentiated hiPSC(TVA+) as well as their differentiating progeny could be infected by recombinant ALV (so-called RCAS virus) with high efficiency. Due to incomplete receptor blocking, even sequential infection of differentiating or undifferentiated TVA(+) cells was possible. In conclusion, the TVA/RCAS system provides an efficient and gentle gene transfer system for hiPSC and extends our possibilities for selective cell targeting and lineage tracing studies.


Asunto(s)
Proteínas Aviares/genética , Virus del Sarcoma Aviar/genética , Ingeniería Genética/métodos , Células Madre Pluripotentes Inducidas/virología , Receptores Virales/genética , Proteínas Aviares/metabolismo , Virus del Sarcoma Aviar/patogenicidad , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Elementos Transponibles de ADN , Citometría de Flujo/métodos , Vectores Genéticos , Humanos , Células Madre Pluripotentes Inducidas/citología , Receptores Virales/metabolismo
4.
J Virol ; 80(1): 353-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16352560

RESUMEN

The envelope of class I viruses can be a target for potent viral inhibitors, such as the human immunodeficiency virus type 1 (HIV-1) inhibitor enfuvirtide, which are derived from the C-terminal heptad repeat (HR2) of the transmembrane (TM) subunit. Resistance to an HR2-based peptide inhibitor of a model retrovirus, subgroup A of the Avian Sarcoma and Leukosis Virus genus (ASLV-A), was studied by examining mutants derived by viral passage in the presence of inhibitor. Variants with reduced sensitivity to inhibitor were readily selected in vitro. Sensitivity determinants were identified for 13 different isolates, all of which mapped to the TM subunit. These determinants were identified in two regions: (i) the N-terminal heptad repeat (HR1) and (ii) the N-terminal segment of TM, between the subunit cleavage site and the fusion peptide. The latter class of mutants identified a region outside of the predicted HR2-binding site that can significantly alter sensitivity to inhibitor. A subset of the HR1 mutants displayed the unanticipated ability to infect nonavian cells. This expanded tropism was associated with increased efficiency of envelope triggering by soluble receptor at low temperatures, as measured by protease sensitivity of the surface subunit (SU) of envelope. In addition, expanded tropism was linked for the most readily triggered mutants with increased sensitivity to neutralization by SU-specific antiserum. These observations depict a class of HR2 peptide-selected mutations with a reduced activation threshold, thereby allowing the utilization of alternative receptors for viral entry.


Asunto(s)
Virus de la Leucosis Aviar/efectos de los fármacos , Virus del Sarcoma Aviar/efectos de los fármacos , Péptidos/farmacología , Tropismo/efectos de los fármacos , Proteínas del Envoltorio Viral/fisiología , Virus de la Leucosis Aviar/patogenicidad , Virus del Sarcoma Aviar/patogenicidad , Línea Celular , Secuencias Repetitivas de Aminoácido , Tropismo/fisiología , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/química
5.
J Virol ; 78(24): 13430-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15564453

RESUMEN

Fusion proteins of enveloped viruses categorized as class I are typified by two distinct heptad repeat domains within the transmembrane subunit. These repeats are important structural elements that assemble into the six-helix bundles characteristic of the fusion-activated envelope trimer. Peptides derived from these domains can be potent and specific inhibitors of membrane fusion and virus infection. To facilitate our understanding of retroviral entry, peptides corresponding to the two heptad repeat domains of the avian sarcoma and leukosis virus subgroup A (ASLV-A) TM subunit of the envelope protein were characterized. Two peptides corresponding to the C-terminal heptad repeat (HR2), offset from one another by three residues, were effective inhibitors of infection, while two overlapping peptides derived from the N-terminal heptad repeat (HR1) were not. Analysis of envelope mutants containing substitutions within the HR1 domain revealed that a single amino acid change, L62A, significantly reduced sensitivity to peptide inhibition. Virus bound to cells at 4 degrees C became sensitive to peptide within the first 5 min of elevating the temperature to 37 degrees C and lost sensitivity to peptide after 15 to 30 min, consistent with a transient intermediate in which the peptide binding site is exposed. In cell-cell fusion experiments, peptide inhibitor sensitivity occurred prior to a fusion-enhancing low-pH pulse. Soluble receptor for ASLV-A induces a lipophilic character in the envelope which can be measured by stable liposome binding, and this activation was found to be unaffected by inhibitory HR2 peptide. Finally, receptor-triggered conformational changes in the TM subunit were also found to be unaffected by inhibitory peptide. These changes are marked by a dramatic shift in mobility on sodium dodecyl sulfate-polyacrylamide gel electrophoresis, from a subunit of 37 kDa to a complex of about 80 kDa. Biotinylated HR2 peptide bound specifically to the 80-kDa complex, demonstrating a surprisingly stable envelope conformation in which the HR2 binding site is exposed. These experiments support a model in which receptor interaction promotes formation of an envelope conformation in which the TM subunit is stably associated with its target membrane and is able to bind a C-terminal peptide.


Asunto(s)
Virus de la Leucosis Aviar/efectos de los fármacos , Virus del Sarcoma Aviar/efectos de los fármacos , Fusión de Membrana/efectos de los fármacos , Péptidos/farmacología , Proteínas Virales de Fusión/química , Secuencia de Aminoácidos , Animales , Virus de la Leucosis Aviar/patogenicidad , Virus del Sarcoma Aviar/patogenicidad , Fusión Celular , Línea Celular , Datos de Secuencia Molecular , Mutación , Péptidos/síntesis química , Péptidos/química , Secuencias Repetitivas de Aminoácido
6.
J Virol ; 78(24): 13489-500, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15564460

RESUMEN

The subgroup A to E avian sarcoma and leukosis viruses (ASLVs) are highly related and are thought to have evolved from a common ancestor. These viruses use distinct cell surface proteins as receptors to gain entry into avian cells. Chickens have evolved resistance to infection by the ASLVs. We have identified the mutations responsible for the block to virus entry in chicken lines resistant to infection by subgroup A ASLVs [ASLV(A)]. The tva genetic locus determines the susceptibility of chicken cells to ASLV(A) viruses. In quail, the ASLV(A) susceptibility allele tva(s) encodes two forms of the Tva receptor; these proteins are translated from alternatively spliced mRNAs. The normal cellular function of the Tva receptor is unknown; however, the extracellular domain contains a 40-amino-acid, cysteine-rich region that is homologous to the ligand binding region of the low-density lipoprotein receptor (LDLR) proteins. The chicken tva(s) cDNAs had not yet been fully characterized; we cloned the chicken tva cDNAs from two lines of subgroup A-susceptible chickens, line H6 and line 0. Two types of chicken tva(s) cDNAs were obtained. These cDNAs encode a longer and shorter form of the Tva receptor homologous to the Tva forms in quail. Two different defects were identified in cDNAs cloned from two different ASLV(A)-resistant inbred chickens, line C and line 7(2). Line C tva(r) contains a single base pair substitution, resulting in a cysteine-to-tryptophan change in the LDLR-like region of Tva. This mutation drastically reduces the binding affinity of Tva(R) for the ASLV(A) envelope glycoproteins. Line 7(2) tva(r2) contains a 4-bp insertion in exon 1 that causes a change in the reading frame, which blocks expression of the Tva receptor.


Asunto(s)
Virus de la Leucosis Aviar/patogenicidad , Virus del Sarcoma Aviar/patogenicidad , Pollos/inmunología , Mutación , Receptores Virales/genética , Secuencia de Aminoácidos , Animales , Leucosis Aviar/inmunología , Leucosis Aviar/virología , Virus de la Leucosis Aviar/metabolismo , Proteínas Aviares , Virus del Sarcoma Aviar/metabolismo , Secuencia de Bases , Células Cultivadas , Embrión de Pollo , Pollos/virología , Datos de Secuencia Molecular , Codorniz , Receptores Virales/química , Receptores Virales/metabolismo , Sarcoma Aviar/inmunología , Sarcoma Aviar/virología , Análisis de Secuencia de ADN
7.
J Virol ; 78(24): 13534-42, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15564464

RESUMEN

The Rous sarcoma virus Gag protein undergoes transient nuclear trafficking during virus assembly. Nuclear import is mediated by a nuclear targeting sequence within the MA domain. To gain insight into the role of nuclear transport in replication, we investigated whether addition of a "classical " nuclear localization signal (NLS) in Gag would affect virus assembly or infectivity. A bipartite NLS derived from nucleoplasmin was inserted into a region of the MA domain of Gag that is dispensable for budding and infectivity. Gag proteins bearing the nucleoplasmin NLS insertion displayed an assembly defect. Mutant virus particles (RC.V8.NLS) were not infectious, although they were indistinguishable from wild-type virions in Gag, Gag-Pol, Env, and genomic RNA incorporation and Gag protein processing. Unexpectedly, postinfection viral DNA synthesis was also normal, as similar amounts of two-long-terminal-repeat junction molecules were detected for RC.V8.NLS and wild type, suggesting that the replication block occurred after nuclear entry of proviral DNA. Phenotypically revertant viruses arose after continued passage in culture, and sequence analysis revealed that the nucleoplasmin NLS coding sequence was deleted from the gag gene. To determine whether the nuclear targeting activity of the nucleoplasmin sequence was responsible for the infectivity defect, two critical basic amino acids in the NLS were altered. This virus (RC.V8.KR/AA) had restored infectivity, and the MA.KR/AA protein showed reduced nuclear localization, comparable to the wild-type MA protein. These data demonstrate that addition of a second NLS, which might direct MA and/or Gag into the nucleus by an alternate import pathway, is not compatible with productive virus infection.


Asunto(s)
Virus del Sarcoma Aviar/fisiología , Núcleo Celular/metabolismo , Productos del Gen gag/química , Señales de Localización Nuclear/genética , Proteínas de la Matriz Viral/química , Replicación Viral , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Virus del Sarcoma Aviar/genética , Virus del Sarcoma Aviar/patogenicidad , Células Cultivadas , Regulación Viral de la Expresión Génica , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Datos de Secuencia Molecular , Mutación , Señales de Localización Nuclear/química , Codorniz , Proteínas Recombinantes de Fusión/metabolismo , Fracciones Subcelulares/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus
8.
Poult Sci ; 83(9): 1479-88, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15384897

RESUMEN

White Leghorn chicks homozygous for B19 MHC haplotype were selected for 18 generations on tumor regression after inoculation in the wing web with an SR-D strain of Rous sarcoma virus (RSV) at 4 wk of age. Each chick was assigned a tumor profile index (TPI) based on age at death and size of the tumor. During 18 generations, 2,010 birds were divergently selected on TPI for either progression or regression of the tumor (P and R lines). A Brody growth curve was fitted for each bird. Brody function parameters included the asymptotic tumor volume (A), the factor for increased growth in progression phase (K1), the factor for decreased growth in regression phase (K2), age at maximum volume (Tmax), and maximum volume of the tumor (Vmax). Tumor growth curves were found to be different according to line, sex, and restriction fragment pattern Y complex Rfp-Y MHC haplotype (Yw*15, Yw*16, and Yw*17). Within the P line, birds from the Yw*16 haplotype reached Vmax at an earlier age than Yw*15 and Yw*17, but with a lower Vmax value. Within the R line, tumor growth curves of birds from Yw*16 and Yw*17 haplotypes were similar. Rank correlations between the different parameters and TPI were low (between -0.26 and 0.36). Heritability estimated by the sire component was high for Vmax (0.73). Heritabilities of Tmax and K2 were moderate (0.20 to 0.23 for Tmax and 0.18 to 0.21 for K2) allowing these traits to be used as selection criteria. Heritabilities of A and K1 were lower than 0.12. Modeling the growth curve should contribute to better distinction between progressors and regressors.


Asunto(s)
Virus del Sarcoma Aviar/patogenicidad , Pollos/crecimiento & desarrollo , Pollos/genética , Enfermedades de las Aves de Corral/patología , Sarcoma Aviar/patología , Animales , Progresión de la Enfermedad , Femenino , Haplotipos/genética , Complejo Mayor de Histocompatibilidad/genética , Masculino , Enfermedades de las Aves de Corral/virología , Regresión Psicológica , Sarcoma Aviar/virología , Factores Sexuales , Carga Tumoral/genética
9.
Cancer Immunol Immunother ; 53(9): 799-808, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15197494

RESUMEN

The formation of a provisional extracellular matrix represents an important step during tumor growth and angiogenesis. Proteins that participate in this process become activated and undergo conformational changes that expose biologically active cryptic sites. Activated matrix proteins express epitopes not found on their native counterparts. We hypothesized that these epitopes may have a restricted tissue distribution, rendering them suitable targets for therapeutic human monoclonal antibodies (huMabs). In this study, we exploited phage antibody display technology and subtractive phage selection to generate human monoclonal antibody fragments that discriminate between the activated and native conformation of the extracellular matrix protein vitronectin. One of the selected antibody fragments, scFv VN18, was used to construct a fully human IgG/kappa monoclonal antibody with an affinity of 9.3 nM. In immunohistochemical analysis, scFv and huMab VN18 recognized activated vitronectin in tumor tissues, whereas hardly any activated vitronectin was detectable in normal tissues. Iodine 123-radiolabeled huMabVN18 was shown to target to Rous sarcoma virus-induced tumors in chickens, an animal model in which the epitope for huMab VN18 is exposed during tumor development. Our results establish activated vitronectin as a potential target for tumor therapy in humans.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Virus del Sarcoma Aviar/patogenicidad , Enfermedades de las Aves de Corral/terapia , Sarcoma Aviar/terapia , Vitronectina/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Sitios de Unión de Anticuerpos , Pollos , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Humanos , Fragmentos de Inmunoglobulinas/inmunología , Región Variable de Inmunoglobulina , Radioisótopos de Yodo , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Fragmentos de Péptidos/inmunología , Biblioteca de Péptidos , Enfermedades de las Aves de Corral/diagnóstico por imagen , Enfermedades de las Aves de Corral/inmunología , Conformación Proteica , Radioinmunodetección , Sarcoma Aviar/diagnóstico por imagen , Sarcoma Aviar/inmunología
10.
Virology ; 318(1): 371-80, 2004 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-14972562

RESUMEN

The chicken TVB(S1) protein serves as the cellular receptor for the cytopathic subgroups B and D avian sarcoma and leukosis viruses (ASLVs) as well as for the non-cytopathic subgroup E ASLV. Previous studies had mapped the subgroup B viral interaction determinants to a region that was located between residues 32 and 46 of TVB(S1) [J. Virol. 76 (2002) 5404]. To gain a greater insight into ASLV Env-receptor interactions and the possible role of these interactions in viral cytopathic effects, we employed a homolog-scanning mutagenesis approach to identify amino acid residues important for subgroup E viral receptor function by exchanging amino acid residues between TVB(S1) and its human homolog, DR5. These studies identified residues Tyr-67, Asn-72, and Asp-73 of TVB(S1) as important subgroup E viral interaction determinants. Intriguingly, these three residues are conserved between TVB(S1) and DR5, demonstrating that the human protein contains critical subgroup E viral interaction determinants, but in this context, they cannot support viral entry. These data confirm that the molecular determinants of the TVB receptor required for subgroup E viral entry are completely distinct from those used by subgroup B viruses.


Asunto(s)
Aminoácidos/química , Virus de la Leucosis Aviar/metabolismo , Virus de la Leucosis Aviar/patogenicidad , Virus del Sarcoma Aviar/metabolismo , Virus del Sarcoma Aviar/patogenicidad , Receptores del Factor de Necrosis Tumoral/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Efecto Citopatogénico Viral , Productos del Gen env/metabolismo , Humanos , Datos de Secuencia Molecular , Mutagénesis , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/genética
11.
J Virol ; 78(2): 683-91, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14694099

RESUMEN

Rong et al. have demonstrated previously that with a few substitutions, the fourth repeat of human low-density lipoprotein (hLDL-A4) receptor can functionally replace the LDL-A module of Tva, the cellular receptor for subgroup A avian sarcoma and leukosis virus (ASLV-A), in viral entry (L. Rong, K. Gendron, and P. Bates, Proc. Natl. Acad. Sci. USA 95:8467-8472, 1998). Here we have shown that swapping the amino terminus of hLDL repeat 5 (hLDL-A5) with that of Tva, in addition to the corresponding substitutions made in human LDL-A4, was required to convert hLDL-A5 into an efficient ASLV-A receptor. These results substantiated our previous findings regarding the role of the specific residues in the viral interaction domain of Tva and demonstrated the critical role of the amino terminus of the Tva LDL-A module in ASLV-A infection. Furthermore, we have shown that the residues between cysteines 2 and 3 of the Tva LDL-A module in a Tva/LDL-A5 chimeric protein can be functionally replaced by the corresponding region of another LDL-A module, human LDL receptor-related protein repeat 22 (LDL-A22), to mediate efficient ASLV-A entry. Since the only conserved feature between the C2-C3 region of LDL-A22 and the Tva LDL-A module is that both contain nine amino acids of which none are conserved, we conclude that the spacing between C2 and C3 of the LDL-A module of Tva is an important determinant for ASLV-A entry. Thus, the present study provides strong evidence to support our hypothesis that one role of the N terminus of the LDL-A module of Tva is to allow proper folding and conformation of the protein for optimal interaction with the viral glycoprotein EnvA in ASLV-A entry.


Asunto(s)
Virus de la Leucosis Aviar/patogenicidad , Virus del Sarcoma Aviar/patogenicidad , Receptores de LDL/química , Receptores de LDL/metabolismo , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Proteínas Aviares , Línea Celular , Cisteína/química , Humanos , Lipoproteínas LDL , Datos de Secuencia Molecular , Codorniz , Receptores de LDL/genética , Receptores Virales/química , Receptores Virales/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
12.
J Virol ; 77(12): 6709-19, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12767991

RESUMEN

Alpharetroviruses provide a useful system for the study of the molecular mechanisms of host range and receptor interaction. These viruses can be divided into subgroups based on diverse receptor usage due to variability within the two host range determining regions, hr1 and hr2, in their envelope glycoprotein SU (gp85). In previous work, our laboratory described selection from a subgroup B avian sarcoma-leukosis virus of an extended-host-range variant (LT/SI) with two adjacent amino acid substitutions in hr1. This virus retains its ability to use the subgroup BD receptor but can also infect QT6/BD cells, which bear a related subgroup E receptor (R. A. Taplitz and J. M. Coffin, J. Virol 71:7814-7819, 1997). Here, we report further analysis of this unusual variant. First, one (L154S) of the two substitutions is sufficient for host range extension, while the other (T155I) does not alter host range. Second, these mutations extend host range to non-avian cell types, including human, dog, cat, mouse, rat, and hamster. Third, interference experiments imply that the mutants interact efficiently with the subgroup BD receptor and possibly the related subgroup E receptor, but they have another means of entry that is not dependent on these interactions. Fourth, binding studies indicate that the mutant SU proteins retain the ability to interact as monomers with subgroup BD and BDE receptors but only bind the subgroup E receptor in the context of an Env trimer. Further, the mutant SU proteins bind well to chicken cells but do not bind any better than wild-type subgroup B to QT6 or human cells, even though the corresponding viruses are capable of infecting these cells.


Asunto(s)
Virus de la Leucosis Aviar/patogenicidad , Virus del Sarcoma Aviar/patogenicidad , Inmunoglobulina G/metabolismo , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Animales , Virus de la Leucosis Aviar/genética , Virus del Sarcoma Aviar/genética , Gatos , Línea Celular , Células Cultivadas , Embrión de Pollo , Pollos , Cricetinae , Perros , Humanos , Inmunoglobulina G/genética , Ratones , Datos de Secuencia Molecular , Mutación , Ratas , Recombinación Genética , Especificidad de la Especie , Transfección , Proteínas del Envoltorio Viral/genética
13.
FEBS Lett ; 543(1-3): 81-6, 2003 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-12753910

RESUMEN

Our data show that in hamster fibroblasts transformed by Rous sarcoma virus (RSV), the phosphoinositide 3'-kinase (PI-3K)/Akt/glycogen synthase kinase 3 antiapoptotic pathway is upregulated and involved in increased protein synthesis through activation of initiation factor eIF2B. Upon inhibition of PI-3K by wortmannin, phosphorylation of 70-kDa ribosomal protein S6 kinase (p70 S6k) and its physiological substrate, ribosomal protein S6, decreased in the non-transformed cells but not in RSV-transformed cells. Thus PI-3K, which is thought to be involved in regulation of p70 S6k, signals to p70 S6k in normal fibroblasts, but it does not appear to be an upstream effector of p70 S6k in fibroblasts transformed by v-src oncogene, suggesting that changes in the PI-3K signalling pathway upstream of p70 S6k are induced by RSV transformation.


Asunto(s)
Factor 2B Eucariótico de Iniciación/fisiología , Proteína Oncogénica pp60(v-src)/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Quinasas S6 Ribosómicas 70-kDa/fisiología , Transducción de Señal , Androstadienos/farmacología , Animales , Virus del Sarcoma Aviar/genética , Virus del Sarcoma Aviar/patogenicidad , Línea Celular , Línea Celular Transformada , Transformación Celular Viral , Cricetinae , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Wortmanina
14.
Proc Natl Acad Sci U S A ; 100(8): 4778-83, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12679521

RESUMEN

Caffeine is an efficient inhibitor of cellular DNA repair, likely through its effects on ATM (ataxia telangiectasia mutated) and ATR (ATM and Rad3-related) kinases. Here, we show that caffeine treatment causes a dose-dependent reduction in the total amount of HIV-1 and avian sarcoma virus retroviral vector DNA that is joined to host DNA in the population of infected cells and also in the number of transduced cells. These changes were observed at caffeine concentrations that had little or no effect on overall cell growth, synthesis, and nuclear import of the viral DNA, or the activities of the viral integrase in vitro. Substantial reductions in the amount of host-viral-joined DNA in the infected population, and in the number of transductants, were also observed in the presence of a dominant-negative form of the ATR protein, ATRkd. After infection, a significant fraction of these cells undergoes cell death. In contrast, retroviral transduction is not impeded in ATM-deficient cells, and addition of caffeine leads to the same reduction that was observed in ATM-proficient cells. These results suggest that activity of the ATR kinase, but not the ATM kinase, is required for successful completion of the viral DNA integration process and/or survival of transduced cells. Components of the cellular DNA damage repair response may represent potential targets for antiretroviral drug development.


Asunto(s)
Proteínas de Ciclo Celular/genética , Daño del ADN , Retroviridae/genética , Retroviridae/patogenicidad , Integración Viral/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Virus del Sarcoma Aviar/efectos de los fármacos , Virus del Sarcoma Aviar/genética , Virus del Sarcoma Aviar/patogenicidad , Virus del Sarcoma Aviar/fisiología , Secuencia de Bases , Cafeína/farmacología , Proteínas de Ciclo Celular/fisiología , Línea Celular Transformada , Reparación del ADN , ADN Viral/genética , ADN Viral/metabolismo , Proteínas de Unión al ADN , Vectores Genéticos , Integrasa de VIH/metabolismo , VIH-1/efectos de los fármacos , VIH-1/genética , VIH-1/patogenicidad , VIH-1/fisiología , Células HeLa , Humanos , Operón Lac , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Retroviridae/fisiología , Transducción Genética , Proteínas Supresoras de Tumor , Integración Viral/efectos de la radiación
15.
Curr Biol ; 12(15): 1301-11, 2002 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-12176358

RESUMEN

BACKGROUND: RNA interference (RNAi) is a newly discovered cellular defense system that is known to suppress replication of genomic parasites in model organisms. It has been widely conjectured that RNAi may also serve as an antiviral system in vertebrates. RESULTS: Retroviral infection could be initiated by electroporation of cloned Rous sarcoma virus (RSV) proviral DNA into the developing chick neural tube. Coelectroporation of proviral DNA and short double-stranded RNAs matching sequences of avain retroviruses, which were designed to induce RNAi against RSV, inhibited viral replication. Replication of RSV after electroporation resulted in disruption of embryonic development and early death, but this, too, could be suppressed by RNAi against the RSV genome. RNAi could also inhibit the growth of RSV and HIV in cell culture. Analysis of the step of the retroviral life cycle that is inhibited by RNAi revealed that it primarily prevented accumulation of the viral RNAs synthesized late during infection. RNA genomes introduced in viral particles early during infection were less sensitive. CONCLUSIONS: RNAi can block retroviral infection in vertebrates. The tissue electroporation method described here should allow RNAi to be used widely to study gene function and control of infection in vertebrate animals.


Asunto(s)
Interferencia de ARN , Retroviridae/genética , Retroviridae/patogenicidad , Animales , Virus del Sarcoma Aviar/genética , Virus del Sarcoma Aviar/patogenicidad , Secuencia de Bases , Embrión de Pollo , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/metabolismo , Sistema Nervioso/embriología , Oligonucleótidos/química , ARN Mensajero/genética , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , ARN Viral/genética , Proteínas Recombinantes de Fusión/metabolismo , Retroviridae/fisiología , Virulencia/genética , Replicación Viral/genética
16.
J Virol ; 76(6): 2848-56, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11861852

RESUMEN

Tva is the cellular receptor for subgroup A avian sarcoma and leukosis virus (ASLV-A). The viral receptor function of Tva is determined by a 40-residue, cysteine-rich motif called the LDL-A module. Here we report the solution structure of the LDL-A module of Tva, determined by nuclear magnetic resonance (NMR) spectroscopy. Although the carboxyl terminus of the Tva LDL-A module has a structure similar to those of other reported LDL-A modules, the amino terminus adopts a different conformation. The LDL-A module of Tva does not contain the signature antiparallel beta-sheet observed in other LDL-A modules, and it is more flexible than other reported LDL-A modules. The LDL-A structure of Tva provides mechanistic insights into how a simple viral receptor functions in retrovirus entry. The side chains of H38 and W48 of Tva, which have been identified as viral contact residues by mutational analysis, are solvent exposed, suggesting that they are directly involved in EnvA binding. However, the side chain of L34, another potential viral contact residue identified previously, is buried inside of the module and forms the hydrophobic core with other residues. Thus L34 likely stabilizes the Tva structure but is not a viral interaction determinant. In addition, we propose that the flexible amino-terminal region of Tva plays an important role in determining specificity in the Tva-EnvA interaction.


Asunto(s)
Virus de la Leucosis Aviar/patogenicidad , Virus del Sarcoma Aviar/patogenicidad , Receptores de LDL/química , Receptores Virales/química , Secuencia de Aminoácidos , Animales , Proteínas Aviares , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Receptores de LDL/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Análisis de Secuencia de ADN
17.
J Virol ; 75(23): 11544-54, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11689636

RESUMEN

The Rous sarcoma virus (RSV) transmembrane (TM) glycoprotein is modified by the addition of palmitic acid. To identify whether conserved cysteines within the hydrophobic anchor region are the site(s) of palmitoylation, and to determine the role of acylation in glycoprotein function, cysteines at residues 164 and 167 of the TM protein were mutated to glycine (C164G, C167G, and C164G/C167G). In CV-1 cells, palmitate was added to env gene products containing single mutations but was absent in the double-mutant Env. Although mutant Pr95 Env precursors were synthesized with wild-type kinetics, the phenotypes of the mutants differed markedly. Env-C164G had properties similar to those of the wild type, while Env-C167G was degraded faster, and Env containing the double mutant C164G/C167G was very rapidly degraded. Degradation occurred after transient plasma membrane expression. The decrease in steady-state surface expression and increased rate of internalization into endosomes and lysosomes paralleled the decrease in palmitoylation observed for the mutants. The phenotypes of mutant viruses were assessed in avian cells in the context of the pATV8R proviral genome. Virus containing the C164G mutation replicated with wild-type kinetics but exhibited reduced peak reverse transcriptase levels. In contrast, viruses containing either the C167G or the C164G/C167G mutation were poorly infectious or noninfectious, respectively. These phenotypes correlated with different degrees of glycoprotein incorporation into virions. Infectious revertants of the double mutant demonstrated the importance of cysteine-167 for efficient plasma membrane expression and Env incorporation. The observation that both cysteines within the membrane-spanning domain are accessible for acylation has implications for the topology of this region, and a model is proposed.


Asunto(s)
Virus del Sarcoma Aviar/patogenicidad , Ácido Palmítico/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Animales , Virus del Sarcoma Aviar/metabolismo , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Productos del Gen env/genética , Productos del Gen env/metabolismo , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Pavos , Proteínas del Envoltorio Viral/química , Virulencia
18.
Immunol Lett ; 75(3): 209-14, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11166377

RESUMEN

As shown earlier, the cells transformed in vitro by various oncogenes, during subsequent in vivo growth were gradually replaced by descendant tumor cells, which co-expressed highly increased H(2)O(2)-catabolizing antioxidant activity (H(2)O(2)(CA)), and the ability to release PGE(2) (PGE(S)) in contact with natural killer cells; v-src was the only oncogene, which in vitro induced cells transformation characterised with the expression of [H(2)O(2)(CA)+PGE(S)] phenotype. Expression of [H(2)O(2)(CA)+PGE(S)] phenotype was providing tumor cells with significantly increased resistance to cytotoxic activities of macrophages and NK cells. However, the possible involvement of [H(2)O(2)(CA)+PGE(S)] phenotype in primary carcinogenesis remained obscure. Here, using three models of the primary virus-induced Syrian hamster tumors we demonstrated that Rous Sarcoma Virus-induced tumors arising after short latent period expressed [H(2)O(2)(CA) + PGE(S)] phenotype at appearance. During the latent periods of SV40- and SA7(C8)-induced tumors the cells expressing [H(2)O(2)(CA)+PGE(S)] phenotype gradually replaced the original [H(2)O(2)(CA)+PGE(S)]-phenotype-negative cell populations. The effectiveness of such selection correlated with the duration of in vivo tumor development. Thus it was shown, that selection of tumor cells expressing [H(2)O(2)(CA)+PGE(S)] phenotype is beginning and may be completed during the latent period of primary carcinogenesis. Taken together, data of this and preceding our studies on [H(2)O(2)(CA)+PGE(S)] phenotype demonstrate that in vivo the host innate immunity antitumor reactions are apparently responsible for the selection of rare tumor cell variants capable to defend themselves against CTA of Mph and NK.


Asunto(s)
Transformación Celular Viral , Inmunidad Innata , Neoplasias/inmunología , Neoplasias/virología , Adenovirus de los Simios/patogenicidad , Adenovirus de los Simios/fisiología , Animales , Virus del Sarcoma Aviar/patogenicidad , Virus del Sarcoma Aviar/fisiología , Cricetinae , Peróxido de Hidrógeno/metabolismo , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Mesocricetus , Neoplasias/patología , Neoplasias Primarias Desconocidas , Fenotipo , Prostaglandinas E/metabolismo , Virus 40 de los Simios/patogenicidad , Virus 40 de los Simios/fisiología , Células Tumorales Cultivadas
19.
J Virol ; 74(8): 3731-9, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10729148

RESUMEN

Short hydrophobic regions referred to as fusion peptide domains (FPDs) at or near the amino terminus of the membrane-anchoring subunit of viral glycoproteins are believed to insert into the host membrane during the initial stage of enveloped viral entry. Avian sarcoma and leukosis viruses (ASLV) are unusual among retroviruses in that the region in the envelope glycoprotein (EnvA) proposed to be the FPD is internal and contains a centrally located proline residue. To begin analyzing the function of this region of EnvA, 20 substitution mutations were introduced into the putative FPD. The mutant envelope glycoproteins were evaluated for effects on virion incorporation, receptor binding, and infection. Interestingly, most of the single-substitution mutations had little effect on any of these processes. In contrast, a bulky hydrophobic substitution for the central proline reduced viral titers 15-fold without affecting virion incorporation or receptor binding, whereas substitution of glycine for the proline had only a nominal effect on EnvA function. Similar to other viral FPDs, the putative ASLV FPD has been modeled as an amphipathic helix where most of the bulky hydrophobic residues form a patch on one face of the helix. A series of alanine insertion mutations designed to interrupt the hydrophobic patch on the helix had differential effects on infectivity, and the results of that analysis together with the results observed with the substitution mutations suggest no correlation between maintenance of the hydrophobic patch and glycoprotein function.


Asunto(s)
Virus de la Leucosis Aviar , Virus del Sarcoma Aviar , Mutación , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Virus de la Leucosis Aviar/química , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/patogenicidad , Virus del Sarcoma Aviar/química , Virus del Sarcoma Aviar/genética , Virus del Sarcoma Aviar/patogenicidad , Virus del Sarcoma Aviar/fisiología , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Epítopos , Datos de Secuencia Molecular , Mutagénesis Insercional , Mutación Puntual , Receptores Virales/metabolismo , Eliminación de Secuencia , Proteínas del Envoltorio Viral/metabolismo , Virión/metabolismo
20.
Poult Sci ; 79(3): 343-8, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10735200

RESUMEN

Rous sarcoma virus-induced tumor outcome is controlled by the MHC (B). Additional data, using controlled segregation in families, has indicated non-MHC effects as well, but few studies have focused on blood groups other than the B complex. Segregating combinations of genes encoding erythrocyte (Ea) alloantigen systems A, C, D, E, H, I, P, and L in B2B5 and B5B5 MHC (B) backgrounds were examined for their effects on Rous sarcomas. Six-week-old chickens were inoculated in the wing-web with 30 pfu of Rous sarcoma virus (RSV). Tumors were scored six times over a 10-wk period. A tumor profile index (TPI) was assigned to each chicken based on the six tumor size scores. Response was evaluated using tumor size at each measurement period, TPI, and mortality. The genotypes of Ea systems A, C, D, E, H, I, and P had no significant effect on any parameter in either B complex population. The Ea-L system had an effect on Rous sarcomas in the B2B5 intermediate responders and B5B5 progressors. Tumor size, TPI, and mortality were all significantly lower in B2B5 L1L1 chickens than in B2B5 L1L2 chickens. Mortality was lower in the B5B5 L1L1 birds than in B5B5 L1L2 chickens. It appears that the Ea-L system, or one closely linked, is acting in a manner independent of the B complex in response to RSV challenge.


Asunto(s)
Virus del Sarcoma Aviar/inmunología , Pollos/virología , Isoantígenos/inmunología , Sarcoma Aviar/inmunología , Animales , Virus del Sarcoma Aviar/patogenicidad , Pollos/inmunología , Femenino , Masculino , Mortalidad , Sarcoma Aviar/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA