Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 86(4): 2021-30, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22171251

RESUMEN

The group of closely related avian sarcoma and leukosis viruses (ASLVs) evolved from a common ancestor into multiple subgroups, A to J, with differential host range among galliform species and chicken lines. These subgroups differ in variable parts of their envelope glycoproteins, the major determinants of virus interaction with specific receptor molecules. Three genetic loci, tva, tvb, and tvc, code for single membrane-spanning receptors from diverse protein families that confer susceptibility to the ASLV subgroups. The host range expansion of the ancestral virus might have been driven by gradual evolution of resistance in host cells, and the resistance alleles in all three receptor loci have been identified. Here, we characterized two alleles of the tva receptor gene with similar intronic deletions comprising the deduced branch-point signal within the first intron and leading to inefficient splicing of tva mRNA. As a result, we observed decreased susceptibility to subgroup A ASLV in vitro and in vivo. These alleles were independently found in a close-bred line of domestic chicken and Indian red jungle fowl (Gallus gallus murghi), suggesting that their prevalence might be much wider in outbred chicken breeds. We identified defective splicing to be a mechanism of resistance to ASLV and conclude that such a type of mutation could play an important role in virus-host coevolution.


Asunto(s)
Alpharetrovirus/fisiología , Proteínas Aviares/genética , Pollos/genética , Predisposición Genética a la Enfermedad , Enfermedades de las Aves de Corral/genética , Empalme del ARN , Receptores Virales/genética , Sarcoma Aviar/genética , Eliminación de Secuencia , Alpharetrovirus/genética , Secuencia de Aminoácidos , Animales , Proteínas Aviares/metabolismo , Secuencia de Bases , Pollos/metabolismo , Pollos/virología , Intrones , Datos de Secuencia Molecular , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/virología , Receptores Virales/metabolismo , Sarcoma Aviar/metabolismo , Sarcoma Aviar/virología
2.
Retrovirology ; 8: 96, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-22099981

RESUMEN

BACKGROUND: Understanding the mechanism by which viruses enter their target cell is an essential part of understanding their infectious cycle. Previous studies have focussed on the multiplicity of viral envelope proteins that need to bind to their cognate receptor to initiate entry. Avian sarcoma and leukosis virus Envelope protein (ASLV Env) mediates entry via a receptor, Tva, which can be attached to the cell surface either by a phospholipid anchor (Tva800) or a transmembrane domain (Tva950). In these studies, we have now investigated the number of target receptors necessary for entry of ASLV Env-pseudotyped virions. RESULTS: Using titration and modelling experiments we provide evidence that binding of more than one receptor, probably two, is needed for entry of virions via Tva800. However, binding of just one Tva950 receptor is sufficient for successful entry. CONCLUSIONS: The different modes of attachment of Tva800 and Tva950 to the cell membrane have important implications for the utilisation of these proteins as receptors for viral binding and/or uptake.


Asunto(s)
Alpharetrovirus/fisiología , Proteínas Aviares/metabolismo , Receptores Virales/metabolismo , Acoplamiento Viral , Internalización del Virus , Animales , Línea Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Ratones , Modelos Biológicos , Proteínas del Envoltorio Viral/metabolismo
3.
J Virol ; 84(13): 6626-35, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20410274

RESUMEN

Accidental insertional activation of proto-oncogenes and potential vector mobilization pose serious challenges for human gene therapy using retroviral vectors. Comparative analyses of integration sites of different retroviral vectors have elucidated distinct target site preferences, highlighting vectors based on the alpharetrovirus Rous sarcoma virus (RSV) as those with the most neutral integration spectrum. To date, alpharetroviral vector systems are based mainly on single constructs containing viral coding sequences and intact long terminal repeats (LTR). Even though they are considered to be replication incompetent in mammalian cells, the transfer of intact viral genomes is unacceptable for clinical applications, due to the risk of vector mobilization and the potentially immunogenic expression of viral proteins, which we minimized by setting up a split-packaging system expressing the necessary viral proteins in trans. Moreover, intact LTRs containing transcriptional elements are capable of activating cellular genes. By removing most of these transcriptional elements, we were able to generate a self-inactivating (SIN) alpharetroviral vector, whose LTR transcriptional activity is strongly reduced and whose transgene expression can be driven by an internal promoter of choice. Codon optimization of the alpharetroviral Gag/Pol expression construct and further optimization steps allowed the production of high-titer self-inactivating vector particles in human cells. We demonstrate proof of principle for the versatility of alpharetroviral SIN vectors for the genetic modification of murine and human hematopoietic cells at a low multiplicity of infection.


Asunto(s)
Alpharetrovirus/fisiología , Terapia Genética/métodos , Vectores Genéticos , Secuencias Repetidas Terminales/genética , Transgenes , Ensamble de Virus , Alpharetrovirus/genética , Animales , Línea Celular , Células Cultivadas , Expresión Génica , Humanos , Ratones , Regiones Promotoras Genéticas , ARN Viral/genética
4.
Curr Opin Cell Biol ; 6(6): 816-24, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7880528

RESUMEN

Analysis of the oncogenes v-erbB and v-erbA and their normal proto-oncogene counterparts has revealed several novel aspects of erythroid differentiation. A new erythroid progenitor capable of extended self-renewal has been described, tyrosine kinase receptors and steroid hormone receptors have been found to cooperate in controlling self-renewal, and dramatic alterations in the cell cycle have been found to accompany induction of terminal differentiation.


Asunto(s)
Alpharetrovirus/fisiología , Eritropoyesis/fisiología , Alpharetrovirus/genética , Animales , Ciclo Celular , Transformación Celular Viral , Pollos , Genes erbA , Genes erbB , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Estrógenos/fisiología , Transcripción Genética
5.
Viruses ; 13(1)2021 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-33477490

RESUMEN

The assembly of a hexameric lattice of retroviral immature particles requires the involvement of cell factors such as proteins and small molecules. A small, negatively charged polyanionic molecule, myo-inositol hexaphosphate (IP6), was identified to stimulate the assembly of immature particles of HIV-1 and other lentiviruses. Interestingly, cryo-electron tomography analysis of the immature particles of two lentiviruses, HIV-1 and equine infectious anemia virus (EIAV), revealed that the IP6 binding site is similar. Based on this amino acid conservation of the IP6 interacting site, it is presumed that the assembly of immature particles of all lentiviruses is stimulated by IP6. Although this specific region for IP6 binding may be unique for lentiviruses, it is plausible that other retroviral species also recruit some small polyanion to facilitate the assembly of their immature particles. To study whether the assembly of retroviruses other than lentiviruses can be stimulated by polyanionic molecules, we measured the effect of various polyanions on the assembly of immature virus-like particles of Rous sarcoma virus (RSV), a member of alpharetroviruses, Mason-Pfizer monkey virus (M-PMV) representative of betaretroviruses, and murine leukemia virus (MLV), a member of gammaretroviruses. RSV, M-PMV and MLV immature virus-like particles were assembled in vitro from truncated Gag molecules and the effect of selected polyanions, myo-inostol hexaphosphate, myo-inositol, glucose-1,6-bisphosphate, myo-inositol hexasulphate, and mellitic acid, on the particles assembly was quantified. Our results suggest that the assembly of immature particles of RSV and MLV was indeed stimulated by the presence of myo-inostol hexaphosphate and myo-inositol, respectively. In contrast, no effect on the assembly of M-PMV as a betaretrovirus member was observed.


Asunto(s)
Membrana Celular/química , Membrana Celular/metabolismo , Interacciones Huésped-Patógeno , Polielectrolitos/química , Retroviridae/fisiología , Ensamble de Virus , Alpharetrovirus/fisiología , Animales , Betaretrovirus/fisiología , Células Cultivadas , Gammaretrovirus/fisiología , Productos del Gen gag/química , Productos del Gen gag/metabolismo , Polielectrolitos/metabolismo , Retroviridae/ultraestructura , Virión
6.
J Virol ; 82(22): 11419-28, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18768966

RESUMEN

We recently identified and cloned the receptor for subgroup C avian sarcoma and leukosis viruses [ASLV(C)], i.e., Tvc, a protein most closely related to mammalian butyrophilins, which are members of the immunoglobulin protein family. The extracellular domain of Tvc contains two immunoglobulin-like domains, IgV and IgC, which presumably each contain a disulfide bond important for native function of the protein. In this study, we have begun to identify the functional determinants of Tvc responsible for ASLV(C) receptor activity. We found that the IgV domain of the Tvc receptor is responsible for interacting with the glycoprotein of ASLV(C). Additional experiments demonstrated that a domain was necessary as a spacer between the IgV domain and the membrane-spanning domain for efficient Tvc receptor activity, most likely to orient the IgV domain a proper distance from the cell membrane. The effects on ASLV(C) glycoprotein binding and infection efficiency were also studied by site-directed mutagenesis of the cysteine residues of Tvc as well as conserved amino acid residues of the IgV Tvc domain compared to other IgV domains. In this initial analysis of Tvc determinants important for interacting with ASLV(C) glycoproteins, at least two aromatic amino acid residues in the IgV domain of Tvc, Trp-48 and Tyr-105, were identified as critical for efficient ASLV(C) infection. Interestingly, one or more aromatic amino acid residues have been identified as critical determinants in the other ASLV(A-E) receptors for a proper interaction with ASLV glycoproteins. This suggests that the ASLV glycoproteins may share a common mechanism of receptor interaction with an aromatic residue(s) on the receptor critical for triggering conformational changes in SU that initiate the fusion process required for efficient virus infection.


Asunto(s)
Alpharetrovirus/fisiología , Membrana Celular/química , Mapeo de Interacción de Proteínas , Receptores Virales/metabolismo , Acoplamiento Viral , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Embrión de Pollo , Pollos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Terciaria de Proteína , Receptores Virales/genética , Alineación de Secuencia , Proteínas del Envoltorio Viral/metabolismo
7.
J Cell Biol ; 91(2 Pt 1): 573-8, 1981 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-6273447

RESUMEN

The transformed or normal phenotype of cultured normal rat kidney cells infected with a temperature-sensitive mutant of avian sarcoma virus is conditional on the temperature at which the cells are grown. Using dye injection techniques, we show that junction-mediated dye transfer is also temperature-sensitive. The extent and rate of transfer between infected cells grown at the transformation-permissive temperature (35 degrees C) is significantly reduced when compared to infected cells grown at the nonpermissive temperature (40.5 degrees C) or uninfected cells grown at either temperature. Infected cells subjected to reciprocal temperature shifts express rapid and reversible alterations of dye transfer capacities, with responses evident by 15 min and completed by 60 min for temperature shifts in either direction. These results suggest that altered junctional capacities may be fundamental to the expression of the ASV-induced, transformed phenotype.


Asunto(s)
Alpharetrovirus/fisiología , Comunicación Celular , Transformación Celular Neoplásica , Transformación Celular Viral , Uniones Intercelulares/fisiología , Animales , Línea Celular , Permeabilidad de la Membrana Celular , Cinética , Mutación , Ratas , Temperatura
8.
Virus Res ; 135(1): 72-82, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18420298

RESUMEN

During retroviral integration, the viral integrase recognizes the attachment (att) sequence (formed by juxtaposition of two LTRs ends) as the substrate of integration. We have developed a self-deleting Avian Leukosis and Sarcoma Viruses (ALSVs)-based retroviral vector carrying an additional copy of the att sequence, between neo and puro genes. We observed that: (i) the resulting NP3Catt vector was produced at neo and puro titers respectively smaller and higher than that of the parental vector devoid of the att sequence; (ii) 61% of NP3Catt proviruses were flanked by LTRs; most of them were deleted of internal sequences, probably during the reverse transcription step; (iii) 31% of clones were deleted of the whole 5' part of their genome and were flanked, in 5', by the additional att sequence and, in 3', by an LTR. Integration of these last proviruses was often imprecise with respect to the viral ends. At total, 77% of proviruses had lost the packaging signal and were not mobilizable by a replication-competent virus and 92% had lost the selectable gene in a single round of replication. Although still to improve, the att vector could be considered as an interesting new safe retroviral vector for gene transfer experiments.


Asunto(s)
Alpharetrovirus/enzimología , Alpharetrovirus/genética , Vectores Genéticos/genética , Integrasas/metabolismo , Eliminación de Secuencia , Integración Viral , Alpharetrovirus/fisiología , Animales , Secuencia de Bases , Línea Celular , Técnicas de Transferencia de Gen , Vectores Genéticos/química , Integrasas/genética , Provirus/enzimología , Provirus/genética , Provirus/fisiología , Codorniz , ARN Viral/química , ARN Viral/genética , Secuencias Repetidas Terminales , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
9.
Oncogene ; 2(4): 317-26, 1988 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2834683

RESUMEN

Cells transformed by the avian erythroblastosis virus S13 contain three proteins derived from the v-sea oncogene, gp155, gp70 (a cleavage product of gp155), and p38. It is not clear whether only one or all three of these proteins are required for transformation by S13. S13 transformed erythroblasts and fibroblasts revert to a normal morphology in the presence of the alpha glucosidase-1 inhibitor castanospermine, whereas cells transformed by the v-src or v-erbB oncogenes are unaffected by this drug. Treatment with castanospermine does not alter the tyrosine kinase autophosphorylation activity of any of the v-sea products, and the synthesis and processing of p38 is unaffected. Castanospermine modifies the structure of the carbohydrate chains of gp155 such that the glucose residues are retained, thereby inhibiting complex chain formation. Analysis of revertant S13 transformed cells shows that the proteolytic cleavage of the modified form of gp155 is inhibited, resulting in a very low yield of a modified form of gp70. There is no detectable effect of castanospermine on the transport of v-sea gene products to the cell surface. However, due to the inhibition of proteolytic cleavage, the modified form of gp155 is now the major v-sea encoded protein expressed on the cell surface. Thus it appears that the cell surface expression of a v-sea encoded protein with tyrosine kinase autophosphorylating activity is insufficient for cell transformation.


Asunto(s)
Alpharetrovirus/genética , Virus de la Leucosis Aviar/genética , Transformación Celular Viral , Indolizinas , Proteínas Oncogénicas Virales/metabolismo , Alcaloides/farmacología , Alpharetrovirus/fisiología , Animales , Transporte Biológico/efectos de los fármacos , Transformación Celular Viral/efectos de los fármacos , Embrión de Pollo , Eritroblastos/patología , Fibroblastos/patología , Glicosilación , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Swainsonina
10.
Oncogene ; 9(2): 537-44, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8290264

RESUMEN

The S13 avian erythroblastosis viral genome encodes an oncogenic tyrosine kinase, termed env-sea, that is capable of transforming fibroblasts and erythroblasts. Although the tyrosine kinase activity of the env-sea protein has been shown to be necessary for transformation, no substrates for this enzyme have been detected in vivo. Here we demonstrate that the recently described shc proteins are phosphorylated on tyrosine residues in both S13 transformed fibroblasts and erythroblasts. Furthermore, using an S13 temperature sensitive mutant, we show that the phosphorylation of the shc proteins occurs concomitantly with the activation of the tyrosine kinase activity of the env-sea protein. These observations make the phosphorylation of the shc proteins a good candidate for being involved in oncogenic signaling by the env-sea oncoprotein.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Eritroblastos/microbiología , Fibroblastos/microbiología , Proteínas Oncogénicas Virales/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas/metabolismo , Fosfolipasas de Tipo C/metabolismo , Tirosina/metabolismo , Alpharetrovirus/fisiología , Animales , Western Blotting , Diferenciación Celular/fisiología , Transformación Celular Viral/genética , Células Cultivadas , Embrión de Pollo , Proteína Adaptadora GRB2 , Proteínas Activadoras de GTPasa , Regulación de la Expresión Génica/genética , Mutación/genética , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Fosfatidilinositol 3-Quinasas , Fosforilación , Pruebas de Precipitina , Ratas , Transducción de Señal/fisiología , Temperatura
11.
Oncogene ; 19(55): 6472-81, 2000 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-11175363

RESUMEN

Studies of retroviral-induced oncogenesis in animal systems led to the initial discovery of viral oncogenes and their cellular homologs, and provided critical insights into their role in the neoplastic process. V-ets, the founding member of the ETS oncogene family, was originally identified as part of the fusion oncogene encoded by the avian acute leukemia virus E26 and subsequent analysis of virus induced leukemias led to the initial isolation of two other members of the ETS gene family. PU.1 was identified as a target of insertional activation in the majority of tumors induced by the murine Spleen Focus Forming virus (SFFV), while fli-1 proved to be the target of Friend murine leukemia virus (F-MuLV) in F-MuLV induced erythroleukemia, as well as that of the 10A1 and Graffi viruses. The common features of the erythroid and myeloid diseases induced by these viruses provided the initial demonstration that these and other members of the ETS family play important roles in hematopoietic development as well as disease. This review provides an overview of the role of ETS genes in retrovirally induced neoplasia, their possible mechanisms of action, and how these viral studies relate to current knowledge of the functions of these genes in hematopoiesis.


Asunto(s)
Transformación Celular Viral/genética , Regulación Neoplásica de la Expresión Génica , Regulación Viral de la Expresión Génica , Familia de Multigenes , Oncogenes , Proteínas Proto-Oncogénicas , Retroviridae/genética , Células 3T3 , Alpharetrovirus/genética , Alpharetrovirus/fisiología , Animales , Virus de la Mieloblastosis Aviar/genética , Virus de la Mieloblastosis Aviar/fisiología , Pollos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Fibroblastos/metabolismo , Hematopoyesis/genética , Humanos , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/fisiología , Ratones , Mutagénesis Insercional , Proteína Proto-Oncogénica c-fli-1 , Provirus/genética , Retroviridae/fisiología , Virus Formadores de Foco en el Bazo/genética , Virus Formadores de Foco en el Bazo/fisiología , Transactivadores/genética , Transactivadores/fisiología
12.
Anticancer Res ; 15(2): 449-54, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7763020

RESUMEN

The influence of cytostatically active alkyllysophospholipid analogs with different chemical structure on IP3 (inositol-1,4,5-trisphosphate) formation and intracellular Ca++ concentration was studied in human mammary epithelial cells before and after transfection with v-erb B oncogene DNA. Transformed cells showed an increased IP3 formation compared with normal cells. In the presence of ALP (alkyllysophospholipid) analogs IP3 formation is inhibited more strongly in transformed cells than in normal cells, dependent on the structure of ALP analogs. Furthermore, these compounds increased [Ca++]i (intracellular Ca++ concentration) in transformed cells much more strongly than in normal cells. From these results, obtained in pursuit of an oncogene-related cancer treatment, it follows that ALP analogs may inhibit processes in signal transduction in cells more strongly after transfection with a defined oncogene than before.


Asunto(s)
Mama/efectos de los fármacos , Inositol 1,4,5-Trifosfato/biosíntesis , Lisofosfolípidos/farmacología , Proteínas Oncogénicas v-erbB/fisiología , Éteres Fosfolípidos/farmacología , Alpharetrovirus/genética , Alpharetrovirus/fisiología , Mama/citología , Mama/metabolismo , Línea Celular Transformada , Transformación Celular Viral , Células Cultivadas , ADN Viral/genética , Relación Dosis-Respuesta a Droga , Humanos , Proteínas Oncogénicas v-erbB/genética , Transducción de Señal/efectos de los fármacos , Transfección
13.
Folia Biol (Praha) ; 41(1): 1-14, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7621964

RESUMEN

Although there are substantial differences between retroviruses originating from avian and primate species, a comparison of these two different biological systems reveals that interaction of these retroviruses with heterologous hosts involves similar biological principles. Retroviral isolates with high replicative capacity in natural targets (e.g. CD4+ lymphocytes and macrophages for human immunodeficiency viruses (HIVs) can infect other cell types [e.g. CD- astrocytes, follicular dendritic cells (FDC) in vivo and/or CD4+ neoplastic T cells in vitro] as well. These viral isolates may have a potential of infecting heterologous cells in vitro and can enlarge their host-range by establishing infection in other species, distantly related. Strains of avian sarcoma/leukemia viruses (ASLV) originating from their natural hosts, chickens, and infectious for other avian species, ducks, can frequently infect mammals (rodents). Similarly, HIV-1 strains infectious for chimpanzees possess capacity of establishing chronic infection in pig-tailed macaques. The broad host-range of retroviral isolates in both viral systems is accompanied by presence of additional structures in viral envelope. These novel or additional envelope structures may recognize alternate viral receptor(s). Moreover, the enlarged host range of primary HIV-1 isolates is evaluated by infection of neoplastic CD4+ permanent cell line, MT2, and serves as a predictive marker of progression of the viral infection toward AIDS.


Asunto(s)
Alpharetrovirus/fisiología , Pollos/virología , VIH/patogenicidad , Alpharetrovirus/inmunología , Animales , Linfocitos T CD4-Positivos/virología , Humanos , Ratones
14.
J Vet Med Sci ; 57(5): 8O1-11, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8593284

RESUMEN

Human and animal herpesviruses are able to stimulate gene expression of various human and animal retroviruses and the activation appears to be a common theme. It is of interest why the retroviruses can be commonly activated by herpesviruses despite clear diversities in each mechanism for retrovirus gene expression. In this review, three typical examples of the interactions; human immunodeficiency virus and human herpesviruses, avian retroviruses and Marek's disease virus, and feline immunodeficiency virus and feline herpesvirus type I will be given to respond the above question on the basis of the comparative study of molecular interactions.


Asunto(s)
Herpesviridae/genética , Herpesviridae/fisiología , Retroviridae/genética , Retroviridae/fisiología , Alpharetrovirus/genética , Alpharetrovirus/fisiología , Animales , Aves , Gatos , Regulación Viral de la Expresión Génica , VIH/genética , VIH/fisiología , Herpesvirus Gallináceo 2/genética , Herpesvirus Gallináceo 2/fisiología , Humanos , Virus de la Inmunodeficiencia Felina/genética , Virus de la Inmunodeficiencia Felina/fisiología
15.
Poult Sci ; 73(11): 1625-32, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7862601

RESUMEN

The effects of presence or absence of individual endogenous virus (ev) genes on production traits was studied in a highly productive commercial layer cross. Age and BW at first egg, egg production, egg weight, and mature BW were recorded for each bird. The birds were examined for presence of ev gene fragments by Southern analysis. A general linear model was used to determine significance of effects of the 21 individual ev fragments on the individual traits and the effects of all ev fragments taken together on each of the traits. Seven significant effects were found for individual ev fragments on individual traits. Four of these involved BW at first egg, and all ev gene fragments taken together had a significant effect on BW at first egg, explaining 17% of total phenotypic variation in this trait. Significant nonlinear correlations were found between total number of ev genes and both BW at first egg and mature BW, with birds having a moderate number of ev genes showing the lowest BW. For age at first egg and egg weight, nonlinear correlations, although not significant, were consistent in sign with those found for BW, implying minimum trait magnitude at moderate number of ev genes. These effects imply that animals with intermediate numbers of ev genes will tend to be favored by commercial selection in layer flocks, whereas birds with either too many or too few ev genes will tend to be culled.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Alpharetrovirus/fisiología , Peso Corporal/fisiología , Pollos/genética , Pollos/fisiología , Genes Virales/fisiología , Oviposición/fisiología , Alpharetrovirus/genética , Animales , Peso Corporal/genética , Pollos/virología , Huevos/normas , Femenino , Masculino , Oviposición/genética , Fenotipo , Polimorfismo Genético , Análisis de Regresión
16.
Viruses ; 6(12): 4811-38, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25490763

RESUMEN

Gene therapy using integrating retroviral vectors has proven its effectiveness in several clinical trials for the treatment of inherited diseases and cancer. However, vector-mediated adverse events related to insertional mutagenesis were also observed, emphasizing the need for safer therapeutic vectors. Paradoxically, alpharetroviruses, originally discovered as cancer-causing agents, have a more random and potentially safer integration pattern compared to gammaretro- and lentiviruses. In this review, we provide a short overview of the history of alpharetroviruses and explain how they can be converted into state-of-the-art gene delivery tools with improved safety features. We discuss development of alpharetroviral vectors in compliance with regulatory requirements for clinical translation, and provide an outlook on possible future gene therapy applications. Taken together, this review is a broad overview of alpharetroviral vectors spanning the bridge from their parental virus discovery to their potential applicability in clinical settings.


Asunto(s)
Alpharetrovirus/fisiología , Terapia Genética/instrumentación , Neoplasias/terapia , Alpharetrovirus/genética , Animales , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Neoplasias/genética , Integración Viral
17.
Virology ; 450-451: 2-12, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24503062

RESUMEN

Lymphoproliferative disease virus (LPDV) is an exogenous oncogenic retrovirus that induces lymphoid tumors in some galliform species of birds. Historically, outbreaks of LPDV have been reported from Europe and Israel. Although the virus has previously never been detected in North America, herein we describe the widespread distribution, genetic diversity, pathogenesis, and evolution of LPDV in the United States. Characterization of the provirus genome of the index LPDV case from North America demonstrated an 88% nucleotide identity to the Israeli prototype strain. Although phylogenetic analysis indicated that the majority of viruses fell into a single North American lineage, a small subset of viruses from South Carolina were most closely related to the Israeli prototype. These results suggest that LPDV was transferred between continents to initiate outbreaks of disease. However, the direction (New World to Old World or vice versa), mechanism, and time frame of the transcontinental spread currently remain unknown.


Asunto(s)
Alpharetrovirus/fisiología , Enfermedades Transmisibles Emergentes/veterinaria , Enfermedades Desatendidas/veterinaria , Enfermedades de las Aves de Corral/virología , Infecciones por Retroviridae/veterinaria , Infecciones Tumorales por Virus/veterinaria , Alpharetrovirus/clasificación , Alpharetrovirus/genética , Alpharetrovirus/aislamiento & purificación , Animales , Carcinogénesis , Enfermedades Transmisibles Emergentes/virología , Evolución Molecular , Datos de Secuencia Molecular , Enfermedades Desatendidas/virología , Filogenia , Enfermedades de las Aves de Corral/diagnóstico , Enfermedades de las Aves de Corral/epidemiología , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Pavos/virología , Estados Unidos/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA