Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
2.
J Virol ; 85(17): 9239-42, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21697469

RESUMEN

v-Abl protein tyrosine kinase encoded by Abelson murine leukemia virus (Ab-MLV) transforms pre-B cells. Transformation requires the phosphatidylinositol 3-kinase (PI3K) pathway. This pathway is antagonized by SH2-containing inositol 5'-phosphatase (SHIP), raising the possibility that v-Abl modulates PI3K signaling through SHIP. Consistent with this, we show that v-Abl expression reduces levels of full-length p145 SHIP in a v-Abl kinase activity-dependent fashion. This event requires signals from the Abl SH2 domain but not the carboxyl terminus. Forced expression of full-length SHIP significantly reduces Ab-MLV pre-B-cell transformation. Therefore, reduction of SHIP protein by v-Abl is a critical component in Ab-MLV transformation.


Asunto(s)
Virus de la Leucemia Murina de Abelson/patogenicidad , Transformación Celular Viral , Interacciones Huésped-Patógeno , Proteínas Oncogénicas v-abl/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Inositol Polifosfato 5-Fosfatasas , Ratones , Células Precursoras de Linfocitos B/virología , Mapeo de Interacción de Proteínas
3.
J Virol ; 82(17): 8383-91, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18579612

RESUMEN

Transformation by Abelson murine leukemia virus (Ab-MLV) is a multistep process in which growth-stimulatory signals from the v-Abl oncoprotein and growth-suppressive signals from the p19(Arf)-p53 tumor suppressor pathway oppose each other and influence the outcome of infection. The process involves a proliferative phase during which highly viable primary transformants expand, followed by a period of marked apoptosis (called "crisis") that is dependent on the presence of p19(Arf) and p53; rare cells that survive this phase emerge as fully transformed and malignant. To understand the way in which v-Abl expression affects p19(Arf) expression, we examined changes in expression of Arf during all stages of Ab-MLV transformation process. As is consistent with the ability of v-Abl to stimulate Myc, a transcription factor known to induce p19(Arf), Myc and Arf are induced soon after infection and p19(Arf) is expressed. At these early time points, the infected cells remain highly viable. The onset of crisis is marked by an increase in p19(Arf) expression and a change in localization of the protein from the nucleoplasm to the nucleolus. These data together suggest that the localization and expression levels of p19(Arf) modulate the effects of the protein during oncogenesis and reveal that the p19(Arf)-mediated response is subject to multiple layers of regulation that influence its function during Ab-MLV-mediated transformation.


Asunto(s)
Virus de la Leucemia Murina de Abelson/genética , Apoptosis , Linfocitos B/virología , Transformación Celular Viral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Animales , Linfocitos B/patología , Línea Celular Transformada , Cruzamientos Genéticos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Citometría de Flujo , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Heterocigoto , Indoles/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH
4.
J Virol ; 82(11): 5307-15, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18367522

RESUMEN

Abelson murine leukemia virus (Ab-MLV) arose from a recombination between gag sequences in Moloney MLV (Mo-MLV) and the c-abl proto-oncogene. The v-Abl oncoprotein encoded by Ab-MLV contains MA, p12, and a portion of CA sequences derived from the gag gene of Mo-MLV. Previous studies indicated that alteration of MA sequences affects the biology of Mo-MLV and Ab-MLV. To understand the role of these sequences in Ab-MLV transformation more fully, alanine substitution mutants that affect Mo-MLV replication were examined in the context of Ab-MLV. Mutations affecting Mo-MLV replication decreased transformation, while alanine mutations in residues dispensable for Mo-MLV replication did not. The altered v-Abl proteins displayed aberrant subcellular localization that correlated to transformation defects. Immunofluorescent analyses suggested that aberrant trafficking of the altered proteins and improper interaction with components of the cytoskeleton were involved in the phenotype. Similar defects in localization were observed when the Gag moiety containing these mutations was expressed in the absence of abl-derived sequences. These results indicate that MA sequences within the Gag moiety of the v-Abl protein contribute to proper localization by playing a dominant role in trafficking of the v-Abl molecule.


Asunto(s)
Virus de la Leucemia Murina de Abelson/metabolismo , Productos del Gen gag/química , Productos del Gen gag/metabolismo , Virus de la Leucemia Murina de Moloney/metabolismo , Proteínas Oncogénicas v-abl/química , Proteínas Oncogénicas v-abl/metabolismo , Virus de la Leucemia Murina de Abelson/química , Virus de la Leucemia Murina de Abelson/genética , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Productos del Gen gag/genética , Modelos Moleculares , Datos de Secuencia Molecular , Virus de la Leucemia Murina de Moloney/química , Virus de la Leucemia Murina de Moloney/genética , Mutación/genética , Oligosacáridos/química , Oligosacáridos/metabolismo , Proteínas Oncogénicas v-abl/genética , Péptidos/química , Péptidos/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia , Homología de Secuencia de Aminoácido
5.
J Virol ; 81(17): 9461-8, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17596313

RESUMEN

Like the v-Onc proteins encoded by many transforming retroviruses, the v-Abl protein is expressed as a Gag-Onc fusion. Although the Gag-derived myristoylation signal targets the v-Abl protein to the plasma membrane, the protein contains the entire MA and p12 sequences and a small number of CA-derived residues. To understand the role of Gag sequences in transformation, mutants lacking portions of these sequences were examined for the effects of these deletions on v-Abl function and localization. Deletion of the N-terminal third of p12 or all of p12 enhanced the transformation of both pre-B cells and NIH 3T3 cells. In contrast, deletions in MA or a deletion removing all of Gag except the first 34 amino acids important for myristoylation highly compromised the ability to transform either cell type. Although all of the mutant proteins retained kinase activity, those defective in transformation were reduced in their ability to activate Erk, suggesting a role for Gag sequences in v-Abl signaling. Immunofluorescence analysis revealed that a v-Abl protein retaining only the first 34 amino acids of Gag localized to the nucleus. These data indicate that Gag sequences are important for normal v-Abl signaling and that they suppress nuclear localization of the molecule.


Asunto(s)
Virus de la Leucemia Murina de Abelson/fisiología , Núcleo Celular/metabolismo , Transformación Celular Viral/fisiología , Productos del Gen gag/fisiología , Proteínas Oncogénicas v-abl/metabolismo , Transporte Activo de Núcleo Celular , Animales , Linfocitos B/virología , Línea Celular , Núcleo Celular/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Productos del Gen gag/genética , Humanos , Ratones , Microscopía Fluorescente , Proteínas Oncogénicas v-abl/análisis , Fosforilación , Transporte de Proteínas , Eliminación de Secuencia
6.
J Virol ; 81(13): 7274-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17428873

RESUMEN

Infection with a recombinant murine-feline gammaretrovirus, MoFe2, or with the parent virus, Moloney murine leukemia virus, caused significant reduction in B-lymphoid differentiation of bone marrow at 2 to 8 weeks postinfection. The suppression was selective, in that myeloid potential was significantly increased by infection. Analysis of cell surface markers and immunoglobulin H gene rearrangements in an in vitro model demonstrated normal B-lymphoid differentiation after infection but significantly reduced viability of differentiating cells. This reduction in viability may confer a selective advantage on undifferentiated lymphoid progenitors in the bone marrow of gammaretrovirus-infected animals and thereby contribute to the establishment of a premalignant state.


Asunto(s)
Linfocitos B/metabolismo , Diferenciación Celular , Virus de la Leucemia Felina/metabolismo , Virus de la Leucemia Murina de Moloney/metabolismo , Células Progenitoras Mieloides/metabolismo , Infecciones por Retroviridae/metabolismo , Infecciones Tumorales por Virus/mortalidad , Animales , Linfocitos B/patología , Linfocitos B/virología , Médula Ósea/metabolismo , Médula Ósea/patología , Médula Ósea/virología , Gatos , Hematopoyesis Extramedular , Ratones , Células Progenitoras Mieloides/patología , Células Progenitoras Mieloides/virología , Infecciones por Retroviridae/patología , Infecciones Tumorales por Virus/patología
7.
Mol Cell Biol ; 25(24): 10940-52, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16314517

RESUMEN

The multifunctional transcription factor TFII-I is tyrosine phosphorylated in response to extracellular growth signals and transcriptionally activates growth-promoting genes. However, whether activation of TFII-I also directly affects the cell cycle profile is unknown. Here we show that under normal growth conditions, TFII-I is recruited to the cyclin D1 promoter and transcriptionally activates this gene. Most strikingly, upon cell cycle arrest resulting from genotoxic stress and p53 activation, TFII-I is ubiquitinated and targeted for proteasomal degradation in a p53- and ATM (ataxia telangiectasia mutated)-dependent manner. Consistent with a direct role of TFII-I in cell cycle regulation and cellular proliferation, stable and ectopic expression of wild-type TFII-I increases cyclin D1 levels, resulting in accelerated entry to and exit from S phase, and overcomes p53-mediated cell cycle arrest, despite radiation. We further show that the transcriptional regulation of cyclin D1 and cell cycle control by TFII-I are dependent on its tyrosine phosphorylation at positions 248 and 611, sites required for its growth signal-mediated transcriptional activity. Taken together, our data define TFII-I as a growth signal-dependent transcriptional activator that is critical for cell cycle control and proliferation and further reveal that genotoxic stress-induced degradation of TFII-I results in cell cycle arrest.


Asunto(s)
Ciclo Celular , Ciclina D1/genética , Factores de Transcripción TFII/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Proteínas de Unión al ADN/metabolismo , Rayos gamma , Humanos , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción TFII/antagonistas & inhibidores , Factores de Transcripción TFII/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinas/metabolismo
8.
J Virol ; 79(18): 11618-26, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16140739

RESUMEN

The Abelson murine leukemia virus (Ab-MLV), like other retroviruses that contain v-onc genes, arose following a recombination event between a replicating retrovirus and a cellular oncogene. Although experimentally validated models have been presented to address the mechanism by which oncogene capture occurs, very little is known about the events that influence emerging viruses following the recombination event that incorporates the cellular sequences. One feature that may play a role is the genetic makeup of the host in which the virus arises; a number of host genes, including oncogenes and tumor suppressor genes, have been shown to affect the pathogenesis of many murine leukemia viruses. To examine how a host gene might affect an emerging v-onc gene-containing retrovirus, we studied the weakly oncogenic Ab-MLV-P90A strain, a mutant that generates highly oncogenic variants in vivo, and compared the viral populations in normal mice and mice lacking the p53 tumor suppressor gene. While variants arose in both p53+/+ and p53-/- tumors, the samples from the wild-type animals contained a more diverse virus population. Differences in virus population diversity were not observed when wild-type and null animals were infected with a highly oncogenic wild-type strain of Ab-MLV. These results indicate that p53, and presumably other host genes, affects the selective forces that operate on virus populations in vivo and likely influences the evolution of oncogenic retroviruses such as Ab-MLV.


Asunto(s)
Virus de la Leucemia Murina de Abelson/genética , Virus de la Leucemia Murina de Abelson/patogenicidad , Genes p53 , Leucemia Experimental/genética , Leucemia Experimental/virología , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/virología , Virus de la Leucemia Murina de Abelson/aislamiento & purificación , Animales , Secuencia de Bases , ADN Viral/genética , Femenino , Variación Genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mutación , Virulencia/genética , Integración Viral/genética
9.
J Virol ; 79(4): 2325-34, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15681433

RESUMEN

The v-Abl protein tyrosine kinase encoded by Abelson murine leukemia virus (Ab-MLV) induces pre-B-cell transformation. Signals emanating from the SH2 domain of the protein are required for transformation, and several proteins bind this region of v-Abl. One such protein is the adaptor molecule Shc, a protein that complexes with Grb2/Sos and facilitates Ras activation, an event associated with Ab-MLV transformation. To test the role this interaction plays in growth and survival of infected pre-B cells, dominant-negative (DN) Shc proteins were coexpressed with v-Abl and transformation was examined. Expression of DN Shc reduced Ab-MLV pre-B-cell transformation and decreased the ability of v-Abl to stimulate Ras activation and Erk phosphorylation in a Raf-dependent but Rac-independent fashion. Further analysis revealed that Shc is required for v-Abl-mediated Raf tyrosine 340 and 341 phosphorylation, an event associated with Erk phosphorylation. In contrast to effects on proliferation, survival of the cells and activation of Akt were not affected by expression of DN Shc. Together, these data reveal that v-Abl-Shc interactions are a critical part of the growth stimulatory signals delivered during transformation but that they do not affect antiapoptotic pathways. Furthermore, these data highlight a novel role for Shc in signaling from v-Abl to Raf.


Asunto(s)
Virus de la Leucemia Murina de Abelson/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Transformación Celular Viral/fisiología , Virus de la Leucemia Murina de Abelson/química , Virus de la Leucemia Murina de Abelson/genética , Animales , Línea Celular , Proliferación Celular , Proteína Adaptadora GRB2 , Regulación Viral de la Expresión Génica , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Proteínas ras/metabolismo
10.
J Virol ; 78(7): 3304-11, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15016851

RESUMEN

Pre-B-cell transformation by Abelson virus (Ab-MLV) is a multistep process in which primary transformants are stimulated to proliferate but subsequently undergo crisis, a period of erratic growth marked by high levels of apoptosis. Inactivation of the p53 tumor suppressor pathway is an important step in this process and can be accomplished by mutation of p53 or down-modulation of p19(Arf), a p53 regulatory protein. Consistent with these data, pre-B cells from either p53 or Ink4a/Arf null mice bypass crisis. However, the Ink4a/Arf locus encodes both p19(Arf) and a second tumor suppressor, p16(Ink4a), that blocks cell cycle progression by inhibiting Cdk4/6. To determine if p16(Ink4a) plays a role in Ab-MLV transformation, primary transformants derived from Arf(-/-) and p16(Ink4a(-/-)) mice were compared. A fraction of those derived from Arf(-/-) animals underwent crisis, and even though all p16(Ink4a(-/-)) primary transformants experienced crisis, these cells became established more readily than cells derived from +/+ mice. Analyses of Ink4a/Arf(-/-) cells infected with a virus that expresses both v-Abl and p16(Ink4a) revealed that p16(Ink4a) expression does not alter cell cycle profiles but does increase the level of apoptosis in primary transformants. These results indicate that both products of the Ink4a/Arf locus influence Ab-MLV transformation and reveal that in addition to its well-recognized effects on the cell cycle, p16(Ink4a) can suppress transformation by inducing apoptosis.


Asunto(s)
Virus de la Leucemia Murina de Abelson/fisiología , Apoptosis , Transformación Celular Viral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Linfocitos B/virología , Línea Celular Transformada , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Eliminación de Gen , Ratones , Células Madre/metabolismo , Células Madre/patología , Células Madre/virología , Proteína p14ARF Supresora de Tumor/deficiencia , Proteína p14ARF Supresora de Tumor/genética , Proteína p14ARF Supresora de Tumor/metabolismo
11.
Blood ; 103(11): 4268-75, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-14976048

RESUMEN

BCR-ABL and v-ABL are oncogenic forms of the Abl tyrosine kinase that can cause leukemias in mice and humans. ABL oncogenes trigger multiple signaling pathways whose contribution to transformation varies among cell types. Activation of phosphoinositide 3-kinase (PI3K) is essential for ABL-dependent proliferation and survival in some cell types, and global PI3K inhibitors can enhance the antileukemia effects of the Abl kinase inhibitor imatinib. Although a significant fraction of BCR-ABL-induced human leukemias are of B-cell origin, little is known about PI3K signaling mechanisms in B-lineage cells transformed by ABL oncogenes. Here we show that activation of class I(A) PI3K and downstream inactivation of FOXO transcription factors are essential for survival of murine pro/pre-B cells transformed by v-ABL or BCR-ABL. In addition, analysis of mice lacking individual PI3K genes indicates that products of the Pik3r1 gene contribute to transformation efficiency by BCR-ABL. These findings establish a role for PI3K signaling in B-lineage transformation by ABL oncogenes.


Asunto(s)
Linfocitos B/fisiología , Transformación Celular Neoplásica/metabolismo , Leucemia/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/genética , Transducción de Señal/fisiología , Animales , División Celular/inmunología , Linaje de la Célula/inmunología , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/genética , Embarazo , Proteínas Quinasas/metabolismo , Serina-Treonina Quinasas TOR , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
J Virol ; 78(4): 1636-44, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14747529

RESUMEN

Suppression of apoptosis is an important feature of the Abelson murine leukemia virus (Ab-MLV) transformation process. During multistep transformation, Ab-MLV-infected pre-B cells undergo p53-dependent apoptosis during the crisis phase of transformation. Even once cells are fully transformed, an active v-Abl protein tyrosine kinase is required to suppress apoptosis because cells transformed by temperature-sensitive (ts) kinase mutants undergo rapid apoptosis after a shift to the nonpermissive temperature. However, inactivation of the v-Abl protein by a temperature shift interrupts signals transmitted via multiple pathways, making it difficult to identify those that are critically important for the suppression of apoptosis. To begin to dissect these pathways, we tested the ability of an SH2 domain Ab-MLV mutant, P120/R273K, to rescue aspects of the ts phenotype of pre-B cells transformed by the conditional kinase domain mutant. The P120/R273K mutant suppressed apoptosis at the nonpermissive temperature, a phenotype correlated with its ability to activate Akt. Apoptosis also was suppressed at the nonpermissive temperature by constitutively active Akt and in p53-null pre-B cells transformed with the ts kinase domain mutant. These data indicate that an intact Src homology 2 (SH2) domain is not critical for apoptosis suppression and suggest that signals transmitted through Akt and p53 play an important role in the response.


Asunto(s)
Virus de la Leucemia Murina de Abelson/fisiología , Apoptosis , Linfocitos B/virología , Células de la Médula Ósea/virología , Transformación Celular Viral , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Virus de la Leucemia Murina de Abelson/genética , Animales , Línea Celular Transformada , Humanos , Ratones , Proteínas Oncogénicas v-abl/genética , Proteínas Oncogénicas v-abl/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt , Proteína p53 Supresora de Tumor/genética , Dominios Homologos src
13.
J Virol ; 77(11): 6208-15, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12743277

RESUMEN

The v-Abl protein encoded by Abelson murine leukemia virus (Ab-MLV) induces transformation of pre-B cells via a two-stage process. An initial proliferative phase during which cells with limited tumorigenic potential expand is followed by a crisis period marked by high levels of apoptosis and erratic growth. Transformants that survive this phase emerge as fully malignant cells and usually contain mutations that disable the p53 tumor suppressor pathway. Consistent with the importance of p53 in this process, pre-B cells from p53 null animals bypass crisis. Thus, the transformation process reflects a balance between signals from the v-Abl protein that drive transformation and those coming from the cellular response to inappropriate growth. One prediction of this hypothesis is that Ab-MLV mutants that are compromised in their ability to transform cells may be less equipped to overcome the effects of p53. To test this idea, we examined the ability of the P120/R273K mutant to transform pre-B cells from wild-type, p53 null, and Ink4a/Arf null mice. The SH2 domain of the v-Abl protein encoded by this mutant contains a substitution that affects the phosphotyrosine-binding pocket, and this mutant is compromised in its ability to transform NIH 3T3 and pre-B cells, especially at 39.5 degrees C. Our data reveal that loss of p53 or Ink4a/Arf locus products complements the transforming defect of the P120/R273K mutant, but it does not completely restore wild-type function. These results indicate that one important transforming function of v-Abl proteins is overcoming the effects of a functional p53 pathway.


Asunto(s)
Virus de la Leucemia Murina de Abelson/patogenicidad , Transformación Celular Viral , Mutación , Proteínas Oncogénicas v-abl/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Células 3T3 , Animales , Apoptosis , Linfocitos B/virología , Línea Celular , Línea Celular Transformada , Humanos , Ratones , Proteínas Oncogénicas v-abl/química , Proteínas Oncogénicas v-abl/metabolismo , Temperatura , Proteína p53 Supresora de Tumor/deficiencia , Dominios Homologos src
14.
J Virol ; 77(8): 4617-25, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12663768

RESUMEN

The v-Abl protein tyrosine kinase encoded by Abelson murine leukemia virus (Ab-MLV) induces transformation of pre-B cells in vivo and in vitro and can transform immortalized fibroblast cell lines in vitro. Although the kinase activity of the protein is required for these events, most previously studied mutants encoding truncated v-Abl proteins that lack the extreme carboxyl terminus retain high transforming capacity in NIH 3T3 cells but transform lymphocytes poorly. To understand the mechanisms responsible for poor lymphoid transformation, mutants expressing a v-Abl protein lacking portions of the COOH terminus were compared for their ability to transform pre-B cells. Although all mutants lacking sequences within the COOH terminus were compromised for lymphoid transformation, loss of amino acids in the central region of the COOH terminus, including those implicated in JAK interaction and DNA binding, decreased transformation twofold or less. In contrast, loss of the extreme COOH terminus rendered the protein unstable and led to rapid proteosome-mediated degradation, a feature that was more prominent when the protein was expressed in Ab-MLV-transformed lymphoid cells. These data indicate that the central portion of the COOH terminus is not essential for lymphoid transformation and reveal that one important function of the COOH terminus is to stabilize the v-Abl protein in lymphoid cells.


Asunto(s)
Virus de la Leucemia Murina de Abelson/fisiología , Transformación Celular Viral , Regulación Viral de la Expresión Génica , Linfocitos/virología , Proteínas Oncogénicas v-abl/química , Células 3T3 , Virus de la Leucemia Murina de Abelson/química , Virus de la Leucemia Murina de Abelson/genética , Animales , Secuencia de Bases , Línea Celular , Línea Celular Transformada , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eliminación de Gen , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Oncogénicas v-abl/genética , Proteínas Oncogénicas v-abl/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA