Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Spine (Phila Pa 1976) ; 46(6): 347-355, 2021 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-33181779

RESUMO

STUDY DESIGN: Randomized controlled trial with 1-year follow up. OBJECTIVE: The aim of this study was to assess whether people with low back pain (LBP) and self-reported physically demanding jobs, benefit from an occupational medicine intervention, in addition to a single hospital consultation and a magnetic resonance imaging, at 1 year of follow-up. Secondly, to examine whether the positive health effects, found in both groups at 6 months, persist at 1-year follow-up. SUMMARY OF BACKGROUND DATA: The prevalence of LBP is high in the working population, resulting in a substantial social and economic burden. Although there are many guidelines available on the management of LBP, including multidisciplinary biopsychosocial rehabilitation, they provide limited guidance on the occupational medicine aspects. METHODS: As reported previously, 305 participants with LBP and self-reported physically demanding jobs were enrolled in the randomized controlled study and randomly allocated to clinical care with additional occupational medicine intervention or clinical care alone. Data were collected at baseline, 6 months, and 1 year. Outcomes included in the present 1-year follow-up study are changes in neuropathic pain (painDETECT questionnaire), severity of pain (0-10 numerical rating scale), disability (Roland Morris Disability Questionnaire), fear-avoidance beliefs (FABQ), physical, and mental quality of life (short-form 36). RESULTS: The study showed no effect of an occupational intervention on neuropathic pain, fear-avoidance beliefs, physical and mental quality of life nor disability measured after 1 year. The positive effects found at 6 months in both groups, remained at 1-year follow-up. CONCLUSION: The results suggest that a thorough clinical consultation, with focus on explaining the cause of pain and instructions to stay active, can promote long-lasting physical and mental health in individuals with LBP. Therefore, additional occupational interventions could focus on altering occupational obstacles on a structural level.Level of Evidence: 2.


Assuntos
Dor Lombar/terapia , Exposição Ocupacional/prevenção & controle , Saúde Ocupacional/tendências , Medicina do Trabalho/tendências , Adulto , Feminino , Seguimentos , Humanos , Dor Lombar/diagnóstico por imagem , Dor Lombar/etiologia , Imageamento por Ressonância Magnética/tendências , Masculino , Pessoa de Meia-Idade , Medicina do Trabalho/métodos , Qualidade de Vida , Autorrelato , Método Simples-Cego , Inquéritos e Questionários
3.
Sci Rep ; 8(1): 14196, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30242273

RESUMO

Heterotopic ossification (HO) is the process of bone formation in tissues that are not usually osseous. It occurs in 60% of those with blast-related amputations. HO can result in reduced range of motion, pain, nerve impingement and can affect prosthesis fitting and is caused by a combination of mechanical, biological, local and systemic factors. As with normal bone formation and remodelling, it is expected that heterotopic bone responds to mechanical stimuli and understanding this relationship can give insight into the pathology. The objective of this research was to investigate whether a physiological 2D computational model that considers both mechanical and biological factors can be used to simulate HO in the residual limb of a trans-femoral amputee. The study found that characteristic morphologies of HO were reproduced by adjusting the loading environment. Significant effects were produced by changing the loading direction on the femur; this is potentially associated with different initial surgical interventions such as muscle myodesis. Also, initial treatment such as negative pressure through a dressing was found to change the shape of heterotopic bone.


Assuntos
Extremidades/fisiopatologia , Fêmur/fisiopatologia , Ossificação Heterotópica/fisiopatologia , Osteogênese/fisiologia , Algoritmos , Amputação Cirúrgica/métodos , Amputados , Humanos , Dor/fisiopatologia , Amplitude de Movimento Articular/fisiologia
4.
Comput Methods Biomech Biomed Engin ; 21(3): 208-218, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29451004

RESUMO

Bone responds to mechanical stimulus and a range of pre-existing finite element models have been suggested to reproduce the internal physiological structure of bone. Inflammation effects are not included in these models, yet inflammation is a key component of bone repair in trauma. Therefore, a model is proposed and tested here that extends these methods to include parameters that could be considered to represent the behaviour of bone remodelling when influenced by inflammation. The proposed model regulates remodelling based on findings from recent studies into the nature of heterotopic ossification, the formation of heterotopic bone, which have revealed information about the nature of bone after high levels of trauma. These parameters include consideration of the distance from the zone of trauma, the density of mesenchymal stem cells, and substrate stiffness as a trigger for cells becoming osteogenic. The method is tested on a two-dimensional plate model and shows that the new extended algorithm can produce a range of structures depending on inputs that could be used in the future to replicate physiological scenarios.


Assuntos
Algoritmos , Remodelação Óssea/fisiologia , Simulação por Computador , Inflamação/patologia , Modelos Biológicos , Ferimentos e Lesões/fisiopatologia , Fenômenos Biomecânicos , Densidade Óssea/fisiologia , Análise de Elementos Finitos , Humanos
7.
J Virol ; 85(17): 9239-42, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21697469

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/patogenicidade , Transformação Celular Viral , Interações Hospedeiro-Patógeno , Proteínas Oncogênicas v-abl/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Inositol Polifosfato 5-Fosfatases , Camundongos , Células Precursoras de Linfócitos B/virologia , Mapeamento de Interação de Proteínas
8.
J Virol ; 82(17): 8383-91, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18579612

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/genética , Apoptose , Linfócitos B/virologia , Transformação Celular Viral , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Animais , Linfócitos B/patologia , Linhagem Celular Transformada , Cruzamentos Genéticos , Inibidor p16 de Quinase Dependente de Ciclina/genética , Citometria de Fluxo , Corantes Fluorescentes/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Heterozigoto , Indóis/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3
9.
Blood ; 112(1): 3, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18574033
10.
J Virol ; 82(11): 5307-15, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18367522

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/metabolismo , Produtos do Gene gag/química , Produtos do Gene gag/metabolismo , Vírus da Leucemia Murina de Moloney/metabolismo , Proteínas Oncogênicas v-abl/química , Proteínas Oncogênicas v-abl/metabolismo , Vírus da Leucemia Murina de Abelson/química , Vírus da Leucemia Murina de Abelson/genética , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Dimerização , Produtos do Gene gag/genética , Modelos Moleculares , Dados de Sequência Molecular , Vírus da Leucemia Murina de Moloney/química , Vírus da Leucemia Murina de Moloney/genética , Mutação/genética , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Proteínas Oncogênicas v-abl/genética , Peptídeos/química , Peptídeos/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
11.
J Virol ; 81(17): 9461-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17596313

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/fisiologia , Núcleo Celular/metabolismo , Transformação Celular Viral/fisiologia , Produtos do Gene gag/fisiologia , Proteínas Oncogênicas v-abl/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linfócitos B/virologia , Linhagem Celular , Núcleo Celular/química , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Produtos do Gene gag/genética , Humanos , Camundongos , Microscopia de Fluorescência , Proteínas Oncogênicas v-abl/análise , Fosforilação , Transporte Proteico , Deleção de Sequência
12.
J Virol ; 81(13): 7274-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17428873

RESUMO

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.


Assuntos
Linfócitos B/metabolismo , Diferenciação Celular , Vírus da Leucemia Felina/metabolismo , Vírus da Leucemia Murina de Moloney/metabolismo , Células Progenitoras Mieloides/metabolismo , Infecções por Retroviridae/metabolismo , Infecções Tumorais por Vírus/mortalidade , Animais , Linfócitos B/patologia , Linfócitos B/virologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Medula Óssea/virologia , Gatos , Hematopoese Extramedular , Camundongos , Células Progenitoras Mieloides/patologia , Células Progenitoras Mieloides/virologia , Infecções por Retroviridae/patologia , Infecções Tumorais por Vírus/patologia
13.
Mol Cell Biol ; 25(24): 10940-52, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16314517

RESUMO

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.


Assuntos
Ciclo Celular , Ciclina D1/genética , Fatores de Transcrição TFII/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Raios gama , Humanos , Camundongos , Células NIH 3T3 , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição TFII/antagonistas & inibidores , Fatores de Transcrição TFII/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinas/metabolismo
14.
J Virol ; 79(18): 11618-26, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16140739

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/genética , Vírus da Leucemia Murina de Abelson/patogenicidade , Genes p53 , Leucemia Experimental/genética , Leucemia Experimental/virologia , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/virologia , Vírus da Leucemia Murina de Abelson/isolamento & purificação , Animais , Sequência de Bases , DNA Viral/genética , Feminino , Variação Genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mutação , Virulência/genética , Integração Viral/genética
15.
J Virol ; 79(4): 2325-34, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15681433

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose , Transformação Celular Viral/fisiologia , Vírus da Leucemia Murina de Abelson/química , Vírus da Leucemia Murina de Abelson/genética , Animais , Linhagem Celular , Proliferação de Células , Proteína Adaptadora GRB2 , Regulação Viral da Expressão Gênica , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Proteínas ras/metabolismo
16.
J Virol ; 78(7): 3304-11, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15016851

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/fisiologia , Apoptose , Transformação Celular Viral , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Animais , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos B/virologia , Linhagem Celular Transformada , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/genética , Deleção de Genes , Camundongos , Células-Tronco/metabolismo , Células-Tronco/patologia , Células-Tronco/virologia , Proteína Supressora de Tumor p14ARF/deficiência , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p14ARF/metabolismo
17.
Blood ; 103(11): 4268-75, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-14976048

RESUMO

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.


Assuntos
Linfócitos B/fisiologia , Transformação Celular Neoplásica/metabolismo , Leucemia/fisiopatologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/genética , Transdução de Sinais/fisiologia , Animais , Divisão Celular/imunologia , Linhagem da Célula/imunologia , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/genética , Gravidez , Proteínas Quinases/metabolismo , Serina-Treonina Quinases TOR , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
J Virol ; 78(4): 1636-44, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14747529

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/fisiologia , Apoptose , Linfócitos B/virologia , Células da Medula Óssea/virologia , Transformação Celular Viral , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Vírus da Leucemia Murina de Abelson/genética , Animais , Linhagem Celular Transformada , Humanos , Camundongos , Proteínas Oncogênicas v-abl/genética , Proteínas Oncogênicas v-abl/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Proteína Supressora de Tumor p53/genética , Domínios de Homologia de src
19.
J Virol ; 77(11): 6208-15, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12743277

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/patogenicidade , Transformação Celular Viral , Mutação , Proteínas Oncogênicas v-abl/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Células 3T3 , Animais , Apoptose , Linfócitos B/virologia , Linhagem Celular , Linhagem Celular Transformada , Humanos , Camundongos , Proteínas Oncogênicas v-abl/química , Proteínas Oncogênicas v-abl/metabolismo , Temperatura , Proteína Supressora de Tumor p53/deficiência , Domínios de Homologia de src
20.
J Virol ; 77(8): 4617-25, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12663768

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Abelson/fisiologia , Transformação Celular Viral , Regulação Viral da Expressão Gênica , Linfócitos/virologia , Proteínas Oncogênicas v-abl/química , Células 3T3 , Vírus da Leucemia Murina de Abelson/química , Vírus da Leucemia Murina de Abelson/genética , Animais , Sequência de Bases , Linhagem Celular , Linhagem Celular Transformada , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Deleção de Genes , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Oncogênicas v-abl/genética , Proteínas Oncogênicas v-abl/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...