Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 7(1): e28568, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22253692

RESUMO

Human cancers often contain genetic alterations that disable G1/S checkpoint control and loss of this checkpoint is thought to critically contribute to cancer generation by permitting inappropriate proliferation and distorting fate-driven cell cycle exit. The identification of cell permeable small molecules that activate the G1/S checkpoint may therefore represent a broadly applicable and clinically effective strategy for the treatment of cancer. Here we describe the identification of several novel small molecules that trigger G1/S checkpoint activation and characterise the mechanism of action for one, CCT020312, in detail. Transcriptional profiling by cDNA microarray combined with reverse genetics revealed phosphorylation of the eukaryotic initiation factor 2-alpha (EIF2A) through the eukaryotic translation initiation factor 2-alpha kinase 3 (EIF2AK3/PERK) as the mechanism of action of this compound. While EIF2AK3/PERK activation classically follows endoplasmic reticulum (ER) stress signalling that sets off a range of different cellular responses, CCT020312 does not trigger these other cellular responses but instead selectively elicits EIF2AK3/PERK signalling. Phosphorylation of EIF2A by EIF2A kinases is a known means to block protein translation and hence restriction point transit in G1, but further supports apoptosis in specific contexts. Significantly, EIF2AK3/PERK signalling has previously been linked to the resistance of cancer cells to multiple anticancer chemotherapeutic agents, including drugs that target the ubiquitin/proteasome pathway and taxanes. Consistent with such findings CCT020312 sensitizes cancer cells with defective taxane-induced EIF2A phosphorylation to paclitaxel treatment. Our work therefore identifies CCT020312 as a novel small molecule chemical tool for the selective activation of EIF2A-mediated translation control with utility for proof-of-concept applications in EIF2A-centered therapeutic approaches, and as a chemical starting point for pathway selective agent development. We demonstrate that consistent with its mode of action CCT020312 is capable of delivering potent, and EIF2AK3 selective, proliferation control and can act as a sensitizer to chemotherapy-associated stresses as elicited by taxanes.


Assuntos
Ativadores de Enzimas/farmacologia , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , eIF-2 Quinase/metabolismo , Animais , Análise por Conglomerados , Ciclina D1/metabolismo , DNA Complementar/genética , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/química , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Paclitaxel/farmacologia , Fosforilação/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
2.
J Biol Chem ; 286(11): 9713-25, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21233209

RESUMO

Cyclin/cyclin-dependent kinase (CDK) complexes are critical regulators of cellular proliferation. A complex network of regulatory mechanisms has evolved to control their activity, including activating and inactivating phosphorylation of the catalytic CDK subunit and inhibition through specific regulatory proteins. Primate herpesviruses, including the oncogenic Kaposi sarcoma herpesvirus, encode cyclin D homologues. Viral cyclins have diverged from their cellular progenitor in that they elicit holoenzyme activity independent of activating phosphorylation by the CDK-activating kinase and resistant to inhibition by CDK inhibitors. Using sequence comparison and site-directed mutagenesis, we performed molecular analysis of the cellular cyclin D and the Kaposi sarcoma herpesvirus-cyclin to delineate the molecular mechanisms behind their different behavior. This provides evidence that a surface recognized for its involvement in the docking of CIP/KIP inhibitors is required and sufficient to modulate cyclin-CDK response to a range of regulatory cues, including INK4 sensitivity and CDK-activating kinase dependence. Importantly, amino acids in this region are critically linked to substrate selection, suggesting that a mutational drift in this surface simultaneously affects function and regulation. Together our work provides novel insight into the molecular mechanisms governing cyclin-CDK function and regulation and defines the biological forces that may have driven evolution of viral cyclins.


Assuntos
Ciclina D/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Evolução Molecular , Herpesvirus Humano 8/enzimologia , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Ciclina D/química , Ciclina D/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/química , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Quinases Ciclina-Dependentes/química , Quinases Ciclina-Dependentes/genética , Humanos , Modelos Biológicos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fosforilação , Análise de Sequência de Proteína , Proteínas Virais/química , Proteínas Virais/genética
3.
J Biol Chem ; 280(43): 35844-58, 2005 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-16115893

RESUMO

Kaposi sarcoma-associated herpes virus (KSHV) encodes a D-like cyclin (K-cyclin) that is thought to contribute to the viral oncogenicity. K-cyclin activates cellular cyclin-dependent kinases (CDK) 4 and 6, generating enzymes with a substrate selectivity deviant from CDK4 and CDK6 activated by D-type cyclins, suggesting different biochemical and biological functions. Here we report the identification of the actin- and calmodulin-binding protein caldesmon (CALD1) as a novel K-cyclin.CDK substrate, which is not phosphorylated by D.CDK. CALD1 plays a central role in the regulation of microfilament organization, consequently controlling cell shape, adhesion, cytokinesis and motility. K-cyclin.CDK6 specifically phosphorylates four Ser/Thr sites in the human CALD1 carboxyl terminus, abolishing CALD1 binding to its effector protein, actin, and its regulator protein, calmodulin. CALD1 is hyperphosphorylated in cells following K-cyclin expression and in KSHV-transformed lymphoma cells. Moreover, expression of exogenous K-cyclin results in microfilament loss and changes in cell morphology; both effects are reliant on CDK catalysis and can be reversed by the expression of a phosphorylation defective CALD1. Together, these data strongly suggest that K-cyclin expression modulates the activity of caldesmon and through this the microfilament functions in cells. These results establish a novel link between KSHV infection and the regulation of the actin cytoskeleton.


Assuntos
Proteínas de Ligação a Calmodulina/química , Quinase 6 Dependente de Ciclina/metabolismo , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/metabolismo , Actinas/química , Animais , Proteínas de Ligação a Calmodulina/metabolismo , Catálise , Cromatografia de Afinidade , Clonagem Molecular , Quinase 4 Dependente de Ciclina/metabolismo , Citoesqueleto/metabolismo , Eletroforese em Gel de Poliacrilamida , Células HeLa , Humanos , Espectrometria de Massas , Camundongos , Microscopia de Fluorescência , Células NIH 3T3 , Peptídeos/química , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/química , Proteína do Retinoblastoma/metabolismo , Sefarose/química , Serina/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Especificidade por Substrato , Treonina/química , Fatores de Tempo
4.
Anal Biochem ; 320(1): 66-74, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12895470

RESUMO

The tumor suppressor protein, pRb, regulates progression through the G1 phase of the cell cycle by its ability to bind to and regulate the activity of a variety of transcription factors. This function of pRb is disabled through its phosphorylation by the cyclin-dependent kinase (CDK) family of serine/threonine kinases. In many human cancers, genetic alteration such as loss of CDK inhibitor function and deregulated G1 cyclin expression leads to inappropriate phosphorylation and hence inactivation of this tumor suppressor. Identification of cell-permeable small molecules that block pRb phosphorylation in these tumors could therefore lead to development of an effective anticancer treatment. As a result, we have developed a high-throughput assay to detect changes in the level of pRb phosphorylation in cells. Signal detection is by a time-resolved fluorescence-based cellular immunosorbant assay on a fixed monolayer of cells. This comprises a mouse monoclonal antibody that recognizes the phosphorylated form of serine 608 on pRb, a known site of CDK phosphorylation, and a Europium-labeled secondary antibody for signal detection. The assay is reproducible and amenable to automation and has been used to screen 2000 compounds in a search for cell-permeable small molecules that will block pRb phosphorylation.


Assuntos
Fluorimunoensaio/métodos , Proteína do Retinoblastoma/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Linhagem Celular Tumoral , Membrana Celular/química , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Humanos , Camundongos , Fosforilação , Purinas/farmacologia , Proteína do Retinoblastoma/antagonistas & inibidores , Roscovitina
5.
Haematologica ; 87(4): 357-62, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11940479

RESUMO

BACKGROUND AND OBJECTIVES: A female patient presented with splenomegaly and lymphocytosis with atypical lymphoid cell morphology. We identified t(2;7)(p12;q21) prompting studies of the translocation breakpoint and its consequences on protein expression to confirm or otherwise the recently reported involvement of CDK6 and IG k genes in the t(2;7) leading to over-expression of CDK6 protein. DESIGN AND METHODS: A variety of clinical and laboratory techniques including cell marker, cytogenetic and histologic studies were applied in order to establish the diagnosis. Fluorescence in situ hybridization (FISH) and Southern blotting were used for mapping the translocation breakpoint and Western blotting for assessing protein expression. RESULTS: Immunophenotyping showed the presence of a B-cell population with strong expression of FMC7, CD22, CD79b, CD5 and k restricted surface immunoglobulins. Based on morphology and immunophenotypic markers the diagnosis of B-cell non-Hodgkin's lymphoma was made. Karyotyping revealed a clone with t(2;7)(p12;q21-22). Evidence for clonal evolution with additional abnormalities including a deletion of the TP53 was present. We established by FISH and Southern blotting that the breakpoint on 7q21-22 fell in a region 66kb telomeric to the previously reported breakpoint for the t(2;7) and was the same as that observed in a t(7;21). CDK6 protein was over-expressed. The patient received alkylating agents and splenectomy and is alive but the lymphocytosis persists with evidence of disease progression. INTERPRETATIONS AND CONCLUSIONS: We have demonstrated that CDK6 expression is dysregulated even when the breakpoint on 7q21-22 is located 66kb upstream from the coding region. Interestingly, the precise assignment of the lymphoma type in our case was not possible even when the splenic histology was analyzed.


Assuntos
Cromossomos Humanos Par 2 , Cromossomos Humanos Par 7 , Quinases Ciclina-Dependentes , Leucemia/genética , Linfoma não Hodgkin/genética , Proteínas Serina-Treonina Quinases/genética , Translocação Genética , Idoso , Mapeamento Cromossômico , Quinase 6 Dependente de Ciclina , Análise Citogenética , Feminino , Regulação da Expressão Gênica , Humanos , Linfoma não Hodgkin/etiologia , Linfoma não Hodgkin/metabolismo , Segunda Neoplasia Primária/genética , Proteínas Serina-Treonina Quinases/metabolismo
6.
Oncogene ; 21(12): 1823-31, 2002 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-11896614

RESUMO

The Kaposi's Sarcoma associated Herpes virus (KSHV) encodes two genes with the potential to affect the activity of the retinoblastoma protein (Rb). Open reading frame (orf) 72 encodes a D type cyclin (kcyc) that can elicit p16INK4a resistant cdk activity and orf73 encodes the latency associated nuclear antigen (LNA) that can bind Rb and neutralize E2F regulation. This indicates that, like papilloma and adenovirus associated malignancies, those associated with KSHV are defective with respect to their Rb pathway. To address this we investigated whether KSHV associated primary effusion lymphoma (PEL) derived cell lines are resistant to growth inhibition by p16INK4a. We provide evidence that ectopic expression of p16INK4a in these cells causes an Rb dependent G1 cell cycle block. Importantly, endogenous p16INK4a expression is not detected in six PEL derived cell lines and four primary PEL samples and examination of the p16INK4a locus shows deletion in two out of six and hypermethylation in four out of six PEL lines. Treatment of the latter with the demethylating agent 5'-aza-2' deoxycytidine leads to re-expression of p16INK4a protein. Taken together these results suggest that p16INK4a loss may be a cellular change frequently associated with PEL. They furthermore argue that despite the presence of KSHV DNA and expression of a latent gene program Rb function is intact in PEL.


Assuntos
Azacitidina/análogos & derivados , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Herpesvirus Humano 8/fisiologia , Linfoma de Células B/metabolismo , Proteína do Retinoblastoma/metabolismo , Sarcoma de Kaposi/metabolismo , Adenoviridae/genética , Azacitidina/farmacologia , Western Blotting , Ilhas de CpG , Inibidor p16 de Quinase Dependente de Ciclina/genética , Quinases Ciclina-Dependentes/metabolismo , Metilação de DNA , Primers do DNA/química , Decitabina , Regulação para Baixo , Deleção de Genes , Herpesvirus Humano 8/efeitos dos fármacos , Humanos , Linfoma de Células B/genética , Linfoma de Células B/virologia , Mutação , Reação em Cadeia da Polimerase , RNA Neoplásico/metabolismo , Proteína do Retinoblastoma/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virologia , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
7.
J Gen Virol ; 83(Pt 1): 179-188, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11752715

RESUMO

LANA, the major latency-associated nuclear antigen of Kaposi's sarcoma herpesvirus/human herpesvirus-8 (KSHV/HHV-8), binds RING3 protein, one of five human homologues of the fsh (female sterile homeotic) gene product of Drosophila. In KSHV/HHV-8-infected cells LANA and the viral episomes accumulate in heterochromatin-associated nuclear bodies. Here we show that in several KSHV/HHV-8-negative cell lines derived from carcinomas, sarcomas and lymphomas, RING3 was expressed at low levels, primarily localized to the euchromatin, and dissociated from the chromosomes during mitosis. In contrast, in KSHV/HHV-8-infected body cavity lymphoma cells the bulk of RING3 localizes to the LANA nuclear bodies and remains associated with the chromosomes during cell division. KSHV/HHV-8-infected body cavity lymphoma cells expressed RING3 at much higher levels than cells without the virus. Transfection of full-length LANA, but not the C terminus alone, greatly induced RING3 gene expression, and LANA and RING3 co-localized even in the transfected cells, in the absence of KSHV/HHV-8 viral DNA. High levels of LANA expression led to the disappearance of heterochromatin in both human and mouse cells. We suggest that LANA and RING3 may create a local euchromatic microenvironment around the viral episomes that are anchored to the heterochromatin.


Assuntos
Herpesvirus Humano 8/metabolismo , Heterocromatina/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Ativo do Núcleo Celular , Proteínas Cromossômicas não Histona , Expressão Gênica , Genoma Viral , Células HeLa , Herpesvirus Humano 8/genética , Humanos , Células Jurkat , Proteínas Serina-Treonina Quinases/genética , Sarcoma de Kaposi/virologia , Fatores de Transcrição , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...