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










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 32(6): 788-96, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22391562

RESUMO

There are few effective treatments for metastatic melanoma. Checkpoint kinase 1 (Chk1) inhibitors are being trialled for their efficacy in enhancing conventional chemotherapeutic agents, but their effectiveness as single agents is not known. We have examined the effectiveness of two novel Chk1 selective inhibitors, AR323 and AR678, in a panel of melanoma cell lines and normal cell types. We demonstrate that these drugs display single-agent activity, with IC50s in the low nanomolar range. The drugs produce cytotoxic effects in cell lines that are most sensitive to these drugs, whereas normal cells are only sensitive to these drugs at the higher concentrations where they have cytostatic activity. The cytotoxic effect is the consequence of inhibition of S-phase Chk1, which drives cells prematurely from late S phase into an aberrant mitosis and results in either failure of cytokinesis or cell death through an apoptotic mechanism. The sensitivity to the Chk1 inhibitors was correlated with the level of endogenous DNA damage indicating replicative stress. Chk1 inhibitors are viable single-agent therapies that target melanoma cells with high levels of endogenous DNA damage. This sensitivity suggests that Chk1 is a critical component of an adaptation to replicative stress in these cells. It also suggests that markers of DNA damage may be useful in identifying the melanomas and potentially other tumour types that are more likely to be sensitive to Chk1 inhibitors as single agents.


Assuntos
Proliferação de Células , Melanoma/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Dano ao DNA/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Fase S/efeitos dos fármacos , Estresse Fisiológico/genética
2.
Mar Biotechnol (NY) ; 11(2): 188-98, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18690486

RESUMO

A wide range of sessile and sedentary marine invertebrates synthesize secondary metabolites that have potential as industrial antifoulants. These antifoulants tend to differ in structure, even between closely related species. Here, we determine if structurally divergent secondary metabolites produced within two sympatric haliclonid demosponges have similar effects on the larvae of a wide range of benthic competitors and potential fouling metazoans (ascidians, molluscs, bryozoans, polychaetes, and sponges). The sponges Haliclona sp. 628 and sp. 1031 synthesize the tetracyclic alkaloid, haliclonacyclamine A (HA), and the long chain alkyl amino alcohol, halaminol A (LA), respectively. Despite structural differences, HA and LA have identical effects on phylogenetically disparate ascidian larvae, inducing rapid larval settlement but preventing subsequent metamorphosis at precisely the same stage. HA and LA also have similar effects on sponge, polychaete, gastropod and bryozoan larvae, inhibiting both settlement and metamorphosis. Despite having identical roles in preventing fouling and colonisation, HA and LA differentially affect the physiology of cultured HeLa human cells, indicating they have different molecular targets. From these data, we infer that the secondary metabolites within marine sponges may emerge by varying evolutionary and biosynthetic trajectories that converge on specific ecological roles.


Assuntos
Haliclona/fisiologia , Feromônios/química , Feromônios/farmacologia , Urocordados/efeitos dos fármacos , Animais , Células HeLa , Humanos , Invertebrados/efeitos dos fármacos , Larva/efeitos dos fármacos , Metamorfose Biológica/efeitos dos fármacos , Controle de Pragas
3.
Oncogene ; 27(31): 4261-8, 2008 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-18372919

RESUMO

Cyclin A/cdk2 has a role in progression through S phase, and a large pool is also activated in G2 phase. Here we report that this G2 phase pool regulates the timing of progression into mitosis. Knock down of cyclin A by siRNA or addition of a specific cdk2 small molecule inhibitor delayed entry into mitosis by delaying cells in G2 phase. The G2 phase-delayed cells contained elevated levels of inactive cyclin B/cdk1. However, increased microtubule nucleation at the centrosomes was observed, and the centrosomes stained for markers of cyclin B/cdk1 activity. Both microtubule nucleation at the centrosomes and the phosphoprotein markers were lost with short-term treatment of the cdk1/2 inhibitor roscovitine but not the Mek1/2 inhibitor U0126. Cyclin A/cdk2 localized at the centrosomes in late G2 phase after separation of the centrosomes but before the start of prophase. Thus G2 phase cyclin A/cdk2 controls the timing of entry into mitosis by controlling the subsequent activation of cyclin B/cdk1, but also has an unexpected role in coordinating the activation of cyclin B/cdk1 at the centrosome and in the nucleus.


Assuntos
Núcleo Celular/fisiologia , Centrossomo/fisiologia , Ciclina A/fisiologia , Quinase 2 Dependente de Ciclina/metabolismo , Mitose , Butadienos/farmacologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Fase G1 , Células HeLa , Humanos , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , Nitrilas/farmacologia , Purinas/farmacologia , RNA Interferente Pequeno , Roscovitina
4.
Oncogene ; 27(10): 1345-54, 2008 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-17828304

RESUMO

Chromosomal passenger proteins have emerged as key players in the regulation of mitosis and cytokinesis. Histone deacetylase inhibitors (HDACi) are a class of anticancer drugs that induce aberrant mitosis and can overcome the spindle assembly checkpoint. Here, we investigate the mechanism by which HDACi disrupt normal mitotic progression and checkpoint function. We demonstrate that HDACi treatment temporarily delays mitotic progression through prometaphase due to activation of the spindle assembly checkpoint. Despite failing to congress chromosomes to the metaphase plate, cells aberrantly segregate their chromosomes and exit mitosis to undergo a failed cytokinesis. We show that this premature exit from mitosis is a form of mitotic slippage. Chromosomal passenger proteins fail to accumulate at the centromere following HDACi treatment. This results in inadequate concentrations of chromosomal passenger proteins at the centromere, which are insufficient to regulate the phosphorylation of the kinetochore checkpoint component BubR1, and an inability to maintain the mitotic arrest. Thus, the centromeric accumulation of chromosomal passenger complex components is critical for regulating kinetochore but not centromeric processes, and failure of this accumulation underlies the HDACi-induced mitotic slippage.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Mitose/efeitos dos fármacos , Cromossomos/efeitos dos fármacos , Cromossomos/enzimologia , Células HeLa , Histona Desacetilases/metabolismo , Humanos , Fase S/efeitos dos fármacos , Fuso Acromático/efeitos dos fármacos
5.
Nucleic Acids Res ; 28(7): 1506-13, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10710416

RESUMO

DNA-PKcs, the catalytic subunit of DNA-dependent protein kinase (DNA-PK), has a phosphoinositol 3-kinase (PI 3-K) domain close to its C-terminus. Cell lines derived from the SCID mouse have been utilised as a model DNA-PKcs-defective system. The SCID mutation results in truncation of DNA-Pkcs at the extreme C-terminus leaving the PI 3-K domain intact. The mutated protein is expressed at low levels in most SCID cell lines, leaving open the question of whether the mutation abolishes kinase activity. Here, we show that a SCID cell line that expresses the mutant protein normally has dramatically impaired kinase activity. We estimate that the residual kinase activity typically present in SCID fibroblast cell lines is at least two orders of magnitude less than that found in control cells. Our results substantiate evidence that DNA-PKcs kinase activity is required for DSB rejoining and V(D)J recombination and show that the extreme C-terminal region of DNA-PKcs, present in PI 3-K-related protein kinases but absent in bona fide PI 3 lipid kinases, is required for DNA-PKcs to function as a protein kinase. We also show that expression of mutant DNA-PKcs protein confers a growth disadvantage, providing an explanation for the lack of DNA-PKcs expression in most SCID cell lines.


Assuntos
Proteínas de Ligação a DNA , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Linfócitos B/enzimologia , Sequência de Bases , Células CHO , Linhagem Celular , Cromossomos Artificiais de Levedura/genética , Sequência Conservada , Cricetinae , Primers do DNA/genética , Reparo do DNA/genética , Reparo do DNA/fisiologia , Proteína Quinase Ativada por DNA , Células-Tronco Hematopoéticas/enzimologia , Camundongos , Camundongos SCID , Mutagênese Sítio-Dirigida , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína
6.
Curr Biol ; 9(13): 699-702, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10395545

RESUMO

The major mechanism for the repair of DNA double-strand breaks (DSBs) in mammalian cells is non-homologous end-joining (NHEJ), a process that involves the DNA-dependent protein kinase [1] [2], XRCC4 and DNA ligase IV [3] [4] [5] [6]. Rodent cells and mice defective in these components are radiation-sensitive and defective in V(D)J-recombination, showing that NHEJ also functions to rejoin DSBs introduced during lymphocyte development [7] [8]. 180BR is a radiosensitive cell line defective in DSB repair, which was derived from a leukaemia patient who was highly sensitive to radiotherapy [9] [10] [11]. We have identified a mutation within a highly conserved motif encompassing the active site in DNA ligase IV from 180BR cells. The mutated protein is severely compromised in its ability to form a stable enzyme-adenylate complex, although residual activity can be detected at high ATP concentrations. Our results characterize the first patient with a defect in an NHEJ component and suggest that a significant defect in NHEJ that leads to pronounced radiosensitivity is compatible with normal human viability and does not cause any major immune dysfunction. The defect, however, may confer a predisposition to leukaemia.


Assuntos
DNA Ligases/genética , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Tolerância a Radiação/genética , Animais , Western Blotting , Linhagem Celular Transformada , DNA Ligase Dependente de ATP , DNA Ligases/metabolismo , Reparo do DNA/genética , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/genética , Fibroblastos/efeitos da radiação , Humanos , Mutação , Proteínas Nucleares , Leucemia-Linfoma Linfoblástico de Células Precursoras/radioterapia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Coelhos , Radiação Ionizante , Análise de Sequência de DNA
7.
C R Acad Sci III ; 322(2-3): 109-12, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10196660

RESUMO

The DNA-dependent protein kinase functions in the repair of DNA double strand breaks (DSBs) and in V(D)J recombination. To gain insight into the function of DNA-PK in this process we have carried out a mutation analysis of Ku80 and DNA-PKcs. Mutations at multiple sites within the N-terminal two thirds of Ku80 result in loss of Ku70/80 interaction, loss of DNA end-binding activity and inability to complement Ku80 defective cell lines. In contrast, mutations in the carboxy terminal region of the protein do not impair DNA end-binding activity but decrease the ability of Ku to activate DNA-PK. To gain insight into important functional domains within DNA-PKcs, we have analysed defective mutants, including the mouse scid cell line, and the rodent mutants, irs-20 and V-3. Mutational changes in the carboxy terminal region have been identified in all cases. Our results strongly suggest that the C-terminus of DNA-PKcs is required for kinase activity.


Assuntos
Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Recombinação Genética , Animais , Linhagem Celular , Cricetinae , Proteína Quinase Ativada por DNA , Camundongos , Mutação , Proteínas Serina-Treonina Quinases/metabolismo , Homologia de Sequência do Ácido Nucleico
8.
Immunity ; 9(3): 355-66, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9768755

RESUMO

The DNA-dependent protein kinase is a mammalian protein complex composed of Ku70, Ku80, and DNA-PKcs subunits that has been implicated in DNA double-strand break repair and V(D)J recombination. Here, by gene targeting, we have constructed a mouse with a disruption in the kinase domain of DNA-PKcs, generating an animal model completely devoid of DNA-PK activity. Our results demonstrate that DNA-PK activity is required for coding but not for signal join formation in mice. Although our DNA-PKcs defective mice closely resemble Scid mice, they differ by having elevated numbers of CD4+CD8+ thymocytes. This suggests that the Scid mice may not represent a null phenotype and may retain some residual DNA-PKcs function.


Assuntos
Proteínas de Ligação a DNA , Marcação de Genes , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Tolerância a Radiação/genética , Imunodeficiência Combinada Severa/genética , Animais , Linfócitos B/citologia , Catálise , Diferenciação Celular/genética , Células Cultivadas , Proteína Quinase Ativada por DNA , Embrião de Mamíferos , Fibroblastos/efeitos da radiação , Genes Codificadores dos Receptores de Linfócitos T/genética , Imunoglobulinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/fisiologia , Estrutura Terciária de Proteína , Recombinação Genética/genética , Linfócitos T/citologia
9.
Nucleic Acids Res ; 26(8): 1965-73, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9518490

RESUMO

The catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs) is a member of a sub-family of phosphatidylinositol (PI) 3-kinases termed PIK-related kinases. A distinguishing feature of this sub-family is the presence of a conserved C-terminal region downstream of a PI 3-kinase domain. Mutants defective in DNA-PKcs are sensitive to ionising radiation and are unable to carry out V(D)J recombination. Irs-20 is a DNA-PKcs-defective cell line with milder gamma-ray sensitivity than two previously characterised mutants, V-3 and mouse scid cells. Here we show that the DNA-PKcs protein from irs-20 cells can bind to DNA but is unable to function as a protein kinase. To verify the defect in irs-20 cells and provide insight into the function and expression of DNA-PKcs in double-strand break repair and V(D)J recombination we introduced YACs encoding human and mouse DNA-PKcs into defective mutants and achieved complementation of the defective phenotypes. Furthermore, in irs-20 we identified a mutation in DNA-PKcs that causes substitution of a lysine for a glutamic acid in the fourth residue from the C-terminus. This represents a strong candidate for the inactivating mutation and provides supportive evidence that the extreme C-terminal motif is important for protein kinase activity.


Assuntos
Sobrevivência Celular/efeitos da radiação , Proteínas de Ligação a DNA , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Células CHO , Linhagem Celular , Cromossomos Artificiais de Levedura , Cricetinae , DNA/metabolismo , Dano ao DNA , DNA Nucleotidiltransferases/metabolismo , Reparo do DNA , Proteína Quinase Ativada por DNA , Relação Dose-Resposta à Radiação , Raios gama , Biblioteca Gênica , Cavalos , Humanos , Camundongos , Camundongos SCID , Proteínas Nucleares , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase , Transfecção , VDJ Recombinases
10.
Oncogene ; 14(16): 1911-21, 1997 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-9150358

RESUMO

The recently cloned gene (ATM) mutated in the human genetic disorder ataxia-telangiectasia (A-T) is involved in DNA damage response at different cell cycle checkpoints and also appears to have a wider role in signal transduction. Antibodies prepared against peptides from the predicted protein sequence detected a approximately 350 kDa protein corresponding to the open reading frame, which was absent in 13/23 A-T homozygotes. Subcellular fractionation, immunoelectronmicroscopy and immunofluorescence showed that the ATM protein is present in the nucleus and cytoplasmic vesicles. This distribution did not change after irradiation. We also provide evidence that ATM protein binds to p53 and this association is defective in A-T cells compatible with the defective p53 response in these cells. These results provide further support for a role for the ATM protein as a sensor of DNA damage and in a more general role in cell signalling, compatible with the broader phenotype of the syndrome.


Assuntos
Ataxia Telangiectasia/genética , Organelas/ultraestrutura , Mutação Puntual , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases , Anticorpos , Proteínas Mutadas de Ataxia Telangiectasia , Western Blotting , Proteínas de Ciclo Celular , Linhagem Celular Transformada , Núcleo Celular/ultraestrutura , Clonagem Molecular , Grânulos Citoplasmáticos/ultraestrutura , Proteínas de Ligação a DNA , Ensaio de Imunoadsorção Enzimática , Herpesvirus Humano 4 , Homozigoto , Humanos , Microscopia Imunoeletrônica , Microssomos/ultraestrutura , Fases de Leitura Aberta , Organelas/metabolismo , Proteínas/análise , Proteínas Recombinantes/análise , Proteínas Recombinantes/biossíntese , Deleção de Sequência , Proteína Supressora de Tumor p53/análise , Proteínas Supressoras de Tumor
11.
J Biol Chem ; 272(14): 9489-95, 1997 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-9083089

RESUMO

A characteristic series of immunological abnormalities are observed in the human genetic disorder ataxia-telangiectasia (A-T). The recent cloning of a gene mutated in this syndrome provides additional evidence for a defect in intracellular signaling in A-T. We have investigated the possibility that signaling through the B cell antigen receptor is one manifestation of the A-T defect. In response to cross-linking of the B cell receptor, several A-T cell lines were defective in their mitogenic response; in addition Ca2+ mobilization from internal stores was either absent or considerably reduced in these cell lines in response to cross-linking. The defect in signaling was not due to difference in expression of surface immunoglobulin. The defective response in A-T cells was also evident in several arms of the intracellular cascade activated by B cell cross-linking. Tyrosine phosphorylation of phospholipase Cgamma1, a key step in activation of the enzyme, was reduced or negligible in some A-T cell lines. This defect in signaling was also seen at the level of Lyn tyrosine kinase activation and its association with and activation of phosphatidylinositol 3-kinase. Our results provide evidence for a role for the ATM gene product in intracellular signaling which may account at least in part for the abnormalities in B cell function in A-T.


Assuntos
Ataxia Telangiectasia/fisiopatologia , Transformação Celular Viral , Herpesvirus Humano 4 , Receptores de Antígenos de Linfócitos B/fisiologia , Transdução de Sinais , Cálcio/metabolismo , Divisão Celular , Linhagem Celular , Humanos , Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinases , Fosfolipase C gama , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de IgG/metabolismo , Fosfolipases Tipo C/metabolismo , Tirosina/metabolismo
12.
Oncogene ; 13(5): 963-70, 1996 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-8806686

RESUMO

The gene mutated in the human genetic disorder ataxia-telangiectasia (A-T) has been described recently (Savitsky et al., 1995a) and the complete coding sequence of this gene, ATM, has been reported (Savitsky et al., 1995b). The derived amino acid sequence demonstrates significant homologies to several proteins containing a phosphatidylinositol 3-kinase (PI3-kinase) domain, including the yeast TOR proteins and the human protein FRAP. Since the TOR and FRAP proteins are targets for the immunosuppressive drug rapamycin, we have investigated the effects of this compound on A-T cells. We report here that 3 A-T cell lines are more resistant than control cells to rapamycin's growth inhibiting effects but were more sensitive to the PI3-kinase inhibitor wortmannin. As expected rapamycin (1 nM) inhibited the rate of exit of control cells from G1 phase but failed to perturb the progression of A-T cells. This difference in cell cycle progress after rapamycin treatment is reflected in ribosomal S6 protein kinase (p70S6k) by both a downward mobility shift on SDS-PAGE and inhibition of activity. Furthermore, the G1 phase cyclin-dependent kinase, cyclin E-cdk2, was rapidly inhibited in control cells post-treatment, whereas in A-T cells it took considerably longer to observe inhibition. There was no evidence that a GST-FKBP12 fusion protein specifically precipitated the ATM protein in the presence of rapamycin in either cell type. These results demonstrate that the ATM protein is not a direct target for rapamycin but its functional loss renders cells more resistant to this compound.


Assuntos
Ataxia Telangiectasia/tratamento farmacológico , Resistência a Medicamentos/genética , Polienos/farmacologia , Proteínas/efeitos dos fármacos , Sequência de Aminoácidos , Androstadienos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Antifúngicos/farmacologia , Ataxia Telangiectasia/patologia , Proteínas Mutadas de Ataxia Telangiectasia , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Transporte/farmacologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/farmacologia , Humanos , Imunossupressores/farmacologia , Dados de Sequência Molecular , Mutação , Nocodazol/farmacologia , Fosfatidilinositol 3-Quinases , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Quinases S6 Ribossômicas , Sirolimo , Proteínas de Ligação a Tacrolimo , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor , Wortmanina
13.
J Biol Chem ; 271(34): 20486-93, 1996 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-8702789

RESUMO

The recent description of a novel gene (ATM) mutated in ataxia-telangiectasia (A-T), with homologies to genes encoding proteins involved in both G1/S and G2/M checkpoint control, points to a common defect in cell cycle control in A-T operating through the cyclin-dependent kinases. In this report we demonstrate that cyclin-dependent kinases are resistant to inhibition by ionizing radiation exposure in A-T cells, and this appears to be due to insufficient induction of WAF1. Exposure of control lymphoblastoid cells to radiation during S phase and in G2 phase causes a rapid inhibition of cyclin A-Cdk2 and cyclin B-Cdc2 activities, respectively. Irradiation led to a 5-20-fold increase in Cdk-associated WAF1 in these cells, which accounts at least in part for the decrease in cyclin-dependent kinase activity. In contrast, radiation did not inhibit any of the cyclin-dependent kinase activities in S phase or G2 phase in A-T cells at short times after irradiation nor was there any significant change in the level of Cdk-associated WAF1 compared to unirradiated cells. These results are similar to those reported previously for the G1 checkpoint and provide additional evidence for the involvement of ATM at multiple points in cell cycle regulation.


Assuntos
Ataxia Telangiectasia/patologia , Quinases relacionadas a CDC2 e CDC28 , Reparo do DNA , Ataxia Telangiectasia/metabolismo , Ciclo Celular , Células Cultivadas , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Humanos , Fosfotirosina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fase S/efeitos da radiação
14.
Int J Radiat Biol ; 69(3): 385-95, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8613688

RESUMO

The molecular basis of radiosensitivity was studied using a cDNA complementation approach to correct radiosensitivity in cells. Four cDNAs of sizes 1.6, 2.0, 2.2 and 2.5 kb were isolated that corrected several aspects of the phenotype of cells from patients with the human genetic disorder ataxia-telangiectasia, characterized by hypersensitivity to ionizing radiation. The criteria used to assess correction included cell viability, induced chromosome aberrations, G2 phase delay and induction of p53 after exposure to radiation. One cDNA (2.5 kb) was identified as the complete sequence of the RNA helicase p68, which was capable of correcting radiosensitivity based on two of the above four criteria, with p53 induction post irradiation being partially corrected. The 2.2 kb cDNA was shown to correspond to the complete sequence of arginyl tRNA synthetase and the other two cDNAs were identical to the 3' untranslated regions (UTR) of the transcription factor TFIIS (1.6 kb) and phospholipase A2 (2.0 kb) respectively. Additional transfections with the 3'UTR (198 nucleotides) of p68 RNA helicase and its inverse sequence revealed that the 3'UTR had the same complementation capacity as the full-length cDNA, whereas the inverse construct failed to complement radiosensitivity. These data provide additional support for a novel role for 3'UTRs in the regulation of gene expression.


Assuntos
Teste de Complementação Genética , Proteínas Quinases , RNA Mensageiro/genética , RNA Mensageiro/efeitos da radiação , Tolerância a Radiação/genética , Ataxia Telangiectasia/genética , Linhagem Celular , Aberrações Cromossômicas , RNA Helicases DEAD-box , Dano ao DNA , DNA Complementar/genética , Regulação da Expressão Gênica , Genes p53/efeitos da radiação , Humanos , Proteínas Nucleares/genética , Fenótipo , Biossíntese de Proteínas , RNA Helicases , RNA Nucleotidiltransferases/genética , Transfecção
16.
Oncogene ; 11(4): 609-18, 1995 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-7651723

RESUMO

We have previously demonstrated that cells from patients with ataxia-telangiectasia (A-T) fail to show initial delay at several cell cycle checkpoints post-irradiation. In addition a defect in the induction of p53 by ionizing radiation was evident. We demonstrate here that the radiation signal transduction pathway operating through p53, its target gene WAF1, cyclin-dependent kinases and the retinoblastoma (Rb) protein is defective in A-T cells. The defective p53 induction after ionizing radiation, observed previously in A-T cells, was also reflected at the functional level using p53-DNA binding activity, transactivation and transfection with wild type p53. Correction of the defect at the G1/S checkpoint was observed when wild type p53 was constitutively expressed in A-T cells. Exposure of control cells to radiation gave rise to p53 induction and as a consequence increased expression of WAF1 mRNA and protein, but A-T cells were defective in this response. As expected the WAF1 response in irradiated control cells resulted in an inhibition of cyclin-dependent kinase activity including cyclin E-cdk2, which plays an important role in the transition from G1 to S phase. No inhibition of cyclin-dependent kinase activity was observed in A-T cells correlating with the delayed WAF1 response. On the contrary an enhancement of cyclin-dependent kinase activity was seen in A-T cells post-irradiation. An accumulation of the hypophosphorylated form of Rb protein occurred in irradiated control cells compatible with the G1/S phase delay observed in these cells after exposure to radiation. In unirradiated A-T cells the amount of Rb protein was much higher compared to controls and it was mainly in the hyperphosphorylated (functionally inactive) form. In addition, accumulation of the hypophosphorylated form of Rb in A-T cells post-irradiation was defective, consistent with the lack of cell cycle arrest. Thus the failure of the G1/S checkpoint in A-T cells after exposure to ionizing radiation is consistent with a defective radiation signal transduction pathway operating through p53.


Assuntos
Ataxia Telangiectasia/patologia , Ciclinas/biossíntese , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Núcleo Celular/metabolismo , Cloranfenicol O-Acetiltransferase/biossíntese , Sequência Consenso , Inibidor de Quinase Dependente de Ciclina p21 , Fase G1 , Genes p53 , Humanos , Luciferases/biossíntese , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Protamina Quinase/metabolismo , Proteínas Recombinantes/biossíntese , Proteína do Retinoblastoma/biossíntese , Fase S , Transdução de Sinais , Ativação Transcricional , Transfecção , Proteína Supressora de Tumor p53/metabolismo , beta-Galactosidase/biossíntese
17.
Int J Radiat Biol ; 66(6 Suppl): S151-6, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7530754

RESUMO

Exposure of mammalian cells to ionizing radiation causes a delay in progression through the cycle at several checkpoints. Cells from patients with ataxia-telangiectasia (A-T) ignore these checkpoint controls postirradiation. The tumour suppressor gene product p53 plays a key role at the G1/S checkpoint preventing the progression of cells into S phase. The induction of p53 by radiation is reduced and/or delayed in A-T cells, which appears to account for the failure of delay at the G1/S checkpoint. We have investigated further this defect in radiation signal transduction in A-T. While the p53 response was defective after radiation, agents that interfered with cell cycle progression such as mimosine, aphidicolin and deprivation of serum led to a normal p53 response in A-T cells. None of these agents caused breaks in DNA, as determined by pulse-field gel electrophoresis, in order to elicit the response. Since this pathway is mediated by protein kinases, we investigated the activity of several of these enzymes in control and A-T cells. Ca+2-dependent and -independent protein kinase C activities were increased by radiation to the same extent in the two cell types, a variety of serine/threonine protein kinase activities were approximately the same and anti-tyrosine antibodies failed to reveal any differences in protein phosphorylation between A-T and control cells. It is not evident what is the nature of the defect in signal transduction in A-T cells. However, it is clear that the p53 response is normal in these cells after exposure to some agents and it is mediated through protein kinase C or another serine/threonine kinase.


Assuntos
Ataxia Telangiectasia/patologia , Ataxia Telangiectasia/fisiopatologia , Naftalenos , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Ciclo Celular/fisiologia , Ciclo Celular/efeitos da radiação , Dano ao DNA , Fibroblastos/efeitos da radiação , Humanos , Linfócitos/efeitos da radiação , Compostos Policíclicos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Proteína Supressora de Tumor p53/efeitos da radiação
18.
Radiat Res ; 138(1 Suppl): S130-3, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8146316

RESUMO

Exposure of mammalian cells to ionizing radiation causes delay in normal progress through the cell cycle at a number of different checkpoints. Abnormalities in these checkpoints have been described for ataxia telangiectasia cells after irradiation. In this report we show that these abnormalities occur at different phases in the cell cycle in several ataxia telangiectasia lymphoblastoid cells. Ataxia telangiectasia cells, synchronized in late G1 phase with either mimosine or aphidicolin and exposed to radiation, showed a reduced delay in entering S phase compared to irradiated control cells. Failure to exhibit G1-phase delay in ataxia telangiectasia cells is accompanied by a reduced ability of radiation to activate the product of the tumor suppressor gene p53, a protein involved in G1/S-phase delay. When the progress of irradiated G1-phase cells was followed into the subsequent G2 and G1 phases ataxia telangiectasia cells showed a more pronounced accumulation in G2 phase than control cells. When cells were irradiated in S phase the extent of delay was more evident in G2 phase and ataxia telangiectasia cells were delayed to a greater extent. These results suggest that the lack of initial delay in both G1 and S phases contributes to the radiosensitivity observed in this syndrome.


Assuntos
Ataxia Telangiectasia/patologia , Ciclo Celular/efeitos da radiação , Linhagem Celular Transformada , Expressão Gênica/efeitos da radiação , Genes p53 , Humanos , Radiação Ionizante , Proteína Supressora de Tumor p53/metabolismo
19.
Int J Radiat Biol ; 65(2): 175-84, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7907115

RESUMO

A number of anomalies have been described in the progression of ataxia-telangiectasia (AT) cells through the cell cycle post-irradiation. Some uncertainty still exists as to whether AT cells show increased or reduced division delay after exposure to ionizing radiation. We have attempted to resolve the apparent inconsistencies that exist by investigating the effects of radiation on AT cells at various stages of the cell cycle. Specific labelling of S phase cells with 5-bromodeoxyuridine (BrdU) followed by irradiation caused a prolonged accumulation of these cells in G2/M phase with only 2-7% of AT cells progressing through to G1 24h post-irradiation. In contrast, 23-28% of control cells irradiated in S phase reached G1 by 24 h after irradiation. As observed previously with AT fibroblasts, AT lymphoblastoid cells irradiated in G1 phase did not experience a delay in entering S phase. After progressing through S phase these cells also were delayed in G2/M, but not to the same extent as irradiated S phase cells. On the other hand, when AT cells were irradiated in G2 phase they showed less delay initially in entry to mitosis and the subsequent G1 phase than did irradiated control cells. The overall results demonstrate that AT cells fail to show an initial delay in transitions between the G1/S and G2/M phases of the cell cycle and in progression through these phases post-irradiation, but in the long-term, after passage through S phase, they experience a prolonged delay in G2/M. Since several AT complementation groups are represented in this study, the cell cycle anomalies appear to be universal in AT. These results implicate deficiencies in control of cell cycle progression in the increased radiosensitivity and cancer predisposition in AT.


Assuntos
Ataxia Telangiectasia/genética , Ciclo Celular/efeitos da radiação , Células Cultivadas , Dano ao DNA/efeitos da radiação , Raios gama , Humanos , Técnicas In Vitro
20.
Cancer Chemother Pharmacol ; 33(5): 399-409, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-8306414

RESUMO

The effects on the cell cycle of two biologically active compounds, bistratene A and cycloxazoline, from the marine ascidian Lissoclinum bistratum were studied in HL-60 human leukemia cells using flow cytometry. Both compounds were shown to cause an apparent accumulation of cells in the G2/M phase. This effect was shown to be both time- and dose-dependent. At the longer time points (30 and 48 h after addition of the compounds) polyploidy was apparent. The fate of cells labeled in the S phase with 5-bromo-2'-deoxyuridine (BrdUrd) was analysed using a bivariate BrdUrd/PI (propidium iodide) technique. Bistratene A and cycloxazoline treatment prevented the majority of BrdUrd-labeled cells from progressing through to the G1 phase. Approximately 50% of the cells were delayed at G2/M, and a significant proportion of cells appeared to be polyploid. Light and electron microscopy revealed the presence of multinucleated cells accounting for the apparent polyploidy. The progression of cells out of the G1 phase was also examined by synchronising cells with mimosine and releasing them from mimosine block in the presence of bistratene A. There was no evidence of a block at the G1/S phase transition or through the S phase since DNA synthesis was not inhibited. The mechanism by which these compounds interfere with cytokinesis is presently unknown but, in the case of bistratene A, may be linked to altered phosphorylation of cellular proteins involved in cell-cycle control.


Assuntos
Acetamidas , Antineoplásicos/farmacologia , Éteres Cíclicos/farmacologia , Leucemia/tratamento farmacológico , Toxinas Marinhas/farmacologia , Peptídeos Cíclicos/farmacologia , Piranos , Divisão Celular/efeitos dos fármacos , Citometria de Fluxo , Fluorescência , Fase G2/efeitos dos fármacos , Humanos , Leucemia/patologia , Mitose/efeitos dos fármacos , Compostos de Espiro , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...