Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Br J Cancer ; 107(2): 291-9, 2012 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-22713662

RESUMO

BACKGROUND: Most solid tumours contain regions of sub-optimal oxygen concentration (hypoxia). Hypoxic cancer cells are more resistant to radiotherapy and represent the most aggressive fraction of a tumour. It is therefore essential that strategies continue to be developed to target hypoxic cancer cells. Inhibition of the DNA damage response (DDR) might be an effective way of sensitising hypoxic tumour cells to radiotherapy. METHODS: Here, we describe the cellular effects of pharmacological inhibition of the apical DDR kinase ATR (Ataxia Telangiectasia and Rad 3 related) with a highly selective inhibitor, VE-821, in hypoxic conditions and its potential as a radiosensitiser. RESULTS: VE-821 was shown to inhibit ATR-mediated signalling in response to replication arrest induced by severe hypoxia. In these same conditions, VE-821 induced DNA damage and consequently increased Ataxia Telangiectasia Mutated-mediated phosphorylation of H2AX and KAP1. Consistently, ATR inhibition sensitised tumour cell lines to a range of oxygen tensions. Most importantly, VE-821 increased radiation-induced loss of viability in hypoxic conditions. Using this inhibitor we have also demonstrated for the first time a link between ATR and the key regulator of the hypoxic response, HIF-1. HIF-1 stabilisation and transcriptional activity were both decreased in response to ATR inhibition. CONCLUSION: These findings suggest that ATR inhibition represents a novel strategy to target tumour cells in conditions relevant to pathophysiology and enhance the efficacy of radiotherapy.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Hipóxia Celular/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Tolerância a Radiação/efeitos dos fármacos , Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Dano ao DNA , Replicação do DNA/efeitos dos fármacos , Células HCT116 , Células HeLa , Histonas/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Pirazinas/farmacologia , Tolerância a Radiação/genética , Radioterapia/métodos , Proteínas Repressoras/genética , Transdução de Sinais/efeitos dos fármacos , Sulfonas/farmacologia , Proteína 28 com Motivo Tripartido
2.
Science ; 241(4871): 1325-8, 1988 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-3045971

RESUMO

A karyotypic analysis was performed on seven independently derived clones of primary rat embryo cells transformed by the ras oncogene plus the cooperating oncogene myc. The transfected oncogenes were sometimes present in amplified copy number, with heterogeneity in the levels of amplification. Some chromosomal features, such as aberrantly banding regions and double-minute chromosomes, typical of cells carrying amplified genes, were also seen in three of the seven cell lines. Underlying this heterogeneity there was an unexpected finding. All seven lines showed a common integration site for ras on the q arm of rat chromosome 3 (3q12), though some lines also had other sites of integration. In four of the lines integration of ras was accompanied by deletion of the p arm of chromosome 3 or its possible translocation to chromosome 12.


Assuntos
Genes ras , Animais , Mapeamento Cromossômico , Amplificação de Genes , Oncogenes , Ratos , Recombinação Genética , Transformação Genética , Translocação Genética
3.
Oncogene ; 26(48): 6851-62, 2007 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-17471232

RESUMO

Caveolin-1 (Cav-1) is an integral transmembrane protein and a critical component in interactions of integrin receptors with cytoskeleton-associated and signaling molecules. Since integrin-mediated cell adhesion generates signals conferring radiation resistance, we examined the effects of small interfering RNA-mediated knockdown of Cav-1 alone or in combination with beta1-integrin or focal adhesion kinase (FAK) on radiation survival and proliferation of pancreatic carcinoma cell lines. Irradiation induced Cav-1 expression in PATU8902, MiaPaCa2 and Panc1 cell lines. The cell lines showed significant radiosensitization after knockdown of Cav-1, beta1-integrin or FAK and cholesterol depletion by beta-cyclodextrin relative to nonspecific controls. Under knockdown conditions, proliferation of non-irradiated and irradiated cells was significantly attenuated relative to controls. These findings correlated with changes in expression or phosphorylation of Akt, glycogen synthase kinase 3beta, Paxillin, Src, c-Jun N-terminal kinase and mitogen-activated protein kinase. Analysis of DNA microarray data revealed a Cav-1 overexpression in a subset of pancreatic ductal adenocarcinoma samples. The data presented show, for the first time, that disruption of interactions of Cav-1 with beta1-integrin or FAK affects radiation survival and proliferation of pancreatic carcinoma cells and suggest that Cav-1 is critical to these processes. These results indicate that strategies targeting Cav-1 may be useful as an approach to improve conventional therapies, including radiotherapy, for pancreatic cancer.


Assuntos
Caveolina 1/metabolismo , Neoplasias Pancreáticas/radioterapia , Tolerância a Radiação/fisiologia , Western Blotting , Adesão Celular , Ciclo Celular/fisiologia , Ciclo Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias , Imunofluorescência , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Perfilação da Expressão Gênica , Humanos , Integrina beta1/química , Integrina beta1/genética , Integrina beta1/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Pancreáticas/metabolismo , Paxilina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais , Raios X
5.
Cancer Res ; 53(5): 1128-35, 1993 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8439958

RESUMO

Ionizing radiation induces a G2 delay in eukaryotic cells. Since mitotic cyclins are required to trigger the transition from G2 into and through mitosis, we chose to investigate their expression after irradiation in HeLa cells. In normally cycling HeLa cells, both cyclin A and B mRNA and protein levels rise dramatically in G2/M and rapidly fall coincident with the completion of mitosis. The rise of cyclin A mRNA at the S/G2 boundary slightly precedes that of cyclin B mRNA. Although the peaks of expression of each of these molecules overlap, cyclin A mRNA and protein diminish before cyclin B. After irradiation in S, cyclin A mRNA and protein levels rose with the same kinetics as in the controls, but ultimately exceeded the levels seen in the control population. Cyclin A mRNA and protein levels remained high throughout the G2 delay induced by irradiation. In contrast, cyclin B mRNA and protein levels did not rise as the irradiated cells entered G2/M. Only later, before the irradiated cells exited from G2/M, did levels of cyclin B reach the levels seen in the unirradiated controls. The decreased amount of cyclin B mRNA and protein was inversely proportional to the dose of radiation. These data indicate that irradiation that results in a G2 delay appears to block cells at a point after production of cyclin A but before cyclin B can be fully expressed and that cells do not exit from the delay until cyclin B is again expressed. Thus, cyclin A and cyclin B expression respond differentially to radiation, with cyclin A rising at the same time as the control and to even higher levels than that seen in the controls, whereas cyclin B shows a temporal delay in expression.


Assuntos
Ciclinas/efeitos da radiação , Sequência de Aminoácidos , Ciclinas/análise , Ciclinas/genética , Dano ao DNA , Fase G2/efeitos da radiação , Células HeLa/efeitos da radiação , Humanos , Dados de Sequência Molecular , RNA Mensageiro/análise , Raios X
6.
Cancer Res ; 51(19): 5113-7, 1991 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-1833048

RESUMO

After exposure to ionizing radiation, eukaryotic cells undergo a division delay which is reflected by increased time spent in the G2 portion of the cell cycle. Recent information identifies increased levels of mitotic cyclins as key biochemical events initiating mitosis. In HeLa cells cyclin B mRNA and protein levels have been shown to increase in G2 and to decrease after division is completed. Cyclin B protein binds to cdc2, resulting in histone kinase activity which is necessary for the initiation of mitosis. Accordingly, we chose to investigate how cyclin B mRNA and protein levels were perturbed by irradiation in order to gain further understanding of the mechanisms by which ionizing radiation leads to a division delay. Our experiments revealed at least two effects on cyclin B regulation which might contribute to the division delay: (a) when HeLa cells were irradiated in S phase, there was a delay in the accumulation of cyclin B mRNA; (b) when cells were radiated in G2 phase, at a time when mRNA levels were increasing, a division delay was induced which coincided with a markedly lowered level of cyclin B protein despite high levels of the mRNA.


Assuntos
Ciclinas/biossíntese , Fase G2/efeitos da radiação , Células HeLa/metabolismo , Northern Blotting , Divisão Celular/efeitos da radiação , Células HeLa/efeitos da radiação , Humanos , RNA Mensageiro/biossíntese , Radiação Ionizante , Fase S/efeitos da radiação , Fatores de Tempo
7.
Cancer Res ; 50(20): 6575-9, 1990 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-2208118

RESUMO

Rat embryo cells (REC) transformed by the H-ras oncogene plus the cooperating oncogene v-myc are highly resistant to ionizing radiation as compared with the nontransformed parent cells, REC, or immortalized REC. In an attempt to understand the potential mechanism of resistance in these cells, the induction and repair of double strand breaks (dsb) in DNA were measured in a H-ras plus v-myc transformed (3.7) and an immortalized REC (mycREC) line using pulsed field gel electrophoresis. Cells were irradiated in the exponential phase of growth, and the amount of DNA dsb present was quantified by measuring the fraction of DNA activity released from the agarose plugs in which cells were embedded. Similar values of the fraction of DNA activity released were measured for both cell lines at equal X-ray doses, after correction for differences in cell cycle distribution, suggesting a similar induction of DNA dsb per Gy. Repair of DNA dsb measured after exposure to 40 Gy of X-rays was similar in both cell lines and displayed a fast and a slow component. The fast component had a 50% repair time of approximately 12 min, and the slow component, 50% repair time of about 3 h. These results suggest that the relative radioresistance of 3.7 cells is not conferred by a decrease in the amount of DNA dsb induced per Gy per dalton or by alterations in the capacity of the cells to repair DNA dsb. It is hypothesized that alterations in the expression of potentially lethal damage underlie this phenomenon.


Assuntos
Transformação Celular Neoplásica , Dano ao DNA , Reparo do DNA , Genes myc , Genes ras , Tolerância a Radiação , Animais , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Ratos
8.
Cancer Res ; 55(5): 995-8, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7867011

RESUMO

Apoptosis has been hypothesized to be mediated through the induction of free radicals via oxidative pathways. Furthermore, it has been proposed that Bcl-2 acts to inhibit apoptosis induced by a wide variety of stimuli by preventing the production of oxygen-derived free radicals. Since the generation of oxygen free radicals is dependent upon oxygen concentration, this hypothesis would lead to the prediction that the concentration of oxygen should affect the induction of apoptosis. In order to test this prediction, we have examined the induction of apoptosis in T-lymphoma cell lines S49.1 and WEHI 7.1 by dexamethasone and by withdrawal of serum from myc-immortalized fibroblasts in 95% oxygen, atmospheric oxygen (20%), and hypoxic conditions of up to 125-fold less oxygen. Culture in 95% oxygen induced apoptosis in all cells tested, confirming that oxidative damage can lead to apoptosis. However, for one cell line, WEHI 7.1, hypoxia also induced apoptosis. Furthermore, for the other cell lines tested, induction of apoptosis by either dexamethasone or by serum withdrawal was not affected by hypoxia. These results are not consistent with the hypothesis that apoptosis is mediated via oxygen-generated free radical formation.


Assuntos
Apoptose/efeitos dos fármacos , Oxigênio/farmacologia , Espécies Reativas de Oxigênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Dexametasona/farmacologia , Radicais Livres , Cinética , Leucemia Experimental/tratamento farmacológico , Leucemia Experimental/patologia , Linfoma de Células T/tratamento farmacológico , Linfoma de Células T/patologia , Oxigênio/metabolismo , Pressão Parcial , Ratos , Transdução de Sinais/fisiologia , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Timoma/tratamento farmacológico , Timoma/patologia , Células Tumorais Cultivadas/efeitos dos fármacos
9.
Cancer Res ; 57(4): 753-8, 1997 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9044856

RESUMO

Irradiation of tumor cells results in a G2 delay, which has been postulated to allow DNA repair and cell survival. The G2 delay after irradiation is marked in HeLa and other cells by delayed expression of cyclin B1. To test whether this depression of cyclin B1 contributes to the G2 delay, we induced cyclin B1 expression in irradiated HeLa cells using a dexamethasone-inducible promoter. Induction of cyclin B1 after radiation abrogated the G2 delay by approximately doubling the rate at which the cells reentered mitosis, whereas dexamethasone itself had no effect. However, overexpression of cyclin B1 did not eliminate the G2 delay in irradiated cells. In unirradiated cells, overexpression of cyclin B1 had no effect on cell cycle progression. Confirmation that reduction of cyclin B1 levels would prolong G2 was provided using antisense oligonucleotides to cyclin B1. These results demonstrate that cyclin B1 levels control the length of the G2 delay following irradiation in HeLa cells but do not exclude additional mechanisms controlling the mitotic delay after irradiation.


Assuntos
Ciclina B , Ciclinas/metabolismo , Fase G2/efeitos da radiação , Biomarcadores , Ciclina B1 , Ciclinas/genética , Dexametasona/farmacologia , Fase G2/efeitos dos fármacos , Fase G2/genética , Vetores Genéticos , Glucocorticoides/farmacologia , Células HeLa/metabolismo , Células HeLa/efeitos da radiação , Humanos , Mitose , Oligonucleotídeos Antissenso/farmacologia , RNA Mensageiro/metabolismo , Transfecção
10.
Cancer Res ; 58(12): 2639-45, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9635591

RESUMO

Treatment of cells with agents that cause DNA damage often results in a delay in G2. There is convincing evidence showing that inhibition of p34cdc2 kinase activation is involved in the DNA damage-induced G2 delay. In this study, we have demonstrated the existence of an additional pathway, independent of the p34cdc2 kinase activation pathway, that leads to a G2 arrest in etoposide-treated cells. Both the X-ray-induced and the etoposide-induced G2 arrest were associated with inhibition of the p34cdc2 H1 kinase activation pathway as judged by p34cdc2 H1 kinase activity and phosphorylation of cdc25C. Caffeine treatment restored these activities after either of the treatments. However, the etoposide-treated cells did not resume cycling, revealing the presence of an alternative pathway leading to a G2 arrest. To explore the possibility that this additional pathway involved phosphorylation of the MPM-2 epitope that is shared by a large family of mitotic phosphoproteins, we monitored the phosphorylation status of the MPM-2 epitope after DNA damage and after treatment with caffeine. Phosphorylation of the MPM-2 epitope was depressed in both X-ray and etoposide-treated cells, and the depression was reversed by caffeine in both cases. The results indicate that the pathway affecting MPM-2 epitope phosphorylation is involved in the G2 delay caused by DNA damage. However, it is not part of the caffeine-insensitive pathway leading to a G2 block seen in etoposide-treated cells.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Cafeína/farmacologia , Proteínas de Ciclo Celular/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Dano ao DNA/efeitos dos fármacos , Etoposídeo/farmacologia , Fase G2/efeitos dos fármacos , Fosfatases cdc25 , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Epitopos/metabolismo , Fase G2/genética , Fase G2/efeitos da radiação , Células HeLa/citologia , Células HeLa/efeitos dos fármacos , Células HeLa/efeitos da radiação , Humanos , Proteínas de Neoplasias/genética , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
11.
Cancer Res ; 60(23): 6597-600, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11118040

RESUMO

Transformation with ras oncogenes results in increased radiation sur vival in many but not all cells. In addition, prenyltransferase inhibitors which inhibit ras proteins by blocking posttranslational modification radiosensitize cells with oncogenic ras. These findings suggest that oncogenic ras contributes to intrinsic radiation resistance. However, because introduction of ras oncogenes does not increase radiation survival in all cells and because prenyltransferase inhibitors target molecules other than ras, these studies left the conclusion that ras increases the intrinsic radi ation resistance of tumor cells in doubt. Here we show that genetic inactivation of K- or N-ras oncogenes in human tumor cells (DLD-1 and HT1080, respectively) leads to increased radiosensitivity. Reintroduction of the activated N-ras gene into the HT1080 line, having lost its mutant allele, resulted in increased radiation resistance. This study lends further support to the hypothesis that expression of activated ras can contribute to intrinsic radiation resistance in human tumor cells and extends this finding to the K- and N- members of the ras family. These findings support the development of strategies that target ras for inactivation in the treatment of cancer.


Assuntos
Genes ras/fisiologia , Tolerância a Radiação/genética , Células Tumorais Cultivadas/efeitos da radiação , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma/radioterapia , Alelos , Apoptose/efeitos da radiação , Divisão Celular/genética , Sobrevivência Celular/efeitos da radiação , Células Clonais , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/radioterapia , Fibrossarcoma/genética , Fibrossarcoma/patologia , Fibrossarcoma/radioterapia , Regulação Neoplásica da Expressão Gênica , Humanos , Células Tumorais Cultivadas/fisiologia
12.
Cancer Res ; 56(8): 1727-30, 1996 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-8620483

RESUMO

Many tumor cells have a greater resistance to ionizing radiation than their normal counterparts, suggesting that the development of drugs that can reduce that radioresistance would potentiate the efficacy of radiation therapy. Because activated H-ras expression has been shown to markedly increase radiation resistance in some transformed cells, the inactivation of H-ras would then be predicted to radiosensitize these tumor cells, while leaving normal cells unaffected. H-ras depends for activity upon farnesylation, which can be blocked by farnesylation inhibitors, including the compound FTI-277. In keeping with this prediction, inhibition of H-ras processing using FTI-277 resulted in higher levels of apoptosis after irradiation and increased radiosensitivity in H-ras-transformed rat embryo cells but did not affect control cells. These experiments suggest that farnesylation inhibitors may prove clinically useful as radiosensitizers of tumors that depend on ras function.


Assuntos
Alquil e Aril Transferases , Apoptose/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Genes ras , Metionina/análogos & derivados , Radiossensibilizantes/farmacologia , Transferases/antagonistas & inibidores , Animais , Apoptose/efeitos da radiação , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Embrião de Mamíferos , Farnesiltranstransferase , Fibroblastos , Genes myc , Genes ras/efeitos dos fármacos , Cinética , Metionina/farmacologia , Ratos , Transfecção , Neoplasias da Bexiga Urinária/genética
13.
Cancer Res ; 58(8): 1754-61, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9563495

RESUMO

The influence of activated ras oncogenes on the sensitivity of human tumor cells to killing by radiation has been an unresolved question in radiobiology. We have examined this question by measuring the radiation sensitivity of human tumor cell lines with oncogenic mutations in their H- or K-ras genes after treatment with prenyltransferase inhibitors that prevent the posttranslational modification of ras required for its activity. Using two measures of clonogenic survival, we have demonstrated radiosensitization in cell lines with oncogenic H-ras mutations or with oncogenic K-ras mutations when ras processing was inhibited by prenyltransferase inhibitor treatment. In contrast, the inhibition of ras processing in cell lines expressing wild-type ras had no effect on radiation-induced cell death. The prenyltransferase inhibitors themselves inhibited clonogenic survival in some cases, but this inhibition did not correlate with ras mutational status. Although treatment with prenyltransferase inhibitors and radiation resulted in a greater reduction of clonogenicity than either treatment alone in cells with wild-type ras, treatment with both agents had a synergistic effect on cell killing in tumor cells with ras mutations. Our results demonstrate that the inhibition of oncogenic ras activity in human tumor cells can reduce the radiation survival of these cells, suggesting that oncogenic ras can contribute to radiation resistance in human tumors. These results further demonstrate the potential of using prenyltransferase inhibitors in combination with radiotherapy in the treatment of human malignancies.


Assuntos
Genes ras/genética , Prenilação de Proteína/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Tolerância a Radiação/efeitos dos fármacos , Western Blotting , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Metionina/análogos & derivados , Metionina/farmacologia , Mutação , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Radiossensibilizantes/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas
14.
Cancer Res ; 50(1): 97-102, 1990 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-2403420

RESUMO

Resistance of tumors to irradiation or chemotherapeutic agents is thought to be one of the reasons why patients who present with early malignancies may not be cured. Much is now known about the molecular mechanisms that underlie drug resistance, but until recently little was known about genetic contributions to radiation resistance. Some evidence now links oncogenes, particularly the ras family of oncogenes, to radiation resistance but heterogeneity between tumors and cell lines has complicated this analysis. Primary rat embryo cells have been chosen as a model system in which the effects on radiation resistance of the H-ras oncogene could be studied on a uniform genetic background. These cells offer several useful advantages. The cells prior to transformation are diploid, and because they have been in culture only for a few passages prior to transformation with the oncogene it is unlikely that any preexisting mutation affecting radiation response could be present. Additionally, the use of rat embryo cells permitted the study of the effects of a second oncogene on the appearance of the radioresistant phenotype. The results show that the activated H-ras oncogene is associated with radiation resistance in primary rat cells after transformation but that the effect of the oncogene by itself is small. However, the oncogene v-myc, which has no effect on radiation resistance by itself, has a synergistic effect on radiation resistance with H-ras. There appear to be differences in the phenotype of radiation resistance associated with these two forms of transfectants. Thus, radiation resistance seen with H-ras by itself is characterized by a change in the slope of the radiation survival curve at high radiation doses but little or no change within the should region of the radiation survival curve. Radiation resistance seen in H-ras plus v-myc transformants is also characterized by an increase in the slope of the curve at high doses but there is also a large effect within the shoulder region of the radiation survival curve. These studies led to the following conclusions: (a) the radioresistant phenotype is not due to preexisting genetic heterogeneity in the cells prior to transfection; (b) the radiation resistant phenotype of cells transformed by H-ras is seen to a greater degree in cells which also contain the v-myc oncogene; (c) the v-myc oncogene may play an important role in the phenotype of radiation resistance at low doses that is within the range most critical for clinical practice.


Assuntos
Sobrevivência Celular/efeitos da radiação , Genes ras , Oncogenes , Proteínas Oncogênicas de Retroviridae/genética , Animais , Ciclo Celular , Linhagem Celular , Células Cultivadas , Células Clonais , Relação Dose-Resposta à Radiação , Embrião de Mamíferos , Cinética , Proteína Oncogênica p55(v-myc) , Proteínas Tirosina Quinases/genética , Ratos , Ratos Endogâmicos
15.
Cancer Res ; 61(5): 2289-93, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11280800

RESUMO

Many tumors contain extensive regions of hypoxia. Because hypoxic cells are markedly more resistant to killing by radiation, repeated attempts have been made to improve the oxygenation of tumors to enhance radiotherapy. We have studied the oxygenation of tumor xenografts in nude mice after treatment with the farnesyltransferase inhibitor L744,832. Hypoxia was assessed by measuring the binding of the hypoxic cell marker pentafluorinated 2-nitroimidazole. We show that xenografts from two tumor cell lines with mutations in H-ras had markedly improved oxygenation after farnesyltransferase treatment. In contrast, xenografts from two tumors without ras mutations had equivalent hypoxia regardless of treatment. The effect on tumor oxygenation could be detected at 3 days and remained after 7 days of treatment. These results indicate that treatment with farnesyltransferase inhibitors can alter the oxygenation of certain tumors and suggest that such treatment might be useful in the radiosensitization of these tumors.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Metionina/análogos & derivados , Metionina/farmacologia , Neoplasias/metabolismo , Oxigênio/metabolismo , Proteínas ras/biossíntese , Animais , Hipóxia Celular/efeitos dos fármacos , Farnesiltranstransferase , Expressão Gênica , Genes ras/genética , Células HT29/efeitos dos fármacos , Células HT29/enzimologia , Células HT29/metabolismo , Humanos , Camundongos , Camundongos Nus , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/genética
16.
Cancer Res ; 61(10): 4278-82, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11358856

RESUMO

The critical pathways determining the resistance of tumor cells to ionizing radiation are poorly defined. Because the ras oncogene, a gene activated in many human cancers treated with radiotherapy, can induce increased radioresistance, we have asked which Ras effector pathways are significant in conferring a survival advantage to tumor cells. The phosphoinositide-3-kinase (PI3K) inhibitor LY294002 radiosensitized cells bearing mutant ras oncogenes, but the survival of cells with wild-type ras was not affected. Inhibition of the PI3K downstream target p70S6K by rapamycin, the Raf-MEK-MAPK pathway with PD98059, or the Ras-MEK kinase-p38 pathway with SB203580 had no effect on radiation survival in cells with oncogenic ras. Expression of active PI3K in cells with wild-type ras resulted in increased radiation resistance that could be inhibited by LY294002. These experiments have indicated the importance of PI3K in mediating enhanced radioresistance and have implicated PI3K as a potential target for specific radiosensitization of tumors.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Tolerância a Radiação/fisiologia , Proteínas ras/fisiologia , Animais , Cromonas/farmacologia , Indução Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Genes ras , Humanos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/biossíntese , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Células Tumorais Cultivadas , Proteínas ras/antagonistas & inibidores , Proteínas ras/genética
17.
Cancer Res ; 60(7): 2018-24, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10766193

RESUMO

Localization and quantitation of 2-nitroimidazole drug binding in low pO2 tumors is a technique that can allow the assessment of hypoxia as a predictive assay. EF5 [2-(2-nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl) acetamide] is such a drug, and it has been shown to be predictive of radiation response in rodent tumors. Using fluorescence immunohistochemical techniques, we provide data on the presence, distribution, and levels of EF5 binding as a surrogate for hypoxia in human head and neck and uterine cervix squamous cell cancers (SCCs). Six patients with SCC were studied. Four patients had head and neck tumors, and two had uterine cervix cancers. The incubation of fresh tissue cubes in EF3 under hypoxic conditions ("reference binding") demonstrated that all tumors were capable of binding drug, and that this binding varied by a factor of 2.9-fold (174.5-516.1) on an absolute fluorescence scale. In the five patients treated at the lowest drug doses (9 mg/kg), in situ binding was quantitatable. For all six patients, the maximum rate of in situ binding varied by a factor of 6.7 between the lowest and highest binding tumor (24.8-160.3) on an absolute fluorescence scale. In tumors with high binding regions, intratumoral heterogeneity was large, extending from minimal fluorescence (<1%) up to 88.6% of reference binding. In tumors with minimal binding, there was little intratumoral heterogeneity. These studies demonstrate substantial heterogeneity of in situ binding between and within individual squamous cell tumors.


Assuntos
Antineoplásicos/farmacocinética , Carcinoma de Células Escamosas/patologia , Hipóxia Celular , Etanidazol/análogos & derivados , Neoplasias de Cabeça e Pescoço/patologia , Hidrocarbonetos Fluorados/farmacocinética , Neoplasias do Colo do Útero/patologia , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Sítios de Ligação , Carcinoma de Células Escamosas/tratamento farmacológico , Etanidazol/efeitos adversos , Etanidazol/farmacocinética , Etanidazol/uso terapêutico , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Hidrocarbonetos Fluorados/efeitos adversos , Hidrocarbonetos Fluorados/uso terapêutico , Masculino , Pessoa de Meia-Idade , Neoplasias do Colo do Útero/tratamento farmacológico
18.
Eur Heart J Cardiovasc Imaging ; 17(11): 1230-1238, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27550664

RESUMO

Magnetic resonance imaging (MRI) is increasingly being used in cardiology to detect heart disease and guide therapy. It is mooted to be a safer alternative to imaging techniques, such as computed tomography (CT) or coronary angiographic imaging. However, there has recently been an increased interest in the potential long-term health risks of MRI, especially in the light of the controversy resulting from a small number of research studies reporting an increase in DNA damage following exposure, with calls to limit its use and avoid unnecessary examination, according to the precautionary principle. Overall the published data are somewhat limited and inconsistent; the ability of MRI to produce DNA lesions has yet to be robustly demonstrated and future experiments should be carefully designed to optimize sensitivity and benchmarked to validate and assess reproducibility. The majority of the current studies have focussed on the initial induction of DNA damage, and this has led to comparisons between the reported induction of γH2AX and implied double-strand break (DSB) yields produced following MRI with induction by imaging techniques using ionizing radiation. However, γH2AX is not only a marker of classical double-ended DSB, but also a marker of stalled replication forks and in certain circumstances stalled DNA transcription. Additionally, ionizing radiation is efficient at producing complex DNA damage, unique to ionizing radiation, with an associated reduction in repairability. Even if the fields associated with MRI are capable of producing DNA damage, the lesions produced will in general be simple, similar to those produced by endogenous processes. It is therefore inappropriate to try and infer cancer risk by simply comparing the yields of γH2AX foci or DNA lesions potentially produced by MRI to those produced by a given exposure of ionizing radiation, which will generally be more biologically effective and have a greater probability of leading to long-term health effects. As a result, it is important to concentrate on more relevant downstream end points (e.g. chromosome aberration production), along with potential mechanisms by which MRI may lead to DNA lesions. This could potentially involve a perturbation in homeostasis of oxidative stress, modifying the background rate of endogenous DNA damage induction. In summary, what the field needs at the moment is more research and less fear mongering.


Assuntos
Dano ao DNA/efeitos da radiação , Imagem Cinética por Ressonância Magnética/efeitos adversos , Tolerância a Radiação/genética , Radiação Ionizante , Doenças Cardiovasculares/diagnóstico por imagem , Estudos de Avaliação como Assunto , Feminino , Humanos , Imagem Cinética por Ressonância Magnética/métodos , Masculino , Avaliação das Necessidades , Pesquisa Qualitativa , Doses de Radiação , Medição de Risco
19.
Oncogene ; 17(25): 3359-63, 1998 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-9916998

RESUMO

The toxicity of radiation to living tissues was discovered soon after the discovery of radioactivity itself and this toxicity is the basis for cancer therapy with radiation. Although this mode of therapy is often effective, its success is far from assured. One major difficulty in the implementation of radiotherapy is that normal tissues are also sensitive to killing by radiation so that treatment is often limited by the tolerance of normal tissues for radiation. Thus methods that sensitize tumor cells while sparing normal tissues could potentially lead to greater success with radiation as a therapy. Oncogenes are frequently altered in tumors, but are not in normal tissue making them potential targets for altering radiosensitivity and apoptosis in tumors.


Assuntos
Apoptose , Radiossensibilizantes/farmacologia , Animais , Apoptose/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Raios X
20.
Oncogene ; 20(27): 3486-96, 2001 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-11429695

RESUMO

All eukaryotic cells manifest cell cycle delay after exposure to DNA damaging agents. It has been proposed that such cell cycle checkpoints may allow DNA repair but direct evidence of such activity during the radiation-induced G2 delay has been lacking. We report here that cells arrested in G2 by radiation (2-3 Gy) and etoposide incorporate bromodeoxyuridine (BrdU) at discrete foci in the nucleus. We detected G2 cells with CENP-F, a nuclear protein maximally expressed in G2. Caffeine and okadaic acid, both established radiosensitizers, inhibit the incorporation of BrdU in G2 cells. Radioresistant HT29 and OVCAR cells demonstrate BrdU foci formation more frequently during the G2 delay when compared to the more radiosensitive A2780 cell line. The repair foci formed during G2 may be followed through mitosis and observed in daughter cells in G1. Taken together, these observations are consistent with the detection of DNA repair activity during the radiation-induced G2 delay after relatively low doses of radiation.


Assuntos
Ciclo Celular/genética , Dano ao DNA , Reparo do DNA , DNA de Neoplasias/genética , Raios gama , Bromodesoxiuridina , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Centrômero/genética , Proteínas Cromossômicas não Histona/genética , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/efeitos da radiação , Etoposídeo/toxicidade , Feminino , Citometria de Fluxo , Fase G2 , Células HeLa , Humanos , Cinética , Proteínas dos Microfilamentos , Neoplasias Ovarianas , Tolerância a Radiação , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA