Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Radiat Res ; 184(1): 105-19, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26114329

RESUMO

During space travel, astronauts are exposed to a wide array of high-linear energy transfer (LET) particles, with differing energies and resulting biological effects. Risk assessment of these exposures carries a large uncertainty predominantly due to the unique track structure of the particle's energy deposition. The complex damage elicited by high charge and energy (HZE) particles results from both lesions along the track core and from energetic electrons, δ rays, generated as a consequence of particle traversal. To better define how cells respond to this complex radiation exposure, a normal hTERT immortalized skin fibroblast cell line was exposed to a defined panel of particles carefully chosen to tease out track structure effects. Phosphorylation kinetics for several key double-strand break (DSB) response proteins (γ-H2AX, pATF2 and pSMC1) were defined after exposure to ten different high-LET radiation qualities and one low-LET radiation (X ray), at two doses (0.5-2 Gy) and time points (2 and 24 h). The results reveal that the lower energy particles (Fe 300, Si 93 and Ti 300 MeV/u), with a narrower track width and higher number and intensity of δ rays, cause the highest degree of persistent damage response. The persistent γ-H2AX signal at lower energies suggests that damage from these exposures are more difficult to resolve, likely due to the greater complexity of the associated DNA lesions. However, different kinetics were observed for the solely ATM-mediated phosphorylations (pATF2 and pSMC1), revealing a shallow induction at early times and a higher level of residual phosphorylation compared to γ-H2AX. The differing phospho-protein profiles exhibited, compared to γ-H2AX, suggests additional functions for these proteins within the cell. The strong correspondence between the predicted curves for energy deposition per nucleosome for each ion/energy combination and the persistent levels of γ-H2AX indicates that the nature of energy distribution defines residual levels of γ-H2AX, an indicator of unrepaired DSBs. Our results suggest that decreasing the energy of a particle results in more complex damage that may increase genomic instability and increase the risk of carcinogenesis.


Assuntos
Radiação Cósmica , Fator 2 Ativador da Transcrição/análise , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Ciclo Celular/efeitos da radiação , Células Cultivadas , Dano ao DNA , Fibroblastos/efeitos da radiação , Histonas/análise , Humanos , Transferência Linear de Energia , Nucleossomos/efeitos da radiação
2.
Radiat Oncol ; 7: 96, 2012 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-22713703

RESUMO

BACKGROUND: Artemis has a defined role in V(D)J recombination and has been implicated in the repair of radiation induced double-strand breaks. However the exact function(s) of Artemis in DNA repair and its preferred substrate(s) in vivo remain undefined. Our previous work suggests that Artemis is important for the repair of complex DNA damage like that inflicted by high Linear Energy Transfer (LET) radiation. To establish the contribution of Artemis in repairing DNA damage caused by various radiation qualities, we evaluated the effect of over-expressing Artemis on cell survival, DNA repair, and cell cycle arrest after exposure to high and low LET radiation. RESULTS: Our data reveal that Artemis over-expression confers marked radioprotection against both types of radiation, although the radioprotective effect was greater following high LET radiation. Inhibitor studies reveal that the radioprotection imparted by Artemis is primarily dependent on DNA-PK activity, and to a lesser extent on ATM kinase activity. Together, these data suggest a DNA-PK dependent role for Artemis in the repair of complex DNA damage. CONCLUSIONS: These findings indicate that Artemis levels significantly influence radiation toxicity in human cells and suggest that Artemis inhibition could be a practical target for adjuvant cancer therapies.


Assuntos
Ciclo Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA , Transferência Linear de Energia , Proteínas Nucleares/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Endonucleases , Citometria de Fluxo , Células HEK293 , Humanos , Cinética , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fatores de Tempo , Proteínas Supressoras de Tumor/metabolismo
3.
Radiat Res ; 174(6): 691-702, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21128792

RESUMO

We developed a mathematical method to analyze flow cytometry data to describe the kinetics of γ-H2AX and pATF2 phosphorylation in normal human fibroblast cells after exposure to various qualities of low-dose radiation. Previously reported flow cytometry kinetics for these DSB repair phospho-proteins revealed that distributions of intensity were highly skewed, severely limiting the detection of differences in the very low-dose range. Distributional analysis revealed significant differences between control and low-dose samples when distributions were compared using the Kolmogorov-Smirnov test. Differences in radiation quality were found in the distribution shapes and when a nonlinear model was used to relate dose and time to the decay of the mean ratio of phospho-protein intensities of irradiated samples to controls. We analyzed cell cycle phase- and radiation quality-dependent characteristic repair times and residual phospho-protein levels with these methods. Characteristic repair times for γ-H2AX were higher after exposure to iron nuclei compared to X rays in G(1) cells and in S/G(2) cells. The RBE in G(1) cells for iron nuclei relative to X rays for γ-H2AX was 2.1 ± 0.6 and 5.0 ± 3.5 at 2 and 24 h after irradiation, respectively. For pATF2, a saturation effect was observed with reduced expression at high doses, especially for iron nuclei, with much slower characteristic repair times (>7 h) compared to X rays. RBEs for pATF2 were 0.7 ± 0.1 and 1.7 ± 0.5 at 2 and 24 h, respectively. Significant differences in γ-H2AX and pATF2 levels when irradiated samples were compared to controls were noted even at the lowest dose analyzed (0.05 Gy). These results show that mathematical models can be applied to flow cytometry data to identify important and subtle differences after exposure to various qualities of low-dose radiation.


Assuntos
Dano ao DNA , Citometria de Fluxo , Ferro , Fosfoproteínas/metabolismo , Raios X/efeitos adversos , Fator 2 Ativador da Transcrição/metabolismo , Ciclo Celular/efeitos da radiação , Células Cultivadas , Radiação Cósmica , Quebras de DNA de Cadeia Dupla , Eletroforese em Gel de Campo Pulsado , Histonas/análise , Humanos , Cinética , Transferência Linear de Energia , Fosforilação
4.
Radiat Res ; 170(3): 353-64, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18763865

RESUMO

To determine whether the physical differences between high- and low-LET radiation are reflected in the biological responses of exposed cells, we detailed phospho-protein profiles of three proteins functional in radiation repair and signal transduction. Detailing gamma-H2AX, pATF2 Ser490/498 and pSMC1 Ser957 kinetics after X-ray and iron-ion exposure also provides a window into understanding the underlying cellular responses. Phosphorylated forms of these proteins have been documented to co-localize at sites of double-strand breaks (DSBs) after low-LET radiation exposures, and two of these phosphorylations, pATF2 and pSMC1, are specifically dependent on ATM. Flow cytometry-based methods were used to quantify total levels of each phospho-protein at various times after irradiation. As expected, we observed a greater induction and persistence in gamma-H2AX after iron-ion (high-LET) exposure compared to X-ray (low-LET) exposure. In contrast, pATF2 and pSMC1 showed markedly lower induction levels after iron-ion exposure compared to equivalent doses of X rays. Quantification of pATF2 and pSMC1 foci revealed fewer cells containing foci and fewer foci per cell after iron-ion compared to X-ray exposure. These findings suggest that ATM responds to DSBs induced by high-LET radiation differently from DSBs induced by low-LET radiation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , DNA/efeitos da radiação , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Linhagem Celular , Relação Dose-Resposta à Radiação , Humanos , Fosforilação/efeitos da radiação , Doses de Radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...