Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 285(1): 1-11, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25655199

RESUMO

The goal of this study was to define pathways regulated by low dose radiation to understand how biological systems respond to subtle perturbations in their environment and prioritize pathways for human health assessment. Using an in vitro 3-D human full thickness skin model, we have examined the temporal response of dermal and epidermal layers to 10 cGy X-ray using transcriptomic, proteomic, phosphoproteomic and metabolomic platforms. Bioinformatics analysis of each dataset independently revealed potential signaling mechanisms affected by low dose radiation, and integrating data shed additional insight into the mechanisms regulating low dose responses in human tissue. We examined direct interactions among datasets (top down approach) and defined several hubs as significant regulators, including transcription factors (YY1, MYC and CREB1), kinases (CDK2, PLK1) and a protease (MMP2). These data indicate a shift in response across time - with an increase in DNA repair, tissue remodeling and repression of cell proliferation acutely (24-72h). Pathway-based integration (bottom up approach) identified common molecular and pathway responses to low dose radiation, including oxidative stress, nitric oxide signaling and transcriptional regulation through the SP1 factor that would not have been identified by the individual data sets. Significant regulation of key downstream metabolites of nitrative stress was measured within these pathways. Among the features identified in our study, the regulation of MMP2 and SP1 was experimentally validated. Our results demonstrate the advantage of data integration to broadly define the pathways and networks that represent the mechanisms by which complex biological systems respond to perturbation.


Assuntos
Fibroblastos/efeitos da radiação , Ensaios de Triagem em Larga Escala , Queratinócitos/efeitos da radiação , Doses de Radiação , Pele/efeitos da radiação , Biologia de Sistemas , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica/efeitos da radiação , Redes Reguladoras de Genes/efeitos da radiação , Genômica , Homeostase , Humanos , Recém-Nascido , Queratinócitos/metabolismo , Queratinócitos/patologia , Masculino , Metabolômica , Estresse Oxidativo/efeitos da radiação , Fosfoproteínas/metabolismo , Mapas de Interação de Proteínas/efeitos da radiação , Proteômica , Transdução de Sinais/efeitos da radiação , Pele/metabolismo , Pele/patologia , Biologia de Sistemas/métodos , Fatores de Tempo
2.
Mutat Res ; 707(1-2): 24-33, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21159317

RESUMO

Epigenetics, including DNA methylation and microRNA (miRNA) expression, could be the missing link in understanding radiation-induced genomic instability (RIGI). This study tests the hypothesis that irradiation induces epigenetic aberrations, which could eventually lead to RIGI, and that the epigenetic aberrations induced by low linear energy transfer (LET) irradiation are different than those induced by high LET irradiations. GM10115 cells were irradiated with low LET X-rays and high LET iron (Fe) ions and evaluated for DNA damage, cell survival and chromosomal instability. The cells were also evaluated for specific locus methylation of nuclear factor-kappa B (NFκB), tumor suppressor in lung cancer 1 (TSLC1) and cadherin 1 (CDH1) gene promoter regions, long interspersed nuclear element 1 (LINE-1) and Alu repeat element methylation, CpG and non-CpG global methylation and miRNA expression levels. Irradiated cells showed increased micronucleus induction and cell killing immediately following exposure, but were chromosomally stable at delayed times post-irradiation. At this same delayed time, alterations in repeat element and global DNA methylation and miRNA expression were observed. Analyses of DNA methylation predominantly showed hypomethylation, however hypermethylation was also observed. We demonstrate that miRNA expression levels can be altered after X-ray irradiation and that these miRNA are involved in chromatin remodeling and DNA methylation. A higher incidence of epigenetic changes was observed after exposure to X-rays than Fe ions even though Fe ions elicited more chromosomal damage and cell killing. This distinction is apparent at miRNA analyses at which only three miRNA involved in two major pathways were altered after high LET irradiations while six miRNA involved in five major pathways were altered after low LET irradiations. This study also shows that the irradiated cells acquire epigenetic changes suggesting that epigenetic aberrations may arise in the cell without initiating chromosomal instability.


Assuntos
Dano ao DNA/efeitos da radiação , Epigênese Genética/efeitos da radiação , Transferência Linear de Energia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Cricetinae , Metilação de DNA/efeitos da radiação , Relação Dose-Resposta à Radiação , Instabilidade Genômica/efeitos da radiação , Humanos , Íons , Ferro , MicroRNAs/efeitos da radiação , Raios X
4.
Mol Cell Biol ; 26(16): 6047-55, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16880516

RESUMO

Ionizing radiation induces delayed genomic instability in human cells, including chromosomal abnormalities and hyperrecombination. Here, we investigate delayed genome instability of cells exposed to UV radiation. We examined homologous recombination-mediated reactivation of a green fluorescent protein (GFP) gene in p53-proficient human cells. We observed an approximately 5-fold enhancement of delayed hyperrecombination (DHR) among cells surviving a low dose of UV-C (5 J/m2), revealed as mixed GFP+/- colonies. UV-B did not induce DHR at an equitoxic (75 J/m2) dose or a higher dose (150 J/m2). UV is known to induce delayed hypermutation associated with increased oxidative stress. We found that hypoxanthine phosphoribosyltransferase (HPRT) mutation frequencies were approximately 5-fold higher in strains derived from GFP+/- (DHR) colonies than in strains in which recombination was directly induced by UV (GFP+ colonies). To determine whether hypermutation was directly caused by hyperrecombination, we analyzed hprt mutation spectra. Large-scale alterations reflecting large deletions and insertions were observed in 25% of GFP+ strains, and most mutants had a single change in HPRT. In striking contrast, all mutations arising in the hypermutable GFP+/- strains were small (1- to 2-base) changes, including substitutions, deletions, and insertions (reminiscent of mutagenesis from oxidative damage), and the majority were compound, with an average of four hprt mutations per mutant. The absence of large hprt deletions in DHR strains indicates that DHR does not cause hypermutation. We propose that UV-induced DHR and hypermutation result from a common source, namely, increased oxidative stress. These two forms of delayed genome instability may collaborate in skin cancer initiation and progression.


Assuntos
Mutagênese/efeitos da radiação , Mutação Puntual/genética , Recombinação Genética/efeitos da radiação , Raios Ultravioleta , Morte Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Éxons/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Hipoxantina Fosforribosiltransferase/genética , Modelos Biológicos , Mutagênese/genética , Recombinação Genética/genética , Células Tumorais Cultivadas
5.
Cancer Res ; 67(3): 1099-104, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17283143

RESUMO

All humans receive some radiation exposure and the risk for radiation-induced cancer at low doses is based on the assumption that there is a linear non-threshold relationship between dose and subsequent effect. Consequently, risk is extrapolated linearly from high radiation doses to very low doses. However, adaptive responses, bystander effects, and death-inducing effect may influence health effects associated with low-dose radiation exposure. Adaptive response is the phenomenon by which cells irradiated with a sublethal radiation dose can become less susceptible to subsequent high-dose radiation exposure. Bystander effects are nontargeted effects observed in cells that were not irradiated but were either in contact with or received soluble signals from irradiated cells. These non-hit bystander cells can exhibit damage typically associated with direct radiation exposure. Death-inducing effect is a phenomenon whereby medium from human-hamster hybrid cells displaying radiation-induced chromosomal instability is toxic to unirradiated parental cells. In this study, we show that human RKO cells do not exhibit adaptive response, bystander effect, or death-inducing effect, as measured by cell killing, or delayed genomic instability in a stably transfected plasmid-based green fluorescent protein assay measuring homologous recombination and delayed mutation/deletion events. However, growth medium conditioned by some chromosomally unstable RKO derivatives induced genomic instability, indicating that these cells can secrete factor(s) that elicit responses in nonirradiated cells. Furthermore, low radiation doses suppressed the induction of delayed genomic instability by a subsequent high dose, indicative of an adaptive response for radiation-induced genomic instability. These results highlight the inherent variability in cellular responses to low-dose radiation exposure and add to the uncertainties associated with evaluating potential hazards at these low doses.


Assuntos
Neoplasias Colorretais/genética , Instabilidade Genômica/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Neoplasias Colorretais/patologia , Neoplasias Colorretais/radioterapia , Relação Dose-Resposta à Radiação , Humanos
6.
Free Radic Biol Med ; 45(12): 1674-81, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18845240

RESUMO

Compounds that can protect cells from the effects of radiation are important for clinical use, in the event of an accidental or terrorist-generated radiation event, and for astronauts traveling in space. One of the major concerns regarding the use of radio-protective agents is that they may protect cells initially, but predispose surviving cells to increased genomic instability later. In this study we used WR-1065, the active metabolite of amifostine, to determine how protection from direct effects of high- and low-LET radiation exposure influences genomic stability. When added 30 min before irradiation and in high concentrations, WR-1065 protected cells from immediate radiation-induced effects as well as from delayed genomic instability. Lower, nontoxic concentrations of WR-1065 did not protect cells from death; however, it was effective in significantly decreasing delayed genomic instability in the progeny of irradiated cells. The observed increase in manganese superoxide dismutase protein levels and activity may provide an explanation for this effect. These results confirm that WR-1065 is protective against both low- and high-LET radiation-induced genomic instability in surviving cells.


Assuntos
Amifostina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Instabilidade Genômica/efeitos dos fármacos , Mercaptoetilaminas/farmacologia , Protetores contra Radiação/farmacologia , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Relação Dose-Resposta à Radiação , Proteínas de Fluorescência Verde/metabolismo , Humanos , Testes para Micronúcleos , Tolerância a Radiação , Superóxido Dismutase/metabolismo , Raios X
7.
Radiat Res ; 170(3): 393-405, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18763867

RESUMO

A summary is provided of presentations and discussions at the NASA Radiation Biomarker Workshop held September 27-28, 2007 at NASA Ames Research Center in Mountain View, CA. Invited speakers were distinguished scientists representing key sectors of the radiation research community. Speakers addressed recent developments in the biomarker and biotechnology fields that may provide new opportunities for health-related assessment of radiation-exposed individuals, including those exposed during long-duration space travel. Topics discussed included the space radiation environment, biomarkers of radiation sensitivity and individual susceptibility, molecular signatures of low-dose responses, multivariate analysis of gene expression, biomarkers in biodefense, biomarkers in radiation oncology, biomarkers and triage after large-scale radiological incidents, integrated and multiple biomarker approaches, advances in whole-genome tiling arrays, advances in mass spectrometry proteomics, radiation biodosimetry for estimation of cancer risk in a rat skin model, and confounding factors. A summary of conclusions is provided at the end of the report.


Assuntos
Bioensaio/métodos , Biomarcadores/análise , Educação , Expressão Gênica/efeitos da radiação , Radiobiologia/métodos , Radiometria/métodos , Animais , Humanos , Doses de Radiação
8.
Mutat Res ; 640(1-2): 74-81, 2008 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-18242642

RESUMO

Radiation induced genomic instability can be perpetuated over time by the transmission of soluble factors. This can occur via cell-to-cell gap junction communication or the secretion/shedding of soluble factors. We have investigated whether our radiation induced chromosomally unstable GM10115 human-hamster hybrid clones secrete factors that can perpetuate the instability phenotype over time. These clones do not have functional gap junctions, but do secrete significant amounts of Interleukin 8 (IL-8) into the culture medium. We then determined whether IL-8 could initiate and or perpetuate genomic instability over time in parental GM10115 cells. Contrary to our hypothesis, IL-8 could induce DNA damage, but was not responsible for the unstable phenotype. Instead it appears that IL-8 secretion provides a pro-survival function in cells that are chromosomally unstable and generally fail to thrive.


Assuntos
Divisão Celular , Sobrevivência Celular , Instabilidade Genômica/efeitos da radiação , Interleucina-8/fisiologia , Animais , Comunicação Celular , Cricetinae , Dano ao DNA , Junções Comunicantes , Humanos , Células Híbridas , Radiação Ionizante , Espécies Reativas de Oxigênio/análise , Transdução de Sinais/efeitos da radiação
9.
Cancer Res ; 66(21): 10377-83, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17079457

RESUMO

Radiation-induced genomic instability (RIGI) manifests as a heritable increased rate of genetic alterations in the progeny of irradiated cells generations after the initial insult. The progeny can show an increased frequency of chromosomal translocations, deletions, mutations, micronuclei, and decreased plating efficiency. What perpetuates RIGI is unclear; however, persistently increased levels of reactive oxygen species (ROS) are frequently associated with genomically unstable clones. Furthermore, addition of free radical scavengers (e.g., DMSO, glycerol, and cationic thiol cysteamine) reduces the incidence of instability after irradiation, implicating a ROS-mediated role in RIGI induction. Because mitochondria are a major natural cellular source of ROS, we tested the hypothesis that mitochondrial dysfunction has a role in maintaining the elevated ROS levels in our irradiated, genetically unstable GM10115 Chinese hamster ovary cells. Amplex Red fluorometry measurements indicate that the relative contribution of uncoupler-sensitive mitochondrial hydrogen peroxide production to total cellular hydrogen peroxide generation is greater in unstable cells. Measurements of mitochondrial DNA levels and cell cytometric fluorescent measurements of Mitotracker Green FM indicate that differences in mitochondrial ROS production are not due to varying mitochondrial levels. However, mitochondrial respiration measured in digitonin-permeabilized cells is impaired in unstable clones. In addition, manganese superoxide dismutase, a major mitochondrial antioxidant enzyme, exhibits increased immunoreactivity but decreased enzyme activity in unstable clones, which along with decreased respiration rates may explain the increased levels of cellular ROS. These studies show that mitochondria from unstable cells are abnormal and likely contribute to the persistent oxidative stress in the unstable clones.


Assuntos
Instabilidade Genômica/efeitos da radiação , Mitocôndrias/fisiologia , Trifosfato de Adenosina/biossíntese , Animais , Células CHO , Cricetinae , Peróxido de Hidrogênio/metabolismo , Espécies Reativas de Oxigênio , Superóxido Dismutase/metabolismo
10.
Curr Biol ; 12(8): 648-53, 2002 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-11967151

RESUMO

Nijmegen breakage syndrome (NBS) is a rare autosomal recessive disorder characterized by microcephaly, immunodeficiency, and predisposition to hematopoietic malignancy. The clinical and cellular phenotypes of NBS substantially overlap those of ataxia-telangiectasia (A-T). NBS is caused by mutation of the NBS1 gene, which encodes a member of the Mre11 complex, a trimeric protein complex also containing Mre11 and Rad50. Several lines of evidence indicate that the ataxia-telangiectasia mutated (ATM) kinase and the Mre11 complex functionally interact. Both NBS and A-T cells exhibit ionizing radiation (IR) sensitivity and defects in the intra S phase checkpoint, resulting in radioresistant DNA synthesis (RDS)-the failure to suppress DNA replication origin firing after IR exposure. NBS1 is phosphorylated by ATM in response to IR, and this event is required for activation of the intra S phase checkpoint (the RDS checkpoint). We derived a murine model of NBS, the Nbs1(DeltaB/DeltaB) mouse. Nbs1(DeltaB/DeltaB) cells are phenotypically identical to those established from NBS patients. The Nbs1(DeltaB) allele was synthetically lethal with ATM deficiency. We propose that the ATM-Mre11 complex DNA damage response pathway is essential and that ATM or the Mre11 complex serves as a nexus to additional components of the pathway.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quebra Cromossômica , Modelos Animais de Doenças , Deleção de Genes , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Southern Blotting , Ciclo Celular , Proteínas de Ciclo Celular/química , Dano ao DNA , Enzimas Reparadoras do DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Epistasia Genética , Fibroblastos , Genes Letais/genética , Humanos , Proteína Homóloga a MRE11 , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutação/genética , Proteínas Nucleares/química , Fenótipo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome , Proteínas Supressoras de Tumor
11.
J Nucl Med ; 48(10): 1683-91, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17873139

RESUMO

Bystander and low-dose-rate effects influence the dose-response relationship in a manner not predicted by current dosimetric methodologies. Radiation-induced bystander effects refer to biologic responses in cells that are not traversed by an ionizing radiation track and, thus, not subject to direct energy deposition; that is, the responses occur in nonirradiated cells. Low-dose-rate hypersensitivity effects have been documented as a reduction in the survival of cells irradiated at dose rates of 0.1-1.0 Gy/h, with total doses ranging from 1.5 to 5 Gy. For humans undergoing external radiotherapy, evidence of bystander events has been observed in the form of abscopal effects, wherein irradiation of one portion of the anatomy affects a portion outside the radiation field, whereas low-dose-rate hypersensitivity has not been described. In this report, the historical literature is briefly reviewed, key experiments are summarized, and current understanding of the factors thought to be involved in the bystander and low-dose-rate effects is conveyed. The mechanisms associated with these events are still being investigated, and questions remain on their impact in radionuclide therapy. Although current findings do not yet sufficiently justify changing traditional dose estimates used to predict the outcomes of radionuclide therapy, it is important to appreciate the potential importance of these effects and to begin revising methods to reflect the emerging empiric and mechanistic knowledge.


Assuntos
Efeito Espectador/fisiologia , Efeito Espectador/efeitos da radiação , Fenômenos Fisiológicos Celulares/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Modelos Biológicos , Neoplasias/fisiopatologia , Neoplasias/radioterapia , Animais , Relação Dose-Resposta à Radiação , Humanos , Neoplasias/patologia , Doses de Radiação
12.
Radiat Res ; 167(4): 465-74, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17388698

RESUMO

RKO36 cells, a subclone of RKO colorectal carcinoma cells that have been stably transfected with the pCMV-EGFP2Xho vector, were grown to confluence and then exposed to either the radioprotector WR-1065, i.e. the active thiol form of amifostine, for 30 min at doses of 40 microM and 4 mM or the cytokine tumor necrosis factor alpha (TNFalpha, TNFA) for 30 min at a concentration of 10 ng/ml and then washed. Total protein was isolated as a function of time up to 32 h after these treatments. Both doses of WR-1065 as well as the concentration of TNFalpha used were effective in elevating intracellular levels of the antioxidant protein SOD2 (also known as MnSOD) at least 15-fold over background levels as determined by Western blot analysis, while measured SOD2 activity was elevated between 5.5- and 6.9-fold. SOD2 reached a maximal level 24 h and 20 h after WR-1065 and TNFalpha treatments, respectively. The antioxidant proteins catalase and glutathione peroxidase (GPX) were also monitored over the 32-h period. In contrast to the robust changes observed in intracellular levels of SOD2 as a function of time after exposure of cells to WR-1065, catalase levels were elevated only 2.6-fold over background as determined by Western blot analysis, while GPX activity was unaffected by WR-1065 exposure. GPX protein levels were extremely low in cells, and analysis of GPX activity using a spectrophotometric method based on the consumption of reduced NADPH also revealed no measurable change as a function of WR-1065 or TNFalpha exposure. RKO36 cells either were irradiated with X rays in the presence of either 40 microM or 4 mM WR-1065 or 10 ng/ml TNFalpha or were irradiated 24 or 20 h later, respectively, when SOD2 protein levels were most elevated. The concentrations and exposure conditions used for WR-1065 and TNFalpha were not cytotoxic and had no effect on plating efficiencies or cell survival compared to untreated controls. No protection or sensitization was observed for cells irradiated in the presence of 40 microM WR-1065 or TNFalpha. Survival was elevated 1.90-fold for cells irradiated in the presence of 4 mM WR-1065. When RKO36 cells were irradiated with 2 Gy 24 h after 40 microM or 4 mM WR-1065 and 20 h after TNFalpha treatments when SOD2 levels were the most increased, survival was elevated 1.42-, 1.48- and 1.36-fold, respectively. This increased survival represents a SOD2-mediated delayed radioprotective effect. SOD2 appears to be an important antioxidant gene whose inducible expression is an important element in adaptive cellular responses in general, and the delayed radioprotective effect in particular. It can be induced by a range of agents including cytoprotective nonprotein thiols such as WR-1065 and pleiotropic cytokines such as TNFalpha.


Assuntos
Sobrevivência Celular/efeitos da radiação , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Mercaptoetilaminas/administração & dosagem , Protetores contra Radiação/administração & dosagem , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/administração & dosagem , Amifostina/administração & dosagem , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Tolerância a Radiação
13.
Mol Cell Biol ; 24(11): 5060-8, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15143196

RESUMO

Exposure to ionizing radiation can result in delayed effects that can be detected in the progeny of an irradiated cell multiple generations after the initial exposure. These effects are described under the rubric of radiation-induced genomic instability and encompass multiple genotoxic endpoints. We have developed a green fluorescence protein (GFP)-based assay and demonstrated that ionizing radiation induces genomic instability in human RKO-derived cells and in human hamster hybrid GM10115 cells, manifested as increased homologous recombination (HR). Up to 10% of cells cultured after irradiation produce mixed GFP(+/-) colonies indicative of delayed HR or, in the case of RKO-derived cells, mutation and deletion. Consistent with prior studies, delayed chromosomal instability correlated with delayed reproductive cell death. In contrast, cells displaying delayed HR showed no evidence of delayed reproductive cell death, and there was no correlation between delayed chromosomal instability and delayed HR, indicating that these forms of genome instability arise by distinct mechanisms. Because delayed hyperrecombination can be induced at doses of ionizing radiation that are not associated with significantly reduced cell viability, these data may have important implications for assessment of radiation risk and understanding the mechanisms of radiation carcinogenesis.


Assuntos
Recombinação Genética/efeitos da radiação , Instabilidade Cromossômica/efeitos da radiação , Humanos , Hibridização in Situ Fluorescente , Projetos de Pesquisa
14.
Mutat Res ; 616(1-2): 159-64, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17134726

RESUMO

Radiation-induced bystander effects refer to those responses occurring in cells that were not subject to energy deposition events following ionizing radiation. These bystander cells may have been neighbors of irradiated cells, or physically separated but subject to soluble secreted signals from irradiated cells. Bystander effects have been observed in vitro and in vivo and for various radiation qualities. In tribute to an old friend and colleague, Anthony V. Carrano, who would have said "well what are the critical questions that should be addressed, and so what?", we review the evidence for non-targeted radiation-induced bystander effects with emphasis on prevailing questions in this rapidly developing research field, and the potential significance of bystander effects in evaluating the detrimental health effects of radiation exposure.


Assuntos
Efeito Espectador , Aberrações Cromossômicas/efeitos da radiação , Radiação Ionizante , Transdução de Sinais , Animais , Cricetinae , Relação Dose-Resposta à Radiação , Instabilidade Genômica , Humanos , Camundongos , Medição de Risco
15.
DNA Repair (Amst) ; 4(9): 958-70, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15996903

RESUMO

The relatively high frequency with which ionizing radiation induces genomic instability suggests that a gene mutation occurring after irradiation is an unlikely cause of the phenotype. To search for mechanism(s) of initiation and perpetuation of this instability phenotype, gene expression profiles of clones exhibiting delayed chromosomal instability were analyzed. Microarray analysis using two pools of isogenic radiation-induced chromosomally unstable clones compared to an irradiated but chromosomally stable clone uncovered a set of 68 differentially expressed genes using two methods of analysis. Unexpectedly, all 68 genes were under-expressed relative to the chromosomally stable reference clone. Further analysis of the candidates placed the differentially expressed genes into pathways implicating differential MAP kinase signaling, ubiquitin/proteasome function, DNA repair, cell cycle control, lipid signaling, nucleotide metabolism, and other potentially disrupted pathways. Validation studies using northern and western blotting, and functional assays concluded that although differences in some of these pathways exist, no single gene or molecular pathway was found to be differentially regulated in all of the chromosomally unstable clones tested. Inferred from these data is that there are multiple potential molecular pathways and/or events that maintain the unstable phenotype, and no single expression pattern is linked to instability in the unstable clones analyzed.


Assuntos
Instabilidade Cromossômica/efeitos da radiação , Cricetinae/genética , Regulação da Expressão Gênica/efeitos da radiação , Expressão Gênica/efeitos da radiação , Animais , Células CHO , Reparo do DNA , Citometria de Fluxo , Amplificação de Genes , Humanos , Análise de Sequência com Séries de Oligonucleotídeos
16.
EXS ; (96): 293-301, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16383023

RESUMO

Ionizing radiation is perhaps the most extensively studied human carcinogen. There have been a number of epidemiological studies on human populations exposed to radiation for medical or occupational reasons, as a result of protracted environmental exposures due to radiation accidents, or after atomic bombings. As a result of these studies exposure to ionizing radiation has been unambiguously linked to cancer causation. While cancer induction is the primary concern and the most important somatic effect of exposure to ionizing radiation, potential health risks do not only involve neoplastic diseases but also somatic mutations that might contribute to birth defects and ocular maladies, and heritable mutations that might impact on disease risks in future generations. Consequantly it is important we understand the long-term health risks associated with exposure to ionizing radiation.


Assuntos
Transformação Celular Neoplásica/efeitos da radiação , Estruturas Celulares/efeitos da radiação , Instabilidade Genômica , Neoplasias Induzidas por Radiação/genética , Neoplasias/genética , Humanos , Radiação Ionizante
18.
Methods Mol Biol ; 314: 43-50, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16673872

RESUMO

A number of ongoing delayed effects have now been described in the progeny of an irradiated cell. These are grouped under the rubric of radiation induced genomic instability. Perhaps the best characterized is the dynamic production of chromosomal rearrangements in some clonally expanded cells surviving irradiation. In this chapter we provide the protocols for irradiation, cell culture, chromosome analysis, and characterization of the status of genomic stability in the context of delayed radiation effects.


Assuntos
Cromossomos Humanos/efeitos da radiação , Instabilidade Genômica , Hibridização in Situ Fluorescente , Radiação Ionizante , Células Cultivadas , Humanos , Coloração e Rotulagem
20.
Cancer Res ; 63(2): 324-8, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12543783

RESUMO

The detrimental effects associated with exposure to ionizing radiation have long been thought to result from the direct targeting of the nucleus leading to DNA damage; however, the emergence of concepts such as radiation-induced genomic instability and bystander effects have challenged this dogma. After cellular exposure to ionizing radiation, we have isolated a number of clones of Chinese hamster-human hybrid GM10115 cells that demonstrate genomic instability as measured by chromosomal destabilization. These clones show dynamic and persistent generation of chromosomal rearrangements multiple generations after the original insult. We hypothesize that these unstable clones maintain this delayed instability phenotype by secreting factors into the culture medium. To test this hypothesis we transferred filtered medium from unstable cells to unirradiated GM10115 cells. No GM10115 cells were able to survive this medium. This phenomenon by which GM10115 cells die when cultured in medium from chromosomally unstable GM10115 clones is the death-inducing effect. Medium transfer experiments indicate that a factor or factors is/are secreted by unstable cells within 8 h of growth in fresh medium and result in cell killing within 24 h. These factors are stable at ambient temperature but do not survive heating or freezing, and are biologically active when diluted with fresh medium. We present the initial description and characterization of the death-inducing effect. This novel epigenetic effect of radiation has implications for radiation risk assessment and for health risks associated with radiation exposure.


Assuntos
Fatores Biológicos/toxicidade , Cromossomos/efeitos da radiação , Animais , Fatores Biológicos/metabolismo , Células CHO , Células Clonais , Cricetinae , Meios de Cultura , Rearranjo Gênico/efeitos da radiação , Humanos , Células Híbridas/efeitos da radiação , Células Híbridas/ultraestrutura , Hibridização in Situ Fluorescente , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA