Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38741937

RESUMO

Introduction: Missions beyond low Earth orbit (LEO) will expose astronauts to ionizing radiation (IR) in the form of solar energetic particles (SEP) and galactic cosmic rays (GCR) including high atomic number and energy (HZE) nuclei. The gastrointestinal (GI) system is documented to be highly radiosensitive with even relatively low dose IR exposures capable of inducing mucosal lesions and disrupting epithelial barrier function. IR is also an established risk factor for colorectal cancer (CRC) with several studies examining long-term GI effects of SEP/GCR exposure using tumor-prone APC mouse models. Studies of acute short-term effects of modeled space radiation exposures in wildtype mouse models are more limited and necessary to better define charged particle-induced GI pathologies and test novel medical countermeasures (MCMs) to promote astronaut safety. Methods: In this study, we performed ground-based studies where male and female C57BL/6J mice were exposed to γ-rays, 50 MeV protons, or 1 GeV/n Fe-56 ions at the NASA Space Radiation Laboratory (NSRL) with histology and immunohistochemistry endpoints measured in the first 24 h post-irradiation to define immediate SEP/GCR-induced GI alterations. Results: Our data show that unlike matched γ-ray controls, acute exposures to protons and iron ions disrupts intestinal function and induces mucosal lesions, vascular congestion, epithelial barrier breakdown, and marked enlargement of mucosa-associated lymphoid tissue. We also measured kinetics of DNA double-strand break (DSB) repair using gamma-H2AX- specific antibodies and apoptosis via TUNEL labeling, noting the induction and disappearance of extranuclear cytoplasmic DNA marked by gamma-H2AX only in the charged particle-irradiated samples. We show that 18 h pre-treatment with curcumin-loaded nanolipoprotein particles (cNLPs) delivered via IV injection reduces DSB-associated foci levels and apoptosis and restore crypt villi lengths. Discussion: These data improve our understanding of physiological alterations in the GI tract immediately following exposures to modeled space radiations and demonstrates effectiveness of a promising space radiation MCM.

2.
Life Sci Space Res (Amst) ; 28: 22-25, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33612176

RESUMO

Foods packaged for future deep-space exploration missions may be prepositioned ahead of astronaut arrival and will be exposed to galactic cosmic rays (GCRs) and solar radiation in deep space at higher levels and different spectrums than those found in low-Earth orbit (LEO). In this study, we have evaluated the impact of a GCR simulation (approximately 0.5 and 5 Gy doses) at the NASA Space Radiation Laboratory (NSRL) on two retort thermostabilized food products that are good sources of radiation labile nutrients (thiamin, vitamin E, or unsaturated fats). No trends or nutritional differences were found between the radiation-treated samples and the control immediately after treatment or one-year after treatment. Small changes in a few nutrients were measured following one-year of storage. Further studies may be needed to confirm these results, as the foods in this study were heterogeneous, and this may have masked meaningful changes due to pouch-to-pouch variations.


Assuntos
Radiação Cósmica , Alimentos/efeitos da radiação , Gorduras Insaturadas/efeitos da radiação , Análise de Alimentos , Inocuidade dos Alimentos , Armazenamento de Alimentos , Voo Espacial , Tiamina/efeitos da radiação , Vitamina E/efeitos da radiação
3.
PLoS Biol ; 18(5): e3000669, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32428004

RESUMO

With exciting new NASA plans for a sustainable return to the moon, astronauts will once again leave Earth's protective magnetosphere only to endure higher levels of radiation from galactic cosmic radiation (GCR) and the possibility of a large solar particle event (SPE). Gateway, lunar landers, and surface habitats will be designed to protect crew against SPEs with vehicle optimization, storm shelter concepts, and/or active dosimetry; however, the ever penetrating GCR will continue to pose the most significant health risks especially as lunar missions increase in duration and as NASA sets its aspirations on Mars. The primary risks of concern include carcinogenesis, central nervous system (CNS) effects resulting in potential in-mission cognitive or behavioral impairment and/or late neurological disorders, degenerative tissue effects including circulatory and heart disease, as well as potential immune system decrements impacting multiple aspects of crew health. Characterization and mitigation of these risks requires a significant reduction in the large biological uncertainties of chronic (low-dose rate) heavy-ion exposures and the validation of countermeasures in a relevant space environment. Historically, most research on understanding space radiation-induced health risks has been performed using acute exposures of monoenergetic single-ion beams. However, the space radiation environment consists of a wide variety of ion species over a broad energy range. Using the fast beam switching and controls systems technology recently developed at the NASA Space Radiation Laboratory (NSRL) at Brookhaven National Laboratory, a new era in radiobiological research is possible. NASA has developed the "GCR Simulator" to generate a spectrum of ion beams that approximates the primary and secondary GCR field experienced at human organ locations within a deep-space vehicle. The majority of the dose is delivered from protons (approximately 65%-75%) and helium ions (approximately 10%-20%) with heavier ions (Z ≥ 3) contributing the remainder. The GCR simulator exposes state-of-the art cellular and animal model systems to 33 sequential beams including 4 proton energies plus degrader, 4 helium energies plus degrader, and the 5 heavy ions of C, O, Si, Ti, and Fe. A polyethylene degrader system is used with the 100 MeV/n H and He beams to provide a nearly continuous distribution of low-energy particles. A 500 mGy exposure, delivering doses from each of the 33 beams, requires approximately 75 minutes. To more closely simulate the low-dose rates found in space, sequential field exposures can be divided into daily fractions over 2 to 6 weeks, with individual beam fractions as low as 0.1 to 0.2 mGy. In the large beam configuration (60 × 60 cm2), 54 special housing cages can accommodate 2 to 3 mice each for an approximately 75 min duration or 15 individually housed rats. On June 15, 2018, the NSRL made a significant achievement by completing the first operational run using the new GCR simulator. This paper discusses NASA's innovative technology solution for a ground-based GCR simulator at the NSRL to accelerate our understanding and mitigation of health risks faced by astronauts. Ultimately, the GCR simulator will require validation across multiple radiogenic risks, endpoints, doses, and dose rates.


Assuntos
Radiação Cósmica , Radiobiologia/instrumentação , Simulação de Ambiente Espacial , Animais , Humanos , Camundongos , Ratos , Voo Espacial
4.
Cancer Res ; 79(14): 3749-3761, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31088835

RESUMO

Glioblastomas are lethal brain tumors that are treated with conventional radiation (X-rays and gamma rays) or particle radiation (protons and carbon ions). Paradoxically, radiation is also a risk factor for GBM development, raising the possibility that radiotherapy of brain tumors could promote tumor recurrence or trigger secondary gliomas. In this study, we determined whether tumor suppressor losses commonly displayed by patients with GBM confer susceptibility to radiation-induced glioma. Mice with Nestin-Cre-driven deletions of Trp53 and Pten alleles were intracranially irradiated with X-rays or charged particles of increasing atomic number and linear energy transfer (LET). Mice with loss of one allele each of Trp53 and Pten did not develop spontaneous gliomas, but were highly susceptible to radiation-induced gliomagenesis. Tumor development frequency after exposure to high-LET particle radiation was significantly higher compared with X-rays, in accordance with the irreparability of DNA double-strand breaks (DSB) induced by high-LET radiation. All resultant gliomas, regardless of radiation quality, presented histopathologic features of grade IV lesions and harbored populations of cancer stem-like cells with tumor-propagating properties. Furthermore, all tumors displayed concomitant loss of heterozygosity of Trp53 and Pten along with frequent amplification of the Met receptor tyrosine kinase, which conferred a stem cell phenotype to tumor cells. Our results demonstrate that radiation-induced DSBs cooperate with preexisting tumor suppressor losses to generate high-grade gliomas. Moreover, our mouse model can be used for studies on radiation-induced development of GBM and therapeutic strategies. SIGNIFICANCE: This study uncovers mechanisms by which ionizing radiation, especially particle radiation, promote GBM development or recurrence.


Assuntos
Neoplasias Encefálicas/genética , Quebras de DNA de Cadeia Dupla , Glioblastoma/genética , Glioma/genética , Neoplasias Induzidas por Radiação/genética , PTEN Fosfo-Hidrolase/genética , Proteína Supressora de Tumor p53/genética , Animais , Neoplasias Encefálicas/patologia , Feminino , Glioblastoma/patologia , Glioma/patologia , Humanos , Transferência Linear de Energia , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gradação de Tumores , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação
5.
Stem Cells Dev ; 27(18): 1237-1256, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29698131

RESUMO

The stem cell compartment of the hematopoietic system constitutes one of the most radiosensitive tissues of the body and leukemias represent one of the most frequent radiogenic cancers with short latency periods. As such, leukemias may pose a particular threat to astronauts during prolonged space missions. Control of hematopoiesis is tightly governed by a specialized bone marrow (BM) microenvironment/niche. As such, any environmental insult that damages cells of this niche would be expected to produce pronounced effects on the types and functionality of hematopoietic/immune cells generated. We recently reported that direct exposure of human hematopoietic stem cells (HSC) to simulated solar energetic particle (SEP) and galactic cosmic ray (GCR) radiation dramatically altered the differentiative potential of these cells, and that simulated GCR exposures can directly induce DNA damage and mutations within human HSC, which led to leukemic transformation when these cells repopulated murine recipients. In this study, we performed the first in-depth examination to define changes that occur in mesenchymal stem cells present in the human BM niche following exposure to accelerated protons and iron ions and assess the impact these changes have upon human hematopoiesis. Our data provide compelling evidence that simulated SEP/GCR exposures can also contribute to defective hematopoiesis/immunity through so-called "biological bystander effects" by damaging the stromal cells that comprise the human marrow microenvironment, thereby altering their ability to support normal hematopoiesis.


Assuntos
Células da Medula Óssea/efeitos da radiação , Radiação Cósmica/efeitos adversos , Hematopoese/efeitos da radiação , Células-Tronco Mesenquimais/efeitos da radiação , Efeito Espectador , Microambiente Celular/efeitos da radiação , Dano ao DNA/efeitos da radiação , Humanos , Ferro/química , Prótons/efeitos adversos , Energia Solar
6.
Mol Cancer Ther ; 17(4): 858-868, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29437879

RESUMO

Carbon ion therapy (CIT) offers several potential advantages for treating cancers compared with X-ray and proton radiotherapy, including increased biological efficacy and more conformal dosimetry. However, CIT potency has not been characterized in primary tumor animal models. Here, we calculate the relative biological effectiveness (RBE) of carbon ions compared with X-rays in an autochthonous mouse model of soft tissue sarcoma. We used Cre/loxP technology to generate primary sarcomas in KrasLSL-G12D/+; p53fl/fl mice. Primary tumors were irradiated with a single fraction of carbon ions (10 Gy), X-rays (20 Gy, 25 Gy, or 30 Gy), or observed as controls. The RBE was calculated by determining the dose of X-rays that resulted in similar time to posttreatment tumor volume quintupling and exponential growth rate as 10 Gy carbon ions. The median tumor volume quintupling time and exponential growth rate of sarcomas treated with 10 Gy carbon ions and 30 Gy X-rays were similar: 27.3 and 28.1 days and 0.060 and 0.059 mm3/day, respectively. Tumors treated with lower doses of X-rays had faster regrowth. Thus, the RBE of carbon ions in this primary tumor model is 3. When isoeffective treatments of carbon ions and X-rays were compared, we observed significant differences in tumor growth kinetics, proliferative indices, and immune infiltrates. We found that carbon ions were three times as potent as X-rays in this aggressive tumor model and identified unanticipated differences in radiation response that may have clinical implications. Mol Cancer Ther; 17(4); 858-68. ©2018 AACR.


Assuntos
Proliferação de Células , Radioterapia com Íons Pesados , Sarcoma Experimental/radioterapia , Sarcoma/radioterapia , Animais , Apoptose , Relação Dose-Resposta à Radiação , Feminino , Masculino , Camundongos , Radiometria , Eficiência Biológica Relativa , Sarcoma/patologia , Sarcoma Experimental/patologia
7.
Radiat Res ; 186(6): 602-613, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27905868

RESUMO

A significant target for radiation-induced effects is the microvascular system, which is critical to healthy tissue function and its pathology is linked to disrupted endothelial barrier function. Low-linear energy transfer (LET) ionizing radiation is a source of noncancer pathologies in humans and little is known about the early events that could initiate subsequent diseases. However, it is well known that gamma radiation causes a very early disruption of the endothelial barrier at doses below those required for cytotoxic effects. After irradiation of human umbilical vein endothelial cells (HUVECs) to doses as low as 2 Gy, transendothelial electrical resistance (TEER) is transiently reduced at 3 h, and the platelet-derived endothothelial cell adhesion molecule (PECAM-1 or CD31) is uncoupled from the cells along with the release of endothelial microparticles (EMPs). In this study, we measured TEER reduction as an indicator of barrier function loss, and specifically examined the shedding of EMPs from human endothelial barrier models after a variety of low-LET irradiations, including photons and charged particles. Our findings showed two TEER responses, dependent on radiation type and environmental conditions. The first response was diminishing oscillations of TEER, which occurred during the first 10 h postirradiation. This response occurred after a 5 Gy proton or helium-ion (1 GeV/n) dose in addition to a 5 Gy gamma or X radiation dose. This occurred only in the presence of multiple growth factors and did not show a dose response, nor was it associated with EMP release. The second response was a single acute drop in TEER at 3 h after photon irradiation. Dose response was observed and was associated with the shedding of EMPs in 2D barrier cultures and in 3D vessel models. In this case, helium-ion and proton irradiations did not induce a drop in TEER or shedding of EMPs. The photon radiation effects was observed both in serum-free media and in the presence of multiple growth factors, indicating that it occurs under a range of environmental conditions. These results show an acute response of the human endothelial barrier that is relevant to photon irradiation. Significantly, it involves the release of EMPs, which have recently attracted attention due to their emerging clinical importance.


Assuntos
Micropartículas Derivadas de Células/efeitos da radiação , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos da radiação , Transferência Linear de Energia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Micropartículas Derivadas de Células/metabolismo , Relação Dose-Resposta à Radiação , Raios gama , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Transporte Proteico/efeitos da radiação , Prótons , Soro/metabolismo , Fatores de Tempo
8.
Life Sci Space Res (Amst) ; 8: 38-51, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26948012

RESUMO

Most accelerator-based space radiation experiments have been performed with single ion beams at fixed energies. However, the space radiation environment consists of a wide variety of ion species with a continuous range of energies. Due to recent developments in beam switching technology implemented at the NASA Space Radiation Laboratory (NSRL) at Brookhaven National Laboratory (BNL), it is now possible to rapidly switch ion species and energies, allowing for the possibility to more realistically simulate the actual radiation environment found in space. The present paper discusses a variety of issues related to implementation of galactic cosmic ray (GCR) simulation at NSRL, especially for experiments in radiobiology. Advantages and disadvantages of different approaches to developing a GCR simulator are presented. In addition, issues common to both GCR simulation and single beam experiments are compared to issues unique to GCR simulation studies. A set of conclusions is presented as well as a discussion of the technical implementation of GCR simulation.


Assuntos
Radiação Cósmica , Laboratórios , Radiobiologia , Pesquisa , Estados Unidos , United States National Aeronautics and Space Administration
9.
Radiat Res ; 180(2): 156-65, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23819597

RESUMO

The hypothesis that mitochondrial dysfunction and increased superoxide levels in thymocytes over expressing Bax (Lck-Bax1 and Lck-Bax38&1) contributes to lymphomagenesis after low-dose radiation was tested. Lck-Bax1 single-transgenic and Lck-Bax38&1 double-transgenic mice were exposed to single whole-body doses of 10 or 100 cGy of (137)Cs or iron ions (1,000 MeV/n, 150 keV/µm) or silicon ions (300 MeV/n, 67 keV/µm). A 10 cGy dose of (137)Cs significantly increased the incidence and onset of thymic lymphomas in female Lck-Bax1 mice. In Lck-Bax38&1 mice, a 100 cGy dose of high-LET iron ions caused a significant dose dependent acceleration of lymphomagenesis in both males and females that was not seen with silicon ions. To determine the contribution of mitochondrial oxidative metabolism, Lck-Bax38&1 over expressing mice were crossed with knockouts of the mitochondrial protein deacetylase, Sirtuin 3 (Sirt3), which regulates superoxide metabolism. Sirt3(-/-)/Lck-Bax38&1 mice demonstrated significant increases in thymocyte superoxide levels and acceleration of lymphomagenesis (P < 0.001). These results show that lymphomagenesis in Bax over expressing animals is enhanced by radiation exposure in both an LET and gender dependent fashion. These findings support the hypothesis that mitochondrial dysfunction leads to increased superoxide levels and accelerates lymphomagenesis in Lck-Bax transgenic mice.


Assuntos
Íons Pesados/efeitos adversos , Transferência Linear de Energia , Linfoma/etiologia , Mitocôndrias/efeitos da radiação , Neoplasias Induzidas por Radiação/etiologia , Estresse Oxidativo , Caracteres Sexuais , Superóxidos/metabolismo , Neoplasias do Timo/etiologia , Irradiação Corporal Total/efeitos adversos , Proteína X Associada a bcl-2/fisiologia , Animais , Radioisótopos de Césio , Relação Dose-Resposta à Radiação , Feminino , Dosagem de Genes , Ferro , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Linfoma/genética , Linfoma/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mitocôndrias/metabolismo , Neoplasias Induzidas por Radiação/genética , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/fisiopatologia , Fosforilação Oxidativa/efeitos da radiação , Doses de Radiação , Tolerância a Radiação/genética , Proteínas Recombinantes de Fusão/fisiologia , Silício , Sirtuína 3/deficiência , Sirtuína 3/genética , Sirtuína 3/fisiologia , Timócitos/metabolismo , Timócitos/patologia , Timócitos/efeitos da radiação , Neoplasias do Timo/genética , Neoplasias do Timo/fisiopatologia , Proteína X Associada a bcl-2/genética
10.
Cancer Biother Radiopharm ; 26(4): 453-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21790310

RESUMO

Space exploration is associated with exposure to 1-3 Gy solar particle radiation and galactic cosmic radiation that could increase cancer rates. Effective nontoxic countermeasures to high linear energy transfer (LET) radiation exposure are highly desirable but currently not available. The aim was to determine whether a single subcutaneous injection of androstenediol (Δ(5) androsten-3ß, 17ß-diol [AED]) could mitigate and restore the mouse hematopoetic system from the radiation-mediated injury of 3 Gy whole-body high LET (56)Fe(26+) exposure. The findings show that postradiation AED treatment has an overall positive and significant beneficial effect to restore the levels of hematopoeitic elements (p<0.001). Androstenediol treatment significantly increased monocyte levels at days 4, 7, and 14 and, similarly, increased red blood cell, hemoglobin, and platelet counts. Flow cytometry analysis 14 days after radiation and AED treatment demonstrated an increase (p<0.05) in bone marrow cells counts. Ex vivo osteoclastogenesis studies show that AED treatment is necessary and advantageous for the development and restoration of osteoclastogenesis after radiation exposure. These findings clearly show that androstenediol functions as a countermeasure to remedy hematopoeitic injury mediated by high LET iron ion radiation. Presently, no other agent has been shown to have such properties.


Assuntos
Androstenodiol/farmacologia , Íons Pesados/efeitos adversos , Sistema Hematopoético/efeitos dos fármacos , Sistema Hematopoético/efeitos da radiação , Lesões Experimentais por Radiação/etiologia , Lesões Experimentais por Radiação/prevenção & controle , Protetores contra Radiação/farmacologia , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/efeitos da radiação , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/efeitos da radiação , Citometria de Fluxo , Injeções Subcutâneas , Ferro/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/efeitos da radiação , Irradiação Corporal Total
11.
Cell Cycle ; 9(10): 1981-5, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20436300

RESUMO

Checkpoint activation benefits DNA homologous recombination repair and therefore protects cells from ionizing radiation (IR)-induced killing. CHK1 is one of the most important checkpoint regulators in mammalian cells. We recently reported that matrix extracellular phosphoglycoprotein/osteoblast factor 45 (MEPE/OF45) stabilizes CHK1 through interacting with CHK1, thus protecting cells from IR-induced killing. The purpose of this study is to investigate whether a small peptide that mimics the key domain of MEPE/OF45 could interact with CHK1 and protect cells from IR-induced killing. We showed here that the synthesized peptide with 18 amino acids (aa) could enter human transformed lymphoblasts when it is linked to fatty acid CH3(CH2)8CO. After the 18 aa peptide entered the human cells, it interacted with CHK1, increased the CHK1 level and induces stronger G2 arrest in the cells following IR. More importantly, the 18 aa peptide could protect the cells from IR-induced killing. Our data indicate that the 18 aa peptide, similar to MEPE/OF45, reduces CHK1 degradation and protects cells from IR-induced killing. We believe that these results provide useful information for drug development in two directions: protect cells from IR induced damage and sensitize cells to radiation therapy.


Assuntos
Ciclo Celular/efeitos dos fármacos , Proteínas da Matriz Extracelular/química , Glicoproteínas/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Fosfoproteínas/química , Proteínas Quinases/metabolismo , Radiação Ionizante , Animais , Western Blotting , Ciclo Celular/efeitos da radiação , Linhagem Celular Transformada , Quinase 1 do Ponto de Checagem , Fase G2/efeitos dos fármacos , Fase G2/efeitos da radiação , Humanos , Peptídeos/química , Ratos
12.
Radiat Res ; 164(4 Pt 2): 545-51, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16187785

RESUMO

The induction of apoptosis, TP53 expression, caspase activation and cell toxicity were investigated after exposure of cells of the human neuronal progenitor cell line Ntera2 (NT2) to low-LET radiation (gamma and X rays). The data indicates that irradiation of NT2 cells quickly induced TP53 expression, which was followed in time by an increase in caspase activity, and ultimately resulted in the induction of apoptosis. Induction of apoptosis was dependent on dose, and the highest levels were measured 48 h after exposure. For comparison, the level of apoptosis induced by high-LET particle radiation (1 GeV/ nucleon iron ions) was also determined and was found to be dependent on dose. The relative biological effectiveness (RBE) was estimated from the slopes of the dose-response curves for the induction of apoptosis. The RBE(max) for apoptosis 48 h after exposure was at least 3.4. In short, exposure to high-LET radiation results in a more efficient and greater induction of apoptosis in human neuronal progenitor cells than low-LET radiation.


Assuntos
Apoptose/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Genes p53 , Íons Pesados/efeitos adversos , Neurônios/efeitos da radiação , Células-Tronco/efeitos da radiação , Caspase 3 , Caspase 9 , Caspases/metabolismo , Células Cultivadas , Raios gama , Humanos , Ferro , Transferência Linear de Energia
13.
Free Radic Biol Med ; 35(5): 495-503, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12927599

RESUMO

Ionizing radiation induces bistranded DNA damage clusters-two or more oxidized bases, abasic, sites or strand breaks on opposing strands within a few helical turns-but it is not known if clusters are also formed in unirradiated DNA in solution or in unirradiated cultured human cells. The frequencies of endogenous oxidized purine clusters (recognized by Escherichia coli Fpg protein), oxidized pyrimidine clusters (recognized by Nth protein), and abasic clusters (cleavage by Nfo protein) were determined using quantitative gel electrophoresis, electronic imaging, and number average length analysis. Methods of DNA isolation and storage were found to affect cluster levels significantly. In bacteriophage T7 DNA prepared using stringent conditions, the frequencies of these clusters were <1/Mbp. In DNA from unirradiated human 28SC monocytes, the levels of such clusters were, at most, a few per gigabase pair.


Assuntos
Ácido Apurínico/efeitos da radiação , Dano ao DNA , DNA Viral/efeitos da radiação , DNA/efeitos da radiação , Estresse Oxidativo/efeitos da radiação , Purinas/efeitos da radiação , Pirimidinas/efeitos da radiação , Bacteriófago T7/genética , DNA-Formamidopirimidina Glicosilase/metabolismo , Desoxirribonuclease (Dímero de Pirimidina)/metabolismo , Desoxirribonuclease IV (Fago T4-Induzido)/metabolismo , Eletroforese em Gel de Ágar , Escherichia coli/virologia , Proteínas de Escherichia coli/metabolismo , Raios gama , Humanos , Monócitos/metabolismo , Monócitos/efeitos da radiação , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...