Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomolecules ; 14(5)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38785926

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is a deadly consequence of radiation exposure to the esophagus. ESCC arises from esophageal epithelial cells that undergo malignant transformation and features a perturbed squamous cell differentiation program. Understanding the dose- and radiation quality-dependence of the esophageal epithelium response to radiation may provide insights into the ability of radiation to promote ESCC. We have explored factors that may play a role in esophageal epithelial radiosensitivity and their potential relationship to ESCC risk. We have utilized a murine three-dimensional (3D) organoid model that recapitulates the morphology and functions of the stratified squamous epithelium of the esophagus to study persistent dose- and radiation quality-dependent changes. Interestingly, although high-linear energy transfer (LET) Fe ion exposure induced a more intense and persistent alteration of squamous differentiation and 53BP1 DNA damage foci levels as compared to Cs, the MAPK/SAPK stress pathway signaling showed similar altered levels for most phospho-proteins with both radiation qualities. In addition, the lower dose of high-LET exposure also revealed nearly the same degree of morphological changes, even though only ~36% of the cells were predicted to be hit at the lower 0.1 Gy dose, suggesting that a bystander effect may be induced. Although p38 and ERK/MAPK revealed the highest levels following high-LET exposure, the findings reveal that even a low dose (0.1 Gy) of both radiation qualities can elicit a persistent stress signaling response that may critically impact the differentiation gradient of the esophageal epithelium, providing novel insights into the pathogenesis of radiation-induced esophageal injury and early stage esophageal carcinogenesis.


Asunto(s)
Células Epiteliales , Esófago , Organoides , Animales , Organoides/efectos de la radiación , Organoides/patología , Ratones , Esófago/efectos de la radiación , Esófago/patología , Células Epiteliales/efectos de la radiación , Células Epiteliales/patología , Células Epiteliales/metabolismo , Daño del ADN , Carcinoma de Células Escamosas de Esófago/patología , Transferencia Lineal de Energía , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Diferenciación Celular/efectos de la radiación , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Tolerancia a Radiación
2.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054780

RESUMEN

Double-strand breaks (DSB) are one of the most lethal forms of DNA damage that, if left unrepaired, can lead to genomic instability, cellular transformation, and cell death. In this work, we examined how repair of transcription activator-like effector nuclease (TALEN)-induced DNA damage was altered when knocking out, or inhibiting a function of, two DNA repair proteins, XRCC4 and MRE11, respectively. We developed a fluorescent reporter assay that uses TALENs to introduce DSB and detected repair by the presence of GFP fluorescence. We observed repair of TALEN-induced breaks in the XRCC4 knockout cells treated with mirin (a pharmacological inhibitor of MRE11 exonuclease activity), albeit with ~40% reduced efficiency compared to normal cells. Editing in the absence of XRCC4 or MRE11 exonuclease was robust, with little difference between the indel profiles amongst any of the groups. Reviewing the transcriptional profiles of the mirin-treated XRCC4 knockout cells showed 307 uniquely differentially expressed genes, a number far greater than for either of the other cell lines (the HeLa XRCC4 knockout sample had 83 genes, and the mirin-treated HeLa cells had 30 genes uniquely differentially expressed). Pathways unique to the XRCC4 knockout+mirin group included differential expression of p53 downstream pathways, and metabolic pathways indicating cell adaptation for energy regulation and stress response. In conclusion, our study showed that TALEN-induced DSBs are repaired, even when a key DSB repair protein or protein function is not operational, without a change in indel profiles. However, transcriptional profiles indicate the induction of unique cellular responses dependent upon the DNA repair protein(s) hampered.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteína Homóloga de MRE11/genética , RNA-Seq , Nucleasas de los Efectores Tipo Activadores de la Transcripción/toxicidad , ADN/metabolismo , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Proteína Homóloga de MRE11/antagonistas & inhibidores , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo
3.
Int J Radiat Oncol Biol Phys ; 109(5): 1521-1532, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33232771

RESUMEN

PURPOSE: Computed tomographic (CT) scans in adolescents have increased dramatically in recent years. However, the effects of cumulative low-dose exposures on the development of radiation sensitive organs, such as the mammary gland, is unknown. The purpose of this work was to define the effects of dose rate on mammary organ formation during puberty, an especially sensitive window in mammary development. We used a fractionated low-dose x-ray exposure to mimic multiple higher dose CT scans, and we hypothesized that fractionated exposure would have less of an effect on the number of mammary gland defects compared with an acute exposure. METHODS AND MATERIALS: Female mice were subjected to fractionated low-dose x-ray exposure (10 cGy/d for 5 days), acute x-ray exposure (1 × 50 cGy), or sham exposure. As the wide genetic diversity in humans can play a role in a person's response to irradiation, 2 genetically diverse mouse strains differing in radiation sensitivity (BALB/c-sensitive; C57BL/6-resistant) were used to investigate the role of genetic background on the magnitude of the effect. RESULTS: Unexpectedly, our data reveal that multiple low-dose exposures produce greater immune and mammary defects for weeks after exposure compared with controls. The most pronounced defects being increased ductal branching in both strains and a greater percentage of terminal end buds in the BALB/c strain of mice exposed to fractionated radiation compared with sham. Radiation-induced defects near the terminal end bud were also increased in both strains. CONCLUSIONS: The findings suggest that fractionated low-dose exposures are potentially more damaging to organ development compared with an equivalent, single acute exposure and that genetic background is an important parameter modifying the severity of these effects.


Asunto(s)
Fraccionamiento de la Dosis de Radiación , Glándulas Mamarias Animales/efectos de la radiación , Maduración Sexual , Anomalías Inducidas por Radiación/etiología , Factores de Edad , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de la radiación , Femenino , Inmunidad Celular/efectos de la radiación , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Ratones Endogámicos BALB C/genética , Ratones Endogámicos C57BL/genética , Morfogénesis/efectos de la radiación , Exposición a la Radiación/efectos adversos , Traumatismos Experimentales por Radiación/etiología , Tolerancia a Radiación/genética , Tomografía Computarizada por Rayos X/efectos adversos
4.
Life Sci Space Res (Amst) ; 25: 28-41, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32414491

RESUMEN

During space travel astronauts will be exposed to a very low, mixed field of radiation containing different high LET particles of varying energies, over an extended period. Thus, defining how human cells respond to these complex low dose exposures is important in ascertaining risk. In the current study, we have chosen to investigate how low doses of three different ion's at various energies uniquely change the kinetics of three different phospho-proteins. A normal hTERT immortalized fibroblast cell line, 82-6, was exposed to a range of lower doses (0.05-0.5 Gy) of radiation of different qualities and energies (Si 1000 MeV/u, Si 300 MeV/u, Si 173 MeV/u, Si 93 MeV/u, Fe 1000 MeV/u, Fe 600 MeV/u, Fe 300 MeV/u, Ti 300 MeV/u, Ti 326 MeV/u, Ti 386 MeV/u), covering a wide span of LET's. Exposed samples were analyzed for the average intensity of signal as a fold over the geometric mean level of the sham controls. Three phospho-proteins known to localize to DNA DSBs following radiation (γH2AX, pATF2, pSMC1) were studied. The kinetics of their response was quantified by flow cytometery at 2 and 24 h post exposure. These studies reveal unique kinetic patterns based on the ion, energy, fluence and time following exposure. In addition, γH2AX phosphorylation patterns are uniquely different from phospho-proteins known to be primarily phosphorylated by ATM. This latter finding suggests that the activating kinase(s), or the phosphatases deactivating these proteins, exhibit differences in their response to various radiation qualities and/ or doses of exposure. Further studies will be needed to better define what the differing kinetics for the kinases activated by the unique radiation qualities plays in the biological effectiveness of the particle.


Asunto(s)
Iones Pesados , Transferencia Lineal de Energía , Fosfoproteínas/efectos de la radiación , Transducción de Señal/efectos de la radiación , Línea Celular , Daño del ADN/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Fosforilación/efectos de la radiación
5.
Life Sci Space Res (Amst) ; 20: 101-112, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30797427

RESUMEN

There exists a wide degree of genetic variation within the normal human population which includes disease free individuals with heterozygote defects in major DNA repair genes. A lack of understanding of how this genetic variation impacts cellular phenotypes that inform cancer risk post heavy ion exposure poses a major limitation in developing personalized cancer risk assessment astronauts. We initiated a pilot study with Human Mammary Epithelial Cell strains (HMEC) derived from wild type, a p16 silenced derivative of wild type, and various genetic variants that were heterozygote for DNA repair genes; BRCA1, BRCA2 and ATM. Cells strains were exposed to different high and low LET radiation qualities to generate both simple and complex lesions and centrosome aberrations were examined as a surrogate marker of genomic instability and cancer susceptibility post different exposures. Our results indicate that centrosome aberration frequency is higher in the genetic variants under study. The aberration frequency increases with dose, complexity of the lesion generated by different radiation qualities and age of the individual. This increase in genomic instability correlates with elevated check-point activation post radiation exposure. These studies suggest that the influence of individual genetics on cell cycle regulation could modify the degree of early genomic instability in response to complex lesions and potentially define cancer predisposition in response to HZE exposure. These results will have significant implications in estimating cancer susceptibility in genetically variant individuals exposed to HZE particles.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/patología , Mama/patología , Aberraciones Cromosómicas , Radiación Cósmica , Variación Genética , Mama/metabolismo , Mama/efectos de la radiación , Neoplasias de la Mama/genética , Neoplasias de la Mama/radioterapia , Células Cultivadas , Daño del ADN , Femenino , Humanos , Fenotipo , Fosfoproteínas , Proyectos Piloto
6.
Aging (Albany NY) ; 9(3): 665-686, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28245431

RESUMEN

Exposures to various DNA damaging agents can deregulate a wide array of critical mechanisms that maintain genome integrity. It is unclear how these processes are impacted by one's age at the time of exposure and the complexity of the DNA lesion. To clarify this, we employed radiation as a tool to generate simple and complex lesions in normal primary human mammary epithelial cells derived from women of various ages. We hypothesized that genomic instability in the progeny of older cells exposed to complex damages will be exacerbated by age-associated deterioration in function and accentuate age-related cancer predisposition. Centrosome aberrations and changes in stem cell numbers were examined to assess cancer susceptibility. Our data show that the frequency of centrosome aberrations proportionately increases with age following complex damage causing exposures. However, a dose-dependent increase in stem cell numbers was independent of both age and the nature of the insult. Phospho-protein signatures provide mechanistic clues to signaling networks implicated in these effects. Together these studies suggest that complex damage can threaten the genome stability of the stem cell population in older people. Propagation of this instability is subject to influence by the microenvironment and will ultimately define cancer risk in the older population.


Asunto(s)
Envejecimiento/patología , Neoplasias de la Mama/patología , Centrosoma/efectos de la radiación , Células Epiteliales/efectos de la radiación , Células Madre/efectos de la radiación , Adulto , Anciano , Células Cultivadas , Daño del ADN/efectos de la radiación , Susceptibilidad a Enfermedades , Células Epiteliales/patología , Femenino , Inestabilidad Genómica , Humanos , Persona de Mediana Edad , Células Madre/fisiología
7.
Life Sci Space Res (Amst) ; 9: 19-47, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27345199

RESUMEN

Robust predictive models are essential to manage the risk of radiation-induced carcinogenesis. Chronic exposure to cosmic rays in the context of the complex deep space environment may place astronauts at high cancer risk. To estimate this risk, it is critical to understand how radiation-induced cellular stress impacts cell fate decisions and how this in turn alters the risk of carcinogenesis. Exposure to the heavy ion component of cosmic rays triggers a multitude of cellular changes, depending on the rate of exposure, the type of damage incurred and individual susceptibility. Heterogeneity in dose, dose rate, radiation quality, energy and particle flux contribute to the complexity of risk assessment. To unravel the impact of each of these factors, it is critical to identify sensitive biomarkers that can serve as inputs for robust modeling of individual risk of cancer or other long-term health consequences of exposure. Limitations in sensitivity of biomarkers to dose and dose rate, and the complexity of longitudinal monitoring, are some of the factors that increase uncertainties in the output from risk prediction models. Here, we critically evaluate candidate early and late biomarkers of radiation exposure and discuss their usefulness in predicting cell fate decisions. Some of the biomarkers we have reviewed include complex clustered DNA damage, persistent DNA repair foci, reactive oxygen species, chromosome aberrations and inflammation. Other biomarkers discussed, often assayed for at longer points post exposure, include mutations, chromosome aberrations, reactive oxygen species and telomere length changes. We discuss the relationship of biomarkers to different potential cell fates, including proliferation, apoptosis, senescence, and loss of stemness, which can propagate genomic instability and alter tissue composition and the underlying mRNA signatures that contribute to cell fate decisions. Our goal is to highlight factors that are important in choosing biomarkers and to evaluate the potential for biomarkers to inform models of post exposure cancer risk. Because cellular stress response pathways to space radiation and environmental carcinogens share common nodes, biomarker-driven risk models may be broadly applicable for estimating risks for other carcinogens.


Asunto(s)
Biomarcadores/metabolismo , Radiación Cósmica/efectos adversos , Neoplasias Inducidas por Radiación/diagnóstico , Relación Dosis-Respuesta en la Radiación , Estudios de Evaluación como Asunto , Humanos , Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/metabolismo , Medición de Riesgo
8.
Mol Cell ; 61(4): 535-546, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26833090

RESUMEN

XPG is a structure-specific endonuclease required for nucleotide excision repair, and incision-defective XPG mutations cause the skin cancer-prone syndrome xeroderma pigmentosum. Truncating mutations instead cause the neurodevelopmental progeroid disorder Cockayne syndrome, but little is known about how XPG loss results in this devastating disease. We identify XPG as a partner of BRCA1 and BRCA2 in maintaining genomic stability through homologous recombination (HRR). XPG depletion causes DNA double-strand breaks, chromosomal abnormalities, cell-cycle delays, defective HRR, inability to overcome replication fork stalling, and replication stress. XPG directly interacts with BRCA2, RAD51, and PALB2, and XPG depletion reduces their chromatin binding and subsequent RAD51 foci formation. Upstream in HRR, XPG interacts directly with BRCA1. Its depletion causes BRCA1 hyper-phosphorylation and persistent chromatin binding. These unexpected findings establish XPG as an HRR protein with important roles in genome stability and suggest how XPG defects produce severe clinical consequences including cancer and accelerated aging.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Síndrome de Cockayne/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Inestabilidad Genómica , Recombinación Homóloga , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Línea Celular Tumoral , Síndrome de Cockayne/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Genoma Humano , Células HeLa , Humanos , Ratones , Proteínas Nucleares/metabolismo , Fosforilación , Recombinasa Rad51/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
9.
Radiat Res ; 184(1): 105-19, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26114329

RESUMEN

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.


Asunto(s)
Radiación Cósmica , Factor de Transcripción Activador 2/análisis , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Ciclo Celular/efectos de la radiación , Células Cultivadas , Daño del ADN , Fibroblastos/efectos de la radiación , Histonas/análisis , Humanos , Transferencia Lineal de Energía , Nucleosomas/efectos de la radiación
10.
Nucleic Acids Res ; 41(2): 933-42, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23221633

RESUMEN

Cellular damage from ionizing radiation (IR) is in part due to DNA damage and reactive oxygen species, which activate DNA damage response (DDR) and cytokine signaling pathways, including the ataxia telangiectasia mutated (ATM) and transforming growth factor (TGF)ß/Smad pathways. Using classic double-strand breaks (DSBs) markers, we studied the roles of Smad proteins in DDR and the crosstalk between TGFß and ATM pathways. We observed co-localization of phospho-Smad2 (pSmad2) and Smad7 with DSB repair proteins following low and high linear energy transfer (LET) radiation in human fibroblasts and epithelial cells. The decays of both foci were similar to that of γH2AX foci. Irradiation with high LET particles induced pSmad2 and Smad7 foci tracks indicating the particle trajectory through cells. pSmad2 foci were absent in S phase cells, while Smad7 foci were present in all phases of cell cycle. pSmad2 (but not Smad7) foci were completely abolished when ATM was depleted or inactivated. In contrast, a TGFß receptor 1 (TGFßR1) inhibitor abrogated Smad7, but not pSmad2 foci at DSBs sites. In summary, we suggest that Smad2 and Smad7 contribute to IR-induced DSB signaling in an ATM or TGFßR1-dependent manner, respectively.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteína Smad2/metabolismo , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Proteínas Supresoras de Tumor/fisiología , Factor de Transcripción Activador 2 , Proteínas de la Ataxia Telangiectasia Mutada , Línea Celular , Histonas/metabolismo , Humanos , Cinética , Transferencia Lineal de Energía , Fosforilación , Radiación Ionizante , Proteína Smad2/análisis , Proteína smad3/análisis , Proteína smad3/metabolismo , Proteína smad7/análisis
11.
PLoS One ; 7(7): e41249, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22844446

RESUMEN

Proton radiotherapy has gained more favor among oncologists as a treatment option for localized and deep-seated tumors. In addition, protons are a major constituent of the space radiation astronauts receive during space flights. The potential for these exposures to lead to, or enhance cancer risk has not been well studied. Our objective is to study the biological effects of low energy protons on epithelial cells and its propensity to enhance transforming growth factor beta 1 (TGFß1)-mediated epithelial-mesenchymal transition (EMT), a process occurring during tumor progression and critical for invasion and metastasis. Non-transformed mink lung epithelial cells (Mv1Lu) and hTERT- immortalized human esophageal epithelial cells (EPC) were used in this study. EMT was identified by alterations in cell morphology, EMT-related gene expression changes determined using real-time PCR, and EMT changes in specific cellular markers detected by immunostaining and western blotting. Although TGFß1 treatment alone is able to induce EMT in both Mv1Lu and EPC cells, low energy protons (5 MeV) at doses as low as 0.1 Gy can enhance TGFß1 induced EMT. Protons alone can also induce a mild induction of EMT. SD208, a potent TGFß Receptor 1 (TGFßR1) kinase inhibitor, can efficiently block TGFß1/Smad signaling and attenuate EMT induction. We suggest a model for EMT after proton irradiation in normal and cancerous tissue based on our results that showed that low and high doses of protons can sensitize normal human epithelial cells to mesenchymal transition, more prominently in the presence of TGFß1, but also in the absence of TGFß1.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/efectos de la radiación , Transición Epitelial-Mesenquimal/efectos de la radiación , Terapia de Protones , Línea Celular , Relación Dosis-Respuesta en la Radiación , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Esófago/citología , Humanos , Fenotipo , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Proteína Smad2/metabolismo , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/efectos de la radiación
12.
Radiat Oncol ; 7: 96, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22713703

RESUMEN

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.


Asunto(s)
Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN , Transferencia Lineal de Energía , Proteínas Nucleares/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Endonucleasas , Citometría de Flujo , Células HEK293 , Humanos , Cinética , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factores de Tiempo , Proteínas Supresoras de Tumor/metabolismo
13.
J Radiat Res ; 53(1): 51-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22302045

RESUMEN

TGFß is a key modulator of the Epithelial-Mesenchymal Transition (EMT), a process important in cancer progression and metastasis, which leads to the suppression of epithelial genes and expression of mesenchymal proteins. Ionizing radiation was found to specifically induce expression of the TGF-ß1 isoform, which can modulate late post-radiation changes and increase the risk of tumor development and metastasis. Interactions between TGFß induced EMT and DNA damage responses have not been fully elucidated, particularly at low doses and following different radiation quality exposures. Further characterization of the relationship between radiation quality, EMT and cancer development is warranted. We investigated whether space radiation induced TGFß dependent EMT, using hTERT immortalized human esophageal epithelial cells (EPC2-hTERT) and non-transformed mink lung epithelial cells (Mv1Lu). We have observed morphologic and molecular alterations in EPC2 and Mv1Lu cells consistent with EMT after pre-treatment with TGFß1. This effect could be efficiently inhibited in both cell lines by the use of a TGFßRI inhibitor. High-energy silicon or iron nuclei were each able to cause a mild induction of EMT, with the inclusion of TGFß1 inducing a greatly enhanced EMT phenotype even when cells were irradiated with doses as low as 0.1 Gy. A further enhancement of EMT was achieved at a higher dose of 2 Gy. TGFßRI inhibitor was able to reverse the EMT induced by the combination of TGFß1 and radiation. These studies indicate that heavy ions, even at a low dose, may trigger the process of TGFß1-induced EMT, and suggest further studies are needed to determine whether the chronic exposures received in space may potentiate this process in astronauts, leading to an increased risk of cancer.


Asunto(s)
Células Epiteliales/efectos de la radiación , Transición Epitelial-Mesenquimal/efectos de la radiación , Iones Pesados/efectos adversos , Hierro , Silicio , Factor de Crecimiento Transformador beta1/farmacología , Animales , Línea Celular Transformada/efectos de los fármacos , Línea Celular Transformada/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Esófago/citología , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Pulmón/citología , Visón , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pteridinas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Proteína Smad2/metabolismo , Proteína smad7/metabolismo , beta Catenina/metabolismo
14.
J Mol Model ; 18(5): 2163-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21947447

RESUMEN

The channel structure of the Ku protein elegantly reveals the mechanistic basis of sequence-independent DNA-end binding, which is essential to genome integrity after exposure to ionizing radiation or in V(D)J recombination. However, contradicting evidence indicates that this protein is also involved in the regulation of gene expression and in other regulatory processes with intact chromosomes. This computational study predicts that a putative DNA binding domain of this protein, the SAP domain, can form DNA-bound complexes with relatively high affinities (ΔG ≈ -20 kcal mol(-1)). The binding modes are searched by low frequency vibration modes driven by the fully flexible docking method while binding affinities are calculated by the molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) method. We find this well defined 5 kDa domain with a helix-extended loop-helix structure is suitable to form favorable electrostatic and hydrophobic interactions with either the major groove or the minor groove of DNA. The calculation also reveals the sequence specified binding preference which may relate to the observed pause sites when Ku translocates along DNA and the perplex binding of Ku with circular DNA.


Asunto(s)
Antígenos Nucleares/química , Simulación por Computador , Reparación del ADN/genética , Proteínas de Unión al ADN/química , ADN/química , Sitios de Unión , Células Eucariotas , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Autoantígeno Ku , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Electricidad Estática , Termodinámica
15.
Mutat Res ; 716(1-2): 76-83, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21889946

RESUMEN

Cells deficient in ATM (product of the gene that is mutated in ataxia telangiectasia patients) or NBS (product of the gene mutated in the Nijmegen breakage syndrome) show increased yields of both simple and complex chromosomal aberrations after high doses (>0.5Gy) of ionizing radiation (X-rays or γ-rays), however less is known on how these cells respond at low dose. Previously we had shown that the increased chromosome aberrations in ATM and NBS defective lines was due to a significantly larger quadratic dose-response term compared to normal fibroblasts for both simple and complex exchanges. The linear dose-response term for simple exchanges was significantly higher in NBS cells compared to wild type cells, but not for AT cells. However, AT cells have a high background level of exchanges compared to wild type or NBS cells that confounds the understanding of low dose responses. To understand the sensitivity differences for high to low doses, chromosomal aberration analysis was first performed at low dose-rates (0.5Gy/d), and results provided further evidence for the lack of sensitivity for exchanges in AT cells below doses of 1Gy. Normal lung fibroblast cells treated with KU-55933, a specific ATM kinase inhibitor, showed increased numbers of exchanges at a dose of 1Gy and higher, but were similar to wild type cells at 0.5Gy or below. These results were confirmed using siRNA knockdown of ATM. The present study provides evidence that the increased radiation sensitivity of AT cells for chromosomal exchanges found at high dose does not occur at low dose.


Asunto(s)
Ataxia Telangiectasia/genética , Aberraciones Cromosómicas/efectos de la radiación , Tolerancia a Radiación/genética , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Línea Celular , Daño del ADN , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta en la Radiación , Fibroblastos , Rayos gamma , Técnicas de Silenciamiento del Gen , Humanos , Morfolinas/farmacología , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Pironas/farmacología , Proteínas Supresoras de Tumor/genética
16.
Radiat Res ; 174(6): 691-702, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21128792

RESUMEN

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.


Asunto(s)
Daño del ADN , Citometría de Flujo , Hierro , Fosfoproteínas/metabolismo , Rayos X/efectos adversos , Factor de Transcripción Activador 2/metabolismo , Ciclo Celular/efectos de la radiación , Células Cultivadas , Radiación Cósmica , Roturas del ADN de Doble Cadena , Electroforesis en Gel de Campo Pulsado , Histonas/análisis , Humanos , Cinética , Transferencia Lineal de Energía , Fosforilación
17.
Radiat Res ; 171(6): 752-63, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19580482

RESUMEN

We studied the effects of DNA double-strand break (DSB) repair deficiencies on chromosomal aberration frequency using low doses (<1 Gy) of gamma rays and high-energy iron ions (LET = 151 keV/microm). Chromosomal aberrations were measured using the fluorescence whole-chromosome painting technique. The cell lines included fibroblasts deficient in ATM (product of the gene that is mutated in ataxia telangiectasia patients) or NBS (product of the gene mutated in the Nijmegen breakage syndrome) and gliomablastoma cells proficient in or lacking DNA-dependent protein kinase (DNA-PK) activity. The yields of both simple and complex chromosomal aberrations were increased in DSB repair-defective cells compared to normal cells; the increase was more than twofold higher for gamma rays compared to iron nuclei. For gamma-ray-induced aberrations, the ATM- and NBS-defective lines were found to have significantly larger quadratic components compared to normal fibroblasts for both simple and complex aberrations, while the linear dose-response term was significantly higher only for the NBS cells. For simple and complex aberrations induced by iron nuclei, regression models preferred purely linear and quadratic dose responses, respectively, for each cell line studied. RBEs were reduced relative to normal cells for all of the DSB repair-defective lines, with the DNA-PK-deficient cells found to have RBEs near unity. The large increase in the quadratic dose-response terms in the DSB repair-deficient cell lines points to the importance of the functions of ATM and NBS in chromatin modifications to facilitate correct DSB repair and to minimize aberration formation. The differences found between AT and NBS cells at lower doses suggest important questions about the applicability of observations of radiation sensitivity at high doses to low-dose exposures.


Asunto(s)
Aberraciones Cromosómicas/efectos de la radiación , Trastornos por Deficiencias en la Reparación del ADN , Rayos gamma/efectos adversos , Hierro , Dosis de Radiación , Ataxia Telangiectasia/genética , Línea Celular , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Trastornos por Deficiencias en la Reparación del ADN/genética , Proteína Quinasa Activada por ADN/deficiencia , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Transferencia Lineal de Energía , Modelos Lineales , Síndrome de Nijmegen/genética , Análisis de Regresión
18.
Radiat Res ; 170(3): 353-64, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18763865

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , ADN/efectos de la radiación , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Línea Celular , Relación Dosis-Respuesta en la Radiación , Humanos , Fosforilación/efectos de la radiación , Dosis de Radiación
19.
Radiat Res ; 169(2): 214-22, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18220463

RESUMEN

We developed a biochemical kinetics approach to describe the repair of double-strand breaks (DSBs) produced by low-LET radiation by modeling molecular events associated with non-homologous end joining (NHEJ). A system of coupled nonlinear ordinary differential equations describes the induction of DSBs and activation pathways for major NHEJ components including Ku70/80, DNA-PKcs, and the ligase IV-XRCC4 heterodimer. The autophosphorylation of DNA-PKcs and subsequent induction of gamma-H2AX foci observed after ionizing radiation exposure were modeled. A two-step model of regulation of repair by DNA-PKcs was developed with an initial step allowing access of other NHEJ components to breaks and a second step limiting access to ligase IV-XRCC4. Our model assumes that the transition from the first to the second step depends on DSB complexity, with a much slower rate for complex DSBs. The model faithfully reproduced several experimental data sets, including DSB rejoining as measured by pulsed-field gel electrophoresis (PFGE) at 10 min postirradiation or longer and quantification of the induction of gamma-H2AX foci. A process that is independent of DNA-PKcs is required for the model to reproduce experimental data for rejoining before 10 min postirradiation. Predictions are made for the behaviors of NHEJ components at low doses and dose rates, and a steady state is found at dose rates of 0.1 Gy/h or lower.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , ADN/química , ADN/genética , Histonas/química , Modelos Genéticos , Bioquímica/métodos , Simulación por Computador , ADN/efectos de la radiación , Histonas/genética , Histonas/efectos de la radiación , Humanos , Cinética , Modelos Químicos
20.
DNA Repair (Amst) ; 7(1): 108-18, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17919995

RESUMEN

NBS1-deficient cells exhibit pronounced radiosensitivity and defects in chromosome integrity after ionizing radiation (IR) exposure, yet show only a minor defect in DNA double-strand break (DSB) rejoining, leaving an as yet unresolved enigma as to the nature of the radiosensitivity of these cells. To further investigate the relationship between radiosensitivity, DSB repair, and chromosome stability, we have compared cytological and molecular assays of DSB misrejoining and repair in NBS1-defective, wild type, and NBS1-complemented cells after IR damage. Our findings suggest a subtle defect in overall DSB rejoining in NBS1-defective cells and uniquely also reveal reduced ability of NBS1-defective cells to rejoin correct ends of DSBs. In agreement with published results, one of two different NBS1-defective cell lines showed a slight defect in overall rejoining of DSBs compared to its complemented counterpart, whereas another NBS line did not show any difference from wild type cells. Significant defects in the correct rejoining of DSBs compared to their respective controls were observed for both NBS1-defective lines. The defect in DSB rejoining and the increased misrejoining detected at the molecular level were also reflected in higher levels of fragments and translocations, respectively, at the chromosomal level. This work provides both molecular and cytological evidence that NBS1-deficient cells have defects in DSB processing and reveals that these molecular events can be manifest cytologically.


Asunto(s)
Aberraciones Cromosómicas , Daño del ADN , Reparación del ADN/genética , Síndrome de Nijmegen/genética , Línea Celular , Reparación del ADN/efectos de la radiación , Citometría de Flujo , Fase G1 , Humanos , Hibridación Fluorescente in Situ , Rayos Infrarrojos , Metafase , Síndrome de Nijmegen/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA