Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cancer Sci ; 106(3): 217-26, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25529563

RESUMEN

Genetic, physiological and environmental factors are implicated in colorectal carcinogenesis. Mutations in the mutL homolog 1 (MLH1) gene, one of the DNA mismatch repair genes, are a main cause of hereditary colon cancer syndromes such as Lynch syndrome. Long-term chronic inflammation is also a key risk factor, responsible for colitis-associated colorectal cancer; radiation exposure is also known to increase colorectal cancer risk. Here, we studied the effects of radiation exposure on inflammation-induced colon carcinogenesis in DNA mismatch repair-proficient and repair-deficient mice. Male and female Mlh1(-/-) and Mlh1(+/+) mice were irradiated with 2 Gy X-rays when aged 2 weeks or 7 weeks and/or were treated with 1% dextran sodium sulfate (DSS) in drinking water for 7 days at 10 weeks old to induce mild inflammatory colitis. No colon tumors developed after X-rays and/or DSS treatment in Mlh1(+/+) mice. Colon tumors developed after DSS treatment alone in Mlh1(-/-) mice, and exposure to radiation prior to DSS treatment increased the number of tumors. Histologically, colon tumors in the mice resembled the subtype of well-to-moderately differentiated adenocarcinomas with tumor-infiltrating lymphocytes of human Lynch syndrome. Immunohistochemistry revealed that expression of both p53 and ß-catenin and loss of p21 and adenomatosis polyposis coli proteins were observed at the later stages of carcinogenesis, suggesting a course of molecular pathogenesis distinct from typical sporadic or colitis-associated colon cancer in humans. In conclusion, radiation exposure could further increase the risk of colorectal carcinogenesis induced by inflammation under the conditions of Mlh1 deficiency.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Adenocarcinoma/genética , Carcinogénesis/genética , Neoplasias del Colon/genética , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Adenocarcinoma/inducido químicamente , Proteína de la Poliposis Adenomatosa del Colon/biosíntesis , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Carcinogénesis/inmunología , Carcinogénesis/efectos de la radiación , Colitis/inducido químicamente , Neoplasias del Colon/inducido químicamente , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Reparación de la Incompatibilidad de ADN/genética , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Femenino , Inflamación/inducido químicamente , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Homólogo 1 de la Proteína MutL , Radiación Ionizante , Proteína p53 Supresora de Tumor/biosíntesis , beta Catenina/biosíntesis
2.
J Appl Toxicol ; 35(12): 1594-600, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25772475

RESUMEN

Renal toxicity is a hallmark of uranium exposure, with uranium accumulating specifically in the S3 segment of the proximal tubules causing tubular damage. As the distribution, concentration and dynamics of accumulated uranium at the cellular level is not well understood, here, we report on high-resolution quantitative in situ measurements by high-energy synchrotron radiation X-ray fluorescence analysis in renal sections from a rat model of uranium-induced acute renal toxicity. One day after subcutaneous administration of uranium acetate to male Wistar rats at a dose of 0.5 mg uranium kg(-1) body weight, uranium concentration in the S3 segment of the proximal tubules was 64.9 ± 18.2 µg g(-1) , sevenfold higher than the mean renal uranium concentration (9.7 ± 2.4 µg g(-1) ). Uranium distributed into the epithelium of the S3 segment of the proximal tubules and highly concentrated uranium (50-fold above mean renal concentration) in micro-regions was found near the nuclei. These uranium levels were maintained up to 8 days post-administration, despite more rapid reductions in mean renal concentration. Two weeks after uranium administration, damaged areas were filled with regenerating tubules and morphological signs of tissue recovery, but areas of high uranium concentration (100-fold above mean renal concentration) were still found in the epithelium of regenerating tubules. These data indicate that site-specific accumulation of uranium in micro-regions of the S3 segment of the proximal tubules and retention of uranium in concentrated areas during recovery are characteristics of uranium behavior in the kidney.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Enfermedades Renales/inducido químicamente , Túbulos Renales Proximales/efectos de los fármacos , Uranio/toxicidad , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Etiquetado Corte-Fin in Situ , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/ultraestructura , Masculino , Ratas Wistar , Toxicocinética , Uranio/farmacocinética
3.
Cell Biol Toxicol ; 30(1): 55-66, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24459009

RESUMEN

The radioprotective agent amifostine is a free radical scavenger that can protect cells from the damaging effects of ionising radiation when administered prior to radiation exposure. However, amifostine has also been shown to protect cells from chromosomal mutations when administered after radiation exposure. As apoptosis is a common mechanism by which cells with mutations are removed from the cell population, we investigated whether amifostine stimulates apoptosis when administered after radiation exposure. We chose to study a relatively low dose which is the maximum radiation dose for radiation emergency workers (0.25 Gy) and a high dose relevant to radiotherapy exposures (6 Gy). Mice were administered 400 mg/kg amifostine 30 min before, or 3 h after, whole-body irradiation with 0.25 or 6 Gy X-rays and apoptosis was analysed 3 or 7 h later in spleen and bone marrow. We observed a significant increase in radiation-induced apoptosis in the spleen of mice when amifostine was administered before or after 0.25 Gy X-rays. In contrast, when a high dose of radiation was used (6 Gy), amifostine caused a reduction in radiation-induced apoptosis 3 h post-irradiation in spleen and bone marrow similar to previously published studies. This is the first study to investigate the effect of amifostine on radiation-induced apoptosis at a relatively low radiation dose and the first to demonstrate that while amifostine can reduce apoptosis from high doses of radiation, it does not mediate the same effect in response to low-dose exposures. These results suggest that there may be a dose threshold at which amifostine protects from radiation-induced apoptosis and highlight the importance of examining a range of radiation doses and timepoints.


Asunto(s)
Amifostina/farmacología , Apoptosis/efectos de los fármacos , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Amifostina/administración & dosificación , Animales , Apoptosis/efectos de la radiación , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/patología , Protectores contra Radiación/administración & dosificación , Bazo/efectos de los fármacos , Bazo/patología , Bazo/efectos de la radiación
4.
Toxicol Appl Pharmacol ; 267(3): 266-75, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23337358

RESUMEN

Assessment of risks associated with childhood exposure to ionizing radiation when combined with chemical carcinogens is of great importance. We studied the age-dependence of the effect of combined exposure to ionizing radiation (IR) and a chemical carcinogen on lung carcinogenesis. Female 1-, 5-, and 22-week-old Wistar rats were locally irradiated on the thorax with X-rays (3.18 Gy) and/or were injected intraperitoneally with N-nitrosobis(2-hydroxypropyl)amine (BHP) (1g/kg body weight) 1 week after X-ray exposure or at 23 weeks of age. Rats were terminated at 90 weeks of age. We found that: (i) the incidence of lung tumors (adenoma and adenocarcinoma) increased slightly as a function of age at X-ray exposure, although this was not statistically significant, while the incidence induced by BHP decreased with increasing age at administration; (ii) combined exposure to X-rays at 5 or 22 weeks with BHP 1 week later enhanced the tumor incidence, and the effect at early-life stage (5 weeks irradiation) was more effective than that at late-life stage (22 weeks irradiation); (iii) combined exposure preferentially enhanced malignant transformation; (iv) although a longer interval between the X-ray and BHP treatments reduced the combined effect, risks of early-life irradiation at 1 or 5 weeks of age lasted into adulthood; (v) adenomas and adenocarcinomas induced by X-ray and/or BHP originated from surfactant apoprotein A-positive alveolar type II cells; and (vi), extracellular signal-regulated kinase pathway activation was observed in half the adenocarcinomas, regardless of the exposure schedule. In conclusion, combined exposure may enhance lung tumorigenesis more synergistically at early-life stage (5 weeks of age) than later-life stage.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/efectos de la radiación , Neoplasias Pulmonares/inducido químicamente , Neoplasias Inducidas por Radiación/inducido químicamente , Nitrosaminas/toxicidad , Tórax/efectos de los fármacos , Tórax/efectos de la radiación , Adenocarcinoma/inducido químicamente , Adenocarcinoma/etiología , Adenocarcinoma/patología , Adenoma/inducido químicamente , Adenoma/etiología , Envejecimiento/efectos de los fármacos , Envejecimiento/efectos de la radiación , Animales , Animales Recién Nacidos , Transformación Celular Neoplásica/patología , Femenino , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/patología , Neoplasias Inducidas por Radiación/patología , Ratas , Ratas Wistar , Tórax/patología
5.
J Appl Toxicol ; 33(7): 685-94, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23619997

RESUMEN

Renal toxicity is the principal health concern after uranium exposure. Children are particularly vulnerable to uranium exposure; with contact with depleted uranium in war zones or groundwater contamination the most likely exposure scenarios. To investigate renal sensitivity to uranium exposure during development, we examined uranium distribution and uranium-induced apoptosis in the kidneys of neonate (7-day-old), prepubertal (25-day-old) and adult (70-day-old) male Wistar rats. Mean renal uranium concentrations increased with both age-at-exposure and exposure level after subcutaneous administration of uranium acetate (UA) (0.1-2 mg kg(-1) body weight). Although less of the injected uranium was deposited in the kidneys of the two younger rat groups, the proportion of the peak uranium content remaining in the kidneys after 2 weeks declined with age-at-exposure, suggesting reduced clearance in younger animals. In situ high-energy synchrotron radiation X-ray fluorescence analysis revealed site-specific accumulation of uranium in the S3 segment of the proximal tubules, distributed in the inner cortex and outer stripe of the outer medulla. Apoptosis and cell loss in the proximal tubules increased with age-at-exposure to 0.5 mg kg(-1) UA. Surprisingly, prepubertal rats were uniquely sensitive to uranium-induced lethality from the higher exposure levels. Observations of increased apoptosis in generating/re-generating tubules particularly in prepubertal rats could help to explain their high mortality rate. Together, our findings suggest that age-at-exposure and exposure level are important parameters for uranium toxicity; uranium tends to persist in developing kidneys after low-level exposures, although renal toxicity is more pronounced in adults.


Asunto(s)
Riñón/crecimiento & desarrollo , Compuestos Organometálicos/toxicidad , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Femenino , Glutamato-Amoníaco Ligasa/metabolismo , Etiquetado Corte-Fin in Situ , Riñón/efectos de los fármacos , Riñón/metabolismo , Médula Renal/efectos de los fármacos , Médula Renal/patología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Compuestos Organometálicos/farmacocinética , Embarazo , Ratas , Ratas Wistar , Espectrometría por Rayos X , Sincrotrones , Distribución Tisular , Uranio/análisis , Uranio/metabolismo
6.
Methods Mol Biol ; 2691: 31-41, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37355535

RESUMEN

The development of in vivo lung cancer models that faithfully mimic the human disease is a crucial research tool for understanding the molecular mechanisms driving tumorigenesis. Subcutaneous transplantation assays are commonly employed, likely due to their amenability to easily monitor tumor growth and the simplistic nature of the technique to deliver tumor cells. Importantly however, subcutaneous tumors grow in a microenvironment that differs from that resident within the lung. To circumvent this limitation, here we describe the development of an intrapulmonary (iPUL) orthotopic transplantation method that enables the delivery of lung cancer cells, with precision, to the left lung lobe of recipient mice. Critically, this allows for the growth of lung cancer cells within their native microenvironment. The coupling of iPUL transplantation with position emission tomography (PET) imaging permits the serial detection of tumors in vivo and serves as a powerful tool to trace lung tumor growth and dissemination over time in mouse disease models.


Asunto(s)
Neoplasias Pulmonares , Humanos , Ratones , Animales , Línea Celular Tumoral , Neoplasias Pulmonares/patología , Pulmón/patología , Trasplante de Neoplasias , Carcinogénesis , Modelos Animales de Enfermedad , Microambiente Tumoral
7.
PLoS One ; 18(1): e0280560, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36662808

RESUMEN

Calorie restriction (CR) suppresses not only spontaneous but also chemical- and radiation-induced carcinogenesis. Our previous study revealed that the cancer-preventive effect of CR is tissue dependent and that CR does not effectively prevent the development of thymic lymphoma (TL). We investigated the association between CR and the genomic alterations of resulting TLs to clarify the underlying resistance mechanism. TLs were obtained from previous and new experiments, in which B6C3F1 mice were exposed to radiation at 1 week of age and fed with a CR or standard (non-CR) diet from 7 weeks throughout their lifetimes. All available TLs were used for analysis of genomic DNA. In contrast to the TLs of the non-CR group, those of the CR group displayed suppression of copy-neutral loss of heterozygosity (LOH) involving relevant tumor suppressor genes (Cdkn2a, Ikzf1, Trp53, Pten), an event regarded as cell division-associated. However, CR did not affect interstitial deletions of those genes, which were observed in both groups. In addition, CR affected the mechanism of Ikzf1 inactivation in TLs: the non-CR group exhibited copy-neutral LOH with duplicated inactive alleles, whereas the CR group showed expression of dominant-negative isoforms accompanying a point mutation or an intragenic deletion. These results suggest that, even though CR reduces cell division-related genomic rearrangements by suppressing cell proliferation, tumors arise via diverse carcinogenic pathways including inactivation of tumor suppressors via interstitial deletions and other mutations. These findings provide a molecular basis for improved prevention strategies that overcome the CR resistance of lymphomagenesis.


Asunto(s)
Neoplasias Inducidas por Radiación , Neoplasias del Timo , Ratones , Animales , Restricción Calórica , Mutación , Neoplasias del Timo/genética , Mutación Puntual , Alelos , Pérdida de Heterocigocidad , Neoplasias Inducidas por Radiación/genética
8.
Cancer Discov ; 13(10): 2228-2247, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37548590

RESUMEN

Therapies that enhance antitumor immunity have altered the natural history of many cancers. Consequently, leveraging nonoverlapping mechanisms to increase immunogenicity of cancer cells remains a priority. Using a novel enzymatic inhibitor of the RNA methyl-transferase METTL3, we demonstrate a global decrease in N6-methyladenosine (m6A) results in double-stranded RNA (dsRNA) formation and a profound cell-intrinsic interferon response. Through unbiased CRISPR screens, we establish dsRNA-sensing and interferon signaling are primary mediators that potentiate T-cell killing of cancer cells following METTL3 inhibition. We show in a range of immunocompetent mouse models that although METTL3 inhibition is equally efficacious to anti-PD-1 therapy, the combination has far greater preclinical activity. Using SPLINTR barcoding, we demonstrate that anti-PD-1 therapy and METTL3 inhibition target distinct malignant clones, and the combination of these therapies overcomes clones insensitive to the single agents. These data provide the mole-cular and preclinical rationale for employing METTL3 inhibitors to promote antitumor immunity in the clinic. SIGNIFICANCE: This work demonstrates that METTL3 inhibition stimulates a cell-intrinsic interferon response through dsRNA formation. This immunomodulatory mechanism is distinct from current immunotherapeutic agents and provides the molecular rationale for combination with anti-PD-1 immune-checkpoint blockade to augment antitumor immunity. This article is featured in Selected Articles from This Issue, p. 2109.


Asunto(s)
Interferones , Metiltransferasas , Animales , Ratones , Interferones/genética , Metiltransferasas/genética , Metiltransferasas/metabolismo , ARN Bicatenario
9.
Pharmaceuticals (Basel) ; 15(6)2022 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-35745647

RESUMEN

The gastrin-releasing peptide receptor (GRPR) is a promising molecular target for imaging and therapy of prostate cancer using bombesin peptides that bind to the receptor with high affinity. Targeted copper theranostics (TCTs) using copper radionuclides, 64Cu for imaging and 67Cu for therapy, offer significant advantages in the development of next-generation theranostics. [64Cu]Cu-SAR-BBN is in clinical development for PET imaging of GRPR-expressing cancers. This study explores the therapeutic efficacy of [67Cu]Cu-SAR-BBN in a pre-clinical mouse model. The peptide was radiolabeled with 67Cu, and specific binding of the radiolabeled peptide towards GRPR-positive PC-3 prostate cancer cells was confirmed with 52.2 ± 1.4% total bound compared to 5.8 ± 0.1% with blocking. A therapy study with [67Cu]Cu-SAR-BBN was conducted in mice bearing PC-3 tumors by injecting 24 MBq doses a total of six times. Tumor growth was inhibited by 93.3% compared to the control group on day 19, and median survival increased from 34.5 days for the control group to greater than 54 days for the treatment group. The ease and stability of the radiochemistry, favorable biodistribution, and the positive tumor inhibition demonstrate the suitability of this copper-based theranostic agent for clinical assessment in the treatment of cancers expressing GRPR.

10.
Biology (Basel) ; 11(3)2022 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-35336821

RESUMEN

The risk of radiation-induced carcinogenesis depends on age at exposure. We previously reported principal causative genes in lymphomas arising after infant or adult exposure to 4-fractionated irradiation as Pten or Ikzf1, respectively, suggesting that cells with mutation in these genes might be the origin of lymphomas arising after irradiation depending on age at exposure. Here, we clarified the age-dependent differences in thymus-cell dynamics in mice during the initial post-irradiation period. The thymocyte number initially decreased, followed by two regeneration phases. During the first regeneration, the proportion of phosphorylated-AKT-positive (p-AKT+) cells in cell-cycle phases S+G2/M of immature CD4-CD8- and CD4+CD8+ thymocytes and in phases G0/G1 of mature CD4+CD8- and CD4-CD8+ thymocytes was significantly greater in irradiated infants than in irradiated adults. During the second regeneration, the proportion of p-AKT+ thymocytes in phases G0/G1 increased in each of the three populations other than CD4-CD8- thymocytes more so than during the first regeneration. Finally, PI3K-AKT-mTOR signaling in infants contributed, at least in part, to biphasic thymic regeneration through the modification of cell proliferation and survival after irradiation, which may be associated with the risk of Pten mutation-associated thymic lymphoma.

11.
Front Oncol ; 11: 580806, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34026597

RESUMEN

BACKGROUND: Muscle wasting (Sarcopenia) is associated with poor outcomes in cancer patients. Early identification of sarcopenia can facilitate nutritional and exercise intervention. Cross-sectional skeletal muscle (SM) area at the third lumbar vertebra (L3) slice of a computed tomography (CT) image is increasingly used to assess body composition and calculate SM index (SMI), a validated surrogate marker for sarcopenia in cancer. Manual segmentation of SM requires multiple steps, which limits use in routine clinical practice. This project aims to develop an automatic method to segment L3 muscle in CT scans. METHODS: Attenuation correction CTs from full body PET-CT scans from patients enrolled in two prospective trials were used. The training set consisted of 66 non-small cell lung cancer (NSCLC) patients who underwent curative intent radiotherapy. An additional 42 NSCLC patients prescribed curative intent chemo-radiotherapy from a second trial were used for testing. Each patient had multiple CT scans taken at different time points prior to and post- treatment (147 CTs in the training and validation set and 116 CTs in the independent testing set). Skeletal muscle at L3 vertebra was manually segmented by two observers, according to the Alberta protocol to serve as ground truth labels. This included 40 images segmented by both observers to measure inter-observer variation. An ensemble of 2.5D fully convolutional neural networks (U-Nets) was used to perform the segmentation. The final layer of U-Net produced the binary classification of the pixels into muscle and non-muscle area. The model performance was calculated using Dice score and absolute percentage error (APE) in skeletal muscle area between manual and automated contours. RESULTS: We trained five 2.5D U-Nets using 5-fold cross validation and used them to predict the contours in the testing set. The model achieved a mean Dice score of 0.92 and an APE of 3.1% on the independent testing set. This was similar to inter-observer variation of 0.96 and 2.9% for mean Dice and APE respectively. We further quantified the performance of sarcopenia classification using computer generated skeletal muscle area. To meet a clinical diagnosis of sarcopenia based on Alberta protocol the model achieved a sensitivity of 84% and a specificity of 95%. CONCLUSIONS: This work demonstrates an automated method for accurate and reproducible segmentation of skeletal muscle area at L3. This is an efficient tool for large scale or routine computation of skeletal muscle area in cancer patients which may have applications on low quality CTs acquired as part of PET/CT studies for staging and surveillance of patients with cancer.

12.
J Nucl Med ; 62(6): 829-832, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33067341

RESUMEN

Radionuclide therapy targeting prostate-specific membrane antigen (PSMA) is promising for prostate cancer. We previously reported a ligand, 64Cu-CuSarbisPSMA, featuring 2 lysine-ureido-glutamate groups. Here, we report the therapeutic potential of 67Cu-CuSarbisPSMA. Methods: Growth of PSMA-positive xenografts was evaluated after treatment with 67Cu-CuSarbisPSMA or 177Lu-LuPSMA imaging and therapy (I&T). Results: At 13 d after injection, tumor growth was similarly inhibited by the 2 tracers in a dose-dependent manner. Survival was comparable after single (30 MBq) or fractionated (2 × 15 MBq, 2 wk apart) administrations. Conclusion:67Cu-CuSarbisPSMA is efficacious in a PSMA-expressing model of prostate cancer.


Asunto(s)
Antígenos de Superficie/química , Radioisótopos de Cobre/química , Glutamato Carboxipeptidasa II/química , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Marcaje Isotópico , Masculino , Neoplasias de la Próstata/patología
13.
Int J Radiat Biol ; 95(10): 1372-1377, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31145030

RESUMEN

Purpose: Projects evaluating the effects of radiation, within the National Institutes of Quantum and Radiological Science and Technology (QST), National Institute of Radiological Sciences (NIRS), have focused on risk analyses for life shortening and cancer prevalence using laboratory animals. Genetic and epigenetic alterations in radiation-induced tumors have been also analyzed with the aim of better understanding mechanisms of radiation carcinogenesis. As well as the economic and practical limitations of repeating such large-scale experiments, ethical considerations make it vital that we store and share the pathological data and samples of the animal experiments for future use. We are now constructing such an archive called the Japan-Storehouse of Animal Radiobiology Experiments (J-SHARE). Methods: J-SHARE records include information such as detailed experimental protocols, necropsy records and photographs of organs at necropsy. For each animal organs and tumor tissues are dissected, and parts are stored as frozen samples at -80 °C. Samples fixed with formalin are also embedded in paraffin blocks for histopathological analyses. Digital copies of stained tissues are being systematically saved using a virtual slide system linked to original records by barcodes. Embedded and frozen tissues are available for molecular analysis. Conclusion: Similar archive systems for radiation biology have also been under construction in the USA and Europe, the Northwestern University Radiation Archive (NURA), and STORE at the BfS, respectively. The J-SHARE will be linked with the sister-archives and made available for collaborative research to institutions and universities all over the world.


Asunto(s)
Acceso a la Información , Histología , Radiobiología/métodos , Experimentación Animal , Animales , Archivos , Carcinogénesis , Bases de Datos Factuales , Humanos , Japón , Registros Médicos , Ratones , Neoplasias/epidemiología , Neoplasias/genética , Neoplasias Inducidas por Radiación/diagnóstico , Neoplasias Inducidas por Radiación/genética , Desarrollo de Programa , Radiobiología/tendencias , Investigación/tendencias , Proyectos de Investigación , Medición de Riesgo
14.
Int J Radiat Biol ; 94(8): 737-742, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29236558

RESUMEN

PURPOSE: Despite decades of research into radiation-induced adaptive responses, where prior irradiation changes the response to subsequent irradiations, the field of radiation oncology relies upon models of tumor control that assume that each radiation therapy fraction reproduces the same effect, known as iso-effect per fraction. Can these radiobiology principles both be true, forming a paradox or is only one of them right? Here, the apparent coexistence of these two contradictory observations is considered, examining how adaptive responses might apply in radiotherapy scenarios that are inconsistent with the majority of adaptive response experimental designs. CONCLUSION: While the iso-effect per fraction assumption would preclude the observation of adaptive responses for cells survival after radiotherapy fractions, this does not preclude the observation of adaptive responses for other endpoints. Adaptive responses for cell survival might also manifest without invalidating the iso-effect principle in practical terms. It may also be the case that instances of both phenomena can be observed under different conditions, but not at the same time.


Asunto(s)
Adaptación Fisiológica/efectos de la radiación , Fraccionamiento de la Dosis de Radiación , Radiobiología , Humanos , Neoplasias/patología , Neoplasias/fisiopatología , Neoplasias/radioterapia
15.
Clin Exp Metastasis ; 35(4): 223-236, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29159430

RESUMEN

Radiation therapy is an effective means of achieving local control in a wide range of primary tumours, with the reduction in the size of the tumour(s) thought to mediate the observed reductions in metastatic spread in clinical trials. However, there is evidence to suggest that the complex changes induced by radiation in the tumour environment can also present metastatic risks that may counteract the long-term efficacy of the treatment. More than 25 years ago, several largely theoretical mechanisms by which radiation exposure might increase metastatic risk were postulated. These include the direct release of tumour cells into the circulation, systemic effects of tumour and normal tissue irradiation and radiation-induced changes in tumour cell phenotype. Here, we review the data that has since emerged to either support or refute these putative mechanisms focusing on how the unique radiobiology underlying modern radiotherapy modalities might alter these risks.


Asunto(s)
Neoplasias Inducidas por Radiación/patología , Neoplasias/patología , Neoplasias/radioterapia , Células Neoplásicas Circulantes/patología , Células Neoplásicas Circulantes/efectos de la radiación , Animales , Humanos , Metástasis de la Neoplasia , Neoplasias/sangre , Radiobiología , Radioterapia/efectos adversos
16.
Sci Rep ; 8(1): 14325, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254198

RESUMEN

Radiation exposure during the peri-pubertal period is a proven risk factor for breast cancer, whereas parity is an established protective factor. The present study investigated whether parity imposes differential protective effects against radiation-induced rat mammary carcinoma depending on the age at exposure. Pre- and post-pubertal female rats, irradiated or left unirradiated, were mated and allowed to nurse until weaning or left unmated. Appearance of mammary tumors was monitored, and serum concentrations of estradiol and progesterone were measured following weaning. Carcinomas were evaluated by immunohistochemistry for estrogen receptor, progesterone receptor, and the cell proliferation marker Ki-67. Parity reduced the risk of carcinoma in unirradiated and pre-pubertally irradiated rats but not post-pubertally irradiated rats. Although radiation exposure increased serum progesterone level, parity after pre-pubertal exposure significantly decreased the elevated progesterone to a normal level, reflecting a protective effect. Moreover, parity significantly decreased the proportion of hormone receptor-positive carcinomas after pre-pubertal exposure. Parity was also related to the observed positive association between progesterone receptor and Ki-67 indices in cancer tissue, implying progesterone receptor-dependent cell proliferation. Thus, parity protects against radiation-induced rat mammary carcinogenesis depending on the age at exposure; the mechanisms may involve changes in hormone levels and cancer tissue.


Asunto(s)
Carcinogénesis/efectos de la radiación , Neoplasias Mamarias Experimentales/patología , Exposición Materna/efectos adversos , Neoplasias Inducidas por Radiación/patología , Paridad , Animales , Femenino , Masculino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Inducidas por Radiación/metabolismo , Embarazo , Ratas
17.
Cancers (Basel) ; 9(11)2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-29077012

RESUMEN

Whilst the near instantaneous physical interaction of radiation energy with living cells leaves little opportunity for inter-individual variation in the initial yield of DNA damage, all the downstream processes in how damage is recognized, repaired or resolved and therefore the ultimate fate of cells can vary across the population. In the clinic, this variability is observed most readily as rare extreme sensitivity to radiotherapy with acute and late tissue toxic reactions. Though some radiosensitivity can be anticipated in individuals with known genetic predispositions manifest through recognizable phenotypes and clinical presentations, others exhibit unexpected radiosensitivity which nevertheless has an underlying genetic cause. Currently, functional assays for cellular radiosensitivity represent a strategy to identify patients with potential radiosensitivity before radiotherapy begins, without needing to discover or evaluate the impact of the precise genetic determinants. Yet, some of the genes responsible for extreme radiosensitivity would also be expected to confer susceptibility to radiation-induced cancer, which can be considered another late adverse event associated with radiotherapy. Here, the utility of functional assays of radiosensitivity for identifying individuals susceptible to radiotherapy-induced second cancer is discussed, considering both the common mechanisms and important differences between stochastic radiation carcinogenesis and the range of deterministic acute and late toxic effects of radiotherapy.

18.
Radiat Res ; 188(2): 235-241, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28581892

RESUMEN

There is a natural tendency to expect that irradiation of an infant organ prior to development-related expansion will result in a higher risk of developing cancer than that of fully-developed adult tissue, and this has generally been observed. However, if tissues also vary in their initial responses to radiation depending on age, the interplay between tissue- and age-dependent risk would potentially be quite complex. We have previously shown opposing age-dependent induction of apoptosis for the intestinal epithelium and hematopoietic cells in mice, but such data are not yet available for the liver. Here, we have examined markers of DNA damage, initiation of DNA damage responses, cell cycle arrest, apoptosis and proliferation, as well as gene expression, in the B6C3F1 mouse liver over the hours and days after irradiation of mice at 1 or 7 weeks of age. We found that induction and resolution of radiation-induced DNA damage is not accompanied by significant changes in these cellular end points in the adult liver, while in infant hepatocytes modest induction of p53 accumulation and p21-mediated cell cycle arrest in a small fraction of damaged cells was overshadowed by a further stimulation of proliferation over the relatively high levels already found in the neonatal liver. We observed distinct expression of genes that regulate cell division between the ages, which may contribute to the differential responses. These data suggest that the growth factor signaling environment of the infant liver may mediate radiation-induced proliferation and increased liver cancer risk after irradiation during early life.


Asunto(s)
Envejecimiento/efectos de la radiación , Hepatocitos/citología , Hepatocitos/efectos de la radiación , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de la radiación , Daño del ADN , Femenino , Regulación de la Expresión Génica/efectos de la radiación , Hepatocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
19.
Environ Mol Mutagen ; 58(8): 592-606, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28921690

RESUMEN

DNA mutations play a crucial role in the origins of cancer, and the clonal expansion of mutant cells is one of the fundamental steps in multistage carcinogenesis. In this study, we correlated tumor incidence in B6C3F1 mice during the period after exposure to N-ethyl-N-nitrosourea (ENU) with the persistence of ENU-induced mutant clones in transgenic gpt delta B6C3F1 mice. The induced gpt mutations afforded no selective advantage in the mouse cells and could be distinguished by a mutational spectrum that is characteristic of ENU treatment. The gpt mutations were passengers of the mutant cell of origin and its daughter cells and thus could be used as neutral markers of clones that arose and persisted in the tissues. Female B6C3F1 mice exposed for 1 month to 200 ppm ENU in the drinking water developed early thymic lymphomas and late liver and lung tumors. To assay gpt mutations, we sampled the thymus, liver, lung, and small intestine of female gpt delta mice at 3 days, 4 weeks, and 8 weeks after the end of ENU exposure. Our results reveal that, in all four tissues, the ENU-induced gpt mutations persisted for weeks after the end of mutagen exposure. Clonal expansion of mutant cells was observed in the thymus and small intestine, with the thymus showing larger clone sizes. These results indicate that the clearance of mutant cells and the potential for clonal expansion during normal tissue growth depends on tissue type and that these factors may affect the sensitivity of different tissues to carcinogenesis. Environ. Mol. Mutagen. 58:592-606, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Carcinogénesis/genética , Proteínas de Escherichia coli/genética , Etilnitrosourea/toxicidad , Mutágenos/toxicidad , Pentosiltransferasa/genética , Animales , Carcinogénesis/efectos de los fármacos , Proteínas de Escherichia coli/biosíntesis , Humanos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/patología , Hígado/efectos de los fármacos , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Transgénicos , Pruebas de Mutagenicidad/métodos , Mutación/efectos de los fármacos , Especificidad de Órganos/efectos de los fármacos , Pentosiltransferasa/biosíntesis , Timo/efectos de los fármacos , Timo/patología
20.
Radiat Res ; 187(2): 210-220, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28135125

RESUMEN

Epidemiology studies have shown that children are at greater overall risk of radiation-induced cancer, but the modifying effect of age at exposure in different tissues is heterogeneous. Early epidemiology findings of increased lung cancer risk with increasing age at the time of exposure have been dismissed, with suggestions that the trend is an artefact from a failure to adequately correct for the effects of tobacco smoking. Yet, differing models used in subsequent analyses have shown that the increased susceptibility with age, counter to the overall solid tumor trend, can either be confirmed or discounted depending on the model parameters used. In this study, we analyzed the induction of tumors in female Wistar rats exposed to increasing thoracic doses of X-ray as neonates, juveniles or young adults, to allow the effect of age at exposure in this early period to be observed in the absence of any interactions with smoking. Histology was used to compare tumor subtypes among groups, and genomic DNA copy number alterations in a number of tumors arising after irradiation at different ages were examined. Induction of lung cancers increased with radiation dose, with the frequency of early occurring lung adenomas greater in rats irradiated at older ages. At the highest dose, the rats irradiated at 5 or 15 weeks of age showed increased age-specific rates of lung adenocarcinomas in later life compared to those irradiated at 1 week of age. However, thoracic mammary gland tumors induced by the highest dose at the later ages significantly decreased the lifespan in these groups, reducing the number of rats at risk of radiation-induced lung adenocarcinoma. There was no induction of mammary tumors outside of the irradiated field. Lung adenocarcinomas showed widespread DNA copy number aberrations at the chromosome level, but the only recurrent lesions were intragenic Fhit deletions and losses on chromosome 4. The results presented here suggest that the risk of radiation-induced lung cancer after irradiation may not monotonically decrease with age, and demonstrate that increasing lung cancer risk with exposure age can be observed independent of corrections for smoking, and that mammary tumors may show a similar relationship with age.


Asunto(s)
Envejecimiento/efectos de la radiación , Neoplasias Pulmonares/etiología , Neoplasias Mamarias Experimentales/etiología , Neoplasias Inducidas por Radiación/etiología , Tórax/efectos de la radiación , Animales , Variaciones en el Número de Copia de ADN/efectos de la radiación , Femenino , Incidencia , Neoplasias Pulmonares/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Inducidas por Radiación/genética , Ratas , Ratas Wistar , Rayos X/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA