Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(7): e0307598, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39042641

RESUMEN

Metformin is a biguanide currently used in the treatment of diabetes mellitus type 2. Besides its anti-glycemic effects, metformin has been reported to induce different cellular pleiotropic effects, depending on concentration and time of treatment. Here we report one administration of metformin (0.5 mM) has radioprotective effects in vitro on BJ human fibroblasts, increasing DNA damage repair and increasing SOD1 expression in the nucleus. Importantly, metformin (200 mg/kg) pre-administration for only 3 days in wild type 129/sv mice, decreases the formation of micronuclei in bone marrow cells and DNA damage in colon and lung tissues compared to control irradiated mice at sub-lethal and lethal doses, increasing the overall survival fraction by 37% after 10Gy total body irradiation. We next pre-treated with metformin and then exposed 129/sv mice, to a galactic cosmic rays simulation (GCRsim), at the NASA Space Radiation Laboratory (NSRL). We found metformin pre-treatment decreases the presence of bone marrow micronuclei and DNA damage in colon and lung tissues and an increase of 8-oxoguanine DNA glycosylase-1 (OGG1) expression. Our data highlight a radioprotective effect of metformin through an indirect modulation of the gene expression involved in the cellular detoxification rather than its effects on mitochondria.


Asunto(s)
Daño del ADN , Hipoglucemiantes , Metformina , Protectores contra Radiación , Metformina/farmacología , Animales , Ratones , Humanos , Protectores contra Radiación/farmacología , Hipoglucemiantes/farmacología , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Fibroblastos/metabolismo , Superóxido Dismutasa/metabolismo , Línea Celular , Pruebas de Micronúcleos , Pulmón/efectos de los fármacos , Pulmón/efectos de la radiación , Pulmón/patología , Pulmón/metabolismo , Masculino
2.
Life Sci Space Res (Amst) ; 41: 43-51, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38670651

RESUMEN

Prolonged manned space flight exposure risks to galactic comic radiation, has led to uncertainties in a variety of health risks. Our previous work, utilizing either single ion or multiple ion radiation exposure conducted at the NSRL (NASA Space Radiation Laboratory, Brookhaven, NY) demonstrated that HZE ion components of the GCR result in persistent inflammatory signaling, increased mutations, and higher rates of cancer initiation and progression. With the development of the 33-beam galactic cosmic radiation simulations (GCRsim) at the NSRL, we can more closely test on earth the radiation environment found in space. With a previously used lung cancer susceptible mouse model (K-rasLA-1), we performed acute exposure experiments lasting 1-2 h, and chronic exposure experiments lasting 2-6 weeks with a total dose of 50 cGy and 75 cGy. We obtained histological samples from a subset of mice 100 days post-irradiation, and the remaining mice were monitored for overall survival up to 1-year post-irradiation. When we compared acute exposures (1-2 hrs.) and chronic exposure (2-6 weeks), we found a trend in the increase of lung adenocarcinoma respectively for a total dose of 50 cGy and 75 cGy. Furthermore, when we added neutron exposure to the 75 cGy of GCRsim, we saw a further increase in the incidence of adenocarcinoma. We interpret these findings to suggest that the risks of carcinogenesis are heightened with doses anticipated during a round trip to Mars, and this risk is magnified when coupled with extra neutron exposure that are expected on the Martian surface. We also observed that risks are reduced when the NASA official 33-beam GCR simulations are provided at high dose rates compared to low dose rates.


Asunto(s)
Radiación Cósmica , Progresión de la Enfermedad , Neoplasias Pulmonares , Neoplasias Inducidas por Radiación , Animales , Radiación Cósmica/efectos adversos , Ratones , Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/patología , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/patología , Vuelo Espacial , Femenino , Masculino
3.
Behav Brain Res ; 419: 113677, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34818568

RESUMEN

In long-term spaceflight, astronauts will face unique cognitive loads and social challenges which will be complicated by communication delays with Earth. It is important to understand the central nervous system (CNS) effects of deep spaceflight and the associated unavoidable exposure to galactic cosmic radiation (GCR). Rodent studies show single- or simple-particle combination exposure alters CNS endpoints, including hippocampal-dependent behavior. An even better Earth-based simulation of GCR is now available, consisting of a 33-beam (33-GCR) exposure. However, the effect of whole-body 33-GCR exposure on rodent behavior is unknown, and no 33-GCR CNS countermeasures have been tested. Here astronaut-age-equivalent (6mo-old) C57BL/6J male mice were exposed to 33-GCR (75cGy, a Mars mission dose). Pre-/during/post-Sham or 33-GCR exposure, mice received a diet containing a 'vehicle' formulation alone or with the antioxidant/anti-inflammatory compound CDDO-EA as a potential countermeasure. Behavioral testing beginning 4mo post-irradiation suggested radiation and diet did not affect measures of exploration/anxiety-like behaviors (open field, elevated plus maze) or recognition of a novel object. However, in 3-Chamber Social Interaction (3-CSI), CDDO-EA/33-GCR mice failed to spend more time exploring a holder containing a novel mouse vs. a novel object (empty holder), suggesting sociability deficits. Also, Vehicle/33-GCR and CDDO-EA/Sham mice failed to discriminate between a novel stranger vs. familiarized stranger mouse, suggesting blunted preference for social novelty. CDDO-EA given pre-/during/post-irradiation did not attenuate the 33-GCR-induced blunting of preference for social novelty. Future elucidation of the mechanisms underlying 33-GCR-induced blunting of preference for social novelty will improve risk analysis for astronauts which may in-turn improve countermeasures.


Asunto(s)
Conducta Animal , Disfunción Cognitiva , Radiación Cósmica/efectos adversos , Ácido Oleanólico/análogos & derivados , Exposición a la Radiación/efectos adversos , Reconocimiento en Psicología , Conducta Social , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/efectos de la radiación , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ácido Oleanólico/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/efectos de la radiación
4.
Life Sci Space Res (Amst) ; 24: 108-115, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31987474

RESUMEN

Most of the research in understanding space radiation-induced cancer progression and risk assessment has been performed using mono-energetic single-ion beams. However, the space radiation environment consists of a wide variety of ion species with a various range of energies. Using the fast beam switching technology developed at the NASA Space Radiation Laboratory (NSRL) at Brookhaven National Laboratory (BNL), ion species can be switched rapidly allowing investigators to use multiple ions with different energies to simulate more closely the radiation environment found in space. Here, we exposed a lung cancer susceptible mouse model (K-rasLA-1) to three sequential ion beams: Proton (H) (120 MeV/n) 20 cGy, Helium (He) (250 MeV/n) 5.0 cGy, and Silicon (Si) (300 MeV/n) 5.0 cGy with a dose rate of 0.5 cGy/min. Using three ion beams we performed whole body irradiation with a total dose of 30 cGy in two different orders: 3B-1 (H→He→Si) and 3B-2 (Si→He→H) and used 30 cGy H single-ion beam as a reference. In this study we show that whole-body irradiation with H→He→Si increases the incidence of premalignant lesions and systemic oxidative stress in mice 100 days post-irradiation more than (Si→He→H) and H only irradiation. Additionally, we observed an increase in adenomas with atypia and adenocarcinomas in H→He→Si irradiated mice but not in (Si→He→H) or H (30 cGy) only irradiated mice. When we used the H→He→Si irradiation sequence but skipped a day before exposing the mice to Si, we did not observe the increased incidence of cancer initiation and progression. We also found that a non-toxic anti-inflammatory, anti-oxidative radioprotector (CDDO-EA) reduced H→He→Si induced oxidative stress and cancer initiation almost back to baseline. Thus, exposure to H→He→Si elicits significant changes in lung cancer initiation that can be mitigated using CDDO-EA.


Asunto(s)
Neoplasias Pulmonares/prevención & control , Neoplasias Inducidas por Radiación/prevención & control , Protección Radiológica/métodos , Animales , Modelos Animales de Enfermedad , Femenino , Neoplasias Pulmonares/etiología , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Transgénicos , Radiación Ionizante , Vuelo Espacial , Irradiación Corporal Total
5.
Life Sci Space Res (Amst) ; 19: 31-42, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30482279

RESUMEN

There are considerable health risks related to ionizing and proton radiation exposure. While there is a long history of health risks associated with ionizing (photon) radiation exposure, there is a limited understanding of the long-term health risks associated with proton radiation exposure. Since proton radiation is becoming more common in cancer therapy, the long-term biological effects of proton radiation remain less well characterized in terms of radiotherapy and well as for astronauts during deep space explorations. In this study, we compared the long-term side effects of proton radiation to equivalent doses of X-rays in the initiation and progression of premalignant lesions in a lung cancer susceptible mouse model (K-rasLA1). We show proton irradiation causes more complex DNA damage that is not completely repaired resulting in increased oxidative stress in the lungs both acutely and persistently. We further observed K-rasLA1 mice irradiated with protons had an increased number and size of initiated and premalignant lesions and adenomas that were often infiltrated with inflammatory cells. Proton irradiated mice had a lower median survival and increased carcinoma incidence as compared to unirradiated controls and X-rays exposed mice. Our conclusion is that exposure to proton irradiation enhances the progression of premalignant lesions to invasive carcinomas through persistent DNA damage, chronic oxidative stress, and immunosuppression.


Asunto(s)
Modelos Animales de Enfermedad , Inflamación/patología , Neoplasias Pulmonares/patología , Neoplasias Inducidas por Radiación/patología , Protones/efectos adversos , Animales , Daño del ADN , Progresión de la Enfermedad , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Inflamación/etiología , Inflamación/metabolismo , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Invasividad Neoplásica , Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/metabolismo , Estrés Oxidativo
6.
Neoplasia ; 20(8): 826-837, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30015158

RESUMEN

Standard and targeted cancer therapies for late-stage cancer patients almost universally fail due to tumor heterogeneity/plasticity and intrinsic or acquired drug resistance. We used the telomerase substrate nucleoside precursor, 6-thio-2'-deoxyguanosine (6-thio-dG), to target telomerase-expressing non-small cell lung cancer cells resistant to EGFR-inhibitors and commonly used chemotherapy combinations. Colony formation assays, human xenografts as well as syngeneic and genetically engineered immune competent mouse models of lung cancer were used to test the effect of 6-thio-dG on targeted therapy- and chemotherapy-resistant lung cancer human cells and mouse models. We observed that erlotinib-, paclitaxel/carboplatin-, and gemcitabine/cisplatin-resistant cells were highly sensitive to 6-thio-dG in cell culture and in mouse models. 6-thio-dG, with a known mechanism of action, is a potential novel therapeutic approach to prolong disease control of therapy-resistant lung cancer patients with minimal toxicities.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Telomerasa/metabolismo , Animales , Línea Celular Tumoral , Desoxiguanosina/análogos & derivados , Desoxiguanosina/farmacología , Femenino , Humanos , Ratones , Ratones Desnudos , Tionucleósidos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
7.
Mol Cancer Ther ; 17(5): 943-951, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29467273

RESUMEN

Truncated APC selective inhibitor-1 (TASIN-1) is a recently identified small molecule that selectively kills colorectal cancer cells that express truncated adenomatous polyposis coli (APC) by reducing cellular cholesterol levels. However, the downstream mechanism responsible for its cytotoxicity is not well understood. In this study, we show that TASIN-1 leads to apoptotic cell death via inducing ER stress-dependent JNK activation in human truncated APC colon cancer cells, accompanied by production of reactive oxygen species (ROS). In addition, TASIN-1 inhibits AKT activity through a cholesterol-dependent manner. Human colon tumor xenografts in immunodeficient mice also show the same TASIN-1 induced molecular mechanisms of tumor cell death as observed in vitro Taken together, cholesterol depletion by TASIN-1 treatment induces apoptotic cell death through activating ER stress/ROS/JNK axis and inhibiting AKT pro-survival signaling in colon cancer cells with truncated APC both in vitro and in vivoMol Cancer Ther; 17(5); 943-51. ©2018 AACR.


Asunto(s)
Apoptosis/efectos de los fármacos , Colesterol/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Piperidinas/farmacología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Células HCT116 , Células HT29 , Humanos , Ratones Desnudos , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo
8.
Sci Rep ; 6: 31669, 2016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27539227

RESUMEN

The effects of radiation in two-dimensional (2D) cell culture conditions may not recapitulate tissue responses as modeled in three-dimensional (3D) organotypic culture. In this study, we determined if the frequency of radiation-induced transformation and cancer progression differed in 3D compared to 2D culture. Telomerase immortalized human bronchial epithelial cells (HBECs) with shTP53 and mutant KRas expression were exposed to various types of radiation (gamma, (+)H, (56)Fe) in either 2D or 3D culture. After irradiation, 3D structures were dissociated and passaged as a monolayer followed by measurement of transformation, cell growth and expression analysis. Cells irradiated in 3D produced significantly fewer and smaller colonies in soft agar than their 2D-irradiated counterparts (gamma P = 0.0004; (+)H P = 0.049; (56)Fe P < 0.0001). The cell culture conditions did not affect cell killing, the ability of cells to survive in a colony formation assay, and proliferation rates after radiation-implying there was no selection against cells in or dissociated from 3D conditions. However, DNA damage repair and apoptosis markers were increased in 2D cells compared to 3D cells after radiation. Ideally, expanding the utility of 3D culture will allow for a better understanding of the biological consequences of radiation exposure.


Asunto(s)
Bronquios/metabolismo , Técnicas de Cultivo de Célula/métodos , Transformación Celular Neoplásica/efectos de la radiación , Daño del ADN , Células Epiteliales/metabolismo , Rayos gamma/efectos adversos , Apoptosis/efectos de la radiación , Bronquios/patología , Línea Celular Transformada , Transformación Celular Neoplásica/patología , Células Epiteliales/patología , Regulación de la Expresión Génica/efectos de la radiación , Humanos
9.
Cancer Res ; 74(21): 6330-40, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25164016

RESUMEN

SNAIL1 has been suggested to regulate breast cancer metastasis based on analyses of human breast tumor transcriptomes and experiments using cancer cell lines and xenografts. However, in vivo genetic experimental support for a role for SNAIL1 in breast cancer metastasis that develops in an immunocompetent tumor microenvironment has not been determined. To address this question, we created a genetic SNAIL1 model by coupling an endogenous SNAIL1 reporter with an inducible SNAIL1 transgene. Using multiple genetic models of breast cancer, we demonstrated that endogenous SNAIL1 expression was restricted to primary tumors that ultimately disseminate. SNAIL1 gene deletion either during the premalignant phase or after primary tumors have reached a palpable size blunted metastasis, indicating that late metastasis was the main driver of metastasis and that this was dependent on SNAIL1. Importantly, SNAIL1 expression during breast cancer metastasis was transient and forced transient, but not continuous. SNAIL1 expression in breast tumors was sufficient to increase metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Factores de Transcripción/biosíntesis , Microambiente Tumoral/genética , Neoplasias de la Mama/patología , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Transducción de Señal/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA