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

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 410: 115360, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33279515

RESUMEN

People living in southwest part of United States are exposed to uranium (U) through drinking water, air, and soil. U is radioactive, but independent of this radioactivity also has important toxicological considerations as an environmental metal. At environmentally relevant concentrations, U is both mutagenic and carcinogenic. Emerging evidence shows that U inhibits DNA repair activity, but how U interacts with DNA repair proteins is still largely unknown. Herein, we report that U directly interacts with the DNA repair protein, Protein Poly (ADP-ribose) Polymerase 1 (PARP-1) through direct binding with the zinc finger motif, resulting in zinc release from zinc finger and DNA binding activity loss of the protein. At the peptide level, instead of direct competition with zinc ion in the zinc finger motif, U does not show thermodynamic advantages over zinc. Furthermore, zinc pre-occupied PARP-1 zinc finger is insensitive to U treatment, but U bound to PARP-1 zinc finger can be partially replaced by zinc. These results provide mechanistic basis on molecular level to U inhibition of DNA repair.


Asunto(s)
Reparación del ADN/fisiología , Reparación del ADN/efectos de la radiación , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/efectos de la radiación , Uranio/metabolismo , Uranio/toxicidad , Secuencia de Aminoácidos , Células Cultivadas , Exposición a Riesgos Ambientales/efectos adversos , Humanos , Recién Nacido , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Poli(ADP-Ribosa) Polimerasa-1/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología
2.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998270

RESUMEN

Immune modulation is a very modern medical field for targeting viral infections. In the race to develop the best immune modulator against viruses, curcumin, as a natural product, is inexpensive, without side effects, and can stimulate very well certain areas of the human immune system. As a bright yellow component of turmeric spice, curcumin has been the subject of thousands of scientific and clinical studies in recent decades to prove its powerful antioxidant properties and anticancer effects. Curcumin has been shown to influence inter- and intracellular signaling pathways, with direct effects on gene expression of the antioxidant proteins and those that regulate the immunity. Experimental studies have shown that curcumin modulates several enzyme systems, reduces nitrosative stress, increases the antioxidant capacity, and decreases the lipid peroxidation, protecting against fatty liver pathogenesis and fibrotic changes. Hepatitis B virus (HBV) affects millions of people worldwide, having sometimes a dramatic evolution to chronic aggressive infection, cirrhosis, and hepatocellular carcinoma. All up-to-date treatments are limited, there is still a gap in the scientific knowledge, and a sterilization cure may not yet be possible with the removal of both covalently closed circular DNA (cccDNA) and the embedded HBV DNA. With a maximum light absorption at 420 nm, the cytotoxicity of curcumin as photosensitizer could be expanded by the intravenous blue laser blood irradiation (IVBLBI) or photobiomodulation in patients with chronic hepatitis B infection, Hepatitis B e-antigen (HBeAg)-positive, noncirrhotic, but nonresponsive to classical therapy. Photobiomodulation increases DNA repair by the biosynthesis of complex molecules with antioxidant properties, the outset of repairing enzyme systems and new phospholipids for regenerating the cell membranes. UltraBioavailable Curcumin and blue laser photobiomodulation could suppress the virus and control better the disease by reducing inflammation/fibrosis and stopping the progression of chronic hepatitis, reversing fibrosis, and diminishing the progression of cirrhosis, and decreasing the incidence of hepatocellular carcinoma. Photodynamic therapy with blue light and curcumin opens new avenues for the effective prevention and cure of chronic liver infections and hepatocellular carcinoma. Blue laser light and UltraBioavailable Curcumin could be a new valuable alternative for medical applications in chronic B viral hepatitis and hepatocarcinoma, saving millions of lives.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma Hepatocelular/radioterapia , Curcumina/uso terapéutico , Hepatitis B Crónica/radioterapia , Cirrosis Hepática/radioterapia , Neoplasias Hepáticas/radioterapia , Terapia por Luz de Baja Intensidad/métodos , Antioxidantes/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/virología , Reparación del ADN/efectos de la radiación , ADN Circular/antagonistas & inhibidores , ADN Circular/genética , ADN Circular/metabolismo , ADN Viral/antagonistas & inhibidores , ADN Viral/genética , ADN Viral/metabolismo , Antígenos e de la Hepatitis B/genética , Antígenos e de la Hepatitis B/inmunología , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/patogenicidad , Virus de la Hepatitis B/efectos de la radiación , Hepatitis B Crónica/complicaciones , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/virología , Humanos , Factores Inmunológicos/uso terapéutico , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Hígado/efectos de la radiación , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/etiología , Cirrosis Hepática/virología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/virología , Fármacos Fotosensibilizantes/uso terapéutico
3.
Cell Cycle ; 18(15): 1770-1783, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31238782

RESUMEN

Although improvements in radiation therapy were made over the years, radioresistance is still a major challenge. Cancer cells are often deficient for DNA repair response, a feature that is currently exploited as a new anti-cancer strategy. In this context, combination of inhibitors targeting complementary pathways is of interest to sensitize cells to radiation. In this work, we used PARP (Olaparib) and RAD51 (B02) inhibitors to radiosensitize cancer cells to proton and X-ray radiation. More particularly, Olaparib and B02 were used at concentration leading to limited cytotoxic (alone or in combination) but increasing cell death when the cells were irradiated. We showed that, although at limited concentration, Olaparib and B02 were able to radiosensitize different cancer cell lines, i.e. lung and pancreatic cancer cells. Antagonistic, additive or synergistic effects were observed and correlated to cell proliferation rate. The inhibitors enhanced persistent DNA damage, delayed apoptosis, prolonged cell cycle arrest and senescence upon irradiation. These results demonstrated that radiation-induced synthetic lethality might widen the therapeutic window, hence extending the use of PARP inhibitors to patients without BRCAness.


Asunto(s)
Neoplasias/radioterapia , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Recombinasa Rad51/antagonistas & inhibidores , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Mutaciones Letales Sintéticas , Células A549 , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Compuestos de Boro/uso terapéutico , Compuestos de Boro/toxicidad , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , 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 , Humanos , Ftalazinas/uso terapéutico , Ftalazinas/toxicidad , Piperazinas/uso terapéutico , Piperazinas/toxicidad , Poli(ADP-Ribosa) Polimerasas/metabolismo , Protones , Radiación Ionizante , Factores de Tiempo
4.
Int J Mol Sci ; 19(8)2018 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-30115874

RESUMEN

Hyperthermia (HT) and molecular targeting agents can be used to enhance the effect of radiotherapy (RT). The purpose of this paper is to evaluate radiation sensitization by HT and different molecular targeting agents (Poly [ADP-ribose] polymerase 1 inhibitor, PARP1-i; DNA-dependent protein kinase catalytic subunit inhibitor, DNA-PKcs-i and Heat Shock Protein 90 inhibitor, HSP90-i) in cervical cancer cell lines. Survival curves of SiHa and HeLa cells, concerning the combined effects of radiation with hyperthermia and PARP1-i, DNA-PKcs-i or HSP90-i, were analyzed using the linear-quadratic model: S(D)/S(0) = exp - (αD + ßD²). The values of the linear-quadratic (LQ) parameters α and ß, determine the effectiveness at low and high doses, respectively. The effects of these sensitizing agents on the LQ parameters are compared to evaluate dose-dependent differences in radio enhancement. Combination of radiation with hyperthermia, PARP1-i and DNA-PKcs-i significantly increased the value of the linear parameter α. Both α and ß were significantly increased for HSP90-i combined with hyperthermia in HeLa cells, though not in SiHa cells. The Homologous Recombination pathway is inhibited by hyperthermia. When hyperthermia is combined with DNA-PKcs-i and PARP1-i, the Non-Homologous End Joining or Alternative Non-Homologous End Joining pathway is also inhibited, leading to a more potent radio enhancement. The observed increments of the α value imply that significant radio enhancement is obtained at clinically-used radiotherapy doses. Furthermore, the sensitizing effects of hyperthermia can be even further enhanced when combined with other molecular targeting agents.


Asunto(s)
Hipertermia Inducida , Terapia Molecular Dirigida , Radiación Ionizante , Neoplasias del Cuello Uterino/terapia , Supervivencia Celular/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de la radiación , Femenino , Células HeLa , Humanos , Resultado del Tratamiento
5.
Molecules ; 23(4)2018 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-29570674

RESUMEN

Skin circadian clock system responds to daily changes, thereby regulating skin functions. Exposure of the skin to UV irradiation induces the expression of matrix metalloproteinase-1 (MMP-1) and causes DNA damage. It has been reported both DNA repair and DNA replication are regulated by the circadian clock in mouse skin. However, the molecular link between circadian clock and MMP-1 has little been investigated. We found PERIOD protein, a morning clock component, represses the expression of MMP-1 in human keratinocytes by using a PER-knockdown strategy. Treatment with siPer3 alleviated the suppression of MMP-1 expression induced by forskolin. Results revealed PER3 suppresses the expression of MMP-1 via cAMP signaling pathway. Additionally, we screened for an activator of PER that could repress the expression of MMP-1 using HaCaT cell line containing PER promoter-luciferase reporter gene. Results showed Lespedeza capitate extract (LCE) increased PER promoter activity. LCE inhibited the expression of MMP-1 and its effect of LCE was abolished in knockdown of PER2 or PER3, demonstrating LCE can repress the expression of MMP-1 through PER. Since circadian clock component PER can regulate MMP-1 expression, it might be a new molecular mechanism to develop therapeutics to alleviate skin aging and skin cancer.


Asunto(s)
Queratinocitos/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Proteínas Circadianas Period/metabolismo , Línea Celular , Relojes Circadianos/genética , Relojes Circadianos/fisiología , AMP Cíclico/metabolismo , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/efectos de la radiación , Lespedeza/química , Metaloproteinasa 1 de la Matriz/genética , Proteínas Circadianas Period/genética , Extractos Vegetales/química , Extractos Vegetales/farmacología , Rayos Ultravioleta
6.
Radiat Oncol ; 12(1): 206, 2017 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-29284495

RESUMEN

BACKGROUND: Tumor Treating Fields (TTFields) are an anti-neoplastic treatment modality delivered via application of alternating electric fields using insulated transducer arrays placed directly on the skin in the region surrounding the tumor. A Phase 3 clinical trial has demonstrated the effectiveness of continuous TTFields application in patients with glioblastoma during maintenance treatment with Temozolomide. The goal of this study was to evaluate the efficacy of combining TTFields with radiation treatment (RT) in glioma cells. We also examined the effect of TTFields transducer arrays on RT distribution in a phantom model and the impact on rat skin toxicity. METHODS: The efficacy of TTFields application after induction of DNA damage by RT or bleomycin was tested in U-118 MG and LN-18 glioma cells. The alkaline comet assay was used to measure repair of DNA lesions. Repair of DNA double strand breaks (DSBs) were assessed by analyzing γH2AX or Rad51 foci. DNA damage and repair signaled by the activation pattern of phospho-ATM (pS1981) and phospho-DNA-PKcs (pS2056) was evaluated by immunoblotting. The absorption of the RT energy by transducer arrays was measured by applying RT through arrays placed on a solid-state phantom. Skin toxicities were tested in rats irradiated daily through the arrays with 2Gy (total dose of 20Gy). RESULTS: TTFields synergistically enhanced the efficacy of RT in glioma cells. Application of TTFields to irradiated cells impaired repair of irradiation- or chemically-induced DNA damage, possibly by blocking homologous recombination repair. Transducer arrays presence caused a minor reduction in RT intensity at 20 mm and 60 mm below the arrays, but led to a significant increase in RT dosage at the phantom surface jeopardizing the "skin sparing effect". Nevertheless, transducer arrays placed on the rat skin during RT did not lead to additional skin reactions. CONCLUSIONS: Administration of TTFields after RT increases glioma cells treatment efficacy possibly by inhibition of DNA damage repair. These preclinical results support the application of TTFields therapy immediately after RT as a viable regimen to enhance RT outcome. Phantom measurements and animal models imply that it may be possible to leave the transducer arrays in place during RT without increasing skin toxicities.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de la radiación , Terapia por Estimulación Eléctrica , Glioma/radioterapia , Fantasmas de Imagen , Enfermedades de la Piel/prevención & control , Animales , Glioma/genética , Glioma/patología , Humanos , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas
7.
Radiat Environ Biophys ; 56(4): 413-422, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28913689

RESUMEN

Many studies suggest that exogenous antioxidants may protect cells against DNA damage caused with ionizing radiation. One of the most powerful antioxidants is lycopene (LYC), a carotenoid derived from tomatoes. The aim of this study was to investigate, using the comet assay, whether LYC can act as protectors/modifiers and prevent DNA damage induced in human blood lymphocytes, as well as to mitigate the effects of radiation exposure. In this project, LYC, dissolved in DMSO at a concentration of 10, 20 or 40 µM/ml of cell suspension, was added to the isolated lymphocytes from human blood at appropriate intervals before or after the X-irradiation at doses of 0.5, 1 and 2 Gy. Cell viability in all groups was maintained at above 70%. The results showed the decrease of DNA damage in cells treated with various concentrations of LYC directly and 1 h before exposure to X-rays compared to the control group exposed to irradiation alone. Contrary results were observed in cells exposed to LYC immediately after exposure to ionizing radiation. The studies confirmed the protective effect of LYC against DNA damage induced by ionizing radiation, but after irradiation the carotenoid did not stimulate of DNA repair and cannot act as modifier. However, supplementation with LYC, especially at lower doses, may be useful in protection from radiation-induced oxidative damage.


Asunto(s)
Carotenoides/farmacología , Linfocitos/efectos de los fármacos , Linfocitos/efectos de la radiación , Protectores contra Radiación/farmacología , Adulto , Daño del ADN , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Femenino , Humanos , Licopeno , Linfocitos/metabolismo , Pruebas de Mutagenicidad , Mutación/efectos de los fármacos , Mutación/efectos de la radiación , Rayos X/efectos adversos
8.
Semin Cancer Biol ; 46: 138-145, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28412456

RESUMEN

Numerous plant products have been used to prevent and manage a wide variety of diseases for centuries. These products are now considered as promising options for the development of more effective and less toxic alternatives to the systems of medicine developed primarily in developed countries in the modern era. Grape seed proanthocyanidins (GSPs) are of great interest due to their anti-carcinogenic effects that have been demonstrated using various tumor models including ultraviolet (UV) radiation-induced non-melanoma skin cancer. In a pre-clinical mouse model supplementation of a control diet (AIN76A) with GSPs at concentrations of 0.2% and 0.5% (w/w) significantly inhibits the growth and multiplicity of UVB radiation-induced skin tumors. In this review, we summarize the evidence that this inhibition of UVB-induced skin tumor development by dietary GSPs is mediated by a multiplicity of coordinated effects including: (i) Promotion of the repair of damaged DNA by nuclear excision repair mechanisms, and (ii) DNA repair-dependent stimulation of the immune system following the functional activation of dendritic cells and effector T cells. Dietary GSPs hold promise for the development of an effective alternative strategy for the prevention of excessive solar UVB radiation exposure-induced skin diseases including the risk of non-melanoma skin cancer in humans.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Proantocianidinas/uso terapéutico , Neoplasias Cutáneas/dietoterapia , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Humanos , Sistema Inmunológico/efectos de la radiación , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/radioterapia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Rayos Ultravioleta/efectos adversos
9.
Int J Biol Macromol ; 94(Pt A): 585-593, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27769934

RESUMEN

Radioprotective effects of the water-soluble polyphenolic glycoconjugates, isolated from flowers of Sanguisorba officinalis L.(SO) and Erigeron canadensis L.(EC), and from leaves of Fragaria vesca L. (FV) and Rubus plicatus Whe. Et N. E. (RP), against γ-radiation-induced toxicity in human peripheral blood lymphocytes were investigated. Cell treatment with glycoconjugates (1, 5 and 25µg/mL) prior exposure to 10/15Gy radiation resulted in concentration-dependent reduction of DNA damage including oxidative DNA lesions (comet assay), substantial inhibition of lipid peroxidation (TBARS) and restoration of superoxide dismutase and S-glutathione transferase activities. Glycoconjugates isolated from SO and EC ensured better protection versus these from RP and FV, with the SO product potential comparable to that of the reference quercetin. Strong antioxidant/radioprotective activity of the SO and EC glycoconjugates could be attributed to high abundance of syringol-type and ferulic acid units in their matrices, respectively. Moreover, polyphenolic glycoconjugates (25µg/mL), including RP and FV products, significantly decreased DNA damage when applied post-radiation suggesting their modulating effects on DNA repair pathways. Preliminary data on the glycoconjugate phenolic structural units, based on GLC/MS of the products of pyrolysis and in situ methylation, in relation to application of plant products as potential radioprotectors is promising and deserves further investigation.


Asunto(s)
Antioxidantes/farmacología , Asteraceae/química , Glicoconjugados/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Protectores contra Radiación/farmacología , Rosaceae/química , Antioxidantes/química , Antioxidantes/aislamiento & purificación , Ensayo Cometa , Ácidos Cumáricos/análisis , Ácidos Cumáricos/química , Fragmentación del ADN/efectos de los fármacos , Fragmentación del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Rayos gamma , Glutatión Transferasa/metabolismo , Glicoconjugados/química , Glicoconjugados/aislamiento & purificación , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/efectos de la radiación , Extractos Vegetales/química , Cultivo Primario de Células , Pirogalol/análogos & derivados , Pirogalol/análisis , Pirogalol/química , Quercetina/farmacología , Protectores contra Radiación/química , Protectores contra Radiación/aislamiento & purificación , Superóxido Dismutasa/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
10.
Radiat Oncol ; 11(1): 153, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27876069

RESUMEN

BACKGROUND: In our previous study, monogalactosyl diacylglycerol (MGDG) purified from spinach was found to have cytotoxic effects in human cancer cell lines. This study further assessed whether MGDG can enhance the cytotoxic effects of radiation in human pancreatic cancer cells in vitro and in vivo. METHODS: Glycoglycerolipids from spinach including MGDG were extracted from dried spinach. The cytotoxicity of MGDG were evaluated by the MTT assay using four human pancreatic cancer cell lines (MIAPaCa-2, AsPC-1, BxPC-3 and PANC-1) and normal human dermal fibroblasts (NHDFs). The effects of radiation and MGDG alone or in combination in MIAPaCa-2 cells was analyzed with the colony forming and apoptosis assays, western blotting and cell cycle and DNA damage analyses (γ-H2AX foci staining and comet assay). The inhibitory effects on tumor growth were assessed in a mouse xenograft tumor model. RESULTS: MGDG showed dose- and time-dependent cytotoxicity, with half-maximal inhibitory concentrations (IC50) in PANC-1, BxPC-3, MIAPaCa-2 and AsPC-1 cells at 72 h of 25.6 ± 2.5, 26.9 ± 1.3, 18.5 ± 1.7, and 22.7 ± 1.9 µM, respectively. The colony forming assay revealed fewer MIAPaCa-2, BxPC-3 and AsPC-1 cell colonies upon treatment with both MGDG and radiation as compared to irradiation alone (P < 0.05). The combination of MGDG and radiation induced a higher proportion of apoptosis in MIAPaCa-2 cells; this effect was associated with increased mitochondrial release of cytochrome c and activation of cleaved poly (ADP-ribose) polymerase and caspase-3. DNA damage was detected and DNA repair mechanisms were more frequently impaired in cells receiving the combination treatment as compared to either one alone. Tumor growth was inhibited to a greater degree in mice treated by intratumoral injection of MGDG combined with irradiation as compared to either one alone (P < 0.05). CONCLUSIONS: This is the first report demonstrating that MGDG enhances the cytotoxicity of radiation to induce apoptosis of cancer cells in vitro and in vivo. Our findings indicate that this therapeutic combination can be an effective strategy for the treatment of pancreatic cancer.


Asunto(s)
Quimioradioterapia/métodos , Galactolípidos/farmacología , Neoplasias Pancreáticas/patología , Extractos Vegetales/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Spinacia oleracea , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Western Blotting , Línea Celular Tumoral , Ensayo Cometa , 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 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Sci Rep ; 6: 32977, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27596356

RESUMEN

Even today, 70 years after Hiroshima and accidents like in Chernobyl and Fukushima, we still have limited knowledge about the health effects of low dose rate (LDR) radiation. Despite their human relevance after occupational and accidental exposure, only few animal studies on the genotoxic effects of chronic LDR radiation have been performed. Selenium (Se) is involved in oxidative stress defence, protecting DNA and other biomolecules from reactive oxygen species (ROS). It is hypothesised that Se deficiency, as it occurs in several parts of the world, may aggravate harmful effects of ROS-inducing stressors such as ionising radiation. We performed a study in the newly established LDR-facility Figaro on the combined effects of Se deprivation and LDR γ exposure in DNA repair knockout mice (Ogg1(-/-)) and control animals (Ogg1(+/-)). Genotoxic effects were seen after continuous radiation (1.4 mGy/h) for 45 days. Chromosomal damage (micronucleus), phenotypic mutations (Pig-a gene mutation of RBC(CD24-)) and DNA lesions (single strand breaks/alkali labile sites) were significantly increased in blood cells of irradiated animals, covering three types of genotoxic activity. This study demonstrates that chronic LDR γ radiation is genotoxic in an exposure scenario realistic for humans, supporting the hypothesis that even LDR γ radiation may induce cancer.


Asunto(s)
Células Sanguíneas/efectos de la radiación , Daño del ADN/efectos de la radiación , ADN Glicosilasas/fisiología , Reparación del ADN/efectos de la radiación , Rayos gamma/efectos adversos , Animales , ADN Glicosilasas/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Estrés Oxidativo/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Selenio/deficiencia
12.
Nutr Cancer ; 68(8): 1349-1356, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27673354

RESUMEN

Ionizing radiation (IR) is a well-documented human carcinogen. The increased use of IR in medical procedures has doubled the annual radiation dose and may increase cancer risk. Genomic instability is an intermediate lesion in IR-induced cancer. We examined whether pomegranate extract (PE) suppresses genomic instability induced by x-rays. Mice were treated orally with PE and exposed to an x-ray dose of 2 Gy. PE intake suppressed x-ray-induced DNA double-strand breaks (DSBs) in peripheral blood and chromosomal damage in bone marrow. We hypothesized that PE-mediated protection against x-ray-induced damage may be due to the upregulation of DSB repair and antioxidant enzymes and/or increase in glutathione (GSH) levels. We found that expression of DSB repair genes was not altered (Nbs1 and Rad50) or was reduced (Mre11, DNA-PKcs, Ku80, Rad51, Rad52 and Brca2) in the liver of PE-treated mice. Likewise, mRNA levels of antioxidant enzymes were reduced (Gpx1, Cat, and Sod2) or were not altered (HO-1 and Sod1) as a function of PE treatment. In contrast, PE-treated mice with and without IR exposure displayed higher hepatic GSH concentrations than controls. Thus, ingestion of pomegranate polyphenols is associated with inhibition of x-ray-induced genomic instability and elevated GSH, which may reduce cancer risk.


Asunto(s)
Reparación del ADN/genética , Inestabilidad Genómica/efectos de la radiación , Lythraceae , Rayos X/efectos adversos , Animales , Antioxidantes/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de la radiación , Enzimas/metabolismo , Glutatión/metabolismo , Histonas/metabolismo , Hígado/metabolismo , Hígado/efectos de la radiación , Ratones Endogámicos C57BL , Extractos Vegetales/farmacología , Protectores contra Radiación/farmacología , Radiografía/efectos adversos
13.
Cell Death Dis ; 7: e2180, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27054335

RESUMEN

Cancer is as much an epigenetic disease as it is a genetic disease, and epigenetic alterations in cancer often serve as potent surrogates for genetic mutations. Because the epigenetic factors involved in the DNA damage response are regulated by multiple elements, therapies to target specific components of the epigenetic machinery can be inefficient. In contrast, therapies aimed at inhibiting the methionine cycle can indirectly inhibit both DNA and protein methylation, and the wide variety of genes and pathways that are affected by these methylations make this global strategy very attractive. In the present study, we propose an adjuvant therapy that targets the epigenetics of the DNA damage response in breast cancer cells and that results in efficient apoptosis and a reduction in distant metastases in vivo. We observed that a combined therapy designed to uncouple adenosine metabolism using dipyridamole in the presence of a new synthetic antifolate, 3-O-(3,4,5-trimethoxybenzoyl)-(-)-catechin, simultaneously and efficiently blocked both the folic cycle and the methionine cycle in breast cancer cells and sensitized these cells to radiotherapy. The treatment impeded the recruitment of 53BP1 and BRCA1 to the chromatin regions flanking DNA double-strand breaks and thereby avoided the DNA damage responses in breast cancer cells that were exposed to ionizing radiation. In addition, this hypomethylating therapy was also efficient in reducing the self-renewal capability of breast cancer-initiating cells and induced reversion of mesenchymal phenotypes in breast cancer cells.


Asunto(s)
Reparación del ADN , Epigénesis Genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Cromatina/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Dipiridamol/metabolismo , Femenino , Antagonistas del Ácido Fólico/farmacología , Histonas/metabolismo , Humanos , Células MCF-7 , Metilación/efectos de los fármacos , Metilación/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
14.
Toxicol In Vitro ; 30(1 Pt B): 552-60, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26362510

RESUMEN

This study aimed to compare the cell stress effects of low and high uranium concentrations and relate them to its localization, precipitate formation, and exposure time. The time-course analysis shows that uranium appears in cell nuclei as a soluble form within 5 min of exposure, and quickly induces expression of antioxidant and DNA repair genes. On the other hand, precipitate formations began at the very beginning of exposure at the 300-µM concentration, but took longer to appear at lower concentrations. Adaptive response might occur at low concentrations but are overwhelmed at high concentrations, especially when uranium precipitates are abundant.


Asunto(s)
Núcleo Celular/efectos de la radiación , Estrés Fisiológico/efectos de la radiación , Uranio/toxicidad , Apoptosis/efectos de la radiación , Reparación del ADN/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Células Hep G2 , Humanos , Estrés Oxidativo/efectos de la radiación , Uranio/farmacocinética
15.
Cancer Res ; 75(8): 1760-9, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25712125

RESUMEN

Glioma stem-like cells (GSC) are a subpopulation of cells in tumors that are believed to mediate self-renewal and relapse in glioblastoma (GBM), the most deadly form of primary brain cancer. In radiation oncology, hyperthermia is known to radiosensitize cells, and it is reemerging as a treatment option for patients with GBM. In this study, we investigated the mechanisms of hyperthermic radiosensitization in GSCs by a phospho-kinase array that revealed the survival kinase AKT as a critical sensitization determinant. GSCs treated with radiation alone exhibited increased AKT activation, but the addition of hyperthermia before radiotherapy reduced AKT activation and impaired GSC proliferation. Introduction of constitutively active AKT in GSCs compromised hyperthermic radiosensitization. Pharmacologic inhibition of PI3K further enhanced the radiosensitizing effects of hyperthermia. In a preclinical orthotopic transplant model of human GBM, thermoradiotherapy reduced pS6 levels, delayed tumor growth, and extended animal survival. Together, our results offer a preclinical proof-of-concept for further evaluation of combined hyperthermia and radiation for GBM treatment.


Asunto(s)
Glioma/terapia , Hipertermia Inducida , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Proteína Oncogénica v-akt/antagonistas & inhibidores , Tolerancia a Radiación , Animales , Muerte Celular/efectos de la radiación , Proliferación Celular/genética , Proliferación Celular/efectos de la radiación , Células Cultivadas , Terapia Combinada , Reparación del ADN/efectos de la radiación , Glioma/genética , Glioma/patología , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/patología , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/metabolismo , Tolerancia a Radiación/genética , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación
16.
PLoS One ; 10(2): e0118819, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25693136

RESUMEN

Bisphenol A (BPA) is a biologically active industrial chemical used in production of consumer products. BPA has become a target of intense public scrutiny following concerns about its association with human diseases such as obesity, diabetes, reproductive disorders, and cancer. Recent studies link BPA with the generation of reactive oxygen species, and base excision repair (BER) is responsible for removing oxidatively induced DNA lesions. Yet, the relationship between BPA and BER has yet to be examined. Further, the ubiquitous nature of BPA allows continuous exposure of the human genome concurrent with the normal endogenous and exogenous insults to the genome, and this co-exposure may impact the DNA damage response and repair. To determine the effect of BPA exposure on base excision repair of oxidatively induced DNA damage, cells compromised in double-strand break repair were treated with BPA alone or co-exposed with either potassium bromate (KBrO3) or laser irradiation as oxidative damaging agents. In experiments with KBrO3, co-treatment with BPA partially reversed the KBrO3-induced cytotoxicity observed in these cells, and this was coincident with an increase in guanine base lesions in genomic DNA. The improvement in cell survival and the increase in oxidatively induced DNA base lesions were reminiscent of previous results with alkyl adenine DNA glycosylase-deficient cells, suggesting that BPA may prevent initiation of repair of oxidized base lesions. With laser irradiation-induced DNA damage, treatment with BPA suppressed DNA repair as revealed by several indicators. These results are consistent with the hypothesis that BPA can induce a suppression of oxidized base lesion DNA repair by the base excision repair pathway.


Asunto(s)
Compuestos de Bencidrilo/efectos adversos , Bromatos/farmacología , Daño del ADN/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Guanina/metabolismo , Fenoles/efectos adversos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN/efectos de los fármacos , ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Fibroblastos/citología , Fibroblastos/efectos de la radiación , Terapia por Luz de Baja Intensidad/efectos adversos , Ratones , Estrés Oxidativo/efectos de los fármacos
17.
J Pineal Res ; 58(2): 189-97, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25623566

RESUMEN

Radiation and adjuvant endocrine therapy are nowadays considered a standard treatment option after surgery in breast cancer. Melatonin exerts oncostatic actions on human breast cancer cells. In the current study, we investigated the effects of a combination of radiotherapy and melatonin on human breast cancer cells. Melatonin (1 mm, 10 µm and 1 nm) significantly inhibited the proliferation of MCF-7 cells. Radiation alone inhibited the MCF-7 cell proliferation in a dose-dependent manner. Pretreatment of breast cancer cells with melatonin 1 wk before radiation led to a significantly greater decrease of MCF-7 cell proliferation compared with radiation alone. Melatonin pretreatment before radiation also decreased G2 -M phase arrest compared with irradiation alone, with a higher percentage of cells in the G0 -G1 phase and a lower percentage of cells in S phase. Radiation alone diminished RAD51 and DNA-protein kinase (PKcs) mRNA expression, two main proteins involved in double-strand DNA break repair. Treatment with melatonin for 7 days before radiation led to a significantly greater decrease in RAD51 and DNA-PKcs mRNA expression compared with radiation alone. Our findings suggest that melatonin pretreatment before radiation sensitizes breast cancer cells to the ionizing effects of radiation by decreasing cell proliferation, inducing cell cycle arrest and downregulating proteins involved in double-strand DNA break repair. These findings may have implications for designing clinical trials using melatonin and radiotherapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Melatonina/farmacología , Radiación Ionizante , Neoplasias de la Mama/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Roturas del ADN de Doble Cadena , Femenino , Humanos , Células MCF-7 , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo
18.
Free Radic Biol Med ; 74: 200-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24992837

RESUMEN

We previously showed that 5 mM ascorbate radiosensitized early passage radioresistant glioblastoma multiforme (GBM) cells derived from one patient tumor. Here we investigate the sensitivity of a panel of cell lines to 5 mM ascorbate and 6 Gy ionizing radiation, made up of three primary human GBM cells, three GBM cell lines, a human glial cell line, and primary human vascular endothelial cells. The response of different cells lines to ascorbate and/or radiation was determined by measuring viability, colony-forming ability, generation and repair of double-stranded DNA breaks (DSBs), cell cycle progression, antioxidant capacity and generation of reactive oxygen species. Individually, radiation and ascorbate both decreased viability and clonogenicity by inducing DNA damage, but had differential effects on cell cycle progression. Radiation led to G2/M arrest in most cells whereas ascorbate caused accumulation in S phase, which was moderately associated with poor DSB repair. While high dose ascorbate radiosensitized all cell lines in clonogenic assays, the sensitivity to radiation, high dose ascorbate, and combined treatment varied between cell lines. Normal glial cells were similar to GBM cells with respect to free radical scavenging potential and effect of treatment on DNA damage and repair, viability, and clonogenicity. Both GBM cells and normal cells coped equally poorly with oxidative stress caused by radiation and/or high dose ascorbate, dependent primarily on their antioxidant and DSB repair capacity.


Asunto(s)
Ácido Ascórbico/farmacología , Quimioradioterapia/métodos , Células Endoteliales/fisiología , Glioblastoma/terapia , Neuroglía/fisiología , Fármacos Sensibilizantes a Radiaciones/farmacología , Línea Celular Tumoral , Supervivencia Celular , Ensayo de Unidades Formadoras de Colonias , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Glioblastoma/patología , Humanos , Neuroglía/efectos de los fármacos , Neuroglía/efectos de la radiación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo
19.
Planta ; 239(5): 1101-11, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24563249

RESUMEN

Screening of 40,000 Arabidopsis FOX (Full-length cDNA Over-eXpressor gene hunting system) lines expressing rice full-length cDNAs brings us to identify four cadmium (Cd)-tolerant lines, one of which carried OsREX1-S as a transgene. OsREX1-S shows the highest levels of identity to Chlamydomonas reinhardtii REX1-S (referred to as CrREX1-S, in which REX denotes Required for Excision) and to yeast and human TFB5s (RNA polymerase II transcription factor B5), both of which are components of the general transcription and DNA repair factor, TFIIH. Transient expression of OsREX1-S consistently localized the protein to the nucleus of onion cells. The newly generated transgenic Arabidopsis plants expressing OsREX1-S reproducibly displayed enhanced Cd tolerance, confirming that the Cd-tolerance of the initial identified line was conferred solely by OsREX1-S expression. Furthermore, transgenic Arabidopsis plants expressing OsREX1-S exhibited ultraviolet-B (UVB) tolerance by reducing the amounts of cyclobutane pyrimidine dimers produced by UVB radiation. Moreover, those transgenic OsREX1-S Arabidopsis plants became resistant to bleomycin (an inducer of DNA strand break) and mitomycin C (DNA intercalating activity), compared to wild type. Our results indicate that OsREX1-S renders host plants tolerant to Cd, UVB radiation, bleomycin and mitomycin C through the enhanced DNA excision repair.


Asunto(s)
Cadmio/toxicidad , Daño del ADN , Reparación del ADN/efectos de la radiación , Oryza/metabolismo , Células Vegetales/efectos de la radiación , Proteínas de Plantas/metabolismo , Factor de Transcripción TFIIH/metabolismo , Rayos Ultravioleta , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/efectos de la radiación , Secuencia de Aminoácidos , Arabidopsis/efectos de los fármacos , Arabidopsis/genética , Arabidopsis/fisiología , Arabidopsis/efectos de la radiación , Bleomicina , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/efectos de la radiación , Reparación del ADN/efectos de los fármacos , ADN Complementario/genética , Humanos , Mitomicina , Datos de Secuencia Molecular , Cebollas/citología , Oryza/efectos de los fármacos , Oryza/efectos de la radiación , Fenotipo , Células Vegetales/efectos de los fármacos , Proteínas de Plantas/química , Plantas Modificadas Genéticamente , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/efectos de la radiación , Dímeros de Pirimidina/metabolismo , Saccharomyces cerevisiae/metabolismo , Plantones/efectos de los fármacos , Plantones/efectos de la radiación , Homología de Secuencia de Aminoácido , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/efectos de la radiación
20.
Environ Mol Mutagen ; 55(5): 436-48, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24500925

RESUMEN

Radiation exposure is a serious threat to biomolecules, particularly DNA, proteins and lipids. Various exogenous substances have been reported to protect these biomolecules. In this study we explored the effect of pre-treatment with G-002M, a mixture of three active derivatives isolated from the rhizomes of Podophyllum hexandrum, on DNA damage response in irradiated human blood leukocytes. Blood was collected from healthy male volunteers, preincubated with G-002M and then irradiated with various doses of radiation. Samples were analyzed using flow cytometry to quantify DNA double strand break (DSB) biomarkers including γ-H2AX, P53BP1 and levels of ligase IV. Blood samples were irradiated in vitro and processed to determine time and dose-dependent kinetics. Semiquantitative RT-PCR was performed at various time points to measure gene expression of DNA-PKcs, Ku80, ATM, and 53BP1; each of these genes is involved in DNA repair signaling. Pre-treatment of blood with G-002M resulted in reduction of γ-H2AX and P53BP1 biomarkers levels and elevated ligase IV levels relative to non-G-002M-treated irradiated cells. These results confirm suppression in radiation-induced DNA DSBs. Samples pre-treated with G-002M and then irradiated also showed significant up-regulation of DNA-PKcs and Ku80 and downregulation of ATM and 53BP1 gene expressions, suggesting that G-002M plays a protective role against DNA damage. The protective effect of G-002M may be due to its ability to scavange radiation-induced free radicals or assist in DNA repair. Further studies are needed to decipher the role of G-002M on signaling molecules involved in radiation-induced DNA damage repair pathways.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Flavonoides/farmacología , Rayos gamma/efectos adversos , Leucocitos/efectos de los fármacos , Podophyllum/química , Protectores contra Radiación/farmacología , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Berberidaceae , Células Cultivadas , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Flavonoides/química , Histonas/genética , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Autoantígeno Ku , Leucocitos/metabolismo , Leucocitos/efectos de la radiación , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Linfocitos/efectos de la radiación , Masculino , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/efectos de la radiación , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 de Unión al Supresor Tumoral P53
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA