Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Radiother Oncol ; 129(1): 84-94, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29685705

RESUMEN

BACKGROUND AND PURPOSE: Previously, we found genistein could sensitize cancer cells to low linear energy transfer (LET) X-rays via inhibiting DNA-PKcs activities. Especially, high-LET heavy ion produces more DNA double strand breaks (DSBs) than low-LET radiation. Thus, the study was designed to investigate the detailed molecular mechanisms of genistein on sensitizing cancer cells to heavy ions. MATERIALS AND METHODS: Human glioblastoma (GBM) cell lines with or without genistein pre-treatment were irradiated with high-LET carbon ions. Cell survival was determined with colony formation assay. DNA DSBs were evaluated by means of detecting γ-H2AX foci and immuno-blotting DSB repair proteins, cell apoptosis was detected using Annexin V and PI staining. The interaction of genistein with DNA-PKcs activation site was estimated by molecular docking in the autodock software. RESULTS: Genistein sensitized DNA-PKcs proficient GBM cells to high-LET carbon ions via delaying the clearance of γ-H2AX foci. Genistein was physically bound to DNA-PKcs and functionally inhibited the phosphorylation of DNA-PKcs. Consequently, the non-homologous end joining (NHEJ) repair of DSBs was inhibited and the homologous recombination (HR) repair was delayed by genistein, thereby leading to an increase in apoptosis in DNA-PKcs proficient GBM cells after irradiation. CONCLUSION: Our study demonstrated that genistein holds promise as a radiosensitizer for enhancing the efficacy of carbon ion radiotherapy against DNA-PKcs proficient GBM via inhibiting DNA-PKcs phosphorylation and subsequently repressing NHEJ and delaying HR repair pathways.


Asunto(s)
Carbono/farmacología , Genisteína/farmacología , Radioterapia de Iones Pesados/métodos , Iones/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Anticarcinógenos/farmacología , Supervivencia Celular/efectos de la radiación , ADN , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/efectos de los fármacos , Glioblastoma/radioterapia , Humanos , Transferencia Lineal de Energía , Simulación del Acoplamiento Molecular , Proteínas Nucleares/efectos de los fármacos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Células Tumorales Cultivadas , Rayos X
2.
DNA Repair (Amst) ; 52: 49-58, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28258841

RESUMEN

Numerous G-protein coupled receptors have been reported to enhance cancer cell survival and resistance to clinically used chemotherapeutics. Recently, hydroxycarboxylic acid receptor 1 (HCAR1) was shown to drive lactate-dependent enhancement of cell survival and metastasis in pancreatic and breast cancers. Furthermore, our previous study confirmed the involvement of HCAR1 in lactate-related enhancement of DNA repair in cervical cancer cells. In the present study, we examined the possible mechanisms of HCAR1-mediated enhancement of DNA repair capacity. We observed that the HCAR1 agonist dihydroxybenzoic acid (DHBA) up-regulated BRCA1 (breast cancer type 1 susceptibility protein) and NBS1 (Nijmegen breakage syndrome 1) expression in HeLa cells. Moreover, HCAR1 silencing decreased mRNA and protein levels of BRCA1 by 30% and 20%, respectively. Immunocytochemical analyses of BRCA1, nibrin and DNA-PKcs indicated an increased accumulation of these proteins in cell nuclei after DHBA stimulation. Subsequently, these changes in the DNA repair protein levels translated into an enhanced DNA repair rate after doxorubicin treatment, as shown by γ-H2AX and comet assay experiments. In contrast, the down-regulation of HCAR1 decreased the efficiency of DNA repair. Finally, we observed the abrogation of DHBA-driven BRCA1 protein up-regulation and enhanced DNA repair following the preincubation of cells with the PKC inhibitor Gö6983. Taken together, our data indicate that lactate receptor/HCAR1 expression in cervical carcinoma cells may contribute to the modulation of cellular DNA repair mechanisms.


Asunto(s)
Proteína BRCA1/genética , Proteínas de Ciclo Celular/genética , Reparación del ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/genética , Hidroxibenzoatos/farmacología , Proteínas Nucleares/genética , Receptores Acoplados a Proteínas G/agonistas , Neoplasias del Cuello Uterino/metabolismo , Proteína BRCA1/efectos de los fármacos , Proteínas de Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayo Cometa , Roturas del ADN de Doble Cadena , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/metabolismo , Proteína Quinasa Activada por ADN/efectos de los fármacos , Doxorrubicina/toxicidad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Cinética , Proteínas Nucleares/efectos de los fármacos , Transducción de Señal , Neoplasias del Cuello Uterino/genética
3.
Cell Biol Int ; 41(6): 622-629, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28328082

RESUMEN

Deregulation of microRNAs (miRNAs) has been implicated in drug resistance in various types of cancers, including malignant melanoma (MM). MiR-488-3p has been reported as a tumor suppressor in several cancers. However, the exact expression patterns of miR-488-3p and the precise molecular mechanisms underlying its role in MM remain largely unknown and require further investigation. In this study, we demonstrated that miR-488-3p is significantly downregulated in MM clinical specimens and cell lines. Ectopic expression of miR-488-3p resulted in markedly increased drug sensitivity of MM cells in vitro and in vivo. The DNA-activated, catalytic polypeptide (PRKDC), which encodes DNA-dependent protein kinase catalytic subunit (DNA-PKcs), was identified as a direct target of miR-488-3p using luciferase reporter assays, qRT-PCR, and western blotting analyses. PRKDC knockdown by small interfering RNA (siRNA) alone promoted sensitivity of MM cells to cisplatin (DDP) while overexpression of PRKDC partially rescued the miR-488-3p-mediated acceleration of sensitivity to DDP in MM cells. Taken together, our results indicate that miR-488-3p serves as a drug resistance sensitizer in MM, supporting its potential as a promising therapeutic candidate.


Asunto(s)
Proteína Quinasa Activada por ADN/efectos de los fármacos , MicroARNs/fisiología , MicroARNs/uso terapéutico , Proteínas Nucleares/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Cisplatino/farmacología , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen/métodos , Humanos , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Interferente Pequeño
4.
Int J Radiat Oncol Biol Phys ; 83(1): e43-52, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22516386

RESUMEN

PURPOSE: We compared radiosensitivity of brain tumor stem cells (BTSCs) with matched nonstem glioma cells, and determined whether gefitinib enhanced BTSC radiosensitivity by inhibiting epidermal growth factor receptor (EGFR)-Akt-DNA-dependent protein kinase (DNA-PK) signaling, followed by enhanced DNA double-stand breaks (DSBs) and inhibition of DSB repair. METHODS AND MATERIALS: Radiosensitivity of stem-like gliomaspheres and nonstem glioma cells (obtained at patient neurosurgical resection) were evaluated by clonogenic assays, γ-H(2)AX immunostaining and cell cycle distribution. Survival of irradiated and nonirradiated NOD-SCID mice intracranially implanted with stem-like gliomaspheres were monitored. Glioma cells treated with gefitinib, irradiation, or both were assayed for clonogenic survival, γ-H(2)AX immunostaining, DNA-PKcs expression, and phosphorylation of EGFR and Akt. RESULTS: Stem-like gliomaspheres displayed BTSC characteristics of self-renewal; differentiation into lineages of neurons, oligodendrocytes, and astrocytes; and initiation of glioma growth in NOD-SCID mice. Irradiation dose-dependently reduced clonogenic survival, induced G(2)/M arrest and increased γ-H(2)AX immunostaining of nonstem glioma cells, but not stem-like gliomaspheres. There was no difference in survival of irradiated and nonirradiated mice implanted with stem-like gliomaspheres. The addition of gefitinib significantly inhibited clonogenic survival, increased γ-H(2)AX immunostaining, and reduced DNA-PKcs expression of irradiated stem-like gliomaspheres, without affecting irradiated-nonstem glioma cells. Gefitinib alone, and when combined with irradiation, inhibited phosphorylation of EGFR (Y1068 and Y1045) and Akt (S473) in stem-like gliomaspheres. In nonstem glioma cells, gefitinib alone inhibited EGFR Y1068 phosphorylation, with further inhibition by combined gefitinib and irradiation. CONCLUSIONS: Stem-like gliomaspheres are resistant to irradiation-induced cytotoxicity, G(2)/M arrest, and DNA DSBs, compared with nonstem glioma cells. Gefitinib differentially enhances radiosensitivity of stem-like gliomaspheres by reducing EGFR-Akt activation and DNA-PKcs expression, accompanied by enhanced irradiation-induced DNA DSBs and inhibition of DSB repair.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/radioterapia , Roturas del ADN de Doble Cadena/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Glioma/radioterapia , Células Madre Neoplásicas/efectos de la radiación , Quinazolinas/farmacología , Tolerancia a Radiación/efectos de los fármacos , Animales , Neoplasias Encefálicas/patología , Proteína Quinasa Activada por ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/metabolismo , Receptores ErbB/metabolismo , Gefitinib , Glioma/patología , Histonas/efectos de los fármacos , Histonas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/patología , Neuroglía/patología , Neuroglía/efectos de la radiación , Fosforilación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Ensayo de Tumor de Célula Madre/métodos
5.
Br J Haematol ; 152(1): 61-71, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21083655

RESUMEN

Defects in the DNA damage response pathway [e.g. del(17p)] are associated with drug-resistant B-cell chronic lymphocytic leukaemia (CLL). We previously demonstrated that over-expression of DNA-dependent protein kinase (DNA-PK) correlates with chemo-resistance and that inhibition of DNA-PK sensitizes CLL cells to chemotherapeutics. Here, we investigated expression of DNA-PK and other proteins that impact on drug resistance, and evaluated the effects of a DNA-PK inhibitor (NU7441) on mitoxantrone-induced cytotoxicity in CLL cells. NU7441 sensitized cells from 42/49 CLL samples to mitoxantrone, with sensitization ranging from 2- to 200-fold Co-culture of CLL cells in conditioned stromal medium increased chemoresistance but did not reduce sensitization by NU7441. Mitoxantrone treatment induced γH2AX foci and NU7441 increased their longevity (24 h). NU7441 prevented mitoxantrone-induced autophosphorylation of the DNA-PK catalytic subunit (DNA-PKcs) at Ser 2056, confirming that DNA-PK participates in repair of mitoxantrone-induced DNA damage. del(17p) cases were more resistant to mitoxantrone than del(13q) cases, but were resensitized (7-16 fold) by co-incubation with NU7441. Expression of DNA-PKcs, Ku80, P-glycoprotein and topoisomerase IIß were significantly higher in del(17p) cases. PRKDC mRNA levels correlated with DNA-PKcs protein expression, which predicted shorter survival. These data confirm the potential of DNA-PK as a therapeutic target in poor prognosis CLL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Leucemia Linfocítica Crónica de Células B/patología , Muerte Celular/efectos de los fármacos , Cromonas/farmacología , Medios de Cultivo Condicionados , Reparación del ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Humanos , Leucemia Linfocítica Crónica de Células B/enzimología , Leucemia Linfocítica Crónica de Células B/genética , Mitoxantrona/farmacología , Morfolinas/farmacología , Proteínas de Neoplasias/metabolismo , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas
6.
Artículo en Inglés | MEDLINE | ID: mdl-22238502

RESUMEN

The study assessed the effect of Chinese herbs of Shenghe Powder (SHP) on the repair capacity of gamma-radiation-induced DNA damage in rat glioma cells (C6) compared with normal human astrocytes (NHA). C6 and NHA Cells treated with SHP and irradiated with 2Gy of gamma radiation. Cells growth inhibition were analysed by MTT assay, DNA damage and repair were evaluated using phosphorylated histone H2AX (γH2AX) at the appointed time. Apoptosis was observed by flow cytometry, and the expression of DNA-dependent protein kinase (DNA-PK) and surviving proteins were assessed by Western blot analysis. SHP depressed the radiation-induced DNA double-strand break and enhanced the DNA repair capacity in NHA, which correlated with promotion of DNA-PK phosphorylation. In contrast, SHP enhanced radiosensitivity of C6 cells, the pre-treatment with SHP resulted in reduced numbers of γH2AX foci in irradiated C6 cells, and decreased the expression of DNA-PK and survivn(P<0.005). It significant effect on inhibition of C6 cell proliferation and induced C6 cells apoptosis in a time-depdendent manner than radiation alone (P<0.001). SHP showed a novel bidirectional function to improve the radioresistance of NHA and enhanced radiosensitivity of C6 cells. This implies that SHP can protect the NHA from radiant damage and enhanced the sensitivity of C6 cells to radiation, which could be attributed to the alteration of survivin DNA-PK in DNA repair processes.


Asunto(s)
Astrocitos/metabolismo , Reparación del ADN/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Glioma/radioterapia , Fitoterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Proteína Quinasa Activada por ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/efectos de la radiación , Rayos gamma , Glioma/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/efectos de la radiación , Ratas
7.
J Neurooncol ; 89(1): 27-35, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18415044

RESUMEN

OBJECTIVE: This study was designed to investigate the relationship between activities of DNA-dependent protein kinase (DNA-PK), its subunits Ku86/Ku70, and sensitivities to cisplatin in human glioma samples. METHODS: Thirty-six glioma samples from patients without prior treatment before neurosurgery were included in this study. The sensitivities to cisplatin as indicated by IC(50) (the inhibitory concentration leading to 50% cell death) were assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenytetrazolium (MTT) assay; activities of DNA-PK and Ku70/Ku86 were analyzed by SigmaTECT DNA-Dependent Protein Kinase Assay System and Ku70/Ku86 DNA Repair Kit, respectively. RESULTS: Sensitivities to cisplatin correlated with the activities of DNA-PK/Ku86, but not with the Ku70 or other clinical parameters such as age, sex of the patients, pathological gradings of the tumors, or tumor size. The levels of DNA-PK activities also associated with pathological grading and Ku86, but not with other clinical parameters. The tumors of the patients who failed to respond to cisplatin-based chemotherapy tended to display higher activity levels of DNA-PK and Ku86. Furthermore, platinum-based chemotherapy did not result in significant changes of DNA-PK/Ku activities in four matched samples before and after chemotherapy. CONCLUSION: Pretreatment determination of DNA-PK/Ku86 activities might be helpful in identifying patients who will actually benefit from platinum-based treatment.


Asunto(s)
Antígenos Nucleares/metabolismo , Neoplasias Encefálicas/enzimología , Cisplatino/farmacología , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Glioma/enzimología , Adulto , Antígenos Nucleares/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Bioensayo , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Cisplatino/uso terapéutico , Proteína Quinasa Activada por ADN/efectos de los fármacos , Proteínas de Unión al ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Glioma/tratamiento farmacológico , Humanos , Autoantígeno Ku , Masculino , Persona de Mediana Edad , Sales de Tetrazolio , Tiazoles , Células Tumorales Cultivadas
8.
Int J Oncol ; 29(5): 1167-72, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17016648

RESUMEN

Vanillin, a naturally occurring food component, has been reported to have anti-mutagenic and anti-metastatic potentials, and to inhibit DNA-PKcs activity. However, vanillin itself exhibits very weak antiproliferative activity. We explored the effects of bromovanin (6-bromine-5-hydroxy-4-methoxybenzaldehyde), a novel vanillin derivative, on survival and cell-cycle progression of human Jurkat leukemia cells. Treatment with >10 microM bromovanin significantly elicited apoptosis and G2/M arrest in Jurkat cells in a dose- and time-dependent manner. Bromovanin-induced DNA double-strand breaks (DSB) were demonstrated by means of comet assay as well as detection of phosphorylated H2AX, a sensitive indicator of DNA DSBs. Immuno-hybridization analysis revealed that the cleavage of procaspase-3 and DNA-PKcs occurred concurrently with bromovanin-induced apoptosis. Furthermore, phosphorylated Akt protein (Ser473), which is catalyzed by DNA-PKcs, as well as phosphorylated GSK3beta (a substrate of activated Akt), markedly decreased in bromovanin-treated Jurkat cells, suggesting that bromovanin leads to inactivation of Akt pathway via cleaving DNA-PKcs. These multiple effects, associated with the regimen of cancer therapeutic strategies, make bromovanin very appealing for future development as a novel anticancer drug.


Asunto(s)
Apoptosis , Benzaldehídos/química , Benzaldehídos/farmacología , Daño del ADN , Proteína Quinasa Activada por ADN/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Ciclo Celular/efectos de los fármacos , ADN/efectos de los fármacos , Humanos , Células Jurkat
9.
Pathol Biol (Paris) ; 54(4): 185-93, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16563661

RESUMEN

A line of investigation in the search for sensitizing tumor cells to chemotherapy or radiotherapy relies on the selection of DNA repair inhibitors. In the area of DNA repair mechanisms, DNA-dependent protein kinase (DNA-PK) represents a key complex. Indeed DNA-PK is involved in the non-homologous end joining (NHEJ) process that corresponds to the major activity responsible for cell survival after ionizing radiation or chemotherapeutic treatment producing DNA double strand breaks. DNA-PK belongs to the PI3-K related kinase family and specific inhibitors have been recently selected and evaluated as radio- and chemo-sensitizers. These drugs, along with other ways to inhibit the DSBs repair process, are presented and discussed.


Asunto(s)
Proteínas de Unión al Calcio/efectos de los fármacos , Daño del ADN , Reparación del ADN , Proteína Quinasa Activada por ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/efectos de la radiación , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Proteínas de Unión al Calcio/efectos de la radiación , Humanos
10.
Free Radic Biol Med ; 39(12): 1650-9, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16298690

RESUMEN

DNA-dependent protein kinase (DNA-PK) is activated in a two-step process whereby the Ku heterodimer first binds to the DNA double-strand breaks (dsbs) and then the DNA-PK catalytic subunit (cs) is recruited to form a repair complex. Oxidative stress is simultaneously generated along with DNA damage by ionizing radiation or chemotherapeutic agents whose impact on the DNA-PK activity has not previously been investigated. Here we show that the DNA damage-induced kinase activity of DNA-PK was modulated by oxidative stress, which was induced along with DNA dsbs in chlorambucil (Cbl)-exposed cells. Pretreatment with the antioxidants, 2(3)-t-butyl-4-hydroxyanisole or N-acetyl-l-cysteine enhanced the amount of DNA-PKcs phosphorylated at threonine 2609 (DNA-PK(pThr2609)) at the DNA dsbs and DNA-PK activity. Conversely, oxidative stress induced by l-buthionine (SR)-sulfoximine or glucose oxidase decreased the DNA-PK activity in Cbl-exposed cells. In addition, DNA-PK(pThr2609) was poorly detectable at the site of DNA dsbs, as shown by colocalization to DNA-end-binding pH2AX or p53BP1. There was no change in the protein levels of DNA-PKcs, Ku70, or Ku86. Data from these studies provide the first evidence that oxidative stress effects posttranslational modification and assembly of DNA-PK complex at DNA dsbs, and thereby repair of DNA dsbs.


Asunto(s)
Clorambucilo/farmacología , Proteína Quinasa Activada por ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Acetilcisteína/farmacología , Hidroxianisol Butilado/farmacología , Línea Celular Tumoral , Daño del ADN , Activación Enzimática/efectos de los fármacos , Humanos , Estrés Oxidativo/fisiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA