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










Base de datos
Intervalo de año de publicación
1.
Adv Anat Embryol Cell Biol ; 235: 81-83, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36525114

RESUMEN

The effects of ionizing radiation on centrosomes have been well documented and reviewed by Saladino et al. (2012) and are only briefly addressed here. These results showed that exposure of tumor cells to ionizing radiation causes centrosome overduplication and the formation of multipolar mitotic spindles, resulting in nuclear fragmentation and subsequent cell death (Sato et al. 2000). By using a variety of cell lines derived from different types of human solid tumors, it was shown that exposure to 10 Gy γ-radiation resulted in a substantial increase in cells containing an abnormally high number of aberrant centrosomes that formed multipolar spindles, resulting in imbalanced chromosome separation followed by mitotic cell death and formation of multi- or micronucleated cells.


Asunto(s)
Centrosoma , Huso Acromático , Humanos , Centrosoma/metabolismo , Centrosoma/patología , Centrosoma/efectos de la radiación , Huso Acromático/metabolismo , Huso Acromático/efectos de la radiación , Segregación Cromosómica , Muerte Celular , Línea Celular
2.
Biochem Biophys Res Commun ; 550: 84-91, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33689884

RESUMEN

The monopolar spindle 1 ((hMps1/TTK) is a serine/threonine kinase that plays an important role in spindle assembly checkpoint signaling. To explore the possible relationship between TTK inhibition and radiosensitivity, we examined whether TTK inhibition influences cellular susceptibility of radiation. And we further revealed its mechanisms. We found that the expression of TTK was obviously higher in liver cancer tissues compared to the normal liver tissues. Kaplan-Meier Plotter demonstrated that patients with low TTK expression levels had a longer overall survival than patients with high TTK expression levels. TTK inhibitor AZ3146 could simulated liver cancer cells to accumulate in the G2/M phase, which ultimately enhances DNA damage with more γ-H2AX foci and more apoptosis and necrosis induced by radiation, which prompted that TTK inhibition sensitized liver cancer cells to radiation. In addition, TTK inhibition altered cell-cycle progression and exacerbated centrosome abnormalities, resulting in enhanced mitotic catastrophe (MC) induced by radiation in a p21-mediated manner. In this study, we present evidences that the TTK inhibitor promotes the radiosensitivity of liver cancer cells through regulating cell cycle in p21-mediated manner in vitro, indicating that TTK inhibitor may be an attractive radiosensitizer for the patients with liver cancer.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/radioterapia , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Tolerancia a Radiación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Centrosoma/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/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 , Histonas/metabolismo , Humanos , Neoplasias Hepáticas/patología , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de la radiación , Necrosis/tratamiento farmacológico , Necrosis/radioterapia , Pronóstico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Análisis de Supervivencia
3.
Sci Transl Med ; 10(456)2018 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158153

RESUMEN

Cockayne syndrome (CS), a hereditary form of premature aging predominantly caused by mutations in the csb gene, affects multiple organs including skin where it manifests with hypersensitivity toward ultraviolet (UV) radiation and loss of subcutaneous fat. There is no curative treatment for CS, and its pathogenesis is only partially understood. Originally considered for its role in DNA repair, Cockayne syndrome group B (CSB) protein most likely serves additional functions. Using CSB-deficient human fibroblasts, Caenorhabditiselegans, and mice, we show that CSB promotes acetylation of α-tubulin and thereby regulates autophagy. At the organ level, chronic exposure of csbm/m mice to UVA radiation caused a severe skin phenotype with loss of subcutaneous fat, inflammation, and fibrosis. These changes in skin tissue were associated with an accumulation of autophagic/lysosomal proteins and reduced amounts of acetylated α-tubulin. At the cellular level, we found that CSB directly interacts with the histone deacetylase 6 (HDAC6) and the α-tubulin acetyltransferase MEC-17. Upon UVA irradiation, CSB is recruited to the centrosome where it colocalizes with dynein and HDAC6. Administration of the pan-HDAC inhibitor SAHA (suberoylanilide hydroxamic acid) enhanced α-tubulin acetylation, improved autophagic function in CSB-deficient models from all three species, and rescued the skin phenotype in csbm/m mice. HDAC inhibition may thus represent a therapeutic option for CS.


Asunto(s)
Autofagia/efectos de los fármacos , Síndrome de Cockayne/patología , Inhibidores de Histona Desacetilasas/farmacología , Lisosomas/metabolismo , Grasa Subcutánea/patología , Acetilación , Animales , Autofagia/efectos de la radiación , Proteínas Relacionadas con la Autofagia/metabolismo , Caenorhabditis elegans/efectos de los fármacos , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Centrosoma/efectos de la radiación , ADN Helicasas/deficiencia , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/deficiencia , Enzimas Reparadoras del ADN/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Lisosomas/efectos de los fármacos , Lisosomas/efectos de la radiación , Ratones , Fenotipo , Proteínas de Unión a Poli-ADP-Ribosa/deficiencia , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Piel/patología , Piel/efectos de la radiación , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/efectos de la radiación , Tubulina (Proteína)/metabolismo , Proteínas Ubiquitinadas/metabolismo , Rayos Ultravioleta , Vorinostat/farmacología
4.
Anticancer Res ; 38(6): 3393-3400, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29848688

RESUMEN

BACKGROUND/AIM: Owing to the frequent observation of centrosome amplification in human cancers, cancer cells have a unique mechanism to suppress detrimental multipolar division by clustering multiple centrosomes into two functional spindle poles, known as centrosome clustering. This study investigated whether inhibition of centrosome clustering enhances the radiation sensitivity of breast cancer cells. MATERIALS AND METHODS: In this study, inhibition of centrosome clustering was examined by using various centrosome-declustering agents and KIFC1 siRNA in three breast cancer cell lines and two normal fibroblast cell lines. The combination effect of radiation and centrosome declustering was evaluated by cell viability, clonogenic, immunofluorescence assay. RESULTS: This study showed that targeting centrosome clustering enhanced the efficacy of radiotherapy of breast cancer cells with less damage to normal cells. Ionizing radiation induced centrosome amplification in breast cancer cells, but not in normal fibroblast cells. Notably, we showed that centrosome declustering efficiently radiosensitized the centrosome-amplified breast cancer cells through induction of multipolar spindles but did not affect the viability of normal fibroblasts in response to irradiation. Furthermore, KIFC1 mediated the radiosensitivity of the centrosome-amplified breast cancer cells. CONCLUSION: Our data provided the first evidence that centrosome clustering is a tumor-selective target for the improvement of radiotherapy in breast cancer cells.


Asunto(s)
Centrosoma/efectos de la radiación , Fibroblastos/efectos de la radiación , Huso Acromático/efectos de la radiación , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , División Celular/genética , División Celular/efectos de la radiación , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Griseofulvina/farmacología , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Células MCF-7 , Fenantrenos/farmacología , Interferencia de ARN , Fármacos Sensibilizantes a Radiaciones/farmacología , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo
5.
Aging (Albany NY) ; 9(3): 665-686, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28245431

RESUMEN

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


Asunto(s)
Envejecimiento/patología , Neoplasias de la Mama/patología , Centrosoma/efectos de la radiación , Células Epiteliales/efectos de la radiación , Células Madre/efectos de la radiación , Adulto , Anciano , Células Cultivadas , Daño del ADN/efectos de la radiación , Susceptibilidad a Enfermedades , Células Epiteliales/patología , Femenino , Inestabilidad Genómica , Humanos , Persona de Mediana Edad , Células Madre/fisiología
6.
PLoS One ; 11(7): e0158236, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27367050

RESUMEN

The embryonic brain is one of the tissues most vulnerable to ionizing radiation. In this study, we showed that ionizing radiation induces apoptosis in the neural progenitors of the mouse cerebral cortex, and that the surviving progenitor cells subsequently develop a considerable amount of supernumerary centrosomes. When mouse embryos at Day 13.5 were exposed to γ-rays, brains sizes were reduced markedly in a dose-dependent manner, and these size reductions persisted until birth. Immunostaining with caspase-3 antibodies showed that apoptosis occurred in 35% and 40% of neural progenitor cells at 4 h after exposure to 1 and 2 Gy, respectively, and this was accompanied by a disruption of the apical layer in which mitotic spindles were positioned in unirradiated mice. At 24 h after 1 Gy irradiation, the apoptotic cells were completely eliminated and proliferation was restored to a level similar to that of unirradiated cells, but numerous spindles were localized outside the apical layer. Similarly, abnormal cytokinesis, which included multipolar division and centrosome clustering, was observed in 19% and 24% of the surviving neural progenitor cells at 48 h after irradiation with 1 and 2 Gy, respectively. Because these cytokinesis aberrations derived from excess centrosomes result in growth delay and mitotic catastrophe-mediated cell elimination, our findings suggest that, in addition to apoptosis at an early stage of radiation exposure, radiation-induced centrosome overduplication could contribute to the depletion of neural progenitors and thereby lead to microcephaly.


Asunto(s)
Centrosoma/patología , Centrosoma/efectos de la radiación , Microcefalia/patología , Células-Madre Neurales/patología , Células-Madre Neurales/efectos de la radiación , Traumatismos Experimentales por Radiación/patología , Animales , Animales Recién Nacidos , Apoptosis/efectos de la radiación , Proliferación Celular/efectos de la radiación , Corteza Cerebral/embriología , Corteza Cerebral/efectos de la radiación , Citocinesis/efectos de la radiación , Femenino , Ratones , Embarazo
7.
Cancer Res ; 76(10): 3036-44, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26896280

RESUMEN

FGF2 signaling in glioblastoma induces resistance to radiotherapy, so targeting FGF2/FGFR pathways might offer a rational strategy for tumor radiosensitization. To investigate this possibility, we evaluated a specific role for FGFR1 in glioblastoma radioresistance as modeled by U87 and LN18 glioblastomas in mouse xenograft models. Silencing FGFR1 decreased radioresistance in a manner associated with radiation-induced centrosome overduplication and mitotic cell death. Inhibiting PLCγ (PLCG1), a downstream effector signaling molecule for FGFR1, was sufficient to produce similar effects, arguing that PLCγ is an essential mediator of FGFR1-induced radioresistance. FGFR1 silencing also reduced expression of HIF1α, which in addition to its roles in hypoxic responses exerts an independent effect on radioresistance. Finally, FGFR1 silencing delayed the growth of irradiated tumor xenografts, in a manner that was associated with reduced HIF1α levels but not blood vessel alterations. Taken together, our results offer a preclinical proof of concept that FGFR1 targeting can degrade radioresistance in glioblastoma, a widespread problem in this tumor, prompting clinical investigations of the use of FGFR1 inhibitors for radiosensitization. Cancer Res; 76(10); 3036-44. ©2016 AACR.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fosfolipasa C gamma/metabolismo , Tolerancia a Radiación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Apoptosis/efectos de la radiación , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Proliferación Celular/efectos de la radiación , Centrosoma/efectos de la radiación , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Desnudos , Mitosis/efectos de la radiación , Fosfolipasa C gamma/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Oncogene ; 35(15): 2003-10, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-26165835

RESUMEN

Genotoxic stresses lead to centrosome amplification, a frequently-observed feature in cancer that may contribute to genome instability and to tumour cell invasion. Here we have explored how the centrosome controls DNA damage responses. For most of the cell cycle, centrosomes consist of two centrioles embedded in the proteinaceous pericentriolar material (PCM). Recent data indicate that the PCM is not an amorphous assembly of proteins, but actually a highly organised scaffold around the centrioles. The large coiled-coil protein, pericentrin, participates in PCM assembly and has been implicated in the control of DNA damage responses (DDRs) through its interactions with checkpoint kinase 1 (CHK1) and microcephalin (MCPH1). CHK1 is required for DNA damage-induced centrosome amplification, whereas MCPH1 deficiency greatly increases the amplification seen after DNA damage. We found that the PCM showed a marked expansion in volume and a noticeable change in higher-order organisation after ionising radiation treatment. PCM expansion was dependent on CHK1 kinase activity and was potentiated by MCPH1 deficiency. Furthermore, pericentrin deficiency or mutation of a separase cleavage site blocked DNA damage-induced PCM expansion. The extent of nuclear CHK1 activation after DNA damage reflected the level of PCM expansion, with a reduction in pericentrin-deficient or separase cleavage site mutant-expressing cells, and an increase in MCPH1-deficient cells that was suppressed by the loss of pericentrin. Deletion of the nuclear export signal of CHK1 led to its hyperphosphorylation after irradiation and reduced centrosome amplification. Deletion of the nuclear localisation signal led to low CHK1 activation and low centrosome amplification. From these data, we propose a feedback loop from the PCM to the nuclear DDR in which CHK1 regulates pericentrin-dependent PCM expansion to control its own activation.


Asunto(s)
Antígenos/fisiología , Proteínas de Ciclo Celular/fisiología , Centrosoma/fisiología , Daño del ADN , Proteínas del Tejido Nervioso/fisiología , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Animales , Antígenos/genética , Sitios de Unión , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Centriolos/metabolismo , Centriolos/ultraestructura , Centrosoma/efectos de la radiación , Centrosoma/ultraestructura , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Pollos , Activación Enzimática , Retroalimentación Fisiológica , Genes Reporteros , Mutación , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas Asociadas a Matriz Nuclear/genética , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo , Separasa/metabolismo
9.
Mol Biol Cell ; 26(25): 4607-17, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26466676

RESUMEN

Accumulating evidence suggests that mitochondrial dynamics is crucial for the maintenance of cellular quality control and function in response to various stresses. However, the role of mitochondrial dynamics in cellular responses to ionizing radiation (IR) is still largely unknown. In this study, we provide evidence that IR triggers mitochondrial fission mediated by the mitochondrial fission protein dynamin-related protein 1 (Drp1). We also show IR-induced mitotic catastrophe (MC), which is a type of cell death associated with defective mitosis, and aberrant centrosome amplification in mouse embryonic fibroblasts (MEFs). These are attenuated by genetic or pharmacological inhibition of Drp1. Whereas radiation-induced aberrant centrosome amplification and MC are suppressed by the inhibition of Plk1 and CDK2 in wild-type MEFs, the inhibition of these kinases is ineffective in Drp1-deficient MEFs. Furthermore, the cyclin B1 level after irradiation is significantly higher throughout the time course in Drp1-deficient MEFs than in wild-type MEFs, implying that Drp1 is involved in the regulation of cyclin B1 level. These findings strongly suggest that Drp1 plays an important role in determining the fate of cells after irradiation via the regulation of mitochondrial dynamics.


Asunto(s)
Dinaminas/genética , Dinámicas Mitocondriales/efectos de la radiación , Mitosis/efectos de la radiación , Animales , Apoptosis/efectos de la radiación , Centrosoma/efectos de la radiación , Ciclina B1/genética , Fibroblastos/efectos de la radiación , Ratones , Mitocondrias/genética , Mitocondrias/efectos de la radiación , Dinámicas Mitocondriales/genética , Rayos X
10.
Oncotarget ; 6(28): 25240-51, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26246475

RESUMEN

Clinical genetic testing of BRCA1 and BRCA2 is commonly performed to identify specific individuals at risk for breast and ovarian cancers who may benefit from prophylactic therapeutic interventions. Unfortunately, it is evident that deleterious BRCA1 alleles demonstrate variable penetrance and that many BRCA1 variants of unknown significance (VUS) exist. In order to further refine hereditary risks that may be associated with specific BRCA1 alleles, we performed gene targeting to establish an isogenic panel of immortalized human breast epithelial cells harboring eight clinically relevant BRCA1 alleles. Interestingly, BRCA1 mutations and VUS had distinct, quantifiable phenotypes relative to isogenic parental BRCA1 wild type cells and controls. Heterozygous cells with known deleterious BRCA1 mutations (185delAG, C61G and R71G) demonstrated consistent phenotypes in radiation sensitivity and genomic instability assays, but showed variability in other assays. Heterozygous BRCA1 VUS cells also demonstrated assay variability, with some VUS demonstrating phenotypes more consistent with deleterious alleles. Taken together, our data suggest that BRCA1 deleterious mutations and VUS can differ in their range of tested phenotypes, suggesting they might impart varying degrees of risk. These results demonstrate that functional isogenic modeling of BRCA1 alleles could aid in classifying BRCA1 mutations and VUS, and determining BRCA allele cancer risk.


Asunto(s)
Proteína BRCA1/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Modelos Genéticos , Mutación , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/efectos de la radiación , Línea Celular Transformada , Proliferación Celular/efectos de la radiación , Centrosoma/metabolismo , Centrosoma/efectos de la radiación , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Inestabilidad Genómica , Herencia , Heterocigoto , Humanos , Fenotipo , Tolerancia a Radiación , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Transfección
11.
Bioorg Med Chem ; 23(13): 3681-6, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25922180

RESUMEN

A series of indole analogs that are synthesized using the scaffold of a potent radiosensitizer, YTR107, were tested for their ability to alter the solubility of phosphorylated nucleophosmin 1 (pNPM1). NPM1 is critical for DNA double strand break (DSB) repair. In response to formation of DNA DSBs, phosphorylated T199 NPM1 binds to ubiquitinated chromatin, in a RNF8/RNF168-dependent manner, forming irradiation-induced foci (IRIF) that promote repair of DNA DSBs. A Western blot assay was developed using lead molecule, YTR107, for the purpose of screening newly synthesized molecules that target pNPM1 in irradiated cells. A colony formation assay was used to demonstrate the radiosensitization properties of the compounds. Compounds that enhanced the extractability of pNPM1 upon radiation treatment possessed radiosensitization properties.


Asunto(s)
Barbitúricos/farmacología , Western Blotting/métodos , Roturas del ADN de Doble Cadena/efectos de la radiación , Indoles/farmacología , Proteínas Nucleares/aislamiento & purificación , Fármacos Sensibilizantes a Radiaciones/farmacología , Línea Celular , Línea Celular Tumoral , Núcleo Celular/química , Núcleo Celular/metabolismo , Núcleo Celular/efectos de la radiación , Centrosoma/química , Centrosoma/metabolismo , Centrosoma/efectos de la radiación , Cromatina/química , Cromatina/metabolismo , Cromatina/efectos de la radiación , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Rayos gamma , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Fosforilación , Ensayo de Tumor de Célula Madre , Ubiquitina/genética , Ubiquitina/metabolismo
12.
Mol Cancer Ther ; 14(7): 1693-704, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25882311

RESUMEN

The aurora kinases constitute one family of serine/threonine kinases whose activity is essential for mitotic progression. The aurora kinases are frequently upregulated in human cancers and are associated with sensitivity to chemotherapy in certain ones. In the present study, we investigated whether aurora kinases could be a target to overcome radioresistance or enhance the radiosensitivity of lung cancer. For that purpose, we determined the therapeutic potential of daurinol, an investigational topoisomerase inhibitor, alone and in combination with radiation, by observing its effect on aurora kinases. Daurinol decreased cell viability and proliferation in human colon and lung cancer cells. Gene expression in daurinol-treated human colon cancer cells was evaluated using RNA microarray. The mRNA expression of 18 genes involved in the mitotic spindle check point, including aurora kinase A (AURKA) and aurora kinase B (AURKB), was decreased in daurinol-treated human colon cancer cells as compared with vehicle-treated cells. As expected, radiation increased expression levels of AURKA and AURKB. This increase was effectively attenuated by siRNAs against AURKA and AURKB, which suppressed cell growth and increased apoptosis under radiation. Furthermore, the expression of AURKA and AURKB was suppressed by daurinol in the presence or absence of radiation in colon and lung cancer cells. Daurinol alone or in combination with radiation decreased lung cancer growth in xenograft mouse models. Our data clearly confirm the antitumor and radiosensitizing activity of daurinol in human lung cancer cells through the inhibition of AURKA and AURKB.


Asunto(s)
Aurora Quinasa A/genética , Aurora Quinasa B/genética , Benzodioxoles/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Naftalenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Aurora Quinasa A/metabolismo , Aurora Quinasa B/metabolismo , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Centrosoma/efectos de los fármacos , Centrosoma/efectos de la radiación , Quimioradioterapia , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Inestabilidad Genómica/efectos de la radiación , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HCT116 , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/radioterapia , Ratones Endogámicos NOD , Ratones SCID , Microscopía Fluorescente , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
13.
Cell Biol Int ; 39(8): 943-53, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25808704

RESUMEN

Primary cilia act as physical-chemical sensors and their functions include the perception of the extracellular milieu, regulation of organogenesis, and cell polarity. In general, these cells are monociliated and the single cilium possesses diverse receptors and channels which are involved in morphogenesis and growth signaling, and are, therefore, important for cell proliferation and differentiation. In this study, we used an in vitro model of C2C12 myoblasts to evaluate the effect of DNA damage induced by gamma ionizing radiation on primary cilia incidence. A significantly higher number of ciliated cells were observed after 1 day post-irradiation with 2-20 Gy when compared with non-irradiated cells. After 3 days post-irradiation, the cilia incidence in cells had decreased slightly when treated with 2, 6, and 10 Gy, although an increase in incidence rate was observed in cells treated with 20 Gy. Multi-ciliated cells were also detected in myoblasts irradiated with 10 and 20 Gy but not in non-irradiated cells or after low irradiation (2-6 Gy). Irradiation also caused a dose-dependent decrease in cell viability and proliferation and corresponding cell cycle arrest. Furthermore, an activation of caspases 3/7, 8, and 9 was observed after higher radiation (10 and 20 Gy) with increased apoptosis. Together, our results show that irradiation by gamma rays promotes myoblast ciliogenesis, with pronounced effects observed after 3 days post-irradiation. We conclude that irradiation doses of 10 and 20 Gy are sufficient to induce cell death and are responsible for the formation of multiple cilia originating from multiple basal bodies.


Asunto(s)
Cilios/efectos de la radiación , Mioblastos/efectos de los fármacos , Apoptosis/efectos de la radiación , Caspasas/metabolismo , Diferenciación Celular/efectos de la radiación , Línea Celular , Proliferación Celular/efectos de la radiación , Centrosoma/efectos de la radiación , Roturas del ADN de Doble Cadena , Daño del ADN , Relación Dosis-Respuesta en la Radiación , Humanos , Mioblastos/citología , Mioblastos/metabolismo , Radiación Ionizante
14.
Int J Radiat Biol ; 91(1): 99-111, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25219679

RESUMEN

PURPOSE: To study the kinetics of gene expression alterations following radiation exposure of isogenic HCT116 p53 +/+ and HCT116 p53-/- cell lines. MATERIALS AND METHODS: Cells were exposed to 5 Gy of irradiation (Cs-137) and genome-wide temporal expression analysis using Illumina bead chip arrays was performed. Signalling pathways were explored using Metacore (Genego). Biological responses including cell cycle checkpoint activation, centrosome amplification and senescence induction were analyzed. RESULTS: Significant differences in the radiation response were observed between the p53+/+ and the p53-/- cell lines. In p53+/+ cells concurrent G1- and G2-arrests were activated followed by senescence induction. Increased expression of genes associated with senescence, senescence associated secretory phenotype (SASP) and repression of genes essential for G2-M transition were detected. P53-/- cells arrested mainly in G2 followed by centrosome amplification, mitotic slippage and a subsequent increase of polyploid cells. Furthermore, changes in expression correlated well with these signs of mitotic catastrophe. CONCLUSIONS: The presence or absence of p53 triggers different signalling cascades with different endpoints. Elucidating these differences is important as it enables improvement of radiation treatment and could be used to develop new combination treatments with specific inhibitors of key regulators of these cell death modalities.


Asunto(s)
Neoplasias Colorrectales/patología , Daño del ADN , Perfilación de la Expresión Génica , Genómica , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Puntos de Control del Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Senescencia Celular/efectos de la radiación , Centrosoma/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Células HCT116 , Humanos , Cinética , Mitosis/efectos de la radiación , Fenotipo
15.
Anticancer Res ; 34(8): 3875-80, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25075008

RESUMEN

AIM: To explore kinetic changes in the gene expression profile during radiation-induced mitotic catastrophes. MATERIALS AND METHODS: Gene expression changes were measured in HPV-infected HeLa Hep2 tumor cells following exposure to 5 Gy of ionizing radiation ((60)Co). Signaling pathways were explored and correlated to the biological responses linked to mitotic catastrophe. RESULTS: Following irradiation a transient G2-arrest was induced. Anaphase bridge formation and centrosome hyperamplification was observed. These phenotypical changes correlated well with the observed gene expression changes. Genes with altered expression were found to be involved in mitotic processes as well as G2- and spindle assembly checkpoints. Also centrosome-associated genes displayed an increased expression. CONCLUSION: This study elucidates specific characteristics in the altered gene expression pattern induced by irradiation, which can be correlated to the events of mitotic catastrophe in HeLa Hep2 cells. Therapeutic strategies modulating these alterations might potentiate future therapy and enhance tumor cell killing.


Asunto(s)
Mitosis/efectos de la radiación , Transcriptoma/efectos de la radiación , Centrosoma/efectos de la radiación , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos
16.
PLoS One ; 9(6): e100819, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24956373

RESUMEN

Beclin 1 interacts with UV-irradiation-resistance-associated gene (UVRAG) to form core complexes that induce autophagy. While cells with defective autophagy are prone to genomic instability that contributes to tumorigenesis, it is unknown whether Beclin1 or UVRAG can regulate the DNA damage/repair response to cancer treatment in established tumor cells. We found that siRNA knockdown of Beclin 1 or UVRAG can increase radiation-induced DNA double strand breaks (DSBs), shown by pATM and γH2Ax, and promote colorectal cancer cell death. Furthermore, knockdown of Beclin 1, UVRAG or ATG5 increased the percentage of irradiated cells with nuclear foci expressing 53BP1, a marker of nonhomologous end joining but not RAD51 (homologous recombination), compared to control siRNA. Beclin 1 siRNA was shown to attenuate UVRAG expression. Cells with a UVRAG deletion mutant defective in Beclin 1 binding showed increased radiation-induced DSBs and cell death compared to cells with ectopic wild-type UVRAG. Knockdown of Beclin 1 or UVRAG, but not ATG5, resulted in a significant increase in centrosome number (γ-tubulin staining) in irradiated cells compared to control siRNA. Taken together, these data indicate that Beclin 1 and UVRAG confer protection against radiation-induced DNA DSBs and may maintain centrosome stability in established tumor cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Centrosoma/metabolismo , Neoplasias Colorrectales/metabolismo , Citoprotección/efectos de la radiación , Daño del ADN , Rayos gamma , Proteínas de la Membrana/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Autofagia/efectos de los fármacos , Autofagia/efectos de la radiación , Proteína 5 Relacionada con la Autofagia , Beclina-1 , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Centrosoma/efectos de los fármacos , Centrosoma/efectos de la radiación , Neoplasias Colorrectales/patología , Citoprotección/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Fluorouracilo/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/efectos de la radiación , Recombinasa Rad51/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
17.
Eur J Cancer ; 49(13): 2884-91, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23747271

RESUMEN

We have previously shown that integrin-linked kinase (ILK) regulates U87 glioblastoma cell radioresistance by modulating the main radiation-induced cell death mechanism in solid tumours, the mitotic cell death. To decipher the biological pathways involved in these mechanisms, we constructed a U87 glioblastoma cell model expressing an inducible shRNA directed against ILK (U87shILK). We then demonstrated that silencing ILK enhanced radiation-induced centrosome overduplication, leading to radiation-induced mitotic cell death. In this model, ionising radiations induce hypoxia-inducible factor 1 alpha (HIF-1α) stabilisation which is inhibited by silencing ILK. Moreover, silencing HIF-1α in U87 cells reduced the surviving fraction after 2 Gy irradiation by increasing cell sensitivity to radiation-induced mitotic cell death and centrosome amplification. Because it is known that HIF-1α controls survivin expression, we then looked at the ILK silencing effect on survivin expression. We show that survivin expression is decreased in U87shILK cells. Furthermore, treating U87 cells with the specific survivin suppressor YM155 significantly increased the percentage of giant multinucleated cells, centrosomal overduplication and thus U87 cell radiosensitivity. In consequence, we decipher here a new pathway of glioma radioresistance via the regulation of radiation-induced centrosome duplication and therefore mitotic cell death by ILK, HIF-1α and survivin. This work identifies new targets in glioblastoma with the intention of radiosensitising these highly radioresistant tumours.


Asunto(s)
Glioblastoma/enzimología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Mitosis/efectos de la radiación , Proteínas Serina-Treonina Quinasas/metabolismo , Tolerancia a Radiación , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , Centrosoma/enzimología , Centrosoma/patología , Centrosoma/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Glioblastoma/genética , Glioblastoma/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/efectos de la radiación , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Transducción de Señal/efectos de la radiación , Survivin , Factores de Tiempo , Transfección
18.
Int J Radiat Oncol Biol Phys ; 86(2): 358-65, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23433796

RESUMEN

PURPOSE: Radiation induces centrosome overduplication, leading to mitotic catastrophe and tumorigenesis. Because mitotic catastrophe is one of the major tumor cell killing factors in high linear energy transfer (LET) radiation therapy and long-term survivors from such treatment have a potential risk of secondary tumors, we investigated LET dependence of radiation-induced centrosome overduplication and the underlying mechanism. METHODS AND MATERIALS: Carbon and iron ion beams (13-200 keV/µm) and γ-rays (0.5 keV/µm) were used as radiation sources. To count centrosomes after IR exposure, human U2OS and mouse NIH3T3 cells were immunostained with antibodies of γ-tubulin and centrin 2. Similarly, Nbs1-, Brca1-, Ku70-, and DNA-PKcs-deficient mouse cells and their counterpart wild-type cells were used for measurement of centrosome overduplication. RESULTS: The number of excess centrosome-containing cells at interphase and the resulting multipolar spindle at mitosis were amplified with increased LET, reaching a maximum level of 100 keV/µm, followed by sharp decrease in frequency. Interestingly, Ku70 and DNA-PKcs deficiencies marginally affected the induction of centrosome overduplication, whereas the cell killings were significantly enhanced. This was in contrast to observation that high LET radiation significantly enhanced frequencies of centrosome overduplication in Nbs1- and Brca1-deficient cells. Because NBS1/BRCA1 is implicated in monoubiquitination of γ-tubulin, we subsequently tested whether it is affected by high LET radiation. As a result, monoubiquitination of γ-tubulin was abolished in 48 to 72 hours after exposure to high LET radiation, although γ-ray exposure slightly decreased it 48 hours postirradiation and was restored to a normal level at 72 hours. CONCLUSIONS: High LET radiation significantly reduces NBS1/BRCA1-mediated monoubiquitination of γ-tubulin and amplifies centrosome overduplication with a peak at 100 keV/µm. In contrast, Ku70 and DNA-PKcs deficiencies mitigate centrosome overduplication, although deficiencies of both NBS1/BRCA1 and Ku70/DNA-PKcs markedly enhance cell killing.


Asunto(s)
Centrosoma/efectos de la radiación , Transferencia Lineal de Energía/fisiología , Tubulina (Proteína)/metabolismo , Animales , Antígenos Nucleares , Proteína BRCA1/deficiencia , Proteína BRCA1/fisiología , Proteínas de Unión al Calcio/deficiencia , Carbono , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/fisiología , Línea Celular Tumoral , Centrosoma/fisiología , Proteína Quinasa Activada por ADN/deficiencia , Proteínas de Unión al ADN/deficiencia , Radioterapia de Iones Pesados/métodos , Humanos , Hierro , Autoantígeno Ku , Ratones , Células 3T3 NIH , Proteínas Nucleares/deficiencia , Proteínas Nucleares/fisiología , Factores de Tiempo
19.
J Cell Sci ; 125(Pt 22): 5353-68, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22956538

RESUMEN

Centrosome duplication is licensed by the disengagement, or 'uncoupling', of centrioles during late mitosis. However, arrest of cells in G2 can trigger premature centriole disengagement. Here, we show that premature disengagement results from untimely activation of the anaphase-promoting complex (APC/C), leading to securin degradation and release of active separase. Although APC/C activation during G2 arrest is dependent on polo-like kinase 1 (Plk1)-mediated degradation of the APC/C inhibitor, early mitotic inhibitor 1 (Emi1), Plk1 also has a second APC/C-independent role in promoting disengagement. Importantly, APC/C and Plk1 activity also stimulates centriole disengagement in response to hydroxyurea or DNA damage-induced cell-cycle arrest and this leads to centrosome amplification. However, the reduplication of disengaged centrioles is dependent on cyclin-dependent kinase 2 (Cdk2) activity and Cdk2 activation coincides with a subsequent inactivation of the APC/C and re-accumulation of cyclin A. Although release from these arrests leads to mitotic entry, the presence of disengaged and/or amplified centrosomes results in the formation of abnormal mitotic spindles that lead to chromosome mis-segregation. Thus, oscillation of APC/C activity during cell cycle arrest promotes both centrosome amplification and genome instability.


Asunto(s)
Puntos de Control del Ciclo Celular , Centrosoma/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de la radiación , Proteínas de Ciclo Celular/metabolismo , Centriolos/efectos de los fármacos , Centriolos/metabolismo , Centriolos/efectos de la radiación , Centrosoma/efectos de los fármacos , Centrosoma/efectos de la radiación , Endopeptidasas/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Células HeLa , Humanos , Hidroxiurea/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Radiación Ionizante , Separasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo , Huso Acromático/efectos de la radiación , Quinasa Tipo Polo 1
20.
Cell Signal ; 24(2): 451-459, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21982881

RESUMEN

BARD1 heterodimerizes with BRCA1, forming an E3 ubiquitin ligase that functions at nuclear foci to repair DNA damage and the centrosome to regulate mitosis. We compared BARD1 recruitment at these structures using fluorescence recovery after photobleaching assays to measure YFP-BARD1 dynamics in live cells. In nuclei at ionizing radiation-induced foci, 20% of the BARD1 pool was immobile and 80% of slow mobility exhibiting a recovery time >500 s. In contrast, at centrosomes 83% of BARD1 was rapidly mobile with extremely fast turnover (recovery time ~20s). The ~25-fold faster exchange of BARD1 at centrosomes correlated with BRCA1-independent recruitment. We mapped key targeting sequences to a combination of the N and C-termini, and showed that mutation of the nuclear export signal reduced centrosome localization by 50%, revealing a role for CRM1. Deletion of the sequence 128-550 increased BARD1 turnover at the centrosome, consistent with a role in transient associations. Conversely, the cancer mutation Q564H reduced turnover by 25%. BARD1 is one of the most highly mobile proteins yet detected at the centrosome, and in contrast to its localization at DNA repair foci, which requires dimerization with BRCA1, targeting of BARD1 to the centrosome occurs prior to heterodimerization and its rapid turnover may provide a mechanism to regulate dimer formation.


Asunto(s)
Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias del Colon/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/efectos de la radiación , Proteína BRCA1/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Centrosoma/metabolismo , Centrosoma/efectos de la radiación , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Dimerización , Fluorescencia , Recuperación de Fluorescencia tras Fotoblanqueo , Rayos gamma , Humanos , Carioferinas/genética , Mitosis/efectos de la radiación , Mutación , Plásmidos , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/efectos de la radiación , Transfección , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...