Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 444
Filtrar
1.
Nature ; 620(7972): 209-217, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37438531

RESUMEN

The human genome functions as a three-dimensional chromatin polymer, driven by a complex collection of chromosome interactions1-3. Although the molecular rules governing these interactions are being quickly elucidated, relatively few proteins regulating this process have been identified. Here, to address this gap, we developed high-throughput DNA or RNA labelling with optimized Oligopaints (HiDRO)-an automated imaging pipeline that enables the quantitative measurement of chromatin interactions in single cells across thousands of samples. By screening the human druggable genome, we identified more than 300 factors that influence genome folding during interphase. Among these, 43 genes were validated as either increasing or decreasing interactions between topologically associating domains. Our findings show that genetic or chemical inhibition of the ubiquitous kinase GSK3A leads to increased long-range chromatin looping interactions in a genome-wide and cohesin-dependent manner. These results demonstrate the importance of GSK3A signalling in nuclear architecture and the use of HiDRO for identifying mechanisms of spatial genome organization.


Asunto(s)
Cromatina , Posicionamiento de Cromosoma , Cromosomas Humanos , Genoma Humano , Glucógeno Sintasa Quinasas , Ensayos Analíticos de Alto Rendimiento , Análisis de la Célula Individual , Humanos , Cromatina/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Posicionamiento de Cromosoma/efectos de los fármacos , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , ADN/análisis , ADN/metabolismo , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Glucógeno Sintasa Quinasas/antagonistas & inhibidores , Glucógeno Sintasa Quinasas/deficiencia , Glucógeno Sintasa Quinasas/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Interfase , Reproducibilidad de los Resultados , ARN/análisis , ARN/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual/métodos , Cohesinas
2.
BMC Cancer ; 20(1): 552, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32539694

RESUMEN

BACKGROUND: The oncogene LSF (encoded by TFCP2) has been proposed as a novel therapeutic target for multiple cancers. LSF overexpression in patient tumors correlates with poor prognosis in particular for both hepatocellular carcinoma and colorectal cancer. The limited treatment outcomes for these diseases and disappointing clinical results, in particular, for hepatocellular carcinoma in molecularly targeted therapies targeting cellular receptors and kinases, underscore the need for molecularly targeting novel mechanisms. LSF small molecule inhibitors, Factor Quinolinone Inhibitors (FQIs), have exhibited robust anti-tumor activity in multiple pre-clinical models, with no observable toxicity. METHODS: To understand how the LSF inhibitors impact cancer cell proliferation, we characterized the cellular phenotypes that result from loss of LSF activity. Cell proliferation and cell cycle progression were analyzed, using HeLa cells as a model cancer cell line responsive to FQI1. Cell cycle progression was studied either by time lapse microscopy or by bulk synchronization of cell populations to ensure accuracy in interpretation of the outcomes. In order to test for biological specificity of targeting LSF by FQI1, results were compared after treatment with either FQI1 or siRNA targeting LSF. RESULTS: Highly similar cellular phenotypes are observed upon treatments with FQI1 and siRNA targeting LSF. Along with similar effects on two cellular biomarkers, inhibition of LSF activity by either mechanism induced a strong delay or arrest prior to metaphase as cells progressed through mitosis, with condensed, but unaligned, chromosomes. This mitotic disruption in both cases resulted in improper cellular division leading to multiple outcomes: multi-nucleation, apoptosis, and cellular senescence. CONCLUSIONS: These data strongly support that cellular phenotypes observed upon FQI1 treatment are due specifically to the loss of LSF activity. Specific inhibition of LSF by either small molecules or siRNA results in severe mitotic defects, leading to cell death or senescence - consequences that are desirable in combating cancer. Taken together, these findings confirm that LSF is a promising target for cancer treatment. Furthermore, this study provides further support for developing FQIs or other LSF inhibitory strategies as treatment for LSF-related cancers with high unmet medical needs.


Asunto(s)
Benzodioxoles/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Quinolonas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzodioxoles/uso terapéutico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , División Celular/efectos de los fármacos , División Celular/genética , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Microscopía Intravital , Terapia Molecular Dirigida/métodos , Neoplasias/genética , Neoplasias/patología , Quinolonas/uso terapéutico , ARN Interferente Pequeño/metabolismo , Imagen de Lapso de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-32087855

RESUMEN

The inherent capacity of individuals to efficiently repair ionizing radiation induced DNA double strand breaks (DSBs) may be inherited, however, it is influenced by several epigenetic and environmental factors. A pilot study tested whether chronic low dose natural radiation exposure influences the rejoining of initial DNA DSBs induced by a 2 Gy γ-irradiation in 22 individuals from high (>1.5 mGy/year) and normal (≤1.5 mGy/year) level natural radiation areas (H&NLNRA) of Kerala. Rejoining of DSBs (during 1 h at 37 °C, immediately after irradiation) was evaluated at the chromosome level in the presence and absence of wortmannin (a potent inhibitor of DSB repair in normal human cells) using a cell fusion-induced premature chromosome condensation (PCC) assay. The PCC assay quantitates DSBs in the form of excess chromosome fragments in human G0 lymphocytes without the requirement for cell division. A quantitative difference was observed in the early rejoining of DNA DSBs between individuals from HLNRA and NLNRA, with HLNRA individuals showing a higher (P = 0.05) mean initial repair ratio. The results indicate an influence of chronic low dose natural radiation on initial DNA DSB repair in inhabitants of HLNRA of the Kerala coast.


Asunto(s)
Radiación de Fondo/efectos adversos , Bioensayo , Reparación del ADN/efectos de los fármacos , Rayos gamma/efectos adversos , Linfocitos/efectos de la radiación , Adulto , Animales , Células CHO , Fusión Celular , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/efectos de la radiación , Cricetulus , ADN/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Humanos , Linfocitos/citología , Linfocitos/efectos de los fármacos , Masculino , Proyectos Piloto , Cultivo Primario de Células , Dosis de Radiación , Wortmanina/farmacología
4.
Pak J Pharm Sci ; 32(6): 2667-2671, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31969300

RESUMEN

Hydrochlorothiazide (HCTZ) belongs to the thiazide diuretics family that is used for the treatment of hypertension. Enalapril is another drug that is used for the treatment of hypertension. Recently, both drugs were combined in a single medication called vaseretic that showed a strong synergistic effect against hypertension. The aim of this investigation is to examine genotoxicity of HCTZ/enalapril on chromosomal damage by measuring the frequency of sister-chromatid exchanges (SCEs) in cultured human lymphocytes. Findings showed that HCTZ (5µg/mL) significantly increased SCEs frequency (P<0.01) in cultured cells relative to the untreated cells. The levels of SCEs induced by Enalapril (10µg/mL) was similar to the level detected in the untreated cultures (P>0.05). Interestingly, SCEs induced by combined treatment were significantly lower than HCTZ alone (P<0.05). Thus, enalapril seems to protect lymphocytes from genotoxicity induced by HCTZ. Neither HCTZ nor enalapril treatment (alone or in combination) induced changes in the mitotic index and the proliferative index (P>0.05). In conclusion, HCTZ increased SCEs in cultured lymphocytes, and this increase is reduced by enalapril.


Asunto(s)
Antihipertensivos/uso terapéutico , Enalapril/farmacología , Hidroclorotiazida/toxicidad , Linfocitos/efectos de los fármacos , Adulto , Antihipertensivos/administración & dosificación , Antihipertensivos/toxicidad , Cromosomas Humanos/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Enalapril/administración & dosificación , Enalapril/uso terapéutico , Humanos , Hidroclorotiazida/administración & dosificación , Hidroclorotiazida/antagonistas & inhibidores , Hidroclorotiazida/uso terapéutico , Hipertensión/tratamiento farmacológico , Masculino , Intercambio de Cromátides Hermanas/efectos de los fármacos , Adulto Joven
5.
Methods Mol Biol ; 1703: 191-215, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29177744

RESUMEN

For analyzing chromosome structural defects that result from topoisomerase II (topo II) dysfunction we have adapted classical cell cycle experiments, classical cytological techniques and the use of a potent topo II inhibitor (ICRF-193). In this chapter, we describe in detail the protocols used and we discuss the rational for our choice and for the adaptations applied. We clarify in which cell cycle stages each of the different chromosomal aberrations induced by inhibiting topo II takes place: lack of chromosome segregation, undercondensation, lack of sister chromatid resolution, and lack of chromosome individualization. We also put these observations into the context of the two topo II-dependent cell cycle checkpoints. In addition, we have devised a system to analyze phenotypes that result when topo II is mutated in human cells. This serves as an alternative strategy to the use of topo II inhibitors to perturb topo II function.


Asunto(s)
Cromosomas Humanos/química , ADN-Topoisomerasas de Tipo II/metabolismo , Mutación , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Inhibidores de Topoisomerasa II/farmacología , Puntos de Control del Ciclo Celular , Aberraciones Cromosómicas , Cromosomas Humanos/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/genética , Dicetopiperazinas , Células HEK293 , Células HeLa , Humanos , Mitosis/efectos de los fármacos , Fenotipo , Piperazinas/farmacología , Proteínas de Unión a Poli-ADP-Ribosa/genética
6.
Toxicol Lett ; 281: 110-118, 2017 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-28964810

RESUMEN

The tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is the most potent risk factor among tobacco-related carcinogens in lung cancer progression and outcomes. Although genetic mutations and chromosome instability have been detected in NNK-induced lung tumors, the oncogenic mechanisms of NNK are not fully understood. Here, we show that NNK increases chromosomal instability by disrupting spindle microtubule (MT) attachment to the kinetochore (KT) and spindle dynamics. Mechanistically, NNK blocks the targeting of p53 to the centrosome during mitosis, leading to chromosome alignment defects in metaphase. Therefore, lung cancer cells with wild-type p53, such as A594 and H226B, are more resistant to the NNK treatment than p53-mutant lung cancer cells, such as A1299 and H226Br. Although NNK does not affect the levels or transcriptional activity of p53, the reduction of the p53 level at the centrosome exacerbates the NNK-induced chromosome alignment defect in A549 and H226B cells. Therefore, p53 protects against NNK-induced chromosome instability by modulating the function of centrosome-localized p53 and not by modulating transcriptional activity. We conclude that NNK may increase the risk of lung cancer progression and poorer outcomes in patients with p53 mutations by perturbing proper mitotic progression and chromosome integrity.


Asunto(s)
Carcinógenos/toxicidad , Centrosoma/efectos de los fármacos , Nicotiana/química , Nitrosaminas/toxicidad , Proteína p53 Supresora de Tumor/metabolismo , Células A549 , Línea Celular Tumoral , Centrosoma/metabolismo , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/genética , Células HeLa , Humanos , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/genética , Mitosis/efectos de los fármacos , Factores de Riesgo , Proteína p53 Supresora de Tumor/genética
7.
J Toxicol Environ Health A ; 80(13-15): 651-660, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28524814

RESUMEN

Results of a number of studies indicate that electroplaters have increased cancer risks as a consequence of exposure to genotoxic metals such as chromium (VI) and nickel. These effects may be due to induction of damage of the genetic material which plays a key role in the etiology of cancer, and it was found that workers in galvanization factories exhibited increased levels of DNA damage. The aim of the present study was to investigate genetic stability in workers of a bright plating factory who are exposed to chromium (Cr) and cobalt (Co). Exfoliated cells were collected from the buccal and nasal mucosa of workers (n = 42) and matched controls (n = 43) and analyzed for induction of micronuclei (MN) which are formed as a consequence of chromosomal aberrations. In addition, other nuclear anomalies namely nuclear buds (Nbuds) which are formed as a consequence of gene amplification and markers indicating different stages of cell death (condensed chromatin, karyorrhexis, karyolysis, and pyknosis) were also assessed. No evidence was noted for induction of MN, but significantly increased rates of Nbuds in cells from both, buccal and nasal mucosa, were found. Parameters which are indicative for cytotoxic effects were more pronounced in nasal cells and rose with duration of employment period. Overall, our findings indicated that no apparent chromosomal damage occurred in bright electroplaters. However, data demonstrated that acute cytotoxic effects may lead to inflammations and/or lesions in epithelia of the respiratory tract of the workers.


Asunto(s)
Cromo/toxicidad , Cobalto/toxicidad , Galvanoplastia , Mucosa Bucal/efectos de los fármacos , Mucosa Nasal/efectos de los fármacos , Exposición Profesional/efectos adversos , Adulto , Muerte Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Cromosomas Humanos/efectos de los fármacos , Femenino , Humanos , Masculino , Micronúcleos con Defecto Cromosómico/inducido químicamente
8.
Methods ; 123: 102-118, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28385536

RESUMEN

The pharmacoepigenome can be defined as the active, noncoding province of the genome including canonical spatial and temporal regulatory mechanisms of gene regulation that respond to xenobiotic stimuli. Many psychotropic drugs that have been in clinical use for decades have ill-defined mechanisms of action that are beginning to be resolved as we understand the transcriptional hierarchy and dynamics of the nucleus. In this review, we describe spatial, temporal and biomechanical mechanisms mediated by psychotropic medications. Focus is placed on a bioinformatics pipeline that can be used both for detection of pharmacoepigenomic variants that discretize drug response and adverse events to improve pharmacogenomic testing, and for the discovery of novel CNS therapeutics. This approach integrates the functional topology and dynamics of the transcriptional hierarchy of the pharmacoepigenome, gene variant-driven identification of pharmacogenomic regulatory domains, and mesoscale mapping for the discovery of novel CNS pharmacodynamic pathways in human brain. Examples of the application of this pipeline are provided, including the discovery of valproic acid (VPA) mediated transcriptional reprogramming of neuronal cell fate following injury, and mapping of a CNS pathway glutamatergic pathway for the mood stabilizer lithium. These examples in regulatory pharmacoepigenomics illustrate how ongoing research using the 4D nucleome provides a foundation to further insight into previously unrecognized psychotropic drug pharmacodynamic pathways in the human CNS.


Asunto(s)
Biología Computacional/métodos , Genoma Humano , Proteínas del Tejido Nervioso/genética , Farmacogenética/métodos , Psicotrópicos/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/metabolismo , Cromosomas Humanos/ultraestructura , Ritmo Circadiano/fisiología , Minería de Datos/métodos , Regulación de la Expresión Génica , Humanos , Litio/uso terapéutico , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Transcripción Genética , Ácido Valproico/uso terapéutico
9.
Molecules ; 22(1)2017 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-28117734

RESUMEN

Hyperforin (HF), a substance that accumulates in the leaves and flowers of Hypericum perforatum L. (St. John's wort), consists of a phloroglucinol skeleton with lipophilic isoprene chains. HF exhibits several medicinal properties and is mainly used as an antidepressant. So far, the antigenotoxicity of HF has not been investigated at the level of primary genetic damage, gene mutations, and chromosome aberrations, simultaneously. The present work is designed to investigate the potential antigenotoxic effects of HF using three different experimental test systems. The antigenotoxic effect of HF leading to the decrease of primary/transient promutagenic genetic changes was detected by the alkaline comet assay on human lymphocytes. The HF antimutagenic effect leading to the reduction of gene mutations was assessed using the Ames test on the standard Salmonella typhimurium (TA97, TA98, and TA100) bacterial strains, and the anticlastogenic effect of HF leading to the reduction of chromosome aberrations was evaluated by the in vitro mammalian chromosome aberration test on the human tumor cell line HepG2 and the non-carcinogenic cell line VH10. Our findings provided evidence that HF showed antigenotoxic effects towards oxidative mutagen zeocin in the comet assay and diagnostic mutagen (4-nitroquinoline-1-oxide) in the Ames test. Moreover, HF exhibited an anticlastogenic effect towards benzo(a)pyrene and cisplatin in the chromosome aberration test.


Asunto(s)
Antimutagênicos/farmacología , Bleomicina/toxicidad , Aberraciones Cromosómicas/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Floroglucinol/análogos & derivados , Salmonella typhimurium/genética , Terpenos/farmacología , Proteínas Bacterianas/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cromosomas Humanos/efectos de los fármacos , Células Hep G2 , Humanos , Leucocitos , Mutación/efectos de los fármacos , Floroglucinol/farmacología
10.
Methods ; 112: 18-24, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27524557

RESUMEN

Biodosimetry is an important tool for triage in the case of large-scale radiological or nuclear emergencies, but traditional microscope-based methods can be tedious and prone to scorer fatigue. While the dicentric chromosome assay (DCA) has been adapted for use in triage situations, it is still time-consuming to create and score slides. Recent adaptations of traditional biodosimetry assays to imaging flow cytometry (IFC) methods have dramatically increased throughput. Additionally, recent improvements in image analysis algorithms in the IFC software have resulted in improved specificity for spot counting of small events. In the IFC method for the dicentric chromosome analysis (FDCA), lymphocytes isolated from whole blood samples are cultured with PHA and Colcemid. After incubation, lymphocytes are treated with a hypotonic solution and chromosomes are isolated in suspension, labelled with a centromere marker and stained for DNA content with DRAQ5. Stained individual chromosomes are analyzed on the ImageStream®X (EMD-Millipore, Billerica, MA) and mono- and dicentric chromosome populations are identified and enumerated using advanced image processing techniques. Both the preparation of the isolated chromosome suspensions as well as the image analysis methods were fine-tuned in order to optimize the FDCA. In this paper we describe the method to identify and score centromeres in individual chromosomes by IFC and show that the FDCA method may further improve throughput for triage biodosimetry in the case of large-scale radiological or nuclear emergencies.


Asunto(s)
Aberraciones Cromosómicas/efectos de la radiación , Cromosomas Humanos/efectos de la radiación , Citometría de Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Exposición a la Radiación/análisis , Radiometría/métodos , Antraquinonas/química , Centrómero/efectos de los fármacos , Centrómero/efectos de la radiación , Centrómero/ultraestructura , Aberraciones Cromosómicas/efectos de los fármacos , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/ultraestructura , Demecolcina/farmacología , Relación Dosis-Respuesta en la Radiación , Humanos , Citometría de Imagen/instrumentación , Linfocitos/efectos de los fármacos , Linfocitos/efectos de la radiación , Fitohemaglutininas/farmacología , Coloración y Etiquetado/métodos
11.
Pak J Pharm Sci ; 29(3 Suppl): 1033-6, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27383496

RESUMEN

This work was carried out to study the genotoxicity of pyridine in vitro on human leucocyte culture. Cyclophosphamide, a well-known carcinogen was used as positive control. The four different concentrations of pyridine and cyclophosphamide showed breaks and pulverization of chromosomes in dose dependent manner. Higher number of pulverization was observed with higher concentration of pyridine (3.25µg/mL). Based on this data, our results confirm that both pyridine and its precursor showed genotoxicity against human lymphocytes.


Asunto(s)
Aberraciones Cromosómicas/inducido químicamente , Cromosomas Humanos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Pruebas de Mutagenicidad , Piridinas/toxicidad , Células Cultivadas , Ciclofosfamida/toxicidad , Roturas del ADN , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Linfocitos/patología , Masculino , Medición de Riesgo , Adulto Joven
12.
Mutagenesis ; 31(3): 309-21, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26443851

RESUMEN

The anti-parasitic benzimidazole flubendazole has been used for many years to treat intestinal infections in humans and animals. Previous genotoxicity studies have shown that the compound is not a bacterial mutagen and a bone marrow micronucleus test, using a formulation that limited systemic absorption, was negative. The purpose of this study is to explore the genotoxicity of flubendazole and its main metabolites in in vitro micronucleus studies and to test a new oral formulation that improves systemic absorption in an in vivo micronucleus test. The isolated metabolites were also screened using the Ames test for bacterial mutagenicity. It was found that flubendazole, like other chemically related benzimidazoles used in anti-parasitic therapies, is a potent aneugen in vitro The hydrolysed metabolite of flubendazole is negative in these tests, but the reduced metabolite (R- and S-forms) shows both aneugenic and clastogenic activity. However, in vitro micronucleus tests of flubendazole in the presence of rat liver S9 gave almost identical signals for aneugenicity as they did in the absence of S9, suggesting that any clastogenicity from the reduced metabolite is not sufficient to change the overall profile. Like flubendazole itself, both metabolites are negative in the Ames test. Analysis of dose-response curves from the in vitro tests, using recently developed point of departure approaches, demonstrate that the aneugenic potency of flubendazole is very similar to related anti-parasitic benzimidazoles, including albendazole, which is used in mass drug administration programmes to combat endemic filarial diseases. The in vivo micronucleus test of the new formulation of flubendazole also showed evidence of induced aneugenicity. Analysis of the in vivo data allowed a reference dose for aneugenicity to be established which can be compared with therapeutic exposures of flubendazole when this has been established. Analysis of the plasma from the animals used in the in vivo micronucleus test showed that there is increased exposure to flubendazole compared with previously tested formulations, as well as significant formation of the non-genotoxic hydrolysed metabolite of flubendazole and small levels of the reduced metabolite. In conclusion, this study shows that flubendazole is a potent aneugen in vitro with similar potency to chemically related benzimidazoles currently used as anti-parasitic therapies. The reduced metabolite also has aneugenic properties as well as clastogenic properties. Treatment with a new formulation of flubendazole that allows increased systemic exposure, compared with previously used formulations, also results in detectable aneugenicity in vivo. Based on the lack of carcinogenicity of this class of benzimidazoles and the intended short-term dosing, it is unlikely that flubendazole treatment will pose a carcinogenic risk to patients.


Asunto(s)
Aneugénicos/toxicidad , Aberraciones Cromosómicas , Daño del ADN , Linfocitos/efectos de los fármacos , Mebendazol/análogos & derivados , Activación Metabólica , Aneugénicos/metabolismo , Animales , Antinematodos/metabolismo , Antinematodos/toxicidad , Células Cultivadas , Cromosomas Humanos/efectos de los fármacos , ADN/efectos de los fármacos , Humanos , Linfocitos/metabolismo , Masculino , Mebendazol/metabolismo , Mebendazol/toxicidad , Pruebas de Micronúcleos , Mutágenos/metabolismo , Mutágenos/toxicidad , Ratas
13.
Artículo en Inglés | MEDLINE | ID: mdl-26520368

RESUMEN

ICRF-187 (Cardioxane™, Chiron) is a catalytic inhibitor of DNA topoisomerase II (Topo II), proposed to act by blocking Topo II-mediated DNA cleavage without stabilizing DNA-Topo II-"cleavable complexes". In this study ICRF-187 was used to evaluate the potential involvement of DNA topoisomerase II in the formation of the radiation-induced chromosome-type aberrations in the G0 phase of the cell cycle in human lymphocytes from three healthy male donors. This is based on many evidences that DNA topoisomerases are involved in DNA recombination, mainly of illegitimate type (non-homologous) both in vitro and in vivo. The results obtained clearly indicated that ICRF-187 did not induce per se any chromosomal damage. When challenged with the non-catalytic Topo II poison VP-16 (etoposide), which acts by stabilizing the "cleavable complex" generating "protein concealed" DSB's and thus chromosomal aberrations, it completely abolished the significant induction of chromosome-type aberrations and formation of dicentric chromosomes. This indicates that ICRF-187 acts effectively as catalytic inhibitor of Topo II. On the other hand, when X-ray treatments were challenged with ICRF-187 using experimental conditions as for VP-16 treatments, no modification of the incidence of chromosome-type aberrations and dicentric chromosomes was observed. On this basis, we conclude that Topo II is not involved in the formation of X-ray-induced chromosome-type aberrations and dicentric chromosomes in human lymphocytes in the G0 phase of the cell cycle.


Asunto(s)
Aberraciones Cromosómicas/efectos de los fármacos , Cromosomas Humanos/efectos de la radiación , ADN-Topoisomerasas de Tipo II/metabolismo , Dexrazoxano/farmacología , Linfocitos/enzimología , Inhibidores de Topoisomerasa II/farmacología , Adulto , Cromosomas Humanos/efectos de los fármacos , Análisis Citogenético , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Etopósido/farmacología , Humanos , Linfocitos/efectos de los fármacos , Linfocitos/efectos de la radiación , Masculino , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase de Descanso del Ciclo Celular/efectos de la radiación , Rayos X
14.
Epigenetics ; 10(4): 329-41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25830902

RESUMEN

Werner Syndrome (WS) is a rare inherited disease characterized by premature aging and increased propensity for cancer. Mutations in the WRN gene can be of several types, including nonsense mutations, leading to a truncated protein form. WRN is a RecQ family member with both helicase and exonuclease activities, and it participates in several cell metabolic pathways, including DNA replication, DNA repair, and telomere maintenance. Here, we reported a novel homozygous WS mutation (c.3767 C > G) in 2 Argentinian brothers, which resulted in a stop codon and a truncated protein (p.S1256X). We also observed increased WRN promoter methylation in the cells of patients and decreased messenger WRN RNA (WRN mRNA) expression. Finally, we showed that the read-through of nonsense mutation pharmacologic treatment with both aminoglycosides (AGs) and ataluren (PTC-124) in these cells restores full-length protein expression and WRN functionality.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Epigénesis Genética , Mutación , Síndrome de Werner/genética , Envejecimiento Prematuro/genética , Aminoglicósidos/farmacología , Apoptosis/efectos de los fármacos , Células Cultivadas , Cromosomas Humanos/efectos de los fármacos , Codón sin Sentido , Daño del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Femenino , Humanos , Masculino , Oxadiazoles/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Síndrome de Werner/tratamiento farmacológico
15.
Carcinogenesis ; 36(1): 160-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25391402

RESUMEN

Formaldehyde (FA) is an economically important industrial chemical to which millions of people worldwide are exposed environmentally and occupationally. Recently, the International Agency for Cancer Research concluded that there is sufficient evidence that FA causes leukemia, particularly myeloid leukemia. To evaluate the biological plausibility of this association, we employed a chromosome-wide aneuploidy study approach, which allows the evaluation of aneuploidy and structural chromosome aberrations (SCAs) of all 24 chromosomes simultaneously, to analyze cultured myeloid progenitor cells from 29 workers exposed to relatively high levels of FA and 23 unexposed controls. We found statistically significant increases in the frequencies of monosomy, trisomy, tetrasomy and SCAs of multiple chromosomes in exposed workers compared with controls, with particularly notable effects for monosomy 1 [P = 6.02E-06, incidence rate ratio (IRR) = 2.31], monosomy 5 (P = 9.01E-06; IRR = 2.24), monosomy 7 (P = 1.57E-05; IRR = 2.17), trisomy 5 (P = 1.98E-05; IRR = 3.40) and SCAs of chromosome 5 (P = 0.024; IRR = 4.15). The detection of increased levels of monosomy 7 and SCAs of chromosome 5 is particularly relevant as they are frequently observed in acute myeloid leukemia. Our findings provide further evidence that leukemia-related cytogenetic changes can occur in the circulating myeloid progenitor cells of healthy workers exposed to FA, which may be a potential mechanism underlying FA-induced leukemogenesis.


Asunto(s)
Aneuploidia , Cromosomas Humanos/efectos de los fármacos , Desinfectantes/efectos adversos , Formaldehído/efectos adversos , Células Progenitoras Mieloides/efectos de los fármacos , Exposición Profesional/efectos adversos , Adulto , Estudios de Casos y Controles , Células Cultivadas , Estudios Transversales , Femenino , Estudios de Seguimiento , Humanos , Hibridación Fluorescente in Situ , Masculino , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patología , Pronóstico
16.
Radiat Prot Dosimetry ; 159(1-4): 52-60, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24743756

RESUMEN

The calyculin A-induced premature chromosome condensation (PCC) assay is a simple and useful method for assessing the cell-cycle distribution in cells, since calyculin A induces chromosome condensation in various phases of the cell cycle. In this study, a novel parameter, the cell-cycle progression index (CPI), in the PCC assay was validated as a novel biomarker for biodosimetry. Peripheral blood was drawn from healthy donors after informed consent was obtained. CPI was investigated using a human peripheral blood lymphocyte (PBL) ex vivo irradiation ((60)Co-gamma rays: ∼0.6 Gy min(-1), or X ray: 1.0 Gy min(-1); 0-10 Gy) model. The calyculin A-induced PCC assay was performed for chromosome preparation. PCC cells were divided into the following five categories according to cell-cycle stage: non-PCC, G1-PCC, S-PCC, G2/M-PCC and M/A-PCC cells. CPI was calculated as the ratio of G2/M-PCC cells to G1-PCC cells. The PCC-stage distribution varied markedly with irradiation doses. The G1-PCC cell fraction was significantly reduced, and the G2/M-PCC cell fraction increased, in 10-Gy-irradiated PBL after 48 h of culture. CPI levels were fitted to an exponential dose-response curve with gamma-ray irradiation [y = 0.6729 + 0.3934 exp(0.5685D), r = 1.0000, p < 0.0001] and X-ray irradiation [y = -0.3743 + 0.9744 exp(0.3321D), r = 0.9999, p < 0.0001]. There were no significant individual (p = 0.853) or gender effects (p = 0.951) on the CPI in the human peripheral blood ex vivo irradiation model. Furthermore, CPI measurements are rapid (< 15 min per case). These results suggest that the CPI is a useful screening tool for the assessment of radiation doses received ranging from 0 to 10 Gy in radiation exposure early after a radiation event, especially after a mass-casualty radiological incident.


Asunto(s)
Bioensayo/métodos , Ciclo Celular/efectos de la radiación , Cromosomas Humanos/efectos de la radiación , Linfocitos/efectos de la radiación , Traumatismos por Radiación/diagnóstico , Carcinógenos/farmacología , Ciclo Celular/genética , Células Cultivadas , Cromosomas Humanos/efectos de los fármacos , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Linfocitos/efectos de los fármacos , Masculino , Toxinas Marinas , Oxazoles/farmacología , Traumatismos por Radiación/genética , Rayos X
17.
J Cell Biochem ; 115(4): 763-71, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24453048

RESUMEN

Genistein, an isoflavone abundantly present in soybeans, possesses anticancer properties and induces growth inhibition including cell cycle arrest and apoptosis. Although abnormal cell division, such as defects in chromosome segregation and spindle formation, and polyploidization have been described, the mechanisms underlying the induction of abnormal cell division are unknown. In this study, we examined the effect of genistein on cell division in cells that are synchronized in M phase, since genistein treatment delays mitotic entry in asynchronous cells. HeLa S3 cells were arrested at the G2 phase and subsequently released into the M phase in presence of genistein. Immunofluorescence staining showed that genistein treatment delays M phase progression. Time-lapse analysis revealed that the delay occurs until anaphase onset. In addition, genistein treatment induces cleavage furrow regression, resulting in the generation of binucleated cells. Central spindle formation, which is essential for cytokinesis, is partially disrupted in genistein-treated cells. Moreover, aberrant chromosome segregation, such as a chromosome bridge and lagging chromosome, occurs through progression of cytokinesis. RhoA, which plays a role in the assembly and constriction of an actomyosin contractile ring, is delocalized from the cortex of the ingressing cleavage furrow. These results suggest that genistein treatment induces binucleated cell formation through cleavage furrow regression, which is accompanied by chromosome bridge formation and RhoA delocalization. Our results provide the mechanism that underlies genistein-induced polyploidization, which may be involved in genistein-induced growth inhibition.


Asunto(s)
Anafase/efectos de los fármacos , Cromosomas Humanos/efectos de los fármacos , Citocinesis/efectos de los fármacos , Genisteína/farmacología , Proteína de Unión al GTP rhoA/metabolismo , Anafase/genética , División Celular/efectos de los fármacos , División Celular/genética , Células HeLa/efectos de los fármacos , Humanos , Mitosis/efectos de los fármacos , Mitosis/genética
18.
Mol Biol Cell ; 25(5): 594-605, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24403607

RESUMEN

The spindle and kinetochore-associated (Ska) protein complex is a heterotrimeric complex required for timely anaphase onset. The major phenotypes seen after small interfering RNA-mediated depletion of Ska are transient alignment defects followed by metaphase arrest that ultimately results in cohesion fatigue. We find that cells depleted of Ska3 arrest at metaphase with only partial degradation of cyclin B1 and securin. In cells arrested with microtubule drugs, Ska3-depleted cells exhibit slower mitotic exit when the spindle checkpoint is silenced by inhibition of the checkpoint kinase, Mps1, or when cells are forced to exit mitosis downstream of checkpoint silencing by inactivation of Cdk1. These results suggest that in addition to a role in fostering kinetochore-microtubule attachment and chromosome alignment, the Ska complex has functions in promoting anaphase onset. We find that both Ska3 and microtubules promote chromosome association of the anaphase-promoting complex/cyclosome (APC/C). Chromosome-bound APC/C shows significantly stronger ubiquitylation activity than cytoplasmic APC/C. Forced localization of Ska complex to kinetochores, independent of microtubules, results in enhanced accumulation of APC/C on chromosomes and accelerated cyclin B1 degradation during induced mitotic exit. We propose that a Ska-microtubule-kinetochore association promotes APC/C localization to chromosomes, thereby enhancing anaphase onset and mitotic exit.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Mitosis/fisiología , Anafase/efectos de los fármacos , Anafase/genética , Anafase/fisiología , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/metabolismo , Ciclina B1/metabolismo , Células HeLa , Humanos , Metafase/efectos de los fármacos , Metafase/genética , Metafase/fisiología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/efectos de los fármacos , Mitosis/genética , Modelos Biológicos , Nocodazol/farmacología , Moduladores de Tubulina/farmacología
19.
Genome Biol ; 14(12): R135, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24330859

RESUMEN

BACKGROUND: Local higher-order chromatin structure, dynamics and composition of the DNA are known to determine double-strand break frequencies and the efficiency of repair. However, how DNA damage response affects the spatial organization of chromosome territories is still unexplored. RESULTS: Our report investigates the effect of DNA damage on the spatial organization of chromosome territories within interphase nuclei of human cells. We show that DNA damage induces a large-scale spatial repositioning of chromosome territories that are relatively gene dense. This response is dose dependent, and involves territories moving from the nuclear interior to the periphery and vice versa. Furthermore, we have found that chromosome territory repositioning is contingent upon double-strand break recognition and damage sensing. Importantly, our results suggest that this is a reversible process where, following repair, chromosome territories re-occupy positions similar to those in undamaged control cells. CONCLUSIONS: Thus, our report for the first time highlights DNA damage-dependent spatial reorganization of whole chromosomes, which might be an integral aspect of cellular damage response.


Asunto(s)
Posicionamiento de Cromosoma/efectos de los fármacos , Cromosomas Humanos/efectos de los fármacos , Reparación del ADN , ADN/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Células Cultivadas , Cromosomas Humanos/genética , Cisplatino/farmacología , Relación Dosis-Respuesta a Droga , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Interfase/efectos de los fármacos
20.
Environ Mol Mutagen ; 54(9): 755-68, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24038327

RESUMEN

Vinyl acetate monomer (VAM) produced rat nasal tumors at concentrations in the hundreds of parts per million. However, VAM is weakly genotoxic in vitro and shows no genotoxicity in vivo. A European Union Risk Assessment concluded that VAM's hydrolysis to acetaldehyde (AA), via carboxylesterase, is a critical key event in VAM's carcinogenic potential. In the following study, we observed increases in micronuclei (MN) and thymidine kinase (Tk) mutants that were dependent on the ability of TK6 cell culture conditions to rapidly hydrolyze VAM to AA. Heat-inactivated horse serum demonstrated a high capacity to hydrolyze VAM to AA; this activity was highly correlated with a concomitant increase in MN. In contrast, heat-inactivated fetal bovine serum (FBS) did not hydrolyze VAM and no increase in MN was observed. AA's ability to induce MN was not impacted by either serum since it directly forms Schiff bases with DNA and proteins. Increased mutant frequency at the Tk locus was similarly mitigated when AA formation was not sufficiently rapid, such as incubating VAM in the presence of FBS for 4 hr. Interestingly, neither VAM nor AA induced mutations at the HPRT locus. Finally, cytotoxicity paralleled genotoxicity demonstrating that a small degree of cytotoxicity occurred prior to increases in MN. These results established 0.25 mM as a consistent concentration where genotoxicity first occurred for both VAM and AA provided VAM is hydrolyzed to AA. This information further informs significant key events related to the mode of action of VAM-induced nasal mucosal tumors in rats.


Asunto(s)
Acetaldehído/efectos adversos , Cromosomas Humanos/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Mutágenos/efectos adversos , Mutación/genética , Timidina Quinasa/genética , Compuestos de Vinilo/efectos adversos , Apoptosis/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Linfocitos/efectos de los fármacos , Pruebas de Micronúcleos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA