Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 446
Filtrar
Más filtros

Publication year range
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Genome Res ; 22(6): 993-1005, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22456607

RESUMEN

Chromosomal common fragile sites (CFSs) are unstable genomic regions that break under replication stress and are involved in structural variation. They frequently are sites of chromosomal rearrangements in cancer and of viral integration. However, CFSs are undercharacterized at the molecular level and thus difficult to predict computationally. Newly available genome-wide profiling studies provide us with an unprecedented opportunity to associate CFSs with features of their local genomic contexts. Here, we contrasted the genomic landscape of cytogenetically defined aphidicolin-induced CFSs (aCFSs) to that of nonfragile sites, using multiple logistic regression. We also analyzed aCFS breakage frequencies as a function of their genomic landscape, using standard multiple regression. We show that local genomic features are effective predictors both of regions harboring aCFSs (explaining ∼77% of the deviance in logistic regression models) and of aCFS breakage frequencies (explaining ∼45% of the variance in standard regression models). In our optimal models (having highest explanatory power), aCFSs are predominantly located in G-negative chromosomal bands and away from centromeres, are enriched in Alu repeats, and have high DNA flexibility. In alternative models, CpG island density, transcription start site density, H3K4me1 coverage, and mononucleotide microsatellite coverage are significant predictors. Also, aCFSs have high fragility when colocated with evolutionarily conserved chromosomal breakpoints. Our models are predictive of the fragility of aCFSs mapped at a higher resolution. Importantly, the genomic features we identified here as significant predictors of fragility allow us to draw valuable inferences on the molecular mechanisms underlying aCFSs.


Asunto(s)
Inestabilidad Cromosómica , Sitios Frágiles del Cromosoma , Genoma Humano , Modelos Genéticos , Elementos Alu , Animales , Afidicolina/farmacología , Centrómero , Rotura Cromosómica , Cromosomas Humanos/efectos de los fármacos , Islas de CpG , Análisis Citogenético , Humanos , Modelos Logísticos , Ratones , Repeticiones de Microsatélite , Reproducibilidad de los Resultados , Sitio de Iniciación de la Transcripción
12.
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
13.
BMC Genomics ; 14: 70, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23368971

RESUMEN

BACKGROUND: An emerging Hi-C protocol has the ability to probe three-dimensional (3D) architecture and capture chromatin interactions in a genome-wide scale. It provides informative results to address how chromatin organization changes contribute to disease/tumor occurrence and progression in response to stimulation of environmental chemicals or hormones. RESULTS: In this study, using MCF7 cells as a model system, we found estrogen stimulation significantly impact chromatin interactions, leading to alteration of gene regulation and the associated histone modification states. Many chromosomal interaction regions at different levels of interaction frequency were identified. In particular, the top 10 hot regions with the highest interaction frequency are enriched with breast cancer specific genes. Furthermore, four types of E2-mediated strong differential (gain- or loss-) chromosomal (intra- or inter-) interactions were classified, in which the number of gain-chromosomal interactions is less than the number of loss-chromosomal interactions upon E2 stimulation. Finally, by integrating with eight histone modification marks, DNA methylation, regulatory elements regions, ERα and Pol-II binding activities, associations between epigenetic patterns and high chromosomal interaction frequency were revealed in E2-mediated gene regulation. CONCLUSIONS: The work provides insight into the effect of chromatin interaction on E2/ERα regulated downstream genes in breast cancer cells.


Asunto(s)
Cromosomas Humanos/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Estradiol/farmacología , Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Genómica , Cromatina/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , Receptor alfa de Estrógeno/metabolismo , Humanos , Células MCF-7
14.
Blood ; 118(9): 2602-8, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21719598

RESUMEN

As peripheral blood has surpassed bone marrow as a predominant source of stem cells for transplantation, use of the cytokine granulocyte colony-stimulating factor (G-CSF) to mobilize peripheral blood stem cells (PBSCs) is increasing. Issues regarding potential genotoxic effects of even short-term, low-dose G-CSF treatment for the healthy donors have been raised. To address the question of chromosomal instability, we used FISH to evaluate the peripheral blood lymphocytes of 22 PBSC donors and 22 matched controls at 5 time points over a 12-month period. The specimens obtained were a pre-G-CSF, followed by collections at the time of PBSC harvest (days 5-7) and at 2, 6, and 12 months after donation. Eight additional PBSC donors provided a single sample at 12 months. Nine loci (mapped to chromosomes 7, 8, 9, 17, 21, and 22) were evaluated for aneuploidy, including 3 mapped to chromosome 7 because of the specific relevance of monosomy 7. Replication timing was evaluated for chromosome 15 and 17 loci. No evidence was found of G-CSF-induced chromosomal instability. This work supports the epidemiologic data that have demonstrated no increased risk for hematologic malignancies in G-CSF-primed PBSC donors.


Asunto(s)
Aneuploidia , Cromosomas Humanos/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/toxicidad , Movilización de Célula Madre Hematopoyética/efectos adversos , Trasplante de Células Madre de Sangre Periférica , Donantes de Tejidos , Células Cultivadas , Deleción Cromosómica , Cromosomas Humanos/ultraestructura , Cromosomas Humanos Par 7 , Estudios de Seguimiento , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Hibridación Fluorescente in Situ , Linfocitos/citología , Linfocitos/efectos de los fármacos , Estudios Prospectivos , Proyectos de Investigación , Trasplante Homólogo
15.
Arch Toxicol ; 87(1): 73-98, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23179754

RESUMEN

Studies about formaldehyde (FA) published since the guideline of 0.1 mg/m(3) by the World Health Organization (WHO) in 2010 have been evaluated; critical effects were eye and nasal (portal-of-entry) irritation. Also, it was considered to prevent long-term effects, including all types of cancer. The majority of the recent toxicokinetic studies showed no exposure-dependent FA-DNA adducts outside the portal-of-entry area and FA-DNA adducts at distant sites were due to endogenously generated FA. The no-observed-adverse-effect level for sensory irritation was 0.5 ppm and recently reconfirmed in hypo- and hypersensitive individuals. Investigation of the relationship between FA exposure and asthma or other airway effects in children showed no convincing association. In rats, repeated exposures showed no point mutation in the p53 and K-Ras genes at ≤15 ppm neither increased cell proliferation, histopathological changes and changes in gene expression at 0.7 ppm. Repeated controlled exposures (0.5 ppm with peaks at 1 ppm) did not increase micronucleus formation in human buccal cells or nasal tissue (0.7 ppm) or in vivo genotoxicity in peripheral blood lymphocytes (0.7 ppm), but higher occupational exposures were associated with genotoxicity in buccal cells and cultivated peripheral blood lymphocytes. It is still valid that exposures not inducing nasal squamous cell carcinoma in rats will not induce nasopharyngeal cancer or lymphohematopoietic malignancies in humans. Reproductive and developmental toxicity are not considered relevant in the absence of sensory irritation. In conclusion, the WHO guideline has been strengthened.


Asunto(s)
Contaminación del Aire Interior/efectos adversos , Formaldehído/toxicidad , Medición de Riesgo/tendencias , Animales , Asma/inducido químicamente , Proliferación Celular/efectos de los fármacos , Niño , Cromosomas Humanos/efectos de los fármacos , Femenino , Formaldehído/farmacocinética , Francia , Regulación de la Expresión Génica/efectos de los fármacos , Genes ras , Guías como Asunto , Humanos , Masculino , Mucosa Bucal/efectos de los fármacos , Mutación , Neoplasias Nasofaríngeas/inducido químicamente , Nivel sin Efectos Adversos Observados , Exposición Profesional , Mutación Puntual , Ratas , Medición de Riesgo/métodos , Distribución Tisular , Pruebas de Toxicidad/métodos , Organización Mundial de la Salud
16.
J Sci Food Agric ; 93(6): 1317-25, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23027643

RESUMEN

BACKGROUND: Quinocetone has been widely used as an animal growth promoter in China. However, available data showed that QCT has potential genotoxicity. This study was conducted to investigate the cytotoxicity and genotoxicity of QCT in human lymphocytes. RESULTS: CCK-8 assay demonstrated the severe inhibitory effects by QCT in a dose- and time-dependent manner. DNA damage analysis using alkalic Comet assay revealed a pronounced increase of DNA fragmentation in cells. In contrast, DNA damage was significantly decreased after incubation with S9 mix. This finding demonstrated that the intermediate metabolites of this drug exerted lower genotoxicity than its parent drugs. We further described chromosomal damage induced by this drug employing cytokinesis-block micronucleus assay. The micronucleus frequency was significantly increased in quinocetone groups as compared to controls. Similar to the observation in Comet assay, incorporation of S9 mix in the cytokinesis-block micronucleus assay could markedly alleviate the chromosomal damage. Moreover, QCT could invoke increase of reactive oxygen species generation in cells. Intriguingly, the toxicity of QCT was more prominent in samples from males than those from females under the same conditions. CONCLUSION: QCT could induce potential cytotoxicity and genotoxicity in human lymphocytes.


Asunto(s)
Citotoxinas/efectos adversos , Daño del ADN , Contaminación de Alimentos , Micronúcleos con Defecto Cromosómico , Mutágenos/efectos adversos , Estrés Oxidativo , Quinoxalinas/efectos adversos , Adulto , Animales , Bovinos , Técnicas de Cultivo de Célula , China , Cromosomas Humanos/efectos de los fármacos , Ensayo Cometa , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Pruebas de Micronúcleos , Pruebas de Mutagenicidad , Especies Reactivas de Oxígeno/metabolismo , Factores Sexuales , Adulto Joven
17.
J Cell Sci ; 123(Pt 15): 2586-95, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20605920

RESUMEN

RINGO/Speedy proteins are direct activators of Cdk1 and Cdk2 that have no sequence homology to cyclins. We have characterized the role in cell-cycle progression of a new human member of this protein family referred to as RINGO C. We show that siRNA-mediated knockdown of RINGO C results in premature mitotic exit with misaligned chromosomes, even in the presence of microtubule poisons. Time-lapse-microscopy experiments suggest that RINGO C is involved in the spindle-assembly checkpoint (SAC). Consistent with this idea, RINGO-C-depleted cells show impaired recruitment of the SAC components Mad2, Bub1 and BubR1. As the checkpoint is overridden, cells display defective chromosome segregation, which leads to an increased number of micronuclei and binucleated structures. Intriguingly, we found that RINGO C can associate with the mitotic kinase Aurora B, and downregulation of RINGO C produces mislocalization of the active form of Aurora B in prometaphase. Taken together, our results indicate a role for RINGO C in the mitotic checkpoint, which might be mediated by defective recruitment of SAC components and deregulation of the activity of Aurora kinase B.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Huso Acromático/metabolismo , Aurora Quinasa B , Aurora Quinasas , Proteínas de Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/genética , Citometría de Flujo , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Cinetocoros/metabolismo , Metafase/efectos de los fármacos , Metafase/genética , Microscopía Confocal , Microscopía Fluorescente , Mitosis/efectos de los fármacos , Mitosis/genética , Nocodazol/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Huso Acromático/efectos de los fármacos
18.
Mol Biol Rep ; 39(1): 251-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21556769

RESUMEN

Glutaraldehyde (GA) is a high production volume chemical that is very reactive with a wide spectrum of medical, scientific and industrial applications. Concerning the genotoxic and carcinogenic effect of GA, controversial results have been reported, while in humans no studies with positive carcinogenic results for GA have been published. However, our previous study concerning the combined effects of exposure to both GA and ionising radiation (IR) in peripheral blood lymphocytes of healthy donors has shown that non-genotoxic doses of the chemical induces a statistically significant increase in chromosomal radiosensitivity. The lack of information concerning the radiosensitizing potential of GA on cancerous cells triggered us to test the radiosensitizing effect of GA on breast cancer cells (MCF7). For this purpose the G2-chromosomal radiosensitivity assay (G2-assay) was used. The assay involves G2-phase irradiation and quantitation of the chromosomal fragility in the subsequent metaphase. The experimental data show that 48 h exposure to GA, at doses that are not clastogenic to MCF7 breast cancer cells enhances G2-chromosomal radiosensitivity of this cell line. In an effort to evaluate whether the observed increase in GAs-induced G2-chromosomal radiosensitization is linked to GA-induced alterations in the cell cycle and feedback control mechanism, Mitotic Index analysis was performed. The results have shown that such a mechanism cannot be directly related to the observed GA-induced increase in G2-chromosomal radiosensitivity. Since increased G2-chromosomal radiosensitivity has been linked with cancer proneness, the radiosensitizing effect of GA at non-clastogenic doses highlights its potential carcinogenic profile.


Asunto(s)
Cromosomas Humanos/efectos de los fármacos , Fase G2/efectos de los fármacos , Glutaral/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Bromodesoxiuridina , Línea Celular Tumoral , Cromosomas Humanos/efectos de la radiación , Femenino , Fase G2/efectos de la radiación , Humanos , Cariotipificación , Índice Mitótico , Intercambio de Cromátides Hermanas , Estadísticas no Paramétricas
19.
Histochem Cell Biol ; 135(3): 263-80, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21347609

RESUMEN

Here, we demonstrate that in HeLa cells, Ser317 of Chk1 undergoes phosphorylation in response to replication stress induced by hydroxyurea. We also demonstrate the existence of constitutive (interphase and mitotic) Chk1 kinase phosphorylation, the translocation of its phosphorylated form from the nucleus to cytoplasm in prometaphase as well as strong labeling of apoptotic nuclei with α-Chk1(S317) antibodies. Additionally, we show that caffeine, 2-aminopurine, staurosporine and sodium metavanadate can induce premature chromosome condensation (PCC) by the abrogation of the S-M checkpoint. Staurosporine appeared to be the most effective PCC inductor, and as in the case of the remaining inductors, the addition of hydroxyurea each time brought about an increase in the number of cells showing PCC symptoms (synergic effect). The forced premature mitosis was accompanied by an increasing index of double-strand breaks marked by the phosphorylation of histone H2AX on Ser139. Moreover, we found that the chemicals used brought about minor actin and tubulin network rearrangements that occurred following either replication stress or drug-induced cell cycle delay. At the same time, it was found that the extent of the cytoskeleton rearrangement did not hinder PCC in all its subperiods, i.e., from PCC-type prophase to PCC-type telophase.


Asunto(s)
Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Histonas/metabolismo , Proteínas Quinasas/metabolismo , Fase S/efectos de los fármacos , 2-Aminopurina/farmacología , Cafeína/farmacología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Células HeLa , Humanos , Fosforilación/efectos de los fármacos , Estaurosporina/farmacología , Relación Estructura-Actividad , Vanadatos/farmacología
20.
Cytometry A ; 79(12): 1016-22, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22052612

RESUMEN

Calyculin A-induced premature chromosome condensation (PCC) assay is a simple and useful method to assess structural and numerical chromosome aberrations in cells. Our hypothesis in this study is that suboptimum calyculin A induction of PCC resulting in fuzzy compactness and/or shortened length chromosomes would decrease the detection sensitivity of numerical and structural chromosome aberrations such as small PCC rings and small excess fragments. In this study, an optimization of calyculin A exposure on chromosome morphology and PCC induction frequency was investigated using a human peripheral blood lymphocyte (PBL) ex vivo irradiation ((60) Co-γ rays; ∼0.6 Gy/min; 0-30 Gy) model. Treatment with calyculin A (50 nM) for 15 and 30 min resulted in 11.3 ± 2.7 and 9.9 ± 1.6-fold increases in the frequency of G(2) /M-PCC cells with extended length chromosomes compared with the 60-min treated group over a broad dose range (0 to 20 Gy), respectively. The G(2) /M-PCC scoring index per PCC in 15- and 30-min treated groups was increased by 1.9 ± 0.2 (P = 0.001) and 1.8 ± 0.2 (P = 0.001) compared with the 60-min treated group over 0-20 Gy, respectively. The G(2) /M-PCC efficiency of 30-min treated group was highest in the three conditions (i.e., 15-, 30-, and 60-min treatment) of calyculin A exposure. Calyculin A (50 nM) treatment for 30 min before the 48-h harvest of mitogen-stimulated human PBL is optimum for the formation of suitable chromosome morphology necessary to assess structural chromosome aberrations induced by exposure to radiation using the chemical induced-PCC assay. Published 2011 Wiley Periodicals, Inc.


Asunto(s)
Aberraciones Cromosómicas/efectos de la radiación , Cromosomas Humanos/efectos de la radiación , Leucocitos Mononucleares/efectos de la radiación , Oxazoles/farmacología , Traumatismos por Radiación/diagnóstico , Bioensayo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Células Cultivadas , Cromosomas Humanos/efectos de los fármacos , Rayos gamma/efectos adversos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Toxinas Marinas , Mitógenos/farmacología , Traumatismos por Radiación/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda