Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Aging (Albany NY) ; 12(14): 14341-14354, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32668413

RESUMEN

Inactivating mutations in the liver kinase B1 (LKB1) tumor suppressor gene underlie Peutz-Jeghers syndrome (PJS) and occur frequently in various human cancers. We previously showed that LKB1 regulates centrosome duplication via PLK1. Here, we report that LKB1 further helps to maintain genomic stability through negative regulation of survivin, a member of the chromosomal passenger complex (CPC) that mediates CPC targeting to the centromere. We found that loss of LKB1 led to accumulation of misaligned and lagging chromosomes at metaphase and anaphase and increased the appearance of multi- and micro-nucleated cells. Ectopic LKB1 expression reduced these features and improved mitotic fidelity in LKB1-deficient cells. Through pharmacological and genetic manipulations, we showed that LKB1-mediated repression of survivin is independent of AMPK, but requires p53. Consistent with the key influence of LKB1 on survivin expression, immunohistochemical analysis indicated that survivin is highly expressed in intestinal polyps from a PJS patient. Lastly, we reaffirm a potential therapeutic avenue to treat LKB1-mutated tumors by demonstrating the increased sensitivity to survivin inhibitors of LKB1-deficient cells.


Asunto(s)
Centrómero/efectos de los fármacos , Genes p53/efectos de los fármacos , Genoma/efectos de los fármacos , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Survivin/biosíntesis , Survivin/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Línea Celular Tumoral , Aberraciones Cromosómicas , Humanos , Pólipos Intestinales/genética , Mitosis/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba/genética
2.
Artículo en Inglés | MEDLINE | ID: mdl-32247554

RESUMEN

BACKGROUND: The cytokinesis-block micronucleus (CBMN) assay is an internationally recognized method for measuring DNA damage after exposure to genotoxic agents, as well as a biomarker for DNA repair and chromosomal instability. The high baseline level of micronuclei (MN) in the healthy population has limited the sensitivity and application of the CBMN assay for the follow-up of exposed populations. We reevaluated the sensitivity of the CBNM assay using semi-automated MN scoring following telomere and centromere (TC) staining after in vitro exposure to genotoxic agents (mitomycin or radiation) or aneugenic agents (vinblastine). MATERIALS AND METHODS: Blood samples from 12 healthy donors were exposed to 137Cs at seven doses from 0.1-4 Gy and cultured for 72 h. Cytochalasin B was added at 46 h of culture. The exposure of chemical agents (mitomycin or vinblastine) was performed after 48 h of culture for 3 h. Cytochalasin B was added after treatment and slides were prepared 24 h after. MN was semi-automatically scored following TC staining. Nucleoplasmic bridges (NPBs) and nuclear buds (NBUDs) were assessed in a human cell line after TC staining. RESULTS: The introduction TC staining to the scoring of MN not only renders MN scoring more efficient and robust, but also permits discrimination between exposure to clastogenic (MN with only telomere signals) and aneugenic agents (MN with both TC signals). The resulting improvement of MN detection led to an increase in the sensitivity of the CBMN assay following low-dose radiation exposure (0.3 versus 0.1 Gy). Hyperradiosensitivity phenomenon was observed after low dose exposure. A dose-response curve was obtained for up to 4 Gy. In addition, TC staining permits assessment of the nature of NPBs and NBUDs as biomarkers for genotoxicity and chromosomal instability. CONCLUSION: These approaches can be potentially used to follow-up populations exposed to genotoxic agents and assess cancer risk.


Asunto(s)
Centrómero/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Pruebas de Mutagenicidad , Telómero/efectos de los fármacos , Aneugénicos/farmacología , Centrómero/genética , Citocinesis/efectos de los fármacos , Citocinesis/genética , Daño del ADN/genética , Humanos , Linfocitos/efectos de los fármacos , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Pruebas de Micronúcleos , Mutágenos/toxicidad , Medición de Riesgo , Telómero/genética
3.
J Cell Biol ; 218(1): 134-149, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30396998

RESUMEN

The centromere is an important genomic locus for chromosomal segregation. Although the centromere is specified by sequence-independent epigenetic mechanisms in most organisms, it is usually composed of highly repetitive sequences, which associate with heterochromatin. We have previously generated various chicken DT40 cell lines containing differently positioned neocentromeres, which do not contain repetitive sequences and do not associate with heterochromatin. In this study, we performed systematic 4C analysis using three cell lines containing differently positioned neocentromeres to identify neocentromere-associated regions at the 3D level. This analysis reveals that these neocentromeres commonly associate with specific heterochromatin-rich regions, which were distantly located from neocentromeres. In addition, we demonstrate that centromeric chromatin adopts a compact structure, and centromere clustering also occurs in vertebrate interphase nuclei. Interestingly, the occurrence of centromere-heterochromatin associations depend on CENP-H, but not CENP-C. Our analyses provide an insight into understanding the 3D architecture of the genome, including the centromeres.


Asunto(s)
Centrómero/ultraestructura , Proteínas Cromosómicas no Histona/genética , Epigénesis Genética , Genoma , Heterocromatina/ultraestructura , Animales , Línea Celular Tumoral , Centrómero/efectos de los fármacos , Centrómero/metabolismo , Pollos , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica/efectos de los fármacos , Citometría de Flujo , Heterocromatina/efectos de los fármacos , Heterocromatina/metabolismo , Ácidos Indolacéticos/farmacología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Linfocitos/ultraestructura , Metiltransferasas/genética , Metiltransferasas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
4.
Drug Chem Toxicol ; 40(4): 457-462, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27936973

RESUMEN

Vanadium is a widely distributed metal in the Earth's surface and is released into the environment by either natural or anthropogenic causes. Vanadium (III) oxide (V2O3) is present in the environment, and many organisms are exposed to this compound; however, its effects at the cellular and genetic levels are still unknown. Therefore, in this study, the ability of V2O3 to induce chromosomal damage and impair cell proliferation was tested on human leukocytes in vitro. The cultures cells were treated for 48 h with different concentrations 2, 4, 8 or 16 µg/mL of V2O3, and we use the sister chromatid exchange's (SCE) test and the viability assay to evaluate the effects. In the results, no change was observed in either the viability or the frequency of SCE; however, a significant increase was observed in the incidence of premature chromatid separation (PCS), and a decrease was observed in both the mitotic index (MI) and the replication index (RI). Therefore, it can be suggested that V2O3 induces a genotoxic effect at the centromere level, indicating that it is a cause of aneuploidy that is capable of altering cell cycle progression.


Asunto(s)
Carcinógenos Ambientales/toxicidad , Centrómero/efectos de los fármacos , Cromátides/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Leucocitos/efectos de los fármacos , Óxidos/toxicidad , Compuestos de Vanadio/toxicidad , Adulto , Aneugénicos/toxicidad , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Centrómero/metabolismo , Cromátides/metabolismo , Humanos , Leucocitos/citología , Leucocitos/inmunología , Leucocitos/metabolismo , Masculino , Índice Mitótico , Pruebas de Mutagenicidad , Concentración Osmolar , Intercambio de Cromátides Hermanas/efectos de los fármacos , Adulto Joven
5.
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
6.
Methods Mol Biol ; 1510: 47-59, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27761812

RESUMEN

The chromosomal passenger complex (CPC) is a key regulator of cell division. Its proper localization during the different phases of mitosis and cytokinesis is crucial for the exertion of its various functions. HDACi treatment has been demonstrated to disturb the centromeric localization of the CPC in tumor cells, thus leading to severe mitotic defects often followed by apoptosis. In this chapter, we describe how HDACi-induced changes of the CPC localization can be analyzed by indirect immunofluorescence using CPC-specific primary and fluorophore-coupled secondary antibodies followed by confocal microscopy.


Asunto(s)
Antineoplásicos/farmacología , Centrómero/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Apoptosis/efectos de los fármacos , Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Centrómero/ultraestructura , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Citocinesis/efectos de los fármacos , Técnica del Anticuerpo Fluorescente/métodos , Células HeLa , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Hidroxiurea/farmacología , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Microscopía Confocal/métodos , Mitosis/efectos de los fármacos , Survivin
7.
PLoS One ; 11(11): e0165873, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27806100

RESUMEN

Heterochromatinisation of pericentromeres, which in mice consist of arrays of major satellite repeats, are important for centromere formation and maintenance of genome stability. The dysregulation of this process has been linked to genomic stress and various cancers. Here we show in mice that the proteasome binds to major satellite repeats and proteasome inhibition by MG132 results in their transcriptional de-repression; this de-repression is independent of cell-cycle perturbation. The transcriptional activation of major satellite repeats upon proteasome inhibition is accompanied by delocalisation of heterochromatin protein 1 alpha (HP1α) from chromocentres, without detectable change in the levels of histone H3K9me3, H3K4me3, H3K36me3 and H3 acetylation on the major satellite repeats. Moreover, inhibition of the proteasome was found to increase the number of chromocentres per cell, reflecting destabilisation of the chromocentre structures. Our findings suggest that the proteasome plays a role in maintaining heterochromatin integrity of pericentromeres.


Asunto(s)
Centrómero/química , Proteínas Cromosómicas no Histona/metabolismo , ADN Satélite/genética , Leupeptinas/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Acetilación , Animales , Centrómero/efectos de los fármacos , Centrómero/genética , Cromatina/química , Cromatina/genética , Homólogo de la Proteína Chromobox 5 , Inestabilidad Cromosómica , ADN Satélite/efectos de los fármacos , Histonas/metabolismo , Hibridación Fluorescente in Situ , Ratones , Células 3T3 NIH , Transcripción Genética/efectos de los fármacos
8.
PLoS One ; 11(4): e0153518, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27082996

RESUMEN

OTSSP167 was recently characterized as a potent inhibitor for maternal embryonic leucine zipper kinase (MELK) and is currently tested in Phase I clinical trials for solid tumors that have not responded to other treatment. Here we report that OTSSP167 abrogates the mitotic checkpoint at concentrations used to inhibit MELK. The abrogation is not recapitulated by RNAi mediated silencing of MELK in cells. Although OTSSP167 indeed inhibits MELK, it exhibits off-target activity against Aurora B kinase in vitro and in cells. Furthermore, OTSSP167 inhibits BUB1 and Haspin kinases, reducing phosphorylation at histones H2AT120 and H3T3 and causing mislocalization of Aurora B and associated chromosomal passenger complex from the centromere/kinetochore. The results suggest that OTSSP167 may have additional mechanisms of action for cancer cell killing and caution the use of OTSSP167 as a MELK specific kinase inhibitor in biochemical and cellular assays.


Asunto(s)
Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Naftiridinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Anticuerpos/farmacología , Aurora Quinasa B/antagonistas & inhibidores , Centrómero/efectos de los fármacos , Centrómero/fisiología , Células HeLa , Humanos , Cinetocoros/efectos de los fármacos , Cinetocoros/fisiología , Células MCF-7 , Mitosis/efectos de los fármacos , Mitosis/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Transducción de Señal/efectos de los fármacos
9.
Cell Cycle ; 15(2): 213-24, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26636626

RESUMEN

Haspin-catalyzed histone H3 threonine 3 (Thr3) phosphorylation facilitates chromosomal passenger complex (CPC) docking at centromeres, regulating indirectly chromosome behavior during somatic mitosis. It is not fully known about the expression and function of H3 with phosphorylated Thr3 (H3T3-P) during meiosis in oocytes. In this study, we investigated the expression and sub-cellular distribution of H3T3-P, as well as its function in mouse oocytes during meiotic division. Western blot analysis revealed that H3T3-P expression was only detected after germinal vesicle breakdown (GVBD), and gradually increased to peak level at metaphase I (MI), but sharply decreased at metaphase II (MII). Immunofluorescence showed H3T3-P was only brightly labeled on chromosomes after GVBD, with relatively high concentration across the whole chromosome axis from pro-metaphase I (pro-MI) to MI. Specially, H3T3-P distribution was exclusively limited to the local space between sister centromeres at MII stage. Haspin inhibitor, 5-iodotubercidin (5-ITu), dose- and time-dependently blocked H3T3-P expression in mouse oocytes. H3T3-P inhibition delayed the resumption of meiosis (GVBD) and chromatin condensation. Moreover, the loss of H3T3-P speeded up the meiotic transition to MII of pro-MI oocytes in spite of the presence of non-aligned chromosomes, even reversed MI-arrest induced with Nocodazole. The inhibition of H3T3-P expression distinguishably damaged MAD1 recruitment on centromeres, which indicates the spindle assembly checkpoint was impaired in function, logically explaining the premature onset of anaphase I. Therefore, Haspin-catalyzed histone H3 phosphorylation is essential for chromatin condensation and the following timely transition from meiosis I to meiosis II in mouse oocytes during meiotic division.


Asunto(s)
Regulación de la Expresión Génica , Histonas/genética , Meiosis/efectos de los fármacos , Oocitos/metabolismo , Treonina/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrómero/efectos de los fármacos , Centrómero/metabolismo , Centrómero/ultraestructura , Cromatina/efectos de los fármacos , Cromatina/metabolismo , Cromatina/ultraestructura , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Células del Cúmulo/citología , Células del Cúmulo/efectos de los fármacos , Células del Cúmulo/metabolismo , Femenino , Histonas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mitosis/efectos de los fármacos , Nocodazol/farmacología , Oocitos/citología , Oocitos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transducción de Señal , Factores de Tiempo , Tubercidina/análogos & derivados , Tubercidina/farmacología
10.
PLoS One ; 10(3): e0119347, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25764183

RESUMEN

Condensin, a central player in eukaryotic chromosomal dynamics, contains five evolutionarily-conserved subunits. Two SMC (structural maintenance of chromosomes) subunits contain ATPase, hinge, and coiled-coil domains. One non-SMC subunit is similar to bacterial kleisin, and two other non-SMC subunits contain HEAT (similar to armadillo) repeats. Here we report isolation and characterization of 21 fission yeast (Schizosaccharomyces pombe) mutants for three non-SMC subunits, created using error-prone mutagenesis that resulted in single-amino acid substitutions. Beside condensation, segregation, and DNA repair defects, similar to those observed in previously isolated SMC and cnd2 mutants, novel phenotypes were observed for mutants of HEAT-repeats containing Cnd1 and Cnd3 subunits. cnd3-L269P is hypersensitive to the microtubule poison, thiabendazole, revealing defects in kinetochore/centromere and spindle assembly checkpoints. Three cnd1 and three cnd3 mutants increased cell size and doubled DNA content, thereby eliminating the haploid state. Five of these mutations reside in helix B of HEAT repeats. Two non-SMC condensin subunits, Cnd1 and Cnd3, are thus implicated in ploidy maintenance.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Reparación del ADN , ADN de Hongos/metabolismo , Ploidias , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Centrómero/efectos de los fármacos , Cromosomas Fúngicos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación , Estructura Terciaria de Proteína , Schizosaccharomyces/metabolismo , Tiabendazol/farmacología
11.
Neoplasia ; 16(10): 861-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25379022

RESUMEN

Human epidermal growth factor receptor 2 (HER2) and topoisomerase II alpha (TOP2A) genes have been proposed as predictive biomarkers of sensitivity to anthracycline chemotherapy. Recently, chromosome 17 centromere enumeration probe (CEP17) duplication has also been associated with increased responsiveness to anthracyclines. However, reports are conflicting and none of these tumor markers can yet be considered a clinically reliable predictor of response to anthracyclines. We studied the association of TOP2A gene alterations, HER2 gene amplification, and CEP17 duplication with response to anthracycline-based neoadjuvant chemotherapy in 140 patients with operable or locally advanced breast cancer. HER2 was tested by fluorescence in situ hybridization and TOP2A and CEP17 by chromogenic in situ hybridization. Thirteen patients (9.3%) achieved pathologic complete response (pCR). HER2 amplification was present in 24 (17.5%) of the tumors. TOP2A amplification occurred in seven tumors (5.1%). CEP17 duplication was detected in 13 patients (9.5%). CEP17 duplication correlated with a higher rate of pCR [odds ratio (OR) 6.55, 95% confidence interval (95% CI) 1.25-34.29, P = .026], and analysis of TOP2A amplification showed a trend bordering on statistical significance (OR 6.97, 95% CI 0.96-50.12, P = .054). TOP2A amplification and CEP17 duplication combined were strongly associated with pCR (OR 6.71, 95% CI 1.66-27.01, P = .007). HER2 amplification did not correlate with pCR. Our results suggest that CEP17 duplication predicts pCR to primary anthracycline-based chemotherapy. CEP17 duplication, TOP2A amplifications, and HER2 amplifications were not associated with prognosis.


Asunto(s)
Antraciclinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Centrómero/efectos de los fármacos , Cromosomas Humanos Par 17 , Adulto , Anciano , Anciano de 80 o más Años , Antibióticos Antineoplásicos/farmacología , Antígenos de Neoplasias/genética , Neoplasias de la Mama/mortalidad , Duplicación Cromosómica , ADN-Topoisomerasas de Tipo II/genética , Proteínas de Unión al ADN/genética , Femenino , Amplificación de Genes , Humanos , Hibridación Fluorescente in Situ , Persona de Mediana Edad , Terapia Neoadyuvante , Proteínas de Unión a Poli-ADP-Ribosa , Pronóstico , Receptor ErbB-2/genética , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento
12.
PLoS One ; 9(6): e95184, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24893165

RESUMEN

Pearson correlation coefficient for expression analysis of the Lymphoma/Leukemia Molecular Profiling Project (LLMPP) demonstrated Aurora A and B are highly correlated with MYC in DLBCL and mantle cell lymphoma (MCL), while both Auroras correlate with BCL2 only in DLBCL. Auroras are up-regulated by MYC dysregulation with associated aneuploidy and resistance to microtubule targeted agents such as vincristine. Myc and Bcl2 are differentially expressed in U-2932, TMD-8, OCI-Ly10 and Granta-519, but only U-2932 cells over-express mutated p53. Alisertib [MLN8237 or M], a highly selective small molecule inhibitor of Aurora A kinase, was synergistic with vincristine [VCR] and rituximab [R] for inhibition of cell proliferation, abrogation of cell cycle checkpoints and enhanced apoptosis versus single agent or doublet therapy. A DLBCL (U-2932) mouse model showed tumor growth inhibition (TGI) of ∼ 10-20% (p = 0.001) for M, VCR and M-VCR respectively, while R alone showed ∼ 50% TGI (p = 0.001). M-R and VCR-R led to tumor regression [TR], but relapsed 10 days after discontinuing therapy. In contrast, M-VCR-R demonstrated TR with no relapse >40 days after stopping therapy with a Kaplan-Meier survival of 100%. Genes that are modulated by M-VCR-R (CENP-C, Auroras) play a role in centromere-kinetochore function in an attempt to maintain mitosis in the presence of synthetic lethality. Together, our data suggest that the interaction between alisertib plus VCR plus rituximab is synergistic and synthetic lethal in Myc and Bcl-2 co-expressing DLBCL. Alisertib plus vincristine plus rituximab [M-VCR-R] may represent a new strategy for DLBCL therapy.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Azepinas/uso terapéutico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirimidinas/uso terapéutico , Vincristina/uso terapéutico , Animales , Anticuerpos Monoclonales de Origen Murino/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Aurora Quinasas/antagonistas & inhibidores , Aurora Quinasas/metabolismo , Azepinas/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Centrómero/efectos de los fármacos , Centrómero/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mitosis/efectos de los fármacos , Invasividad Neoplásica , Pirimidinas/farmacología , Rituximab , Proteína p53 Supresora de Tumor/metabolismo , Vincristina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Prostate ; 74(10): 999-1011, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24802614

RESUMEN

BACKGROUND: Previous studies have shown that COX-2 inhibitors inhibit cancer cell proliferation. However, the molecular mechanism remains elusive. METHODS: Prostate cancer LNCaP, 22Rv1, and PC3 cells were cultured and treated with the COX-2 inhibitors celecoxib and CAY10404. Knockdown of COX-2 in LNCaP cells was carried out using lentiviral vector-loaded COX-2 shRNA. Cell cycle progression and cell proliferation were analyzed by flow cytometry, microscopy, cell counting, and the MTT assay. The antagonists of EP1, EP2, EP3, and EP4 were used to examine the effects of the PGE2 signaling. The effect of COX-2 inhibitors and COX-2 knockdown on expression of the kinetochore/centromere genes and proteins was determined by RT-PCR and immunoblotting. RESULTS: Treatment with the COX-2 inhibitors celecoxib and CAY10404 or knockdown of COX-2 significantly inhibited prostate cancer cell proliferation. Flow-cytometric analysis and immunofluorescent staining confirmed the cell cycle arrested at the G2/M phase. Biochemical analysis showed that inhibition of COX-2 or suppression of COX-2 expression induced a dramatic down-regulation of key proteins in the kinetochore/centromere assembly, such as ZWINT, Cdc20, Ndc80, CENP-A, Bub1, and Plk1. Furthermore, the EP1 receptor antagonist SC51322, but not the EP2, EP3, and EP4 receptor antagonists, produced similar effects to the COX-2 inhibitors on cell proliferation and down-regulation of kinetochore/centromere proteins, suggesting that the effect of the COX-2 inhibition is through inactivation of the EP1 receptor signaling. CONCLUSIONS: Our studies indicate that inhibition of COX-2 can arrest prostate cancer cell cycle progression through inactivation of the EP1 receptor signaling and down-regulation of kinetochore/centromere proteins.


Asunto(s)
Centrómero/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Cinetocoros/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Autoantígenos/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Centrómero/metabolismo , Proteína A Centromérica , Proteínas Cromosómicas no Histona/genética , Ciclooxigenasa 2/fisiología , Regulación hacia Abajo , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Cinetocoros/metabolismo , Masculino , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasas/fisiología , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Subtipo EP1 de Receptores de Prostaglandina E/fisiología
14.
Chromosoma ; 123(1-2): 129-46, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24013524

RESUMEN

Sister chromatid cohesion is regulated by cohesin complexes and topoisomerase IIα. Although relevant studies have shed some light on the relationship between these two mechanisms of cohesion during mammalian mitosis, their interplay during mammalian meiosis remains unknown. In the present study, we have studied the dynamics of topoisomerase IIα in relation to that of the cohesin subunits RAD21 and REC8, the shugoshin-like 2 (Schizosaccharomyces pombe) (SGOL2) and the polo-like kinase 1-interacting checkpoint helicase (PICH), during both male mouse meiotic divisions. Our results strikingly show that topoisomerase IIα appears at stretched strands connecting the sister kinetochores of segregating early anaphase II chromatids, once the cohesin complexes have been removed from the centromeres. Moreover, the number and length of these topoisomerase IIα-connecting strands increase between lagging chromatids at anaphase II after the chemical inhibition of the enzymatic activity of topoisomerase IIα by etoposide. Our results also show that the etoposide-induced inhibition of topoisomerase IIα is not able to rescue the loss of centromere cohesion promoted by the absence of the shugoshin SGOL2 during anaphase I. Taking into account our results, we propose a two-step model for the sequential release of centromeric cohesion during male mammalian meiosis II. We suggest that the cohesin removal is a prerequisite for the posterior topoisomerase IIα-mediated resolution of persisting catenations between segregating chromatids during anaphase II.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/metabolismo , Mamíferos/metabolismo , Meiosis , Anafase/efectos de los fármacos , Animales , Centrómero/efectos de los fármacos , Cromátides/efectos de los fármacos , Cromátides/metabolismo , Etopósido/farmacología , Masculino , Meiosis/efectos de los fármacos , Metafase/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mitosis/efectos de los fármacos , Cohesinas
15.
G3 (Bethesda) ; 3(8): 1301-10, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-23733889

RESUMEN

The severity of the toxic side effects of chemotherapy varies among patients, and much of this variation is likely genetically based. Here, we use the model system Drosophila melanogaster to genetically dissect the toxicity of methotrexate (MTX), a drug used primarily to treat childhood acute lymphoblastic leukemia and rheumatoid arthritis. We use the Drosophila Synthetic Population Resource, a panel of recombinant inbred lines derived from a multiparent advanced intercross, and quantify MTX toxicity as a reduction in female fecundity. We identify three quantitative trait loci (QTL) affecting MTX toxicity; two colocalize with the fly orthologs of human genes believed to mediate MTX toxicity and one is a novel MTX toxicity gene with a human ortholog. A fourth suggestive QTL spans a centromere. Local single-marker association scans of candidate gene exons fail to implicate amino acid variants as the causative single-nucleotide polymorphisms, and we therefore hypothesize the causative variation is regulatory. In addition, the effects at our mapped QTL do not conform to a simple biallelic pattern, suggesting multiple causative factors underlie the QTL mapping results. Consistent with this observation, no single single-nucleotide polymorphism located in or near a candidate gene can explain the QTL mapping signal. Overall, our results validate D. melanogaster as a model for uncovering the genetic basis of chemotoxicity and suggest the genetic basis of MTX toxicity is due to a handful of genes each harboring multiple segregating regulatory factors.


Asunto(s)
Drosophila melanogaster/efectos de los fármacos , Fertilidad/efectos de los fármacos , Metotrexato/toxicidad , Animales , Centrómero/efectos de los fármacos , Centrómero/genética , Mapeo Cromosómico , Elementos Transponibles de ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Glutatión Peroxidasa/genética , Humanos , Modelos Genéticos , Fenotipo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo
16.
Mutat Res ; 755(1): 73-80, 2013 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-23726961

RESUMEN

When characterizing the genotoxicity of chemicals that induce micronuclei, it is practical to be able to classify the chemicals as aneugens or clastogens. This classification gives information on the mechanistic properties of chemicals and is indispensable for setting the threshold safety margins for genotoxicity in pharmaceutical development. A widely used method for detecting aneugens is fluorescence in situ hybridization (FISH) but, even though the rat is an experimental animal generally used in preclinical studies in drug development, DNA probes that hybridize to all the centromeres of rat chromosomes have not yet been established. In the present study, in addition to the previously known satellite I sequence, we identified two novel satellite sequences, satellite II and satellite III, from the rat genome database. DNA probes with a mixture of these satellite DNA sequences were used to establish a FISH method for pan-centromeric staining of rat chromosomes. To confirm the feasibility of the method, vinblastine (VBS) and mitomycin C (MMC) were administered to rats as a typical aneugen and clastogen, respectively. Micronucleated polychromatic erythrocytes (MNPCE) from bone marrow were enriched by sorting in flow cytometry and subjected to the FISH method. As a result, the ratio of centromere-positive MNPCE increased in VBS-treated rats but not in MMC-treated ones. Since the FISH method using the novel DNA probes clearly discriminates the aneugens from the clastogens, we suggest this method as a useful tool for providing mechanistic information for micronucleus induction in vivo.


Asunto(s)
Aneugénicos/toxicidad , Médula Ósea/efectos de los fármacos , Centrómero/efectos de los fármacos , Sondas de ADN , Hibridación Fluorescente in Situ , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Mutágenos/toxicidad , Animales , Antibióticos Antineoplásicos/toxicidad , Secuencia de Bases , Centrómero/genética , Cromosomas/genética , Colchicina/toxicidad , ADN Satélite/análisis , ADN Satélite/genética , Eritrocitos/efectos de los fármacos , Citometría de Flujo , Masculino , Pruebas de Micronúcleos , Mitomicina/toxicidad , Datos de Secuencia Molecular , Ratas , Homología de Secuencia de Ácido Nucleico , Moduladores de Tubulina/toxicidad , Vinblastina/toxicidad
17.
PLoS One ; 8(1): e53702, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23349730

RESUMEN

Integrin-linked kinase (ILK) localizes to both focal adhesions and centrosomes in distinct multiprotein complexes. Its dual function as a kinase and scaffolding protein has been well characterized at focal adhesions, where it regulates integrin-mediated cell adhesion, spreading, migration and signaling. At the centrosomes, ILK regulates mitotic spindle organization and centrosome clustering. Our previous study showed various spindle defects after ILK knockdown or inhibition that suggested alteration in microtubule dynamics. Since ILK expression is frequently elevated in many cancer types, we investigated the effects of ILK overexpression on microtubule dynamics. We show here that overexpressing ILK in HeLa cells was associated with a shorter duration of mitosis and decreased sensitivity to paclitaxel, a chemotherapeutic agent that suppresses microtubule dynamics. Measurement of interphase microtubule dynamics revealed that ILK overexpression favored microtubule depolymerization, suggesting that microtubule destabilization could be the mechanism behind the decreased sensitivity to paclitaxel, which is known to stabilize microtubules. Conversely, the use of a small molecule inhibitor selective against ILK, QLT-0267, resulted in suppressed microtubule dynamics, demonstrating a new mechanism of action for this compound. We further show that treatment of HeLa cells with QLT-0267 resulted in higher inter-centromere tension in aligned chromosomes during mitosis, slower microtubule regrowth after cold depolymerization and the presence of a more stable population of spindle microtubules. These results demonstrate that ILK regulates microtubule dynamics in both interphase and mitotic cells.


Asunto(s)
Interfase , Microtúbulos/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Compuestos Azo/farmacología , Centrómero/efectos de los fármacos , Centrómero/metabolismo , Células HeLa , Humanos , Interfase/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Mitosis/efectos de los fármacos , Paclitaxel/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Factores de Tiempo
18.
J Appl Toxicol ; 33(6): 426-33, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22081495

RESUMEN

The mechanism of genotoxic potential of the cancer chemotherapeutic drugs amsacrine and nocodazole in mouse bone marrow was investigated using a micronucleus test complemented by fluorescence in situ hybridization assay with mouse centromeric and telomeric DNA probes. In animals treated with different doses of amsacrine (0.5-12 mg kg(-1) ), the frequencies of micronucleated polychromatic erythrocytes increased significantly after treatment with 9 and 12 mg kg(-1) . A statistically significant increase in micronuclei frequency was also detected for 75 mg kg(-1) nocodazole (two exposures, spaced 24 h apart). Both compounds caused significant suppressions of erythroblast proliferation at higher doses. Furthermore, the present study demonstrated for the first time that amsacrine has high incidences of clastogenicity and low incidences of aneugenicity whereas nocodazole has high incidences of aneugenicity and low incidences of clastogenicity during mitotic phases in vivo. The assay also showed that chromosomes can be enclosed in the micronuclei before and after centromere separation. Therefore, the clinical use of these genotoxic drugs must be weighed against the risks of the development of chromosomal aberrations in cancer patients and medical personnel exposed to drug regimens that include these chemicals.


Asunto(s)
Amsacrina/toxicidad , Antineoplásicos/toxicidad , Células de la Médula Ósea/efectos de los fármacos , Mutágenos , Nocodazol/toxicidad , Animales , Antibióticos Antineoplásicos/toxicidad , Centrómero/efectos de los fármacos , Colchicina/toxicidad , Citogenética , Sondas de ADN , Relación Dosis-Respuesta a Droga , Eritrocitos/efectos de los fármacos , Eritrocitos/ultraestructura , Hibridación in Situ , Técnicas In Vitro , Masculino , Ratones , Pruebas de Micronúcleos , Mitomicina/toxicidad
19.
Cell Biol Toxicol ; 28(5): 331-42, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22843076

RESUMEN

Genistein, due to its recognized chemopreventive and antitumour potential, is a molecule of interest as a lead compound in drug design. Recently, we found that the novel genistein derivative, [7-O-(2,3,4,6-tetra-O-acetyl-ß-D-galactopyranosyl)-(1 → 4)-(6-O-acetyl-hex-2-ene-α-D-erythro pyranosyl)genistein, named G21, induced aberrations in mitotic spindle formation. In the presented study, we investigated the properties of G21 relevant to its genotoxic activity. The inhibition of topoisomerase IIα activity was evaluated in decatenation assay and immunoband depletion assay, the covalent DNA-topoisomerase IIα complexes and histone É£H2AX were detected immunofluorescently. Genotoxic effects of the tested compounds were assessed in micronucleation assay. The presence of centromeres in the micronuclei and the multiplication of centrosomes were evaluated in fluorescence immunolabelled specimens. The inhibition of tubulin polymerization was measured spectrophotometrically. We found that both tested drugs were able to inhibit topoisomerase II activity; however, G21, in contrast to genistein, blocked this enzyme at the concentration far exceeding cytotoxic IC(50). We also found that both compounds caused micronucleation in DU 145 prostate cancer cells, but in contrast to genistein, G21 exhibited aneugenic activity, manifested by the presence of centromeres in micronuclei formed in cells treated with the drug. Aneugenic properties of G21 resulted from the inhibition of tubulin polymerization and centrosome disruption, not observed in the presence of genistein. The study supports and extends our previous observations that the mechanisms of cytotoxicity of genistein and its new glycosidic derivative-G21 are significantly different.


Asunto(s)
Aneugénicos/farmacología , Disacáridos/química , Genisteína/análogos & derivados , Genisteína/farmacología , Glicósidos/farmacología , Inhibidores de Topoisomerasa II/farmacología , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Centrómero/efectos de los fármacos , Centrómero/patología , Centrosoma/efectos de los fármacos , Centrosoma/patología , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Técnica del Anticuerpo Fluorescente , Genisteína/química , Glicósidos/química , Histonas/metabolismo , Humanos , Masculino , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Polimerizacion , Inhibidores de Topoisomerasa II/química , Tubulina (Proteína)/metabolismo
20.
J Exp Ther Oncol ; 9(3): 221-30, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22070054

RESUMEN

Diets rich in fruits and vegetables have been associated with reduced risks for many types of cancers. Avocado (Persea americana Mill.) is a widely consumed fruit containing many cancer preventing nutrients, vitamins and phytochemicals. Studies have shown that phytochemicals extracted from the avocado fruit selectively induce cell cycle arrest, inhibit growth, and induce apoptosis in precancerous and cancer cell lines. Our recent studies indicate that phytochemicals extracted with 50% Methanol from avocado fruits help in proliferation of human lymphocyte cells and decrease chromosomal aberrations induced by cyclophosphamide. Among three concentrations (100 mg, 150 mg and 200 mg per Kg Body Weight), the most effective conc. of extract was 200 mg/Kg Body Wt. It decreased significant level of numerical and structural aberrations (breaks, premature centromeric division etc. up to 88%, p < 0.0001)), and accrocentric associtation within D & G group (up to 78%, p = 0.0008). These studies suggest that phytochemicals from the avocado fruit can be utilized for making active chemoprotective ingredient for lowering the side effect of chemotherapy like cyclophosphamide in cancer therapy.


Asunto(s)
Aberraciones Cromosómicas/efectos de los fármacos , Frutas/química , Persea/química , Extractos Vegetales/farmacología , Células Cultivadas , Centrómero/efectos de los fármacos , Centrómero/genética , Quimioprevención , Aberraciones Cromosómicas/inducido químicamente , Ciclofosfamida/toxicidad , Humanos , Linfocitos , Metafase/genética , Extractos Vegetales/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA