Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 16(10): e1008623, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33052904

RESUMO

Plant cells undergo two types of cell cycles-the mitotic cycle in which DNA replication is coupled to mitosis, and the endocycle in which DNA replication occurs in the absence of cell division. To investigate DNA replication programs in these two types of cell cycles, we pulse labeled intact root tips of maize (Zea mays) with 5-ethynyl-2'-deoxyuridine (EdU) and used flow sorting of nuclei to examine DNA replication timing (RT) during the transition from a mitotic cycle to an endocycle. Comparison of the sequence-based RT profiles showed that most regions of the maize genome replicate at the same time during S phase in mitotic and endocycling cells, despite the need to replicate twice as much DNA in the endocycle and the fact that endocycling is typically associated with cell differentiation. However, regions collectively corresponding to 2% of the genome displayed significant changes in timing between the two types of cell cycles. The majority of these regions are small with a median size of 135 kb, shift to a later RT in the endocycle, and are enriched for genes expressed in the root tip. We found larger regions that shifted RT in centromeres of seven of the ten maize chromosomes. These regions covered the majority of the previously defined functional centromere, which ranged between 1 and 2 Mb in size in the reference genome. They replicate mainly during mid S phase in mitotic cells but primarily in late S phase of the endocycle. In contrast, the immediately adjacent pericentromere sequences are primarily late replicating in both cell cycles. Analysis of CENH3 enrichment levels in 8C vs 2C nuclei suggested that there is only a partial replacement of CENH3 nucleosomes after endocycle replication is complete. The shift to later replication of centromeres and possible reduction in CENH3 enrichment after endocycle replication is consistent with a hypothesis that centromeres are inactivated when their function is no longer needed.


Assuntos
Período de Replicação do DNA/genética , Replicação do DNA/efeitos dos fármacos , Raízes de Plantas/genética , Zea mays/genética , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/genética , Centrômero/efeitos dos fármacos , Centrômero/genética , Replicação do DNA/genética , Período de Replicação do DNA/efeitos dos fármacos , DNA de Plantas/efeitos dos fármacos , DNA de Plantas/genética , Desoxiuridina/análogos & derivados , Desoxiuridina/farmacologia , Endocitose/efeitos dos fármacos , Meristema/efeitos dos fármacos , Meristema/genética , Mitose/efeitos dos fármacos , Mitose/genética , Nucleossomos/efeitos dos fármacos , Raízes de Plantas/efeitos dos fármacos , Raízes de Plantas/crescimento & desenvolvimento , Fase S/genética , Zea mays/crescimento & desenvolvimento
2.
Development ; 144(19): 3475-3486, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28851706

RESUMO

Mouse oocytes carrying DNA damage arrest in meiosis I, thereby preventing creation of embryos with deleterious mutations. The arrest is dependent on activation of the spindle assembly checkpoint, which results in anaphase-promoting complex (APC) inhibition. However, little is understood about how this checkpoint is engaged following DNA damage. Here, we find that within minutes of DNA damage checkpoint proteins are assembled at the kinetochore, not at damage sites along chromosome arms, such that the APC is fully inhibited within 30 min. Despite this robust response, there is no measurable loss in k-fibres, or tension across the bivalent. Through pharmacological inhibition we observed that the response is dependent on Mps1 kinase, aurora kinase and Haspin. Using oocyte-specific knockouts we find the response does not require the DNA damage response kinases ATM or ATR. Furthermore, checkpoint activation does not occur in response to DNA damage in fully mature eggs during meiosis II, despite the divisions being separated by just a few hours. Therefore, mouse oocytes have a unique ability to sense DNA damage rapidly by activating the checkpoint at their kinetochores.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Dano ao DNA , Cinetocoros/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular , Meiose , Oócitos/citologia , Oócitos/metabolismo , Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Animais , Aurora Quinases/metabolismo , Centrômero/efeitos dos fármacos , Centrômero/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinetocoros/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Meiose/efeitos dos fármacos , Camundongos , Modelos Biológicos , Oócitos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo
3.
Methods ; 112: 18-24, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27524557

RESUMO

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.


Assuntos
Aberrações Cromossômicas/efeitos da radiação , Cromossomos Humanos/efeitos da radiação , Citometria por Imagem/métodos , Interpretação de Imagem Assistida por Computador/métodos , Exposição à Radiação/análise , Radiometria/métodos , Antraquinonas/química , Centrômero/efeitos dos fármacos , Centrômero/efeitos da radiação , Centrômero/ultraestrutura , Aberrações Cromossômicas/efeitos dos fármacos , Cromossomos Humanos/efeitos dos fármacos , Cromossomos Humanos/ultraestrutura , Demecolcina/farmacologia , Relação Dose-Resposta à Radiação , Humanos , Citometria por Imagem/instrumentação , Linfócitos/efeitos dos fármacos , Linfócitos/efeitos da radiação , Fito-Hemaglutininas/farmacologia , Coloração e Rotulagem/métodos
4.
Genes Cells ; 21(1): 65-87, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26696398

RESUMO

Autophagy is a conserved intracellular degradation system, which contributes to development and differentiation of various organisms. Yeast cells undergo meiosis under nitrogen-starved conditions and require autophagy for meiosis initiation. However, the precise roles of autophagy in meiosis remain unclear. Here, we show that autophagy is required for efficient meiosis progression and proper meiotic chromosome segregation in fission yeast. Autophagy-defective strains bearing a mutation in the autophagy core factor gene atg1, atg7, or atg14 exhibit deformed nuclear structures during meiosis. These mutant cells require an extracellular nitrogen supply for meiosis progression following their entry into meiosis and show delayed meiosis progression even with a nitrogen supply. In addition, they show frequent chromosome dissociation from the spindle together with spindle overextension, forming extra nuclei. Furthermore, Aurora kinase, which regulates chromosome segregation and spindle elongation, is significantly increased at the centromere and spindle in the mutant cells. Aurora kinase down-regulation eliminated delayed initiation of meiosis I and II, chromosome dissociation, and spindle overextension, indicating that increased Aurora kinase activity may cause these aberrances in the mutant cells. Our findings show a hitherto unrecognized relationship of autophagy with the nuclear structure, regulation of cell cycle progression, and chromosome segregation in meiosis.


Assuntos
Autofagia , Segregação de Cromossomos , Meiose , Schizosaccharomyces/citologia , Schizosaccharomyces/genética , Anáfase/efeitos dos fármacos , Aurora Quinases/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/genética , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Centrômero/efeitos dos fármacos , Centrômero/metabolismo , Segregação de Cromossomos/efeitos dos fármacos , Clonagem Molecular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Genes de Insetos , Espaço Intracelular/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Meiose/efeitos dos fármacos , Meiose/genética , Mutação/genética , Nitrogênio/farmacologia , Fenótipo , Schizosaccharomyces/efeitos dos fármacos , Schizosaccharomyces/enzimologia , Proteínas de Schizosaccharomyces pombe/metabolismo , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Esporos Fúngicos/efeitos dos fármacos , Esporos Fúngicos/fisiologia
5.
Drug Chem Toxicol ; 40(4): 457-462, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27936973

RESUMO

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.


Assuntos
Carcinógenos Ambientais/toxicidade , Centrômero/efeitos dos fármacos , Cromátides/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Óxidos/toxicidade , Compostos de Vanádio/toxicidade , Adulto , Aneugênicos/toxicidade , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Centrômero/metabolismo , Cromátides/metabolismo , Humanos , Leucócitos/citologia , Leucócitos/imunologia , Leucócitos/metabolismo , Masculino , Índice Mitótico , Testes de Mutagenicidade , Concentração Osmolar , Troca de Cromátide Irmã/efeitos dos fármacos , Adulto Jovem
6.
Chromosoma ; 123(1-2): 129-46, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24013524

RESUMO

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.


Assuntos
Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mamíferos/metabolismo , Meiose , Anáfase/efeitos dos fármacos , Animais , Centrômero/efeitos dos fármacos , Cromátides/efeitos dos fármacos , Cromátides/metabolismo , Etoposídeo/farmacologia , Masculino , Meiose/efeitos dos fármacos , Metáfase/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Mitose/efeitos dos fármacos , Coesinas
7.
Cytogenet Genome Res ; 147(1): 17-23, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26606131

RESUMO

The frequency of cells containing micronuclei (MN) and the presence of centromeres in these MN were analyzed in lymphocytes of 98 men from Southern Bohemia. Forty-six of them had worked at the uranium processing plant 'MAPE Mydlovary' which was closed in 1991, and 52 men were controls from the same area. FISH using human pan-centromeric chromosome paint was employed to detect centromere-positive (CEN+) and -negative (CEN-) MN. A total of 1,000 binucleated cells (BNC) per participant were analyzed after cytochalasin B treatment. All BNC with MN (CEN+ or CEN-) were recorded. No differences were found between formerly exposed workers and the control group, neither in the total frequency of cells with MN per 1,000 BNC (mean levels ± SD, 9.1 ± 3.1 and 9.8 ± 2.5, respectively) nor in the percentage of CEN- MN, which were equal (50 ± 18 and 49 ± 17, respectively). Also, there was no difference between individuals living in the 3 villages closest to the uranium processing plant and those living further away. Considering the fact that effective doses of the workers at MAPE Mydlovary were overall similar to those of former uranium miners in whom higher frequencies of CEN- MN have been found more than 10 years after they had finished working underground, these results are somewhat surprising. A more detailed analysis of the exposures indicates that uranium miners received a greater percentage of their effective dose from the inhalation of radon and its daughters, whereas uranium processing workers received it from the incorporation of long-lived radioactive nuclides such as uranium. If, as has been suggested before, the higher level of DNA damage in miners is due to induced genomic instability, then this phenomenon may be related to radon exposure rather than exposure to uranium.


Assuntos
Centrômero/ultraestrutura , Linfócitos/ultraestrutura , Micronúcleos com Defeito Cromossômico/estatística & dados numéricos , Mineração , Exposição Ocupacional , Idoso , Idoso de 80 Anos ou mais , Centrômero/efeitos dos fármacos , Citocalasina B/farmacologia , República Tcheca , Humanos , Hibridização in Situ Fluorescente , Linfócitos/efeitos dos fármacos , Masculino , Testes para Micronúcleos , Pessoa de Meia-Idade , Radiometria , Radônio/toxicidade , Urânio/toxicidade
8.
Prostate ; 74(10): 999-1011, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24802614

RESUMO

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.


Assuntos
Centrômero/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Cinetocoros/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Autoantígenos/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Centrômero/metabolismo , Proteína Centromérica A , Proteínas Cromossômicas não Histona/genética , Ciclo-Oxigenase 2/fisiologia , Regulação para Baixo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Cinetocoros/metabolismo , Masculino , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinases/fisiologia , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Receptores de Prostaglandina E Subtipo EP1/fisiologia
9.
Proc Natl Acad Sci U S A ; 108(9): 3767-72, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21321216

RESUMO

Members of the eukaryotic phylum Apicomplexa are the cause of important human diseases including malaria, toxoplasmosis, and cryptosporidiosis. These obligate intracellular parasites produce new invasive stages through a complex budding process. The budding cycle is remarkably flexible and can produce varied numbers of progeny to adapt to different host-cell niches. How this complex process is coordinated remains poorly understood. Using Toxoplasma gondii as a genetic model, we show that a key element to this coordination is the centrocone, a unique elaboration of the nuclear envelope that houses the mitotic spindle. Exploiting transgenic parasite lines expressing epitope-tagged centromeric H3 variant CenH3, we identify the centromeres of T. gondii chromosomes by hybridization of chromatin immunoprecipitations to genome-wide microarrays (ChIP-chip). We demonstrate that centromere attachment to the centrocone persists throughout the parasite cell cycle and that centromeres localize to a single apical region within the nucleus. Centromere sequestration provides a mechanism for the organization of the Toxoplasma nucleus and the maintenance of genome integrity.


Assuntos
Ciclo Celular , Centrômero/metabolismo , Toxoplasma/citologia , Toxoplasma/metabolismo , Ciclo Celular/efeitos dos fármacos , Centrômero/efeitos dos fármacos , Imunoprecipitação da Cromatina , Quebra Cromossômica/efeitos dos fármacos , Etoposídeo/farmacologia , Imunofluorescência , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação/efeitos dos fármacos , Mitose/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Reprodutibilidade dos Testes , Toxoplasma/efeitos dos fármacos
10.
Mutat Res ; 755(1): 73-80, 2013 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-23726961

RESUMO

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.


Assuntos
Aneugênicos/toxicidade , Medula Óssea/efeitos dos fármacos , Centrômero/efeitos dos fármacos , Sondas de DNA , Hibridização in Situ Fluorescente , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Mutagênicos/toxicidade , Animais , Antibióticos Antineoplásicos/toxicidade , Sequência de Bases , Centrômero/genética , Cromossomos/genética , Colchicina/toxicidade , DNA Satélite/análise , DNA Satélite/genética , Eritrócitos/efeitos dos fármacos , Citometria de Fluxo , Masculino , Testes para Micronúcleos , Mitomicina/toxicidade , Dados de Sequência Molecular , Ratos , Homologia de Sequência do Ácido Nucleico , Moduladores de Tubulina/toxicidade , Vimblastina/toxicidade
11.
J Appl Toxicol ; 33(6): 426-33, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22081495

RESUMO

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.


Assuntos
Amsacrina/toxicidade , Antineoplásicos/toxicidade , Células da Medula Óssea/efeitos dos fármacos , Mutagênicos , Nocodazol/toxicidade , Animais , Antibióticos Antineoplásicos/toxicidade , Centrômero/efeitos dos fármacos , Colchicina/toxicidade , Citogenética , Sondas de DNA , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Eritrócitos/ultraestrutura , Hibridização In Situ , Técnicas In Vitro , Masculino , Camundongos , Testes para Micronúcleos , Mitomicina/toxicidade
12.
Dev Cell ; 12(1): 17-30, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17199038

RESUMO

The centromere is the chromosomal site that joins to microtubules during mitosis for proper segregation. Determining the location of a centromere-specific histone H3 called CENP-A at the centromere is vital for understanding centromere structure and function. Here, we report the identification of three human proteins essential for centromere/kinetochore structure and function, hMis18alpha, hMis18beta, and M18BP1, the complex of which is accumulated specifically at the telophase-G1 centromere. We provide evidence that such centromeric localization of hMis18 is essential for the subsequent recruitment of de novo-synthesized CENP-A. If any of the three is knocked down by RNAi, centromere recruitment of newly synthesized CENP-A is rapidly abolished, followed by defects such as misaligned chromosomes, anaphase missegregation, and interphase micronuclei. Tricostatin A, an inhibitor to histone deacetylase, suppresses the loss of CENP-A recruitment to centromeres in hMis18alpha RNAi cells. Telophase centromere chromatin may be primed or licensed by the hMis18 complex and RbAp46/48 to recruit CENP-A through regulating the acetylation status in the centromere.


Assuntos
Autoantígenos/metabolismo , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular , Centrômero/efeitos dos fármacos , Proteína Centromérica A , Proteínas Cromossômicas não Histona/química , Segregação de Cromossomos/efeitos dos fármacos , Sequência Consenso , Genoma Humano/efeitos dos fármacos , Células HeLa , Humanos , Ácidos Hidroxâmicos/farmacologia , Metáfase/efeitos dos fármacos , Dados de Sequência Molecular , Mutação/genética , Filogenia , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myb/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Telófase/efeitos dos fármacos , Vertebrados
13.
Cell Biol Toxicol ; 28(5): 331-42, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22843076

RESUMO

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.


Assuntos
Aneugênicos/farmacologia , Dissacarídeos/química , Genisteína/análogos & derivados , Genisteína/farmacologia , Glicosídeos/farmacologia , Inibidores da Topoisomerase II/farmacologia , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Centrômero/efeitos dos fármacos , Centrômero/patologia , Centrossomo/efeitos dos fármacos , Centrossomo/patologia , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Imunofluorescência , Genisteína/química , Glicosídeos/química , Histonas/metabolismo , Humanos , Masculino , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Polimerização , Inibidores da Topoisomerase II/química , Tubulina (Proteína)/metabolismo
14.
Cytogenet Genome Res ; 134(2): 151-62, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21555880

RESUMO

An interstitial constriction located on the long arm of rye chromosome 5R (5RL) shows neocentromeric activity at meiosis. In some meiocytes this region is strongly stretched orienting with the true centromere to opposite poles at metaphase I, and keeping sister chromatid cohesion at anaphase I. We found previously that the frequency of neocentric activity varied dramatically in different generations suggesting the effect of environmental factors. Here we studied the behavior of the 5RL neocentromere in mono- and ditelosomic 5RL, and mono-, and disomic 5R wheat-rye addition lines, untreated and treated with an organophosphate pesticide. The treated plants form neocentromeres with an about 4.5-fold increased frequency compared to untreated ones, demonstrating that the pesticide promotes neocentric activity. The neocentromere was activated irrespectively of the pairing configuration or the presence of a complete or truncated 5R centromere. Fluorescence in situ hybridization (FISH) with 2 repetitive sequences (UCM600 and pSc119.2) present at the constriction showed kinetic activity at several locations within this region. Immunostaining with anti-α-tubulin showed that treated plants have abnormal spindles in 46% of the metaphase I cells, indicating that disturbances in spindle formation might promote neocentromere activation.


Assuntos
Centrômero/efeitos dos fármacos , Organofosfatos/farmacologia , Praguicidas/farmacologia , Secale/efeitos dos fármacos , Secale/genética , Metáfase , Monossomia , Secale/citologia , Triticum/genética
15.
Cytogenet Genome Res ; 132(4): 297-303, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21307636

RESUMO

Global climate change, i.e. higher and more variable temperatures, and a gain in soil salinity are increasing plant stress with direct consequences on crop yield and quality levels. Rice productivity is strongly affected by abiotic stress conditions. The regulation of chromatin structure in response to environmental stress is poorly understood. We investigated the interphase chromatin organization from rice plants in non-stress versus stress conditions. We have used a cytogenetic approach, based on fluorescence in situ hybridization (FISH) with 45S, 5S rDNA and centromeric probes on rice tissue sections. The abiotic stress conditions included cold, heat and mild salinity and were applied during seed germination. In contrast to cold, saline and heat stresses caused extensive decondensation of 45S rDNA chromatin and also an increase in the distance between the 2 homologous 5S rDNA loci. 5-Azacytidine (5-AC), a DNA hypomethylating drug, greatly increased 45S rDNA chromatin decondensation and interestingly was able to induce polarization of centromeres in rice interphase nuclei. The abiotic stresses tested did not perturb the spatial position of centromeres, typically with circular arrangement around the nucleolus. The results suggest a role for chromatin plasticity in a world of climate changes.


Assuntos
Metilação de DNA , Interfase/genética , Oryza/genética , Estresse Fisiológico/genética , Azacitidina/farmacologia , Centrômero/efeitos dos fármacos , Centrômero/genética , Cromatina/efeitos dos fármacos , Cromatina/metabolismo , Cromossomos de Plantas/efeitos dos fármacos , Cromossomos de Plantas/genética , Cromossomos de Plantas/metabolismo , Temperatura Baixa , DNA Ribossômico/genética , Temperatura Alta , Hibridização in Situ Fluorescente , Interfase/efeitos dos fármacos , Oryza/efeitos dos fármacos , Salinidade , Estresse Fisiológico/efeitos dos fármacos
16.
Mutagenesis ; 26(6): 771-81, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21775299

RESUMO

There is some evidence that the mouse lymphoma TK assay (MLA) can detect aneugens, and this is accepted in the current International Conference on Harmonisation guidance for testing pharmaceuticals. However, whether or not it can be used as a reliable screen for aneugenicity has been the subject of debate. Consequently, aneugens with diverse mechanisms of action were tested in the MLA using 24-h exposure. No evidence of increased mutant frequency was seen with noscapine, diazepam or colchicine and increases were seen with taxol, carbendazim, econazole and chloral hydrate only at high levels of toxicity (for all but one taxol concentration survival reduced to ≤10% of control). None of these agents would be unequivocally classified as positive using currently accepted criteria. The largest increases in mutant number were seen with taxol and carbendazim; therefore, trifluorothymidine (TFT)-resistant clones resulting from treatment with them were cultured and analysed for chromosome 11 copy number using fluorescent in situ hybridisation (FISH) and loss of heterozygosity (LOH). High concentrations of these aneugens induced LOH at all loci examined indicating only one chromosome 11 was present but, perhaps surprisingly, all were found to have two copies of chromosome 11 using FISH. This would be consistent with loss of the tk(+) chromosome 11b with concomitant duplication of chromosome 11a, which has been proposed as a likely mechanism for induction of TFT-resistant clones. However, it was also surprising that analysis of centromere size showed that almost all the clones had both small and large centromeres, i.e. suggesting the presence of both chromosomes 11a and 11b. In conclusion, it appears that the TFT-resistant mutants resulting from treatment with toxic concentrations of some aneugens such as taxol and carbendazim have undergone complex genetic changes. However, these data show that the MLA cannot be used as a routine screen to detect aneugens.


Assuntos
Aneugênicos/análise , Ensaios Enzimáticos/métodos , Linfoma/metabolismo , Timidina Quinase/metabolismo , Aneugênicos/toxicidade , Animais , Linhagem Celular Tumoral , Centrômero/efeitos dos fármacos , Centrômero/metabolismo , Cromossomos de Mamíferos/genética , Dosagem de Genes/efeitos dos fármacos , Dosagem de Genes/genética , Hibridização in Situ Fluorescente , Cariotipagem , Perda de Heterozigosidade/efeitos dos fármacos , Perda de Heterozigosidade/genética , Camundongos , Repetições de Microssatélites/genética , Reação em Cadeia da Polimerase
17.
J Exp Ther Oncol ; 9(3): 221-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22070054

RESUMO

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.


Assuntos
Aberrações Cromossômicas/efeitos dos fármacos , Frutas/química , Persea/química , Extratos Vegetais/farmacologia , Células Cultivadas , Centrômero/efeitos dos fármacos , Centrômero/genética , Quimioprevenção , Aberrações Cromossômicas/induzido quimicamente , Ciclofosfamida/toxicidade , Humanos , Linfócitos , Metáfase/genética , Extratos Vegetais/toxicidade
18.
Mol Hum Reprod ; 16(9): 665-84, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20406800

RESUMO

Mitotic centromere-associated kinesin (MCAK) is an ATP-dependent microtubule (MT) depolymerase regulated by Aurora kinase (AURK) phosphorylation and implicated in resolution of improper MT attachments in mitosis. Distribution of MCAK was studied in oocyte maturation by anti-MCAK antibody, anti-tubulin antibody, anti-AURKB antibody and anti-centromere antibody (ACA) and by the expression of MCAK-enhanced green fluorescent protein fusion protein in maturing mouse oocytes. Function was assessed by knockdown of MCAK and Mad2, by inhibiting AURK or the proteasome, by live imaging with polarization microscope and by chromosomal analysis. The results show that MCAK is transiently recruited to the nucleus and transits to spindle poles, ACA-positive domains and chiasmata at prometaphase I. At metaphase I and II, it is present at centrosomes and centromeres next to AURKB and checkpoint proteins Mad2 and BubR1. It is retained at centromeres at telophase I and also at the midbody. Knockdown of MCAK causes a delay in chromosome congression but does not prevent bipolar spindle assembly. MCAK knockdown also induces a meiosis I arrest, which is overcome by knockdown of Mad2 resulting in chiasma resolution, chromosome separation, formation of aberrant meiosis II spindles and increased hypoploidy. In conclusion, MCAK appears to possess a unique distribution and function in oocyte maturation. It is required for meiotic progression from meiosis I to meiosis II associated with silencing of the spindle assembly checkpoint. Alterations in abundance and activity of MCAK, as implicated in aged oocytes, may therefore contribute to the loss of control of cell cycle and chromosome behaviour, thus increasing risk for errors in chromosome segregation and aneuploidy.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centrômero/enzimologia , Cinesinas/metabolismo , Meiose , Mitose , Oócitos/enzimologia , Fuso Acromático/enzimologia , Animais , Aurora Quinase B , Aurora Quinases , Proteínas de Ciclo Celular/genética , Nucléolo Celular/enzimologia , Células Cultivadas , Centrômero/efeitos dos fármacos , Segregação de Cromossomos , Inibidores de Cisteína Proteinase/farmacologia , Feminino , Cinesinas/genética , Proteínas Mad2 , Camundongos , Microinjeções , Oócitos/efeitos dos fármacos , Fosforilação , Ploidias , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Interferência de RNA , Proteínas Recombinantes de Fusão/metabolismo , Fuso Acromático/efeitos dos fármacos , Fatores de Tempo
19.
Mol Cell Biol ; 27(3): 949-62, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17101789

RESUMO

Cellular information is encoded genetically in the DNA nucleotide sequence and epigenetically by the "histone code," DNA methylation, and higher-order packaging of DNA into chromatin. Cells possess intricate mechanisms to sense and repair damage to DNA and the genetic code. However, nothing is known of the mechanisms, if any, that repair and/or compensate for damage to epigenetically encoded information, predicted to result from perturbation of DNA and histone modifications or other changes in chromatin structure. Here we show that primary human cells respond to a variety of small molecules that perturb DNA and histone modifications by recruiting HP1 proteins to sites of altered pericentromeric heterochromatin. This response is essential to maintain the HP1-binding kinetochore protein hMis12 at kinetochores and to suppress catastrophic mitotic defects. Recruitment of HP1 proteins to pericentromeres depends on histone H3.3 variant deposition, mediated by the HIRA histone chaperone. These data indicate that defects in pericentromeric epigenetic heterochromatin modifications initiate a dynamic HP1-dependent response that rescues pericentromeric heterochromatin function and is essential for viable progression through mitosis.


Assuntos
Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Heterocromatina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Células Cultivadas , Centrômero/efeitos dos fármacos , Centrômero/metabolismo , Homólogo 5 da Proteína Cromobox , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Heterocromatina/efeitos dos fármacos , Chaperonas de Histonas , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Chaperonas Moleculares/metabolismo , Transporte Proteico/efeitos dos fármacos , Fatores de Transcrição/metabolismo
20.
Acta Biochim Biophys Sin (Shanghai) ; 42(10): 677-87, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20817931

RESUMO

Previous studies have shown that histone deacetylase inhibitors (HDACis) can kill cancer cells. In addition, HDACis can induce mitotic catastrophe in cancer cells due to insufficient localization of chromosomal passenger complex (CPC) to the centromere. However, the mechanisms behind these phenomena remain unclear. In this study, we found that a HDACi, FK228, affected multiple epigenetic modification characteristics of the centromere, including enhanced acetylation of histone H3 lysine 9 (H3K9), decreased trimethylation of H3K9, and decreased phosphorylation of histone H3 serine 10 (H3S10) and centromere protein A (CENP-A). These epigenetic changes implied that H3K9 hyperacetylation inhibits the CPC recruitment, induces impaired centromere assembly and function, and eventually leads to aberrant mitosis. These data suggested that hypoacetylation of histone in the pericentromere is the most important landmark for recruiting CPC and leading to the mitotic catastrophe in HDACi-induced killing of cancer cells.


Assuntos
Depsipeptídeos/farmacologia , Histonas/metabolismo , Lisina/metabolismo , Fuso Acromático/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Antibióticos Antineoplásicos/farmacologia , Autoantígenos/metabolismo , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Centrômero/efeitos dos fármacos , Centrômero/metabolismo , Proteína Centromérica A , Proteínas Cromossômicas não Histona/metabolismo , Humanos , Metilação/efeitos dos fármacos , Microscopia de Fluorescência , Mitose/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Serina/metabolismo , Fuso Acromático/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA