Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Life Sci ; 277: 119556, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-33945829

RESUMEN

OBJECTIVE: Poly(ADP-ribose) polymerase1 (PARP1) interacts and poly(ADP-ribosyl)ates telomere repeat binding factor 2 (TRF2), which acts as a platform to recruit a large number of proteins at the telomere. Since the discovery of TRF2-SLX4 interaction, SLX4 is becoming the key player in telomere length (TL) maintenance and repair by telomere sister chromatid exchange (T-SCE). Defective TL maintenance pathway results in a spectrum of diseases called telomeropathies like dyskeratosis congenita, aplastic anemia, fanconi anemia, cancer. We aimed to study the role of SLX4 and PARP1 on each other's telomere localization, T-SCE, and TL maintenance in human telomerase-negative osteosarcoma U2OS cells to understand some of the molecular mechanisms of telomere homeostasis. MATERIALS AND METHODS: We checked the role of SLX4 and PARP1 on each other's telomere localization by telomere immunofluorescence. We have cloned full-length wild-type and catalytically inactive mutant PARP1 to understand the role of poly(ADP-ribosyl)ation reaction by PARP1 in telomere length homeostasis. TL of U2OS cells was measured by Q-FISH. T-SCE was measured by Telomere-FISH. KEY FINDINGS: We observed that SLX4 has no role in the telomere localization of PARP1. However, reduced localization of SLX4 at undamaged and damaged telomere upon PARP1 depletion was reversed by overexpression of exogenous wild-type PARP1 but not by overexpression of catalytically inactive mutant PARP1. PARP1 depletion synergized SLX4 depletion-mediated reduction of T-SCE. Furthermore, SLX4 depletion elongated TL, and combined insufficiency of SLX4 with PARP1 further elongated TL. CONCLUSION: So, PARP1 controls SLX4 recruitment at telomere by poly(ADP-ribosyl)ation reaction, thereby regulating SLX4-mediated T-SCE and TL homeostasis.


Asunto(s)
Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Recombinasas/metabolismo , Intercambio de Cromátides Hermanas/fisiología , Línea Celular Tumoral , Cromátides/metabolismo , Cromátides/fisiología , Reparación del ADN , Homeostasis , Humanos , Poli(ADP-Ribosa) Polimerasa-1/fisiología , Poli(ADP-Ribosa) Polimerasas/genética , Recombinasas/genética , Recombinasas/fisiología , Telomerasa/metabolismo , Telómero/fisiología , Homeostasis del Telómero/fisiología , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
2.
STAR Protoc ; 1(3): 100202, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33377096

RESUMEN

Sister chromatid interactions are a key step to ensure the successful segregation of sister chromatids after replication. Our knowledge about this phenomenon is mostly based on microscopy approaches, which have some constraints such as resolution limit and the impossibility of studying several genomic positions at the same time. Here, we present a protocol for Hi-SC2, a high-throughput sequencing-based method, to monitor sister chromatid contacts after replication at high resolution throughout the genome, which we applied to study cohesion in Vibrio cholerae. For complete details on the use and execution of this protocol, please refer to Espinosa et al. (2020).


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Intercambio de Cromátides Hermanas/fisiología , Animales , Cromátides/metabolismo , Segregación Cromosómica , Biología Computacional/métodos , Replicación del ADN , Humanos , Mitosis , Intercambio de Cromátides Hermanas/genética , Vibrio cholerae/genética
3.
Life Sci Alliance ; 3(12)2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33106324

RESUMEN

Chromosome fusion is a frequent intermediate in oncogenic chromosome rearrangements and has been proposed to cause multiple tumor-driving abnormalities. In conventional experimental systems, however, these abnormalities were often induced by randomly induced chromosome fusions involving multiple different chromosomes. It was therefore not well understood whether a single defined type of chromosome fusion, which is reminiscent of a sporadic fusion in tumor cells, has the potential to cause chromosome instabilities. Here, we developed a human cell-based sister chromatid fusion visualization system (FuVis), in which a single defined sister chromatid fusion is induced by CRISPR/Cas9 concomitantly with mCitrine expression. The fused chromosome subsequently developed extra-acentric chromosomes, including chromosome scattering, indicative of chromothripsis. Live-cell imaging and statistical modeling indicated that sister chromatid fusion generated micronuclei (MN) in the first few cell cycles and that cells with MN tend to display cell cycle abnormalities. The powerful FuVis system thus demonstrates that even a single sporadic sister chromatid fusion can induce chromosome instability and destabilize the cell cycle through MN formation.


Asunto(s)
Inestabilidad Cromosómica/genética , Análisis de la Célula Individual/métodos , Intercambio de Cromátides Hermanas/fisiología , Sistemas CRISPR-Cas/genética , Ciclo Celular/genética , División Celular/genética , Cromátides/genética , Cromátides/patología , Cromátides/fisiología , Inestabilidad Cromosómica/fisiología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Ingeniería Genética/métodos , Células HCT116 , Humanos , Microscopía Fluorescente/métodos , Neoplasias/genética , Intercambio de Cromátides Hermanas/genética
4.
Fertil Steril ; 112(2): 336-342.e3, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31103288

RESUMEN

OBJECTIVE: To determine factors affecting unbalanced chromosomal rearrangement originating from parental inversion and interchromosomal effect occurrence in blastocysts from inversion carriers. DESIGN: Retrospective study. SETTING: University-affiliated center. PATIENT(S): Couples with one partner carrying inversion underwent preimplantation genetic testing for chromosomal structural rearrangement cycles. INTERVENTION(S): Not applicable. MAIN OUTCOME MEASURE(S): Unbalanced rearrangement embryo rate, normal embryo rate, interchromosomal effect. RESULT(S): Preimplantation genetic testing was performed for 576 blastocysts from 57 paracentric (PAI) and 94 pericentric (PEI) inversion carriers. The percentage of normal/balanced blastocysts was significantly higher in PAI than PEI carriers (70.4% vs. 57.5%). Logistic regression indicated the inverted segment size ratio was a statistically significant risk factor for abnormality from parental inversion in both PEI and PAI. The optimal cutoff values to predict unbalanced rearrangement risk were 35.7% and 57%. In PAI, rates of abnormality from parental inversion were 0% and 12.1% in the <35.7% and ≥35.7% groups, respectively, with no gender difference. For PEI, the rates of abnormality from parental inversion were 7.9% and 33.1% in the <57% and ≥57% groups, respectively. In the ≥57% group, the rate of unbalanced rearrangement was significantly higher from paternal than maternal inversion (43.3% vs. 23.6%). In inversion carriers, 21,208 chromosomes were examined, and 187 (0.88%) malsegregations were identified from structurally normal chromosomes. In controls, 56,488 chromosomes were assessed, and 497 (0.88%) aneuploidies were identified, indicating no significant difference. CONCLUSION(S): The risk of unbalanced rearrangement is affected by the ratio of inverted segment size in both PAI and PEI carriers and is associated with gender.


Asunto(s)
Inversión Cromosómica , Segregación Cromosómica/genética , Pruebas Genéticas , Meiosis/genética , Diagnóstico Preimplantación , Adulto , Blastocisto/citología , Blastocisto/metabolismo , Estudios de Casos y Controles , Inversión Cromosómica/embriología , Inversión Cromosómica/genética , Inversión Cromosómica/estadística & datos numéricos , Cruzamientos Genéticos , Femenino , Pruebas Genéticas/métodos , Pruebas Genéticas/estadística & datos numéricos , Humanos , Masculino , Diagnóstico Preimplantación/estadística & datos numéricos , Estudios Retrospectivos , Intercambio de Cromátides Hermanas/genética , Intercambio de Cromátides Hermanas/fisiología , Adulto Joven
5.
J Reprod Dev ; 65(1): 57-66, 2019 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-30464155

RESUMEN

Mammalian oocyte quality degrades over time after ovulation in vitro, which can cause fatal defects such as chromosomal aneuploidy. As various oocyte manipulations employed in assisted reproductive technology are time consuming, post-ovulatory aging is a serious problem to overcome in reproductive medicine or ova research. In this study, we investigated the effects of postovulatory aging on the incidence of chromosome aneuploidy during meiosis II, with a focus on the expression of functional proteins from the spindle assembly checkpoint (SAC). Chromosome analysis was used to assess the rate of aneuploidy in in vitro aged oocytes, or in early embryos derived from aged oocytes. Immunofluorescent staining was used to detect the localization of MAD2, which is a SAC signal that monitors the correct segregation of sister chromatids. Immunoblotting was used to quantify cohesin subunits, which are adhesion factors connecting sister chromatids at the metaphase II (MII) centromere. It was shown that post-ovulatory oocyte aging inhibits MAD2 localization to the sister kinetochore. Furthermore, oocyte aging prevented cohesin subunits from being maintained or degraded at the appropriate time. These data suggest that the destabilization of SAC signaling causes sister chromatid segregation errors in MII oocytes, and consequently increases the incidence of aneuploidy in early embryos. Our findings have provided distinct evidence that the post-ovulatory aging of oocytes might also be a risk factor for aneuploidy, irrespective of maternal age.


Asunto(s)
Aneuploidia , Senescencia Celular/fisiología , Meiosis/fisiología , Oocitos/fisiología , Ovulación/fisiología , Huso Acromático/fisiología , Animales , Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Embrión de Mamíferos/química , Femenino , Fertilización In Vitro , Técnica del Anticuerpo Fluorescente , Técnicas de Maduración In Vitro de los Oocitos , Cinetocoros/química , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Proteínas Mad2/análisis , Masculino , Ratones , Ratones Endogámicos ICR , Oocitos/química , Factores de Riesgo , Intercambio de Cromátides Hermanas/fisiología , Cohesinas
6.
Curr Biol ; 28(17): 2837-2844.e3, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30122528

RESUMEN

The fidelity of mitosis depends on cohesive forces that keep sister chromatids together. This is mediated by cohesin that embraces sister chromatid fibers from the time of their replication until the subsequent mitosis [1-3]. Cleavage of cohesin marks anaphase onset, where single chromatids are dragged to the poles by the mitotic spindle [4-6]. Cohesin cleavage should only occur when all chromosomes are properly bio-oriented to ensure equal genome distribution and prevent random chromosome segregation. Unscheduled loss of sister chromatid cohesion is prevented by a safeguard mechanism known as the spindle assembly checkpoint (SAC) [7, 8]. To identify specific conditions capable of restoring defects associated with cohesion loss, we screened for genes whose depletion modulates Drosophila wing development when sister chromatid cohesion is impaired. Cohesion deficiency was induced by knockdown of the acetyltransferase separation anxiety (San)/Naa50, a cohesin complex stabilizer [9-12]. Several genes whose function impacts wing development upon cohesion loss were identified. Surprisingly, knockdown of key SAC proteins, Mad2 and Mps1, suppressed developmental defects associated with San depletion. SAC impairment upon cohesin removal, triggered by San depletion or artificial removal of the cohesin complex, prevented extensive genome shuffling, reduced segregation defects, and restored cell survival. This counterintuitive phenotypic suppression was caused by an intrinsic bias for efficient chromosome biorientation at mitotic entry, coupled with slow engagement of error-correction reactions. Thus, in contrast to SAC's role as a safeguard mechanism for mitotic fidelity, removal of this checkpoint alleviates mitotic errors when sister chromatid cohesion is compromised.


Asunto(s)
Drosophila melanogaster/citología , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Mitosis/fisiología , Intercambio de Cromátides Hermanas/fisiología , Animales
7.
Nat Commun ; 8: 14618, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28262707

RESUMEN

Aberrant DNA replication is a major source of the mutations and chromosomal rearrangements associated with pathological disorders. In bacteria, several different DNA lesions are repaired by homologous recombination, a process that involves sister chromatid pairing. Previous work in Escherichia coli has demonstrated that sister chromatid interactions (SCIs) mediated by topological links termed precatenanes, are controlled by topoisomerase IV. In the present work, we demonstrate that during the repair of mitomycin C-induced lesions, topological links are rapidly substituted by an SOS-induced sister chromatid cohesion process involving the RecN protein. The loss of SCIs and viability defects observed in the absence of RecN were compensated by alterations in topoisomerase IV, suggesting that the main role of RecN during DNA repair is to promote contacts between sister chromatids. RecN also modulates whole chromosome organization and RecA dynamics suggesting that SCIs significantly contribute to the repair of DNA double-strand breaks (DSBs).


Asunto(s)
Cromátides/metabolismo , Daño del ADN/fisiología , ADN Bacteriano/metabolismo , Escherichia coli/fisiología , Intercambio de Cromátides Hermanas/fisiología , Proteínas Bacterianas/fisiología , Segregación Cromosómica/fisiología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Replicación del ADN/fisiología , Enzimas de Restricción del ADN/fisiología , Topoisomerasa de ADN IV/fisiología , ADN Bacteriano/efectos de los fármacos , Mitomicina/farmacología , Rec A Recombinasas/fisiología , Respuesta SOS en Genética/efectos de los fármacos , Respuesta SOS en Genética/fisiología
8.
Nat Commun ; 6: 8399, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26423134

RESUMEN

Warsaw breakage syndrome (WABS) is caused by defective DDX11, a DNA helicase that is essential for chromatid cohesion. Here, a paired genome-wide siRNA screen in patient-derived cell lines reveals that WABS cells do not tolerate partial depletion of individual APC/C subunits or the spindle checkpoint inhibitor p31(comet). A combination of reduced cohesion and impaired APC/C function also leads to fatal mitotic arrest in diploid RPE1 cells. Moreover, WABS cell lines, and several cancer cell lines with cohesion defects, display a highly increased response to a new cell-permeable APC/C inhibitor, apcin, but not to the spindle poison paclitaxel. Synthetic lethality of APC/C inhibition and cohesion defects strictly depends on a functional mitotic spindle checkpoint as well as on intact microtubule pulling forces. This indicates that the underlying mechanism involves cohesion fatigue in response to mitotic delay, leading to spindle checkpoint re-activation and lethal mitotic arrest. Our results point to APC/C inhibitors as promising therapeutic agents targeting cohesion-defective cancers.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/fisiología , Intercambio de Cromátides Hermanas/fisiología , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Segregación Cromosómica , Humanos , Mitosis/fisiología , Morfolinas/farmacología , Nocodazol/farmacología , Paclitaxel/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Intercambio de Cromátides Hermanas/efectos de los fármacos , Moduladores de Tubulina/farmacología
9.
Cold Spring Harb Perspect Biol ; 7(3): a015776, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25663668

RESUMEN

In this article, we will discuss the biochemistry of mitosis in eukaryotic cells. We will focus on conserved principles that, importantly, are adapted to the biology of the organism. It is vital to bear in mind that the structural requirements for division in a rapidly dividing syncytial Drosophila embryo, for example, are markedly different from those in a unicellular yeast cell. Nevertheless, division in both systems is driven by conserved modules of antagonistic protein kinases and phosphatases, underpinned by ubiquitin-mediated proteolysis, which create molecular switches to drive each stage of division forward. These conserved control modules combine with the self-organizing properties of the subcellular architecture to meet the specific needs of the cell. Our discussion will draw on discoveries in several model systems that have been important in the long history of research on mitosis, and we will try to point out those principles that appear to apply to all cells, compared with those in which the biochemistry has been specifically adapted in a particular organism.


Asunto(s)
Fenómenos Bioquímicos/fisiología , Quinasas Ciclina-Dependientes/metabolismo , Mitosis/fisiología , Fosfoproteínas Fosfatasas/metabolismo , Transducción de Señal/fisiología , Intercambio de Cromátides Hermanas/fisiología , Animales , Levaduras
10.
Cell Cycle ; 13(15): 2370-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25483188

RESUMEN

Sister chromatid separation creates a sudden loss of tension on kinetochores, which could, in principle, re-activate the spindle checkpoint in anaphase. This so-called "anaphase problem" is probably avoided by timely inactivation of cyclin B1-Cdk1, which may prevent the spindle tension sensing Aurora B kinase from destabilizing kinetochore-microtubule interactions as they lose tension in anaphase. However, exactly how spindle checkpoint re-activation is prevented remains unclear. Here, we investigated how different degrees of cyclin B1 stabilization affected the spindle checkpoint in metaphase and anaphase. Cells expressing a strongly stabilized (R42A) mutant of cyclin B1 degraded APC/C(Cdc20) substrates normally, showing that checkpoint release was not inhibited by high cyclin B1-Cdk1 activity. However, after this initial wave of APC/C(Cdc20) activity, the spindle checkpoint returned in cells with uncohesed sister chromatids. Expression of a lysine mutant of cyclin B1 that is degraded only slightly inefficiently allowed a normal metaphase-to-anaphase transition. Strikingly, however, the spindle checkpoint returned in cells that had not degraded the cyclin B1 mutant 10-15 min after anaphase onset. When cyclin B1 remained in late anaphase, cytokinesis stalled, and translocation of INCENP from separated sister chromatids to the spindle midzone was blocked. This late anaphase arrest required the activity of Aurora B and Mps1. In conclusion, our results reveal that complete removal of cyclin B1 is essential to prevent the return of the spindle checkpoint following sister chromatid disjunction. Speculatively, increasing activity of APC/C(Cdc20) in late anaphase helps to keep cyclin B1 levels low.


Asunto(s)
Ciclina B1/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Proteolisis , Intercambio de Cromátides Hermanas/fisiología , Aurora Quinasa B/metabolismo , Proteína Quinasa CDC2 , Proteínas Cdc20/metabolismo , Línea Celular Tumoral , Ciclina B1/genética , Quinasas Ciclina-Dependientes/metabolismo , Humanos , Lisina/metabolismo , Proteína 1 de Superficie de Merozoito/metabolismo , Mutación
11.
Cell Cycle ; 13(23): 3623-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25457919

RESUMEN

Meiosis is a crucial process of sexual reproduction by forming haploid gametes from diploid precursor cells. It involves 2 subsequent divisions (meiosis I and meiosis II) after one initial round of DNA replication. Homologous monocentric chromosomes are separated during the first and sister chromatids during the second meiotic division. The faithful segregation of monocentric chromosomes is realized by mono-orientation of fused sister kinetochores at metaphase I and by bi-orientation of sister kinetochores at metaphase II. Conventionally this depends on a 2-step loss of cohesion, along chromosome arms during meiosis I and at sister centromeres during meiosis II.


Asunto(s)
Meiosis/fisiología , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Intercambio de Cromátides Hermanas/fisiología
12.
Environ Monit Assess ; 186(4): 2195-204, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24292950

RESUMEN

The search for reliable biomarkers of human exposure to benzene and its derivatives is still subject of research. Many of the proposed biomarkers have limitations ranging from the low sensitivity to the wide variability of results. Thus, the aim of our study was to assess the frequencies of chromosomal abnormalities (CA) and sister chromatid exchanges (SCE) in workers of gas stations, with (cases, n = 19) and without (local controls, n = 6) risk of exposure to benzene and its derivatives, comparing them with the results from the general population (external controls, n = 38). The blood dosages of benzene, toluene, and xylenes were measured in all participants. Blood solvent levels were compared with the findings obtained in cytogenetic evaluation and a research protocol which included data of the workplace, lifestyle, and health of the individuals. We did not detect the presence of benzene and its derivatives and did not find chromosomal damage that may be associated with the gas station activity in cases. Moreover, although we found an association of increased SCE and the working time in the local controls, the values found for SCE are within normal limits. Thus, our evaluation of SCE and CA reflected the levels of benzene and its derivatives observed in the blood. We believe, therefore, that SCE and CA may actually constitute possible tests for the evaluation of these exposures. However, we believe that further studies, including individuals at risk, are important to confirm this assertion.


Asunto(s)
Contaminantes Ocupacionales del Aire/toxicidad , Benceno/toxicidad , Exposición Profesional/análisis , Adulto , Contaminantes Ocupacionales del Aire/análisis , Contaminantes Ocupacionales del Aire/metabolismo , Benceno/análisis , Benceno/metabolismo , Biomarcadores/metabolismo , Aberraciones Cromosómicas/inducido químicamente , Femenino , Gasolina/toxicidad , Humanos , Masculino , Exposición Profesional/estadística & datos numéricos , Intercambio de Cromátides Hermanas/fisiología , Adulto Joven
13.
Plant J ; 73(1): 154-65, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22974522

RESUMEN

Replication factor C1 (RFC1), which is conserved in eukaryotes, is involved in DNA replication and checkpoint control. However, a RFC1 product participating in DNA repair at meiosis has not been reported in Arabidopsis. Here, we report functional characterization of AtRFC1 through analysis of the rfc1-2 mutant. The rfc1-2 mutant displayed normal vegetative growth but showed silique sterility because the male gametophyte was arrested at the uninucleus microspore stage and the female at the functional megaspore stage. Expression of AtRFC1 was concentrated in the reproductive organ primordia, meiocytes and developing gametes. Chromosome spreads showed that pairing and synapsis were normal, and the chromosomes were broken when desynapsis began at late prophase I, and chromosome fragments remained in the subsequent stages. For this reason, homologous chromosomes and sister chromatids segregated unequally, leading to pollen sterility. Immunolocalization revealed that the AtRFC1 protein localized to the chromosomes during zygotene and pachytene in wild-type but were absent in the spo11-1 mutant. The chromosome fragmentation of rfc1-2 was suppressed by spo11-1, indicating that AtRFC1 acted downstream of AtSPO11-1. The similar chromosome behavior of rad51 rfc1-2 and rad51 suggests that AtRFC1 may act with AtRAD51 in the same pathway. In summary, AtRFC1 is required for DNA double-strand break repair during meiotic homologous recombination of Arabidopsis.


Asunto(s)
Proteínas de Arabidopsis/fisiología , Arabidopsis/genética , Reparación del ADN/genética , Meiosis/genética , Reparación del ADN por Recombinación/genética , Proteína de Replicación C/fisiología , Arabidopsis/fisiología , Cromosomas de las Plantas/genética , Cromosomas de las Plantas/fisiología , Reparación del ADN/fisiología , Meiosis/fisiología , Óvulo Vegetal/fisiología , Polen/fisiología , Reparación del ADN por Recombinación/fisiología , Intercambio de Cromátides Hermanas/fisiología
14.
Dev Cell ; 23(1): 112-23, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22814604

RESUMEN

Complete dissociation of sister chromatid cohesion and subsequent induction of poleward movement of disjoined sisters are two essential events underlying chromosome segregation; however, how cells coordinate these two processes is not well understood. Here, we developed a fluorescence-based sensor for the protease separase that mediates cohesin cleavage. We found that separase undergoes an abrupt activation shortly before anaphase onset in the vicinity of chromosomes. This activation profile of separase depends on the abilities of two of its binding proteins, securin and cyclin B1, to inhibit its protease activity and target it to chromosomes. Subsequent to its proteolytic activation, separase then binds to and inhibits a subset of cyclin B1-cdk1, which antagonizes cdk1-mediated phosphorylation on chromosomes and facilitates poleward movement of sisters in anaphase. Therefore, by consecutively acting as a protease and a cdk1 inhibitor, separase coordinates two key processes to achieve simultaneous and abrupt separation of sister chromatids.


Asunto(s)
Proteína Quinasa CDC2/antagonistas & inhibidores , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Endopeptidasas/fisiología , Proteínas Nucleares/genética , Fosfoproteínas/genética , Segregación Cromosómica/fisiología , Ciclina B1/metabolismo , Proteínas de Unión al ADN , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Microscopía Fluorescente/métodos , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Securina , Separasa , Intercambio de Cromátides Hermanas/fisiología , Cohesinas
15.
Mol Cell ; 47(3): 396-409, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22704558

RESUMEN

Completion of DNA replication after replication stress depends on PCNA, which undergoes monoubiquitination to stimulate direct bypass of DNA lesions by specialized DNA polymerases or is polyubiquitinated to promote recombination-dependent DNA synthesis across DNA lesions by template switching mechanisms. Here we report that the ZRANB3 translocase, a SNF2 family member related to the SIOD disorder SMARCAL1 protein, is recruited by polyubiquitinated PCNA to promote fork restart following replication arrest. ZRANB3 depletion in mammalian cells results in an increased frequency of sister chromatid exchange and DNA damage sensitivity after treatment with agents that cause replication stress. Using in vitro biochemical assays, we show that recombinant ZRANB3 remodels DNA structures mimicking stalled replication forks and disassembles recombination intermediates. We therefore propose that ZRANB3 maintains genomic stability at stalled or collapsed replication forks by facilitating fork restart and limiting inappropriate recombination that could occur during template switching events.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN/fisiología , Inestabilidad Genómica/fisiología , Poliubiquitina/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estrés Fisiológico/genética , Secuencia de Aminoácidos , Línea Celular Tumoral , Daño del ADN/fisiología , ADN Helicasas/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Datos de Secuencia Molecular , Osteosarcoma , Unión Proteica/fisiología , Recombinación Genética/fisiología , Intercambio de Cromátides Hermanas/fisiología , Ubiquitinación/fisiología
16.
Exp Cell Res ; 318(12): 1361-6, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22564894

RESUMEN

During mitosis, kinetochores couple chromosomes to the dynamic tips of spindle microtubules. These attachments convert chemical energy stored in the microtubule lattice into mechanical energy, generating force to move chromosomes. In addition to mediating robust microtubule attachments, kinetochores also integrate and respond to regulatory signals that ensure the accuracy of chromosome segregation during each cell division. Signals for corrective detachment act specifically on kinetochore-microtubule attachments that fail to generate normal levels of tension, although it is unclear how tension is sensed and how the attachments are released. In this review, we discuss the mechanisms by which kinetochore-microtubule attachments generate force during chromosome biorientation, and the pathways of maturation and regulation that lead to the formation of correct attachments.


Asunto(s)
Cinetocoros/metabolismo , Mitosis/genética , Intercambio de Cromátides Hermanas/fisiología , Animales , Segregación Cromosómica/genética , Segregación Cromosómica/fisiología , Humanos , Cinetocoros/fisiología , Microtúbulos/genética , Microtúbulos/metabolismo , Microtúbulos/fisiología , Mitosis/fisiología , Modelos Biológicos , Multimerización de Proteína/genética , Multimerización de Proteína/fisiología , Intercambio de Cromátides Hermanas/genética
17.
Exp Cell Res ; 318(12): 1386-93, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22472347

RESUMEN

Cells use ring-like structured protein complexes for various tasks in DNA dynamics. The tripartite cohesin ring is particularly suited to determine chromosome architecture, for it is large and dynamic, may acquire different forms, and is involved in several distinct nuclear processes. This review focuses on cohesin's role in structuring chromosomes during mitotic and meiotic cell divisions and during interphase.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Cromosomas/química , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/genética , Cromosomas/metabolismo , Cromosomas/ultraestructura , Humanos , Interfase/genética , Interfase/fisiología , Meiosis/genética , Meiosis/fisiología , Mitosis/genética , Mitosis/fisiología , Modelos Biológicos , Conformación de Ácido Nucleico , Intercambio de Cromátides Hermanas/genética , Intercambio de Cromátides Hermanas/fisiología , Cohesinas
18.
J Biol Chem ; 287(7): 5091-101, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22187435

RESUMEN

Inactivating mutations in the breast cancer susceptibility gene BRCA2 cause gross chromosomal rearrangements. Chromosome structural instability in the absence of BRCA2 is thought to result from defective homology-directed DNA repair. Here, we show that BRCA2 links the fidelity of telomere maintenance with genetic integrity. Absence of BRCA2 resulted in signs of dysfunctional telomeres, such as telomere shortening, erosions, and end fusions in proliferating mouse fibroblasts. BRCA2 localized to the telomeres in S phase in an ATR-dependent manner, and its absence resulted in the accumulation of common fragile sites, particularly at the G-rich lagging strand, and increased the telomere sister chromatid exchange in unchallenged cells. The incidence of common fragile sites and telomere sister chromatid exchange increased markedly after treatment with replication inhibitors. Congruently, telomere-induced foci were frequently observed in the absence of Brca2, denoting activation of the DNA damage response and abnormal chromosome end joining. These telomere end fusions constituted a significant portion of chromosome aberrations in Brca2-deficient cells. Our results suggest that BRCA2 is required for telomere homeostasis and may be particularly important for the replication of G-rich telomeric lagging strands.


Asunto(s)
Proteína BRCA2/metabolismo , Fibroblastos/metabolismo , Homeostasis/fisiología , Fase S/fisiología , Telómero/metabolismo , Animales , Proteína BRCA2/genética , Células Cultivadas , Aberraciones Cromosómicas , Daño del ADN/fisiología , Fibroblastos/citología , Humanos , Ratones , Ratones Noqueados , Intercambio de Cromátides Hermanas/fisiología , Telómero/genética
19.
PLoS Genet ; 7(3): e1001329, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21423721

RESUMEN

The chiasma is a structure that forms between a pair of homologous chromosomes by crossover recombination and physically links the homologous chromosomes during meiosis. Chiasmata are essential for the attachment of the homologous chromosomes to opposite spindle poles (bipolar attachment) and their subsequent segregation to the opposite poles during meiosis I. However, the overall function of chiasmata during meiosis is not fully understood. Here, we show that chiasmata also play a crucial role in the attachment of sister chromatids to the same spindle pole and in their co-segregation during meiosis I in fission yeast. Analysis of cells lacking chiasmata and the cohesin protector Sgo1 showed that loss of chiasmata causes frequent bipolar attachment of sister chromatids during anaphase. Furthermore, high time-resolution analysis of centromere dynamics in various types of chiasmate and achiasmate cells, including those lacking the DNA replication checkpoint factor Mrc1 or the meiotic centromere protein Moa1, showed the following three outcomes: (i) during the pre-anaphase stage, the bipolar attachment of sister chromatids occurs irrespective of chiasma formation; (ii) the chiasma contributes to the elimination of the pre-anaphase bipolar attachment; and (iii) when the bipolar attachment remains during anaphase, the chiasmata generate a bias toward the proper pole during poleward chromosome pulling that results in appropriate chromosome segregation. Based on these results, we propose that chiasmata play a pivotal role in the selection of proper attachments and provide a backup mechanism that promotes correct chromosome segregation when improper attachments remain during anaphase I.


Asunto(s)
Cromátides/metabolismo , Segregación Cromosómica/fisiología , Cromosomas Fúngicos/metabolismo , Meiosis/fisiología , Schizosaccharomyces/genética , Intercambio de Cromátides Hermanas/fisiología , Anafase , Proteínas de Ciclo Celular/genética , Centrómero/metabolismo , Centrómero/ultraestructura , Cromátides/ultraestructura , Proteínas Cromosómicas no Histona/genética , Cromosomas Fúngicos/genética , Cromosomas Fúngicos/ultraestructura , Proteínas de Unión al ADN/genética , Eliminación de Gen , Proteínas Mad2 , Proteínas Nucleares/genética , Schizosaccharomyces/citología , Proteínas de Schizosaccharomyces pombe/genética
20.
Cell Cycle ; 10(2): 222-8, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21224723

RESUMEN

DNA double-strand breaks (DSBs) are arguably the most important lesions induced by ionizing radiation (IR) since unrepaired or mis-repaired DSBs can lead to chromosomal aberrations and cell death. The two major pathways to repair IR-induced DSBs are non-homologous end-joining (NHEJ) and homologous recombination (HR). Perhaps surprisingly, NHEJ represents the predominant pathway in the G1 and G2 phases of the cell cycle, but HR also contributes and repairs a subset of IR-induced DSBs in G2. Following S-phase-dependent genotoxins, HR events give rise to sister chromatid exchanges (SCEs), which can be detected cytogenetically in mitosis. Here, we describe that HR occurring in G2-irradiated cells also generates SCEs in ~50% of HR events. Since HR of IR-induced DSBs in G2 is a slow process, SCE formation in G2-irradiated cells requires several hours. During this time, irradiated S-phase cells can also reach mitosis, which has contributed to the widely held belief that SCEs form only during S phase. We describe procedures to measure SCEs exclusively in G2-irradiated cells and provide evidence that following IR cells do not need to progress through S phase in order to form SCEs.


Asunto(s)
Radiación Ionizante , Intercambio de Cromátides Hermanas/fisiología , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Reparación del ADN , Fase G2 , Histonas/metabolismo , Humanos , Mitosis , Recombinación Genética , Fase S
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...