Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 477
Filtrar
Más filtros

Intervalo de año de publicación
1.
Plant Cell ; 35(5): 1496-1512, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-36753568

RESUMEN

Cell divisions are accurately positioned to generate cells of the correct size and shape. In plant cells, the new cell wall is built in the middle of the cell by vesicles trafficked along an antiparallel microtubule and a microfilament array called the phragmoplast. The phragmoplast expands toward a specific location at the cell cortex called the division site, but how it accurately reaches the division site is unclear. We observed microtubule arrays that accumulate at the cell cortex during the telophase transition in maize (Zea mays) leaf epidermal cells. Before the phragmoplast reaches the cell cortex, these cortical-telophase microtubules transiently interact with the division site. Increased microtubule plus end capture and pausing occur when microtubules contact the division site-localized protein TANGLED1 or other closely associated proteins. Microtubule capture and pausing align the cortical microtubules perpendicular to the division site during telophase. Once the phragmoplast reaches the cell cortex, cortical-telophase microtubules are incorporated into the phragmoplast primarily by parallel bundling. The addition of microtubules into the phragmoplast promotes fine-tuning of the positioning at the division site. Our hypothesis is that division site-localized proteins such as TANGLED1 organize cortical microtubules during telophase to mediate phragmoplast positioning at the final division plane.


Asunto(s)
Arabidopsis , Zea mays , Zea mays/genética , Citocinesis , Telofase , Microtúbulos/metabolismo , Mitosis
2.
EMBO Rep ; 25(7): 3137-3159, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38877171

RESUMEN

Junctions between the endoplasmic reticulum (ER) and the outer membrane of the nuclear envelope (NE) physically connect both organelles. These ER-NE junctions are essential for supplying the NE with lipids and proteins synthesized in the ER. However, little is known about the structure of these ER-NE junctions. Here, we systematically study the ultrastructure of ER-NE junctions in cryo-fixed mammalian cells staged in anaphase, telophase, and interphase by correlating live cell imaging with three-dimensional electron microscopy. Our results show that ER-NE junctions in interphase cells have a pronounced hourglass shape with a constricted neck of 7-20 nm width. This morphology is significantly distinct from that of junctions within the ER network, and their morphology emerges as early as telophase. The highly constricted ER-NE junctions are seen in several mammalian cell types, but not in budding yeast. We speculate that the unique and highly constricted ER-NE junctions are regulated via novel mechanisms that contribute to ER-to-NE lipid and protein traffic in higher eukaryotes.


Asunto(s)
Retículo Endoplásmico , Mitosis , Membrana Nuclear , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Membrana Nuclear/metabolismo , Membrana Nuclear/ultraestructura , Humanos , Animales , Núcleo Celular/metabolismo , Células HeLa , Interfase , Telofase
3.
Mol Cell ; 61(5): 774-787, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26942680

RESUMEN

Centromeres are specialized chromatin domains specified by the centromere-specific CENP-A nucleosome. The stable inheritance of vertebrate centromeres is an epigenetic process requiring deposition of new CENP-A nucleosomes by HJURP. We show HJURP is recruited to centromeres through a direct interaction between the HJURP centromere targeting domain and the Mis18α-ß C-terminal coiled-coil domains. We demonstrate Mis18α and Mis18ß form a heterotetramer through their C-terminal coiled-coil domains. Mis18α-ß heterotetramer formation is required for Mis18BP1 binding and centromere recognition. S. pombe contains a single Mis18 isoform that forms a homotetramer, showing tetrameric Mis18 is conserved from fission yeast to humans. HJURP binding disrupts the Mis18α-ß heterotetramer and removes Mis18α from centromeres. We propose stable binding of Mis18 to centromeres in telophase licenses them for CENP-A deposition. Binding of HJURP deposits CENP-A at centromeres and facilitates the removal of Mis18, restricting CENP-A deposition to a single event per cell cycle.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Centrómero/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Telofase , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Autoantígenos/genética , Autoantígenos/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proteína A Centromérica , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Datos de Secuencia Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Transducción de Señal , Transfección
4.
J Cell Sci ; 134(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34387316

RESUMEN

Dephosphorylation of lamin A, which triggers nuclear lamina reconstitution, is crucial for the completion of mitosis. However, the specific phosphatase and regulatory mechanism that allow timely lamin A dephosphorylation remain unclear. Here, we report that RepoMan (also known as CDCA2), a regulatory subunit of protein phosphatase 1γ (PP1γ) is transiently modified with SUMO-2 at K762 during late telophase. SUMOylation of RepoMan markedly enhanced its binding affinity with lamin A. Moreover, SUMOylated RepoMan contributes to lamin A recruitment to telophase chromosomes and dephosphorylation of the mitotic lamin A phosphorylation. Expression of a SUMO-2 mutant that has a defective interaction with the SUMO-interacting motif (SIM) resulted in failure of the lamin A and RepoMan association, along with abrogation of lamin A dephosphorylation and subsequent nuclear lamina formation. These findings strongly suggest that RepoMan recruits lamin A through SUMO-SIM interaction. Thus, transient SUMOylation of RepoMan plays an important role in the spatiotemporal regulation of lamin A dephosphorylation and the subsequent nuclear lamina formation at the end of mitosis.


Asunto(s)
Lamina Tipo A , Sumoilación , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Mitosis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Telofase
5.
J Mol Recognit ; 36(7): e3019, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37061787

RESUMEN

Cell mechanics is a factor that determines cell growth, migration, proliferation, or differentiation, as well as trafficking inside the cytoplasm and organization of organelles. Knowledge about cell mechanics is critical to gaining insight into these biological processes. Here, we used atomic force microscopy to examine the elasticity, an important parameter of cell mechanics, of non-adherent Jurkat leukemic T-cells in both interphase and mitotic phases. We found that the elasticity of an individual cell does not significantly change at interphase. When a cell starts to divide, its elasticity increases in the transition from metaphase to telophase during normal division while the cell is stiffened right after it enters mitosis during abnormal division. At the end of the division, the cell elasticity gradually returned to the value of the mother cell. These changes may originate from the changes in cell surface tension during modulating actomyosin at the cleavage furrow, redistributing cell organelles, and constricting the contractile ring to sever mother cell to form daughters. The difference in elasticity patterns suggests that there is a discrepancy in the redistribution of the cell organelles during normal and abnormal division.


Asunto(s)
Mitosis , Linfocitos T , Ciclo Celular , Telofase , Interfase
6.
Cell ; 132(5): 771-82, 2008 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-18329364

RESUMEN

Toward the end of mitosis, neighboring chromosomes gather closely to form a compact cluster. This is important for reassembling the nuclear envelope around the entire chromosome mass but not individual chromosomes. By analyzing mice and cultured cells lacking the expression of chromokinesin Kid/kinesin-10, we show that Kid localizes to the boundaries of anaphase and telophase chromosomes and contributes to the shortening of the anaphase chromosome mass along the spindle axis. Loss of Kid-mediated anaphase chromosome compaction often causes the formation of multinucleated cells, specifically at oocyte meiosis II and the first couple of mitoses leading to embryonic death. In contrast, neither male meiosis nor somatic mitosis after the morula-stage is affected by Kid deficiency. These data suggest that Kid-mediated anaphase/telophase chromosome compaction prevents formation of multinucleated cells. This protection is especially important during the very early stages of development, when the embryonic cells are rich in ooplasm.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Proteínas de Unión al ADN/metabolismo , Cinesinas/metabolismo , Membrana Nuclear/metabolismo , Anafase , Animales , Blastómeros/metabolismo , Cruzamientos Genéticos , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Células HeLa , Humanos , Masculino , Ratones , Telofase
7.
J Cell Sci ; 132(14)2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31315909

RESUMEN

An acto-myosin contractile ring, which forms after anaphase onset and is highly regulated in time and space, mediates cytokinesis, the final step of mitosis. The chromosomal passenger complex (CPC), composed of Aurora-B kinase, INCENP, borealin and survivin (also known as BIRC5), regulates various processes during mitosis, including cytokinesis. It is not understood, however, how CPC regulates cytokinesis. We show that survivin binds to non-muscle myosin II (NMII), regulating its filament assembly. Survivin and NMII interact mainly in telophase, and Cdk1 regulates their interaction in a mitotic-phase-specific manner, revealing the mechanism for the specific timing of survivin-NMII interaction during mitosis. The survivin-NMII interaction is indispensable for cytokinesis, and its disruption leads to multiple mitotic defects. We further show that only the survivin homodimer binds to NMII, attesting to the biological importance for survivin homodimerization. We suggest a novel function for survivin in regulating the spatio-temporal formation of the acto-NMII contractile ring during cytokinesis and we elucidate the role of Cdk1 in regulating this process.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Citocinesis , Miosina Tipo II/metabolismo , Survivin/metabolismo , Proteína Quinasa CDC2/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Mitosis , Modelos Biológicos , Miosina Tipo II/química , Fosforilación , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Telofase
8.
Nature ; 522(7555): 236-9, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-26040713

RESUMEN

During telophase, the nuclear envelope (NE) reforms around daughter nuclei to ensure proper segregation of nuclear and cytoplasmic contents. NE reformation requires the coating of chromatin by membrane derived from the endoplasmic reticulum, and a subsequent annular fusion step to ensure that the formed envelope is sealed. How annular fusion is accomplished is unknown, but it is thought to involve the p97 AAA-ATPase complex and bears a topological equivalence to the membrane fusion event that occurs during the abscission phase of cytokinesis. Here we show that the endosomal sorting complex required for transport-III (ESCRT-III) machinery localizes to sites of annular fusion in the forming NE in human cells, and is necessary for proper post-mitotic nucleo-cytoplasmic compartmentalization. The ESCRT-III component charged multivesicular body protein 2A (CHMP2A) is directed to the forming NE through binding to CHMP4B, and provides an activity essential for NE reformation. Localization also requires the p97 complex member ubiquitin fusion and degradation 1 (UFD1). Our results describe a novel role for the ESCRT machinery in cell division and demonstrate a conservation of the machineries involved in topologically equivalent mitotic membrane remodelling events.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Membrana Nuclear/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Línea Celular , Cromatina/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/deficiencia , Humanos , Péptidos y Proteínas de Señalización Intracelular , Fusión de Membrana , Mitosis , Transporte de Proteínas , Proteínas/metabolismo , Telofase
9.
PLoS Genet ; 14(5): e1007388, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29813053

RESUMEN

It is widely accepted in eukaryotes that the cleavage furrow only initiates after mitosis completion. In fission yeast, cytokinesis requires the synthesis of a septum tightly coupled to cleavage furrow ingression. The current cytokinesis model establishes that simultaneous septation and furrow ingression only initiate after spindle breakage and mitosis exit. Thus, this model considers that although Cdk1 is inactivated at early-anaphase, septation onset requires the long elapsed time until mitosis completion and full activation of the Hippo-like SIN pathway. Here, we studied the precise timing of septation onset regarding mitosis by exploiting both the septum-specific detection with the fluorochrome calcofluor and the high-resolution electron microscopy during anaphase and telophase. Contrarily to the existing model, we found that both septum and cleavage furrow start to ingress at early anaphase B, long before spindle breakage, with a slow ingression rate during anaphase B, and greatly increasing after telophase onset. This shows that mitosis and cleavage furrow ingression are not concatenated but simultaneous events in fission yeast. We found that the timing of septation during early anaphase correlates with the cell size and is regulated by the corresponding levels of SIN Etd1 and Rho1. Cdk1 inactivation was directly required for timely septation in early anaphase. Strikingly the reduced SIN activity present after Cdk1 loss was enough to trigger septation by immediately inducing the medial recruitment of the SIN kinase complex Sid2-Mob1. On the other hand, septation onset did not depend on the SIN asymmetry establishment, which is considered a hallmark for SIN activation. These results recalibrate the timing of key cytokinetic events in fission yeast; and unveil a size-dependent control mechanism that synchronizes simultaneous nuclei separation with septum and cleavage furrow ingression to safeguard the proper chromosome segregation during cell division.


Asunto(s)
Anafase/fisiología , Proteínas de Ciclo Celular/fisiología , Citocinesis/fisiología , Proteínas de Schizosaccharomyces pombe/fisiología , Schizosaccharomyces/fisiología , Huso Acromático/fisiología , Bencenosulfonatos/química , Proteína Quinasa CDC2/fisiología , Núcleo Celular/fisiología , Microscopía Electrónica de Transmisión , Microscopía Fluorescente/métodos , Proteínas Quinasas/fisiología , Schizosaccharomyces/ultraestructura , Huso Acromático/ultraestructura , Telofase/fisiología , Factores de Tiempo , Proteínas de Unión al GTP rho/fisiología
10.
Int J Mol Sci ; 22(6)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33802105

RESUMEN

Several lines of evidence suggest the existence in the eukaryotic cells of a tight, yet largely unexplored, connection between DNA replication and sister chromatid cohesion. Tethering of newly duplicated chromatids is mediated by cohesin, an evolutionarily conserved hetero-tetrameric protein complex that has a ring-like structure and is believed to encircle DNA. Cohesin is loaded onto chromatin in telophase/G1 and converted into a cohesive state during the subsequent S phase, a process known as cohesion establishment. Many studies have revealed that down-regulation of a number of DNA replication factors gives rise to chromosomal cohesion defects, suggesting that they play critical roles in cohesion establishment. Conversely, loss of cohesin subunits (and/or regulators) has been found to alter DNA replication fork dynamics. A critical step of the cohesion establishment process consists in cohesin acetylation, a modification accomplished by dedicated acetyltransferases that operate at the replication forks. Defects in cohesion establishment give rise to chromosome mis-segregation and aneuploidy, phenotypes frequently observed in pre-cancerous and cancerous cells. Herein, we will review our present knowledge of the molecular mechanisms underlying the functional link between DNA replication and cohesion establishment, a phenomenon that is unique to the eukaryotic organisms.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromátides/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica/fisiología , Replicación del ADN/fisiología , Fase G1/fisiología , Telofase/fisiología , Animales , Humanos , Cohesinas
11.
Ecotoxicol Environ Saf ; 192: 110328, 2020 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-32078840

RESUMEN

In this study pillar[5]arene (P5) and a quinoline-functionalized pillar[5]arene (P5-6Q) which is used for detecting radioactive element, gas adsorption and toxic ions were synthesized. These materials were characterized by Nuclear Magnetic Resonance (NMR), Fourier Transform Infrared (FTIR), elemental analysis, melting point, Mass Spectroscopy, Scanning Electron Microscopy (SEM) and Zeta Potential. The cytotoxic and genotoxic potential of P5 and P5-6Q at distinct concentrations of 12.5, 25, 50, and 100 µg/mL were also investigated by Allium ana-telophase and comet assays on Allium cepa roots and Drosophila melanogaster haemocytes. P5 and P5-6Q showed dose dependent cytotoxic effect by decreasing mitotic index (MI) and genotoxic effect by increasing chromosomal aberrations (CAs such as disturbed anaphase-telophase, polyploidy, stickiness, chromosome laggards and bridges) and DNA damage at the exposed concentrations. These changes in P5-6Q were lower than P5. Further research is necessary to clarify the cytotoxic and genotoxic action mechanisms of P5 and P5-6Q at molecular levels.


Asunto(s)
Calixarenos/toxicidad , Daño del ADN , Drosophila melanogaster/efectos de los fármacos , Cebollas/efectos de los fármacos , Anafase/efectos de los fármacos , Animales , Calixarenos/química , Aberraciones Cromosómicas , Ensayo Cometa , Citotoxinas/química , Citotoxinas/toxicidad , Drosophila melanogaster/genética , Hemocitos/efectos de los fármacos , Índice Mitótico , Cebollas/genética , Raíces de Plantas/efectos de los fármacos , Quinolinas/síntesis química , Quinolinas/química , Quinolinas/toxicidad , Telofase/efectos de los fármacos
12.
Genes Chromosomes Cancer ; 58(5): 305-316, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30387289

RESUMEN

Some regions of the genome, notably common fragile sites (CFSs), are hypersensitive to replication stress and often involved in the generation of gross chromosome rearrangements in cancer cells. CFSs nest within very large genes and display cell-type-dependent instability. Fragile or not, large genes tend to replicate late in S-phase. A number of data now show that transcription perturbs replication completion across the body of large genes, particularly upon replication stress. However, the molecular mechanisms by which transcription elicits such under-replication and subsequent instability remain unclear. We present here our view of the mechanisms responsible for CFS under-replication and those allowing the cells to cope with this problem in G2 and mitosis. We notably focus on the major role played by the FANC proteins in the protection of CFSs from S phase up to late mitosis. We finally discuss a possible rationale for the conservation of large genes across vertebrate evolution.


Asunto(s)
Sitios Frágiles del Cromosoma , Fase S/genética , Telofase/genética , Animales , Evolución Molecular , Humanos
13.
J Biol Chem ; 292(37): 15254-15265, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28620047

RESUMEN

Squamous cell carcinoma-related oncogene (SCCRO)/DCUN1D1, a component of the neddylation E3 complex, regulates the activity of the cullin-RING-ligase type of ubiquitination E3s by promoting neddylation of cullin family members. Studies have shown that SCCRO regulates proliferation in vitro and in vivo Here we show that inactivation of SCCRO results in prolonged mitotic time because of delayed and/or failed abscission. The effects of SCCRO on abscission involve its role in neddylation and localization of Cul3 to the midbody. The Cul3 adaptor KLHL21 mediates the effects of SCCRO on abscission, as it fails to localize to the midbody in SCCRO-deficient cells during abscission, and its inactivation resulted in phenotypic changes identical to SCCRO inactivation. Ubiquitination-promoted turnover of Aurora B at the midbody was deficient in SCCRO- and KLHL21-deficient cells, suggesting that it is the target of Cul3KLHL21 at the midbody. Correction of abscission delays in SCCRO-deficient cells with addition of an Aurora B inhibitor at the midbody stage suggests that Aurora B is the target of SCCRO-promoted Cul3KLHL21 activity. The activity of other Cul3-anchored complexes, including Cul3KLHL9/KLHL13, was intact in SCCRO-deficient cells, suggesting that SCCRO selectively, rather than collectively, neddylates cullins in vivo Combined, these findings support a model in which the SCCRO, substrate, and substrate adaptors cooperatively provide tight control of neddylation and cullin-RING-ligase activity in vivo.


Asunto(s)
Proteínas Cullin/metabolismo , Proteínas de Microfilamentos/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/metabolismo , Ubiquitinas/metabolismo , Sustitución de Aminoácidos , Animales , Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Biomarcadores/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Proteínas Cullin/química , Proteínas Cullin/genética , Proteínas del Citoesqueleto , Embrión de Mamíferos/citología , Péptidos y Proteínas de Señalización Intracelular , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Microscopía Confocal , Mutación , Proteína NEDD8 , Multimerización de Proteína , Transporte de Proteínas , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Telofase , Imagen de Lapso de Tiempo
14.
Plant Physiol ; 173(1): 863-871, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27881728

RESUMEN

A sensitive and dynamically responsive auxin signaling reporter based on the DII domain of the INDOLE-3-ACETIC ACID28 (IAA28, DII) protein from Arabidopsis (Arabidopsis thaliana) was modified for use in maize (Zea mays). The DII domain was fused to a yellow fluorescent protein and a nuclear localization sequence to simplify quantitative nuclear fluorescence signal. DII degradation dynamics provide an estimate of input signal into the auxin signaling pathway that is influenced by both auxin accumulation and F-box coreceptor concentration. In maize, the DII-based marker responded rapidly and in a dose-dependent manner to exogenous auxin via proteasome-mediated degradation. Low levels of DII-specific fluorescence corresponding to high endogenous auxin signaling occurred near vasculature tissue and the outer layer and glume primordia of spikelet pair meristems and floral meristems, respectively. In addition, high DII levels were observed in cells during telophase and early G1, suggesting that low auxin signaling at these stages may be important for cell cycle progression.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Ácidos Indolacéticos/metabolismo , Telofase/fisiología , Factores de Transcripción/metabolismo , Zea mays/citología , Proteínas de Arabidopsis/genética , Fase G1/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ácidos Indolacéticos/farmacología , Meristema/genética , Meristema/metabolismo , Plantas Modificadas Genéticamente , Dominios Proteicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Imagen de Lapso de Tiempo , Factores de Transcripción/genética , Zea mays/efectos de los fármacos , Zea mays/genética , Zea mays/metabolismo
15.
Exp Cell Res ; 360(2): 347-357, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28942021

RESUMEN

Cell division, in which duplicated chromosomes are separated into two daughter cells, is the most dynamic event during cell proliferation. Chromosome movement is powered mainly by microtubules, which vary in morphology and are organized into characteristic structures according to mitotic progression. During the later stages of mitosis, antiparallel microtubules form the spindle midzone, and the irregular formation of the midzone often leads to failure of cytokinesis, giving rise to the unequal segregation of chromosomes. However, it is difficult to analyze the morphology of these microtubules because microtubules in the antiparallel overlaps of microtubule-plus ends in the midzone are embedded in highly electron-dense matrices, impeding the access of anti-tubulin antibodies to their epitopes during immunofluorescence staining. Here, we developed a novel method to visualize selectively antiparallel microtubule overlaps in the midzone. When cells are air-dried before fixation, aligned α-tubulin staining is observed and colocalized with PRC1 in the center of the midzone of anaphase and telophase cells, suggesting that antiparallel microtubule overlaps can be visualized by this method. In air-dried cells, mCherry-α-tubulin fluorescence and ß-tubulin staining show almost the same pattern as α-tubulin staining in the midzone, suggesting that the selective visualization of antiparallel microtubule overlaps in air-dried cells is not attributed to an alteration of the antigenicity of α-tubulin. Taxol treatment extends the microtubule filaments of the midzone in air-dried cells, and nocodazole treatment conversely decreases the number of microtubules, suggesting that unstable microtubules are depolymerized during the air-drying method. It is of note that the air-drying method enables the detection of the disruption of the midzone and premature midzone formation upon Aurora B and Plk1 inhibition, respectively. These results suggest that the air-drying method is suitable for visualizing microtubules in the antiparallel overlaps of microtubule-plus ends of the midzone and for detecting their effects on midzone formation.


Asunto(s)
Anafase , Técnica del Anticuerpo Fluorescente/métodos , Microtúbulos/metabolismo , Huso Acromático/metabolismo , Telofase , Animales , Células Cultivadas , Segregación Cromosómica/fisiología , Citocinesis/fisiología , Células HeLa , Humanos , Microtúbulos/ultraestructura , Mitosis , Huso Acromático/ultraestructura , Porcinos
16.
J Cell Sci ; 128(18): 3466-77, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26224877

RESUMEN

The metazoan nucleus breaks down and reassembles during each cell division. Upon mitotic exit, the successful reestablishment of an interphase nucleus requires the coordinated reorganization of chromatin and formation of a functional nuclear envelope. Here, we report that the histone demethylase LSD1 (also known as KDM1A) plays a crucial role in nuclear assembly at the end of mitosis. Downregulation of LSD1 in cells extends telophase and impairs nuclear pore complex assembly. In vitro, LSD1 demethylase activity is required for the recruitment of MEL28 (also known as ELYS and AHCTF1) and nuclear envelope precursor vesicles to chromatin, crucial steps in nuclear reassembly. Accordingly, the formation of a closed nuclear envelope and nuclear pore complex assembly are impaired upon depletion of LSD1 or inhibition of its activity. Our results identify histone demethylation by LSD1 as a new regulatory mechanism linking the chromatin state and nuclear envelope formation at the end of mitosis.


Asunto(s)
Ensamble y Desensamble de Cromatina , Histona Demetilasas/metabolismo , Membrana Nuclear/metabolismo , Telofase/fisiología , Animales , Células HeLa , Humanos , Xenopus laevis
17.
Genes Cells ; 21(9): 978-93, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27458047

RESUMEN

ICRF-193 [meso-4,4-(2,3-butanediyl)-bis(2,6-piperazinedione)] is a complex-stabilizing inhibitor of DNA topoisomerase II (topo II) that is used as an effective anticancer drug. ICRF-193 inhibits topo II catalytic activity in vitro and blocks nuclear division in vivo. Here, we examined the effects of ICRF-193 treatment on chromatin behavior and spindle dynamics using detailed live mitotic cell analysis in the fission yeast, Schizosaccharomyces pombe. Time-lapse movie analysis showed that ICRF-193 treatment leads to an elongation of presumed chromatin fibers connected to kinetochores during mid-mitosis. Anaphase spindles begin to arch, and eventually spindle poles come together abruptly, as if the spindle snapped at the point of spindle microtubule overlap in telophase. Segregating chromosomes appeared as elastic clumps and subsequently pulled back and merged. The snapped spindle phenotype was abolished by microtubule destabilization after thiabendazole treatment, accompanied by unequal chromosome segregation or severe defects in spindle extension. Thus, we conclude that ICRF-193-treated, unseparated sister chromatids pulling toward opposite spindle poles produce the arched and snapped telophase spindle. ICRF-193 treatment increased DNA content, suggesting that the failure of sister chromatids to separate properly in anaphase, causes the spindle to break in telophase, resulting in polyploidization.


Asunto(s)
Piperazinas/farmacología , Schizosaccharomyces/efectos de los fármacos , Huso Acromático/efectos de los fármacos , Telofase/efectos de los fármacos , Anafase/efectos de los fármacos , Anafase/fisiología , Antineoplásicos/farmacología , Proteínas de Ciclo Celular/genética , División del Núcleo Celular , Cromátides/efectos de los fármacos , Cromátides/genética , Cromátides/metabolismo , Segregación Cromosómica , Dicetopiperazinas , Cinetocoros/metabolismo , Microtúbulos/efectos de los fármacos , Mitosis , Ploidias , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Huso Acromático/fisiología , Telofase/fisiología , Inhibidores de Topoisomerasa II/farmacología
18.
Reprod Biol Endocrinol ; 15(1): 69, 2017 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851440

RESUMEN

BACKGROUND: Polo-like kinase 1 (Plk1), as a characteristic regulator in meiosis, organizes multiple biological events of cell division. Although Plk1 has been implicated in various functions in somatic cell mitotic processes, considerably less is known regarding its function during the transition from metaphase I (MI) to metaphase II (MII) stage in oocyte meiotic progression. METHODS: In this study, the possible role of Plk1 during the MI-to-MII stage transition in pig oocytes was addressed. Initially, the spatiotemporal expression and subcellular localization pattern of Plk1 were revealed in pig oocytes from MI to MII stage using indirect immunofluorescence and confocal microscopy imaging techniques combined with western blot analyses. Moreover, a highly selective Plk1 inhibitor, GSK461364, was used to determine the potential role of Plk1 during this MI-to-MII transition progression. RESULTS: Upon expression, Plk1 exhibited a specific dynamic intracellular localization, and co-localization of Plk1 with α-tubulin was revealed in the meiotic spindle of pig oocyte during the transition from MI to MII stage. GSK461364 treatment significantly blocked the first polar body (pbI) emission in a dose-dependent manner and resulted in a failure of meiotic maturation, with a larger percentage of the GSK461364-treated oocytes arresting in the anaphase-telophase I (ATI) stage. Further subcellular structure examination results showed that inhibition of Plk1 with GSK461364 had no visible effect on spindle assembly but caused a significantly higher proportion of the treated oocytes to have obvious defects in homologous chromosome segregation at ATI stage. CONCLUSIONS: Thus, these results indicate that Plk1 plays an essential role during the meiosis I/meiosis II transition in porcine oocytes, and the regulation is associated with Plk1's effects on homologous chromosome segregation in the ATI stage.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Segregación Cromosómica/genética , Meiosis/genética , Oocitos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Porcinos/genética , Anafase/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Femenino , Metafase/genética , Oocitos/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Fracciones Subcelulares , Telofase/genética , Distribución Tisular , Quinasa Tipo Polo 1
19.
20.
Bioessays ; 37(3): 257-66, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25470791

RESUMEN

Here we discuss a "chromosome separation checkpoint" that might regulate the anaphase-telophase transition. The concept of cell cycle checkpoints was originally proposed to account for extrinsic control mechanisms that ensure the order of cell cycle events. Several checkpoints have been shown to regulate major cell cycle transitions, namely at G1-S and G2-M. At the onset of mitosis, the prophase-prometaphase transition is controlled by several potential checkpoints, including the antephase checkpoint, while the spindle assembly checkpoint guards the metaphase-anaphase transition. Our hypothesis is based on the recently uncovered feedback control mechanism that delays chromosome decondensation and nuclear envelope reassembly until effective separation of sister chromatids during anaphase is achieved. A central player in this potential checkpoint is the establishment of a constitutive, midzone-based Aurora B phosphorylation gradient that monitors the position of chromosomes along the spindle axis. We propose that this surveillance mechanism represents an additional step towards ensuring mitotic fidelity.


Asunto(s)
Anafase , Telofase , Animales , Aurora Quinasa B/metabolismo , Proteína Quinasa CDC2/metabolismo , Línea Celular , Segregación Cromosómica , Drosophila melanogaster , Humanos , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA