Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 306
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 38: 25-48, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35395166

RESUMEN

The anaphase-promoting complex/cyclosome (APC/C) represents a large multisubunit E3-ubiquitin ligase complex that controls the unidirectional progression through the cell cycle by the ubiquitination of specific target proteins, marking them for proteasomal destruction. Although the APC/C's role is largely conserved among eukaryotes, its subunit composition and target spectrum appear to be species specific. In this review, we focus on the plant APC/C complex, whose activity correlates with different developmental processes, including polyploidization and gametogenesis. After an introduction into proteolytic control by ubiquitination, we discuss the composition of the plant APC/C and the essential nature of its core subunits for plant development. Subsequently, we describe the APC/C activator subunits and interactors, most being plant specific. Finally, we provide a comprehensive list of confirmed and suspected plant APC/C target proteins. Identification of growth-related targets might offer opportunities to increase crop yield and resilience of plants to climate change by manipulating APC/C activity.


Asunto(s)
Anafase , Plantas , Ciclosoma-Complejo Promotor de la Anafase/genética , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Plantas/genética , Plantas/metabolismo , Ubiquitinación , Ubiquitinas/metabolismo
2.
Cell ; 171(4): 918-933.e20, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29033132

RESUMEN

Posttranslational modification with ubiquitin chains controls cell fate in all eukaryotes. Depending on the connectivity between subunits, different ubiquitin chain types trigger distinct outputs, as seen with K48- and K63-linked conjugates that drive protein degradation or complex assembly, respectively. Recent biochemical analyses also suggested roles for mixed or branched ubiquitin chains, yet without a method to monitor endogenous conjugates, the physiological significance of heterotypic polymers remained poorly understood. Here, we engineered a bispecific antibody to detect K11/K48-linked chains and identified mitotic regulators, misfolded nascent polypeptides, and pathological Huntingtin variants as their endogenous substrates. We show that K11/K48-linked chains are synthesized and processed by essential ubiquitin ligases and effectors that are mutated across neurodegenerative diseases; accordingly, these conjugates promote rapid proteasomal clearance of aggregation-prone proteins. By revealing key roles of K11/K48-linked chains in cell-cycle and quality control, we establish heterotypic ubiquitin conjugates as important carriers of biological information.


Asunto(s)
Anticuerpos Biespecíficos/análisis , Transducción de Señal , Ubiquitina/metabolismo , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclo Celular , Humanos , Mitosis , Biosíntesis de Proteínas , Ubiquitinación
3.
Mol Cell ; 84(17): 3254-3270.e9, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39153474

RESUMEN

The individualization of chromosomes during early mitosis and their clustering upon exit from cell division are two key transitions that ensure efficient segregation of eukaryotic chromosomes. Both processes are regulated by the surfactant-like protein Ki-67, but how Ki-67 achieves these diametric functions has remained unknown. Here, we report that Ki-67 radically switches from a chromosome repellent to a chromosome attractant during anaphase in human cells. We show that Ki-67 dephosphorylation during mitotic exit and the simultaneous exposure of a conserved basic patch induce the RNA-dependent formation of a liquid-like condensed phase on the chromosome surface. Experiments and coarse-grained simulations support a model in which the coalescence of chromosome surfaces, driven by co-condensation of Ki-67 and RNA, promotes clustering of chromosomes. Our study reveals how the switch of Ki-67 from a surfactant to a liquid-like condensed phase can generate mechanical forces during genome segregation that are required for re-establishing nuclear-cytoplasmic compartmentalization after mitosis.


Asunto(s)
Segregación Cromosómica , Cromosomas Humanos , Antígeno Ki-67 , Mitosis , Humanos , Antígeno Ki-67/metabolismo , Antígeno Ki-67/genética , Células HeLa , Cromosomas Humanos/metabolismo , Cromosomas Humanos/genética , Fosforilación , Anafase
4.
Annu Rev Cell Dev Biol ; 33: 1-22, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28992437

RESUMEN

During my graduate work with Keith Porter, I became fascinated by the mitotic spindle, an interest that has motivated much of my scientific work ever since. I began spindle studies by using electron microscopes, instruments that have made significant contributions to our understanding of spindle organization. Such instruments have helped to elucidate the distributions of spindle microtubules, the interactions among them, their molecular polarity, and their associations with both kinetochores and spindle poles. Our lab has also investigated some processes of spindle physiology: microtubule dynamics, the actions of microtubule-associated proteins (including motor enzymes), the character of forces generated by specific spindle components, and factors that control mitotic progression. Here, I give a personal perspective on some of this intellectual history and on what recent discoveries imply about the mechanisms of chromosome motion.


Asunto(s)
Cromosomas/metabolismo , Enzimas/metabolismo , Microtúbulos/metabolismo , Mitosis , Proteínas Motoras Moleculares/metabolismo , Movimiento (Física) , Animales , Cromosomas/ultraestructura , Humanos , Microtúbulos/ultraestructura
5.
Mol Cell ; 82(1): 90-105.e13, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34942119

RESUMEN

Neurodevelopmental cognitive disorders provide insights into mechanisms of human brain development. Here, we report an intellectual disability syndrome caused by the loss of APC7, a core component of the E3 ubiquitin ligase anaphase promoting complex (APC). In mechanistic studies, we uncover a critical role for APC7 during the recruitment and ubiquitination of APC substrates. In proteomics analyses of the brain from mice harboring the patient-specific APC7 mutation, we identify the chromatin-associated protein Ki-67 as an APC7-dependent substrate of the APC in neurons. Conditional knockout of the APC coactivator protein Cdh1, but not Cdc20, leads to the accumulation of Ki-67 protein in neurons in vivo, suggesting that APC7 is required for the function of Cdh1-APC in the brain. Deregulated neuronal Ki-67 upon APC7 loss localizes predominantly to constitutive heterochromatin. Our findings define an essential function for APC7 and Cdh1-APC in neuronal heterochromatin regulation, with implications for understanding human brain development and disease.


Asunto(s)
Subunidad Apc7 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Encéfalo/enzimología , Heterocromatina/metabolismo , Discapacidad Intelectual/enzimología , Células-Madre Neurales/enzimología , Neurogénesis , Adolescente , Animales , Antígenos CD , Subunidad Apc7 del Ciclosoma-Complejo Promotor de la Anafase/genética , Conducta Animal , Encéfalo/crecimiento & desarrollo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Heterocromatina/genética , Humanos , Lactante , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Discapacidad Intelectual/psicología , Inteligencia , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis , Mutación , Células-Madre Neurales/patología , Proteolisis , Transducción de Señal , Síndrome , Ubiquitinación , Adulto Joven
6.
EMBO J ; 43(7): 1325-1350, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38321267

RESUMEN

Exit from mitosis is brought about by dramatic changes in the phosphoproteome landscape. A drop in Cyclin-dependent kinase (Cdk) activity, the master regulatory kinase, and activation of counteracting phosphatases such as Cdc14 in budding yeast, results in ordered substrate dephosphorylation, allowing entry into a new cell cycle and replication licensing. In meiosis however, two cell divisions have to be executed without intermediate DNA replication, implying that global phosphorylation and dephosphorylation have to be adapted to the challenges of meiosis. Using a global time-resolved phosphoproteomics approach in budding yeast, we compared the phosphoproteome landscape between mitotic exit and the transition from meiosis I to meiosis II. We found that unlike exit from mitosis, Cdk phosphomotifs remain mostly stably phosphorylated at the end of meiosis I, whereas a majority of Cdk-unrelated motifs are reset by dephosphorylation. However, inducing an artificial drop of Cdk at metaphase of meiosis I leads to ordered substrate dephosphorylation, comparable to mitosis, indicating that phosphoregulation of substrates at the end of meiosis I is thus mainly qualitatively rather than quantitatively ordered.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomycetales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Saccharomycetales/genética , Saccharomycetales/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Mitosis , Fosforilación , Meiosis
7.
EMBO J ; 43(5): 666-694, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38279026

RESUMEN

The efficacy of current antimitotic cancer drugs is limited by toxicity in highly proliferative healthy tissues. A cancer-specific dependency on the microtubule motor protein KIF18A therefore makes it an attractive therapeutic target. Not all cancers require KIF18A, however, and the determinants underlying this distinction remain unclear. Here, we show that KIF18A inhibition drives a modest and widespread increase in spindle assembly checkpoint (SAC) signaling from kinetochores which can result in lethal mitotic delays. Whether cells arrest in mitosis depends on the robustness of the metaphase-to-anaphase transition, and cells predisposed with weak basal anaphase-promoting complex/cyclosome (APC/C) activity and/or persistent SAC signaling through metaphase are uniquely sensitive to KIF18A inhibition. KIF18A-dependent cancer cells exhibit hallmarks of this SAC:APC/C imbalance, including a long metaphase-to-anaphase transition, and slow mitosis overall. Together, our data reveal vulnerabilities in the cell division apparatus of cancer cells that can be exploited for therapeutic benefit.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase , Neoplasias , Humanos , Ciclosoma-Complejo Promotor de la Anafase/genética , Dineínas , Cinesinas/genética , Cinetocoros , Mitosis , Neoplasias/genética
8.
Genes Dev ; 34(17-18): 1239-1251, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32820038

RESUMEN

A maize chromosome variant called abnormal chromosome 10 (Ab10) converts knobs on chromosome arms into neocentromeres, causing their preferential segregation to egg cells in a process known as meiotic drive. We previously demonstrated that the gene Kinesin driver (Kindr) on Ab10 encodes a kinesin-14 required to mobilize neocentromeres made up of the major tandem repeat knob180. Here we describe a second kinesin-14 gene, TR-1 kinesin (Trkin), that is required to mobilize neocentromeres made up of the minor tandem repeat TR-1. Trkin lies in a 4-Mb region of Ab10 that is not syntenic with any other region of the maize genome and shows extraordinary sequence divergence from Kindr and other kinesins in plants. Despite its unusual structure, Trkin encodes a functional minus end-directed kinesin that specifically colocalizes with TR-1 in meiosis, forming long drawn out neocentromeres. TRKIN contains a nuclear localization signal and localizes to knobs earlier in prophase than KINDR. The fact that TR-1 repeats often co-occur with knob180 repeats suggests that the current role of the TRKIN/TR-1 system is to facilitate the meiotic drive of the KINDR/knob180 system.


Asunto(s)
Centrómero/genética , Centrómero/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Zea mays/genética , Zea mays/metabolismo , Cromosomas de las Plantas/genética , Genes de Plantas/genética , Meiosis , Modelos Genéticos , Transporte de Proteínas/genética
9.
Genes Dev ; 34(17-18): 1110-1112, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32873577

RESUMEN

Maize heterochromatic knobs cheat female meiosis by forming neocentromeres that bias their segregation into the future egg cell. In this issue of Genes & Development, Swentowsky and colleagues (pp. 1239-1251) show that two types of knobs, those composed of 180-bp and TR1 sequences, recruit their own novel and divergent kinesin-14 family members to form neocentromeres.


Asunto(s)
Genoma de Planta , Zea mays/genética , Centrómero/genética , Genoma de Planta/genética , Cinesinas/genética , Cinesinas/metabolismo , Meiosis/genética
10.
Proc Natl Acad Sci U S A ; 121(25): e2323009121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38875144

RESUMEN

Error correction is central to many biological systems and is critical for protein function and cell health. During mitosis, error correction is required for the faithful inheritance of genetic material. When functioning properly, the mitotic spindle segregates an equal number of chromosomes to daughter cells with high fidelity. Over the course of spindle assembly, many initially erroneous attachments between kinetochores and microtubules are fixed through the process of error correction. Despite the importance of chromosome segregation errors in cancer and other diseases, there is a lack of methods to characterize the dynamics of error correction and how it can go wrong. Here, we present an experimental method and analysis framework to quantify chromosome segregation error correction in human tissue culture cells with live cell confocal imaging, timed premature anaphase, and automated counting of kinetochores after cell division. We find that errors decrease exponentially over time during spindle assembly. A coarse-grained model, in which errors are corrected in a chromosome-autonomous manner at a constant rate, can quantitatively explain both the measured error correction dynamics and the distribution of anaphase onset times. We further validated our model using perturbations that destabilized microtubules and changed the initial configuration of chromosomal attachments. Taken together, this work provides a quantitative framework for understanding the dynamics of mitotic error correction.


Asunto(s)
Segregación Cromosómica , Cinetocoros , Microtúbulos , Mitosis , Huso Acromático , Humanos , Cinetocoros/metabolismo , Huso Acromático/metabolismo , Microtúbulos/metabolismo , Anafase , Modelos Biológicos , Células HeLa
11.
Proc Natl Acad Sci U S A ; 121(30): e2319574121, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39024113

RESUMEN

Regulated cell cycle progression ensures homeostasis and prevents cancer. In proliferating cells, premature S phase entry is avoided by the E3 ubiquitin ligase anaphasepromoting complex/cyclosome (APC/C), although the APC/C substrates whose degradation restrains G1-S progression are not fully known. The APC/C is also active in arrested cells that exited the cell cycle, but it is not clear whether APC/C maintains all types of arrest. Here, by expressing the APC/C inhibitor, EMI1, we show that APC/C activity is essential to prevent S phase entry in cells arrested by pharmacological cyclin-dependent kinases 4 and 6 (CDK4/6) inhibition (Palbociclib). Thus, active protein degradation is required for arrest alongside repressed cell cycle gene expression. The mechanism of rapid and robust arrest bypass from inhibiting APC/C involves CDKs acting in an atypical order to inactivate retinoblastoma-mediated E2F repression. Inactivating APC/C first causes mitotic cyclin B accumulation which then promotes cyclin A expression. We propose that cyclin A is the key substrate for maintaining arrest because APC/C-resistant cyclin A, but not cyclin B, is sufficient to induce S phase entry. Cells bypassing arrest from CDK4/6 inhibition initiate DNA replication with severely reduced origin licensing. The simultaneous accumulation of S phase licensing inhibitors, such as cyclin A and geminin, with G1 licensing activators disrupts the normal order of G1-S progression. As a result, DNA synthesis and cell proliferation are profoundly impaired. Our findings predict that cancers with elevated EMI1 expression will tend to escape CDK4/6 inhibition into a premature, underlicensed S phase and suffer enhanced genome instability.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Humanos , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclosoma-Complejo Promotor de la Anafase/genética , Línea Celular Tumoral , Fase S/efectos de los fármacos , Piridinas/farmacología , Piperazinas/farmacología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Factores de Transcripción E2F/metabolismo , Factores de Transcripción E2F/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Ciclinas/metabolismo , Ciclinas/genética , Proteínas F-Box
12.
EMBO J ; 41(3): e108823, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942047

RESUMEN

Polyubiquitination by E2 and E3 enzymes is crucial to cell cycle control, epigenetic regulation, and development. The hallmark of the E2 family is the ubiquitin (Ub)-conjugating (UBC) domain that forms a dynamic thioester conjugate with ubiquitin (E2~Ub). Numerous studies have focused on E2 surfaces, such as the N-terminal and crossover helices, that directly interact with an E3 or the conjugated ubiquitin to stabilize the active, "closed" state of the E2~Ub. However, it remains unclear how other E2 surfaces regulate ubiquitin transfer. Here, we demonstrate the helix-turn-helix (HTH) motif of the UBC tunes the intrinsic polyubiquitination activity through distinct functions in different E2s. Interestingly, the E2HTH motif is repurposed in UBE2S and UBE2R2 to interact with the conjugated or acceptor ubiquitin, respectively, modulating ubiquitin transfer. Furthermore, we propose that Anaphase-Promoting Complex/Cyclosome binding to the UBE2SHTH reduces the conformational space of the flexible E2~Ub, demonstrating an atypical E3-dependent activation mechanism. Altogether, we postulate the E2HTH motif evolved to provide new functionalities that can be harnessed by E3s and permits additional regulation to facilitate specific E2-E3-mediated polyubiquitination.


Asunto(s)
Enzimas Ubiquitina-Conjugadoras/química , Secuencias de Aminoácidos , Dominio Catalítico , Humanos , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo
13.
Mol Cell ; 66(3): 398-410.e4, 2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475874

RESUMEN

Replication stress and mitotic abnormalities are key features of cancer cells. Temporarily paused forks are stabilized by the intra-S phase checkpoint and protected by the association of Rad51, which prevents Mre11-dependent resection. However, if a fork becomes dysfunctional and cannot resume, this terminally arrested fork is rescued by a converging fork to avoid unreplicated parental DNA during mitosis. Alternatively, dysfunctional forks are restarted by homologous recombination. Using fission yeast, we report that Rad52 and the DNA binding activity of Rad51, but not its strand-exchange activity, act to protect terminally arrested forks from unrestrained Exo1-nucleolytic activity. In the absence of recombination proteins, large ssDNA gaps, up to 3 kb long, occur behind terminally arrested forks, preventing efficient fork merging and leading to mitotic sister chromatid bridging. Thus, Rad52 and Rad51 prevent temporarily and terminally arrested forks from degrading and, despite the availability of converging forks, converting to anaphase bridges causing aneuploidy and cell death.


Asunto(s)
Replicación del ADN , ADN de Hongos/biosíntesis , ADN de Cadena Simple/biosíntesis , Mitosis/fisiología , Origen de Réplica , Schizosaccharomyces/metabolismo , Intercambio de Cromátides Hermanas , Aneuploidia , Cromosomas Fúngicos/genética , Cromosomas Fúngicos/metabolismo , Roturas del ADN de Cadena Simple , ADN de Hongos/genética , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Viabilidad Microbiana , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/crecimiento & desarrollo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Factores de Tiempo
14.
Mol Cell ; 68(3): 605-614.e4, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100057

RESUMEN

Cohesins establish sister chromatid cohesion during S phase and are removed when cohesin Scc1 is cleaved by separase at anaphase onset. During this process, cohesin Smc3 undergoes a cycle of acetylation: Smc3 acetylation by Eco1 in S phase stabilizes cohesin association with chromosomes, and its deacetylation by Hos1 in anaphase allows re-use of Smc3 in the next cell cycle. Here we find that Smc3 deacetylation by Hos1 has a more immediate effect in the early anaphase of budding yeast. Hos1 depletion significantly delayed sister chromatid separation and segregation. Smc3 deacetylation facilitated removal of cohesins from chromosomes without changing Scc1 cleavage efficiency, promoting dissolution of cohesion. This action is probably due to disengagement of Smc1-Smc3 heads prompted by de-repression of their ATPase activity. We suggest Scc1 cleavage per se is insufficient for efficient dissolution of cohesion in early anaphase; subsequent Smc3 deacetylation, triggered by Scc1 cleavage, is also required.


Asunto(s)
Anafase , Proteínas de Ciclo Celular/metabolismo , Cromátides/enzimología , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica , Histona Desacetilasas/metabolismo , Histona Demetilasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Acetilación , Proteínas de Ciclo Celular/genética , Cromátides/genética , Proteínas Cromosómicas no Histona/genética , Histona Desacetilasas/genética , Histona Demetilasas/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética , Separasa/genética , Separasa/metabolismo , Transducción de Señal , Factores de Tiempo , Cohesinas
15.
Cell Mol Life Sci ; 81(1): 251, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38847937

RESUMEN

The Smc5/6 complex is a highly conserved molecular machine involved in the maintenance of genome integrity. While its functions largely depend on restraining the fork remodeling activity of Mph1 in yeast, the presence of an analogous Smc5/6-FANCM regulation in humans remains unknown. We generated human cell lines harboring mutations in the NSE1 subunit of the Smc5/6 complex. Point mutations or truncations in the RING domain of NSE1 result in drastically reduced Smc5/6 protein levels, with differential contribution of the two zinc-coordinating centers in the RING. In addition, nse1-RING mutant cells display cell growth defects, reduced replication fork rates, and increased genomic instability. Notably, our findings uncover a synthetic sick interaction between Smc5/6 and FANCM and show that Smc5/6 controls fork progression and chromosome disjunction in a FANCM-independent manner. Overall, our study demonstrates that the NSE1 RING domain plays vital roles in Smc5/6 complex stability and fork progression through pathways that are not evolutionary conserved.


Asunto(s)
Proteínas de Ciclo Celular , Replicación del ADN , Inestabilidad Genómica , Humanos , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/genética , Dominios Proteicos , Estabilidad Proteica , Mutación , Línea Celular , ADN Helicasas
16.
Proc Natl Acad Sci U S A ; 119(39): e2204068119, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-36122237

RESUMEN

Chromosome segregation requires load-bearing interactions across kinetochore fibers and antiparallel microtubule bundles, which constitute the spindle midzone. Mechanical properties of kinetochore fibers have been characterized during metaphase, when the mitotic spindle achieves steady state. However, it has been difficult to probe the mechanics of the spindle midzone that elongates during anaphase. Here, we combine superresolution expansion and electron microscopies, lattice light-sheet imaging, and laser microsurgery to examine how midzone organization sets its mechanics. We find that individual midzone bundles extend out to multiple positions across chromosomes and form multiple apparent microtubule-based connections with each other. Across the spindle's short axis, these microtubule bundles exhibit restricted, submicrometer-amplitude motions, which are weakly correlated on <10s timescales. Severing individual midzone bundles near their center does not substantially affect positions of neighboring bundles, nor the overall structural stability of the midzone. In contrast, severing multiple midzone bundles or individual bundles at their chromosome-proximal ends significantly displaces neighboring microtubule bundles. Together, these data suggest a model wherein multiple midzone connections both reinforce its structure and mechanically isolate individual bundles from local perturbations. This feature sets the robust midzone architecture to accommodate disruptions, including those which result from lagging chromosomes, and achieve stereotypic outputs, such as proper chromosome separation.


Asunto(s)
Anafase , Huso Acromático , Segregación Cromosómica , Cinetocoros , Microtúbulos
17.
Proc Natl Acad Sci U S A ; 119(10): e2123363119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35235450

RESUMEN

During mitosis, from late prophase onward, sister chromatids are connected along their entire lengths by axis-linking chromatin/structure bridges. During prometaphase/metaphase, these bridges ensure that sister chromatids retain a parallel, paranemic relationship, without helical coiling, as they undergo compaction. Bridges must then be removed during anaphase. Motivated by these findings, the present study has further investigated the process of anaphase sister separation. Morphological and functional analyses of mammalian mitoses reveal a three-stage pathway in which interaxis bridges play a prominent role. First, sister chromatid axes globally separate in parallel along their lengths, with concomitant bridge elongation, due to intersister chromatin pushing forces. Sister chromatids then peel apart progressively from a centromere to telomere region(s), step-by-step. During this stage, poleward spindle forces dramatically elongate centromere-proximal bridges, which are then removed by a topoisomerase IIα­dependent step. Finally, in telomere regions, widely separated chromatids remain invisibly linked, presumably by catenation, with final separation during anaphase B. During this stage increased separation of poles and/or chromatin compaction appear to be the driving force(s). Cohesin cleavage licenses these events, likely by allowing bridges to respond to imposed forces. We propose that bridges are not simply removed during anaphase but, in addition, play an active role in ensuring smooth and synchronous microtubule-mediated sister separation. Bridges would thereby be the topological gatekeepers of sister chromatid relationships throughout all stages of mitosis.


Asunto(s)
Anafase , Cromátides , Intercambio de Cromátides Hermanas , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Humanos , Cohesinas
18.
J Cell Sci ; 135(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35237831

RESUMEN

Mammalian oocytes are arrested at meiotic prophase I. The dual-specificity phosphatase CDC25B is essential for cyclin-dependent kinase 1 (CDK1) activation that drives resumption of meiosis. CDC25B reverses the inhibitory effect of the protein kinases WEE1 and MYT1 on CDK1 activation. Cdc25b-/- female mice are infertile because oocytes cannot activate CDK1. To identify a role for CDC25B following resumption of meiosis, we restored CDK1 activation in Cdc25b-/- oocytes by inhibiting WEE1 and MYT1, or expressing EGFP-CDC25A or constitutively active EGFP-CDK1 from microinjected complementary RNAs. Forced CDK1 activation in Cdc25b-/- oocytes allowed resumption of meiosis, but oocytes mostly arrested at metaphase I (MI) with intact spindles. Similarly, approximately a third of Cdc25b+/- oocytes with a reduced amount of CDC25B arrested in MI. MI-arrested Cdc25b-/- oocytes also displayed a transient decrease in CDK1 activity similar to Cdc25b+/+ oocytes during the MI-MII transition, whereas Cdc25b+/- oocytes exhibited only a partial anaphase-promoting complex/cyclosome activation and anaphase I entry. Thus, CDC25B is necessary for the resumption of meiosis and the MI-MII transition.


Asunto(s)
Meiosis , Oocitos , Anafase , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Animales , Femenino , Mamíferos , Metafase , Ratones , Oocitos/metabolismo , Fosfatasas cdc25
19.
Annu Rev Genet ; 50: 45-66, 2016 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-27893964

RESUMEN

Chromosomal instability (CIN), the persistent inability of a cell to faithfully segregate its genome, is a feature of many cancer cells. It stands to reason that CIN enables the acquisition of multiple cancer hallmarks; however, there is a growing body of evidence suggesting that CIN impairs cellular fitness and prevents neoplastic transformation. Here, we suggest a new perspective to reconcile this apparent paradox and share an unexpected link between aneuploidy and aging that was discovered through attempts to investigate the CIN-cancer relationship. Additionally, we provide a comprehensive overview of the function and regulation of the anaphase-promoting complex, an E3 ubiquitin ligase that mediates high-fidelity chromosome segregation, and describe the mechanisms that lead to whole-chromosome gain or loss. With this review, we aim to expand our understanding of the role of CIN in cancer and aging with the long-term objective of harnessing this information for the advancement of patient care.


Asunto(s)
Envejecimiento/genética , Aneuploidia , Neoplasias/genética , Ciclosoma-Complejo Promotor de la Anafase/genética , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Cromátides , Inestabilidad Cromosómica , Humanos
20.
Mol Cell ; 64(6): 1144-1153, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27939943

RESUMEN

The Spindle Assembly Checkpoint (SAC) ensures genomic stability by preventing sister chromatid separation until all chromosomes are attached to the spindle. It catalyzes the production of the Mitotic Checkpoint Complex (MCC), which inhibits Cdc20 to inactivate the Anaphase Promoting Complex/Cyclosome (APC/C). Here we show that two Cdc20-binding motifs in BubR1 of the recently identified ABBA motif class are crucial for the MCC to recognize active APC/C-Cdc20. Mutating these motifs eliminates MCC binding to the APC/C, thereby abolishing the SAC and preventing cells from arresting in response to microtubule poisons. These ABBA motifs flank a KEN box to form a cassette that is highly conserved through evolution, both in the arrangement and spacing of the ABBA-KEN-ABBA motifs, and association with the amino-terminal KEN box required to form the MCC. We propose that the ABBA-KEN-ABBA cassette holds the MCC onto the APC/C by binding the two Cdc20 molecules in the MCC-APC/C complex.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/genética , Proteínas Cdc20/genética , Puntos de Control de la Fase M del Ciclo Celular , Proteínas Serina-Treonina Quinasas/genética , Secuencias de Aminoácidos , Ciclosoma-Complejo Promotor de la Anafase/química , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Animales , Arabidopsis/genética , Arabidopsis/metabolismo , Evolución Biológica , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas Cdc20/química , Proteínas Cdc20/metabolismo , Ciona intestinalis/genética , Ciona intestinalis/metabolismo , Secuencia Conservada , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Expresión Génica , Células HeLa , Humanos , Mutación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Imagen de Lapso de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA