Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
1.
Cells ; 10(8)2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34440628

RESUMEN

Tubulin post-translational modifications regulate microtubule properties and functions. Mitotic spindle microtubules are highly modified. While tubulin detyrosination promotes proper mitotic progression by recruiting specific microtubule-associated proteins motors, tubulin acetylation that occurs on specific microtubule subsets during mitosis is less well understood. Here, we show that siRNA-mediated depletion of the tubulin acetyltransferase ATAT1 in epithelial cells leads to a prolonged prometaphase arrest and the formation of monopolar spindles. This results from collapse of bipolar spindles, as previously described in cells deficient for the mitotic kinase PLK1. ATAT1-depleted mitotic cells have defective recruitment of PLK1 to centrosomes, defects in centrosome maturation and thus microtubule nucleation, as well as labile microtubule-kinetochore attachments. Spindle bipolarity could be restored, in the absence of ATAT1, by stabilizing microtubule plus-ends or by increasing PLK1 activity at centrosomes, demonstrating that the phenotype is not just a consequence of lack of K-fiber stability. We propose that microtubule acetylation of K-fibers is required for a recently evidenced cross talk between centrosomes and kinetochores.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centrosoma/enzimología , Células Epiteliales/enzimología , Microtúbulos/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Huso Acromático/enzimología , Acetilación , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Animales , Células LLC-PK1 , Proteínas de Microtúbulos/genética , Proteínas de Microtúbulos/metabolismo , Microtúbulos/genética , Mitosis , Transducción de Señal , Huso Acromático/genética , Porcinos , Quinasa Tipo Polo 1
2.
Sci Rep ; 11(1): 2616, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33510346

RESUMEN

Src-family tyrosine kinases (SFKs) play important roles in a number of signal transduction events during mitosis, such as spindle formation. A relationship has been reported between SFKs and the mitotic spindle; however, the underlying mechanisms remain unclear. We herein demonstrated that SFKs accumulated in the centrosome region at the onset of mitosis. Centrosomal Fyn increased in the G2 phase in a microtubule polymerization-dependent manner. A mass spectrometry analysis using mitotic spindle preparations was performed to identify tyrosine-phosphorylated substrates. Protein regulator of cytokinesis 1 (PRC1) and kinastrin/small kinetochore-associated protein (kinastrin/SKAP) were identified as SFK substrates. SFKs mainly phosphorylated PRC1 at Tyr-464 and kinastrin at Tyr-87. Although wild-type PRC1 is associated with microtubules, phosphomimetic PRC1 impaired the ability to bind microtubules. Phosphomimetic kinastrin at Tyr-87 also impaired binding with microtubules. Collectively, these results suggest that tyrosine phosphorylation of PRC1 and kinastrin plays a role in their delocalization from microtubules during mitosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centrosoma/enzimología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Huso Acromático/enzimología , Ciclo Celular , Células HeLa , Humanos , Fosforilación
3.
J Cell Biol ; 219(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32497213

RESUMEN

Nearly six decades ago, Lewis Wolpert proposed the relaxation of the polar cell cortex by the radial arrays of astral microtubules as a mechanism for cleavage furrow induction. While this mechanism has remained controversial, recent work has provided evidence for polar relaxation by astral microtubules, although its molecular mechanisms remain elusive. Here, using C. elegans embryos, we show that polar relaxation is achieved through dynein-mediated removal of myosin II from the polar cortexes. Mutants that position centrosomes closer to the polar cortex accelerated furrow induction, whereas suppression of dynein activity delayed furrowing. We show that dynein-mediated removal of myosin II from the polar cortexes triggers a bidirectional cortical flow toward the cell equator, which induces the assembly of the actomyosin contractile ring. These results provide a molecular mechanism for the aster-dependent polar relaxation, which works in parallel with equatorial stimulation to promote robust cytokinesis.


Asunto(s)
Actomiosina/metabolismo , Anafase , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Centrosoma/enzimología , Citocinesis , Dineínas/metabolismo , Microtúbulos/enzimología , Miosina Tipo II/metabolismo , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Dineínas/genética , Microtúbulos/genética , Mutación , Miosina Tipo II/genética , Transducción de Señal
4.
J Pathol ; 251(3): 310-322, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32315081

RESUMEN

The phenotypic spectrum of colorectal cancer (CRC) is remarkably diverse, with seemingly endless variations in cell shape, mitotic figures and multicellular configurations. Despite this morphological complexity, histological grading of collective phenotype patterns provides robust prognostic stratification in CRC. Although mechanistic understanding is incomplete, previous studies have shown that the cortical protein ezrin controls diversification of cell shape, mitotic figure geometry and multicellular architecture, in 3D organotypic CRC cultures. Because ezrin is a substrate of Src tyrosine kinase that is frequently overexpressed in CRC, we investigated Src regulation of ezrin and morphogenic growth in 3D CRC cultures. Here we show that Src perturbations disrupt CRC epithelial spatial organisation. Aberrant Src activity suppresses formation of the cortical ezrin cap that anchors interphase centrosomes. In CRC cells with a normal centrosome number, these events lead to mitotic spindle misorientation, perturbation of cell cleavage, abnormal epithelial stratification, apical membrane misalignment, multilumen formation and evolution of cribriform multicellular morphology, a feature of low-grade cancer. In isogenic CRC cells with centrosome amplification, aberrant Src signalling promotes multipolar mitotic spindle formation, pleomorphism and morphological features of high-grade cancer. Translational studies in archival human CRC revealed associations between Src intensity, multipolar mitotic spindle frequency and high-grade cancer morphology. Collectively, our study reveals Src regulation of CRC morphogenic growth via ezrin-centrosome engagement and uncovers combined perturbations underlying transition to high-grade CRC morphology. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Centrosoma/enzimología , Neoplasias Colorrectales/enzimología , Proteínas del Citoesqueleto/metabolismo , Mitosis , Familia-src Quinasas/metabolismo , Células CACO-2 , Centrosoma/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Proteínas del Citoesqueleto/genética , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Células HCT116 , Humanos , Clasificación del Tumor , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Familia-src Quinasas/genética
5.
Cancer Res ; 80(8): 1693-1706, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32054769

RESUMEN

A significant therapeutic challenge for patients with cancer is resistance to chemotherapies such as taxanes. Overexpression of LIN9, a transcriptional regulator of cell-cycle progression, occurs in 65% of patients with triple-negative breast cancer (TNBC), a disease commonly treated with these drugs. Here, we report that LIN9 is further elevated with acquisition of taxane resistance. Inhibiting LIN9 genetically or by suppressing its expression with a global BET inhibitor restored taxane sensitivity by inducing mitotic progression errors and apoptosis. While sustained LIN9 is necessary to maintain taxane resistance, there are no inhibitors that directly repress its function. Hence, we sought to discover a druggable downstream transcriptional target of LIN9. Using a computational approach, we identified NIMA-related kinase 2 (NEK2), a regulator of centrosome separation that is also elevated in taxane-resistant cells. High expression of NEK2 was predictive of low survival rates in patients who had residual disease following treatment with taxanes plus an anthracycline, suggesting a role for this kinase in modulating taxane sensitivity. Like LIN9, genetic or pharmacologic blockade of NEK2 activity in the presence of paclitaxel synergistically induced mitotic abnormalities in nearly 100% of cells and completely restored sensitivity to paclitaxel, in vitro. In addition, suppressing NEK2 activity with two distinct small molecules potentiated taxane response in multiple in vivo models of TNBC, including a patient-derived xenograft, without inducing toxicity. These data demonstrate that the LIN9/NEK2 pathway is a therapeutically targetable mediator of taxane resistance that can be leveraged to improve response to this core chemotherapy. SIGNIFICANCE: Resistance to chemotherapy is a major hurdle for treating patients with cancer. Combining NEK2 inhibitors with taxanes may be a viable approach for improving patient outcomes by enhancing mitotic defects induced by taxanes alone.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Mitosis/efectos de los fármacos , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Paclitaxel/farmacología , Taxoides/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Senescencia Celular , Centrosoma/enzimología , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Xenoinjertos , Humanos , Mitosis/genética , Quinasas Relacionadas con NIMA/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Paclitaxel/administración & dosificación , Tasa de Supervivencia , Taxoides/administración & dosificación , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad , Ensayo de Tumor de Célula Madre , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba
6.
Cytoskeleton (Hoboken) ; 75(11): 481-494, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30414309

RESUMEN

During cell division, duplicated genetic material is separated into two distinct daughter cells. This process is essential for initial tissue formation during development and to maintain tissue integrity throughout an organism's lifetime. To ensure the efficacy and efficiency of this process, the cell employs a variety of regulatory and signaling proteins that function as mitotic regulators and checkpoint proteins. One vital mitotic regulator is polo-like kinase 1 (PLK1), a highly conserved member of the polo-like kinase family. Unique from its paralogues, it functions specifically during mitosis as a regulator of cell division. PLK1 is spatially and temporally enriched at three distinct subcellular locales; the mitotic centrosomes, kinetochores, and the cytokinetic midbody. These localization patterns allow PLK1 to phosphorylate specific downstream targets to regulate mitosis. In this review, we will explore how polo-like kinases were originally discovered and diverged into the five paralogues (PLK1-5) in mammals. We will then focus specifically on the most conserved, PLK1, where we will discuss what is known about how its activity is modulated, its role during the cell cycle, and new, innovative tools that have been developed to examine its function and interactions in cells.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Mitosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Centrosoma/enzimología , Citocinesis/fisiología , Humanos , Cinetocoros/enzimología , Transducción de Señal/fisiología , Quinasa Tipo Polo 1
7.
Mol Cancer Res ; 16(11): 1785-1797, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30018032

RESUMEN

Aurora A kinase (AURKA) is a master cell-cycle regulator that is often dysregulated in human cancers. Its overexpression has been associated with genome instability and oncogenic transformation. The protein kinase D (PKD) family is an emerging therapeutic target of cancer. Aberrant PKD activation has been implicated in tumor growth and survival, yet the underlying mechanisms remain to be elucidated. This study identified, for the first time, a functional crosstalk between PKD2 and Aurora A kinase in cancer cells. The data demonstrate that PKD2 is catalytically active during the G2-M phases of the cell cycle, and inactivation or depletion of PKD2 causes delay in mitotic entry due to downregulation of Aurora A, an effect that can be rescued by overexpression of Aurora A. Moreover, PKD2 localizes in the centrosome with Aurora A by binding to γ-tubulin. Knockdown of PKD2 caused defects in centrosome separation, elongated G2 phase, mitotic catastrophe, and eventually cell death via apoptosis. Mechanistically, PKD2 interferes with Fbxw7 function to protect Aurora A from ubiquitin- and proteasome-dependent degradation. Taken together, these results identify PKD as a cell-cycle checkpoint kinase that positively modulates G2-M transition through Aurora A kinase in mammalian cells.Implications: PKD2 is a novel cell-cycle regulator that promotes G2-M transition by modulating Aurora A kinase stability in cancer cells and suggests the PKD2/Aurora A kinase regulatory axis as new therapeutic targets for cancer treatment. Mol Cancer Res; 16(11); 1785-97. ©2018 AACR.


Asunto(s)
Aurora Quinasa A/metabolismo , Centrosoma/enzimología , Proteínas Quinasas/metabolismo , Ciclo Celular/fisiología , División Celular/fisiología , Centrosoma/metabolismo , Regulación hacia Abajo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Fase G2/fisiología , Células HeLa , Humanos , Células PC-3 , Proteína Quinasa D2 , Ubiquitinación
8.
J Pathol ; 244(4): 445-459, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29520890

RESUMEN

Histological grading provides prognostic stratification of colorectal cancer (CRC) by scoring heterogeneous phenotypes. Features of aggressiveness include aberrant mitotic spindle configurations, chromosomal breakage, and bizarre multicellular morphology, but pathobiology is poorly understood. Protein kinase C zeta (PKCz) controls mitotic spindle dynamics, chromosome segregation, and multicellular patterns, but its role in CRC phenotype evolution remains unclear. Here, we show that PKCz couples genome segregation to multicellular morphology through control of interphase centrosome anchoring. PKCz regulates interdependent processes that control centrosome positioning. Among these, interaction between the cytoskeletal linker protein ezrin and its binding partner NHERF1 promotes the formation of a localized cue for anchoring interphase centrosomes to the cell cortex. Perturbation of these phenomena induced different outcomes in cells with single or extra centrosomes. Defective anchoring of a single centrosome promoted bipolar spindle misorientation, multi-lumen formation, and aberrant epithelial stratification. Collectively, these disturbances induce cribriform multicellular morphology that is typical of some categories of low-grade CRC. By contrast, defective anchoring of extra centrosomes promoted multipolar spindle formation, chromosomal instability (CIN), disruption of glandular morphology, and cell outgrowth across the extracellular matrix interface characteristic of aggressive, high-grade CRC. Because PKCz enhances apical NHERF1 intensity in 3D epithelial cultures, we used an immunohistochemical (IHC) assay of apical NHERF1 intensity as an indirect readout of PKCz activity in translational studies. We show that apical NHERF1 IHC intensity is inversely associated with multipolar spindle frequency and high-grade morphology in formalin-fixed human CRC samples. To conclude, defective PKCz control of interphase centrosome anchoring may underlie distinct categories of mitotic slippage that shape the development of low- or high-grade CRC phenotypes. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Centrosoma/enzimología , Neoplasias Colorrectales/enzimología , Interfase , Proteína Quinasa C/metabolismo , Células CACO-2 , Proliferación Celular , Forma de la Célula , Inestabilidad Cromosómica , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Humanos , Clasificación del Tumor , Fenotipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteína Quinasa C/genética , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo
9.
Sci Rep ; 8(1): 3353, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29463826

RESUMEN

The hexameric AAA ATPase VPS4 facilitates ESCRT III filament disassembly on diverse intracellular membranes. ESCRT III components and VPS4 have been localized to the ciliary transition zone and spindle poles and reported to affect centrosome duplication and spindle pole stability. How the canonical ESCRT pathway could mediate these events is unclear. We studied the association of VPS4 with centrosomes and found that GFP-VPS4 was a dynamic component of both mother and daughter centrioles. A mutant, VPS4EQ, which can't hydrolyze ATP, was less dynamic and accumulated at centrosomes. Centrosome localization of the VPS4EQ mutant, caused reduced γ-tubulin levels at centrosomes and consequently decreased microtubule growth and altered centrosome positioning. In addition, preventing VPS4 ATP hydrolysis nearly eliminated centriolar satellites and paused ciliogensis after formation of the ciliary vesicle. Zebrafish embryos injected with GFP-VPS4EQ mRNA were less viable, exhibited developmental defects and had fewer cilia in Kupffer's vesicle. Surprisingly, ESCRT III proteins seldom localized to centrosomes and their depletion did not lead to these phenotypes. Our data support an ESCRT III-independent function for VPS4 at the centrosome and reveal that this evolutionary conserved AAA ATPase influences diverse centrosome functions and, as a result, global cellular architecture and development.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/análisis , Centrosoma/enzimología , Centrosoma/metabolismo , Cilios/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/análisis , Tubulina (Proteína)/metabolismo , ATPasas de Translocación de Protón Vacuolares/análisis , Células 3T3 , ATPasas Asociadas con Actividades Celulares Diversas/genética , Animales , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , Pez Cebra
10.
J Biol Chem ; 293(8): 2939-2948, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29326161

RESUMEN

Activating transcription factor 5 (ATF5) is a member of the ATF/cAMP response element-binding protein family of transcription factors. ATF5 regulates stress responses and cell survival, proliferation, and differentiation and also plays a role in viral infections, cancer, diabetes, schizophrenia, and the olfactory system. Moreover, it was found to also have a critical cell cycle-dependent structural function at the centrosome. However, the mechanism that controls the localization of ATF5 at the centrosome is unclear. Here we report that ATF5 is small ubiquitin-like modifier (SUMO) 2/3-modified at a conserved SUMO-targeting consensus site in various types of mammalian cells. We found that SUMOylation of ATF5 is elevated in the G1 phase of the cell cycle and diminished in the G2/M phase. ATF5 SUMOylation disrupted the interaction of ATF5 with several centrosomal proteins and dislodged ATF5 from the centrosome at the end of the M phase. Of note, blockade of ATF5 SUMOylation deregulated the centrosome cycle, impeded ATF5 translocation from the centrosome, and caused genomic instability and G2/M arrest in HeLa cells. Our results indicate that ATF5 SUMOylation is an essential mechanism that regulates ATF5 localization and function at the centrosome.


Asunto(s)
Factores de Transcripción Activadores/metabolismo , Centrosoma/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación , Ubiquitinas/metabolismo , Factores de Transcripción Activadores/química , Factores de Transcripción Activadores/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Línea Celular , Centrosoma/enzimología , Secuencia de Consenso , Secuencia Conservada , Eliminación de Gen , Inestabilidad Genómica , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microscopía Fluorescente , Mutagénesis Sitio-Dirigida , Mutación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Transporte de Proteínas , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/antagonistas & inhibidores , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/química , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Ubiquitinas/antagonistas & inhibidores , Ubiquitinas/química , Ubiquitinas/genética
11.
Mol Cells ; 40(12): 925-934, 2017 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-29237113

RESUMEN

The Cdc6 protein is essential for the initiation of chromosomal replication and functions as a licensing factor to maintain chromosome integrity. During the S and G2 phases of the cell cycle, Cdc6 has been found to inhibit the recruitment of pericentriolar material (PCM) proteins to the centrosome and to suppress centrosome over-duplication. In this report, we analyzed the correlation between these two functions of Cdc6 at the centrosome. Cdc6 depletion increased the population of cells showing centrosome over-duplication and premature centrosome separation; Cdc6 expression reversed these changes. Deletion and fusion experiments revealed that the 18 amino acid residues (197-214) of Cdc6, which were fused to the Cdc6-centrosomal localization signal, suppressed centrosome over-duplication and premature centrosome separation. Cdc6 mutant proteins that showed defective ATP binding or hydrolysis did not exhibit a significant difference in suppressing centrosome over-duplication, compared to the wild type protein. In contrast to the Cdc6-mediated inhibition of PCM protein recruitment to the centrosome, the independence of Cdc6 on its ATPase activity for suppressing centrosome over-duplication, along with the difference between the Cdc6 protein regions participating in the two functions, suggested that Cdc6 controls centrosome duplication in a manner independent of its recruitment of PCM proteins to the centrosome.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrosoma/fisiología , Replicación del ADN/fisiología , Proteínas Nucleares/metabolismo , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/deficiencia , Línea Celular Tumoral , Centriolos/metabolismo , Centrosoma/enzimología , Centrosoma/metabolismo , Humanos , Proteínas Nucleares/deficiencia , Transfección
12.
Sci Rep ; 7(1): 15536, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-29138491

RESUMEN

Polo-like kinase 1 (Plk1) is a crucial cell cycle regulator by its specific localization and activity during cell cycle. It has been shown that the phosphorylation and ubiquitylation of Plk1 are required for its own activation and localization. Here, we report that SUMOylation regulates the activity of Plk1 in the lepidopteran insect of Bombyx mori. In the absence of SUMOylation, it causes the lost localization of Plk1 on centrosomes and kinetochores, as well as an uneven distribution in midzone. We further identify that the putative SUMOylation site of Bombyx Plk1 at lysine 466 is required for its localization on centrosomes, and K466 mutation in Plk1 could influence its interaction with Smt3/Ubc9 complex. These findings are also confirmed by Drosophila Polo and human Plk1, which together reveals a conserved role of Plk1 SUMOylation in mammals. Moreover, conjugation of Smt3 to Plk1 SUMOylation mutant promotes its localization on centrosomes and kinetochores, and rescues functional defects of chromosome alignment in cells depleted of endogenous Plk1. Altogether, the present data indicate that the SUMOylation of Plk1 could participate in proper chromosome alignment and segregation during mitosis, and provides a novel layer for the regulation of Plk1 localization and activity throughout cell cycle.


Asunto(s)
Bombyx/citología , Bombyx/enzimología , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Proteínas de Insectos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Sumoilación , Animales , Bombyx/genética , Proteínas de Ciclo Celular/genética , Centrosoma/enzimología , Segregación Cromosómica , Drosophila/metabolismo , Cinetocoros/enzimología , Mitosis , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Quinasa Tipo Polo 1
13.
J Biol Chem ; 292(31): 12874-12884, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28620049

RESUMEN

The X-linked deubiquitinase USP9X has been implicated in multiple pathological disorders including malignancies and X-linked intellectual disability. However, its biological function and substrate repertoire remain to be investigated. In this study, we utilized the tandem mass tag labeling assay to identify USP9X-regulated proteins and revealed that the expression of multiple genes is altered in USP9X-deficient cells. Interestingly, we showed that USP9X promotes stabilization of centrosome proteins PCM1 and CEP55 through its catalytic activity. Remarkably, we demonstrated that USP9X is physically associated and spatially co-localized with PCM1 and CEP55 in the centrosome, and we revealed that either PCM1 or CEP55 loss resulted in impairment of USP9X centrosome localization. Moreover, we showed that USP9X is required for centrosome duplication, and this effect is dependent on its catalytic activity and its N-terminal module, which is responsible for physical association of USP9X with PCM1 and CEP55. Collectively, our experiments identified USP9X as an integral component of the centrosome where it functions to stabilize PCM1 and CEP55 and promote centrosome biogenesis.


Asunto(s)
Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrosoma/enzimología , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Sustitución de Aminoácidos , Autoantígenos/química , Autoantígenos/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Centrosoma/metabolismo , Eliminación de Gen , Humanos , Inmunoprecipitación , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/genética , Oligopéptidos/genética , Oligopéptidos/metabolismo , Biogénesis de Organelos , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Transporte de Proteínas , Proteómica/métodos , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/genética
14.
Mol Biol Cell ; 28(8): 1011-1020, 2017 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-28228549

RESUMEN

The budding yeast Polo-like kinase Cdc5 is a key regulator of many mitotic events. Cdc5 coordinates its functions spatially and temporally by changing its localization during the cell cycle: Cdc5 is imported into the nucleus in G2 phase and released to the cytoplasm in anaphase, where it accumulates at the bud neck. Cdc5 also localizes to the spindle pole bodies (SPBs) from S phase until the end of mitosis. Whether Cdc5 changes its SPB population during the cell cycle is not known. We find that Cdc5 localizes to distinct SPB subpopulations, depending on the mitotic stage. Cdc5 localizes to the nuclear side of the SPBs during metaphase and early anaphase and to the cytoplasmic surface of the SPBs during late anaphase. Cdc14 is necessary to relocalize Cdc5 from the nuclear SPB plaque. Accumulation of Cdc5 at the daughter SPB in late anaphase is controlled by Bfa1. We also show that Cdc5 and Bfa1 are found in spatially distinct locations at the SPBs during G2/M arrest after DNA damage. Collectively our data reveal that Cdc5 is a dynamic component of the SPBs during mitosis and provide new insight into its regulation during both late mitotic events and DNA damage-induced G2/M arrest.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centrosoma/enzimología , Mitosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Saccharomycetales/citología , Saccharomycetales/enzimología , Cuerpos Polares del Huso/metabolismo , Ciclo Celular/fisiología , Núcleo Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomycetales/genética , Huso Acromático/metabolismo
15.
Elife ; 52016 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-27885983

RESUMEN

Asymmetric stem cell division is often accompanied by stereotypical inheritance of the mother and daughter centrosomes. However, it remains unknown whether and how stem cell centrosomes are uniquely regulated and how this regulation may contribute to stem cell fate. Here we identify Klp10A, a microtubule-depolymerizing kinesin of the kinesin-13 family, as the first protein enriched in the stem cell centrosome in Drosophila male germline stem cells (GSCs). Depletion of klp10A results in abnormal elongation of the mother centrosomes in GSCs, suggesting the existence of a stem cell-specific centrosome regulation program. Concomitant with mother centrosome elongation, GSCs form asymmetric spindle, wherein the elongated mother centrosome organizes considerably larger half spindle than the other. This leads to asymmetric cell size, yielding a smaller differentiating daughter cell. We propose that klp10A functions to counteract undesirable asymmetries that may result as a by-product of achieving asymmetries essential for successful stem cell divisions.


Asunto(s)
División Celular , Centrosoma/enzimología , Proteínas de Drosophila/metabolismo , Células Germinativas/fisiología , Cinesinas/metabolismo , Células Madre/fisiología , Animales , Drosophila , Masculino
16.
Sci Rep ; 6: 27401, 2016 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-27264542

RESUMEN

Dynamic phosphorylation of Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7 heptad-repeats in the C-terminal domain (CTD) of the large subunit coordinates progression of RNA polymerase (Pol) II through the transcription cycle. Here, we describe an M phase-specific form of Pol II phosphorylated at Thr4, but not at Tyr1, Ser2, Ser5, and Ser7 residues. Thr4 phosphorylated Pol II binds to centrosomes and midbody and interacts with the Thr4-specific Polo-like kinase 1. Binding of Pol II to centrosomes does not require the CTD but may involve subunits of the non-canonical R2TP-Prefoldin-like complex, which bind to and co-localize with Pol II at centrosomes. CTD Thr4 mutants, but not Ser2 and Ser5 mutants, display severe mitosis and cytokinesis defects characterized by multipolar spindles and polyploid cells. We conclude that proper M phase progression of cells requires binding of Pol II to centrosomes to facilitate regulation of mitosis and cytokinesis in a CTD Thr4-P dependent manner.


Asunto(s)
División Celular , ARN Polimerasa II/metabolismo , Treonina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Centrosoma/enzimología , Humanos , Peso Molecular , Mutación , Fosforilación , Dominios Proteicos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN Polimerasa II/química , Treonina/genética , Quinasa Tipo Polo 1
17.
Nat Commun ; 6: 10072, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26656453

RESUMEN

TANK Binding Kinase 1 (TBK1) is a non-canonical IκB kinase that contributes to KRAS-driven lung cancer. Here we report that TBK1 plays essential roles in mammalian cell division. Specifically, levels of active phospho-TBK1 increase during mitosis and localize to centrosomes, mitotic spindles and midbody, and selective inhibition or silencing of TBK1 triggers defects in spindle assembly and prevents mitotic progression. TBK1 binds to the centrosomal protein CEP170 and to the mitotic apparatus protein NuMA, and both CEP170 and NuMA are TBK1 substrates. Further, TBK1 is necessary for CEP170 centrosomal localization and binding to the microtubule depolymerase Kif2b, and for NuMA binding to dynein. Finally, selective disruption of the TBK1-CEP170 complex augments microtubule stability and triggers defects in mitosis, suggesting that TBK1 functions as a mitotic kinase necessary for microtubule dynamics and mitosis.


Asunto(s)
Centrosoma/enzimología , Microtúbulos/fisiología , Mitosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Línea Celular , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas
18.
Mol Biol Cell ; 26(23): 4187-96, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26378257

RESUMEN

In many animals, female meiotic spindles are assembled in the absence of centrosomes, the major microtubule (MT)-organizing centers. How MTs are formed and organized into meiotic spindles is poorly understood. Here we report that, in Caenorhabditis elegans, Aurora A kinase/AIR-1 is required for the formation of spindle microtubules during female meiosis. When AIR-1 was depleted or its kinase activity was inhibited in C. elegans oocytes, although MTs were formed around chromosomes at germinal vesicle breakdown (GVBD), they were decreased during meiotic prometaphase and failed to form a bipolar spindle, and chromosomes were not separated into two masses. Whereas AIR-1 protein was detected on and around meiotic spindles, its kinase-active form was concentrated on chromosomes at prometaphase and on interchromosomal MTs during late anaphase and telophase. We also found that AIR-1 is involved in the assembly of short, dynamic MTs in the meiotic cytoplasm, and these short MTs were actively incorporated into meiotic spindles. Collectively our results suggest that, after GVBD, the kinase activity of AIR-1 is continuously required for the assembly and/or stabilization of female meiotic spindle MTs.


Asunto(s)
Aurora Quinasa A/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Meiosis/fisiología , Huso Acromático/enzimología , Animales , Caenorhabditis elegans/citología , Técnicas de Cultivo de Célula , Centrosoma/enzimología , Femenino , Microtúbulos/metabolismo , Oocitos/enzimología
20.
J Cell Biol ; 210(1): 45-62, 2015 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-26124292

RESUMEN

Coordination of cell growth and proliferation in response to nutrient supply is mediated by mammalian target of rapamycin (mTOR) signaling. In this study, we report that Mio, a highly conserved member of the SEACAT/GATOR2 complex necessary for the activation of mTORC1 kinase, plays a critical role in mitotic spindle formation and subsequent chromosome segregation by regulating the proper concentration of active key mitotic kinases Plk1 and Aurora A at centrosomes and spindle poles. Mio-depleted cells showed reduced activation of Plk1 and Aurora A kinase at spindle poles and an impaired localization of MCAK and HURP, two key regulators of mitotic spindle formation and known substrates of Aurora A kinase, resulting in spindle assembly and cytokinesis defects. Our results indicate that a major function of Mio in mitosis is to regulate the activation/deactivation of Plk1 and Aurora A, possibly by linking them to mTOR signaling in a pathway to promote faithful mitotic progression.


Asunto(s)
Aurora Quinasa A/fisiología , Proteínas de Ciclo Celular/metabolismo , Centrosoma/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Secuencia de Aminoácidos , Activación Enzimática , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Cinesinas/metabolismo , Mitosis , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Huso Acromático/metabolismo , Quinasa Tipo Polo 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...