Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Cancer Res ; 78(21): 6282-6296, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30166419

RESUMEN

The targeting of telomerase and telomere maintenance mechanisms represents a promising therapeutic approach for various types of cancer. In this work, we designed a new protocol to screen for and rank the efficacy of compounds specifically targeting telomeres and telomerase. This approach used two isogenic cell lines containing a circular human artificial chromosome (HAC, lacking telomeres) and a linear HAC (containing telomeres) marked with the EGFP transgene; compounds that target telomerase or telomeres should preferentially induce loss of the linear HAC but not the circular HAC. Our assay allowed quantification of chromosome loss by routine flow cytometry. We applied this dual-HAC assay to rank a set of known and newly developed compounds, including G-quadruplex (G4) ligands. Among the latter group, two compounds, Cu-ttpy and Pt-ttpy, induced a high rate of linear HAC loss with no significant effect on the mitotic stability of a circular HAC. Analysis of the mitotic phenotypes induced by these drugs revealed an elevated rate of chromatin bridges in late mitosis and cytokinesis as well as UFB (ultrafine bridges). Chromosome loss after Pt-ttpy or Cu-ttpy treatment correlated with the induction of telomere-associated DNA damage. Overall, this platform enables identification and ranking of compounds that greatly increase chromosome mis-segregation rates as a result of telomere dysfunction and may expedite the development of new therapeutic strategies for cancer treatment.Significance: An assay provides a unique opportunity to screen thousands of chemical compounds for their ability to inactivate replication of telomeric ends in cancer cells and holds potential to lay the foundation for the discovery of new treatments for cancer. Cancer Res; 78(21); 6282-96. ©2018 AACR.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Telomerasa/antagonistas & inhibidores , Telómero/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Cromatina , Cromosomas , Cromosomas Artificiales Humanos , Daño del ADN , Diseño de Fármacos , Células HCT116 , Humanos , Ácidos Hidroxámicos/farmacología , Mitosis , Neoplasias/genética , Transgenes
3.
Nat Commun ; 9(1): 2071, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29789663

RESUMEN

The original PDF version of this Article listed the authors as "Marcus J.G.W. Ladds," where it should have read "Marcus J. G. W. Ladds, Ingeborg M. M. van Leeuwen, Catherine J. Drummond et al.#".Also in the PDF version, it was incorrectly stated that "Correspondence and requests for materials should be addressed to S. Lín.", instead of the correct "Correspondence and requests for materials should be addressed to S. Laín."This has been corrected in the PDF version of the Article. The HTML version was correct from the time of publication.

4.
Nat Commun ; 9(1): 1107, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29549331

RESUMEN

The development of non-genotoxic therapies that activate wild-type p53 in tumors is of great interest since the discovery of p53 as a tumor suppressor. Here we report the identification of over 100 small-molecules activating p53 in cells. We elucidate the mechanism of action of a chiral tetrahydroindazole (HZ00), and through target deconvolution, we deduce that its active enantiomer (R)-HZ00, inhibits dihydroorotate dehydrogenase (DHODH). The chiral specificity of HZ05, a more potent analog, is revealed by the crystal structure of the (R)-HZ05/DHODH complex. Twelve other DHODH inhibitor chemotypes are detailed among the p53 activators, which identifies DHODH as a frequent target for structurally diverse compounds. We observe that HZ compounds accumulate cancer cells in S-phase, increase p53 synthesis, and synergize with an inhibitor of p53 degradation to reduce tumor growth in vivo. We, therefore, propose a strategy to promote cancer cell killing by p53 instead of its reversible cell cycle arresting effect.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Indazoles/farmacología , Neoplasias/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dihidroorotato Deshidrogenasa , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/química , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Proteolisis/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética
6.
Chromosome Res ; 24(4): 495-509, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27924426

RESUMEN

It is widely accepted that heterochromatin is necessary to maintain genomic stability. However, direct experimental evidence supporting this is slim. Previous studies using either enzyme inhibitors, gene knockout or knockdown studies all are subject to the caveat that drugs may have off-target effects and enzymes that modify chromatin proteins to support heterochromatin formation may also have numerous other cellular targets as well. Here, we describe PREditOR (protein reading and editing of residues), a synthetic biology approach that allows us to directly remove heterochromatin from cells without either drugs or global interference with gene function. We find that removal of heterochromatin perturbs mitotic progression and causes a dramatic increase in chromosome segregation defects, possibly as a result of interfering with the normal centromeric localization of the chromosomal passenger complex.


Asunto(s)
Heterocromatina/fisiología , Biología Sintética/métodos , Línea Celular , Centrómero/metabolismo , Segregación Cromosómica , Inestabilidad Genómica , Humanos , Mitosis
7.
Front Cell Dev Biol ; 3: 73, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26636082

RESUMEN

Aurora kinases comprise a family of highly conserved serine-threonine protein kinases that play a pivotal role in the regulation of cell cycle. Aurora kinases are not only involved in the control of multiple processes during cell division but also coordinate chromosomal and cytoskeletal events, contributing to the regulation of checkpoints and ensuring the smooth progression of the cell cycle. Because of their fundamental contribution to cell cycle regulation, Aurora kinases were originally identified in independent genetic screens designed to find genes involved in the regulation of cell division. The first aurora mutant was part of a collection of mutants isolated in C. Nusslein-Volhard's laboratory. This collection was screened in D. M. Glover's laboratory in search for mutations disrupting the centrosome cycle in embryos derived from homozygous mutant mothers. The mutants identified were given names related to the "polar regions," and included not only aurora but also the equally famous polo. Ipl1, the only Aurora in yeast, was identified in a genetic screen looking for mutations that caused chromosome segregation defects. The discovery of a second Aurora-like kinase in mammals opened a new chapter in the research of Aurora kinases. The rat kinase AIM was found to be highly homologous to the fly and yeast proteins, but localized at the midzone and midbody and was proposed to have a role in cytokinesis. Homologs of the equatorial Aurora (Aurora B) were identified in metazoans ranging from flies to humans. Xenopus Aurora B was found to be in a complex with the chromosomal passenger INCENP, and both proteins were shown to be essential in flies for chromosome structure, segregation, central spindle formation and cytokinesis. Fifteen years on, Aurora kinase research is an active field of research. After the successful introduction of the first anti-mitotic agents in cancer therapy, both Auroras have become the focus of attention as targets for the development of new anti-cancer drugs. In this review we will aim to give a historical overview of the research on Aurora kinases, highlighting the most relevant milestones in the advance of the field.

8.
Open Biol ; 4(11): 140162, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25376909

RESUMEN

Cell cycle progression is regulated by members of the cyclin-dependent kinase (CDK), Polo and Aurora families of protein kinases. The levels of expression and localization of the key regulatory kinases are themselves subject to very tight control. There is increasing evidence that crosstalk between the mitotic kinases provides for an additional level of regulation. We have previously shown that Aurora B activates Polo kinase at the centromere in mitosis, and that the interaction between Polo and the chromosomal passenger complex (CPC) component INCENP is essential in this activation. In this report, we show that Polo kinase is required for the correct localization and activity of the CPC in meiosis and mitosis. Study of the phenotype of different polo allele combinations compared to the effect of chemical inhibition revealed significant differences in the localization and activity of the CPC in diploid tissues. Our results shed new light on the mechanisms that control the activity of Aurora B in meiosis and mitosis.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Meiosis , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Aurora Quinasa B/metabolismo , Dominio Catalítico , Diploidia , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Datos de Secuencia Molecular , Mutación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas
9.
J Cell Biol ; 199(5): 719-21, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23185028

RESUMEN

Before the final step of cytokinesis, termed abscission, dividing cells need to ensure that the cleavage plane is clear of chromatin. In this issue, Kotadia et al. (2012. J. Cell Biol. http://dx.doi.org/jcb.201208041) show that in Drosophila melanogaster, larval neuroblasts elongate to allow segregation of extra-long chromatids and clearance of the midzone, thereby avoiding cytokinesis failure and aneuploidy.


Asunto(s)
Adaptación Fisiológica , Forma de la Célula/fisiología , Cromátides/metabolismo , Segregación Cromosómica , Citocinesis , Drosophila melanogaster/citología , Células-Madre Neurales/citología , Animales
10.
Nat Rev Mol Cell Biol ; 13(12): 789-803, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23175282

RESUMEN

Successful cell division requires the precise and timely coordination of chromosomal, cytoskeletal and membrane trafficking events. These processes are regulated by the competing actions of protein kinases and phosphatases. Aurora B is one of the most intensively studied kinases. In conjunction with inner centromere protein (INCENP), borealin (also known as Dasra) and survivin it forms the chromosomal passenger complex (CPC). This complex targets to different locations at differing times during mitosis, where it regulates key mitotic events: correction of chromosome-microtubule attachment errors; activation of the spindle assembly checkpoint; and construction and regulation of the contractile apparatus that drives cytokinesis. Our growing understanding of the CPC has seen it develop from a mere passenger riding on the chromosomes to one of the main controllers of mitosis.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Mitosis , Animales , Aurora Quinasa B , Aurora Quinasas , Proteínas de Ciclo Celular/fisiología , Centrómero/fisiología , Drosophila melanogaster/fisiología , Humanos , Ratones , Microtúbulos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Huso Acromático/fisiología
11.
Open Biol ; 2(7): 120095, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22870391

RESUMEN

At the end of cell division, the cytoplasmic bridge joining the daughter cells is severed through a process that involves scission of the plasma membrane. The presence of chromatin bridges 'stuck' in the division plane is sensed by the chromosomal passenger complex (CPC) component Aurora B kinase, triggering a checkpoint that delays abscission until the chromatin bridges have been resolved. Recent work has started to shed some light on the molecular mechanism by which the CPC controls the timing of abscission.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Cromatina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Aurora Quinasa B , Aurora Quinasas , Membrana Celular/genética , Membrana Celular/metabolismo , Cromatina/genética , Células HeLa , Humanos , Proteínas Serina-Treonina Quinasas/genética
12.
Cell Cycle ; 11(8): 1490-5, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22433949

RESUMEN

The events of cell division are regulated by a complex interplay between kinases and phosphatases. Cyclin-dependent kinases (Cdks), polo-like kinases (Plks) and Aurora kinases play central roles in this process. Polo kinase (Plk1 in humans) regulates a wide range of events in mitosis and cytokinesis. To ensure the accuracy of these processes, polo activity itself is subject to complex regulation. Phosphorylation of polo in its T loop (or activation loop) increases its kinase activity several-fold. It has been shown that Aurora A kinase, with its co-factor Bora, activates Plk1 in G(2), and that this is essential for recovery from cell cycle arrest induced by DNA damage. In a recent article published in PLoS Biology, we report that Drosophila polo is activated by Aurora B kinase at centromeres, and that this is crucial for polo function in regulating chromosome dynamics in prometaphase. Our results suggest that this regulatory pathway is conserved in humans. Here, we propose a model for the collaboration between Aurora B and polo in the regulation of kinetochore attachment to microtubules in early mitosis. Moreover, we suggest that Aurora B could also function to activate Polo/Plk1 in cytokinesis. Finally, we discuss recent findings and open questions regarding the activation of polo and polo-like kinases by different kinases in mitosis, cytokinesis and other processes.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Aurora Quinasa B , Aurora Quinasas , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/química , Citocinesis , Daño del ADN , Humanos , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Mitosis , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/química , Quinasa Tipo Polo 1
13.
Cell Cycle ; 11(7): 1269, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22426423
14.
PLoS Biol ; 10(1): e1001250, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22291575

RESUMEN

The coordinated activities at centromeres of two key cell cycle kinases, Polo and Aurora B, are critical for ensuring that the two sister kinetochores of each chromosome are attached to microtubules from opposite spindle poles prior to chromosome segregation at anaphase. Initial attachments of chromosomes to the spindle involve random interactions between kinetochores and dynamic microtubules, and errors occur frequently during early stages of the process. The balance between microtubule binding and error correction (e.g., release of bound microtubules) requires the activities of Polo and Aurora B kinases, with Polo promoting stable attachments and Aurora B promoting detachment. Our study concerns the coordination of the activities of these two kinases in vivo. We show that INCENP, a key scaffolding subunit of the chromosomal passenger complex (CPC), which consists of Aurora B kinase, INCENP, Survivin, and Borealin/Dasra B, also interacts with Polo kinase in Drosophila cells. It was known that Aurora A/Bora activates Polo at centrosomes during late G2. However, the kinase that activates Polo on chromosomes for its critical functions at kinetochores was not known. We show here that Aurora B kinase phosphorylates Polo on its activation loop at the centromere in early mitosis. This phosphorylation requires both INCENP and Aurora B activity (but not Aurora A activity) and is critical for Polo function at kinetochores. Our results demonstrate clearly that Polo kinase is regulated differently at centrosomes and centromeres and suggest that INCENP acts as a platform for kinase crosstalk at the centromere. This crosstalk may enable Polo and Aurora B to achieve a balance wherein microtubule mis-attachments are corrected, but proper attachments are stabilized allowing proper chromosome segregation.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/metabolismo , Cinetocoros/enzimología , Proteínas Serina-Treonina Quinasas/genética , Animales , Aurora Quinasa B , Aurora Quinasas , Técnicas de Cultivo de Célula , Proteínas Cromosómicas no Histona/genética , Segregación Cromosómica/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Células HeLa , Humanos , Microtúbulos/metabolismo , Mitosis/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Huso Acromático/genética , Huso Acromático/metabolismo
15.
J Cell Sci ; 123(Pt 16): 2823-33, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20663916

RESUMEN

Aurora kinases are central regulators of mitotic-spindle assembly, chromosome segregation and cytokinesis. Aurora B is a member of the chromosomal passenger complex (CPC) with crucial functions in regulation of the attachment of kinetochores to microtubules and in cytokinesis. We report here that Aurora B contains a conserved SUMO modification motif within its kinase domain. Aurora B can bind SUMO peptides in vitro when bound to the IN-box domain of its CPC partner INCENP. Mutation of Lys207 to arginine (Aurora B(K207R)) impairs the formation of conjugates of Aurora B and SUMO in vivo. Expression of the SUMO-null form of Aurora B results in abnormal chromosome segregation and cytokinesis failure and it is not able to rescue mitotic defects in Aurora-B-knockout cells. These defects are accompanied by increased levels of the CPC on chromosome arms and defective centromeric function, as detected by decreased phosphorylation of the Aurora-B substrate CENP-A. The Aurora-B(K207R) mutant does not display reduced kinase activity, suggesting that functional defects are probably a consequence of the altered localization, rather than decreased intrinsic kinase activity. These data suggest that SUMOylation of Aurora B modulates its function, possibly by mediating the extraction of CPC complexes from chromosome arms during prometaphase.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteína SUMO-1/metabolismo , Animales , Aurora Quinasa B , Aurora Quinasas , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica , Citocinesis/genética , Células HeLa , Humanos , Ratones , Mutación , Proteínas Serina-Treonina Quinasas/genética , Huso Acromático/genética , Huso Acromático/metabolismo , Sumoilación , Transfección
16.
Curr Opin Cell Biol ; 21(6): 796-805, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19836940

RESUMEN

The conserved Aurora family of protein kinases have emerged as crucial regulators of mitosis and cytokinesis. Despite their high degree of homology, Aurora A and B have very distinctive localisations and functions: Aurora A associates with the spindle poles to regulate entry into mitosis, centrosome maturation and spindle assembly; Aurora B is a member of the Chromosomal Passenger Complex (CPC) that transfers from the inner centromere in early mitosis to the spindle midzone, equatorial cortex and midbody in late mitosis and cytokinesis. Aurora B functions include regulation of chromosome-microtubule interactions, cohesion, spindle stability and cytokinesis. This review will focus on how interacting proteins make this functional diversity possible by targeting the kinases to different subcellular locations and regulating their activity.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Aurora Quinasa B , Aurora Quinasas , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Cromosomas/metabolismo , Humanos , Microtúbulos/metabolismo , Mitosis , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/genética , Intercambio de Cromátides Hermanas/genética , Huso Acromático/metabolismo
18.
Development ; 135(19): 3239-46, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18755775

RESUMEN

Spindle formation in female meiosis differs from mitosis in many animals, as it takes place independently of centrosomes, and the molecular requirements of this pathway remain to be understood. Here, we report two crucial roles of Incenp, an essential subunit of the chromosomal passenger complex (the Aurora B complex), in centrosome-independent spindle formation in Drosophila female meiosis. First, the initial assembly of spindle microtubules is drastically delayed in an incenp mutant. This clearly demonstrates, for the first time, a crucial role for Incenp in chromosome-driven spindle microtubule assembly in living oocytes. Additionally, Incenp is necessary to stabilise the equatorial region of the metaphase I spindle, in contrast to mitosis, where the equivalent function becomes prominent after anaphase onset. Our analysis suggests that Subito, a kinesin-6 protein, cooperates with Incenp for this latter function, but not in microtubule assembly. We propose that the two functions of Incenp are part of the mechanisms that compensate for the lack of centrosomes during meiotic spindle formation.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Proteínas de Drosophila/fisiología , Drosophila/citología , Drosophila/fisiología , Meiosis/fisiología , Animales , Animales Modificados Genéticamente , Centrosoma/fisiología , Proteínas Cromosómicas no Histona/genética , Drosophila/genética , Proteínas de Drosophila/genética , Femenino , Genes de Insecto , Cinesinas/fisiología , Meiosis/genética , Metafase/genética , Metafase/fisiología , Microtúbulos/fisiología , Mutación
19.
Biochem Soc Trans ; 36(Pt 3): 367-70, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18481960

RESUMEN

The CPC (chromosomal passenger complex) performs essential roles in the regulation and co-ordination of chromosomal and cytoskeletal events during mitosis and meiosis. The first functional analyses showed evidence of a role of the CPC in the regulation of cytokinesis. In this review, I summarize what we have learned since then about the role of the CPC in the late stages of mitosis and cytokinesis.


Asunto(s)
Cromosomas/metabolismo , Citocinesis , Actomiosina/metabolismo , Animales , Proteínas del Citoesqueleto/metabolismo , Humanos , Filamentos Intermedios/metabolismo , Huso Acromático/metabolismo
20.
Cell ; 131(2): 230-1, 2007 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17956723
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA