Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 607(7919): 604-609, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35831506

RESUMEN

Chromosome segregation errors during cell divisions generate aneuploidies and micronuclei, which can undergo extensive chromosomal rearrangements such as chromothripsis1-5. Selective pressures then shape distinct aneuploidy and rearrangement patterns-for example, in cancer6,7-but it is unknown whether initial biases in segregation errors and micronucleation exist for particular chromosomes. Using single-cell DNA sequencing8 after an error-prone mitosis in untransformed, diploid cell lines and organoids, we show that chromosomes have different segregation error frequencies that result in non-random aneuploidy landscapes. Isolation and sequencing of single micronuclei from these cells showed that mis-segregating chromosomes frequently also preferentially become entrapped in micronuclei. A similar bias was found in naturally occurring micronuclei of two cancer cell lines. We find that segregation error frequencies of individual chromosomes correlate with their location in the interphase nucleus, and show that this is highest for peripheral chromosomes behind spindle poles. Randomization of chromosome positions, Cas9-mediated live tracking and forced repositioning of individual chromosomes showed that a greater distance from the nuclear centre directly increases the propensity to mis-segregate. Accordingly, chromothripsis in cancer genomes9 and aneuploidies in early development10 occur more frequently for larger chromosomes, which are preferentially located near the nuclear periphery. Our findings reveal a direct link between nuclear chromosome positions, segregation error frequencies and micronucleus content, with implications for our understanding of tumour genome evolution and the origins of specific aneuploidies during development.


Asunto(s)
Aneuploidia , Posicionamiento de Cromosoma , Segregación Cromosómica , Cromosomas , Proteína 9 Asociada a CRISPR , Línea Celular , Línea Celular Tumoral , Segregación Cromosómica/genética , Cromosomas/genética , Cromosomas/metabolismo , Cromotripsis , Crecimiento y Desarrollo/genética , Humanos , Interfase , Micronúcleos con Defecto Cromosómico , Mitosis , Neoplasias/genética , Neoplasias/patología , Organoides/citología , Organoides/metabolismo , Análisis de Secuencia de ADN , Análisis de la Célula Individual
2.
Chromosome Res ; 31(3): 25, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37640903

RESUMEN

An abnormal chromosome number, or aneuploidy, underlies developmental disorders and is a common feature of cancer, with different cancer types exhibiting distinct patterns of chromosomal gains and losses. To understand how specific aneuploidies emerge in certain tissues and how they contribute to disease development, various methods have been developed to alter the karyotype of mammalian cells and mice. In this review, we provide an overview of both classic and novel strategies for inducing or selecting specific chromosomal gains and losses in human and murine cell systems. We highlight how these customized aneuploidy models helped expanding our knowledge of the consequences of specific aneuploidies to (cancer) cell physiology.


Asunto(s)
Aneuploidia , Aberraciones Cromosómicas , Humanos , Animales , Ratones , Cariotipificación , Cariotipo , Mamíferos
3.
EMBO Rep ; 22(7): e52295, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33973335

RESUMEN

The shugoshin proteins are universal protectors of centromeric cohesin during mitosis and meiosis. The binding of human hSgo1 to the PP2A-B56 phosphatase through a coiled-coil (CC) region mediates cohesion protection during mitosis. Here we undertook a structure function analysis of the PP2A-B56-hSgo1 complex, revealing unanticipated aspects of complex formation and function. We establish that a highly conserved pocket on the B56 regulatory subunit is required for hSgo1 binding and cohesion protection during mitosis in human somatic cells. Consistent with this, we show that hSgo1 blocks the binding of PP2A-B56 substrates containing a canonical B56 binding motif. We find that PP2A-B56 bound to hSgo1 dephosphorylates Cdk1 sites on hSgo1 itself to modulate cohesin interactions. Collectively our work provides important insight into cohesion protection during mitosis.


Asunto(s)
Proteínas de Ciclo Celular , Proteína Fosfatasa 2 , Proteína Quinasa CDC2 , Proteínas de Ciclo Celular/genética , Centrómero , Humanos , Meiosis , Mitosis , Proteína Fosfatasa 2/genética
4.
Cell ; 132(2): 233-46, 2008 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-18243099

RESUMEN

Maintenance of chromosomal stability relies on coordination between various processes that are critical for proper chromosome segregation in mitosis. Here we show that monopolar spindle 1 (Mps1) kinase, which is essential for the mitotic checkpoint, also controls correction of improper chromosome attachments. We report that Borealin/DasraB, a member of the complex that regulates the Aurora B kinase, is directly phosphorylated by Mps1 on residues that are crucial for Aurora B activity and chromosome alignment. As a result, cells lacking Mps1 kinase activity fail to efficiently align chromosomes due to impaired Aurora B function at centromeres, leaving improper attachments uncorrected. Strikingly, Borealin/DasraB bearing phosphomimetic mutations restores Aurora B activity and alignment in Mps1-depleted cells. Mps1 thus coordinates attachment error correction and checkpoint signaling, two crucial responses to unproductive chromosome attachments.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Cromosomas Humanos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Alelos , Aurora Quinasa B , Aurora Quinasas , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Activación Enzimática , Células HeLa , Humanos , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Mutación , Fosforilación , Plásmidos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Huso Acromático/metabolismo , Transfección
5.
Mol Cell ; 57(5): 824-835, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25661489

RESUMEN

Regulated recruitment of the kinase-adaptor complex BUB1/BUB3 to kinetochores is crucial for correcting faulty chromosome-spindle attachments and for spindle assembly checkpoint (SAC) signaling. BUB1/BUB3 localizes to kinetochores by binding phosphorylated MELT motifs (MELpT) in the kinetochore scaffold KNL1. Human KNL1 has 19 repeats that contain a MELT-like sequence. The repeats are, however, larger than MELT, and repeat sequences can vary significantly. Using systematic screening, we show that only a limited number of repeats is "active." Repeat activity correlates with the presence of a vertebrate-specific SHT motif C-terminal to the MELT sequence. SHT motifs are phosphorylated by MPS1 in a manner that requires prior phosphorylation of MELT. Phospho-SHT (SHpT) synergizes with MELpT in BUB3/BUB1 binding in vitro and in cells, and human BUB3 mutated in a predicted SHpT-binding surface cannot localize to kinetochores. Our data show sequential multisite regulation of the KNL1-BUB1/BUB3 interaction and provide mechanistic insight into evolution of the KNL1-BUB3 interface.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Células HeLa , Humanos , Immunoblotting , Cinetocoros/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/genética , Proteínas Asociadas a Microtúbulos/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Nocodazol/farmacología , Fosforilación , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Interferencia de ARN , Secuencias Repetitivas de Aminoácido/genética , Homología de Secuencia de Aminoácido , Imagen de Lapso de Tiempo , Moduladores de Tubulina/farmacología
6.
Chromosoma ; 123(1-2): 25-42, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24091645

RESUMEN

The ultimate goal of cell division is equal transmission of the duplicated genome to two new daughter cells. Multiple surveillance systems exist that monitor proper execution of the cell division program and as such ensure stability of our genome. One widely studied protein complex essential for proper chromosome segregation and execution of cytoplasmic division (cytokinesis) is the chromosomal passenger complex (CPC). This highly conserved complex consists of Borealin, Survivin, INCENP, and Aurora B kinase, and has a dynamic localization pattern during mitosis and cytokinesis. Not surprisingly, it also performs various functions during these phases of the cell cycle. In this review, we will give an overview of the latest insights into the regulation of CPC localization and discuss if and how specific localization impacts its diverse functions in the dividing cell.


Asunto(s)
División Celular , Cromosomas/metabolismo , Animales , Centrómero/metabolismo , Retroalimentación Fisiológica , Humanos , Huso Acromático/metabolismo , Factores de Tiempo
7.
Biochem Soc Trans ; 43(1): 23-32, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25619243

RESUMEN

The evolutionary conserved chromosomal passenger complex (CPC) is essential for faithful transmission of the genome during cell division. Perturbation of this complex in cultured cells gives rise to chromosome segregation errors and cytokinesis failure and as a consequence the ploidy status of the next generation of cells is changed. Aneuploidy and chromosomal instability (CIN) is observed in many human cancers, but whether this may be caused by deregulation of the CPC is unknown. In the present review, we discuss if and how a dysfunctional CPC could contribute to CIN in cancer.


Asunto(s)
Inestabilidad Cromosómica , Segregación Cromosómica , Neoplasias/genética , Animales , Aurora Quinasa B/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Citocinesis , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Complejos Multiproteicos/fisiología , Neoplasias/metabolismo , Neoplasias/patología , Survivin
8.
Eur J Immunol ; 43(7): 1914-24, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23616302

RESUMEN

Survivin, an inhibitor of apoptosis family molecule, has been proposed as a crucial intermediate in the signaling pathways leading to T-cell development, proliferation, and expansion. However, the importance of survivin to T-cell-driven inflammatory responses has not been demonstrated. Here, we show that survivin transgenic mice exhibit an increased antigen-driven Th2 lung inflammation and that constitutive expression of survivin reversed the defective lung inflammation even in the absence of OX40 costimulation. We found that OX40-deficient mice were compromised in generating Th2 cells, airway eosinophilia, and IgE responses. In contrast, OX40-deficient/survivin transgenic mice generated normal Th2 responses and exhibited strong lung inflammation. These results suggest that OX40 costimulation crucially engages survivin during antigen-mediated Th2 responses. These findings also promote the notion that OX40 costimulation regulates allergic responses or lung inflammation by targeting survivin thereby enhancing T-cell proliferation and resulting in more differentiated Th2 cells in the allergic inflammatory response.


Asunto(s)
Hipersensibilidad/inmunología , Proteínas Inhibidoras de la Apoptosis/inmunología , Neumonía/inmunología , Receptores OX40/inmunología , Proteínas Represoras/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular/inmunología , Hipersensibilidad/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neumonía/metabolismo , Receptores OX40/deficiencia , Proteínas Represoras/metabolismo , Survivin , Células Th2/citología
9.
EMBO Rep ; 13(9): 847-54, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22732840

RESUMEN

Aurora B localization to mitotic centromeres, which is required for proper chromosome alignment during mitosis, relies on Haspin-dependent histone H3 phosphorylation and on Bub1-dependent histone H2A phosphorylation--which interacts with Borealin through a Shugoshin (Sgo) intermediate. We demonstrate that Mps1 stimulates the latter recruitment axis. Mps1 activity enhances H2A-T120ph and is critical for Sgo1 recruitment to centromeres, thereby promoting Aurora B centromere recruitment in early mitosis. Importantly, chromosome biorientation defects caused by Mps1 inhibition are improved by restoring Aurora B centromere recruitment. As Mps1 kinetochore localization reciprocally depends on Aurora B, we propose that this Aurora B-Mps1 recruitment circuitry cooperates with the Aurora B-Haspin feedback loop to ensure rapid centromere accumulation of Aurora B at the onset of mitosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Aurora Quinasa B , Aurora Quinasas , Células HeLa , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mitosis , Fosforilación
10.
Mol Cell Proteomics ; 11(5): 47-59, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22267324

RESUMEN

To understand how the chromosomal passenger complex ensures chromosomal stability, it is crucial to identify its substrates and to find ways to specifically inhibit the enzymatic core of the complex, Aurora B. We therefore developed a chemical genetic approach to selectively inhibit human Aurora B. By mutating the gatekeeper residue Leu-154 in the kinase active site, the ATP-binding pocket was enlarged, but kinase function was severely disrupted. A unique second site suppressor mutation was identified that rescued kinase activity in the Leu-154 mutant and allowed the accommodation of bulky N(6)-substituted adenine analogs. Using this analog-sensitive Aurora B kinase, we found that retention of the chromosomal passenger complex at the centromere depends on Aurora B kinase activity. Furthermore, analog-sensitive Aurora B was able to use bulky ATPγS analogs and could thiophosphorylate multiple proteins in cell extracts. Utilizing an unbiased approach for kinase substrate mapping, we identified several novel substrates of Aurora B, including the nucleosomal-binding protein HMGN2. We confirmed that HMGN2 is a bona fide Aurora B substrate in vivo and show that its dynamic association to chromatin is controlled by Aurora B.


Asunto(s)
Proteína HMGN2/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/genética , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Aurora Quinasa B , Aurora Quinasas , Dominio Catalítico , Extractos Celulares/química , Línea Celular Tumoral , Centrómero , Secuencia Conservada , Difosfatos/farmacología , Histonas/metabolismo , Humanos , Leucina/genética , Datos de Secuencia Molecular , Complejos Multiproteicos/metabolismo , Mutagénesis Sitio-Dirigida , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo
11.
J Cell Sci ; 124(Pt 4): 622-34, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21266467

RESUMEN

Precise control of the attachment strength between kinetochores and spindle microtubules is essential to preserve genomic stability. Aurora B kinase has been implicated in regulating the stability of kinetochore-microtubule attachments but its relevant kinetochore targets in cells remain unclear. Here, we identify multiple serine residues within the N-terminus of the kinetochore protein Hec1 that are phosphorylated in an Aurora-B-kinase-dependent manner during mitosis. On all identified target sites, Hec1 phosphorylation at kinetochores is high in early mitosis and decreases significantly as chromosomes bi-orient. Furthermore, once dephosphorylated, Hec1 is not highly rephosphorylated in response to loss of kinetochore-microtubule attachment or tension. We find that a subpopulation of Aurora B kinase remains localized at the outer kinetochore even upon Hec1 dephosphorylation, suggesting that Hec1 phosphorylation by Aurora B might not be regulated wholly by spatial positioning of the kinase. Our results define a role for Hec1 phosphorylation in kinetochore-microtubule destabilization and error correction in early mitosis and for Hec1 dephosphorylation in maintaining stable attachments in late mitosis.


Asunto(s)
Cinetocoros/metabolismo , Microtúbulos/metabolismo , Mitosis , Proteínas Nucleares/metabolismo , Aurora Quinasa B , Aurora Quinasas , Proteínas del Citoesqueleto , Células HeLa , Humanos , Cinetocoros/química , Microtúbulos/química , Microtúbulos/genética , Proteínas Nucleares/genética , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica
12.
Curr Opin Cell Biol ; 18(6): 616-22, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16962308

RESUMEN

Survivin has been proposed to inhibit apoptosis and to regulate cell division. However, controversy still exists as to whether Survivin can indeed execute these distinct functions and if Survivin somehow coordinates apoptosis and (abnormal) cell division. Recent evidence has demonstrated that Survivin acts as a subunit of the chromosomal passenger complex, which is essential for proper chromosome segregation and cytokinesis. Within this complex, the mitotic kinase Aurora B acts as the enzymatic core, whereas Survivin dictates chromosomal passenger complex localization. This function of Survivin appears to be conserved throughout evolution. Although these findings do not exclude a role for Survivin as apoptosis inhibitor, they make a very strong case for Survivin as mitotic regulator.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/genética , Proteínas de Neoplasias/metabolismo , Animales , Aurora Quinasa B , Aurora Quinasas , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica/genética , Evolución Molecular , Humanos , Proteínas Inhibidoras de la Apoptosis , Sustancias Macromoleculares/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/genética , Huso Acromático/metabolismo , Survivin
13.
Exp Cell Res ; 318(12): 1407-20, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22472345

RESUMEN

The Chromosomal Passenger Complex (CPC) consisting of Aurora B kinase, INCENP, Survivin and Borealin, is essential for genomic stability by controlling multiple processes during both nuclear and cytoplasmic division. In mitosis it ensures accurate segregation of the duplicated chromosomes by regulating the mitotic checkpoint, destabilizing incorrectly attached spindle microtubules and by promoting the axial shortening of chromosomal arms in anaphase. During cytokinesis the CPC most likely prevents chromosome damage by imposing an abscission delay when a chromosome bridge connects the two daughter cells. Moreover, by controlling proper cytoplasmic division, the CPC averts tetraploidization. This review describes recent insights on how the CPC is capable of conducting its various functions in the dividing cell to ensure chromosomal stability.


Asunto(s)
División Celular/genética , Proteínas Cromosómicas no Histona/fisiología , Complejos Multiproteicos/fisiología , Animales , Aurora Quinasa B , Aurora Quinasas , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , División Celular/fisiología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Citocinesis/genética , Citocinesis/fisiología , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Inhibidoras de la Apoptosis/fisiología , Modelos Biológicos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Survivin
14.
Trends Cell Biol ; 32(2): 165-176, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34663523

RESUMEN

The Chromosomal Passenger Complex (CPC) regulates a plethora of processes during multiple stages of nuclear and cytoplasmic division. Early during mitosis, the CPC is recruited to centromeres and kinetochores, and ensures that the duplicated chromosomes become properly connected to microtubules from opposite poles of the mitotic spindle. Progression into anaphase is accompanied by a striking relocation of the CPC from centromeres to the antiparallel microtubule overlaps of the anaphase spindle and to the equatorial cortex. This translocation requires direct interactions of the CPC with the kinesin-6 family member MKLP2/KIF20A, and the inactivation of cyclin B-cyclin-dependent kinase-1 (CDK1). Here, we review recent progress in the regulation of this relocation event. Furthermore, we discuss why the CPC must be relocated during early anaphase in light of recent advances in the functions of the CPC post metaphase.


Asunto(s)
Anafase , Proteínas Cromosómicas no Histona , Aurora Quinasa B/genética , Centrómero , Humanos , Microtúbulos , Mitosis , Huso Acromático
15.
J Cell Biol ; 221(8)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35776132

RESUMEN

Centromere association of the chromosomal passenger complex (CPC; Borealin-Survivin-INCENP-Aurora B) and Sgo1 is crucial for chromosome biorientation, a process essential for error-free chromosome segregation. Phosphorylated histone H3 Thr3 (H3T3ph; directly recognized by Survivin) and histone H2A Thr120 (H2AT120ph; indirectly recognized via Sgo1), together with CPC's intrinsic nucleosome-binding ability, facilitate CPC centromere recruitment. However, the molecular basis for CPC-Sgo1 binding and how their physical interaction influences CPC centromere localization are lacking. Here, using an integrative structure-function approach, we show that the "histone H3-like" Sgo1 N-terminal tail-Survivin BIR domain interaction acts as a hotspot essential for CPC-Sgo1 assembly, while downstream Sgo1 residues and Borealin contribute for high-affinity binding. Disrupting Sgo1-Survivin interaction abolished CPC-Sgo1 assembly and perturbed CPC centromere localization and function. Our findings reveal that Sgo1 and H3T3ph use the same surface on Survivin to bind CPC. Hence, it is likely that these interactions take place in a spatiotemporally restricted manner, providing a rationale for the Sgo1-mediated "kinetochore-proximal" CPC centromere pool.


Asunto(s)
Proteínas de Ciclo Celular , Centrómero , Histonas , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Histonas/genética , Histonas/metabolismo , Cinetocoros/metabolismo , Fosforilación , Survivin/genética , Survivin/metabolismo
16.
Cell Rep ; 39(1): 110584, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385742

RESUMEN

Elevated expression of non-receptor tyrosine kinase FER is an independent prognosticator that correlates with poor survival of high-grade and basal/triple-negative breast cancer (TNBC) patients. Here, we show that high FER levels are also associated with improved outcomes after adjuvant taxane-based combination chemotherapy in high-risk, HER2-negative patients. In TNBC cells, we observe a causal relation between high FER levels and sensitivity to taxanes. Proteomics and mechanistic studies demonstrate that FER regulates endosomal recycling, a microtubule-dependent process that underpins breast cancer cell invasion. Using chemical genetics, we identify DCTN2 as a FER substrate. Our work indicates that the DCTN2 tyrosine 6 is essential for the development of tubular recycling domains in early endosomes and subsequent propagation of TNBC cell invasion in 3D. In conclusion, we show that high FER expression promotes endosomal recycling and represents a candidate predictive marker for the benefit of adjuvant taxane-containing chemotherapy in high-risk patients, including TNBC patients.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama/metabolismo , Hidrocarburos Aromáticos con Puentes/farmacología , Hidrocarburos Aromáticos con Puentes/uso terapéutico , Endosomas/metabolismo , Femenino , Humanos , Taxoides/farmacología , Taxoides/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo
17.
Chromosoma ; 119(4): 405-13, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20354862

RESUMEN

Error-free chromosome segregation requires that all chromosomes biorient on the mitotic spindle. The motor protein Centromere-associated protein E (CENP-E) facilitates chromosome congression by mediating the lateral sliding of sister chromatids along existing K-fibers, while the mitotic kinase Aurora B detaches kinetochore-microtubule interactions that are not bioriented. Whether these activities cooperate to promote efficient chromosome biorientation and timely anaphase onset is not known. We here show that the chromosomes that fail to congress after CENP-E depletion displayed high centromeric Aurora B kinase activity. This activity destabilized spindle pole proximal kinetochore-microtubule interactions resulting in a checkpoint-dependent mitotic delay that allowed CENP-E-independent chromosome congression, thus reducing chromosome segregation errors. This shows that Aurora B keeps the mitotic checkpoint active by destabilizing kinetochore fibers of polar chromosomes to permit chromosome congression in CENP-E-compromised cells and implies that this kinase normally prevents pole proximal syntelic attachments to allow CENP-E-mediated congression of mono-oriented chromosomes.


Asunto(s)
Anafase/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica/fisiología , Cromosomas Humanos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Aurora Quinasa B , Aurora Quinasas , Células HeLa , Humanos , Cinetocoros/metabolismo , Microscopía Fluorescente/métodos , Microtúbulos/metabolismo , Mitosis , ARN Interferente Pequeño , Huso Acromático/metabolismo
18.
Hum Reprod ; 26(7): 1868-81, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21493633

RESUMEN

BACKGROUND: Human embryos generated by IVF demonstrate a high incidence of chromosomal segregation errors during the cleavage divisions. To analyse underlying molecular mechanisms, we investigated the behaviour of the chromosomal passenger complex (CPC) in human oocytes and embryos. This important mitotic regulatory complex comprises the inner centromere protein (INCENP), survivin, borealin and Aurora B, or the meiotic kinase Aurora C. METHODS: We analysed mRNA expression by quantitative RT-PCR of all CPC members in human oocytes, tripronuclear (3PN) zygotes, 2-cell and 4-cell embryos developed from 3PN zygotes, plus good-quality cryopreserved 8-cell, morula and blastocyst stage embryos. Protein expression and localization of CPC members were investigated by immunofluorescence in oocytes and embryos arrested at prometaphase. Histone H3S10 phosphorylation was investigated as an indicator of a functional CPC. RESULTS: INCENP, survivin and borealin were detected at the inner centromere of prometaphase chromosomes in all stages investigated. Whereas Aurora B and C are both present in oocytes, Aurora C becomes the most prominent kinase in the CPC during the first three embryonic cell cycles. Moreover, Aurora C mRNA was up-regulated with Aurora B after activation of the embryonic genome and both proteins were detected in early Day 4 embryos. Subsequently, only Aurora B was detected in blastocysts. CONCLUSIONS: In contrast to somatic cells, our results point to a specific role for Aurora C in the CPC during human preimplantation embryo development. Although, the presence of Aurora C in itself may not explain the high chromosome segregation error rate, the data presented here provide novel information regarding possible mechanisms.


Asunto(s)
Blastocisto/enzimología , Proteínas Cromosómicas no Histona/fisiología , Desarrollo Embrionario/genética , Proteínas Serina-Treonina Quinasas/fisiología , Aurora Quinasa B , Aurora Quinasa C , Aurora Quinasas , Blastocisto/metabolismo , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas Inhibidoras de la Apoptosis/análisis , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Oocitos/enzimología , Oocitos/metabolismo , Embarazo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/metabolismo , Espermatocitos/enzimología , Survivin
19.
J Cell Biol ; 173(6): 833-7, 2006 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-16769825

RESUMEN

During mitosis, the chromosomal passenger complex (CPC) orchestrates highly different processes, such as chromosome alignment, histone modification, and cytokinesis. Proper and timely localization of this complex is the key to precise control over the enzymatic core of the CPC, the Aurora-B kinase. We discuss the molecular mechanisms by which the CPC members direct the dynamic localization of the complex throughout cell division. Also, we summarize posttranslational modifications that occur on the CPC and discuss their roles in regulating localization and function of this mitotic complex.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Mitosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Aurora Quinasas , Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Fosforilación , Procesamiento Proteico-Postraduccional , Huso Acromático/metabolismo
20.
J Cell Biol ; 220(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33404606

RESUMEN

How chromatin bridges are relayed to the chromosomal passenger complex (CPC) during mammalian cell division is unknown. In this issue, Petsalaki and Zachos (2020. J. Cell Biol.https://doi.org/10.1083/jcb.202008029) show that the DNA damage checkpoint kinases ATM and Chk2 signal to the CPC to associate with a pool of cytoskeletal regulators, MKLP2-Cep55, in the midbody center and to delay abscission.


Asunto(s)
Citocinesis , Huso Acromático , Animales , Aurora Quinasa B/genética , Proteínas de Ciclo Celular/genética , División Celular , Células HeLa , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA