Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
PLoS One ; 11(4): e0153518, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27082996

RESUMEN

OTSSP167 was recently characterized as a potent inhibitor for maternal embryonic leucine zipper kinase (MELK) and is currently tested in Phase I clinical trials for solid tumors that have not responded to other treatment. Here we report that OTSSP167 abrogates the mitotic checkpoint at concentrations used to inhibit MELK. The abrogation is not recapitulated by RNAi mediated silencing of MELK in cells. Although OTSSP167 indeed inhibits MELK, it exhibits off-target activity against Aurora B kinase in vitro and in cells. Furthermore, OTSSP167 inhibits BUB1 and Haspin kinases, reducing phosphorylation at histones H2AT120 and H3T3 and causing mislocalization of Aurora B and associated chromosomal passenger complex from the centromere/kinetochore. The results suggest that OTSSP167 may have additional mechanisms of action for cancer cell killing and caution the use of OTSSP167 as a MELK specific kinase inhibitor in biochemical and cellular assays.


Asunto(s)
Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Naftiridinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Anticuerpos/farmacología , Aurora Quinasa B/antagonistas & inhibidores , Centrómero/efectos de los fármacos , Centrómero/fisiología , Células HeLa , Humanos , Cinetocoros/efectos de los fármacos , Cinetocoros/fisiología , Células MCF-7 , Mitosis/efectos de los fármacos , Mitosis/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Transducción de Señal/efectos de los fármacos
3.
Proc Natl Acad Sci U S A ; 111(33): 12019-24, 2014 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-25092294

RESUMEN

The mitotic (or spindle assembly) checkpoint system delays anaphase until all chromosomes are correctly attached to the mitotic spindle. When the checkpoint is active, a Mitotic Checkpoint Complex (MCC) assembles and inhibits the ubiquitin ligase Anaphase-Promoting Complex/Cyclosome (APC/C). MCC is composed of the checkpoint proteins Mad2, BubR1, and Bub3 associated with the APC/C activator Cdc20. When the checkpoint signal is turned off, MCC is disassembled and the checkpoint is inactivated. The mechanisms of the disassembly of MCC are not sufficiently understood. We have previously observed that ATP hydrolysis is required for the action of the Mad2-binding protein p31(comet) to disassemble MCC. We now show that HeLa cell extracts contain a factor that promotes ATP- and p31(comet)-dependent disassembly of a Cdc20-Mad2 subcomplex and identify it as Thyroid Receptor Interacting Protein 13 (TRIP13), an AAA-ATPase known to interact with p31(comet). The joint action of TRIP13 and p31(comet) also promotes the release of Mad2 from MCC, participates in the complete disassembly of MCC and abrogates checkpoint inhibition of APC/C. We propose that TRIP13 plays centrally important roles in the sequence of events leading to MCC disassembly and checkpoint inactivation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Portadoras/fisiología , Proteínas de Ciclo Celular/fisiología , Mitosis , Proteínas Nucleares/fisiología , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Cdc20/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células HeLa , Humanos , Proteínas Mad2/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica
4.
J Biol Chem ; 289(34): 23928-37, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25012665

RESUMEN

The mitotic checkpoint (or spindle assembly checkpoint) is a fail-safe mechanism to prevent chromosome missegregation by delaying anaphase onset in the presence of defective kinetochore-microtubule attachment. The target of the checkpoint is the E3 ubiquitin ligase anaphase-promoting complex/cyclosome. Once all chromosomes are properly attached and bioriented at the metaphase plate, the checkpoint needs to be silenced. Previously, we and others have reported that TRIP13 AAA-ATPase binds to the mitotic checkpoint-silencing protein p31(comet). Here we show that endogenous TRIP13 localizes to kinetochores. TRIP13 knockdown delays metaphase-to-anaphase transition. The delay is caused by prolonged presence of the effector for the checkpoint, the mitotic checkpoint complex, and its association and inhibition of the anaphase-promoting complex/cyclosome. These results suggest that TRIP13 is a novel mitotic checkpoint-silencing protein. The ATPase activity of TRIP13 is essential for its checkpoint function, and interference with TRIP13 abolished p31(comet)-mediated mitotic checkpoint silencing. TRIP13 overexpression is a hallmark of cancer cells showing chromosomal instability, particularly in certain breast cancers with poor prognosis. We suggest that premature mitotic checkpoint silencing triggered by TRIP13 overexpression may promote cancer development.


Asunto(s)
Proteínas Portadoras/fisiología , Mitosis/fisiología , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas Mad2/metabolismo , Microscopía Fluorescente , Proteínas Nucleares/metabolismo , Interferencia de ARN
5.
Dev Cell ; 21(6): 1077-91, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22137763

RESUMEN

Here, we report a cell-intrinsic mechanism by which oncogenic RAS promotes senescence while predisposing cells to senescence bypass by allowing for secondary hits. We show that oncogenic RAS inactivates the BRCA1 DNA repair complex by dissociating BRCA1 from chromatin. This event precedes senescence-associated cell cycle exit and coincides with the accumulation of DNA damage. Downregulation of BRIP1, a physiological partner of BRCA1 in the DNA repair pathway, triggers BRCA1 chromatin dissociation. Conversely, ectopic BRIP1 rescues BRCA1 chromatin dissociation and suppresses RAS-induced senescence and the DNA damage response. Significantly, cells undergoing senescence do not exhibit a BRCA1-dependent DNA repair response when exposed to DNA damage. Overall, our study provides a molecular basis by which oncogenic RAS promotes senescence. Because DNA damage has the potential to produce additional "hits" that promote senescence bypass, our findings may also suggest one way a small minority of cells might bypass senescence and contribute to cancer development.


Asunto(s)
Proteína BRCA1/metabolismo , Cromatina/genética , Cromatina/metabolismo , Reparación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Genes ras , ARN Helicasas/metabolismo , Ciclo Celular , Línea Celular , Transformación Celular Neoplásica/genética , Senescencia Celular/genética , Senescencia Celular/fisiología , Daño del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes BRCA1 , Humanos , ARN Helicasas/genética
6.
Clin Chim Acta ; 412(23-24): 2267-71, 2011 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-21888900

RESUMEN

BACKGROUND: Anticentromere autoantibodies have been reported to be associated with scleroderma and serve as a marker in different rheumatic diseases in humans. Major centromere autoantigens described so far include constitutive kinetochore proteins such as CENPA, CENPB, CENPC and CENPH and facultative proteins such as CENPE, CENPF and INCENP. We examined the inner kinetochore component CENPI as a new putative centromere autoantigen in scleroderma patients. METHODS: To test for the presence of CENPI centromere autoantibodies, 72 sera from patients with systemic lupus erythematosus and systemic sclerosis were assayed by immunofluorescence and further tested by immunoblots with an Nt-CENPI recombinant protein. RESULTS: 8 out of 31 (25.8%) patients diagnosed of scleroderma or Undifferentiated Connective Tissue Disease (UCTD) produced anti-CENPI autoantibodies. Epitopes were demonstrated to be located mainly but not exclusively in the N-terminal domain of the human CENPI protein. Five of the 8 (62.5%) CENPI positive sera also had other autoantibodies related to primary biliary cirrhosis. Further, two patients (25%) with anti-CENPI autoantibodies had concurrent diagnosis of primary biliary cirrhosis. CONCLUSIONS: This study demonstrates that CENPI, a centromere protein that localizes to the inner kinetochore structure, is a human autoantigen. The significance of anti-CENPI autoantibodies could be relevant in scleroderma patients as a marker for concurrent autoimmune liver disease.


Asunto(s)
Autoanticuerpos/inmunología , Proteínas de Unión al ADN/inmunología , Hepatopatías/inmunología , Esclerodermia Sistémica/inmunología , Epítopos/inmunología , Técnica del Anticuerpo Fluorescente , Humanos
7.
Cell ; 137(4): 672-84, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19450515

RESUMEN

Chromosome segregation requires assembly of kinetochores on centromeric chromatin to mediate interactions with spindle microtubules and control cell-cycle progression. To elucidate the protein architecture of human kinetochores, we developed a two-color fluorescence light microscopy method that measures average label separation, Delta, at <5 nm accuracy. Delta analysis of 16 proteins representing core structural complexes spanning the centromeric chromatin-microtubule interface, when correlated with mechanical states of spindle-attached kinetochores, provided a nanometer-scale map of protein position and mechanical properties of protein linkages. Treatment with taxol, which suppresses microtubule dynamics and activates the spindle checkpoint, revealed a specific switch in kinetochore architecture. Cumulatively, Delta analysis revealed that compliant linkages are restricted to the proximity of chromatin, suggested a model for how the KMN (KNL1/Mis12 complex/Ndc80 complex) network provides microtubule attachment and generates pulling forces from depolymerization, and identified an intrakinetochore molecular switch that may function in controlling checkpoint activity.


Asunto(s)
Cinetocoros/química , Cinetocoros/metabolismo , Microtúbulos/química , Microtúbulos/metabolismo , Proteínas del Citoesqueleto , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Metafase , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares
9.
Front Biosci (Landmark Ed) ; 14(10): 3733-9, 2009 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-19273306

RESUMEN

Nuclear Autoantigen of 14 kDa (NA14) was originally identified using the serum of a Sjögren's syndrome (SS) patient as probe in screening a human testis cDNA expression library. To date there is no report in the systematic analysis of the prevalence of autoantibodies to NA14. In this study, anti-NA14 was determined in several rheumatic diseases from independent cohorts in the US and Japan. The prevalence of anti-NA14 were 18/132 (13.6%) in primary SS, 0/50 (0%) secondary SS, 2/100 (2%) SLE, 1/43 (2.3%) scleroderma, 0/54 (0%) rheumatoid arthritis, 1/29 (3.4%) polymyositis/dermatomyositis, and 0/58 (0%) normal healthy controls. The frequencies of anti-NA14 positive sera in primary SS are statistically greater than normal healthy controls (p=0.006), secondary SS (p=0.044), and other rheumatic diseases. Furthermore, among 11 anti-NA14 positive primary SS sera, 4/11 (36.3%) sera were negative for both anti-SS-A/Ro and SS-B/La antibodies. Thus anti-NA14 autoantibodies may be useful for the discrimination of primary versus secondary SS and serve as a diagnostic marker for primary SS especially in seronegative (anti-SS-A/Ro and anti-SS-B/La antibodies negative) patients with SS.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Biomarcadores/sangre , Proteínas Nucleares/inmunología , Síndrome de Sjögren/inmunología , Especificidad de Anticuerpos , Autoanticuerpos/sangre , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Humanos
10.
Mol Biol Cell ; 20(5): 1289-301, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19116315

RESUMEN

Cohesin is an essential protein complex required for sister chromatid cohesion. Cohesin associates with chromosomes and establishes sister chromatid cohesion during interphase. During metaphase, a small amount of cohesin remains at the chromosome-pairing domain, mainly at the centromeres, whereas the majority of cohesin resides in the cytoplasm, where its functions remain unclear. We describe the mitosis-specific recruitment of cohesin to the spindle poles through its association with centrosomes and interaction with nuclear mitotic apparatus protein (NuMA). Overexpression of NuMA enhances cohesin accumulation at spindle poles. Although transient cohesin depletion does not lead to visible impairment of normal spindle formation, recovery from nocodazole-induced spindle disruption was significantly impaired. Importantly, selective blocking of cohesin localization to centromeres, which disrupts centromeric sister chromatid cohesion, had no effect on this spindle reassembly process, clearly separating the roles of cohesin at kinetochores and spindle poles. In vitro, chromosome-independent spindle assembly using mitotic extracts was compromised by cohesin depletion, and it was rescued by addition of cohesin that was isolated from mitotic, but not S phase, cells. The combined results identify a novel spindle-associated role for human cohesin during mitosis, in addition to its function at the centromere/kinetochore regions.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Mitosis , Huso Acromático/metabolismo , Animales , Antígenos Nucleares/química , Antígenos Nucleares/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Centrosoma/metabolismo , Centrosoma/ultraestructura , Pollos/metabolismo , Cromátides/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Células HeLa , Humanos , Nocodazol/farmacología , Proteínas Asociadas a Matriz Nuclear/química , Proteínas Asociadas a Matriz Nuclear/metabolismo , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Huso Acromático/efectos de los fármacos , Huso Acromático/ultraestructura , Cohesinas
11.
Proc Natl Acad Sci U S A ; 105(27): 9181-5, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18591651

RESUMEN

The mitotic checkpoint system ensures the fidelity of chromosome segregation by preventing the completion of mitosis in the presence of any misaligned chromosome. When activated, it blocks the initiation of anaphase by inhibiting the ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C). Little is known about the biochemical mechanisms by which this system inhibits APC/C, except for the existence of a mitotic checkpoint complex (MCC) inhibitor of APC/C composed of the APC/C activator Cdc20 associated with the checkpoint proteins Mad2, BubR1, and Bub3. We have been studying the mechanisms of the mitotic checkpoint system in extracts that reproduce its downstream events. We found that inhibitory factors are associated with APC/C in the checkpoint-arrested state, which can be recovered from immunoprecipitates. Only a part of the inhibitory activity was caused by MCC [Braunstein I, Miniowitz S, Moshe Y, Hershko A (2007) Proc Natl Acad Sci USA 104:4870-4875]. Here, we show that during exit from checkpoint, rapid disassembly of MCC takes place while APC/C is still inactive. This observation suggested the possible involvement of multiple factors in the regulation of APC/C by the mitotic checkpoint. We have separated a previously unknown inhibitor of APC/C from MCC. This inhibitor, called mitotic checkpoint factor 2 (MCF2), is associated with APC/C only in the checkpoint-arrested state. The inhibition of APC/C by both MCF2 and MCC was decreased at high concentrations of Cdc20. We propose that both MCF2 and MCC inhibit APC/C by antagonizing Cdc20, possibly by interaction with the Cdc20-binding site of APC/C.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Mitosis , Complejos de Ubiquitina-Proteína Ligasa/antagonistas & inhibidores , Ciclosoma-Complejo Promotor de la Anafase , Proteínas Cdc20 , Células HeLa , Humanos , Factores de Tiempo
12.
Cancer Res ; 68(10): 3724-32, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18483255

RESUMEN

X-irradiation-induced DNA damage perturbs the G(1), S, and G(2) phases of the cell cycle. The behavior of cells after they have experienced a DNA damage checkpoint delay is poorly characterized. We therefore examined the fates of irradiated tumor cells that have overcome a prolonged G(2) checkpoint delay. Most irradiated cells progressed through mitosis without significant delay, but failed to complete cytokinesis as they remained tethered to each other at the midbody. We observed that the movement of centrioles at the time of cytokinesis was impaired in the irradiated, bridged cells. We attribute the perturbation of centriole dynamics to the presence of chromatin bridges that spanned the daughter cells. The bridged cells exhibited different fates that included death, fusion that formed multinucleated cells, or another round of mitosis with no noticeable cell cycle delays. The presence of gammaH2AX foci in the bridge as well as in the separated nuclei indicated that cells were proliferating despite the presence of DNA damage. It seems that DNA damage checkpoints were not reactivated in cells that overrode a prolonged G(2) delay. Cells deficient in ATM, H2AX, XRCC3, or ligase 4 exhibited a higher frequency of radiation-induced bridges than controls, suggesting that the DNA bridges resulted from inadequate DNA repair. These data show a previously unappreciated cytologic hallmark of DNA damage in dividing cells. Chromatin bridges that interfere with cytokinesis are likely to contribute to the replication failure and clonogenic death of cells exposed to irradiation.


Asunto(s)
Cromatina/química , Citocinesis , Daño del ADN , Fase G2 , Animales , Ciclo Celular , Cromatina/metabolismo , Fibroblastos/metabolismo , Células HeLa , Humanos , Ratones , Mitosis , Modelos Biológicos , ARN Interferente Pequeño/metabolismo , Radiación Ionizante , Rayos X
13.
Mol Cell ; 29(6): 729-41, 2008 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-18374647

RESUMEN

SUMOylation is essential for cell-cycle regulation in invertebrates; however, its functions during the mammalian cell cycle are largely uncharacterized. Mammals express three SUMO paralogs: SUMO-1, SUMO-2, and SUMO-3 (SUMO-2 and SUMO-3 are 96% identical and referred to as SUMO-2/3). We found that SUMO-2/3 localize to centromeres and condensed chromosomes, whereas SUMO-1 localizes to the mitotic spindle and spindle midzone, indicating that SUMO paralogs regulate distinct mitotic processes in mammalian cells. Consistent with this, global inhibition of SUMOylation caused a prometaphase arrest due to defects in targeting the microtubule motor protein CENP-E to kinetochores. CENP-E was found to be modified specifically by SUMO-2/3 and to possess SUMO-2/3 polymeric chain-binding activity essential for kinetochore localization. Our findings indicate that SUMOylation is a key regulator of the mammalian cell cycle, with SUMO-1 and SUMO-2/3 modification of different proteins regulating distinct processes.


Asunto(s)
Ciclo Celular/fisiología , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cinetocoros/metabolismo , Mitosis/fisiología , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Cisteína Endopeptidasas/metabolismo , ADN-Topoisomerasas/metabolismo , Genes Reporteros , Células HeLa , Humanos , Cinética , Metafase , Unión Proteica
14.
Biochem Biophys Res Commun ; 370(2): 213-9, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18361916

RESUMEN

Alterations in the expression and activity of the centrosomal kinase, Aurora-A/STK15, affect genomic stability, disrupt the fidelity of centrosome duplication, and induce cellular transformation. A mitotic spindle-associated protein, astrin/DEEPEST, was identified as an Aurora-A interacting protein by a two-hybrid screen. Astrin and Aurora-A co-express at mitosis and co-localize to mitotic spindles. RNAi-mediated depletion of astrin abolishes the localization of Aurora-A on mitotic spindles and leads to a moderate mitotic cell cycle delay, which resembles the mitotic arrest phenotypes in siAurora-A treated cells. However, depletion of Aurora-A does not affect astrin localization, and co-depletion of both astrin and Aurora-A causes a mitotic arrest phenotype similar to depletion of siAurora-A alone. These results suggest that astrin acts upstream of Aurora-A to regulate its mitotic spindle localization.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/enzimología , Aurora Quinasa A , Aurora Quinasas , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Centrosoma/enzimología , Epistasis Genética , Células HeLa , Humanos , Mitosis/efectos de los fármacos , Mitosis/genética , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Saccharomyces cerevisiae/genética , Técnicas del Sistema de Dos Híbridos
15.
J Cell Biol ; 177(3): 413-24, 2007 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-17485487

RESUMEN

hSgo2 (previously annotated as Tripin) was recently reported to be a new inner centromere protein that is essential for centromere cohesion (Kitajima et al., 2006). In this study, we show that hSgo2 exhibits a dynamic distribution pattern, and that its localization depends on the BUB1 and Aurora B kinases. hSgo2 is concentrated at the inner centromere of unattached kinetochores, but extends toward the kinetochores that are under tension. This localization pattern is reminiscent of MCAK, which is a microtubule depolymerase that is believed to be a key component of the error correction mechanism at kinetochores. Indeed, we found that hSgo2 is essential for MCAK to localize to the centromere. Delocalization of MCAK accounts for why cells depleted of hSgo2 exhibit kinetochore attachment defects that go uncorrected, despite a transient delay in the onset of anaphase. Consequently, these cells exhibit a high frequency of lagging chromosomes when they enter anaphase. We confirmed that hSgo2 is associated with PP2A, and we propose that it contributes to the spatial regulation of MCAK activity within inner centromere and kinetochore.


Asunto(s)
Anafase/fisiología , Proteínas de Ciclo Celular/metabolismo , Cinesinas/metabolismo , Cinetocoros/metabolismo , Aurora Quinasa B , Aurora Quinasas , Células HeLa , Humanos , Cinetocoros/ultraestructura , Fosfoproteínas Fosfatasas/metabolismo , Unión Proteica/fisiología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/fisiología
16.
Cell ; 128(1): 20-1, 2007 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-17218250

RESUMEN

In this issue, Baumann et al. (2007) identify a helicase PICH that localizes to "threads" that remain connected between sister kinetochores after they have separated in anaphase. These threads are thought to be catenated centromeric DNA. PICH contributes to the mitotic checkpoint by recruiting Mad2 to kinetochores and is proposed to regulate checkpoint signaling by monitoring tension at centromeres.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN Helicasas/metabolismo , Cinetocoros/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , Cromatina/metabolismo , ADN Helicasas/química , ADN Helicasas/deficiencia , Humanos , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Huso Acromático/metabolismo , Especificidad por Sustrato , Quinasa Tipo Polo 1
17.
J Cell Biol ; 175(1): 41-53, 2006 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-17030981

RESUMEN

We report the interactions amongst 20 proteins that specify their assembly to the centromere-kinetochore complex in human cells. Centromere protein (CENP)-A is at the top of a hierarchy that directs three major pathways, which are specified by CENP-C, -I, and Aurora B. Each pathway consists of branches that intersect to form nodes that may coordinate the assembly process. Complementary EM studies found that the formation of kinetochore trilaminar plates depends on the CENP-I/NUF2 branch, whereas CENP-C and Aurora B affect the size, shape, and structural integrity of the plates. We found that hMis12 is not constitutively localized at kinetochores, and that it is not essential for recruiting CENP-I. Our studies also revealed that kinetochores in HeLa cells contain an excess of CENP-A, of which approximately 10% is sufficient to promote the assembly of normal levels of kinetochore proteins. We elaborate on a previous model that suggested kinetochores are assembled from repetitive modules (Zinkowski, R.P., J. Meyne, and B.R. Brinkley. 1991. J. Cell Biol. 113:1091-110).


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Cinetocoros/metabolismo , Modelos Genéticos , Aurora Quinasa B , Aurora Quinasas , Autoantígenos/metabolismo , Autoantígenos/fisiología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteína A Centromérica , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Células HeLa , Humanos , Cinetocoros/ultraestructura , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología
18.
Rheumatol Int ; 26(4): 298-303, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15700116

RESUMEN

Anti-centromere antibody (ACA) has been reported to be associated with Sjogren's syndrome (SS) and the clinical significance of anti-CENP-H antibody remains unknown. To determine the clinical significance of anti-CENP-H and anti-centromere antibodies in primary SS, sera from 62 patients with primary SS and 40 normal controls were examined for anti-SS-A/SS-B antibodies, ACA and anti-CENP-H antibodies, by enzyme-linked immunosorbent assay and indirect immunofluorescence (IIF), respectively. Of the 62 serum samples with primary SS, 17 were positive with ACA and anti-CENP-H antibodies. Sera from SS patients with anti-CENP-H and ACA antibodies do not contain anti-SS-A/Ro and/or anti-SS-B/La antibodies. No anti-CENP-H antibody was found in sera of normal controls. An increased frequency of ACA and anti-CENP-H antibodies was found for the first time in patients with SS. Anti-CENP-H antibodies and anti-SS-A/Ro or anti-SS-B/La antibodies are present mutually exclusive. Patients with anti-CENP-H antibodies had a lower frequency of rheumatoid factor (RF). SS can be subdivided serologically into two groups; group one with anti-SS-A/Ro and/or anti-SS-B/La antibody, group two with ACA and/or anti-CENP-H antibodies. We recommend that ACA or anti-CENP-H antibodies should be considered as one of the serological markers for SS.


Asunto(s)
Anticuerpos Antinucleares/sangre , Autoantígenos/inmunología , Centrómero/inmunología , Proteínas Cromosómicas no Histona/inmunología , Síndrome de Sjögren/inmunología , Animales , Autoantígenos/biosíntesis , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibroblastos/citología , Técnica del Anticuerpo Fluorescente Indirecta , Células HeLa , Humanos , Técnicas para Inmunoenzimas , Ciervo Muntjac , Conejos , Factor Reumatoide/sangre , Síndrome de Sjögren/diagnóstico
19.
Trends Cell Biol ; 15(11): 589-98, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16214339

RESUMEN

The vertebrate kinetochore is a complex structure that specifies the attachments between the chromosomes and microtubules of the spindle and is thus essential for accurate chromosome segregation. Kinetochores are assembled on centromeric chromatin through complex pathways that are coordinated with the cell cycle. In the light of recent discoveries on how proteins assemble onto kinetochores and interact with each other, we review these findings in this article (which is part of the Chromosome Segregation and Aneuploidy series), and discuss their implications for the current mitotic checkpoint models - the template model and the two-step model. The template model proposes that Mad1-Mad2 at kinetochores acts as a template to change the conformation of another binding molecule of Mad2. This templated change in conformation is postulated as a mechanism for the amplification of the 'anaphase wait' signal. The two-step model proposes that the mitotic checkpoint complex (MCC) is the kinetochore-independent anaphase inhibitor, and the role of the unaligned kinetochore is to sensitize the anaphase-promoting complex/cyclosome (APC/C) to MCC-mediated inhibition.


Asunto(s)
Cinetocoros/fisiología , Ciclosoma-Complejo Promotor de la Anafase , Animales , Proteínas de Ciclo Celular/fisiología , Segregación Cromosómica/fisiología , Humanos , Cinetocoros/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Microtúbulos/metabolismo , Modelos Biológicos , Unión Proteica , Complejos de Ubiquitina-Proteína Ligasa/fisiología
20.
Biochim Biophys Acta ; 1744(2): 89-92, 2005 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15950749

RESUMEN

Two splice variants of Nek2 kinase, a member of the NIMA-related family, have been identified as Nek2A and Nek2B. Nek2A regulates centrosome disjunction, spindle formation checkpoint signaling, and faithful chromosome segregation. A specific role for Nek2B has not yet been identified. Here, we have examined the distinct roles of Nek2A and Nek2B using timelapse video microscopy to follow the fate of cells progressing through the cell cycle in the absence of either Nek2A or Nek2B. We show that the down-regulation of Nek2B leads to a mitotic delay in the majority of cells. Upon exiting mitosis, cells exhibit mitotic defects such as the formation of multinucleated cells. Such phenotypes are not observed in cells that exit mitosis in the absence of Nek2A. These observations suggest that Nek2B may be required for the execution of mitotic exit.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Ciclo Celular , Procesamiento de Imagen Asistido por Computador , Proteínas Serina-Treonina Quinasas/fisiología , Empalme Alternativo , Proteínas de Ciclo Celular/genética , Centrosoma/fisiología , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/fisiología , Microscopía por Video , Mitosis/genética , Peso Molecular , Quinasas Relacionadas con NIMA , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/farmacología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...