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











Base de datos
Intervalo de año de publicación
1.
Animal Model Exp Med ; 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38247322

RESUMEN

BACKGROUND: Cholangiocarcinoma (CCA), a malignancy that arises from biliary epithelial cells, has a dismal prognosis, and few targeted therapies are available. Aurora B, a key mitotic regulator, has been reported to be involved in the progression of various tumors, yet its role in CCA is still unclarified. METHODS: Human CCA tissues and murine spontaneous CCA models were used to assess Aurora B expression in CCA. A loss-of-function model was constructed in CCA cells to determine the role of Aurora B in CCA progression. Subcutaneous and liver orthotopic xenograft models were used to assess the therapeutic potential of Aurora B inhibitors in CCA. RESULTS: In murine spontaneous CCA models, Aurora B was significantly upregulated. Elevated Aurora B expression was also observed in 62.3% of human specimens in our validation cohort (143 CCA specimens), and high Aurora B expression was positively correlated with pathological parameters of tumors and poor survival. Knockdown of Aurora B by siRNA and heteroduplex oligonucleotide (HDO) or an Aurora B kinase inhibitor (AZD1152) significantly suppressed CCA progression via G2/M arrest induction. An interaction between Aurora B and c-Myc was found in CCA cells. Targeting Aurora B significantly reduced this interaction and accelerated the proteasomal degradation of c-Myc, suggesting that Aurora B promoted the malignant properties of CCA by stabilizing c-Myc. Furthermore, sequential application of AZD1152 or Aurora B HDO drastically improved the efficacy of gemcitabine in CCA. CONCLUSIONS: Aurora B plays an essential role in CCA progression by modulating c-Myc stability and represents a new target for treatment and chemosensitization in CCA.

2.
Biochem Biophys Res Commun ; 681: 180-185, 2023 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-37783115

RESUMEN

A bioluminescent immunoassay system was developed to determine serine/threonine protein kinase activity using an aequorin-labeled monoclonal antibody and a synthetic peptide as the substrate. A monoclonal antibody against the synthetic phosphorylated serine peptide (K9P peptide) of histone H3 (19 amino acid residues), referred to as the H3S10P antibody, was chemically conjugated to maleimide-activated aequorin to prepare aequorin-labeled H3S10P (AQ-S-H3S10P). For the serine/threonine kinase assay, a non-phosphorylated serine peptide (K9C peptide) coated on a microplate was incubated with serine/threonine protein kinase in the presence of ATP and Mg2+. The resulting phosphorylated K9C peptides (K9P peptide) were identified using AQ-S-H3S10P. Thus, after the removal of unbound AQ-S-H3S10P though washing, the serine/threonine kinase activity was determined by the luminescence activity of aequorin from AQ-S-H3S10P bound to the K9P peptide. This assay system, in combination with the K9C peptide and AQ-S-H3S10P, could be used to screen inhibitors of various serine/threonine protein kinases in general.


Asunto(s)
Aequorina , Anticuerpos Monoclonales , Aequorina/metabolismo , Anticuerpos Monoclonales/metabolismo , Inmunoensayo/métodos , Péptidos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Treonina/metabolismo , Especificidad por Sustrato
3.
Int J Mol Sci ; 24(5)2023 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-36902447

RESUMEN

Tumor Treating Fields (TTFields) were incorporated into the treatment of glioblastoma, the most malignant brain tumor, after showing an effect on progression-free and overall survival in a phase III clinical trial. The combination of TTFields and an antimitotic drug might further improve this approach. Here, we tested the combination of TTFields with AZD1152, an Aurora B kinase inhibitor, in primary cultures of newly diagnosed (ndGBM) and recurrent glioblastoma (rGBM). AZD1152 concentration was titrated for each cell line and 5-30 nM were used alone or in addition to TTFields (1.6 V/cm RMS; 200 kHz) applied for 72 h using the inovitro™ system. Cell morphological changes were visualized by conventional and confocal laser microscopy. The cytotoxic effects were determined by cell viability assays. Primary cultures of ndGBM and rGBM varied in p53 mutational status; ploidy; EGFR expression and MGMT-promoter methylation status. Nevertheless; in all primary cultures; a significant cytotoxic effect was found following TTFields treatment alone and in all but one, a significant effect after treatment with AZD1152 alone was also observed. Moreover, in all primary cultures the combined treatment had the most pronounced cytotoxic effect in parallel with morphological changes. The combined treatment of TTFields and AZD1152 led to a significant reduction in the number of ndGBM and rGBM cells compared to each treatment alone. Further evaluation of this approach, which has to be considered as a proof of concept, is warranted, before entering into early clinical trials.


Asunto(s)
Antineoplásicos , Glioblastoma , Humanos , Aurora Quinasa B/metabolismo , Recurrencia Local de Neoplasia , Antineoplásicos/farmacología
4.
Open Biol ; 12(10): 220197, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36196534

RESUMEN

Cytokinesis in eukaryotes is regulated by a Polo-like kinase-mediated and Aurora B kinase-mediated signalling pathway that promotes the assembly of the actomyosin contractile ring, a cytokinesis machinery conserved across evolution from yeast to humans. Trypanosoma brucei, an early divergent parasitic protozoan, employs an actomyosin-independent mechanism for its unusual cytokinesis that is controlled by a regulatory pathway comprising the Polo-like kinase TbPLK, the Aurora B kinase TbAUK1 and multiple trypanosomatid-specific regulators. However, whether any of these trypanosomatid-specific regulators function as substrates of TbPLK and/or TbAUK1 and how they cooperate with TbPLK and TbAUK1 to promote cytokinesis remain unknown. Here, we demonstrate that TbPLK and TbAUK1 phosphorylate the cytokinesis regulators CIF1 and CIF2 on multiple sites within their intrinsically disordered regions. We further show that TbPLK localization depends on its interaction with CIF1 from S/G2 phases, that TbPLK maintains CIF1 and CIF2 localization from G2 phase until early mitosis, and that TbAUK1 maintains CIF1 and CIF2 localization from late mitosis. Finally, we demonstrate that the cytokinesis regulators CIF4 and FPRC are not substrates of TbPLK and TbAUK1, and that they function upstream of TbPLK and TbAUK1 in the cytokinesis regulatory pathway. Together, these results provide insights into the functional interplay and the order of actions between the two protein kinases and the trypanosomatid-specific cytokinesis regulators in T. brucei.


Asunto(s)
Trypanosoma brucei brucei , Actomiosina/metabolismo , Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Citocinesis/fisiología , Humanos , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Trypanosoma brucei brucei/metabolismo
5.
Bioessays ; 44(5): e2100246, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35261042

RESUMEN

Correct chromosome segregation in mitosis relies on chromosome biorientation, in which sister kinetochores attach to microtubules from opposite spindle poles prior to segregation. To establish biorientation, aberrant kinetochore-microtubule interactions must be resolved through the error correction process. During error correction, kinetochore-microtubule interactions are exchanged (swapped) if aberrant, but the exchange must stop when biorientation is established. In this article, we discuss recent findings in budding yeast, which have revealed fundamental molecular mechanisms promoting this "swap and stop" process for error correction. Where relevant, we also compare the findings in budding yeast with mechanisms in higher eukaryotes. Evidence suggests that Aurora B kinase differentially regulates kinetochore attachments to the microtubule end and its lateral side and switches relative strength of the two kinetochore-microtubule attachment modes, which drives the exchange of kinetochore-microtubule interactions to resolve aberrant interactions. However, Aurora B kinase, recruited to centromeres and inner kinetochores, cannot reach its targets at kinetochore-microtubule interface when tension causes kinetochore stretching, which stops the kinetochore-microtubule exchange once biorientation is established.


Asunto(s)
Cinetocoros , Saccharomycetales , Aurora Quinasa B/genética , Segregación Cromosómica , Microtúbulos/fisiología , Mitosis
6.
Cancer Cell ; 39(9): 1245-1261.e6, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34388376

RESUMEN

The clinical success of EGFR inhibitors in EGFR-mutant lung cancer is limited by the eventual development of acquired resistance. We hypothesize that enhancing apoptosis through combination therapies can eradicate cancer cells and reduce the emergence of drug-tolerant persisters. Through high-throughput screening of a custom library of ∼1,000 compounds, we discover Aurora B kinase inhibitors as potent enhancers of osimertinib-induced apoptosis. Mechanistically, Aurora B inhibition stabilizes BIM through reduced Ser87 phosphorylation, and transactivates PUMA through FOXO1/3. Importantly, osimertinib resistance caused by epithelial-mesenchymal transition (EMT) activates the ATR-CHK1-Aurora B signaling cascade and thereby engenders hypersensitivity to respective kinase inhibitors by activating BIM-mediated mitotic catastrophe. Combined inhibition of EGFR and Aurora B not only efficiently eliminates cancer cells but also overcomes resistance beyond EMT.


Asunto(s)
Acrilamidas/farmacología , Compuestos de Anilina/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Aurora Quinasa B/antagonistas & inhibidores , Proteína 11 Similar a Bcl2/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología
7.
Int J Mol Sci ; 22(6)2021 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-33804687

RESUMEN

Cell proliferation and sexual reproduction require the faithful segregation of chromosomes. Chromosome segregation is driven by the interaction of chromosomes with the spindle, and the attachment of chromosomes to the proper spindle poles is essential. Initial attachments are frequently erroneous due to the random nature of the attachment process; however, erroneous attachments are selectively eliminated. Proper attachment generates greater tension at the kinetochore than erroneous attachments, and it is thought that attachment selection is dependent on this tension. However, studies of meiotic chromosome segregation suggest that attachment elimination cannot be solely attributed to tension, and the precise mechanism of selective elimination of erroneous attachments remains unclear. During attachment elimination, chromosomes oscillate between the spindle poles. A recent study on meiotic chromosome segregation in fission yeast has suggested that attachment elimination is coupled to chromosome oscillation. In this review, the possible contribution of chromosome oscillation in the elimination of erroneous attachment is discussed in light of the recent finding.


Asunto(s)
Segregación Cromosómica , Cinetocoros/metabolismo , Meiosis , Microtúbulos/metabolismo , Mitosis , Animales , Centrómero/genética , Centrómero/metabolismo , Humanos , Modelos Biológicos , Unión Proteica , Huso Acromático
8.
Bioorg Med Chem Lett ; 30(23): 127556, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32941989

RESUMEN

A series of novel 4-substituted phthalazinones as Aurora B kinase inhibitors was synthesized and evaluated the anti-proliferative activities against A549, HCT116, MCF-7 and HepG2 cells. 1-(4-(2-((4-Oxo-3,4-dihydrophthalazin-1-yl)amino)ethyl) phenyl)-3-(3-(trifluoromethyl)phenyl)urea (17b) exhibited the most potent anti-proliferative activity against HCT116 cells with IC50 value of 4.35 ± 1.21 µM, as well as the moderate Aurora B inhibitory activity with the IC50 value of 142 nM. Furthermore, 17b inhibited the phosphorylation of Aurora B on Thr232, leading to cell cycle arrest in the G2/M phase by down-regulating the expression of CyclinB1 and Cdc2 proteins, and apoptosis by up-regulating the expression of BAD and Bax proteins in HCT116 cells. In addition, a docking study revealed that 17b could form key hydrogen bonds with Ala173, Glu171 and Glu177 in Aurora B. All the results reveal that 17b is worthy of further development as an Aurora B kinase inhibitor.


Asunto(s)
Antineoplásicos/farmacología , Aurora Quinasa B/antagonistas & inhibidores , Ftalazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Aurora Quinasa B/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Pruebas de Enzimas , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Fosforilación/efectos de los fármacos , Ftalazinas/síntesis química , Ftalazinas/metabolismo , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Relación Estructura-Actividad
9.
Clin Exp Hepatol ; 6(2): 77-84, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32728623

RESUMEN

AIM OF THE STUDY: Microwave ablation (MWA) for treatment of hepatocellular carcinoma (HCC) is a new promising modality. The prognosis after treatment is mainly linked to the recurrence. We aimed to investigate the predictive value of α-fetoprotein (AFP) score and Aurora B kinase (AURKB) in HCC recurrence after MWA. MATERIAL AND METHODS: A cross-sectional study where 25 early-stage HCC patients (Barcelona Clinic Liver Cancer 0/A-B) were treated with MWA. Tumor biopsies were obtained just prior to MWA and assessed for WHO pathological grade and AURKB expression by immunohistochemistry. AFP score was calculated and a cut-off value of 2 classifies patients into high and low risk of recurrence. After achieving complete ablation, patients were followed every 3 months for 1 year by triphasic CT to detect recurrence. RESULTS: Child-Pugh classification has no significant impact on prognosis of HCC after MWA (χ2 = 1.924, p = 0.165). Serum AFP level and AFP score can effectively predict the response to MWA among HCC patients (χ2 = 6.451, MC p = 0.031) (χ2 = 9.0, p = 0.003), respectively. AFP score was strongly associated with the pathological grade of the tumor (r = 0.467, p = 0.019). AURKB was over-expressed in tumoral more than non-tumoral specimens (p < 0.001). It was correlated with the size of the tumor, the number of tumor nodules and the pathological grade of the tumor (p < 0.05) but has no role in predicting recurrence after MWA (p = 0.869). CONCLUSIONS: AFP score but not AURKB can predict the risk of recurrence of HCC after MWA.

10.
Cancer Chemother Pharmacol ; 85(4): 773-783, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32144432

RESUMEN

PURPOSE: Acute lymphoblastic leukemia (ALL) is curable with standardized chemotherapy. However, the development of novel therapies is still required, especially for patients with relapsed or refractory disease. By utilizing an in vitro drug screening system, active molecular targeting agents against ALL were explored in this study. METHODS: By the in vitro drug sensitivity test, 81 agents with various actions were screened for their cytotoxicity in a panel of 22 ALL cell lines and ALL clinical samples. The drug effect score (DES) was calculated from the dose-response of each drug for comparison among drugs or samples. Normal peripheral blood mononuclear cells were also applied onto the drug screening to provide the reference control values. The drug combination effect was screened based on the Bliss independent model, and validated by the improved isobologram method. RESULTS: On sensitivity screening in a cell line panel, barasertib-HQPA which is an active metabolite of barasertib, an aurora B kinase inhibitor, alisertib, an aurora A kinase inhibitor, and YM155, a survivin inhibitor, were effective against the broadest range of ALL cells. The DES of barasertib-HQPA was significantly higher in ALL clinical samples compared to the reference value. There were significant correlations in DES between barasertib-HQPA and vincristine or docetaxel. In the drug combination assay, barasertib-HQPA and eribulin showed additive to synergistic effects. CONCLUSION: Aurora B kinase was identified to be an active therapeutic target in a broad range of ALL cells. Combination therapy of barasertib and a microtubule-targeting drug is of clinical interest.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Aurora Quinasa A/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Quinazolinas/farmacología , Ciclo Celular , Proliferación Celular , Docetaxel/administración & dosificación , Quimioterapia Combinada , Furanos/administración & dosificación , Ensayos Analíticos de Alto Rendimiento , Humanos , Cetonas/administración & dosificación , Fosforilación , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico , Células Tumorales Cultivadas , Vincristina/administración & dosificación
11.
Cancer Cell ; 37(2): 200-215.e5, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-32049046

RESUMEN

Deregulation of MYC plays an essential role in T cell acute lymphoblastic leukemia (T-ALL), yet the mechanisms underlying its deregulation remain elusive. Herein, we identify a molecular mechanism responsible for reciprocal activation between Aurora B kinase (AURKB) and MYC. AURKB directly phosphorylates MYC at serine 67, counteracting GSK3ß-directed threonine 58 phosphorylation and subsequent FBXW7-mediated proteasomal degradation. Stabilized MYC, in concert with T cell acute lymphoblastic leukemia 1 (TAL1), directly activates AURKB transcription, constituting a positive feedforward loop that reinforces MYC-regulated oncogenic programs. Therefore, inhibitors of AURKB induce prominent MYC degradation concomitant with robust leukemia cell death. These findings reveal an AURKB-MYC regulatory circuit that underlies T cell leukemogenesis, and provide a rationale for therapeutic targeting of oncogenic MYC via AURKB inhibition.


Asunto(s)
Aurora Quinasa B/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/inmunología , Linfocitos T/inmunología , Animales , Aurora Quinasa A/genética , Aurora Quinasa A/inmunología , Aurora Quinasa B/inmunología , Línea Celular Tumoral , Proteína 7 que Contiene Repeticiones F-Box-WD/inmunología , Humanos , Ratones , Fosforilación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/inmunología , Pez Cebra
12.
Development ; 147(1)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31806662

RESUMEN

Although cytokinesis has been intensely studied, the way it is executed during development is not well understood, despite a long-standing appreciation that various aspects of cytokinesis vary across cell and tissue types. To address this, we investigated cytokinesis during the invariant Caenorhabditis elegans embryonic divisions and found several parameters that are altered at different stages in a reproducible manner. During early divisions, furrow ingression asymmetry and midbody inheritance is consistent, suggesting specific regulation of these events. During morphogenesis, we found several unexpected alterations to cytokinesis, including apical midbody migration in polarizing epithelial cells of the gut, pharynx and sensory neurons. Aurora B kinase, which is essential for several aspects of cytokinesis, remains apically localized in each of these tissues after internalization of midbody ring components. Aurora B inactivation disrupts cytokinesis and causes defects in apical structures, even if inactivated post-mitotically. Therefore, we demonstrate that cytokinesis is implemented in a specialized way during epithelial polarization and that Aurora B has a role in the formation of the apical surface.


Asunto(s)
Aurora Quinasa B/fisiología , Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/embriología , Caenorhabditis elegans/enzimología , Citocinesis , Morfogénesis , Animales , Caenorhabditis elegans/citología , Polaridad Celular , Citocinesis/fisiología , Dendritas/fisiología , Embrión no Mamífero/citología , Células Epiteliales/fisiología , Intestinos/embriología , Neuronas/citología , Faringe/embriología , Propiedades de Superficie
13.
Biology (Basel) ; 8(3)2019 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-31357447

RESUMEN

The primary goal of cytokinesis is to produce two daughter cells, each having a full set of chromosomes. To achieve this, cells assemble a dynamic structure between segregated sister chromatids called the contractile ring, which is made up of filamentous actin, myosin-II, and other regulatory proteins. Constriction of the actomyosin ring generates a cleavage furrow that divides the cytoplasm to produce two daughter cells. Decades of research have identified key regulators and underlying molecular mechanisms; however, many fundamental questions remain unanswered and are still being actively investigated. This review summarizes the key findings, computational modeling, and recent advances in understanding of the molecular mechanisms that control the formation of the cleavage furrow and cytokinesis.

14.
Cell Cycle ; 18(17): 2006-2025, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31306061

RESUMEN

A single inner centromere protein (INCENP) found throughout eukaryotes modulates Aurora B kinase activity and chromosomal passenger complex (CPC) localization, which is essential for timely mitotic progression. It has been proposed that INCENP might act as a rheostat to regulate Aurora B activity through mitosis, with successively higher activity threshold levels for chromosome alignment, the spindle checkpoint, anaphase spindle transfer and finally spindle elongation and cytokinesis. It remains mechanistically unclear how this would be achieved. Here, we reveal that the urochordate, Oikopleura dioica, possesses two INCENP paralogs, which display distinct localizations and subfunctionalization in order to complete M-phase. INCENPa was localized on chromosome arms and centromeres by prometaphase, and modulated Aurora B activity to mediate H3S10/S28 phosphorylation, chromosome condensation, spindle assembly and transfer of the CPC to the central spindle. Polo-like kinase (Plk1) recruitment to CDK1 phosphorylated INCENPa was crucial for INCENPa-Aurora B enrichment on centromeres. The second paralog, INCENPb was enriched on centromeres from prometaphase, and relocated to the central spindle at anaphase onset. In the absence of INCENPa, meiotic spindles failed to form, and homologous chromosomes did not segregate. INCENPb was not required for early to mid M-phase events but became essential for the activity and localization of Aurora B on the central spindle and midbody during cytokinesis in order to allow abscission to occur. Together, our results demonstrate that INCENP paralog switching on centromeres modulates Aurora B kinase localization, thus chronologically regulating CPC functions during fast embryonic divisions in the urochordate O. dioica. Abbreviations: CCAN: constitutive centromere-associated network; CENPs: centromere proteins; cmRNA: capped messenger RNA; CPC: chromosomal passenger complex; INCENP: inner centromere protein; Plk1: polo-like kinase 1; PP1: protein phosphatase 1; PP2A: protein phosphatase 2A; SAC: spindle assembly checkpoint; SAH: single α-helix domain.


Asunto(s)
Aurora Quinasa B/genética , Proteínas Cromosómicas no Histona/genética , Cromosomas/genética , Mitosis/genética , Proteína Quinasa CDC2/genética , Proteínas de Ciclo Celular/genética , Segregación Cromosómica/genética , Citocinesis/genética , Humanos , Cinetocoros/metabolismo , Fosforilación/genética , Plancton/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Huso Acromático/genética , Quinasa Tipo Polo 1
15.
Curr Biol ; 29(12): 1999-2008.e4, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31178324

RESUMEN

The large length scale of Xenopus laevis eggs facilitates observation of bulk cytoplasm dynamics far from the cortex during cytokinesis. The first furrow ingresses through the egg midplane, which is demarcated by chromosomal passenger complex (CPC) localized on microtubule bundles at the boundary between asters. Using an extract system, we found that local kinase activity of the Aurora B kinase (AURKB) subunit of the CPC caused disassembly of F-actin and keratin between asters and local softening of the cytoplasm as assayed by flow patterns. Beads coated with active CPC mimicked aster boundaries and caused AURKB-dependent disassembly of F-actin and keratin that propagated ∼40 µm without microtubules and much farther with microtubules present. Consistent with extract observations, we observed disassembly of the keratin network between asters in zygotes fixed before and during 1st cytokinesis. We propose that active CPC at aster boundaries locally reduces cytoplasmic stiffness by disassembling actin and keratin networks. Possible functions of this local disassembly include helping sister centrosomes move apart after mitosis, preparing a soft path for furrow ingression, and releasing G-actin from internal networks to build cortical networks that support furrow ingression.


Asunto(s)
Actinas/metabolismo , Aurora Quinasa B/metabolismo , Queratinas/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Microtúbulos , Óvulo/crecimiento & desarrollo , Huso Acromático , Xenopus laevis/embriología
16.
Cell Rep ; 26(11): 2875-2889.e3, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30865880

RESUMEN

The segregation of chromosomes is a critical step during cell division. This process is driven by the elongation of spindle microtubules and is tightly regulated by checkpoint mechanisms. It is unknown whether microtubules affect checkpoint responses as passive contributors or active regulators of the process. We show here that interphase microtubules are essential to temporally restrict the effects of DNA replication stress to S phase in Saccharomyces cerevisiae. Tubulin mutants hypersensitive to DNA damage experience a strong but delayed mitotic checkpoint arrest after exposure to genotoxic stress in S phase. This untimely arrest is dependent on the Aurora B kinase but, surprisingly, not on the DNA damage checkpoint. Impaired microtubule-kinetochore interaction is the apparent cause for this unusual phenotype. Collectively, our results reveal that core components of microtubules potentiate the detection of DNA lesions created in S phase, thereby suppressing untimely activation of mitotic checkpoints after DNA replication stress.


Asunto(s)
Aurora Quinasa B/genética , Replicación del ADN , Interfase , Microtúbulos/metabolismo , Mitosis , Aurora Quinasa B/metabolismo , Daño del ADN , Cinetocoros/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
Elife ; 82019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30758285

RESUMEN

Microtubules (MTs) are essential for cleavage furrow positioning during cytokinesis, but the mechanisms by which MT-derived signals spatially define regions of cortical contractility are unresolved. In this study cytokinesis regulators visualized in Drosophila melanogaster (Dm) cells were found to localize to and track MT plus-ends during cytokinesis. The RhoA GEF Pebble (Dm ECT2) did not evidently tip-track, but rather localized rapidly to cortical sites contacted by MT plus-tips, resulting in RhoA activation and enrichment of myosin-regulatory light chain. The MT plus-end localization of centralspindlin was compromised following EB1 depletion, which resulted in a higher incidence of cytokinesis failure. Centralspindlin plus-tip localization depended on the C-terminus and a putative EB1-interaction motif (hxxPTxh) in RacGAP50C. We propose that MT plus-end-associated centralspindlin recruits a cortical pool of Dm ECT2 upon physical contact to activate RhoA and to trigger localized contractility.


Asunto(s)
Citocinesis , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Microtúbulos/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo , Secuencias de Aminoácidos , Anafase/efectos de los fármacos , Animales , Concanavalina A/farmacología , Citocinesis/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Microtúbulos/efectos de los fármacos , Miosinas/metabolismo , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
18.
J Biochem ; 165(5): 433-446, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30590679

RESUMEN

Heterochromatin protein 1 (HP1) is an evolutionarily conserved chromosomal protein that plays a crucial role in heterochromatin-mediated gene silencing. We previously showed that mammalian HP1α is constitutively phosphorylated at its N-terminal serine residues by casein kinase II (CK2), and that this phosphorylation enhances HP1α's binding specificity for nucleosomes containing lysine 9-methylated histone H3 (H3K9me). Although the presence of additional HP1α phosphorylation during mitosis was reported more than a decade ago, its biological significance remains largely elusive. Here we found that mitosis-specific HP1α phosphorylation affected HP1α's ability to bind chromatin. Using biochemical and mutational analyses, we showed that HP1α's mitotic phosphorylation was located in its hinge region and was reversibly regulated by Aurora B kinase and serine/threonine phosphatases. In addition, chromatin fractionation and electrophoretic mobility shift assays revealed that hinge region-phosphorylated HP1α was preferentially dissociated from mitotic chromatin and exhibited a reduced DNA-binding activity. Although HP1's mitotic behaviour was previously linked to H3 serine 10 phosphorylation, which blocks the binding of HP1's chromodomain to H3K9me3, our findings suggest that mitotic phosphorylation in HP1α's hinge region also contributes to changes in HP1α's association with mitotic chromatin.


Asunto(s)
Ciclo Celular , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Mitosis , Aurora Quinasa B/metabolismo , Homólogo de la Proteína Chromobox 5 , ADN/metabolismo , Células HEK293 , Células HeLa , Histonas/metabolismo , Humanos , Fosforilación , Unión Proteica , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2C/metabolismo
20.
Elife ; 72018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30044223

RESUMEN

Proper kinetochore-microtubule attachments, mediated by the NDC80 complex, are required for error-free chromosome segregation. Erroneous attachments are corrected by the tension dependence of kinetochore-microtubule interactions. Here, we present a method, based on fluorescence lifetime imaging microscopy and Förster resonance energy transfer, to quantitatively measure the fraction of NDC80 complexes bound to microtubules at individual kinetochores in living human cells. We found that NDC80 binding is modulated in a chromosome autonomous fashion over prometaphase and metaphase, and is predominantly regulated by centromere tension. We show that this tension dependency requires phosphorylation of the N-terminal tail of Hec1, a component of the NDC80 complex, and the proper localization of Aurora B kinase, which modulates NDC80 binding. Our results lead to a mathematical model of the molecular basis of tension-dependent NDC80 binding to kinetochore microtubules in vivo.


Asunto(s)
Cinetocoros/metabolismo , Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Aurora Quinasa B/metabolismo , Calibración , Línea Celular Tumoral , Simulación por Computador , Proteínas del Citoesqueleto , Transferencia Resonante de Energía de Fluorescencia , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metafase , Modelos Biológicos , Método de Montecarlo , Proteínas Serina-Treonina Quinasas/metabolismo , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA