Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Cell ; 137(4): 672-84, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19450515

RESUMO

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.


Assuntos
Cinetocoros/química , Cinetocoros/metabolismo , Microtúbulos/química , Microtúbulos/metabolismo , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Metáfase , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Nucleares
2.
J Pathol ; 245(4): 502-513, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29862509

RESUMO

Hepatitis B virus (HBV) is an aetiological factor for liver cirrhosis and hepatocellular carcinoma (HCC). Despite current antiviral therapies that successfully reduce the viral load in patients with chronic hepatitis B, persistent hepatitis B surface antigen (HBsAg) remains a risk factor for HCC. To explore whether intrahepatic viral antigens contribute directly to hepatocarcinogenesis, we monitored the mitotic progression of HBV-positive cells. Cytokinesis failure was increased in HBV-positive HepG2.2.15 and 1.3ES2 cells, as well as in HuH-7 cells transfected with a wild-type or X-deficient HBV construct, but not in cells transfected with an HBsAg-deficient construct. We show that expression of viral large surface antigen (LHBS) was sufficient to induce cytokinesis failure of immortalized hepatocytes. Premitotic defects with DNA damage and G2 /M checkpoint attenuation preceded cytokinesis in LHBS-positive cells, and ultimately resulted in hyperploidy. Inhibition of polo-like kinase-1 (Plk1) not only restored the G2 /M checkpoint in these cells, but also suppressed LHBS-mediated in vivo tumourigenesis. Finally, a positive correlation between intrahepatic LHBS expression and hepatocyte hyperploidy was detected in >70% of patients with chronic hepatitis B. We conclude that HBV LHBS provokes hyperploidy by inducing DNA damage and upregulation of Plk1; the former results in atypical chromatin structures, and the latter attenuates the function of the G2 /M DNA damage checkpoint. Our data uncover a mechanism by which genomic integrity of hepatocytes is disrupted by viral LHBS. These findings highlight the role of intrahepatic surface antigen as an oncogenic risk factor in the development of HCC. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Carcinoma Hepatocelular/virologia , Citocinese , Antígenos de Superfície da Hepatite B/metabolismo , Vírus da Hepatite B/metabolismo , Hepatite B Crônica/virologia , Hepatócitos/virologia , Neoplasias Hepáticas/virologia , Ploidias , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular/metabolismo , Transformação Celular Viral , Dano ao DNA , Modelos Animais de Doenças , Pontos de Checagem da Fase G2 do Ciclo Celular , Células Hep G2 , Antígenos de Superfície da Hepatite B/genética , Vírus da Hepatite B da Marmota/genética , Vírus da Hepatite B da Marmota/metabolismo , Vírus da Hepatite B/genética , Hepatite B Crônica/genética , Hepatite B Crônica/metabolismo , Hepatite B Crônica/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/transplante , Interações Hospedeiro-Patógeno , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Marmota , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Quinase 1 Polo-Like
3.
Proc Natl Acad Sci U S A ; 111(33): 12019-24, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-25092294

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Transporte/fisiologia , Proteínas de Ciclo Celular/fisiologia , Mitose , Proteínas Nucleares/fisiologia , ATPases Associadas a Diversas Atividades Celulares , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Cdc20/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células HeLa , Humanos , Proteínas Mad2/metabolismo , Proteínas Nucleares/metabolismo , Ligação Proteica
4.
Mol Cell ; 29(6): 729-41, 2008 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-18374647

RESUMO

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.


Assuntos
Ciclo Celular/fisiologia , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cinetocoros/metabolismo , Mitose/fisiologia , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Cisteína Endopeptidases/metabolismo , DNA Topoisomerases/metabolismo , Genes Reporter , Células HeLa , Humanos , Cinética , Metáfase , Ligação Proteica
5.
J Biol Chem ; 289(34): 23928-37, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25012665

RESUMO

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.


Assuntos
Proteínas de Transporte/fisiologia , Mitose/fisiologia , ATPases Associadas a Diversas Atividades Celulares , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/metabolismo , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Proteínas Mad2/metabolismo , Microscopia de Fluorescência , Proteínas Nucleares/metabolismo , Interferência de RNA
7.
J Biol Chem ; 286(24): 21173-9, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21525009

RESUMO

The mitotic checkpoint maintains genomic stability by ensuring that chromosomes are accurately segregated during mitosis. When the checkpoint is activated, the mitotic checkpoint complex (MCC), assembled from BUBR1, BUB3, CDC20, and MAD2, directly binds and inhibits the anaphase-promoting complex/cyclosome (APC/C) until all chromosomes are properly attached and aligned. The mechanisms underlying MCC assembly and MCC-APC/C interaction are not well characterized. Here, we show that a novel interaction between BUBR1 and closed MAD2 (C-MAD2) is essential for MCC-mediated inhibition of APC/C. Intriguingly, Arg(133) and Gln(134) in C-MAD2 are required for BUBR1 interaction. The same residues are also critical for MAD2 dimerization and MAD2 binding to p31(comet), a mitotic checkpoint silencing protein. Along with previously characterized BUBR1-CDC20 and C-MAD2-CDC20 interactions, our results underscore the integrity of the MCC for its activity and suggest the fundamental importance of the MAD2 αC helix in modulating mitotic checkpoint activation and silencing.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Regulação da Expressão Gênica , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Anáfase , Ciclossomo-Complexo Promotor de Anáfase , Dimerização , Inativação Gênica , Células HeLa , Humanos , Proteínas Mad2 , Masculino , Próstata/metabolismo , Fuso Acromático/metabolismo , Transfecção , Ubiquitina-Proteína Ligases/metabolismo
8.
J Cell Biol ; 177(3): 413-24, 2007 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-17485487

RESUMO

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.


Assuntos
Anáfase/fisiologia , Proteínas de Ciclo Celular/metabolismo , Cinesinas/metabolismo , Cinetocoros/metabolismo , Aurora Quinase B , Aurora Quinases , Células HeLa , Humanos , Cinetocoros/ultraestrutura , Fosfoproteínas Fosfatases/metabolismo , Ligação Proteica/fisiologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico/fisiologia
9.
Nat Cell Biol ; 5(4): 341-5, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12640463

RESUMO

The kinetochore, a macromolecular complex located at the centromere of chromosomes, provides essential functions for accurate chromosome segregation. Kinetochores contain checkpoint proteins that monitor attachments between the kinetochore and microtubules to ensure that cells do not exit mitosis in the presence of unaligned chromosomes. Here we report that human CENP-I, a constitutive protein of the kinetochore that shares limited similarity with Mis6 of Schizosaccharomyces pombe, is required for the localization of CENP-F and the checkpoint proteins MAD1 and MAD2 to kinetochores. Depletion of CENP-I from kinetochores causes the cell cycle to delay in G2. Although monopolar chromosomes in CENP-I-depleted cells fail to establish bipolar connections, the cells are unable to arrest in mitosis. These cells are transiently delayed in mitosis in a MAD2-dependent manner, even though their kinetochores are depleted of MAD2. The delay is extended considerably when the number of unattached kinetochores is increased. This suggests that no single unattached kinetochore in CENP-I-depleted cells can arrest mitosis. The collective output from many unattached kinetochores is required to reach a threshold signal of 'wait for anaphase' to sustain a prolonged mitotic arrest.


Assuntos
Proteínas de Transporte , Núcleo Celular/genética , Proteínas de Ligação a DNA/genética , Células Eucarióticas/metabolismo , Genes cdc/fisiologia , Cinetocoros/metabolismo , Mitose/genética , Antineoplásicos/farmacologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/deficiência , Células Eucarióticas/citologia , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Células HeLa , Humanos , Proteínas Mad2 , Proteínas dos Microfilamentos , Microtúbulos/genética , Microtúbulos/metabolismo , Nocodazol/farmacologia , Proteínas Nucleares , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transporte Proteico/genética , RNA Interferente Pequeno/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas de Schizosaccharomyces pombe
10.
J Cell Biol ; 175(1): 41-53, 2006 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-17030981

RESUMO

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).


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Cinetocoros/metabolismo , Modelos Genéticos , Aurora Quinase B , Aurora Quinases , Autoantígenos/metabolismo , Autoantígenos/fisiologia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Proteína Centromérica A , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Células HeLa , Humanos , Cinetocoros/ultraestrutura , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia
11.
Proc Natl Acad Sci U S A ; 105(27): 9181-5, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18591651

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mitose , Complexos Ubiquitina-Proteína Ligase/antagonistas & inibidores , Ciclossomo-Complexo Promotor de Anáfase , Proteínas Cdc20 , Células HeLa , Humanos , Fatores de Tempo
12.
Mol Cancer Res ; 19(10): 1651-1665, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34158391

RESUMO

Nestin, a class IV intermediate filament protein, is generally considered as a putative marker of neural stem and progenitor cells in the central nervous system. Glioma is a common type of adult brain tumors, and glioblastoma (GBM) represents the most aggressive form of glioma. Here, we report that Nestin expression is significantly upregulated in human GBM, compared with other types of glioma. Nestin knockdown or deletion in U251 cells and tumor cells from GBM patients derived xenografts resulted in G2-M arrest, finally leading to apoptosis in tumor cells. Using proximity-dependent biotin identification method, we identified ßII-tubulin as an interacting protein of Nestin in U251 cells. Nestin stabilized ßII-tubulin in U251 cells through physical interaction. Knockdown of Nestin or ßII-tubulin disrupted spindle morphology in tumor cells. Our studies further revealed that Nestin deficiency in U251 cells and GBM PDX cells repressed tumor growth upon transplantation. Finally, we found that Nestin deficiency sensitized GBM cells to microtubule-destabilizing drugs such as vinblastine and vincristine. Our studies demonstrate the essential functions and underlying mechanisms of Nestin in the growth and drug response of GBM cells. IMPLICATIONS: Through interaction with ßII-tubulin, Nestin facilitates cell-cycle progression and spindle assembly of tumor cells in glioblastoma.


Assuntos
Neoplasias Encefálicas/metabolismo , Ciclo Celular/fisiologia , Glioblastoma/metabolismo , Nestina/metabolismo , Fuso Acromático/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Glioma/metabolismo , Células HEK293 , Humanos , Camundongos Nus , Camundongos SCID , Tubulina (Proteína)/metabolismo
13.
Trends Cell Biol ; 15(11): 589-98, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16214339

RESUMO

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.


Assuntos
Cinetocoros/fisiologia , Ciclossomo-Complexo Promotor de Anáfase , Animais , Proteínas de Ciclo Celular/fisiologia , Segregação de Cromossomos/fisiologia , Humanos , Cinetocoros/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/metabolismo , Modelos Biológicos , Ligação Proteica , Complexos Ubiquitina-Proteína Ligase/fisiologia
14.
J Cell Biol ; 160(7): 1017-27, 2003 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-12668657

RESUMO

Anumber of proteins are recruited to nuclear foci upon exposure to double-strand DNA damage, including 53BP1 and Rad51, but the precise role of these DNA damage-induced foci remain unclear. Here we show in a variety of human cell lines that histone deacetylase (HDAC) 4 is recruited to foci with kinetics similar to, and colocalizes with, 53BP1 after exposure to agents causing double-stranded DNA breaks. HDAC4 foci gradually disappeared in repair-proficient cells but persisted in repair-deficient cell lines or cells irradiated with a lethal dose, suggesting that resolution of HDAC4 foci is linked to repair. Silencing of HDAC4 via RNA interference surprisingly also decreased levels of 53BP1 protein, abrogated the DNA damage-induced G2 delay, and radiosensitized HeLa cells. Our combined results suggest that HDAC4 is a critical component of the DNA damage response pathway that acts through 53BP1 and perhaps contributes in maintaining the G2 cell cycle checkpoint.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA , Histona Desacetilases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Nucleares/metabolismo , Fosfoproteínas , Proteínas Repressoras/metabolismo , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Reparo do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta à Radiação , Etoposídeo/farmacologia , Fase G2 , Raios gama/efeitos adversos , Histona Desacetilases/efeitos dos fármacos , Histona Desacetilases/efeitos da radiação , Humanos , Ácidos Hidroxâmicos/farmacologia , Cinética , Mutação , Proteínas Nucleares/genética , Inibidores da Síntese de Ácido Nucleico/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/efeitos dos fármacos , Proteínas Repressoras/efeitos da radiação , Células Tumorais Cultivadas , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
15.
Biochem Biophys Res Commun ; 370(2): 213-9, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18361916

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Fuso Acromático/enzimologia , Aurora Quinase A , Aurora Quinases , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/genética , Centrossomo/enzimologia , Epistasia Genética , Células HeLa , Humanos , Mitose/efeitos dos fármacos , Mitose/genética , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , Saccharomyces cerevisiae/genética , Técnicas do Sistema de Duplo-Híbrido
16.
Mol Biol Cell ; 16(3): 1043-55, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15616196

RESUMO

The temporal and spatial regulation of cytokinesis requires an interaction between the anaphase mitotic spindle and the cell cortex. However, the relative roles of the spindle asters or the central spindle bundle are not clear in mammalian cells. The central spindle normally serves as a platform to localize key regulators of cell cleavage, including passenger proteins. Using time-lapse and immunofluorescence analysis, we have addressed the consequences of eliminating the central spindle by ablation of PRC1, a microtubule bundling protein that is critical to the formation of the central spindle. Without a central spindle, the asters guide the equatorial cortical accumulation of anillin and actin, and of the passenger proteins, which organize into a subcortical ring in anaphase. Furrowing goes to completion, but abscission to create two daughter cells fails. We conclude the central spindle bundle is required for abscission but not for furrowing in mammalian cells.


Assuntos
Proteínas de Ciclo Celular/fisiologia , RNA Interferente Pequeno/genética , Actinas/genética , Anáfase , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas Contráteis/genética , Citocinese , Citoesqueleto/metabolismo , Células HeLa , Humanos , Microscopia de Fluorescência , Microscopia de Contraste de Fase , Microscopia de Vídeo , Microtúbulos/metabolismo , Mitose , RNA Interferente Pequeno/metabolismo , Fuso Acromático , Fatores de Tempo
17.
Curr Biol ; 14(7): 611-7, 2004 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-15062103

RESUMO

RanGAP1 is the activating protein for the Ran GTPase. Vertebrate RanGAP1 is conjugated to a small ubiquitin-like protein, SUMO-1. This modification promotes association of RanGAP1 with the interphase nuclear pore complex (NPC) through binding to the nucleoporin RanBP2, also known as Nup358. During mitosis, RanGAP1 is concentrated at kinetochores in a microtubule- (MT) and SUMO-1-dependent fashion. RanBP2 is also abundantly found on kinetochores in mitosis. Here we show that ablation of proteins required for MT-kinetochore attachment (Hec1/Ndc80, Nuf2 ) disrupts RanGAP1 and RanBP2 targeting to kinetochores. No similar disruption was observed after ablation of proteins nonessential for MT-kinetochore interactions (CENP-I, Bub1, CENP-E ). Acquisition of RanGAP1 and RanBP2 by kinetochores is temporally correlated in untreated cells with MT attachment. These patterns of accumulation suggest a loading mechanism wherein the RanGAP1-RanBP2 complex may be transferred along the MT onto the kinetochore. Depletion of RanBP2 caused mislocalization of RanGAP1, Mad1, Mad2, CENP-E, and CENP-F, as well as loss of cold-stable kinetochore-MT interactions and accumulation of mitotic cells with multipolar spindles and unaligned chromosomes. Taken together, our observations indicate that RanBP2 and RanGAP1 are targeted as a single complex that is both regulated by and essential for stable kinetochore-MT association.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Mitose/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Primers do DNA , Células HeLa , Humanos , Indóis , Microscopia de Fluorescência , Chaperonas Moleculares , Interferência de RNA , Proteína SUMO-1/metabolismo
18.
Mol Biol Cell ; 14(4): 1379-91, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12686595

RESUMO

The Zeste-White 10 (ZW10) and Rough Deal (ROD) proteins are part of a complex necessary for accurate chromosome segregation. This complex recruits cytoplasmic dynein to the kinetochore and participates in the spindle checkpoint. We used immunoaffinity chromatography and mass spectroscopy to identify the Drosophila proteins in this complex. We found that the complex contains an additional protein we name Zwilch. Zwilch localizes to kinetochores and kinetochore microtubules in a manner identical to ZW10 and ROD. We have also isolated a zwilch mutant, which exhibits the same mitotic phenotypes associated with zw10 and rod mutations: lagging chromosomes at anaphase and precocious sister chromatid separation upon activation of the spindle checkpoint. Zwilch's role within the context of this complex is evolutionarily conserved. The human Zwilch protein (hZwilch) coimmunoprecipitates with hZW10 and hROD from HeLa cell extracts and localizes to the kinetochores at prometaphase. Finally, we discuss immunoaffinity chromatography results that suggest the existence of a weak interaction between the ZW10/ROD/Zwilch complex and the kinesin-like kinetochore component CENP-meta.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Cinetocoros/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Drosophila/citologia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Genes de Insetos , Células HeLa , Humanos , Imuno-Histoquímica , Substâncias Macromoleculares , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/genética , Mitose , Dados de Sequência Molecular , Mutação , Fenótipo , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Fuso Acromático/metabolismo
19.
Mol Biol Cell ; 14(4): 1638-51, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12686615

RESUMO

We have determined that the previously identified dual-specificity protein kinase TTK is the human orthologue of the yeast MPS1 kinase. Yeast MPS1 (monopolar spindle) is required for spindle pole duplication and the spindle checkpoint. Consistent with the recently identified vertebrate MPS1 homologues, we found that hMPS1 is localized to centrosomes and kinetochores. In addition, hMPS1 is part of a growing list of kinetochore proteins that are localized to nuclear pores. hMPS1 is required by cells to arrest in mitosis in response to spindle defects and kinetochore defects resulting from the loss of the kinesin-like protein, CENP-E. The pattern of kinetochore localization of hMPS1 in CENP-E defective cells suggests that their interaction with the kinetochore is sensitive to microtubule occupancy rather than kinetochore tension. hMPS1 is required for MAD1, MAD2 but not hBUB1, hBUBR1 and hROD to bind to kinetochores. We localized the kinetochore targeting domain in hMPS1 and found that it can abrogate the mitotic checkpoint in a dominant negative manner. Last, hMPS1 was found to associate with the anaphase promoting complex, thus raising the possibility that its checkpoint functions extend beyond the kinetochore.


Assuntos
Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona/metabolismo , Cinetocoros/metabolismo , Mitose/fisiologia , Proteínas Quinases , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Anáfase/fisiologia , Sequência de Bases , Centrossomo/metabolismo , Proteínas Cromossômicas não Histona/genética , Clonagem Molecular , DNA Complementar/genética , Células HeLa , Humanos , Técnicas In Vitro , Interfase/fisiologia , Poro Nuclear/metabolismo , Proteínas Nucleares , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/metabolismo , Fuso Acromático/metabolismo
20.
Mol Biol Cell ; 14(6): 2262-76, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12808028

RESUMO

The receptor for hyaluronan-mediated motility (RHAMM), an acidic coiled coil protein, has previously been characterized as a cell surface receptor for hyaluronan, and a microtubule-associated intracellular hyaluronan binding protein. In this study, we demonstrate that a subset of cellular RHAMM localizes to the centrosome and functions in the maintenance of spindle integrity. We confirm a previous study showing that the amino terminus of RHAMM interacts with microtubules and further demonstrate that a separate carboxy-terminal domain is required for centrosomal targeting. This motif overlaps the defined hyaluronan binding domain and bears 72% identity to the dynein interaction domain of Xklp2. RHAMM antibodies coimmunprecipitate dynein IC from Xenopus and HeLa extracts. Deregulation of RHAMM expression inhibits mitotic progression and affects spindle architecture. Structure, localization, and function, along with phylogenetic analysis, suggests that RHAMM may be a new member of the TACC family. Thus, we demonstrate a novel centrosomal localization and mitotic spindle-stabilizing function for RHAMM. Moreover, we provide a potential mechanism for this function in that RHAMM may cross-link centrosomal microtubules, through a direct interaction with microtubules and an association with dynein.


Assuntos
Centrossomo/metabolismo , Dineínas/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Receptores de Hialuronatos/metabolismo , Fuso Acromático/metabolismo , Mapeamento Cromossômico , Genes Reporter , Humanos , Interfase/fisiologia , Microtúbulos/metabolismo , Mitose/fisiologia , Filogenia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa