Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Chromosome Res ; 31(3): 25, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37640903

RESUMO

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


Assuntos
Aneuploidia , Aberrações Cromossômicas , Humanos , Animais , Camundongos , Cariotipagem , Cariótipo , Mamíferos
2.
Nature ; 607(7919): 604-609, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35831506

RESUMO

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


Assuntos
Aneuploidia , Posicionamento Cromossômico , Segregação de Cromossomos , Cromossomos , Proteína 9 Associada à CRISPR , Linhagem Celular , Linhagem Celular Tumoral , Segregação de Cromossomos/genética , Cromossomos/genética , Cromossomos/metabolismo , Cromotripsia , Crescimento e Desenvolvimento/genética , Humanos , Interfase , Micronúcleos com Defeito Cromossômico , Mitose , Neoplasias/genética , Neoplasias/patologia , Organoides/citologia , Organoides/metabolismo , Análise de Sequência de DNA , Análise de Célula Única
3.
Cell Rep ; 39(1): 110584, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35385742

RESUMO

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


Assuntos
Neoplasias da Mama , Neoplasias de Mama Triplo Negativas , Neoplasias da Mama/metabolismo , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Hidrocarbonetos Aromáticos com Pontes/uso terapêutico , Endossomos/metabolismo , Feminino , Humanos , Taxoides/farmacologia , Taxoides/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo
4.
Nat Genet ; 53(8): 1187-1195, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34211178

RESUMO

Central to tumor evolution is the generation of genetic diversity. However, the extent and patterns by which de novo karyotype alterations emerge and propagate within human tumors are not well understood, especially at single-cell resolution. Here, we present 3D Live-Seq-a protocol that integrates live-cell imaging of tumor organoid outgrowth and whole-genome sequencing of each imaged cell to reconstruct evolving tumor cell karyotypes across consecutive cell generations. Using patient-derived colorectal cancer organoids and fresh tumor biopsies, we demonstrate that karyotype alterations of varying complexity are prevalent and can arise within a few cell generations. Sub-chromosomal acentric fragments were prone to replication and collective missegregation across consecutive cell divisions. In contrast, gross genome-wide karyotype alterations were generated in a single erroneous cell division, providing support that aneuploid tumor genomes can evolve via punctuated evolution. Mapping the temporal dynamics and patterns of karyotype diversification in cancer enables reconstructions of evolutionary paths to malignant fitness.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Análise de Célula Única/métodos , Proliferação de Células/genética , Cromatina/genética , Cromossomos Humanos , Dosagem de Genes , Humanos , Cariótipo , Cariotipagem , Microscopia Confocal , Mitose , Organoides/crescimento & desenvolvimento , Organoides/patologia , Fuso Acromático/genética
5.
J Cell Biol ; 218(4): 1250-1264, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30728176

RESUMO

Cytokinesis begins upon anaphase onset. An early step involves local activation of the small GTPase RhoA, which triggers assembly of an actomyosin-based contractile ring at the equatorial cortex. Here, we delineated the contributions of PLK1 and Aurora B to RhoA activation and cytokinesis initiation in human cells. Knock-down of PRC1, which disrupts the spindle midzone, revealed the existence of two pathways that can initiate cleavage furrow ingression. One pathway depends on a well-organized spindle midzone and PLK1, while the other depends on Aurora B activity and centralspindlin at the equatorial cortex and can operate independently of PLK1. We further show that PLK1 inhibition sequesters centralspindlin onto the spindle midzone, making it unavailable for Aurora B at the equatorial cortex. We propose that PLK1 activity promotes the release of centralspindlin from the spindle midzone through inhibition of PRC1, allowing centralspindlin to function as a regulator of spindle midzone formation and as an activator of RhoA at the equatorial cortex.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Citocinese , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/enzimologia , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fuso Acromático/enzimologia , Animais , Aurora Quinase B/genética , Aurora Quinase B/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/genética , Ativação Enzimática , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/genética , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Fuso Acromático/genética , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Quinase 1 Polo-Like
6.
Nat Rev Cancer ; 19(1): 32-45, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30523339

RESUMO

Whole-genome and centrosome duplication as a consequence of cytokinesis failure can drive tumorigenesis in experimental model systems. However, whether cytokinesis failure is in fact an important cause of human cancers has remained unclear. In this Review, we summarize evidence that whole-genome-doubling events are frequently observed in human cancers and discuss the contribution that cytokinesis defects can make to tumorigenesis. We provide an overview of the potential causes of cytokinesis failure and discuss how tetraploid cells that are generated through cytokinesis defects are used in cancer as a transitory state on the route to aneuploidy. Finally, we discuss how cytokinesis defects can facilitate genetic diversification within the tumour to promote cancer development and could constitute the path of least resistance in tumour evolution.


Assuntos
Citocinese/genética , Neoplasias/genética , Neoplasias/patologia , Aneuploidia , Animais , Carcinogênese/genética , Carcinogênese/patologia , Centrossomo/patologia , Genoma/genética , Humanos
7.
J Cell Biol ; 217(1): 163-177, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29187526

RESUMO

Precise regulation of kinetochore-microtubule attachments is essential for successful chromosome segregation. Central to this regulation is Aurora B kinase, which phosphorylates kinetochore substrates to promote microtubule turnover. A critical target of Aurora B is the N-terminal "tail" domain of Hec1, which is a component of the NDC80 complex, a force-transducing link between kinetochores and microtubules. Although Aurora B is regarded as the "master regulator" of kinetochore-microtubule attachment, other mitotic kinases likely contribute to Hec1 phosphorylation. In this study, we demonstrate that Aurora A kinase regulates kinetochore-microtubule dynamics of metaphase chromosomes, and we identify Hec1 S69, a previously uncharacterized phosphorylation target site in the Hec1 tail, as a critical Aurora A substrate for this regulation. Additionally, we demonstrate that Aurora A kinase associates with inner centromere protein (INCENP) during mitosis and that INCENP is competent to drive accumulation of the kinase to the centromere region of mitotic chromosomes. These findings reveal that both Aurora A and B contribute to kinetochore-microtubule attachment dynamics, and they uncover an unexpected role for Aurora A in late mitosis.


Assuntos
Aurora Quinase A/metabolismo , Aurora Quinase B/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos/fisiologia , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Animais , Linhagem Celular Tumoral , Centrômero/metabolismo , Proteínas do Citoesqueleto , Células HeLa , Humanos , Metáfase/fisiologia , Fosforilação , Potoroidae , Ligação Proteica/fisiologia , Fuso Acromático/metabolismo
8.
PLoS One ; 12(6): e0179514, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28640891

RESUMO

The CRISPR/Cas9 system is a highly effective tool for genome editing. Key to robust genome editing is the efficient delivery of the CRISPR/Cas9 machinery. Viral delivery systems are efficient vehicles for the transduction of foreign genes but commonly used viral vectors suffer from a limited capacity in the genetic information they can carry. Baculovirus however is capable of carrying large exogenous DNA fragments. Here we investigate the use of baculoviral vectors as a delivery vehicle for CRISPR/Cas9 based genome-editing tools. We demonstrate transduction of a panel of cell lines with Cas9 and an sgRNA sequence, which results in efficient knockout of all four targeted subunits of the chromosomal passenger complex (CPC). We further show that introduction of a homology directed repair template into the same CRISPR/Cas9 baculovirus facilitates introduction of specific point mutations and endogenous gene tags. Tagging of the CPC recruitment factor Haspin with the fluorescent reporter YFP allowed us to study its native localization as well as recruitment to the cohesin subunit Pds5B.


Assuntos
Baculoviridae/genética , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Linhagem Celular Tumoral , Técnicas de Inativação de Genes , Genoma Humano/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação Puntual , Proteínas Serina-Treonina Quinases/genética
9.
Eur J Hum Genet ; 24(12): 1752-1760, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27530628

RESUMO

The oral-facial-digital (OFD) syndromes comprise a group of related disorders with a combination of oral, facial and digital anomalies. Variants in several ciliary genes have been associated with subtypes of OFD syndrome, yet in most OFD patients the underlying cause remains unknown. We investigated the molecular basis of disease in two brothers with OFD type II, Mohr syndrome, by performing single-nucleotide polymorphism (SNP)-array analysis on the brothers and their healthy parents to identify homozygous regions and candidate genes. Subsequently, we performed whole-exome sequencing (WES) on the family. Using WES, we identified compound heterozygous variants c.[464G>C];[1226G>A] in NIMA (Never in Mitosis Gene A)-Related Kinase 1 (NEK1). The novel variant c.464G>C disturbs normal splicing in an essential region of the kinase domain. The nonsense variant c.1226G>A, p.(Trp409*), results in nonsense-associated alternative splicing, removing the first coiled-coil domain of NEK1. Candidate variants were confirmed with Sanger sequencing and alternative splicing assessed with cDNA analysis. Immunocytochemistry was used to assess cilia number and length. Patient-derived fibroblasts showed severely reduced ciliation compared with control fibroblasts (18.0 vs 48.9%, P<0.0001), but showed no significant difference in cilia length. In conclusion, we identified compound heterozygous deleterious variants in NEK1 in two brothers with Mohr syndrome. Ciliation in patient fibroblasts is drastically reduced, consistent with a ciliary defect pathogenesis. Our results establish NEK1 variants involved in the etiology of a subset of patients with OFD syndrome type II and support the consideration of including (routine) NEK1 analysis in patients suspected of OFD.


Assuntos
Códon sem Sentido , Quinase 1 Relacionada a NIMA/genética , Síndromes Orofaciodigitais/genética , Processamento Alternativo , Células Cultivadas , Criança , Cílios/patologia , Exoma , Fibroblastos/metabolismo , Fibroblastos/patologia , Heterozigoto , Humanos , Lactente , Masculino , Síndromes Orofaciodigitais/patologia , Irmãos
10.
Dev Cell ; 34(4): 466-74, 2015 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-26256213

RESUMO

Sister-chromatid disjunction in anaphase requires the resolution of DNA catenanes by topoisomerase II together with Plk1-interacting checkpoint helicase (PICH) and Bloom's helicase (BLM). We here identify Rif1 as a factor involved in the resolution of DNA catenanes that are visible as ultrafine DNA bridges (UFBs) in anaphase to which PICH and BLM localize. Rif1, which during interphase functions downstream of 53BP1 in DNA repair, is recruited to UFBs in a PICH-dependent fashion, but independently of 53BP1 or BLM. Similar to PICH and BLM, Rif1 promotes the resolution of UFBs: its depletion increases the frequency of nucleoplasmic bridges and RPA70-positive UFBs in late anaphase. Moreover, in the absence of Rif1, PICH, or BLM, more nuclear bodies with damaged DNA arise in ensuing G1 cells, when chromosome decatenation is impaired. Our data reveal a thus far unrecognized function for Rif1 in the resolution of UFBs during anaphase to protect genomic integrity.


Assuntos
Anáfase , DNA/metabolismo , Instabilidade Genômica , Proteínas de Ligação a Telômeros/metabolismo , Proteína Quinase CDC2/metabolismo , Cromátides , Dano ao DNA , Fase G1 , Células HeLa , Humanos , Células MCF-7 , Micronúcleo Germinativo/metabolismo , Transporte Proteico
11.
Cell Rep ; 11(4): 508-15, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25892238

RESUMO

Correction of faulty kinetochore-microtubule attachments is essential for faithful chromosome segregation and dictated by the opposing activities of Aurora B kinase and PP1 and PP2A phosphatases. How kinase and phosphatase activities are appropriately balanced is less clear. Here, we show that a centromeric pool of PP2A-B56 counteracts Aurora B T-loop phosphorylation and is recruited to centromeres through Shugoshin-1 (Sgo1). In non-transformed RPE-1 cells, Aurora B, Sgo1, and PP2A-B56 are enriched on centromeres and levels diminish as chromosomes establish bi-oriented attachments. Elevating Sgo1 levels at centromeres recruits excess PP2A-B56, and this counteracts Aurora B kinase activity, undermining efficient correction of kinetochore-microtubule attachment errors. Conversely, Sgo1-depleted cells display reduced centromeric localization of Aurora B, whereas the remaining kinase is hyperactive due to concomitant reduction of centromeric PP2A-B56. Our data suggest that Sgo1 can tune the stability of kinetochore-microtubule attachments through recruitment of PP2A-B56 that balances Aurora B activity at the centromere.


Assuntos
Aurora Quinase B/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrômero/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Pareamento Cromossômico , Humanos
12.
Biochem Soc Trans ; 43(1): 23-32, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25619243

RESUMO

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


Assuntos
Instabilidade Cromossômica , Segregação de Cromossomos , Neoplasias/genética , Animais , Aurora Quinase B/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Citocinese , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Complexos Multiproteicos/fisiologia , Neoplasias/metabolismo , Neoplasias/patologia , Survivina
13.
Eur J Immunol ; 43(7): 1914-24, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23616302

RESUMO

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


Assuntos
Hipersensibilidade/imunologia , Proteínas Inibidoras de Apoptose/imunologia , Pneumonia/imunologia , Receptores OX40/imunologia , Proteínas Repressoras/imunologia , Células Th2/imunologia , Animais , Diferenciação Celular/imunologia , Hipersensibilidade/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pneumonia/metabolismo , Receptores OX40/deficiência , Proteínas Repressoras/metabolismo , Survivina , Células Th2/citologia
14.
Mol Cell Proteomics ; 11(5): 47-59, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22267324

RESUMO

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


Assuntos
Proteína HMGN2/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/genética , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Aurora Quinase B , Aurora Quinases , Domínio Catalítico , Extratos Celulares/química , Linhagem Celular Tumoral , Centrômero , Sequência Conservada , Difosfatos/farmacologia , Histonas/metabolismo , Humanos , Leucina/genética , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Mutagênese Sítio-Dirigida , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo
15.
Nat Rev Cancer ; 10(12): 825-41, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21102634

RESUMO

Large numbers of inhibitors for polo-like kinases and aurora kinases are currently being evaluated as anticancer drugs. Interest in these drugs is fuelled by the idea that these kinases have unique functions in mitosis. Within the polo-like kinase family, the emphasis for targeted therapies has been on polo-like kinase 1 (PLK1), and in the aurora kinase family drugs have been developed to specifically target aurora kinase A (AURKA; also known as STK6) and/or aurora kinase B (AURKB; also known as STK12). Information on the selectivity of these compounds in vivo is limited, but it is likely that off-target effects within the same kinase families will affect efficacy and toxicity profiles. In addition, it is becoming clear that interplay between polo-like kinases and aurora kinases is much more extensive than initially anticipated, and that both kinase families are important factors in the response to classical chemotherapeutics that damage the genome or the mitotic spindle. In this Review we discuss the implications of these novel insights on the clinical applicability of polo-like kinase and aurora kinase inhibitors.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Aurora Quinase A , Aurora Quinase B , Aurora Quinases , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/antagonistas & inibidores , Cromátides/fisiologia , Citocinese , Humanos , Cinetocoros/fisiologia , Mitose , Neoplasias/etiologia , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Quinase 1 Polo-Like
16.
Science ; 323(5919): 1350-3, 2009 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-19150808

RESUMO

Successful cell division requires that chromosomes attach to opposite poles of the mitotic spindle (bi-orientation). Aurora B kinase regulates chromosome-spindle attachments by phosphorylating kinetochore substrates that bind microtubules. Centromere tension stabilizes bi-oriented attachments, but how physical forces are translated into signaling at individual centromeres is unknown. Using fluorescence resonance energy transfer-based biosensors to measure localized phosphorylation dynamics in living cells, we found that phosphorylation of an Aurora B substrate at the kinetochore depended on its distance from the kinase at the inner centromere. Furthermore, repositioning Aurora B closer to the kinetochore prevented stabilization of bi-oriented attachments and activated the spindle checkpoint. Thus, centromere tension can be sensed by increased spatial separation of Aurora B from kinetochore substrates, which reduces phosphorylation and stabilizes kinetochore microtubules.


Assuntos
Centrômero/metabolismo , Cromossomos Humanos/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fuso Acromático/metabolismo , Aurora Quinase B , Aurora Quinases , Autoantígenos/metabolismo , Técnicas Biossensoriais , Linhagem Celular Tumoral , Centrômero/enzimologia , Proteína Centromérica A , Cromátides/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Transferência Ressonante de Energia de Fluorescência , Células HeLa , Humanos , Mitose , Modelos Biológicos , Fosforilação , Proteínas Recombinantes de Fusão/metabolismo
17.
Biochim Biophys Acta ; 1786(1): 60-72, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18662747

RESUMO

The Aurora protein kinase family (consisting of Aurora-A, -B and -C) is an important group of enzymes that controls several aspects of cell division in mammalian cells. Dysfunction of these kinases has been associated with a failure to maintain a stable chromosome content, a state that can contribute to tumourigenesis. Additionally, Aurora-A is frequently found amplified in a variety of tumour types and displays oncogenic activity. On the other hand, therapeutic inhibition of these kinases has shown great promise as potential anti-cancer treatment, most likely because of their essential roles during cell division. This review will focus on our present understanding of the different roles played by these kinases, their regulation throughout cell division, their deregulation in human cancers and on the progress that is made in targeting these important regulators in the treatment of cancer.


Assuntos
Divisão Celular/fisiologia , Neoplasias/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Anáfase/fisiologia , Aurora Quinases , Centrossomo/fisiologia , Cromátides/fisiologia , Citocinese/fisiologia , Ativação Enzimática , Humanos , Mitose/fisiologia , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Fuso Acromático/fisiologia
18.
Cell ; 132(2): 233-46, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18243099

RESUMO

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


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Cromossomos Humanos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Alelos , Aurora Quinase B , Aurora Quinases , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Ativação Enzimática , Células HeLa , Humanos , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Mutação , Fosforilação , Plasmídeos , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/metabolismo , Fuso Acromático/metabolismo , Transfecção
19.
Mol Biol Cell ; 18(11): 4553-64, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17699588

RESUMO

The chromosomal passenger complex (CPC) is a critical regulator of chromosome segregation during mitosis by correcting nonbipolar microtubule-kinetochore interactions. By severing these interactions, the CPC is thought to create unattached kinetochores that are subsequently sensed by the spindle assembly checkpoint (SAC) to prevent premature mitotic exit. We now show that spindle checkpoint function of the CPC and its role in eliminating nonbipolar attachments can be uncoupled. Replacing the chromosomal passenger protein INCENP with a mutant allele that lacks its coiled-coil domain results in an overt defect in a SAC-mediated mitotic arrest in response to taxol treatment, indicating that this domain is critical for CPC function in spindle checkpoint control. Surprisingly, this mutant could restore alignment and cytokinesis during unperturbed cell divisions and was capable of resolving syntelic attachments. Also, Aurora-B kinase was localized and activated normally on centromeres in these cells, ruling out a role for the coiled-coil domain in general Aurora-B activation. Thus, mere microtubule destabilization of nonbipolar attachments by the CPC is insufficient to install a checkpoint-dependent mitotic arrest, and additional, microtubule destabilization-independent CPC signaling toward the spindle assembly checkpoint is required for this arrest, potentially through amplification of the unattached kinetochore-derived checkpoint signal.


Assuntos
Cromossomos/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Aurora Quinase B , Aurora Quinases , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Deleção de Genes , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
20.
Cell Cycle ; 6(10): 1220-30, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17457057

RESUMO

Survivin operates in a complex with aurora B kinase and is phosphorylated by it on threonine 117 in vitro. Here we ask whether phosphorylation of survivin by aurora B kinase regulates its function during mitosis in vivo. Using a phospho-specific antibody we first establish that survivin is phosphorylated at T117 during mitosis and is present at the midbody during cytokinesis. Next we use two independent RNAi complementation approaches to investigate threonine 117 mutants in survivin depleted cells. Our data suggest that while non-phosphorylatable survivin, survivin(T117A), can substitute for the wild type protein, a phosphomimic, survivin(T117E) cannot restore viability, nor can it complement chromosome congression and spindle checkpoint defects that arise due to depletion of endogenous survivin. Fluorescence imaging and fluorescence recovery after photobleaching analysis suggest that the phosphomimic has reduced affinity for centromeres compared with the non-phosphorylatable form. We conclude that survivin is phosphorylated at T117 during mitosis, and once phosphorylated, dephosphorylation is crucial for chromosome congression and progression into anaphase.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/fisiologia , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Aurora Quinase B , Aurora Quinases , Linhagem Celular Tumoral , Centrômero/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Humanos , Immunoblotting , Proteínas Inibidoras de Apoptose , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Mutação/genética , Proteínas de Neoplasias/genética , Fosforilação , Interferência de RNA , Survivina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA