Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Nature ; 590(7846): 486-491, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33505028

RESUMEN

Selective targeting of aneuploid cells is an attractive strategy for cancer treatment1. However, it is unclear whether aneuploidy generates any clinically relevant vulnerabilities in cancer cells. Here we mapped the aneuploidy landscapes of about 1,000 human cancer cell lines, and analysed genetic and chemical perturbation screens2-9 to identify cellular vulnerabilities associated with aneuploidy. We found that aneuploid cancer cells show increased sensitivity to genetic perturbation of core components of the spindle assembly checkpoint (SAC), which ensures the proper segregation of chromosomes during mitosis10. Unexpectedly, we also found that aneuploid cancer cells were less sensitive than diploid cells to short-term exposure to multiple SAC inhibitors. Indeed, aneuploid cancer cells became increasingly sensitive to inhibition of SAC over time. Aneuploid cells exhibited aberrant spindle geometry and dynamics, and kept dividing when the SAC was inhibited, resulting in the accumulation of mitotic defects, and in unstable and less-fit karyotypes. Therefore, although aneuploid cancer cells could overcome inhibition of SAC more readily than diploid cells, their long-term proliferation was jeopardized. We identified a specific mitotic kinesin, KIF18A, whose activity was perturbed in aneuploid cancer cells. Aneuploid cancer cells were particularly vulnerable to depletion of KIF18A, and KIF18A overexpression restored their response to SAC inhibition. Our results identify a therapeutically relevant, synthetic lethal interaction between aneuploidy and the SAC.


Asunto(s)
Aneuploidia , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Neoplasias/patología , Cariotipo Anormal/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Segregación Cromosómica/efectos de los fármacos , Diploidia , Genes Letales , Humanos , Cinesinas/deficiencia , Cinesinas/genética , Cinesinas/metabolismo , Neoplasias/genética , Huso Acromático/efectos de los fármacos , Mutaciones Letales Sintéticas/efectos de los fármacos , Mutaciones Letales Sintéticas/genética , Factores de Tiempo
2.
Proc Natl Acad Sci U S A ; 115(8): E1779-E1788, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29432173

RESUMEN

Numerous posttranslational modifications have been described in kinesins, but their consequences on motor mechanics are largely unknown. We investigated one of these-acetylation of lysine 146 in Eg5-by creating an acetylation mimetic lysine to glutamine substitution (K146Q). Lysine 146 is located in the α2 helix of the motor domain, where it makes an ionic bond with aspartate 91 on the neighboring α1 helix. Molecular dynamics simulations predict that disrupting this bond enhances catalytic site-neck linker coupling. We tested this using structural kinetics and single-molecule mechanics and found that the K146Q mutation increases motor performance under load and coupling of the neck linker to catalytic site. These changes convert Eg5 from a motor that dissociates from the microtubule at low load into one that is more tightly coupled and dissociation resistant-features shared by kinesin 1. These features combined with the increased propensity to stall predict that the K146Q Eg5 acetylation mimetic should act in the cell as a "brake" that slows spindle pole separation, and we have confirmed this by expressing this modified motor in mitotically active cells. Thus, our results illustrate how a posttranslational modification of a kinesin can be used to fine tune motor behavior to meet specific physiological needs.


Asunto(s)
Cinesinas/química , Cinesinas/metabolismo , Mitosis/fisiología , Secuencia de Aminoácidos , Fenómenos Biomecánicos , Células HeLa , Humanos , Modelos Moleculares , Mutación , Conformación Proteica
3.
Mol Cell ; 43(5): 764-75, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21884977

RESUMEN

Metaphase chromosome positioning depends on Kif18A, a kinesin-8 that accumulates at and suppresses the dynamics of K-MT plus ends. By engineering Kif18A mutants that suppress MT dynamics but fail to concentrate at K-MT plus ends, we identify a mechanism that allows Kif18A to accumulate at K-MT plus ends to a level required to suppress chromosome movements. Enrichment of Kif18A at K-MT plus ends depends on its C-terminal tail domain, while the ability of Kif18A to suppress MT growth is conferred by the N-terminal motor domain. The Kif18A tail contains a second MT-binding domain that diffuses along the MT lattice, suggesting that it tethers the motor to the MT track. Consistently, the tail enhances Kif18A processivity and is crucial for it to accumulate at K-MT plus ends. The heightened processivity of Kif18A, conferred by its tail domain, thus promotes concentration of Kif18A at K-MT plus ends, where it suppresses their dynamics to control chromosome movements.


Asunto(s)
Cinesinas/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Posicionamiento de Cromosoma , Células HeLa , Humanos , Cinesinas/genética
4.
Dev Biol ; 402(2): 253-262, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25824710

RESUMEN

Genome integrity in the developing germ line is strictly required for fecundity. In proliferating somatic cells and in germ cells, there are mitotic checkpoint mechanisms that ensure accurate chromosome segregation and euploidy. There is growing evidence of mitotic cell cycle components that are uniquely required in the germ line to ensure genome integrity. We previously showed that the primary phenotype of germ cell deficient 2 (gcd2) mutant mice is infertility due to germ cell depletion during embryogenesis. Here we show that the underlying mutation is a mis-sense mutation, R308K, in the motor domain of the kinesin-8 family member, KIF18A, a protein that is expressed in a variety of proliferative tissues and is a key regulator of chromosome alignment during mitosis. Despite the conservative nature of the mutation, we show that its functional consequences are equivalent to KIF18A deficiency in HeLa cells. We also show that somatic cells progress through mitosis, despite having chromosome alignment defects, while germ cells with similar chromosome alignment defects undergo mitotic arrest and apoptosis. Our data provide evidence for differential requirements for chromosome alignment in germ and somatic cells and show that Kif18a is one of a growing number of genes that are specifically required for cell cycle progression in proliferating germ cells.


Asunto(s)
Proteínas de Ciclo Celular/genética , Células Germinativas/fisiología , Cinesinas/genética , Mitosis/fisiología , Animales , Apoptosis/fisiología , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Clonación Molecular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Vectores Genéticos/genética , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Cinesinas/metabolismo , Ratones , Mitosis/genética , Mutación Missense/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
J Cell Physiol ; 229(10): 1301-5, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24683081

RESUMEN

The proper organization and segregation of chromosomes during cell division is essential to the preservation of genomic integrity. To understand the mechanisms that spatially control the arrangement and dynamics of mitotic chromosomes requires imaging assays to quantitatively resolve their positions and movements. Here, we will discuss analytical approaches to investigate the position-dependent control of mitotic chromosomes in cultured cells. These methods can be used to dissect the specific contributions of mitotic proteins to the molecular control of chromosome dynamics.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Segregación Cromosómica , Cromosomas/metabolismo , Cinetocoros/metabolismo , Mitosis , Imagen Molecular/métodos , Animales , Células Cultivadas , Cromosomas/ultraestructura , Humanos , Cinética
6.
J Cell Physiol ; 229(12): 1881-3, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24905602

RESUMEN

Chromosome segregation and spindle microtubule dynamics are strictly coordinated during cell division in order to preserve genomic integrity. Alterations in the genome that affect microtubule stability and spindle assembly during mitosis may contribute to genomic instability and cancer predisposition, but directly testing this potential link poses a significant challenge. Germ-line mutations in tumor suppressor genes that predispose patients to cancer and alter spindle microtubule dynamics offer unique opportunities to investigate the relationship between spindle dysfunction and carcinogenesis. Mutations in two such tumor suppressors, adenomatous polyposis coli (APC) and Shwachman-Bodian-Diamond syndrome (SBDS), affect multifunctional proteins that have been well characterized for their roles in Wnt signaling and interphase ribosome assembly, respectively. Less understood, however, is how their shared involvement in stabilizing the microtubules that comprise the mitotic spindle contributes to cancer predisposition. Here, we briefly discuss the potential for mutations in APC and SBDS as informative tools for studying the impact of mitotic spindle dysfunction on cellular transformation.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Insuficiencia Pancreática Exocrina/genética , Lipomatosis/genética , Microtúbulos/genética , Neoplasias/genética , Huso Acromático/genética , Proteína de la Poliposis Adenomatosa del Colon/genética , Enfermedades de la Médula Ósea/patología , Carcinogénesis/genética , Segregación Cromosómica/genética , Insuficiencia Pancreática Exocrina/patología , Inestabilidad Genómica , Mutación de Línea Germinal , Humanos , Lipomatosis/patología , Mitosis/genética , Neoplasias/patología , Síndrome de Shwachman-Diamond
7.
Mol Biol Cell ; 35(1): ar3, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37903223

RESUMEN

Kinesins support many diverse cellular processes, including facilitating cell division through mechanical regulation of the mitotic spindle. However, how kinesin activity is controlled to facilitate this process is not well understood. Interestingly, posttranslational modifications have been identified within the enzymatic region of all 45 mammalian kinesins, but the significance of these modifications has gone largely unexplored. Given the critical role of the enzymatic region in facilitating nucleotide and microtubule binding, it may serve as a primary site for kinesin regulation. Consistent with this idea, a phosphomimetic mutation at S357 in the neck-linker of KIF18A alters the localization of KIF18A within the spindle from kinetochore microtubules to nonkinetochore microtubules at the periphery of the spindle. Changes in localization of KIF18A-S357D are accompanied by defects in mitotic spindle positioning and the ability to promote mitotic progression. This altered localization pattern is mimicked by a shortened neck-linker mutant, suggesting that KIF18A-S357D may cause the motor to adopt a shortened neck-linker-like state that decreases KIF18A accumulation at the plus-ends of kinetochore microtubules. These findings demonstrate that posttranslational modifications in the enzymatic region of kinesins could be important for biasing their localization to particular microtubule subpopulations.


Asunto(s)
Cinesinas , Cinetocoros , Microtúbulos , Animales , Humanos , Células HeLa , Cinesinas/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Mitosis , Huso Acromático/metabolismo
8.
Front Mol Biosci ; 11: 1328077, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38410188

RESUMEN

Background: The mitotic kinesin, KIF18A, is required for proliferation of cancer cells that exhibit chromosome instability (CIN), implicating it as a promising target for treatment of a subset of aggressive tumor types. Determining regions of the KIF18A protein to target for inhibition will be important for the design and optimization of effective small molecule inhibitors. Methods: In this study, we used cultured cell models to investigate the effects of mutating S284 within the alpha-4 helix of KIF18A, which was previously identified as a phosphorylated residue. Results: Mutations in S284 cause relocalization of KIF18A from the plus-ends of spindle microtubules to the spindle poles. Furthermore, KIF18A S284 mutants display loss of KIF18A function and fail to support proliferation in CIN tumor cells. Interestingly, similar effects on KIF18A localization and function were seen after treatment of CIN cells with KIF18A inhibitory compounds that are predicted to interact with residues within the alpha-4 helix. Conclusion: These data implicate the KIF18A alpha-4 helix as an effective target for inhibition and demonstrate that small molecules targeting KIF18A selectively limit CIN tumor cell proliferation and result in phenotypically similar effects on mitosis at the single cell level compared to genetic perturbations.

9.
J Cell Physiol ; 228(4): 835-45, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23018647

RESUMEN

Malignant mesothelioma (MM) is an intractable tumor of the peritoneal and pleural cavities primarily linked to exposure to asbestos. Recently, we described an interplay between mitochondrial-derived oxidants and expression of FOXM1, a redox-responsive transcription factor that has emerged as a promising therapeutic target in solid malignancies. Here we have investigated the effects of nitroxides targeted to mitochondria via triphenylphosphonium (TPP) moieties on mitochondrial oxidant production, expression of FOXM1 and peroxiredoxin 3 (PRX3), and cell viability in MM cells in culture. Both Mito-carboxy-proxyl (MCP) and Mito-TEMPOL (MT) caused dose-dependent increases in mitochondrial oxidant production that was accompanied by inhibition of expression of FOXM1 and PRX3 and loss of cell viability. At equivalent concentrations TPP, CP, and TEMPOL had no effect on these endpoints. Live cell ratiometric imaging with a redox-responsive green fluorescent protein targeted to mitochondria (mito-roGFP) showed that MCP and MT, but not CP, TEMPOL, or TPP, rapidly induced mitochondrial fragmentation and swelling, morphological transitions that were associated with diminished ATP levels and increased production of mitochondrial oxidants. Mdivi-1, an inhibitor of mitochondrial fission, did not rescue mitochondria from fragmentation by MCP. Immunofluorescence microscopy experiments indicate a fraction of FOXM1 coexists in the cytoplasm with mitochondrial PRX3. Our results indicate that MCP and MT inhibit FOXM1 expression and MM tumor cell viability via perturbations in redox homeostasis caused by marked disruption of mitochondrial architecture, and suggest that both compounds, either alone or in combination with thiostrepton or other agents, may provide credible therapeutic options for the management of MM.


Asunto(s)
Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/biosíntesis , Mesotelioma/metabolismo , Mitocondrias/metabolismo , Oxidantes/metabolismo , Peroxiredoxina III/antagonistas & inhibidores , Peroxiredoxina III/biosíntesis , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Citoplasma/efectos de los fármacos , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/fisiología , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/genética , Homeostasis/fisiología , Humanos , Mesotelioma/patología , Mitocondrias/efectos de los fármacos , Dinámicas Mitocondriales/efectos de los fármacos , Dinámicas Mitocondriales/genética , Dinámicas Mitocondriales/fisiología , Compuestos Organofosforados/farmacología , Oxidación-Reducción/efectos de los fármacos , Peroxiredoxina III/genética , Peroxiredoxina III/metabolismo , Quinazolinonas/farmacología
10.
Dev Cell ; 14(2): 252-62, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18267093

RESUMEN

During vertebrate cell division, chromosomes oscillate with periods of smooth motion interrupted by abrupt reversals in direction. These oscillations must be spatially constrained in order to align and segregate chromosomes with high fidelity, but the molecular mechanism for this activity is uncertain. We report here that the human kinesin-8 Kif18A has a primary role in the control of chromosome oscillations. Kif18A accumulates as a gradient on kinetochore microtubules in a manner dependent on its motor activity. Quantitative analyses of kinetochore movements reveal that Kif18A reduces the amplitude of preanaphase oscillations and slows poleward movement during anaphase. Thus, the microtubule-depolymerizing kinesin Kif18A has the unexpected function of suppressing chromosome movements. Based on these findings, we propose a molecular model in which Kif18A regulates kinetochore microtubule dynamics to control mitotic chromosome positioning.


Asunto(s)
Posicionamiento de Cromosoma , Cromosomas Humanos/metabolismo , Cinesinas/metabolismo , Cinetocoros/metabolismo , Mitosis , Anafase , Polaridad Celular , Células HeLa , Humanos , Microtúbulos/metabolismo , Modelos Biológicos , Transporte de Proteínas , Huso Acromático/metabolismo
11.
bioRxiv ; 2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-37205510

RESUMEN

Kinesins support many diverse cellular processes, including facilitating cell division through mechanical regulation of the mitotic spindle. However, how kinesin activity is controlled to facilitate this process is not well understood. Interestingly, post-translational modifications have been identified within the enzymatic region of all 45 mammalian kinesins, but the significance of these modifications has gone largely unexplored. Given the critical role of the enzymatic region in facilitating nucleotide and microtubule binding, it may serve as a primary site for kinesin regulation. Consistent with this idea, a phosphomimetic mutation at S357 in the neck-linker of KIF18A alters the localization of KIF18A within the spindle from kinetochore microtubules to peripheral microtubules. Changes in localization of KIF18A-S357D are accompanied by defects in mitotic spindle positioning and the ability to promote mitotic progression. This altered localization pattern is mimicked by a shortened neck-linker mutant, suggesting that KIF18A-S357D may cause the motor to adopt a shortened neck-linker like state that prevents KIF18A from accumulating at the plus-ends of kinetochore microtubules. These findings demonstrate that post-translational modifications in the enzymatic region of kinesins could be important for biasing their localization to particular microtubule subpopulations.

12.
bioRxiv ; 2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37905069

RESUMEN

The mitotic kinesin, KIF18A, is required for proliferation of cancer cells that exhibit chromosome instability (CIN), implicating it as a promising target for treatment of a subset of aggressive tumor types. Determining regions of the KIF18A protein to target for inhibition will be important for the design and optimization of effective small molecule inhibitors. In this study, we investigated the effects of mutating S284 within the alpha-4 helix of KIF18A, which was previously identified as a phosphorylated residue. Mutations in S284 cause relocalization of KIF18A from the plus-ends of spindle microtubules to the spindle poles. Furthermore, KIF18A S284 mutants display loss of KIF18A function and fail to support proliferation in CIN tumor cells. Interestingly, similar effects on KIF18A localization and function were seen after treatment of CIN cells with KIF18A inhibitory compounds that are predicted to interact with residues within the alpha-4 helix. These data implicate the KIF18A alpha-4 helix as an effective target for inhibition and demonstrate that small molecules targeting KIF18A selectively limit CIN tumor cell proliferation and result in phenotypically similar effects on mitosis at the single cell level compared to genetic perturbations.

13.
Methods Mol Biol ; 2415: 139-149, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34972951

RESUMEN

The chromokinesin KIF22 (Kid, kinesin-10 family) is the primary generator of polar ejection forces, which contribute to chromosome positioning and alignment in mitotic cells. Assessment of KIF22 function requires quantitative comparison of relative polar ejection forces between experimental conditions. This is facilitated by the generation of monopolar spindles to reduce the impact of bioriented microtubule attachment at kinetochores on chromosome positions and increase the dependence of chromosome positions on chromokinesin activity. Radial profile plots measure the intensity of chromatin signal in concentric circles around the poles of monopolar cells and represent an expedient quantitative measure of relative polar ejection forces. As such, this assay can be used to measure changes in polar ejection forces resulting from chromokinesin depletion or perturbation.


Asunto(s)
Cromosomas , Cinesinas , Cromosomas/genética , Proteínas de Unión al ADN/genética , Cinetocoros , Microtúbulos , Mitosis , Proteínas Nucleares/genética , Huso Acromático
14.
Elife ; 112022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35730929

RESUMEN

The chromokinesin KIF22 generates forces that contribute to mitotic chromosome congression and alignment. Mutations in the α2 helix of the motor domain of KIF22 have been identified in patients with abnormal skeletal development, and we report the identification of a patient with a novel mutation in the KIF22 tail. We demonstrate that pathogenic mutations do not result in a loss of KIF22's functions in early mitosis. Instead, mutations disrupt chromosome segregation in anaphase, resulting in reduced proliferation, abnormal daughter cell nuclear morphology, and, in a subset of cells, cytokinesis failure. This phenotype could be explained by a failure of KIF22 to inactivate in anaphase. Consistent with this model, constitutive activation of the motor via a known site of phosphoregulation in the tail phenocopied the effects of pathogenic mutations. These results suggest that the motor domain α2 helix may be an important site for regulation of KIF22 activity at the metaphase to anaphase transition. In support of this conclusion, mimicking phosphorylation of α2 helix residue T158 also prevents inactivation of KIF22 in anaphase. These findings demonstrate the importance of both the head and tail of the motor in regulating the activity of KIF22 and offer insight into the cellular consequences of preventing KIF22 inactivation and disrupting force balance in anaphase.


Asunto(s)
Anafase , Segregación Cromosómica , Proteínas de Unión al ADN , Cinesinas , Proteínas Nucleares , Proteínas de Unión al ADN/genética , Cinesinas/genética , Metafase , Mitosis , Mutación , Proteínas Nucleares/genética , Huso Acromático
15.
Curr Biol ; 18(2): R81-3, 2008 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-18211848

RESUMEN

Establishment of proper attachments between chromosomes and microtubules is essential for the accurate division of the genome. Two recent studies indicate that these attachments are facilitated by the geometry of chromosomes and the bipolar arrangement of spindle microtubules.


Asunto(s)
Cromosomas/metabolismo , Microtúbulos/metabolismo , Huso Acromático/metabolismo , División del Núcleo Celular/fisiología , Cinetocoros/metabolismo , Saccharomyces cerevisiae
16.
J Cell Biol ; 220(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34515734

RESUMEN

Micronuclei, whole or fragmented chromosomes spatially separated from the main nucleus, are associated with genomic instability and have been identified as drivers of tumorigenesis. Paradoxically, Kif18a mutant mice produce micronuclei due to asynchronous segregation of unaligned chromosomes in vivo but do not develop spontaneous tumors. We report here that micronuclei in Kif18a mutant mice form stable nuclear envelopes. Challenging Kif18a mutant mice via deletion of the Trp53 gene led to formation of thymic lymphoma with elevated levels of micronuclei. However, loss of Kif18a had modest or no effect on survival of Trp53 homozygotes and heterozygotes, respectively. Micronuclei in cultured KIF18A KO cells form stable nuclear envelopes characterized by increased recruitment of nuclear envelope components and successful expansion of decondensing chromatin compared with those induced by nocodazole washout or radiation. Lagging chromosomes were also positioned closer to the main chromatin masses in KIF18A KO cells. These data suggest that not all micronuclei actively promote tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Núcleo Celular/genética , Cinesinas/genética , Membrana Nuclear/genética , Animales , Línea Celular , Cromatina/genética , Cromosomas/genética , Daño del ADN/genética , Femenino , Inestabilidad Genómica/genética , Humanos , Masculino , Ratones
17.
Nat Commun ; 12(1): 1213, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33619254

RESUMEN

Chromosomal instability (CIN) is a hallmark of tumor cells caused by changes in the dynamics and control of microtubules that compromise the mitotic spindle. Thus, CIN cells may respond differently than diploid cells to treatments that target mitotic spindle regulation. Here, we test this idea by inhibiting a subset of kinesin motor proteins involved in mitotic spindle control. KIF18A is required for proliferation of CIN cells derived from triple negative breast cancer or colorectal cancer tumors but is not required in near-diploid cells. Following KIF18A inhibition, CIN tumor cells exhibit mitotic delays, multipolar spindles, and increased cell death. Sensitivity to KIF18A knockdown is strongly correlated with centrosome fragmentation, which requires dynamic microtubules but does not depend on bipolar spindle formation or mitotic arrest. Our results indicate the altered spindle microtubule dynamics characteristic of CIN tumor cells can be exploited to reduce the proliferative capacity of CIN cells.


Asunto(s)
Inestabilidad Cromosómica , Cinesinas/metabolismo , Neoplasias/genética , Neoplasias/patología , Puntos de Control del Ciclo Celular , Muerte Celular , Línea Celular Tumoral , Proliferación Celular , Centrosoma/metabolismo , Humanos , Microtúbulos/metabolismo , Mitosis , Modelos Biológicos , Nocodazol/farmacología , Paclitaxel/farmacología , Huso Acromático/metabolismo
18.
Sci Adv ; 7(47): eabj9812, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34797717

RESUMEN

Kinesins are regulated in space and time to ensure activation only in the presence of cargo. Kinesin-binding protein (KIFBP), which is mutated in Goldberg-Shprintzen syndrome, binds to and inhibits the catalytic motor heads of 8 of 45 kinesin superfamily members, but the mechanism remains poorly defined. Here, we used cryo­electron microscopy and cross-linking mass spectrometry to determine high-resolution structures of KIFBP alone and in complex with two mitotic kinesins, revealing structural remodeling of kinesin by KIFBP. We find that KIFBP remodels kinesin motors and blocks microtubule binding (i) via allosteric changes to kinesin and (ii) by sterically blocking access to the microtubule. We identified two regions of KIFBP necessary for kinesin binding and cellular regulation during mitosis. Together, this work further elucidates the molecular mechanism of KIFBP-mediated kinesin inhibition and supports a model in which structural rearrangement of kinesin motor domains by KIFBP abrogates motor protein activity.

19.
Curr Biol ; 17(9): R326-8, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17470346

RESUMEN

During mitosis, chromosomes must become aligned at the equator of the mitotic spindle before segregation. Recent work suggests that a kinesin-8 motor uses a unique combination of activities to regulate this process.


Asunto(s)
Anafase/fisiología , Segregación Cromosómica/fisiología , Cinesinas/metabolismo , Proteínas Motoras Moleculares/metabolismo , Humanos , Cinetocoros/metabolismo , Microtúbulos/metabolismo
20.
Elife ; 92020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31958056

RESUMEN

Kinesin-5 motors organize mitotic spindles by sliding apart microtubules. They are homotetramers with dimeric motor and tail domains at both ends of a bipolar minifilament. Here, we describe a regulatory mechanism involving direct binding between tail and motor domains and its fundamental role in microtubule sliding. Kinesin-5 tails decrease microtubule-stimulated ATP-hydrolysis by specifically engaging motor domains in the nucleotide-free or ADP states. Cryo-EM reveals that tail binding stabilizes an open motor domain ATP-active site. Full-length motors undergo slow motility and cluster together along microtubules, while tail-deleted motors exhibit rapid motility without clustering. The tail is critical for motors to zipper together two microtubules by generating substantial sliding forces. The tail is essential for mitotic spindle localization, which becomes severely reduced in tail-deleted motors. Our studies suggest a revised microtubule-sliding model, in which kinesin-5 tails stabilize motor domains in the microtubule-bound state by slowing ATP-binding, resulting in high-force production at both homotetramer ends.


Asunto(s)
Cinesinas/metabolismo , Microtúbulos/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Microscopía por Crioelectrón , Humanos , Hidrólisis , Cinesinas/química , Cinesinas/ultraestructura , Cinética , Unión Proteica , Dominios Proteicos , Huso Acromático/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda