Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 43(5): 764-75, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21884977

RESUMO

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.


Assuntos
Cinesinas/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Posicionamento Cromossômico , Células HeLa , Humanos , Cinesinas/genética
2.
J Biol Chem ; 291(41): 21350-21362, 2016 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-27531749

RESUMO

The molecular basis for control of the cytoskeleton by the Arf GTPase-activating protein AGAP1 has not been characterized. AGAP1 is composed of G-protein-like (GLD), pleckstrin homology (PH), Arf GAP, and ankyrin repeat domains. Kif2A was identified in screens for proteins that bind to AGAP1. The GLD and PH domains of AGAP1 bound the motor domain of Kif2A. Kif2A increased GAP activity of AGAP1, and a protein composed of the GLD and PH domains of AGAP1 increased ATPase activity of Kif2A. Knockdown (KD) of Kif2A or AGAP1 slowed cell migration and accelerated cell spreading. The effect of Kif2A KD on spreading could be rescued by expression of Kif2A-GFP or FLAG-AGAP1, but not by Kif2C-GFP. The effect of AGAP1 KD could be rescued by FLAG-AGAP1, but not by an AGAP1 mutant that did not bind Kif2A efficiently, ArfGAP1-HA or Kif2A-GFP. Taken together, the results support the hypothesis that the Kif2A·AGAP1 complex contributes to control of cytoskeleton remodeling involved in cell movement.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Cinesinas/metabolismo , Animais , Bovinos , Células Cultivadas , Proteínas Ativadoras de GTPase/química , Proteínas Ativadoras de GTPase/genética , Células HeLa , Humanos , Cinesinas/química , Cinesinas/genética
3.
bioRxiv ; 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38617353

RESUMO

Centrosomes are the principal microtubule-organizing centers of the cell and play an essential role in mitotic spindle function. Centrosome biogenesis is achieved by strict control of protein acquisition and phosphorylation prior to mitosis. Defects in this process promote fragmentation of pericentriolar material culminating in multipolar spindles and chromosome missegregation. Centriolar satellites, membrane-less aggrupations of proteins involved in the trafficking of proteins toward and away from the centrosome, are thought to contribute to centrosome biogenesis. Here we show that the microtubule plus-end directed kinesin motor Kif9 localizes to centriolar satellites and regulates their pericentrosomal localization during interphase. Lack of Kif9 leads to aggregation of satellites closer to the centrosome and increased centrosomal protein degradation that disrupts centrosome maturation and results in chromosome congression and segregation defects during mitosis. Our data reveal roles for Kif9 and centriolar satellites in the regulation of cellular proteostasis and mitosis.

4.
Dev Cell ; 14(2): 252-62, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18267093

RESUMO

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.


Assuntos
Posicionamento Cromossômico , Cromossomos Humanos/metabolismo , Cinesinas/metabolismo , Cinetocoros/metabolismo , Mitose , Anáfase , Polaridade Celular , Células HeLa , Humanos , Microtúbulos/metabolismo , Modelos Biológicos , Transporte Proteico , Fuso Acromático/metabolismo
5.
Curr Protoc ; 3(12): e965, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38153181

RESUMO

Protein activity is generally functionally integrated and spatially restricted to key locations within the cell. Knocksideways experiments allow researchers to rapidly move proteins to alternate or ectopic regions of the cell and assess the resultant cellular response. Briefly, individual proteins to be tested using this approach must be modified with moieties that dimerize under treatment with rapamycin to promote the experimental spatial relocalizations. CRISPR technology enables researchers to engineer modified protein directly in cells while preserving proper protein levels because the engineered protein will be expressed from endogenous promoters. Here we provide straightforward instructions to engineer tagged, rapamycin-relocalizable proteins in cells. The protocol is described in the context of our work with the microtubule depolymerizer MCAK/Kif2C, but it is easily adaptable to other genes and alternate tags such as degrons, optogenetic constructs, and other experimentally useful modifications. Off-target effects are minimized by testing for the most efficient target site using a split-GFP construct. This protocol involves no proprietary kits, only plasmids available from repositories (such as addgene.org). Validation, relocalization, and some example novel discoveries obtained working with endogenous protein levels are described. A graduate student with access to a fluorescence microscope should be able to prepare engineered cells with spatially controllable endogenous protein using this protocol. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Choosing a target site for gene modification Basic Protocol 2: Design of gRNA(s) for targeted gene modification Basic Protocol 3: Split-GFP test for target efficiency Basic Protocol 4: Design of the recombination template and analytical primers Support Protocol 1: Design of primers for analytical PCR Basic Protocol 5: Transfection, isolation, and validation of engineered cells Support Protocol 2: Stable transfection of engineered cells with binding partners.


Assuntos
Sistemas CRISPR-Cas , RNA Guia de Sistemas CRISPR-Cas , Animais , Humanos , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Animais Geneticamente Modificados , Linhagem Celular , Sirolimo
7.
Neurooncol Adv ; 3(1): vdaa165, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33506204

RESUMO

BACKGROUND: Glioma is sensitive to microtubule-targeting agents (MTAs), but most MTAs do not cross the blood brain barrier (BBB). To address this limitation, we developed the new chemical entity, ST-401, a brain-penetrant MTA. METHODS: Synthesis of ST-401. Measures of MT assembly and dynamics. Cell proliferation and viability of patient-derived (PD) glioma in culture. Measure of tumor microtube (TM) parameters using immunofluorescence analysis and machine learning-based workflow. Pharmacokinetics (PK) and experimental toxicity in mice. In vivo antitumor activity in the RCAS/tv-a PDGFB-driven glioma (PDGFB-glioma) mouse model. RESULTS: We discovered that ST-401 disrupts microtubule (MT) function through gentle and reverisible reduction in MT assembly that triggers mitotic delay and cell death in interphase. ST-401 inhibits the formation of TMs, MT-rich structures that connect glioma to a network that promotes resistance to DNA damage. PK analysis of ST-401 in mice shows brain penetration reaching antitumor concentrations, and in vivo testing of ST-401 in a xenograft flank tumor mouse model demonstrates significant antitumor activity and no over toxicity in mice. In the PDGFB-glioma mouse model, ST-401 enhances the therapeutic efficacies of temozolomide (TMZ) and radiation therapy (RT). CONCLUSION: Our study identifies hallmarks of glioma tumorigenesis that are sensitive to MTAs and reports ST-401 as a promising chemical scaffold to develop brain-penetrant MTAs.

8.
J Cell Biol ; 169(3): 391-7, 2005 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-15883193

RESUMO

MCAK is a member of the kinesin-13 family of microtubule (MT)-depolymerizing kinesins. We show that the potent MT depolymerizer MCAK tracks (treadmills) with the tips of polymerizing MTs in living cells. Tip tracking of MCAK is inhibited by phosphorylation and is dependent on the extreme COOH-terminal tail of MCAK. Tip tracking is not essential for MCAK's MT-depolymerizing activity. We propose that tip tracking is a mechanism by which MCAK is preferentially localized to regions of the cell that modulate the plus ends of MTs.


Assuntos
Compartimento Celular/fisiologia , Polaridade Celular/fisiologia , Cinesinas/metabolismo , Microtúbulos/metabolismo , Animais , Células CHO , Cricetinae , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Microscopia de Vídeo , Microtúbulos/ultraestrutura , Fosforilação , Polímeros/metabolismo , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/fisiologia , Proteínas Recombinantes de Fusão/metabolismo
9.
Mol Biol Cell ; 18(8): 2970-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17538014

RESUMO

The human genome has three unique genes coding for kinesin-13 proteins called Kif2a, Kif2b, and MCAK (Kif2c). Kif2a and MCAK have documented roles in mitosis, but the function of Kif2b has not been defined. Here, we show that Kif2b is expressed at very low levels in cultured cells and that GFP-Kif2b localizes predominately to centrosomes and midbodies, but also to spindle microtubules and transiently to kinetochores. Kif2b-deficient cells assemble monopolar or disorganized spindles. Chromosomes in Kif2b-deficient cells show typical kinetochore-microtubule attachments, but the velocity of movement is reduced approximately 80% compared with control cells. Some Kif2b-deficient cells attempt anaphase, but the cleavage furrow regresses and cytokinesis fails. Like Kif2a-deficient cells, bipolar spindle assembly can be restored to Kif2b-deficient cells by simultaneous deficiency of MCAK or Nuf2 or treatment with low doses of nocodazole. However, Kif2b-deficient cells are unique in that they assemble bipolar spindles when the pole focusing activities of NuMA and HSET are perturbed. These data demonstrate that Kif2b function is required for spindle assembly and chromosome movement and that the microtubule depolymerase activities of Kif2a, Kif2b, and MCAK fulfill distinct functions during mitosis in human cells.


Assuntos
Cinesinas/metabolismo , Mitose , Linhagem Celular , Cromossomos Humanos/metabolismo , Citocinese , Humanos , Cinetocoros/metabolismo , Filogenia , Proteínas Recombinantes de Fusão/metabolismo , Fuso Acromático/metabolismo
10.
Curr Biol ; 30(4): 610-623.e5, 2020 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-31928876

RESUMO

Neuronal axons terminate as synaptic boutons that form stable yet plastic connections with their targets. Synaptic bouton development relies on an underlying network of both long-lived and dynamic microtubules that provide structural stability for the boutons while also allowing for their growth and remodeling. However, a molecular-scale mechanism that explains how neurons appropriately balance these two microtubule populations remains a mystery. We hypothesized that α-tubulin acetyltransferase (αTAT), which both stabilizes long-lived microtubules against mechanical stress via acetylation and has been implicated in promoting microtubule dynamics, could play a role in this process. Using the Drosophila neuromuscular junction as a model, we found that non-enzymatic dαTAT activity limits the growth of synaptic boutons by affecting dynamic, but not stable, microtubules. Loss of dαTAT results in the formation of ectopic boutons. These ectopic boutons can be similarly suppressed by resupplying enzyme-inactive dαTAT or by treatment with a low concentration of the microtubule-targeting agent vinblastine, which acts to suppress microtubule dynamics. Biophysical reconstitution experiments revealed that non-enzymatic αTAT1 activity destabilizes dynamic microtubules but does not substantially impact the stability of long-lived microtubules. Further, during microtubule growth, non-enzymatic αTAT1 activity results in increasingly extended tip structures, consistent with an increased rate of acceleration of catastrophe frequency with microtubule age, perhaps via tip structure remodeling. Through these mechanisms, αTAT enriches for stable microtubules at the expense of dynamic ones. We propose that the specific suppression of dynamic microtubules by non-enzymatic αTAT activity regulates the remodeling of microtubule networks during synaptic bouton development.


Assuntos
Acetiltransferases/metabolismo , Drosophila melanogaster/metabolismo , Junção Neuromuscular/fisiologia , Terminações Pré-Sinápticas/fisiologia , Animais , Drosophila melanogaster/enzimologia , Drosophila melanogaster/crescimento & desenvolvimento , Larva/enzimologia , Larva/crescimento & desenvolvimento , Larva/metabolismo
11.
Dev Cell ; 6(2): 253-68, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14960279

RESUMO

Chromosome orientation and alignment within the mitotic spindle requires the Aurora B protein kinase and the mitotic centromere-associated kinesin (MCAK). Here, we report the regulation of MCAK by Aurora B. Aurora B inhibited MCAK's microtubule depolymerizing activity in vitro, and phospho-mimic (S/E) mutants of MCAK inhibited depolymerization in vivo. Expression of either MCAK (S/E) or MCAK (S/A) mutants increased the frequency of syntelic microtubule-kinetochore attachments and mono-oriented chromosomes. MCAK phosphorylation also regulates MCAK localization: the MCAK (S/E) mutant frequently localized to the inner centromere while the (S/A) mutant concentrated at kinetochores. We also detected two different binding sites for MCAK using FRAP analysis of the different MCAK mutants. Moreover, disruption of Aurora B function by expression of a kinase-dead mutant or RNAi prevented centromeric targeting of MCAK. These results link Aurora B activity to MCAK function, with Aurora B regulating MCAK's activity and its localization at the centromere and kinetochore.


Assuntos
Ciclo Celular/fisiologia , Centrômero/metabolismo , Cinesinas/metabolismo , Mitose/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Antineoplásicos Fitogênicos/farmacologia , Aurora Quinase B , Aurora Quinases , Autorradiografia , Células CHO , Ciclo Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Cromossomos/metabolismo , Cricetinae , Eletroforese em Gel Bidimensional , Recuperação de Fluorescência Após Fotodegradação/métodos , Imunofluorescência/métodos , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Técnicas In Vitro , Cinetocoros/metabolismo , Proteínas Luminescentes/metabolismo , Mitose/efeitos dos fármacos , Modelos Biológicos , Mutação , Nocodazol/farmacologia , Paclitaxel/farmacologia , Fosforilação , RNA Interferente Pequeno/farmacologia , Ratos , Transfecção
12.
J Cell Biol ; 159(4): 557-62, 2002 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-12446739

RESUMO

Unlike most kinesins, mitotic centromere-associated kinesin (MCAK) does not translocate along the surface of microtubules (MTs), but instead depolymerizes them. Among the motile kinesins, refinements that are unique for specific cellular functions, such as directionality and processivity, are under the control of a "neck" domain adjacent to the ATP-hydrolyzing motor domain. Despite its apparent lack of motility, MCAK also contains a neck domain. We found that deletions and alanine substitutions of highly conserved positively charged residues in the MCAK neck domain significantly reduced MT depolymerization activity. Furthermore, substitution of MCAK's neck domain with either the positively charged KIF1A K-loop or poly-lysine rescues the loss of MT-depolymerizing activity observed in the neckless MCAK mutant. We propose that the neck, analogously to the K-loop, interacts electrostatically with the tubulin COOH terminus to permit diffusional translocation of MCAK along the surface of MTs. This weak-binding interaction may also play an important role in processivity of MCAK-induced MT depolymerization.


Assuntos
Cinesinas/metabolismo , Microtúbulos/metabolismo , Polímeros/metabolismo , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Cricetinae , Humanos , Cinesinas/química , Cinesinas/genética , Proteínas Motores Moleculares/metabolismo , Dados de Sequência Molecular , Mutação , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência
13.
Biophys J ; 95(11): 5216-27, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18775962

RESUMO

Although the mechanism by which a kinesin-1 molecule moves individually along a microtubule is quite well-understood, the way that many kinesin-1 motor proteins bound to the same cargo move together along a microtubule is not. We identified a 60-amino-acid-long domain, termed Hinge 1, in kinesin-1 from Drosophila melanogaster that is located between the coiled coils of the neck and stalk domains. Its deletion reduces microtubule gliding speed in multiple-motor assays but not single-motor assays. Hinge 1 thus facilitates the cooperation of motors by preventing them from impeding each other. We addressed the structural basis for this phenomenon. Video-microscopy of single microtubule-bound full-length motors reveals the sporadic occurrence of high-compliance states alternating with longer-lived, low-compliance states. The deletion of Hinge 1 abolishes transitions to the high-compliance state. Based on Fourier transform infrared, circular dichroism, and fluorescence spectroscopy of Hinge 1 peptides, we propose that low-compliance states correspond to an unexpected structured organization of the central Hinge 1 region, whereas high-compliance states correspond to the loss of that structure. We hypothesize that strain accumulated during multiple-kinesin motility populates the high-compliance state by unfolding helical secondary structure in the central Hinge 1 domain flanked by unordered regions, thereby preventing the motors from interfering with each other in multiple-motor situations.


Assuntos
Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Movimento , Sequência de Aminoácidos , Animais , Elasticidade , Humanos , Camundongos , Microtúbulos/metabolismo , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Multimerização Proteica , Estabilidade Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Temperatura
14.
Eur J Med Chem ; 159: 74-89, 2018 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-30268825

RESUMO

Small molecules that target microtubules (MTs) represent promising therapeutics to treat certain types of cancer, including glioblastoma multiform (GBM). We synthesized modified carbazoles and evaluated their antitumor activity in GBM cells in culture. Modified carbazoles with an ethyl moiety linked to the nitrogen of the carbazole and a carbonyl moiety linked to distinct biaromatic rings exhibited remarkably different killing activities in human GBM cell lines and patient-derived GBM cells, with IC50 values from 67 to >10,000 nM. Measures of the activity of modified carbazoles with tubulin and microtubules coupled to molecular docking studies show that these compounds bind to the colchicine site of tubulin in a unique low interaction space that inhibits tubulin assembly. The modified carbazoles reported here represent novel chemical tools to better understand how small molecules disrupt MT functions and kill devastating cancers such as GBM.


Assuntos
Antineoplásicos/farmacologia , Carbazóis/farmacologia , Glioblastoma/tratamento farmacológico , Microtúbulos/efeitos dos fármacos , Antineoplásicos/síntese química , Antineoplásicos/química , Carbazóis/síntese química , Carbazóis/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Glioblastoma/patologia , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Relação Estrutura-Atividade
15.
Science ; 362(6415): 705-709, 2018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30409885

RESUMO

We describe a general computational approach to designing self-assembling helical filaments from monomeric proteins and use this approach to design proteins that assemble into micrometer-scale filaments with a wide range of geometries in vivo and in vitro. Cryo-electron microscopy structures of six designs are close to the computational design models. The filament building blocks are idealized repeat proteins, and thus the diameter of the filaments can be systematically tuned by varying the number of repeat units. The assembly and disassembly of the filaments can be controlled by engineered anchor and capping units built from monomers lacking one of the interaction surfaces. The ability to generate dynamic, highly ordered structures that span micrometers from protein monomers opens up possibilities for the fabrication of new multiscale metamaterials.


Assuntos
Biologia Computacional/métodos , Engenharia de Proteínas/métodos , Proteínas/química , Microscopia Crioeletrônica , Escherichia coli , Conformação Proteica em alfa-Hélice , Dobramento de Proteína , Estrutura Secundária de Proteína , Proteínas/genética
16.
Nat Cell Biol ; 19(4): 384-390, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28263957

RESUMO

Microtubules tether centrosomes together during interphase. How this is accomplished and what benefit it provides to the cell is not known. We have identified a bipolar, minus-end-directed kinesin, Kif25, that suppresses centrosome separation. Kif25 is required to prevent premature centrosome separation during interphase. We show that premature centrosome separation leads to microtubule-dependent nuclear translocation, culminating in eccentric nuclear positioning that disrupts the cortical spindle positioning machinery. The activity of Kif25 during interphase is required to maintain a centred nucleus to ensure the spindle is stably oriented at the onset of mitosis.


Assuntos
Centrossomo/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Mitose , Multimerização Proteica , Fuso Acromático/metabolismo , Animais , Núcleo Celular/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HCT116 , Células HeLa , Humanos , Células LLC-PK1 , Suínos
17.
Mol Biol Cell ; 27(8): 1300-9, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26912793

RESUMO

Depletion of microtubule (MT) regulators can initiate stable alterations in MT assembly rates that affect chromosome instability and mitotic spindle function, but the manner by which cellular MT assembly rates can stably increase or decrease is not understood. To investigate this phenomenon, we measured the response of microtubule assembly to both rapid and long-term loss of MT regulators MCAK/Kif2C and Kif18A. Depletion of MCAK/Kif2C by siRNA stably decreases MT assembly rates in mitotic spindles, whereas depletion of Kif18A stably increases rates of assembly. Surprisingly, this is not phenocopied by rapid rapamycin-dependent relocalization of MCAK/Kif2C and Kif18A to the plasma membrane. Instead, this treatment yields opposite affects on MT assembly. Rapidly increased MT assembly rates are balanced by a decrease in nucleated microtubules, whereas nucleation appears to be maximal and limiting for decreased MT assembly rates and also for long-term treatments. We measured amplified tubulin synthesis during long-term depletion of MT regulators and hypothesize that this is the basis for different phenotypes arising from long-term versus rapid depletion of MT regulators.


Assuntos
Cinesinas/metabolismo , Microtúbulos/metabolismo , Membrana Celular/metabolismo , Células HCT116/citologia , Células HCT116/efeitos dos fármacos , Células HeLa/citologia , Células HeLa/efeitos dos fármacos , Humanos , Cinesinas/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/efeitos dos fármacos , Microtúbulos/genética , RNA Interferente Pequeno , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sirolimo/farmacologia , Fuso Acromático/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo
18.
Mol Cancer Ther ; 15(9): 2018-29, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27325686

RESUMO

Glioblastoma multiforme is a devastating and intractable type of cancer. Current antineoplastic drugs do not improve the median survival of patients diagnosed with glioblastoma multiforme beyond 14 to 15 months, in part because the blood-brain barrier is generally impermeable to many therapeutic agents. Drugs that target microtubules (MT) have shown remarkable efficacy in a variety of cancers, yet their use as glioblastoma multiforme treatments has also been hindered by the scarcity of brain-penetrant MT-targeting compounds. We have discovered a new alkylindole compound, ST-11, that acts directly on MTs and rapidly attenuates their rate of assembly. Accordingly, ST-11 arrests glioblastoma multiforme cells in prometaphase and triggers apoptosis. In vivo analyses reveal that unlike current antitubulin agents, ST-11 readily crosses the blood-brain barrier. Further investigation in a syngeneic orthotopic mouse model of glioblastoma multiforme shows that ST-11 activates caspase-3 in tumors to reduce tumor volume without overt toxicity. Thus, ST-11 represents the first member of a new class of brain-penetrant antitubulin therapeutic agents. Mol Cancer Ther; 15(9); 2018-29. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Microtúbulos/metabolismo , Moduladores de Tubulina/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Camundongos , Nanopartículas , Projetos Piloto , Solubilidade , Moduladores de Tubulina/administração & dosagem , Moduladores de Tubulina/farmacocinética , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
19.
FEBS Lett ; 572(1-3): 80-4, 2004 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-15304328

RESUMO

Mitotic-centromere-associated kinesin (MCAK) is a member of the KIN I (internal motor domain) subfamily of kinesin related proteins. MCAK and its homologues destabilize microtubules both in cells and in vitro. Here, we analyzed the effects of MCAK in the presence and absence of ATP on the dynamic instability behavior of steady state microtubules assembled from purified HeLa cell tubulin. In the presence of ATP, substoichiometric levels of full length MCAK and a segment (A182) consisting of the motor and neck domains strongly increased the catastrophe frequency of the microtubules. These data demonstrate that MCAK is a microtubule-catastrophe promoting factor in vitro, and support the hypothesis that MCAK may serve as a catastrophe-promoting factor in cells.


Assuntos
Trifosfato de Adenosina/metabolismo , Cinesinas/metabolismo , Microtúbulos/enzimologia , Clonagem Molecular , Células HeLa , Humanos , Cinética , Microtúbulos/patologia , Proteínas Recombinantes/metabolismo
20.
J Cell Biol ; 197(2): 231-7, 2012 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-22492725

RESUMO

Mitotic centromere-associated kinesin (MCAK) is a microtubule-depolymerizing kinesin-13 member that can track with polymerizing microtubule tips (hereafter referred to as tip tracking) during both interphase and mitosis. MCAK tracks with microtubule tips by binding to end-binding proteins (EBs) through the microtubule tip localization signal SKIP, which lies N terminal to MCAK's neck and motor domain. The functional significance of MCAK's tip-tracking behavior during mitosis has never been explained. In this paper, we identify and define a mitotic function specific to the microtubule tip-associated population of MCAK: negative regulation of microtubule length within the assembling bipolar spindle. This function depends on MCAK's ability to bind EBs and track with polymerizing nonkinetochore microtubule tips. Although this activity antagonizes centrosome separation during bipolarization, it ultimately benefits the dividing cell by promoting robust kinetochore attachments to the spindle microtubules.


Assuntos
Cinesinas/genética , Cinesinas/metabolismo , Cinetocoros/metabolismo , Microtúbulos/fisiologia , Fuso Acromático/fisiologia , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrômero/metabolismo , Centrossomo/metabolismo , Células HeLa , Humanos , Cinesinas/antagonistas & inibidores , Mitose , Proteínas Nucleares/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Pirimidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/genética , Fuso Acromático/ultraestrutura , Tionas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA