Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Crit Rev Clin Lab Sci ; : 1-31, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38344808

RESUMO

KIF2C/MCAK (KIF2C) is the most well-characterized member of the kinesin-13 family, which is critical in the regulation of microtubule (MT) dynamics during mitosis, as well as interphase. This systematic review briefly describes the important structural elements of KIF2C, its regulation by multiple molecular mechanisms, and its broad cellular functions. Furthermore, it systematically summarizes its oncogenic potential in malignant progression and performs a meta-analysis of its prognostic value in cancer patients. KIF2C was shown to be involved in multiple crucial cellular processes including cell migration and invasion, DNA repair, senescence induction and immune modulation, which are all known to be critical during the development of malignant tumors. Indeed, an increasing number of publications indicate that KIF2C is aberrantly expressed in multiple cancer entities. Consequently, we have highlighted its involvement in at least five hallmarks of cancer, namely: genome instability, resisting cell death, activating invasion and metastasis, avoiding immune destruction and cellular senescence. This was followed by a systematic search of KIF2C/MCAK's expression in various malignant tumor entities and its correlation with clinicopathologic features. Available data were pooled into multiple weighted meta-analyses for the correlation between KIF2Chigh protein or gene expression and the overall survival in breast cancer, non-small cell lung cancer and hepatocellular carcinoma patients. Furthermore, high expression of KIF2C was correlated to disease-free survival of hepatocellular carcinoma. All meta-analyses showed poor prognosis for cancer patients with KIF2Chigh expression, associated with a decreased overall survival and reduced disease-free survival, indicating KIF2C's oncogenic potential in malignant progression and as a prognostic marker. This work delineated the promising research perspective of KIF2C with modern in vivo and in vitro technologies to further decipher the function of KIF2C in malignant tumor development and progression. This might help to establish KIF2C as a biomarker for the diagnosis or evaluation of at least three cancer entities.

2.
Cell ; 136(5): 865-75, 2009 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-19269365

RESUMO

Kinetochores couple chromosomes to the assembling and disassembling tips of microtubules, a dynamic behavior that is fundamental to mitosis in all eukaryotes but poorly understood. Genetic, biochemical, and structural studies implicate the Ndc80 complex as a direct point of contact between kinetochores and microtubules, but these approaches provide only a static view. Here, using techniques for manipulating and tracking individual molecules in vitro, we demonstrate that the Ndc80 complex is capable of forming the dynamic, load-bearing attachments to assembling and disassembling tips required for coupling in vivo. We also establish that Ndc80-based coupling likely occurs through a biased diffusion mechanism and that this activity is conserved from yeast to humans. Our findings demonstrate how an ensemble of Ndc80 complexes may provide the combination of plasticity and strength that allows kinetochores to maintain load-bearing tip attachments during both microtubule assembly and disassembly.


Assuntos
Cinetocoros/metabolismo , Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citologia
3.
Biochem J ; 478(8): 1617-1629, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33909027

RESUMO

Historically, the diffusion of chemical signals through the cell was thought to occur within a cytoplasmic soup bounded by the plasma membrane. This theory was predicated on the notion that all regulatory enzymes are soluble and moved with a Brownian motion. Although enzyme compartmentalization was initially rebuffed by biochemists as a 'last refuge of a scoundrel', signal relay through macromolecular complexes is now accepted as a fundamental tenet of the burgeoning field of spatial biology. A-Kinase anchoring proteins (AKAPs) are prototypic enzyme-organizing elements that position clusters of regulatory proteins at defined subcellular locations. In parallel, the primary cilium has gained recognition as a subcellular mechanosensory organelle that amplifies second messenger signals pertaining to metazoan development. This article highlights advances in our understanding of AKAP signaling within the primary cilium and how defective ciliary function contributes to an increasing number of diseases known as ciliopathies.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Cílios/metabolismo , Ciliopatias/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Mecanotransdução Celular/genética , Proteínas de Ancoragem à Quinase A/genética , Animais , Compartimento Celular , Cílios/genética , Cílios/ultraestrutura , Ciliopatias/genética , Ciliopatias/patologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Células Eucarióticas/citologia , Células Eucarióticas/metabolismo , Regulação da Expressão Gênica , Humanos , Família Multigênica
4.
J Biol Chem ; 295(40): 13784-13797, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-32732289

RESUMO

Mitogenic signals that regulate cell division often proceed through multienzyme assemblies within defined intracellular compartments. The anchoring protein Gravin restricts the action of mitotic kinases and cell-cycle effectors to defined mitotic structures. In this report we discover that genetic deletion of Gravin disrupts proper accumulation and asymmetric distribution of γ-tubulin during mitosis. We utilize a new precision pharmacology tool, Local Kinase Inhibition, to inhibit the Gravin binding partner polo-like kinase 1 at spindle poles. Using a combination of gene-editing approaches, quantitative imaging, and biochemical assays, we provide evidence that disruption of local polo-like kinase 1 signaling underlies the γ-tubulin distribution defects observed with Gravin loss. Our study uncovers a new role for Gravin in coordinating γ-tubulin recruitment during mitosis and illuminates the mechanism by which signaling enzymes regulate this process at a distinct subcellular location.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ciclo Celular/genética , Células HEK293 , Células HeLa , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Fuso Acromático/genética , Tubulina (Proteína)/genética , Quinase 1 Polo-Like
5.
Mol Cell ; 48(4): 547-59, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23063527

RESUMO

The mitogenic and second-messenger signals that promote cell proliferation often proceed through multienzyme complexes. The kinase-anchoring protein Gravin integrates cAMP and calcium/phospholipid signals at the plasma membrane by sequestering protein kinases A and C with G protein-coupled receptors. In this report we define a role for Gravin as a temporal organizer of phosphorylation-dependent protein-protein interactions during mitosis. Mass spectrometry, molecular, and cellular approaches show that CDK1/Cyclin B1 phosphorylates Gravin on threonine 766 to prime the recruitment of the polo-like kinase Plk1 at defined phases of mitosis. Fluorescent live-cell imaging reveals that cells depleted of Gravin exhibit mitotic defects that include protracted prometaphase and misalignment of chromosomes. Moreover, a Gravin T766A phosphosite mutant that is unable to interact with Plk1 negatively impacts cell proliferation. In situ detection of phospho-T766 Gravin in biopsy sections of human glioblastomas suggests that this phosphorylation event might identify malignant neoplasms.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Animais , Proteínas de Ciclo Celular/genética , Divisão Celular , Proliferação de Células , Humanos , Camundongos , Mitose , Fosforilação , Ligação Proteica , Células Tumorais Cultivadas , Quinase 1 Polo-Like
6.
Glia ; 67(8): 1558-1570, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31058365

RESUMO

GPR124 is involved in embryonic development and remains expressed by select organs. The importance of GPR124 during development suggests that its aberrant expression might participate in tumor growth. Here we show that both increases and decreases in GPR124 expression in glioblastoma cells reduce cell proliferation by differentially altering the duration mitotic progression. Using mass spectrometry-based proteomics, we discovered that GPR124 interacts with ch-TOG, a known regulator of both microtubule (MT)-plus-end assembly and mitotic progression. Accordingly, changes in GPR124 expression and ch-TOG similarly affect MT assembly measured by real-time microscopy in cells. Our study describes a novel molecular interaction involving GPR124 and ch-TOG at the plasma membrane that controls glioblastoma cell proliferation by modifying MT assembly rates and controlling the progression of distinct phases of mitosis.


Assuntos
Neoplasias Encefálicas/metabolismo , Proliferação de Células/fisiologia , Glioblastoma/metabolismo , Microtúbulos/metabolismo , Mitose/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Encéfalo/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Feminino , Expressão Gênica , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade
7.
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
8.
Crit Rev Biochem Mol Biol ; 51(4): 228-45, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27146484

RESUMO

Chromosome stability is ensured by precisely fine-tuned dynamics of mitotic spindles, which are controlled by a network of various microtubule-associated and interacting proteins including the kinesin-13 family. The best characterized member of this family is the mitotic centromere-associated kinesin (MCAK). By efficiently depolymerizing microtubules, MCAK influences various key events during mitosis. MCAK itself is regulated by its interaction partners, its intrinsic conformation switch and the phosphorylation of mitotic kinases like Aurora A/B, cyclin-dependent kinase 1 and Polo-like kinase 1. Perturbing its regulation alters MCAK's conformation, catalytic activity, subcellular localization and stability, leading further to mitotic defects in spindle formation and chromosome movement. Indeed, MCAK is aberrantly regulated in various cancer types, which is linked to increased invasiveness, metastasis and drug resistance. In the current review, we summarize recently published data concerning MCAK, correlate its conformation changes with its depolymerization activity and function, propose a model of its regulation by multiple mitotic kinases and highlight its potential involvement in oncogenesis and drug resistance.


Assuntos
Regulação da Expressão Gênica , Cinesinas/genética , Fuso Acromático/metabolismo , Instabilidade Cromossômica , Resistencia a Medicamentos Antineoplásicos , Humanos , Cinesinas/química , Cinesinas/metabolismo , Cinesinas/fisiologia , Invasividade Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação , Conformação Proteica
9.
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
10.
J Mol Cell Cardiol ; 93: 32-43, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26902968

RESUMO

Microtubules (MTs) have many roles in ventricular myocytes, including structural stability, morphological integrity, and protein trafficking. However, despite their functional importance, dynamic MTs had never been visualized in living adult myocytes. Using adeno-associated viral vectors expressing the MT-associated protein plus end binding protein 3 (EB3) tagged with EGFP, we were able to perform live imaging and thus capture and quantify MT dynamics in ventricular myocytes in real time under physiological conditions. Super-resolution nanoscopy revealed that EB1 associated in puncta along the length of MTs in ventricular myocytes. The vast (~80%) majority of MTs grew perpendicular to T-tubules at a rate of 0.06µm∗s(-1) and growth was preferentially (82%) confined to a single sarcomere. Microtubule catastrophe rate was lower near the Z-line than M-line. Hydrogen peroxide increased the rate of catastrophe of MTs ~7-fold, suggesting that oxidative stress destabilizes these structures in ventricular myocytes. We also quantified MT dynamics after myocardial infarction (MI), a pathological condition associated with increased production of reactive oxygen species (ROS). Our data indicate that the catastrophe rate of MTs increases following MI. This contributed to decreased transient outward K(+) currents by decreasing the surface expression of Kv4.2 and Kv4.3 channels after MI. On the basis of these data, we conclude that, under physiological conditions, MT growth is directionally biased and that increased ROS production during MI disrupts MT dynamics, decreasing K(+) channel trafficking.


Assuntos
Ventrículos do Coração/metabolismo , Microtúbulos/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Animais , Camundongos , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/metabolismo , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Ligação Proteica , Transporte Proteico , Tubulina (Proteína)/metabolismo
11.
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.

12.
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
13.
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
14.
Semin Cell Dev Biol ; 21(3): 260-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20109570

RESUMO

Kinesins are enzymes that use the energy of ATP to perform mechanical work. There are approximately 14 families of kinesins within the kinesin superfamily. Family classification is derived primarily from alignments of the sequences of the core motor domain. For this reason, the enzymatic behavior and motility of each motor generally reflects its family. At the cellular level, kinesin motors perform a variety of functions during cell division and within the mitotic spindle to ensure that chromosomes are segregated with the highest fidelity possible. The cellular functions of these motors are intimately related to their mechanical and enzymatic properties at the single molecule level. For this reason, motility studies designed to evaluate the activity of purified molecular motors are a requirement in order to understand, mechanistically, how these motors make the mitotic spindle work and what can cause the spindle to fail. This review will focus on a selection of illustrative kinesins, which have been studied at the molecular level in order to inform our understanding of their function in cells. In addition, the review will endeavor to point out some kinesins that have been studied extensively but which still lack sufficient molecular underpinnings to fully predict their contribution to spindle function.


Assuntos
Cinesinas/fisiologia , Fuso Acromático , Trifosfato de Adenosina/metabolismo , Animais , Divisão Celular , Cromossomos/ultraestrutura , Humanos , Microtúbulos/metabolismo , Modelos Biológicos , Proteínas Motores Moleculares/metabolismo , Proteínas de Plantas/metabolismo
16.
J Biol Chem ; 286(45): 39269-81, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21890631

RESUMO

Cell movement requires the coordinated reception, integration, and processing of intracellular signals. We have discovered that the protein kinase A anchoring protein AKAP220 interacts with the cytoskeletal scaffolding protein IQGAP1 to influence cell motility. AKAP220/IQGAP1 networks receive and integrate calcium and cAMP second messenger signals and position signaling enzymes near their intended substrates at leading edges of migrating cells. IQGAP1 supports calcium/calmodulin-dependent association of factors that modulate microtubule dynamics. AKAP220 suppresses GSK-3ß and positions this kinase to allow recruitment of the plus-end microtubule tracking protein CLASP2. Gene silencing of AKAP220 alters the rate of microtubule polymerization and the lateral tracking of growing microtubules and retards cell migration in metastatic human cancer cells. This reveals an unappreciated role for this anchored kinase/microtubule effector protein network in the propagation of cell motility.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Movimento Celular/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Cálcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Linhagem Celular Tumoral , AMP Cíclico/genética , AMP Cíclico/metabolismo , Inativação Gênica , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/genética , Microtúbulos/metabolismo , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo
17.
Curr Opin Cell Biol ; 17(1): 82-8, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15661523

RESUMO

The fact that some kinesin-related proteins can destabilize microtubules is now a well-established fact. However, the contribution that these kinesins make to cellular function is just coming into focus. Key structural and kinetic studies on the mechanism of microtubule depolymerization by these kinesins have provided a framework for understanding their cellular regulation and function. Completion of some of the genome sequences and recent technological advances enabling the rapid depletion of cellular proteins in metazoans have clarified the functional role and level of cooperation between members of the depolymerizing kinesin families. Recent studies utilizing these technologies have revealed how these kinesins play an integral role in the mechanics of mitotic spindle assembly, chromosome segregation and the shaping of connections in the brain.


Assuntos
Cinesinas/química , Microtúbulos/ultraestrutura , Trifosfato de Adenosina/química , Adenilil Imidodifosfato/química , Animais , Humanos , Hidrólise , Cinética , Microtúbulos/química , Microtúbulos/metabolismo , Mitose , Modelos Biológicos , Neurônios/metabolismo , Fuso Acromático
18.
J Cell Biol ; 174(6): 839-49, 2006 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-16954346

RESUMO

Tubulin-tyrosine ligase (TTL), the enzyme that catalyzes the addition of a C-terminal tyrosine residue to alpha-tubulin in the tubulin tyrosination cycle, is involved in tumor progression and has a vital role in neuronal organization. We show that in mammalian fibroblasts, cytoplasmic linker protein (CLIP) 170 and other microtubule plus-end tracking proteins comprising a cytoskeleton-associated protein glycine-rich (CAP-Gly) microtubule binding domain such as CLIP-115 and p150 Glued, localize to the ends of tyrosinated microtubules but not to the ends of detyrosinated microtubules. In vitro, the head domains of CLIP-170 and of p150 Glued bind more efficiently to tyrosinated microtubules than to detyrosinated polymers. In TTL-null fibroblasts, tubulin detyrosination and CAP-Gly protein mislocalization correlate with defects in both spindle positioning during mitosis and cell morphology during interphase. These results indicate that tubulin tyrosination regulates microtubule interactions with CAP-Gly microtubule plus-end tracking proteins and provide explanations for the involvement of TTL in tumor progression and in neuronal organization.


Assuntos
Fibroblastos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Tubulina (Proteína)/metabolismo , Tirosina/metabolismo , Animais , Células Cultivadas , Complexo Dinactina , Fibroblastos/ultraestrutura , Interfase/fisiologia , Camundongos , Microtúbulos/ultraestrutura , Proteínas do Tecido Nervoso/metabolismo , Peptídeo Sintases/genética , Peptídeo Sintases/metabolismo , Polímeros/metabolismo , Estrutura Terciária de Proteína/fisiologia , Fuso Acromático/metabolismo , Fuso Acromático/ultraestrutura
19.
EMBO Rep ; 10(8): 857-65, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19543227

RESUMO

The microtubule (MT) cytoskeleton orchestrates the cellular plasticity and dynamics that underlie morphogenesis and cell division. Growing MT plus ends have emerged as dynamic regulatory machineries in which specialized proteins-called plus-end tracking proteins (+TIPs)-bind to and control the plus-end dynamics that are essential for cell division and migration. However, the molecular mechanisms underlying the plus-end regulation by +TIPs at spindle and astral MTs have remained elusive. Here, we show that TIP150 is a new +TIP that binds to end-binding protein 1 (EB1) in vitro and co-localizes with EB1 at the MT plus ends in vivo. Suppression of EB1 eliminates the plus-end localization of TIP150. Interestingly, TIP150 also binds to mitotic centromere-associated kinesin (MCAK), an MT depolymerase that localizes to the plus end of MTs. Suppression of TIP150 diminishes the plus-end localization of MCAK. Importantly, aurora B-mediated phosphorylation disrupts the TIP150-MCAK association in vitro. We reason that TIP150 facilitates the EB1-dependent loading of MCAK onto MT plus ends and orchestrates the dynamics at the plus end of MTs.


Assuntos
Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Células HeLa , Humanos , Imuno-Histoquímica , Proteínas Associadas aos Microtúbulos/genética , Dados de Sequência Molecular , Ligação Proteica , Homologia de Sequência de Aminoácidos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA