Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Mol Biol Cell ; 18(5): 1909-17, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17344472

RESUMO

The microtubule cytoskeleton is differentially regulated by a diverse array of proteins during interphase and mitosis. Op18/stathmin (Op18) and microtubule-associated protein (MAP)4 have been ascribed opposite general microtubule-directed activities, namely, microtubule destabilization and stabilization, respectively, both of which can be inhibited by phosphorylation. Here, using three human cell models, we depleted cells of Op18 and/or MAP4 by expression of interfering hairpin RNAs and we analyzed the resulting phenotypes. We found that the endogenous levels of Op18 and MAP4 have opposite and counteractive activities that largely govern the partitioning of tubulin dimers in the microtubule array at interphase. Op18 and MAP4 were also found to be the downstream targets of Ca(2+)- and calmodulin-dependent protein kinase IV and PAR-1/MARK2 kinase, respectively, that control the demonstrated counteractive phosphorylation-mediated regulation of tubulin dimer partitioning. Furthermore, to address mechanisms regulating microtubule polymerization in response to cell signals, we developed a system for inducible gene product replacement. This approach revealed that site-specific phosphorylation of Op18 is both necessary and sufficient for polymerization of microtubules in response to the multifaceted signaling event of stimulation of the T cell antigen receptor complex, which activates several signal transduction pathways.


Assuntos
Interfase/fisiologia , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Sequência de Bases , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Linhagem Celular , DNA/genética , Dimerização , Humanos , Células Jurkat , Células K562 , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Quaternária de Proteína , Transdução de Sinais , Estatmina/antagonistas & inibidores , Estatmina/genética , Estatmina/metabolismo , Transfecção
2.
Mol Biol Cell ; 17(7): 2921-30, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16624860

RESUMO

Op18/stathmin (Op18) is a phosphorylation-regulated microtubule destabilizer that is frequently overexpressed in tumors. The importance of Op18 in malignancy was recently suggested by identification of a somatic Q18-->E mutation of Op18 in an adenocarcinoma. We addressed the functional consequences of aberrant Op18 expression in leukemias by analyzing the cell cycle of K562 cells either depleted of Op18 by expression of interfering hairpin RNA or induced to express wild-type or Q18E substituted Op18. We show here that although Op18 depletion increases microtubule density during interphase, the density of mitotic spindles is essentially unaltered and cells divide normally. This is consistent with phosphorylation-inactivation of Op18 during mitosis. Overexpression of wild-type Op18 results in aneugenic activities, manifest as aberrant mitosis, polyploidization, and chromosome loss. One particularly significant finding was that the aneugenic activity of Op18 was dramatically increased by the Q18-->E mutation. The hyperactivity of mutant Op18 is apparent in its unphosphorylated state, and this mutation also suppresses phosphorylation-inactivation of the microtubule-destabilizing activity of Op18 without any apparent effect on its phosphorylation status. Thus, although Op18 is dispensable for mitosis, the hyperactive Q18-->E mutant, or overexpressed wild-type Op18, exerts aneugenic effects that are likely to contribute to chromosomal instability in tumors.


Assuntos
Instabilidade Cromossômica , Leucemia/genética , Fuso Acromático/metabolismo , Estatmina/genética , Estatmina/metabolismo , Aneugênicos/metabolismo , Ciclo Celular/genética , Humanos , Leucemia/metabolismo , Leucemia/patologia , Microtúbulos/metabolismo , Mitose/genética , Mutação , Fosforilação , Interferência de RNA , Estatmina/antagonistas & inibidores , Tubulina (Proteína)/metabolismo
3.
Mol Biol Cell ; 14(9): 3716-29, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12972559

RESUMO

The Op18/stathmin family of microtubule regulators includes the ubiquitous cytosolic Op18/stathmin (Op18) and the neuronal, primarily Golgi-associated proteins SCG10 and RB3, which all form ternary complexes with two head-to-tail-aligned tubulin heterodimers. To understand the physiological significance of previously observed differences in ternary complex stability, we have fused each of the heterodimer-binding regions of these three proteins with the CD2 cell surface protein to generate confined plasma membrane localization of the resulting CD2 chimeras. Herein, we show that, in contrast to constitutively active CD2-Op18-tetraA, both the CD2-SCG10 and CD2-RB3 chimeras sequestered tubulin at the plasma membrane, which results in >35% reduction of cytosolic tubulin heterodimer levels and consequent delayed formation of mitotic spindles. However, all three CD2 chimeras, including the tubulin sequestration-incompetent CD2-Op18-tetraA, destabilize interphase microtubules. Given that microtubules are in extensive contact with the plasma membrane during interphase, but not during mitosis, these findings indicate that Op18-like proteins have the potential to destabilize microtubules by both sequestration and direct interaction with microtubules. However, the differences in tubulin binding observed in cells also indicate conceptual differences between the functions of low-abundance neural family members, which will accumulate tubulin at specific cellular compartments, and the abundant cytosolic Op18 protein, which will not.


Assuntos
Proteínas dos Microtúbulos , Microtúbulos/metabolismo , Fatores de Crescimento Neural/metabolismo , Fosfoproteínas/metabolismo , Tubulina (Proteína)/metabolismo , Antígenos CD2 , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Dimerização , Imunofluorescência , Humanos , Interfase/fisiologia , Células K562 , Proteínas de Membrana , Microtúbulos/fisiologia , Mitose/fisiologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Ligação Proteica , Estrutura Secundária de Proteína/genética , Estrutura Secundária de Proteína/fisiologia , Proteínas Recombinantes , Fuso Acromático/metabolismo , Fuso Acromático/fisiologia , Estatmina , Ressonância de Plasmônio de Superfície , Tubulina (Proteína)/fisiologia
4.
Mol Biol Cell ; 25(10): 1594-607, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24648497

RESUMO

Septins are filament-forming proteins important for organizing the cortex of animal and fungal cells. In mammals, 13 septin paralogues were recently shown to assemble into core heterohexamer and heterooctamer complexes, which serve as building blocks for apolar filamentous structures that differ among cell types. To determine how tissue-specific septin paralogue expression may shape core heteromer repertoires and thereby modulate properties of septin filaments, we devised protocols to analyze native septin heteromers with distinct numbers of subunits. Our evidence based on genetically manipulated human cells supports and extends recent concepts of homology subgroup-restricted assembly into distinct categories of apolar heterohexamers and heterooctamers. We also identify a category of tetramers that have a subunit composition equivalent to an octameric building block. These atypical tetramers are prevalent in lymphocytes and neural tissues, in which octamers are abundant but hexamers are rare. Our results can be explained by tissue-specific expression of SEPT3 subgroup members: SEPT3, SEPT9, and SEPT12. These serve as cognate subunits in either heterooctamers or atypical tetramers but exhibit different preferences in various tissues. The identified tissue-specific repertoires of septin heteromers provide insights into how higher-order septin structures with differential properties and stabilities may form in diverse animal cell types.


Assuntos
Agregados Proteicos/fisiologia , Septinas/metabolismo , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Humanos , Células Jurkat , Neurônios/metabolismo , Polimerização , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína , Interferência de RNA , RNA Interferente Pequeno , Septinas/biossíntese , Septinas/genética , Linfócitos T/metabolismo
5.
Mol Biol Cell ; 23(21): 4242-55, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956766

RESUMO

Septin-family proteins assemble into rod-shaped heteromeric complexes that form higher-order arrangements at the cell cortex, where they serve apparently conserved functions as diffusion barriers and molecular scaffolds. There are 13 confirmed septin paralogues in mammals, which may be ubiquitous or tissue specific. Septin hetero-oligomerization appears homology subgroup directed, which in turn determines the subunit arrangement of six- to eight-subunit core heteromers. Here we address functional properties of human SEPT9, which, due to variable mRNA splicing, exists as multiple isoforms that differ between tissues. Myeloid K562 cells express three SEPT9 isoforms, all of which have an equal propensity to hetero-oligomerize with SEPT7-containing hexamers to generate octameric heteromers. However, due to limiting amounts of SEPT9, K562 cells contain both hexameric and octameric heteromers. To generate cell lines with controllable hexamer-to-octamer ratios and that express single SEPT9 isoforms, we developed a gene product replacement strategy. By this means we identified SEPT9 isoform-specific properties that either facilitate septin heteromer polymerization along microtubules or modulate the size range of submembranous septin disks-a prevalent septin structure in nonadhered cells. Our findings show that the SEPT9 expression level directs the hexamer-to-octamer ratio, and that the isoform composition and expression level together determine higher-order arrangements of septins.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mamíferos/metabolismo , Microtúbulos/metabolismo , Multimerização Proteica , Septinas/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Humanos , Interfase/efeitos dos fármacos , Células K562 , Microtúbulos/efeitos dos fármacos , Concentração Osmolar , Paclitaxel/farmacologia , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerização Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Septinas/química , Septinas/genética , Solubilidade
6.
Mol Biol Cell ; 22(17): 3152-64, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21737677

RESUMO

Septins are conserved GTP-binding proteins that assemble into lateral diffusion barriers and molecular scaffolds. Vertebrate genomes contain 9-17 septin genes that encode both ubiquitous and tissue-specific septins. Expressed septins may assemble in various combinations through both heterotypic and homotypic G-domain interactions. However, little is known regarding assembly states of mammalian septins and mechanisms directing ordered assembly of individual septins into heteromeric units, which is the focus of this study. Our analysis of the septin system in cells lacking or overexpressing selected septins reveals interdependencies coinciding with previously described homology subgroups. Hydrodynamic and single-particle data show that individual septins exist solely in the context of stable six- to eight-subunit core heteromers, all of which contain SEPT2 and SEPT6 subgroup members and SEPT7, while heteromers comprising more than six subunits also contain SEPT9. The combined data suggest a generic model for how the temporal order of septin assembly is homology subgroup-directed, which in turn determines the subunit arrangement of native heteromers. Because mammalian cells normally express multiple members and/or isoforms of some septin subgroups, our data also suggest that only a minor fraction of native heteromers are arranged as perfect palindromes.


Assuntos
Complexos Multiproteicos/metabolismo , Multimerização Proteica , Septinas/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos , Microscopia Eletrônica de Transmissão , Peso Molecular , Complexos Multiproteicos/isolamento & purificação , Complexos Multiproteicos/ultraestrutura , Ligação Proteica , Estabilidade Proteica , Interferência de RNA , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Septinas/química , Septinas/genética , Solubilidade
7.
Mol Biol Cell ; 22(23): 4588-601, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21998205

RESUMO

Septin family proteins oligomerize through guanosine 5'-triphosphate-binding domains into core heteromers, which in turn polymerize at the cleavage furrow of dividing fungal and animal cells. Septin assemblies during the interphase of animal cells remain poorly defined and are the topic of this report. In this study, we developed protocols for visualization of authentic higher-order assemblies using tagged septins to effectively replace the endogenous gene product within septin core heteromers in human cells. Our analysis revealed that septins assemble into microtubule-supported, disk-like structures at the plasma membrane. In the absence of cell substrate adhesion, this is the predominant higher-order arrangement in interphase cells and each of the seven to eight septin family members expressed by the two analyzed cell types appears equally represented. However, studies of myeloid and lymphoid cell model systems revealed cell type-specific alterations of higher-order septin arrangements in response to substrate adhesion. Live-cell observations suggested that all higher-order septin assemblies are mutually exclusive with plasma membrane regions undergoing remodeling. The combined data point to a mechanism by which densely arranged cortical microtubules, which are typical for nonadhered spherical cells, support plasma membrane-bound, disk-like septin assemblies.


Assuntos
Membrana Celular/metabolismo , Microtúbulos/metabolismo , Septinas/metabolismo , Adesão Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Membrana Celular/ultraestrutura , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células Jurkat , Células K562 , Leucemia Eritroblástica Aguda/patologia , Permeabilidade , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Septinas/genética , Linfócitos T/metabolismo , Linfócitos T/ultraestrutura
8.
Exp Cell Res ; 314(6): 1367-77, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18262179

RESUMO

Op18/stathmin (Op18) is a phosphorylation-regulated and differentially expressed microtubule-destabilizing protein in animal cells. Op18 regulates tubulin monomer-polymer partitioning of the interphase microtubule system and forms complexes with tubulin heterodimers. Recent reports have shown that specific tubulin-folding cofactors and related proteins may disrupt tubulin heterodimers. We therefore investigated whether Op18 protects unpolymerized tubulin from such disruptive activities. Our approach was based on inducible overexpression of two tubulin-disrupting proteins, namely TBCE, which is required for tubulin biogenesis, and E-like, which has been proposed to regulate tubulin turnover and microtubule stability. Expression of either of these proteins was found to cause a rapid degradation of both alpha-tubulin and beta-tubulin subunits of unpolymerized, but not polymeric, tubulin heterodimers. We found that depletion of Op18 by means of RNA interference increased the susceptibility of tubulin to TBCE or E-like mediated disruption, while overexpressed Op18 exerted a tubulin-protective effect. Tubulin protection was shown to depend on Op18 levels, binding affinity, and the partitioning between tubulin monomers and polymers. Hence, the present study reveals that Op18 at physiologically relevant levels functions to preserve the integrity of tubulin heterodimers, which may serve to regulate tubulin turnover rates.


Assuntos
Leucemia/metabolismo , Leucemia/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Chaperonas Moleculares/metabolismo , Estatmina/metabolismo , Dimerização , Regulação Leucêmica da Expressão Gênica , Humanos , Células K562 , Cinesinas/metabolismo , Leucemia/genética , Proteínas de Membrana/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estatmina/genética , Tubulina (Proteína)/metabolismo
9.
Mol Biol Cell ; 19(7): 2897-906, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18434595

RESUMO

Op18/stathmin (Op18), a conserved microtubule-depolymerizing and tubulin heterodimer-binding protein, is a major interphase regulator of tubulin monomer-polymer partitioning in diverse cell types in which Op18 is abundant. Here, we addressed the question of whether the microtubule regulatory function of Op18 includes regulation of tubulin heterodimer synthesis. We used two human cell model systems, K562 and Jurkat, combined with strategies for regulatable overexpression or depletion of Op18. Although Op18 depletion caused extensive overpolymerization and increased microtubule content in both cell types, we did not detect any alteration in polymer stability. Interestingly, however, we found that Op18 mediates positive regulation of tubulin heterodimer content in Jurkat cells, which was not observed in K562 cells. By analysis of cells treated with microtubule-poisoning drugs, we found that Jurkat cells regulate tubulin mRNA levels by a posttranscriptional mechanism similarly to normal primary cells, whereas this mechanism is nonfunctional in K562 cells. We present evidence that Op18 mediates posttranscriptional regulation of tubulin mRNA in Jurkat cells through the same basic autoregulatory mechanism as microtubule-poisoning drugs. This, combined with potent regulation of tubulin monomer-polymer partitioning, enables Op18 to exert global regulation of the microtubule system.


Assuntos
Regulação da Expressão Gênica , Interfase , Proteínas dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Estatmina/metabolismo , Colchicina/farmacologia , Dimerização , Humanos , Células Jurkat , Células K562 , Fenótipo , Polímeros/química , Processamento Pós-Transcricional do RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tubulina (Proteína)/química
10.
EMBO J ; 24(6): 1256-66, 2005 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-15775983

RESUMO

MCAK, a member of the kinesin-13 family, is a microtubule (MT) depolymerase that is necessary to ensure proper kinetochore MT attachment during spindle formation. Regulation of MCAK activity and localization is controlled in part by Aurora B kinase at the centromere. Here we analyzed human cells depleted of the ubiquitous Ca(2+)/calmodulin-dependent protein kinase IIgamma isoform (CaMKIIgamma) by RNA interference and found that CaMKIIgamma was necessary to suppress MCAK depolymerase activity in vivo. A functional overlap with TOGp, a MT regulator known to counteract MCAK, was suggested by similar CaMKIIgamma- and TOGp-depletion phenotypes, namely disorganized multipolar spindles. A replicating vector system, which permits inducible overexpression in cells that simultaneously synthesize interfering short hairpin RNAs, was used to dissect the functional interplay between CaMKIIgamma, TOGp, and MCAK. Our results revealed two distinct but functionally overlapping mechanisms for negative regulation of the cytosolic/centrosomal pool of MCAK. These two mechanisms, involving CaMKIIgamma and TOGp, respectively, are both essential for spindle bipolarity in a normal physiological context, but not in MCAK-depleted cells.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Cinesinas/metabolismo , Fuso Acromático/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Divisão Celular/genética , Divisão Celular/fisiologia , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Conformação de Ácido Nucleico , Interferência de RNA , RNA Interferente Pequeno/genética
11.
EMBO J ; 23(3): 627-37, 2004 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-14749730

RESUMO

XMAP215/TOGp family members and KinI kinesins are conserved microtubule (MT)-regulatory proteins, and have been viewed as possessing prominent antagonistic stabilizing/destabilizing activities that must be balanced. Here, interdependencies between TOGp and the KinI kinesin MCAK were analyzed in human leukemia cells. A system was established that permits inducible overexpression in homogeneous cell populations that simultaneously synthesize interfering short hairpin RNAs. We present evidence that the functional interplay of TOGp and MCAK proteins is manifested as three distinct phenotypes during the cell cycle. The first involves a role for TOGp in protecting spindle MTs from MCAK activity at the centrosome, which appears essential to prevent the formation of disorganized multipolar spindles. The second phenotype involves TOGp-dependent counteraction of excessive MCAK activity during mitosis, which recapitulates the previously established plus-end specific counteractive activities in vitro. The third involves an unexpected destabilization of the interphase MTs by overexpressed TOGp, a phenotype that requires endogenous MCAK. We hypothesize that TOGp-dependent prevention of MCAK-mediated spindle disorganization, as evidenced by depletion experiments, reflects a primary physiological role for TOGp in human somatic cells.


Assuntos
Interfase/fisiologia , Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/fisiologia , Fuso Acromático/metabolismo , Humanos , Células K562
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA