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










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 6984, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34848715

RESUMO

Eukaryotic cells have evolved highly orchestrated protein catabolic machineries responsible for the timely and selective disposal of proteins and organelles, thereby ensuring amino acid recycling. However, how protein degradation is coordinated with amino acid supply and protein synthesis has remained largely elusive. Here we show that the mammalian proteasome undergoes liquid-liquid phase separation in the nucleus upon amino acid deprivation. We termed these proteasome condensates SIPAN (Starvation-Induced Proteasome Assemblies in the Nucleus) and show that these are a common response of mammalian cells to amino acid deprivation. SIPAN undergo fusion events, rapidly exchange proteasome particles with the surrounding milieu and quickly dissolve following amino acid replenishment. We further show that: (i) SIPAN contain K48-conjugated ubiquitin, (ii) proteasome inhibition accelerates SIPAN formation, (iii) deubiquitinase inhibition prevents SIPAN resolution and (iv) RAD23B proteasome shuttling factor is required for SIPAN formation. Finally, SIPAN formation is associated with decreased cell survival and p53-mediated apoptosis, which might contribute to tissue fitness in diverse pathophysiological conditions.


Assuntos
Aminoácidos/metabolismo , Apoptose/fisiologia , Núcleo Celular/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inanição , Animais , Autoantígenos , Linhagem Celular Tumoral , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Eucarióticas , Exercício Físico , Fibroblastos , Humanos , Camundongos , Nutrientes , Biossíntese de Proteínas , Proteólise , Estresse Fisiológico , Ubiquitina
2.
Nat Commun ; 12(1): 3637, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131133

RESUMO

KIF14 is a mitotic kinesin whose malfunction is associated with cerebral and renal developmental defects and several cancers. Like other kinesins, KIF14 couples ATP hydrolysis and microtubule binding to the generation of mechanical work, but the coupling mechanism between these processes is still not fully clear. Here we report 20 high-resolution (2.7-3.9 Å) cryo-electron microscopy KIF14-microtubule structures with complementary functional assays. Analysis procedures were implemented to separate coexisting conformations of microtubule-bound monomeric and dimeric KIF14 constructs. The data provide a comprehensive view of the microtubule and nucleotide induced KIF14 conformational changes. It shows that: 1) microtubule binding, the nucleotide species, and the neck-linker domain govern the transition between three major conformations of the motor domain; 2) an undocked neck-linker prevents the nucleotide-binding pocket to fully close and dampens ATP hydrolysis; 3) 13 neck-linker residues are required to assume a stable docked conformation; 4) the neck-linker position controls the hydrolysis rather than the nucleotide binding step; 5) the two motor domains of KIF14 dimers adopt distinct conformations when bound to the microtubule; and 6) the formation of the two-heads-bound-state introduces structural changes in both motor domains of KIF14 dimers. These observations provide the structural basis for a coordinated chemo-mechanical kinesin translocation model.


Assuntos
Cinesinas/química , Cinesinas/metabolismo , Proteínas Oncogênicas/química , Proteínas Oncogênicas/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação , Microscopia Crioeletrônica , Cinesinas/genética , Ligantes , Camundongos , Microtúbulos/química , Microtúbulos/genética , Microtúbulos/metabolismo , Simulação de Acoplamento Molecular , Proteínas Oncogênicas/genética , Ligação Proteica , Conformação Proteica , Domínios Proteicos
3.
Development ; 147(14)2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32665245

RESUMO

Amputation of a salamander limb triggers a regeneration process that is perfect. A limited number of genes have been studied in this context and even fewer have been analyzed functionally. In this work, we use the BMP signaling inhibitor LDN193189 on Ambystoma mexicanum to explore the role of BMPs in regeneration. We find that BMP signaling is required for proper expression of various patterning genes and that its inhibition causes major defects in the regenerated limbs. Fgf8 is downregulated when BMP signaling is blocked, but ectopic injection of either human or axolotl protein did not rescue the defects. By administering LDN193189 treatments at different time points during regeneration, we show clearly that limb regeneration progresses in a proximal to distal fashion. This demonstrates that BMPs play a major role in patterning of regenerated limbs and that regeneration is a progressive process like development.


Assuntos
Ambystoma mexicanum/metabolismo , Proteínas de Anfíbios/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Extremidades/fisiologia , Regeneração/fisiologia , Transdução de Sinais , Ambystoma mexicanum/crescimento & desenvolvimento , Proteínas de Anfíbios/genética , Animais , Proteínas Morfogenéticas Ósseas/genética , Proliferação de Células/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Fator de Transcrição MSX1/genética , Fator de Transcrição MSX1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo
4.
Elife ; 92020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31951198

RESUMO

DNA double strand breaks (DSBs) have detrimental effects on cell survival and genomic stability, and are related to cancer and other human diseases. In this study, we identified microtubule-depolymerizing kinesin Kif2C as a protein associated with DSB-mimicking DNA templates and known DSB repair proteins in Xenopus egg extracts and mammalian cells. The recruitment of Kif2C to DNA damage sites was dependent on both PARP and ATM activities. Kif2C knockdown or knockout led to accumulation of endogenous DNA damage, DNA damage hypersensitivity, and reduced DSB repair via both NHEJ and HR. Interestingly, Kif2C depletion, or inhibition of its microtubule depolymerase activity, reduced the mobility of DSBs, impaired the formation of DNA damage foci, and decreased the occurrence of foci fusion and resolution. Taken together, our study established Kif2C as a new player of the DNA damage response, and presented a new mechanism that governs DSB dynamics and repair.


DNA can be damaged in many ways, and a double strand break is one of the most dangerous. This occurs when both strands of the double helix snap at the same time, leaving two broken ends. When cells detect this kind of damage, they race to get it fixed as quickly as possible. Fixing these double strand breaks is thought to involve the broken ends being moved to 'repair centers' in the nucleus of the cell, but it was unclear how the broken ends were moved. One possibility was that the cells transport the broken ends along protein filaments called microtubules. Cells can assemble these track-like filaments on-demand to carry cargo attached to molecular motors called kinesins. However, this type of transport happens outside of the cell's nucleus, and while there are different kinesin proteins localized inside the nucleus, their roles are largely unknown. In an effort to understand how broken DNA ends are repaired, Zhu, Paydar et al. conducted experiments that simulated double strand breaks and examined the proteins that responded. The first set of experiments involved mixing cut pieces of DNA with extracts taken from frog eggs or human cells. Zhu, Paydar et al. found that one kinesin called Kif2C stuck to the DNA fragments, and attached to many proteins known to play a role in DNA damage repair. Kif2C had previously been shown to help separate the chromosomes during cell division. To find out more about its potential role in DNA repair, Zhu, Paydar et al. then used a laser to create breaks in the DNA of living human cells and tracked Kif2C movement. The kinesin arrived within 60 seconds of the DNA damage and appeared to transport the cut DNA ends to 'repair centers'. Getting rid of Kif2C, or blocking its activity, had dire effects on the cells' abilities to mobilize and repair breaks to its DNA. Without the molecular motor, fewer double strand breaks were repaired, and so DNA damage started to build up. Defects in double strand break repair happen in many human diseases, including cancer. Many cancer treatments damage the DNA of cancer cells, sometimes in combination with drugs that stop cells from building and using their microtubule transport systems. Understanding the new role of Kif2C in DNA damage repair could therefore help optimize these treatment combinations.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Recombinação Homóloga , Cinesinas/fisiologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microtúbulos/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Ligação Proteica , Xenopus
5.
Hum Mol Genet ; 29(5): 766-784, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-31919497

RESUMO

By using the Cre-mediated genetic switch technology, we were able to successfully generate a conditional knock-in mouse, bearing the KIF2A p.His321Asp missense point variant, identified in a subject with malformations of cortical development. These mice present with neuroanatomical anomalies and microcephaly associated with behavioral deficiencies and susceptibility to epilepsy, correlating with the described human phenotype. Using the flexibility of this model, we investigated RosaCre-, NestinCre- and NexCre-driven expression of the mutation to dissect the pathophysiological mechanisms underlying neurodevelopmental cortical abnormalities. We show that the expression of the p.His321Asp pathogenic variant increases apoptosis and causes abnormal multipolar to bipolar transition in newborn neurons, providing therefore insights to better understand cortical organization and brain growth defects that characterize KIF2A-related human disorders. We further demonstrate that the observed cellular phenotypes are likely to be linked to deficiency in the microtubule depolymerizing function of KIF2A.


Assuntos
Comportamento Animal , Cinesinas/fisiologia , Malformações do Desenvolvimento Cortical/patologia , Mutação , Neurônios/patologia , Proteínas Repressoras/fisiologia , Animais , Masculino , Malformações do Desenvolvimento Cortical/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo
6.
Proc Natl Acad Sci U S A ; 116(48): 24056-24065, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31712417

RESUMO

Viruses have transformed our understanding of mammalian RNA processing, including facilitating the discovery of the methyl-7-guanosine (m7G) cap on the 5' end of RNAs. The m7G cap is required for RNAs to bind the eukaryotic translation initiation factor eIF4E and associate with the translation machinery across plant and animal kingdoms. The potyvirus-derived viral genome-linked protein (VPg) is covalently bound to the 5' end of viral genomic RNA (gRNA) and associates with host eIF4E for successful infection. Divergent models to explain these observations proposed either an unknown mode of eIF4E engagement or a competition of VPg for the m7G cap-binding site. To dissect these possibilities, we resolved the structure of VPg, revealing a previously unknown 3-dimensional (3D) fold, and characterized the VPg-eIF4E complex using NMR and biophysical techniques. VPg directly bound the cap-binding site of eIF4E and competed for m7G cap analog binding. In human cells, VPg inhibited eIF4E-dependent RNA export, translation, and oncogenic transformation. Moreover, VPg formed trimeric complexes with eIF4E-eIF4G, eIF4E bound VPg-luciferase RNA conjugates, and these VPg-RNA conjugates were templates for translation. Informatic analyses revealed structural similarities between VPg and the human kinesin EG5. Consistently, EG5 directly bound eIF4E in a similar manner to VPg, demonstrating that this form of engagement is relevant beyond potyviruses. In all, we revealed an unprecedented modality for control and engagement of eIF4E and show that VPg-RNA conjugates functionally engage eIF4E. As such, potyvirus VPg provides a unique model system to interrogate eIF4E.


Assuntos
Fator de Iniciação 4E em Eucariotos/química , Potyvirus/genética , Biossíntese de Proteínas/fisiologia , RNA/química , Ribonucleoproteínas/química , Proteínas Virais/química , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Fator de Iniciação 4E em Eucariotos/metabolismo , Humanos , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Dobramento de Proteína , Capuzes de RNA/química , Processamento Pós-Transcricional do RNA , Ribonucleoproteínas/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/fisiologia
7.
Hum Mol Genet ; 28(5): 778-795, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30388224

RESUMO

Mutations in KIF14 have previously been associated with either severe, isolated or syndromic microcephaly with renal hypodysplasia (RHD). Syndromic microcephaly-RHD was strongly reminiscent of clinical ciliopathies, relating to defects of the primary cilium, a signalling organelle present on the surface of many quiescent cells. KIF14 encodes a mitotic kinesin, which plays a key role at the midbody during cytokinesis and has not previously been shown to be involved in cilia-related functions. Here, we analysed four families with fetuses presenting with the syndromic form and harbouring biallelic variants in KIF14. Our functional analyses showed that the identified variants severely impact the activity of KIF14 and likely correspond to loss-of-function mutations. Analysis in human fetal tissues further revealed the accumulation of KIF14-positive midbody remnants in the lumen of ureteric bud tips indicating a shared function of KIF14 during brain and kidney development. Subsequently, analysis of a kif14 mutant zebrafish line showed a conserved role for this mitotic kinesin. Interestingly, ciliopathy-associated phenotypes were also present in mutant embryos, supporting a potential direct or indirect role for KIF14 at cilia. However, our in vitro and in vivo analyses did not provide evidence of a direct role for KIF14 in ciliogenesis and suggested that loss of kif14 causes ciliopathy-like phenotypes through an accumulation of mitotic cells in ciliated tissues. Altogether, our results demonstrate that KIF14 mutations result in a severe syndrome associating microcephaly and RHD through its conserved function in cytokinesis during kidney and brain development.


Assuntos
Anormalidades Congênitas/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Nefropatias/congênito , Rim/anormalidades , Cinesinas/genética , Mutação com Perda de Função , Microcefalia/genética , Proteínas Oncogênicas/genética , Animais , Anormalidades Congênitas/metabolismo , Citocinese/genética , Modelos Animais de Doenças , Feminino , Imunofluorescência , Genes Letais , Estudos de Associação Genética/métodos , Loci Gênicos , Humanos , Rim/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Masculino , Microcefalia/metabolismo , Microcefalia/patologia , Proteínas Oncogênicas/química , Proteínas Oncogênicas/metabolismo , Linhagem , Fenótipo , Relação Estrutura-Atividade , Peixe-Zebra
8.
Nat Commun ; 9(1): 2628, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29980677

RESUMO

Kinesin-13 proteins are major microtubule (MT) regulatory factors that catalyze removal of tubulin subunits from MT ends. The class-specific "neck" and loop 2 regions of these motors are required for MT depolymerization, but their contributing roles are still unresolved because their interactions with MT ends have not been observed directly. Here we report the crystal structure of a catalytically active kinesin-13 monomer (Kif2A) in complex with two bent αß-tubulin heterodimers in a head-to-tail array, providing a view of these interactions. The neck of Kif2A binds to one tubulin dimer and the motor core to the other, guiding insertion of the KVD motif of loop 2 in between them. AMPPNP-bound Kif2A can form stable complexes with tubulin in solution and trigger MT depolymerization. We also demonstrate the importance of the neck in modulating ATP turnover and catalytic depolymerization of MTs. These results provide mechanistic insights into the catalytic cycles of kinesin-13.


Assuntos
Cinesinas/metabolismo , Microtúbulos/metabolismo , Polimerização , Multimerização Proteica , Tubulina (Proteína)/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Humanos , Cinesinas/química , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/metabolismo
10.
J Mol Biol ; 429(5): 715-731, 2017 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-28147230

RESUMO

Cytokinesis of animal cells requires the assembly of a contractile ring, which promotes daughter cell splitting. Anillin is a conserved scaffold protein involved in organizing the structural components of the contractile ring including filamentous actin (F-actin), myosin, and septins and in forming the subsequent midbody ring. Like other metazoan homologs, Drosophila anillin contains a conserved domain that can bind and bundle F-actin, but the importance and molecular details of its interaction with F-actin remain unclear. Here, we show that in a depletion-and-rescue assay in Drosophila S2 cells, anillin lacking the entire actin-binding domain (ActBD) exhibits defective cortical localization during mitosis and a greatly diminished ability to support cytokinesis. Using in vitro binding assays and electron microscopy on recombinant fragments, we determine that the anillin ActBD harbors three distinct actin-binding sites (ABS 1-3). We show that each ABS binds to a distinct place on F-actin. Importantly, ABS1 and ABS3 partially overlap on the surface of actin and, therefore, interact with F-actin in a mutually exclusive fashion. Although ABS2 and ABS3 are sufficient for bundling, ABS1 contributes to the overall F-actin bundling activity of anillin and enables anillin to switch between two actin-bundling morphologies and promote the formation of three-dimensional F-actin bundles. Finally, we show that in live S2 cells, ABS2 and ABS3 are each required and together sufficient for the robust cortical localization of the ActBD during cytokinesis. Collectively, our structural, biochemical, and cell biological data suggest that multiple anillin-actin interaction modes promote the faithful progression of cytokinesis.


Assuntos
Actinas/metabolismo , Proteínas Contráteis/metabolismo , Citocinese , Domínios e Motivos de Interação entre Proteínas , Animais , Drosophila/metabolismo , Processamento de Imagem Assistida por Computador , Mitose , Miosinas , Septinas
11.
J Clin Invest ; 126(12): 4569-4584, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27797342

RESUMO

Current chemotherapies for T cell acute lymphoblastic leukemia (T-ALL) efficiently reduce tumor mass. Nonetheless, disease relapse attributed to survival of preleukemic stem cells (pre-LSCs) is associated with poor prognosis. Herein, we provide direct evidence that pre-LSCs are much less chemosensitive to existing chemotherapy drugs than leukemic blasts because of a distinctive lower proliferative state. Improving therapies for T-ALL requires the development of strategies to target pre-LSCs that are absolutely dependent on their microenvironment. Therefore, we designed a robust protocol for high-throughput screening of compounds that target primary pre-LSCs maintained in a niche-like environment, on stromal cells that were engineered for optimal NOTCH1 activation. The multiparametric readout takes into account the intrinsic complexity of primary cells in order to specifically monitor pre-LSCs, which were induced here by the SCL/TAL1 and LMO1 oncogenes. We screened a targeted library of compounds and determined that the estrogen derivative 2-methoxyestradiol (2-ME2) disrupted both cell-autonomous and non-cell-autonomous pathways. Specifically, 2-ME2 abrogated pre-LSC viability and self-renewal activity in vivo by inhibiting translation of MYC, a downstream effector of NOTCH1, and preventing SCL/TAL1 activity. In contrast, normal hematopoietic stem/progenitor cells remained functional. These results illustrate how recapitulating tissue-like properties of primary cells in high-throughput screening is a promising avenue for innovation in cancer chemotherapy.


Assuntos
Estradiol/análogos & derivados , Células-Tronco Neoplásicas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Microambiente Tumoral/efeitos dos fármacos , 2-Metoxiestradiol , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Estradiol/farmacologia , Humanos , Células Jurkat , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Camundongos , Células-Tronco Neoplásicas/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Microambiente Tumoral/genética , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cell Rep ; 17(7): 1755-1763, 2016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27829147

RESUMO

Karyotype diversity is a hallmark of solid tumors that contributes to intratumor heterogeneity. This diversity is generated by persistent chromosome mis-segregation associated with chromosomal instability (CIN). CIN correlates with tumor relapse and is thought to promote drug resistance by creating a vast genomic landscape through which karyotypically unique clones survive lethal drug selection. We explore this proposition using a small molecule (UMK57) that suppresses chromosome mis-segregation in CIN cancer cells by potentiating the activity of the kinesin-13 protein MCAK. Sublethal doses of UMK57 destabilize kinetochore-microtubule (k-MT) attachments during mitosis to increase chromosome segregation fidelity. Surprisingly, chromosome mis-segregation rebounds in UMK57-treated cancer cells within a few days. This rapid relapse is driven by alterations in the Aurora B signaling pathway that hyper-stabilize k-MT attachments and is reversible following UMK57 removal. Thus, cancer cells display adaptive resistance to therapies targeting CIN through rapid and reversible changes to mitotic signaling networks.


Assuntos
Antineoplásicos/farmacologia , Instabilidade Cromossômica/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias/patologia , Aurora Quinase B/metabolismo , Linhagem Celular Tumoral , Cromossomos Humanos/metabolismo , Humanos , Cinesinas/metabolismo
13.
J Cell Biol ; 207(2): 201-11, 2014 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-25332165

RESUMO

Drosophila melanogaster Polo and its human orthologue Polo-like kinase 1 fulfill essential roles during cell division. Members of the Polo-like kinase (Plk) family contain an N-terminal kinase domain (KD) and a C-terminal Polo-Box domain (PBD), which mediates protein interactions. How Plks are regulated in cytokinesis is poorly understood. Here we show that phosphorylation of Polo by Aurora B is required for cytokinesis. This phosphorylation in the activation loop of the KD promotes the dissociation of Polo from the PBD-bound microtubule-associated protein Map205, which acts as an allosteric inhibitor of Polo kinase activity. This mechanism allows the release of active Polo from microtubules of the central spindle and its recruitment to the site of cytokinesis. Failure in Polo phosphorylation results in both early and late cytokinesis defects. Importantly, the antagonistic regulation of Polo by Aurora B and Map205 in cytokinesis reveals that interdomain allosteric mechanisms can play important roles in controlling the cellular functions of Plks.


Assuntos
Aurora Quinase B/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/citologia , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Aurora Quinase B/metabolismo , Células Cultivadas , Citocinese , Proteínas de Drosophila/análise , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Modelos Moleculares , Fosforilação , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/fisiologia
14.
J Biol Chem ; 289(40): 27418-31, 2014 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-25135640

RESUMO

Structural maintenance of chromosome (SMC) proteins are key organizers of chromosome architecture and are essential for genome integrity. They act by binding to chromatin and connecting distinct parts of chromosomes together. Interestingly, their potential role in providing connections between chromatin and the mitotic spindle has not been explored. Here, we show that yeast SMC proteins bind directly to microtubules and can provide a functional link between microtubules and DNA. We mapped the microtubule-binding region of Smc5 and generated a mutant with impaired microtubule binding activity. This mutant is viable in yeast but exhibited a cold-specific conditional lethality associated with mitotic arrest, aberrant spindle structures, and chromosome segregation defects. In an in vitro reconstitution assay, this Smc5 mutant also showed a compromised ability to protect microtubules from cold-induced depolymerization. Collectively, these findings demonstrate that SMC proteins can bind to and stabilize microtubules and that SMC-microtubule interactions are essential to establish a robust system to maintain genome integrity.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Instabilidade Genômica , Microtúbulos/química , Microtúbulos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Sítios de Ligação , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Genoma Fúngico , Microtúbulos/genética , Ligação Proteica , Estabilidade Proteica , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
15.
J Mol Biol ; 426(17): 2997-3015, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-24949858

RESUMO

The mitotic kinesin motor protein KIF14 is essential for cytokinesis during cell division and has been implicated in cerebral development and a variety of human cancers. Here we show that the mouse KIF14 motor domain binds tightly to microtubules and does not display typical nucleotide-dependent changes in this affinity. It also has robust ATPase activity but very slow motility. A crystal structure of the ADP-bound form of the KIF14 motor domain reveals a dramatically opened ATP-binding pocket, as if ready to exchange its bound ADP for Mg·ATP. In this state, the central ß-sheet is twisted ~10° beyond the maximal amount observed in other kinesins. This configuration has only been seen in the nucleotide-free states of myosins-known as the "rigor-like" state. Fitting of this atomic model to electron density maps from cryo-electron microscopy indicates a distinct binding configuration of the motor domain to microtubules. We postulate that these properties of KIF14 are well suited for stabilizing midbody microtubules during cytokinesis.


Assuntos
Cinesinas/química , Microtúbulos/química , Difosfato de Adenosina/química , Adenosina Trifosfatases/química , Sequência de Aminoácidos , Animais , Domínio Catalítico , Cristalografia por Raios X , Cinética , Camundongos , Microtúbulos/ultraestrutura , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Multimerização Proteica , Estabilidade Proteica , Estrutura Secundária de Proteína
16.
FEBS Lett ; 588(14): 2315-20, 2014 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-24859087

RESUMO

The kinesin-13 family of microtubule depolymerases is a major regulator of microtubule dynamics. RNA interference-induced knockdown studies have highlighted their importance in many cell division processes including spindle assembly and chromosome segregation. Since microtubule turnovers and most mitotic events are relatively rapid (in minutes or seconds), developing tools that offer faster control over protein functions is therefore essential to more effectively interrogate kinesin-13 activities in living cells. Here, we report the identification and characterization of a selective allosteric kinesin-13 inhibitor, DHTP. Using high resolution microscopy, we show that DHTP is cell permeable and can modulate microtubule dynamics in cells.


Assuntos
Cinesinas/antagonistas & inibidores , Pirimidinas/química , Tiazolidinas/química , Moduladores de Tubulina/química , Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Regulação Alostérica , Animais , Bovinos , Avaliação Pré-Clínica de Medicamentos , Humanos , Cinesinas/química , Microtúbulos/química , Multimerização Proteica
17.
J Cell Biol ; 202(2): 251-60, 2013 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-23857773

RESUMO

Ezrin, Radixin, and Moesin (ERM) proteins play important roles in many cellular processes including cell division. Recent studies have highlighted the implications of their metastatic potential in cancers. ERM's role in these processes is largely attributed to their ability to link actin filaments to the plasma membrane. In this paper, we show that the ERM protein Moesin directly binds to microtubules in vitro and stabilizes microtubules at the cell cortex in vivo. We identified two evolutionarily conserved residues in the FERM (4.1 protein and ERM) domains of ERMs that mediated the association with microtubules. This ERM-microtubule interaction was required for regulating spindle organization in metaphase and cell shape transformation after anaphase onset but was dispensable for bridging actin filaments to the metaphase cortex. These findings provide a molecular framework for understanding the complex functional interplay between the microtubule and actin cytoskeletons mediated by ERM proteins in mitosis and have broad implications in both physiological and pathological processes that require ERMs.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Microtúbulos/metabolismo , Citoesqueleto de Actina/genética , Anáfase , Animais , Linhagem Celular , Membrana Celular/metabolismo , Clonagem Molecular , Proteínas do Citoesqueleto/genética , Drosophila/genética , Drosophila/metabolismo , Humanos , Interfase , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/genética , Metáfase , Microtúbulos/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/metabolismo , Fuso Acromático/genética , Fuso Acromático/metabolismo , Proteína Vermelha Fluorescente
18.
J Cell Biol ; 182(3): 421-8, 2008 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-18678707

RESUMO

Although assembly of the mitotic spindle is known to be a precisely controlled process, regulation of the key motor proteins involved remains poorly understood. In eukaryotes, homotetrameric kinesin-5 motors are required for bipolar spindle formation. Eg5, the vertebrate kinesin-5, has two modes of motion: an adenosine triphosphate (ATP)-dependent directional mode and a diffusive mode that does not require ATP hydrolysis. We use single-molecule experiments to examine how the switching between these modes is controlled. We find that Eg5 diffuses along individual microtubules without detectable directional bias at close to physiological ionic strength. Eg5's motility becomes directional when bound between two microtubules. Such activation through binding cargo, which, for Eg5, is a second microtubule, is analogous to known mechanisms for other kinesins. In the spindle, this might allow Eg5 to diffuse on single microtubules without hydrolyzing ATP until the motor is activated by binding to another microtubule. This mechanism would increase energy and filament cross-linking efficiency.


Assuntos
Cinesinas/metabolismo , Microtúbulos/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Difusão , Dimerização , Proteínas de Fluorescência Verde/metabolismo , Cinesinas/química , Concentração Osmolar , Estrutura Quaternária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Suínos , Xenopus , Proteínas de Xenopus/química
19.
Anal Chem ; 80(7): 2419-25, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18321079

RESUMO

We describe a rapid and efficient method for the identification of phosphopeptides, which we term mass spectrometric (MS) phosphopeptide fingerprinting. The method involves quantitative comparison of proteolytic peptides from native versus completely dephosphorylated proteins. Dephosphorylation of serine, threonine, and tyrosine residues is achieved by in-gel treatment of the separated proteins with hydrogen fluoride (HF). This chemical dephosphorylation results in enrichment of those unmodified peptides that correspond to previously phosphorylated peptides. Quantitative comparison of the signal-to-noise ratios of peaks in the treated versus untreated samples are used to identify phosphopeptides, which can be confirmed and further studied by tandem mass spectrometry (MS/MS). We have applied this method to identify eight known phosphorylation sites of Xenopus Aurora A kinase, as well as several novel sites in the Xenopus chromosome passenger complex (CPC).


Assuntos
Espectrometria de Massas/métodos , Mapeamento de Peptídeos/métodos , Fosfopeptídeos/análise , Fosfopeptídeos/química , Sequência de Aminoácidos , Animais , Aurora Quinases , Cromossomos/química , Cromossomos/metabolismo , Estrutura Molecular , Peso Molecular , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Xenopus laevis
20.
Curr Opin Cell Biol ; 19(1): 36-42, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17174541

RESUMO

While the metaphase spindle maintains a constant shape and size during cell division, its major component microtubules are continuously being polymerized, depolymerized and transported towards the two spindle poles in a process called microtubule poleward flux. This process has been observed in all metazoan cells. Recent studies have indicated that Kinesin-5s, which can drive the relative sliding of microtubules, and kinesin-13s, which regulate microtubule polymerization, are directly involved in microtubule poleward flux. The availability of molecular and chemical tools to perturb protein functions together with improvements in imaging and analytical methods have allowed the examination of these two kinesins' roles in poleward flux at high temporal and spatial resolution. These advances have shed some light on the molecular mechanisms that drive microtubule poleward flux.


Assuntos
Cinetocoros/fisiologia , Microtúbulos/fisiologia , Proteínas Motores Moleculares/fisiologia , Fuso Acromático/fisiologia , Animais , Humanos , Cinesinas/fisiologia , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...