Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 257
Filtrar
1.
J Cell Sci ; 136(7)2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-37039101

RESUMO

Finely tuned regulation of transport protein localization is vital for epithelial function. The Na+-HCO3- co-transporter NBCn1 (also known as SLC4A7) is a key contributor to epithelial pH homeostasis, yet the regulation of its subcellular localization is not understood. Here, we show that a predicted N-terminal ß-sheet and short C-terminal α-helical motif are essential for NBCn1 plasma membrane localization in epithelial cells. This localization was abolished by cell-cell contact disruption, and co-immunoprecipitation (co-IP) and proximity ligation (PLA) revealed NBCn1 interaction with E-cadherin and DLG1, linking it to adherens junctions and the Scribble complex. NBCn1 also interacted with RhoA and localized to lamellipodia and filopodia in migrating cells. Finally, analysis of native and GFP-tagged NBCn1 localization, subcellular fractionation, co-IP with Arl13B and CEP164, and PLA of NBCn1 and tubulin in mitotic spindles led to the surprising conclusion that NBCn1 additionally localizes to centrosomes and primary cilia in non-dividing, polarized epithelial cells, and to the spindle, centrosomes and midbodies during mitosis. We propose that NBCn1 traffics between lateral junctions, the leading edge and cell division machinery in Rab11 endosomes, adding new insight to the role of NBCn1 in cell cycle progression.


Assuntos
Membrana Celular , Centrossomo , Cílios , Simportadores de Sódio-Bicarbonato , Fuso Acromático , Humanos , Animais , Ratos , Membrana Celular/química , Cílios/química , Centrossomo/química , Fuso Acromático/química , Simportadores de Sódio-Bicarbonato/análise , Simportadores de Sódio-Bicarbonato/metabolismo , Ciclo Celular , AMP Cíclico/metabolismo , Polaridade Celular , Células Epiteliais/metabolismo
2.
J Cell Biol ; 220(7)2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-33983387

RESUMO

Centrosomes are composed of a centriolar core surrounded by pericentriolar material that nucleates microtubules. The ubiquitin ligase TRIM37 localizes to centrosomes, but its centrosomal roles are not yet defined. We show that TRIM37 does not control centriole duplication, structure, or the ability of centrioles to form cilia but instead prevents assembly of an ectopic centrobin-scaffolded structured condensate that forms by budding off of centrosomes. In ∼25% of TRIM37-deficient cells, the condensate organizes an ectopic spindle pole, recruiting other centrosomal proteins and acquiring microtubule nucleation capacity during mitotic entry. Ectopic spindle pole-associated transient multipolarity and multipolar segregation in TRIM37-deficient cells are suppressed by removing centrobin, which interacts with and is ubiquitinated by TRIM37. Thus, TRIM37 ensures accurate chromosome segregation by preventing the formation of centrobin-scaffolded condensates that organize ectopic spindle poles. Mutations in TRIM37 cause the disorder mulibrey nanism, and patient-derived cells harbor centrobin condensate-organized ectopic poles, leading us to propose that chromosome missegregation is a pathological mechanism in this disorder.


Assuntos
Microtúbulos/genética , Mitose/genética , Fuso Acromático/genética , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética , Proteínas de Ciclo Celular , Centríolos/genética , Centrossomo/química , Segregação de Cromossomos/genética , Humanos , Mutação/genética , Polos do Fuso/genética , Ubiquitina/genética
3.
Dev Cell ; 56(7): 1000-1013.e6, 2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33725482

RESUMO

Lysosome-related organelles (LROs) are endosomal compartments carrying tissue-specific proteins, which become enlarged in Chediak-Higashi syndrome (CHS) due to mutations in LYST. Here, we show that Drosophila Mauve, a counterpart of LYST, suppresses vesicle fusion events with lipid droplets (LDs) during the formation of yolk granules (YGs), the LROs of the syncytial embryo, and opposes Rab5, which promotes fusion. Mauve localizes on YGs and at spindle poles, and it co-immunoprecipitates with the LDs' component and microtubule-associated protein Minispindles/Ch-TOG. Minispindles levels are increased at the enlarged YGs and diminished around centrosomes in mauve-derived mutant embryos. This leads to decreased microtubule nucleation from centrosomes, a defect that can be rescued by dominant-negative Rab5. Together, this reveals an unanticipated link between endosomal vesicles and centrosomes. These findings establish Mauve/LYST's role in regulating LRO formation and centrosome behavior, a role that could account for the enlarged LROs and centrosome positioning defects at the immune synapse of CHS patients.


Assuntos
Centrossomo/metabolismo , Grânulos Citoplasmáticos/ultraestrutura , Proteínas de Drosophila/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Proteínas de Transporte Vesicular/fisiologia , Animais , Linhagem Celular , Centrossomo/química , Síndrome de Chediak-Higashi , Grânulos Citoplasmáticos/química , Drosophila/química , Drosophila/embriologia , Drosophila/metabolismo , Proteínas de Drosophila/análise , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Feminino , Humanos , Lisossomos , Proteínas Associadas aos Microtúbulos/genética , Mutação , Oócitos/química , Fuso Acromático/química , Proteínas de Transporte Vesicular/análise , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética
4.
Elife ; 92020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32851976

RESUMO

Asymmetric astral microtubule organization drives the polarized orientation of the S. cerevisiae mitotic spindle and primes the invariant inheritance of the old spindle pole body (SPB, the yeast centrosome) by the bud. This model has anticipated analogous centrosome asymmetries featured in self-renewing stem cell divisions. We previously implicated Spc72, the cytoplasmic receptor for the gamma-tubulin nucleation complex, as the most upstream determinant linking SPB age, functional asymmetry and fate. Here we used structured illumination microscopy and biochemical analysis to explore the asymmetric landscape of nucleation sites inherently built into the spindle pathway and under the control of cyclin-dependent kinase (CDK). We show that CDK enforces Spc72 asymmetric docking by phosphorylating Nud1/centriolin. Furthermore, CDK-imposed order in the construction of the new SPB promotes the correct balance of nucleation sites between the nuclear and cytoplasmic faces of the SPB. Together these contributions by CDK inherently link correct SPB morphogenesis, age and fate.


Assuntos
Centrossomo/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae , Ciclo Celular/genética , Ciclo Celular/fisiologia , Centrossomo/química , Quinases Ciclina-Dependentes/genética , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Fuso Acromático/genética , Fuso Acromático/metabolismo , Corpos Polares do Fuso/genética , Corpos Polares do Fuso/metabolismo
5.
Cells ; 9(6)2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32526902

RESUMO

Centrioles are microtubule-based cellular structures present in most human cells that build centrosomes and cilia. Proliferating cells have only two centrosomes and this number is stringently maintained through the temporally and spatially controlled processes of centriole assembly and segregation. The assembly of new centrioles begins in early S phase and ends in the third G1 phase from their initiation. This lengthy process of centriole assembly from their initiation to their maturation is characterized by numerous structural and still poorly understood biochemical changes, which occur in synchrony with the progression of cells through three consecutive cell cycles. As a result, proliferating cells contain three structurally, biochemically, and functionally distinct types of centrioles: procentrioles, daughter centrioles, and mother centrioles. This age difference is critical for proper centrosome and cilia function. Here we discuss the centriole assembly process as it occurs in somatic cycling human cells with a focus on the structural, biochemical, and functional characteristics of centrioles of different ages.


Assuntos
Centríolos/química , Centríolos/ultraestrutura , Centrossomo/química , Centrossomo/ultraestrutura , Envelhecimento , Humanos
6.
Structure ; 28(8): 910-921.e4, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32433990

RESUMO

FAM46C, a non-canonical poly(A) polymerase, is frequently mutated in multiple myeloma. Loss of function of FAM46C promotes cell survival of multiple myeloma, suggesting a tumor-suppressive role. FAM46C is also essential for fastening sperm head and flagellum, indispensable for male fertility. The molecular mechanisms of these functions of FAM46C remain elusive. We report the crystal structure of FAM46C to provide the basis for its poly(A) polymerase activity and rationalize mutations associated with multiple myeloma. In addition, we found that FAM46C interacts directly with the serine/threonine kinase Plk4, the master regulator of centrosome duplication. We present the structure of FAM46C in complex with the Cryptic Polo-Box 1-2 domains of Plk4. Our structure-based mutational analyses show that the interaction with Plk4 recruits FAM46C to centrosomes. Our data suggest that Plk4-mediated localization of FAM46C enables its regulation of centrosome structure and functions, which may underlie the roles for FAM46C in cell proliferation and sperm development.


Assuntos
Nucleotidiltransferases/química , Proteínas Serina-Treonina Quinases/química , Sítios de Ligação , Linhagem Celular Tumoral , Centrossomo/química , Centrossomo/metabolismo , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , Mutação , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
7.
Elife ; 82019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31872801

RESUMO

Deciphering how signaling enzymes operate within discrete microenvironments is fundamental to understanding biological processes. A-kinase anchoring proteins (AKAPs) restrict the range of action of protein kinases within intracellular compartments. We exploited the AKAP targeting concept to create genetically encoded platforms that restrain kinase inhibitor drugs at distinct subcellular locations. Local Kinase Inhibition (LoKI) allows us to ascribe organelle-specific functions to broad specificity kinases. Using chemical genetics, super resolution microscopy, and live-cell imaging we discover that centrosomal delivery of Polo-like kinase 1 (Plk1) and Aurora A (AurA) inhibitors attenuates kinase activity, produces spindle defects, and prolongs mitosis. Targeted inhibition of Plk1 in zebrafish embryos illustrates how centrosomal Plk1 underlies mitotic spindle assembly. Inhibition of kinetochore-associated pools of AurA blocks phosphorylation of microtubule-kinetochore components. This versatile precision pharmacology tool enhances investigation of local kinase biology.


Assuntos
Proteínas de Ancoragem à Quinase A/genética , Aurora Quinase A/genética , Proteínas de Ciclo Celular/genética , Mitose/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Animais , Aurora Quinase A/química , Proteínas de Ciclo Celular/química , Centrossomo/química , Centrossomo/ultraestrutura , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Cinetocoros/química , Microtúbulos/genética , Fosforilação/genética , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/química , Proteínas Proto-Oncogênicas/química , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Quinase 1 Polo-Like
8.
Sci Rep ; 9(1): 19126, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31836782

RESUMO

Gold nanoparticles of different sizes, shapes, and decorations exert a variety of effects on biological systems. We report a novel mechanism of action of chemically modified, tryptone-stabilized gold nanoparticles (T-GNPs) in the triple-negative breast cancer (TNBC) cell line, MDA-MB-231. The T-GNPs, synthesized using HAuCl4.3H2O and tryptone and characterized by an assortment of spectroscopy techniques combined with high-resolution electron microscopy, demonstrated strong antiproliferative and anti-clonogenic potential against MDA-MB-231 cells, arresting them at the G1 phase of the cell cycle and promoting apoptosis. The molecular mechanism of action of these particles involved induction of unipolar clustering and hyper amplification of the supernumerary centrosomes (a distinctive feature of many tumour cells, including TNBC cells). The clustering was facilitated by microtubules with suppressed dynamicity. Mass spectrometry-assisted proteomic analysis revealed that the T-GNP-induced G1 arrest was facilitated, at least in part, by downregulation of ribosome biogenesis pathways. Due to the presence of supernumerary centrosomes in many types of tumour cells, we propose chemical induction of their unipolar clustering as a potential therapeutic strategy.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Centrossomo/química , Ouro/química , Nanopartículas Metálicas/química , Peptonas/química , Neoplasias de Mama Triplo Negativas/genética , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Fase G1/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial , Microscopia Eletrônica , Proteômica , Espécies Reativas de Oxigênio , Espectrofotometria
9.
J Cell Biol ; 218(12): 3977-3985, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31636117

RESUMO

During mitosis, the centrosome expands its capacity to nucleate microtubules. Understanding the mechanisms of centrosomal microtubule nucleation is, however, constrained by a lack of knowledge of the amount of soluble and polymeric tubulin at mitotic centrosomes. Here we combined light microscopy and serial-section electron tomography to measure the amount of dimeric and polymeric tubulin at mitotic centrosomes in early C. elegans embryos. We show that a C. elegans one-cell stage centrosome at metaphase contains >10,000 microtubules with a total polymer concentration of 230 µM. Centrosomes concentrate soluble α/ß tubulin by about 10-fold over the cytoplasm, reaching peak values of 470 µM, giving a combined total monomer and polymer tubulin concentration at centrosomes of up to 660 µM. These findings support in vitro data suggesting that microtubule nucleation in C. elegans centrosomes is driven in part by concentrating soluble tubulin.


Assuntos
Caenorhabditis elegans/química , Centrossomo/química , Mitose , Tubulina (Proteína)/química , Animais , Centrossomo/ultraestrutura , Citoplasma/química , Dimerização , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Metáfase , Microscopia Eletrônica , Microtúbulos/química , Nocodazol/farmacologia , Polímeros/química , Interferência de RNA , Solubilidade
10.
Trends Cell Biol ; 29(8): 612-622, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31076235

RESUMO

There is currently intense interest in the idea that many membraneless organelles might assemble through phase separation of their constituent molecules into biomolecular 'condensates' that have liquid-like properties. This idea is intuitively appealing, especially for complex organelles such as centrosomes, where a liquid-like structure would allow the many constituent molecules to diffuse and interact with one another efficiently. I discuss here recent studies that either support the concept of a liquid-like centrosome or suggest that centrosomes are assembled upon a more solid, stable scaffold. I suggest that it may be difficult to distinguish between these possibilities. I argue that the concept of biomolecular condensates is an important advance in cell biology, with potentially wide-ranging implications, but it seems premature to conclude that centrosomes, and perhaps other membraneless organelles, are necessarily best described as liquid-like phase-separated condensates.


Assuntos
Centrossomo/metabolismo , Animais , Biologia Celular , Centrossomo/química , Humanos , Organelas/química , Organelas/metabolismo , Transição de Fase
11.
J Genet Genomics ; 45(8): 433-442, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30174135

RESUMO

In most kinds of animal cells, the centrosome serves as the main microtubule organizing center (MTOC) that nucleates microtubule arrays throughout the cytoplasm to maintain cell structure, cell division and intracellular transport. Whereas in epithelial cells, non-centrosomal MTOCs are established in the apical domain for generating asymmetric microtubule fibers and cilia in epithelial cells for the organ morphogenesis during embryonic development. However, the mechanism by which MTOCs localize to the apical domain in epithelial cells remains largely unknown. Here, we show that Mid1ip1b has a close interaction with γ-tubulin protein, the central component of MTOC, and modulates lumen opening of the neural tube, gut, intestine, and kidney of zebrafish. Knockdown or dominant negative effect of Mid1ip1b resulted in failure of lumen formation of the organs as aforementioned. Moreover, the non-centrosomal MTOCs were unable to orientate to the apical domain in Mid1ip1b knockdown epithelial cells, and the centrosomal MTOCs were inaccurately placed in the apical domain, resulting in defective formation of asymmetric microtubules and misplacement of cilia in the apical domain. These data uncover a molecule that controls the proper localization of MTOCs in the apical domain in epithelial cells for organ morphogenesis during embryonic development.


Assuntos
Centrossomo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Epiteliais/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Centrossomo/química , Proteínas do Citoesqueleto/genética , Mucosa Intestinal/metabolismo , Intestinos/embriologia , Rim/embriologia , Rim/metabolismo , Microtúbulos/genética , Microtúbulos/metabolismo , Transporte Proteico , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
12.
Biosystems ; 173: 191-206, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30142359

RESUMO

BACKGROUND: Myriads of signaling pathways in a single cell function to achieve the highest spatio-temporal integration. Data are accumulating on the role of electromechanical soliton-like waves in signal transduction processes. Theoretical studies strongly suggest feasibility of both classical and quantum computing involving microtubules. AIM: A theoretical study of the role of the complex composed of the plasma membrane and the microtubule-based cytoskeleton as a system that transmits, stores and processes information. METHODS: Theoretical analysis presented here refers to (i) the Penrose-Hameroff theory of consciousness (Orchestrated Objective Reduction; Orch OR), (ii) the description of the centrosome as a reference system for construction of the 3D map of the cell proposed by Regolini, (iii) the Heimburg-Jackson model of the nerve pulse propagation along axons' lipid bilayer as soliton-like electro-mechanical waves. RESULTS AND CONCLUSION: The ideas presented in this paper provide a qualitative model for the decision-making processes in a living cell undergoing a differentiation process. OUTLOOK: This paper paves the way for the real-time live-cell observation of information processing by microtubule-based cytoskeleton and cell fate decision making.


Assuntos
Membrana Celular/metabolismo , Centrossomo/química , Transdução de Sinais , Estresse Mecânico , Actinas/química , Animais , Apoptose , Linhagem da Célula , Citoesqueleto/metabolismo , Tomada de Decisões , Humanos , Íons , Bicamadas Lipídicas/química , Microtúbulos/metabolismo
13.
Open Biol ; 8(6)2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29899122

RESUMO

Centrosome aberrations disrupt tissue architecture and may confer invasive properties to cancer cells. Here we show that structural centrosome aberrations, induced by overexpression of either Ninein-like protein (NLP) or CEP131/AZI1, sensitize polarized mammalian epithelia to basal cell extrusion. While unperturbed epithelia typically dispose of damaged cells through apical dissemination into luminal cavities, certain oncogenic mutations cause a switch in directionality towards basal cell extrusion, raising the potential for metastatic cell dissemination. Here we report that NLP-induced centrosome aberrations trigger the preferential extrusion of damaged cells towards the basal surface of epithelial monolayers. This switch in directionality from apical to basal dissemination coincides with a profound reorganization of the microtubule cytoskeleton, which in turn prevents the contractile ring repositioning that is required to support extrusion towards the apical surface. While the basal extrusion of cells harbouring NLP-induced centrosome aberrations requires exogenously induced cell damage, structural centrosome aberrations induced by excess CEP131 trigger the spontaneous dissemination of dying cells towards the basal surface from MDCK cysts. Thus, similar to oncogenic mutations, structural centrosome aberrations can favour basal extrusion of damaged cells from polarized epithelia. Assuming that additional mutations may promote cell survival, this process could sensitize epithelia to disseminate potentially metastatic cells.


Assuntos
Centrossomo/química , Células Epiteliais/citologia , Proteínas dos Microtúbulos/genética , Animais , Técnicas de Cultura de Células , Linhagem Celular , Polaridade Celular , Centrossomo/metabolismo , Cães , Células Epiteliais/metabolismo , Humanos , Células Madin Darby de Rim Canino , Proteínas dos Microtúbulos/metabolismo , Mitose
14.
Elife ; 72018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29741480

RESUMO

The two centrioles of the centrosome in quiescent cells are inherently asymmetric structures that differ in age, morphology and function. How these asymmetric properties are established and maintained during quiescence remains unknown. Here, we show that a daughter centriole-associated ciliopathy protein, Cep120, plays a critical inhibitory role at daughter centrioles. Depletion of Cep120 in quiescent mouse and human cells causes accumulation of pericentriolar material (PCM) components including pericentrin, Cdk5Rap2, ninein and Cep170. The elevated PCM levels result in increased microtubule-nucleation activity at the centrosome. Consequently, loss of Cep120 leads to aberrant dynein-dependent trafficking of centrosomal proteins, dispersal of centriolar satellites, and defective ciliary assembly and signaling. Our results indicate that Cep120 helps to maintain centrosome homeostasis by inhibiting untimely maturation of the daughter centriole, and defines a potentially new molecular defect underlying the pathogenesis of ciliopathies such as Jeune Asphyxiating Thoracic Dystrophy and Joubert syndrome.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centríolos/metabolismo , Centrossomo/metabolismo , Homeostase , Animais , Linhagem Celular , Centrossomo/química , Ciliopatias/fisiopatologia , Humanos , Camundongos
15.
Trends Cell Biol ; 28(7): 574-588, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29571882

RESUMO

Microtubule organization has a crucial role in regulating cell architecture. The geometry of microtubule arrays strongly depends on the distribution of sites responsible for microtubule nucleation and minus-end attachment. In cycling animal cells, the centrosome often represents a dominant microtubule-organizing center (MTOC). However, even in cells with a radial microtubule system, many microtubules are not anchored at the centrosome, but are instead linked to the Golgi apparatus or other structures. Non-centrosomal microtubules predominate in many types of differentiated cell and in mitotic spindles. In this review, we discuss recent advances in understanding how the organization of centrosomal and non-centrosomal microtubule networks is controlled by proteins involved in microtubule nucleation and specific factors that recognize free microtubule minus ends and regulate their localization and dynamics.


Assuntos
Centrossomo/metabolismo , Complexo de Golgi/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Animais , Centrossomo/química , Complexo de Golgi/química , Humanos , Centro Organizador dos Microtúbulos/química , Microtúbulos/química
16.
Proc Natl Acad Sci U S A ; 115(10): E2246-E2253, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29463719

RESUMO

The centrosome linker proteins C-Nap1, rootletin, and CEP68 connect the two centrosomes of a cell during interphase into one microtubule-organizing center. This coupling is important for cell migration, cilia formation, and timing of mitotic spindle formation. Very little is known about the structure of the centrosome linker. Here, we used stimulated emission depletion (STED) microscopy to show that each C-Nap1 ring at the proximal end of the two centrioles organizes a rootletin ring and, in addition, multiple rootletin/CEP68 fibers. Rootletin/CEP68 fibers originating from the two centrosomes form a web-like, interdigitating network, explaining the flexible nature of the centrosome linker. The rootletin/CEP68 filaments are repetitive and highly ordered. Staggered rootletin molecules (N-to-N and C-to-C) within the filaments are 75 nm apart. Rootletin binds CEP68 via its C-terminal spectrin repeat-containing region in 75-nm intervals. The N-to-C distance of two rootletin molecules is ∼35 to 40 nm, leading to an estimated minimal rootletin length of ∼110 nm. CEP68 is important in forming rootletin filaments that branch off centrioles and to modulate the thickness of rootletin fibers. Thus, the centrosome linker consists of a vast network of repeating rootletin units with C-Nap1 as ring organizer and CEP68 as filament modulator.


Assuntos
Centríolos/metabolismo , Centrossomo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas/metabolismo , Motivos de Aminoácidos , Centríolos/química , Centríolos/genética , Centrossomo/química , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Células HeLa , Humanos , Interfase , Microscopia , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/genética , Ligação Proteica , Proteínas/química , Proteínas/genética , tRNA Metiltransferases
17.
Biochemistry ; 57(1): 30-37, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29256606

RESUMO

Centrosomes are self-assembling, micron-scale, nonmembrane bound organelles that nucleate microtubules (MTs) and organize the microtubule cytoskeleton of the cell. They orchestrate critical cellular processes such as ciliary-based motility, vesicle trafficking, and cell division. Much is known about the role of the centrosome in these contexts, but we have a less comprehensive understanding of how the centrosome assembles and generates microtubules. Studies over the past 10 years have fundamentally shifted our view of these processes. Subdiffraction imaging has probed the amorphous haze of material surrounding the core of the centrosome revealing a complex, hierarchically organized structure whose composition and size changes profoundly during the transition from interphase to mitosis. New biophysical insights into protein phase transitions, where a diffuse protein spontaneously separates into a locally concentrated, nonmembrane bounded compartment, have provided a fresh perspective into how the centrosome might rapidly condense from diffuse cytoplasmic components. In this Perspective, we focus on recent findings that identify several centrosomal proteins that undergo phase transitions. We discuss how to reconcile these results with the current model of the underlying organization of proteins in the centrosome. Furthermore, we reflect on how these findings impact our understanding of how the centrosome undergoes self-assembly and promotes MT nucleation.


Assuntos
Bioquímica/métodos , Centrossomo/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Modelos Biológicos , Animais , Bioquímica/tendências , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Centrossomo/química , Humanos , Interfase , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/metabolismo , Centro Organizador dos Microtúbulos/química , Microtúbulos/química , Mitose , Transição de Fase , Multimerização Proteica , Transporte Proteico
18.
Mol Cell Biol ; 38(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29061732

RESUMO

The migration of chromosomes during mitosis is mediated primarily by kinesins that bind to the chromosomes and move along the microtubules, exerting pulling and pushing forces on the centrosomes. We report that a DNA replication protein, Sld5, localizes to the centrosomes, resisting the microtubular pulling forces experienced during chromosome congression. In the absence of Sld5, centriolar satellites, which normally cluster around the centrosomes, are dissipated throughout the cytoplasm, resulting in the loss of their known function of recruiting the centrosomal protein, pericentrin. We observed that Sld5-deficient centrosomes lacking pericentrin were unable to endure the CENP-E- and Kid-mediated microtubular forces that converge on the centrosomes during chromosome congression, resulting in monocentriolar and acentriolar spindle poles. The minus-end-directed kinesin-14 motor protein, HSET, sustains the traction forces that mediate centrosomal fragmentation in Sld5-depleted cells. Thus, we report that a DNA replication protein has an as yet unknown function of ensuring spindle pole resistance to traction forces exerted during chromosome congression.


Assuntos
Centríolos/metabolismo , Centrossomo/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centríolos/genética , Centrossomo/química , Proteínas Cromossômicas não Histona/genética , Cromossomos Humanos/metabolismo , Dano ao DNA , Células HeLa , Humanos , Interfase/fisiologia , Microtúbulos/química , Microtúbulos/fisiologia , Mitose , Polos do Fuso/fisiologia , Polos do Fuso/ultraestrutura , Imagem com Lapso de Tempo
19.
J Biol Chem ; 292(50): 20410-20411, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29247130

RESUMO

The scaffolding protein AKAP350A is known to localize to the centrosome and the Golgi, but the molecular details of its function at the centrosome remain elusive. Using structure-function analyses, protein interaction assays, and super-resolution microscopy, Kolobova et al. now identify AKAP350A's specific location and protein partners at the centrosome. The authors further define an autoregulatory mechanism that likely controls AKAP350A's ability to nucleate microtubule growth.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Centrossomo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Modelos Moleculares , Proteínas de Ancoragem à Quinase A/química , Animais , Proteínas de Ciclo Celular , Centrossomo/química , Proteínas do Citoesqueleto/química , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/metabolismo , Centro Organizador dos Microtúbulos/química , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Transporte Proteico
20.
Biochem Biophys Res Commun ; 491(4): 980-985, 2017 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-28765046

RESUMO

S100A6 is a Ca2+-signal transducer that interacts with numerous proteins and regulates their biochemical functions. Here we identified a centrosomal protein, FOR20 (FOP-related protein of 20 kDa) as a novel S100A6 target by screening protein microarrays carrying 19,676 recombinant GST-fused human proteins. Binding experiments revealed that S100A6 interacts with the N-terminal region (residues 1-30) of FOR20 in a Ca2+-dependent manner in vitro and in living cells. Several S100 proteins including S100A1, A2, A4, A11, B also exhibited Ca2+-dependent interactions with FOR20 as well as S100A6. We found that two distantly related centrosomal proteins, FOP and OFD1, also possess N-terminal regions with a significant sequence similarity to the putative S100A6-binding site (residues 1-30) in FOR20 and are capable of binding to S100A6 in a Ca2+-dependent manner. Taken together, these results may indicate that S100A6 interacts with FOR20 and related centrosomal proteins through a conserved N-terminal domain, suggesting a novel Ca2+-dependent regulation of centrosomal function.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas/química , Proteínas/metabolismo , Proteínas S100/química , Proteínas S100/metabolismo , Animais , Células COS , Células Cultivadas , Centrossomo/química , Centrossomo/metabolismo , Chlorocebus aethiops , Células HeLa , Humanos , Análise Serial de Proteínas , Ligação Proteica , Proteína A6 Ligante de Cálcio S100 , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA