Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 473, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35078983

ABSTRACT

The gamma-tubulin ring complex (γ-TuRC) is the principal microtubule nucleation template in vertebrates. Recent cryo-EM reconstructions visualized the intricate quaternary structure of the γ-TuRC, containing more than thirty subunits, raising fundamental questions about γ-TuRC assembly and the role of actin as an integral part of the complex. Here, we reveal the structural mechanism underlying modular γ-TuRC assembly and identify a functional role of actin in microtubule nucleation. During γ-TuRC assembly, a GCP6-stabilized core comprising GCP2-3-4-5-4-6 is expanded by stepwise recruitment, selective stabilization and conformational locking of four pre-formed GCP2-GCP3 units. Formation of the lumenal bridge specifies incorporation of the terminal GCP2-GCP3 unit and thereby leads to closure of the γ-TuRC ring in a left-handed spiral configuration. Actin incorporation into the complex is not relevant for γ-TuRC assembly and structural integrity, but determines γ-TuRC geometry and is required for efficient microtubule nucleation and mitotic chromosome alignment in vivo.


Subject(s)
Actins/chemistry , Cryoelectron Microscopy/methods , Microtubule-Associated Proteins/chemistry , Microtubule-Organizing Center/chemistry , Microtubules/chemistry , Tubulin/chemistry , Actins/metabolism , Cell Line , Humans , Microtubule-Associated Proteins/isolation & purification , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Tubulin/metabolism
2.
Curr Opin Cell Biol ; 68: 124-131, 2021 02.
Article in English | MEDLINE | ID: mdl-33190097

ABSTRACT

Microtubules are essential cytoskeletal elements assembled from αß-tubulin dimers. In high eukaryotes, microtubule nucleation, the de novo assembly of a microtubule from its minus end, is initiated by the γ-tubulin ring complex (γ-TuRC). Despite many years of research, the structural and mechanistic principles of the microtubule nucleation machinery remained poorly understood. Only recently, cryoelectron microscopy studies uncovered the molecular organization and potential activation mechanisms of γ-TuRC. In vitro assays further deciphered the spatial and temporal cooperation between γ-TuRC and additional factors, for example, the augmin complex, the phase separation protein TPX2, and the microtubule polymerase XMAP215. These breakthroughs deepen our understanding of microtubule nucleation mechanisms and will link the assembly of individual microtubules to the organization of cellular microtubule networks.


Subject(s)
Microtubule-Organizing Center/chemistry , Microtubules/chemistry , Tubulin/chemistry , Animals , Cryoelectron Microscopy , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/ultrastructure , Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/ultrastructure , Microtubules/metabolism , Microtubules/ultrastructure , Polymerization , Tubulin/metabolism , Tubulin/ultrastructure
3.
Virchows Arch ; 478(2): 327-334, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32710188

ABSTRACT

Urothelial carcinoma (UC) comprises two subtypes, low grade (LG-UC) and high grade (HG-UC), with different pathological and clinical behavior. LG-UC and HG-UC are classified based on cellular and structural atypia of pathological findings. The mechanisms responsible for maintaining structural atypia, such as the disturbance of nuclear polarity, remain unclear. In this study, we studied microtubule-organizing center (MTOC)-mediated nuclear polarity in UC subtypes. We evaluated six cases with normal urothelium (NU), 10 LG-UC cases, and 10 HG-UC cases by double immunofluorescence staining of γ-tubulin as a marker of MTOC and E-cadherin as a marker of each cell border. The number and position of γ-tubulin dots of expression in more than 100 cells per case were assessed using the spatial relationship with the nucleus and surface-basal axis. We found one γ-tubulin dot in most normal and tumor cells, and more than two γ-tubulin dots in 4.6% of NU cells, 6.1% of LG-UC cells, and 9.8% of HG-UC cells. More than three γ-tubulin dots were found only in 1.2% of HG-UC cells. Surface side positioning of γ-tubulin was found in 77.4% of normal urothelial cells, 63.8% of LG-UC cells, and 39.2% of HG-UC cells, whereas aberrant lateral and basal side positioning of γ-tubulin was found in 22.6% of normal urothelial cells, 36.1% of LG-UC cells, and 60.8% of HG-UC cells. We concluded that numerical and positional aberrations of MTOC in UC cases were strongly correlated with both cellular and structural atypia as well as abnormal cell proliferation.


Subject(s)
Carcinoma/pathology , Cell Nucleus/pathology , Microtubule-Organizing Center/pathology , Urinary Bladder Neoplasms/pathology , Urothelium/pathology , Adult , Aged , Aged, 80 and over , Antigens, CD/analysis , Biomarkers, Tumor/analysis , Cadherins/analysis , Carcinoma/chemistry , Carcinoma/surgery , Cell Nucleus/chemistry , Cell Proliferation , Female , Humans , Male , Microtubule-Organizing Center/chemistry , Middle Aged , Neoplasm Grading , Tubulin/analysis , Urinary Bladder Neoplasms/chemistry , Urinary Bladder Neoplasms/surgery , Urothelium/chemistry , Urothelium/surgery
4.
J Cell Biol ; 217(7): 2417-2428, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29875259

ABSTRACT

Microtubules (MTs) must be generated from precise locations to form the structural frameworks required for cell shape and function. MTs are nucleated by the γ-tubulin ring complex (γ-TuRC), but it remains unclear how γ-TuRC gets to the right location. Augmin has been suggested to be a γ-TuRC targeting factor and is required for MT nucleation from preexisting MTs. To determine augmin's architecture and function, we purified Xenopus laevis augmin from insect cells. We demonstrate that augmin is sufficient to target γ-TuRC to MTs by in vitro reconstitution. Augmin is composed of two functional parts. One module (tetramer-II) is necessary for MT binding, whereas the other (tetramer-III) interacts with γ-TuRC. Negative-stain electron microscopy reveals that both tetramers fit into the Y-shape of augmin, and MT branching assays reveal that both are necessary for MT nucleation. The finding that augmin can directly bridge MTs with γ-TuRC via these two tetramers adds to our mechanistic understanding of how MTs can be nucleated from preexisting MTs.


Subject(s)
Cell Cycle Proteins/chemistry , Microtubule-Associated Proteins/chemistry , Spindle Apparatus/genetics , Tubulin/chemistry , Animals , Cell Cycle Proteins/ultrastructure , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/ultrastructure , Microtubule-Organizing Center/chemistry , Microtubules/chemistry , Microtubules/genetics , Protein Binding , Protein Conformation , Spindle Apparatus/chemistry , Tubulin/genetics , Xenopus laevis/genetics
5.
Trends Cell Biol ; 28(7): 574-588, 2018 07.
Article in English | MEDLINE | ID: mdl-29571882

ABSTRACT

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.


Subject(s)
Centrosome/metabolism , Golgi Apparatus/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Animals , Centrosome/chemistry , Golgi Apparatus/chemistry , Humans , Microtubule-Organizing Center/chemistry , Microtubules/chemistry
6.
Biochemistry ; 57(1): 30-37, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29256606

ABSTRACT

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.


Subject(s)
Biochemistry/methods , Centrosome/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Models, Biological , Animals , Biochemistry/trends , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Centrosome/chemistry , Humans , Interphase , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/chemistry , Microtubules/chemistry , Mitosis , Phase Transition , Protein Multimerization , Protein Transport
7.
J Biol Chem ; 292(50): 20410-20411, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29247130

ABSTRACT

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.


Subject(s)
A Kinase Anchor Proteins/metabolism , Centrosome/metabolism , Cytoskeletal Proteins/metabolism , Microtubule-Organizing Center/metabolism , Models, Molecular , A Kinase Anchor Proteins/chemistry , Animals , Cell Cycle Proteins , Centrosome/chemistry , Cytoskeletal Proteins/chemistry , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport
8.
J Biol Chem ; 292(39): 16267-16276, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28827311

ABSTRACT

The centrosome serves as a major microtubule-organizing center (MTOC). The Cdc6 protein is a component of the pre-replicative complex and a licensing factor for the initiation of chromosome replication and localizes to centrosomes during the S and G2 phases of the cfell cycle of human cells. This cell cycle-dependent localization of Cdc6 to the centrosome motivated us to investigate whether Cdc6 negatively regulates MTOC activity and to determine the integral proteins that comprise the pericentriolar material (PCM). Time-lapse live-cell imaging of microtubule regrowth revealed that Cdc6 depletion increased microtubule nucleation at the centrosomes and that expression of Cdc6 in Cdc6-depleted cells reversed this effect. This increase and decrease in microtubule nucleation correlated with the centrosomal intensities of PCM proteins such as γ-tubulin, pericentrin, CDK5 regulatory subunit-associated protein 2 (CDK5RAP2), and centrosomal protein 192 (Cep192). The regulation of microtubule nucleation and the recruitment of PCM proteins to the centrosome required Cdc6 ATPase activity, as well as a centrosomal localization of Cdc6. These results suggest a novel function for Cdc6 in coordinating centrosome assembly and function.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/metabolism , Microtubule-Organizing Center/metabolism , Nuclear Proteins/metabolism , Amino Acid Substitution , Antigens/metabolism , Biomarkers/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line, Tumor , Centrosome/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Gene Deletion , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Microscopy, Fluorescence , Microtubule-Organizing Center/chemistry , Mutagenesis, Site-Directed , Mutation , Nerve Tissue Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Transport , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Time-Lapse Imaging , Tubulin/metabolism
9.
Curr Opin Cell Biol ; 44: 93-101, 2017 02.
Article in English | MEDLINE | ID: mdl-27666167

ABSTRACT

The process of cellular differentiation requires the distinct spatial organization of the microtubule cytoskeleton, the arrangement of which is specific to cell type. Microtubule patterning does not occur randomly, but is imparted by distinct subcellular sites called microtubule-organizing centers (MTOCs). Since the discovery of MTOCs fifty years ago, their study has largely focused on the centrosome. All animal cells use centrosomes as MTOCs during mitosis. However in many differentiated cells, MTOC function is reassigned to non-centrosomal sites to generate non-radial microtubule organization better suited for new cell functions, such as mechanical support or intracellular transport. Here, we review the current understanding of non-centrosomal MTOCs (ncMTOCs) and the mechanisms by which they form in differentiating animal cells.


Subject(s)
Microtubule-Organizing Center/chemistry , Microtubule-Organizing Center/metabolism , Animals , Cell Differentiation , Centrosome/metabolism , Cytoskeleton , Humans , Microtubules/metabolism , Mitosis
10.
Mycologia ; 108(3): 528-32, 2016.
Article in English | MEDLINE | ID: mdl-26951366

ABSTRACT

To spatially resolve genetic differences at the cellular level, the laser-capture microdissection technique was developed. With this method cells can be cut from tissues with a laser beam and analyzed for DNA, RNA or protein composition. Here we adapted the technique to isolate septal microtubule-organizing center (MTOC)-associated proteins in Aspergillus nidulans About 3000 septa were collected and subjected to peptide fingerprinting by mass-spectrometric analysis. We identified the microtubule polymerase AlpA and found it interacts with ApsB specifically at sMTOCs, suggesting that AlpA might be involved in the assembly or the functioning of this protein complex.


Subject(s)
Aspergillus nidulans/chemistry , Laser Capture Microdissection/methods , Aspergillus nidulans/genetics , Aspergillus nidulans/growth & development , Aspergillus nidulans/metabolism , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Hyphae/chemistry , Hyphae/genetics , Hyphae/growth & development , Hyphae/metabolism , Microtubule-Organizing Center/chemistry , Microtubule-Organizing Center/metabolism , Protein Binding
11.
J Reprod Dev ; 61(6): 541-8, 2015.
Article in English | MEDLINE | ID: mdl-26346254

ABSTRACT

Polymerase (RNA) II (DNA directed) polypeptide A (RPB1) is the largest subunit of RNA polymerase II (RNAPII), and phosphorylation of its C-terminal domain (CTD) is required for transcription initiation, elongation and RNA processing. Little is known about the CTD phosphorylation pattern and potential function during cell division when transcription is silenced. In this study, we assessed the protein expression and subcellular distribution of RPB1 during mouse oocyte meiotic division. Western blot analysis revealed that the RPB1 CTD was highly phosphorylated on Ser2 (pRPB1(Ser2)), Ser5 (pRPB1(Ser5)) and Ser7 (pRPB1(Ser7)). High and stable expression of pRPB1(Ser2) and pRPB1(Ser5) was detected from germinal vesicle (GV) to Metaphase II (MII) stage. In contrast, pRPB1(Ser7) only emerged after germinal vesicle breakdown (GVBD) and gradually increased to its peak level at metaphase I (MI) and MII. Immunofluorescence demonstrated that pRPB1(Ser2), pRPB1(Ser5) and pRPB1(Ser7) were pronouncedly aggregated within the nucleus of GV oocytes with a non-surrounded nucleolus (NSN) but very faintly labeled in oocytes with a surrounded nucleolus (SN). After meiotic resumption, pRPB1(Ser2) was again detected at spindle poles and co-localized with key microtubule organizing center (MTOC) components, pericentrin and γ-tubulin. pRPB1(Ser5) and pRPB1(Ser7) were assembled as filamentous aggregates and co-localized with microtubules throughout the spindle structure, responding to spindle-disturbing drugs, nocodazole or taxol, in pattern strongly similar to microtubules. pRPB1(Ser2) and pRPB1(Ser5) were constantly localized on chromosomes, with a relatively high concentration in centromere areas. Taken together, our data suggest that the CTD is highly phosphorylated and may be required for accurate chromosome segregation in mouse oocytes during meiosis.


Subject(s)
Meiosis/physiology , Oocytes/chemistry , RNA Polymerase II/analysis , Animals , Blotting, Western , Centromere/chemistry , Cytoplasm/chemistry , Female , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred Strains , Microtubule-Organizing Center/chemistry , Phosphorylation
12.
Cell Mol Life Sci ; 71(11): 2119-34, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24413667

ABSTRACT

During meiosis, telomeres cluster and promote homologous chromosome pairing. Telomere clustering depends on conserved SUN and KASH domain nuclear membrane proteins, which form a complex called the linker of nucleoskeleton and cytoskeleton (LINC) and connect telomeres with the cytoskeleton. It has been thought that LINC-mediated cytoskeletal forces induce telomere clustering. However, how cytoskeletal forces induce telomere clustering is not fully understood. Recent study of fission yeast has shown that the LINC complex forms the microtubule-organizing center (MTOC) at the telomere, which has been designated as the "telocentrosome", and that microtubule motors gather telomeres via telocentrosome-nucleated microtubules. This MTOC-dependent telomere clustering might be conserved in other eukaryotes. Furthermore, the MTOC-dependent clustering mechanism appears to function in various other biological events. This review presents an overview of the current understanding of the mechanism of meiotic telomere clustering and discusses the universality of the MTOC-dependent clustering mechanism.


Subject(s)
Meiosis , Microtubule-Organizing Center/physiology , Microtubules/genetics , Schizosaccharomyces/genetics , Gene Expression Regulation , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Organizing Center/chemistry , Microtubules/chemistry , Microtubules/metabolism , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Nuclear Envelope/chemistry , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Matrix/chemistry , Nuclear Matrix/genetics , Nuclear Matrix/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Structure, Tertiary , Schizosaccharomyces/chemistry , Schizosaccharomyces/metabolism , Telomere/chemistry , Telomere/genetics , Telomere/metabolism
13.
Cell Mol Life Sci ; 69(21): 3651-64, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22744750

ABSTRACT

Dictyostelium centrosomes consist of a layered core structure surrounded by a microtubule-nucleating corona. At the G2/M transition, the corona dissociates and the core structure duplicates, yielding two spindle pole bodies. Finally, in telophase, the spindle poles mature into two new, complete centrosomes. CP55 was identified in a centrosomal proteome analysis. It is a component of the centrosomal core structure, and persists at the centrosome throughout the entire cell cycle. FRAP experiments revealed that during interphase the majority of centrosomal GFP-CP55 is immobile, which indicates a structural task of CP55 at the centrosome. The CP55null mutant is characterized by increased ploidy, a less structured, slightly enlarged corona, and by supernumerary, cytosolic MTOCs, containing only corona proteins and lacking a core structure. Live cell imaging showed that supernumerary MTOCs arise in telophase. Lack of CP55 also caused premature recruitment of the corona organizer CP148 to mitotic spindle poles, already in metaphase instead of telophase. Forces transmitted through astral microtubules may expel prematurely acquired or loosely attached corona fragments into the cytosol, where they act as independent MTOCs. CP55null cells were also impaired in growth, most probably due to difficulties in centrosome splitting during prophase. Furthermore, although they were still capable of phagocytosis, they appeared unable to utilize phagocytosed nutrients. This inability may be attributed to their partially disorganized Golgi apparatus.


Subject(s)
Centrosome/metabolism , Dictyostelium/metabolism , Protozoan Proteins/metabolism , Cell Division , Dictyostelium/cytology , Dictyostelium/genetics , Gene Knockout Techniques , Golgi Apparatus/metabolism , Interphase , Microtubule-Organizing Center/chemistry , Microtubule-Organizing Center/metabolism , Mitosis , Phagocytosis , Ploidies , Protozoan Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spindle Apparatus/metabolism
14.
J Immunol ; 187(11): 5824-33, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22043013

ABSTRACT

The cytoskeletal adaptor protein paxillin localizes to the microtubule organizing center (MTOC) in T cells and, upon target cell binding, is recruited to the supramolecular activation complex (SMAC). We mapped the region of paxillin that associates with both the MTOC and SMAC to the leucine-aspartic acid (LD) domains and showed that a protein segment containing LD2-4 was sufficient for MTOC and SMAC recruitment. Examination of the localization of paxillin at the SMAC revealed that paxillin localizes to the peripheral area of the SMAC along with LFA-1, suggesting that LFA-1 may contribute to its recruitment. LFA-1 or CD3 engagement alone was insufficient for paxillin recruitment because there was no paxillin accumulation at the site of CTL contact with anti-LFA-1- or anti-CD3-coated beads. In contrast, paxillin accumulation was detected when beads coated with both anti-CD3 and anti-LFA-1 were bound to CTL, suggesting that signals from both the TCR and LFA-1 are required for paxillin accumulation. Paxillin was shown to be phosphorylated downstream of ERK, but when we generated a mutation (S83A/S130A) that abolished the mobility shift as a result of phosphorylation, we found that paxillin still bound to the MTOC and was recruited to the SMAC. Furthermore, ERK was not absolutely required for MTOC reorientation in CTL that require ERK for killing. Finally, expression of the LD2-4 region of paxillin substantially reduced MTOC reorientation. These studies demonstrated that paxillin is recruited, through its LD domains, to sites of integrin engagement and may contribute to MTOC reorientation required for directional degranulation.


Subject(s)
Lymphocyte Activation/physiology , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Paxillin/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Animals , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Blotting, Western , Cell Degranulation/physiology , Immunological Synapses/chemistry , Immunological Synapses/immunology , Immunological Synapses/metabolism , Immunoprecipitation , Leucine/chemistry , Leucine/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microtubule-Organizing Center/chemistry , Microtubule-Organizing Center/ultrastructure , Mutagenesis, Site-Directed , Paxillin/chemistry , Paxillin/immunology , Polymerase Chain Reaction , Protein Structure, Tertiary , Protein Transport/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
15.
Nat Commun ; 2: 430, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21847104

ABSTRACT

It is well known that the parallel order of microtubules in the plant cell cortex defines the direction of cell expansion, yet it remains unclear how microtubule orientation is controlled, especially on a cell-wide basis. Here we show through 4D imaging and computational modelling that plant cell polyhedral geometry provides spatial input that determines array orientation and heterogeneity. Microtubules depolymerize when encountering sharp cell edges head-on, whereas those oriented parallel to those sharp edges remain. Edge-induced microtubule depolymerization, however, is overcome by the microtubule-associated protein CLASP, which accumulates at specific cell edges, enables microtubule growth around sharp edges and promotes formation of microtubule bundles that span adjacent cell faces. By computationally modelling dynamic 'microtubules on a cube' with edges differentially permissive to microtubule passage, we show that the CLASP-edge complex is a 'tuneable' microtubule organizer, with the inherent flexibility to generate the numerous cortical array patterns observed in nature.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Cell Polarity , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Arabidopsis/chemistry , Arabidopsis/cytology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubule-Organizing Center/chemistry , Microtubules/chemistry , Microtubules/genetics , Microtubules/metabolism , Molecular Structure , Protein Structure, Tertiary , Protein Transport
16.
J Cell Sci ; 124(Pt 8): 1207-13, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21444751

ABSTRACT

Although the fission yeast Schizosaccharomyces pombe contains many of the γ-tubulin ring complex (γ-TuRC)-specific proteins of the γ-tubulin complex (γ-TuC), several questions about the organizational state and function of the fission yeast γ-TuC in vivo remain unresolved. Using 3×GFP-tagged γ-TuRC-specific proteins, we show here that γ-TuRC-specific proteins are present at all microtubule organizing centers in fission yeast and that association of γ-TuRC-specific proteins with the γ-tubulin small complex (γ-TuSC) does not depend on Mto1, which is a key regulator of the γ-TuC. Through sensitive imaging in mto1Δ mutants, in which cytoplasmic microtubule nucleation is abolished, we unexpectedly found that γ-TuC incapable of nucleating microtubules can nevertheless associate with microtubule minus-ends in vivo. The presence of γ-TuC at microtubule ends is independent of γ-TuRC-specific proteins and strongly correlates with the stability of microtubule ends. Strikingly, microtubule bundles lacking γ-TuC at microtubule ends undergo extensive treadmilling in vivo, apparently induced by geometrical constraints on plus-end growth. Our results indicate that microtubule stabilization by the γ-TuC, independently of its nucleation function, is important for maintaining the organization and dynamic behavior of microtubule arrays in vivo.


Subject(s)
Microtubules/chemistry , Microtubules/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Tubulin/metabolism , Microtubule-Organizing Center/chemistry , Microtubule-Organizing Center/metabolism , Microtubules/genetics , Protein Binding , Protein Stability , Schizosaccharomyces/chemistry , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/genetics , Tubulin/genetics
17.
Dev Biol ; 320(2): 414-25, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18602096

ABSTRACT

Defects in meiotic spindle structure can lead to chromosome segregation errors and genomic instability. In this study the potential role of protein kinase C delta (PKCdelta) on meiotic spindle organization was evaluated in mouse oocytes. PKCdelta was previously shown to be phosphorylated during meiotic maturation and concentrate on the meiotic spindle during metaphases I and II. Currently we show that when phosphorylated on Threonine 505 (pPKCdelta(Thr505)), within the activation loop of its C4 domain, PKCdelta expression was restricted to the meiotic spindle poles and a few specific cytoplasmic foci. In addition, pPKCdelta(Thr505) co-localized with two key microtubule organizing center (MTOC)-associated proteins, pericentrin and gamma-tubulin. An interaction between pPKCdelta(Thr505) and pericentrin as well as gamma-tubulin was confirmed by co-immunoprecipitation analysis using both fetal fibroblast cells and oocytes. Notably, targeted knockdown of PKCdelta expression in oocytes using short interfering RNAs effectively reduced pPKCdelta(Thr505) protein expression at MTOCs and leads to a significant (P < 0.05) disruption of meiotic spindle organization and chromosome alignment during MI and MII. Moreover, both gamma-tubulin and pericentrin expression at MTOCs were decreased in pPKCdelta(Thr505)-depleted oocytes. In sum, these results indicate that pPKCdelta(Thr505) interacts with MTOC-associated proteins and plays a role in meiotic spindle organization in mammalian oocytes.


Subject(s)
Antigens/metabolism , Microtubule-Organizing Center/chemistry , Protein Kinase C-delta/physiology , Spindle Apparatus/metabolism , Tubulin/metabolism , Animals , Chromosome Segregation , Embryo, Mammalian , Female , Metaphase , Mice , Microtubule-Associated Proteins/metabolism , Oocytes , Phosphorylation , Protein Kinase C-delta/metabolism
18.
Mol Reprod Dev ; 74(1): 76-87, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16929525

ABSTRACT

Although it has been known for over a century that sea urchin eggs are polarized cells, very little is known about the mechanism responsible for establishing and maintaining polarity. Our previous studies of microtubule organization during sea urchin oogenesis described a cortical microtubule-organizing center (MTOC) present during germinal vesicle (GV) migration in large oocytes. This MTOC was localized within the future animal pole of the mature egg. In this study we have used electron microscopy and immunocytochemistry to characterize the structure of this MTOC and have established that this organelle appears prior to GV migration. We show that the cortical MTOC contains all the components of a centrosome, including a pair of centrioles. Although a centrosome proper was not found in small oocytes, the centriole pair in these cells was always found in association with a striated rootlet, a structural remnant of the flagellar apparatus present in precursor germinal cells (PGCs). The centrioles/striated rootlet complex was asymmetrically localized to the side of the oocyte closest to the gonadal wall. These data are consistent with the previously proposed hypothesis that in echinoderms the polarity of the PGCs in the germinal epithelium influences the final polarity of the mature egg.


Subject(s)
Cell Polarity , Microtubule-Organizing Center/metabolism , Oocytes/growth & development , Oogenesis , Strongylocentrotus/growth & development , Animals , Antigens/analysis , Centrioles/ultrastructure , Centrosome/ultrastructure , Female , Microtubule-Organizing Center/chemistry , Microtubule-Organizing Center/ultrastructure , Oocytes/chemistry , Oocytes/ultrastructure , Strongylocentrotus/ultrastructure , Tubulin/analysis
19.
Development ; 133(20): 3963-72, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16971473

ABSTRACT

The gamma-tubulin ring complex (gammaTuRC) forms an essential template for microtubule nucleation in animal cells. The molecular composition of the gammaTuRC has been described; however, the functions of the subunits proposed to form the cap structure remain to be characterized in vivo. In Drosophila, the core components of the gammaTuRC are essential for mitosis, whereas the cap component Grip75 is not required for viability but functions in bicoid RNA localization during oogenesis. The other cap components have not been analyzed in vivo. We report the functional characterization of the cap components Grip128 and Grip75. Animals with mutations in Dgrip128 or Dgrip75 are viable, but both males and females are sterile. Both proteins are required for the formation of distinct sets of microtubules, which facilitate bicoid RNA localization during oogenesis, the formation of the central microtubule aster connecting the meiosis II spindles in oocytes and cytokinesis in male meiosis. Grip75 and Grip128 anchor the axoneme at the nucleus during sperm elongation. We propose that Grip75 and Grip128 are required to tether microtubules at specific microtubule-organizing centers, instead of being required for general microtubule nucleation. The gammaTuRC cap structure may be essential only for non-centrosome-based microtubule functions.


Subject(s)
Drosophila Proteins/physiology , Drosophila/growth & development , Meiosis , Microtubule-Associated Proteins/physiology , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/analysis , Drosophila Proteins/genetics , Female , Homeodomain Proteins/genetics , Male , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/genetics , Microtubule-Organizing Center/chemistry , Microtubules/chemistry , Mutation , Oocytes/chemistry , Oocytes/cytology , Oocytes/metabolism , Oogenesis/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Spermatogenesis/genetics , Spermatozoa/chemistry , Spermatozoa/cytology , Spermatozoa/metabolism , Trans-Activators/genetics , Tubulin/analysis , Tubulin/genetics , Tubulin/physiology
20.
Mol Biol Cell ; 17(5): 2212-22, 2006 May.
Article in English | MEDLINE | ID: mdl-16481403

ABSTRACT

Microtubule-organizing centers (MTOCs) concentrate microtubule nucleation, attachment and bundling factors and thus restrict formation of microtubule arrays in spatial and temporal manner. How MTOCs occur remains an exciting question in cell biology. Here, we show that the transforming acidic coiled coil-related protein Mia1p/Alp7p functions in emergence of large MTOCs in interphase fission yeast cells. We found that Mia1p was a microtubule-binding protein that preferentially localized to the minus ends of microtubules and was associated with the sites of microtubule attachment to the nuclear envelope. Cells lacking Mia1p exhibited less microtubule bundles. Microtubules could be nucleated and bundled but were frequently released from the nucleation sites in mia1delta cells. Mia1p was required for stability of microtubule bundles and persistent use of nucleation sites both in interphase and postanaphase array dynamics. The gamma-tubulin-rich material was not organized in large perinuclear or microtubule-associated structures in mia1delta cells. Interestingly, absence of microtubules in dividing wild-type cells prevented appearance of large gamma-tubulin-rich MTOC structures in daughters. When microtubule polymerization was allowed, MTOCs were efficiently assembled de novo. We propose a model where MTOC emergence is a self-organizing process requiring the continuous association of microtubules with nucleation sites.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Nuclear Envelope/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Cell Cycle , Gene Deletion , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/genetics , Microtubule-Organizing Center/chemistry , Microtubules/chemistry , Models, Biological , Schizosaccharomyces/genetics , Schizosaccharomyces/ultrastructure , Schizosaccharomyces pombe Proteins/analysis , Schizosaccharomyces pombe Proteins/genetics , Spindle Apparatus/metabolism , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...