Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
Science ; 375(6581): eabj3944, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35143306

ABSTRACT

Human oocytes are prone to assembling meiotic spindles with unstable poles, which can favor aneuploidy in human eggs. The underlying causes of spindle instability are unknown. We found that NUMA (nuclear mitotic apparatus protein)-mediated clustering of microtubule minus ends focused the spindle poles in human, bovine, and porcine oocytes and in mouse oocytes depleted of acentriolar microtubule-organizing centers (aMTOCs). However, unlike human oocytes, bovine, porcine, and aMTOC-free mouse oocytes have stable spindles. We identified the molecular motor KIFC1 (kinesin superfamily protein C1) as a spindle-stabilizing protein that is deficient in human oocytes. Depletion of KIFC1 recapitulated spindle instability in bovine and aMTOC-free mouse oocytes, and the introduction of exogenous KIFC1 rescued spindle instability in human oocytes. Thus, the deficiency of KIFC1 contributes to spindle instability in human oocytes.


Subject(s)
Cell Cycle Proteins/metabolism , Kinesins/deficiency , Oocytes/physiology , Oocytes/ultrastructure , Spindle Apparatus/physiology , Spindle Poles/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Cattle , Dynactin Complex/metabolism , Dyneins/metabolism , Female , Humans , Kinesins/genetics , Kinesins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/physiology , Microtubule-Organizing Center/ultrastructure , Microtubules/metabolism , Recombinant Proteins/metabolism , Spindle Apparatus/ultrastructure , Spindle Poles/ultrastructure , Swine
2.
PLoS Pathog ; 18(1): e1010223, 2022 01.
Article in English | MEDLINE | ID: mdl-35077503

ABSTRACT

Transmission of malaria-causing parasites to mosquitoes relies on the production of gametocyte stages and their development into gametes. These stages display various microtubule cytoskeletons and the architecture of the corresponding microtubule organisation centres (MTOC) remains elusive. Combining ultrastructure expansion microscopy (U-ExM) with bulk proteome labelling, we first reconstructed in 3D the subpellicular microtubule network which confers cell rigidity to Plasmodium falciparum gametocytes. Upon activation, as the microgametocyte undergoes three rounds of endomitosis, it also assembles axonemes to form eight flagellated microgametes. U-ExM combined with Pan-ExM further revealed the molecular architecture of the bipartite MTOC coordinating mitosis with axoneme formation. This MTOC spans the nuclear membrane linking cytoplasmic basal bodies to intranuclear bodies by proteinaceous filaments. In P. berghei, the eight basal bodies are concomitantly de novo assembled in a SAS6- and SAS4-dependent manner from a deuterosome-like structure, where centrin, γ-tubulin, SAS4 and SAS6 form distinct subdomains. Basal bodies display a fusion of the proximal and central cores where centrin and SAS6 are surrounded by a SAS4-toroid in the lumen of the microtubule wall. Sequential nucleation of axonemes and mitotic spindles is associated with a dynamic movement of γ-tubulin from the basal bodies to the intranuclear bodies. This dynamic architecture relies on two non-canonical regulators, the calcium-dependent protein kinase 4 and the serine/arginine-protein kinase 1. Altogether, these results provide insights into the molecular organisation of a bipartite MTOC that may reflect a functional transition of a basal body to coordinate axoneme assembly with mitosis.


Subject(s)
Axoneme/ultrastructure , Gametogenesis/physiology , Microscopy/methods , Microtubule-Organizing Center/ultrastructure , Mitosis/physiology , Plasmodium/physiology , Animals , Mice , Plasmodium/ultrastructure
3.
Nat Commun ; 12(1): 6042, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34654813

ABSTRACT

Centriole biogenesis and maintenance are crucial for cells to generate cilia and assemble centrosomes that function as microtubule organizing centers (MTOCs). Centriole biogenesis and MTOC function both require the microtubule nucleator γ-tubulin ring complex (γTuRC). It is widely accepted that γTuRC nucleates microtubules from the pericentriolar material that is associated with the proximal part of centrioles. However, γTuRC also localizes more distally and in the centriole lumen, but the significance of these findings is unclear. Here we identify spatially and functionally distinct subpopulations of centrosomal γTuRC. Luminal localization is mediated by augmin, which is linked to the centriole inner scaffold through POC5. Disruption of luminal localization impairs centriole integrity and interferes with cilium assembly. Defective ciliogenesis is also observed in γTuRC mutant fibroblasts from a patient suffering from microcephaly with chorioretinopathy. These results identify a non-canonical role of augmin-γTuRC in the centriole lumen that is linked to human disease.


Subject(s)
Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Microtubule-Associated Proteins/isolation & purification , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins/ultrastructure , Cell Line , Centrioles/ultrastructure , Centrosome/metabolism , Centrosome/ultrastructure , Cilia , Female , Humans , Male , Mice , Microtubule-Associated Proteins/ultrastructure , Microtubule-Organizing Center/ultrastructure , Microtubules/metabolism , Neurons
4.
Dev Biol ; 478: 1-12, 2021 10.
Article in English | MEDLINE | ID: mdl-34147472

ABSTRACT

Dorsal root ganglion (DRG) neurons are the predominant cell type that innervates the vertebrate skin. They are typically described as pseudounipolar cells that have central and peripheral axons branching from a single root exiting the cell body. The peripheral axon travels within a nerve to the skin, where free sensory endings can emerge and branch into an arbor that receives and integrates information. In some immature vertebrates, DRG neurons are preceded by Rohon-Beard (RB) neurons. While the sensory endings of RB and DRG neurons function like dendrites, we use live imaging in zebrafish to show that they have axonal plus-end-out microtubule polarity at all stages of maturity. Moreover, we show both cell types have central and peripheral axons with plus-end-out polarity. Surprisingly, in DRG neurons these emerge separately from the cell body, and most cells never acquire the signature pseudounipolar morphology. Like another recently characterized cell type that has multiple plus-end-out neurites, ganglion cells in Nematostella, RB and DRG neurons maintain a somatic microtubule organizing center even when mature. In summary, we characterize key cellular and subcellular features of vertebrate sensory neurons as a foundation for understanding their function and maintenance.


Subject(s)
Ganglia, Spinal/ultrastructure , Microtubules/ultrastructure , Sensory Receptor Cells/ultrastructure , Skin/innervation , Animals , Animals, Genetically Modified , Axons/physiology , Axons/ultrastructure , Cell Body/ultrastructure , Cell Polarity , Dendrites/physiology , Drosophila/cytology , Drosophila/growth & development , Ganglia, Spinal/physiology , Microtubule-Organizing Center/ultrastructure , Sea Anemones/cytology , Sea Anemones/growth & development , Sea Anemones/ultrastructure , Sensory Receptor Cells/physiology , Zebrafish
5.
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
6.
Cell Microbiol ; 23(3): e13284, 2021 03.
Article in English | MEDLINE | ID: mdl-33124706

ABSTRACT

The eukaryotic cell cycle is typically divided into distinct phases with cytokinesis immediately following mitosis. To ensure proper cell division, each phase is tightly coordinated via feedback controls named checkpoints. During its asexual replication cycle, the malaria parasite Plasmodium falciparum undergoes multiple asynchronous rounds of mitosis with segregation of uncondensed chromosomes followed by nuclear division with intact nuclear envelope. The multi-nucleated schizont is then subjected to a single round of cytokinesis that produces dozens of daughter cells called merozoites. To date, no cell cycle checkpoints have been identified that regulate the Plasmodium spp. mode of division. Here, we identify the Plasmodium homologue of the Mini-Chromosome Maintenance Complex Binding Protein (PfMCMBP), which co-purified with the Mini-Chromosome Maintenance (MCM) complex, a replicative helicase required for genomic DNA replication. By conditionally depleting PfMCMBP, we disrupt nuclear morphology and parasite proliferation without causing a block in DNA replication. By immunofluorescence microscopy, we show that PfMCMBP depletion promotes the formation of mitotic spindle microtubules with extensions to more than one DNA focus and abnormal centrin distribution. Strikingly, PfMCMBP-deficient parasites complete cytokinesis and form aneuploid merozoites with variable cellular and nuclear sizes. Our study demonstrates that the parasite lacks a robust checkpoint response to prevent cytokinesis following aberrant karyokinesis.


Subject(s)
Cell Nucleus Division , Cytokinesis , Minichromosome Maintenance Proteins/metabolism , Plasmodium falciparum/cytology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Chromosomes/metabolism , Chromosomes/ultrastructure , Gene Knockdown Techniques , Merozoites/cytology , Merozoites/growth & development , Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/ultrastructure , Nuclear Proteins/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Schizonts/physiology
7.
Virology ; 549: 13-24, 2020 10.
Article in English | MEDLINE | ID: mdl-32805585

ABSTRACT

Filamentous mammalian orthoreovirus (MRV) viral factories (VFs) are membrane-less cytosolic inclusions in which virus transcription, replication of dsRNA genome segments, and packaging of virus progeny into newly synthesized virus cores take place. In infected cells, the MRV µ2 protein forms punctae in the enlarged region of the filamentous VFs that are co-localized with γ-tubulin and resistant to nocodazole treatment, and permitted microtubule (MT)-extension, features common to MT-organizing centers (MTOCs). Using a previously established reconstituted VF model, we addressed the functions of MT-components and MTOCs concerning their roles in the formation of filamentous VFs. Indeed, the MTOC markers γ-tubulin and centrin were redistributed within the VF-like structures (VFLS) in a µ2-dependent manner. Moreover, the MT-nucleation centers significantly increased in numbers, and γ-tubulin was pulled-down in a binding assay when co-expressed with histidine-tagged-µ2 and µNS. Thus, µ2, by interaction with γ-tubulin, can modulate MTOCs localization and function according to viral needs.


Subject(s)
Host-Pathogen Interactions/genetics , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Orthoreovirus, Mammalian/genetics , Tubulin/genetics , Viral Proteins/genetics , Animals , Cell Line , Chlorocebus aethiops , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation , Microtubule-Organizing Center/drug effects , Microtubule-Organizing Center/ultrastructure , Microtubule-Organizing Center/virology , Microtubules/drug effects , Microtubules/ultrastructure , Microtubules/virology , Nocodazole/pharmacology , Orthoreovirus, Mammalian/drug effects , Orthoreovirus, Mammalian/metabolism , Signal Transduction , Tubulin/metabolism , Tubulin Modulators/pharmacology , Viral Proteins/metabolism , Virus Replication/drug effects
8.
Nat Cell Biol ; 22(3): 297-309, 2020 03.
Article in English | MEDLINE | ID: mdl-32066907

ABSTRACT

Non-centrosomal microtubule-organizing centres (ncMTOCs) have a variety of roles that are presumed to serve the diverse functions of the range of cell types in which they are found. ncMTOCs are diverse in their composition, subcellular localization and function. Here we report a perinuclear MTOC in Drosophila fat body cells that is anchored by the Nesprin homologue Msp300 at the cytoplasmic surface of the nucleus. Msp300 recruits the microtubule minus-end protein Patronin, a calmodulin-regulated spectrin-associated protein (CAMSAP) homologue, which functions redundantly with Ninein to further recruit the microtubule polymerase Msps-a member of the XMAP215 family-to assemble non-centrosomal microtubules and does so independently of the widespread microtubule nucleation factor γ-Tubulin. Functionally, the fat body ncMTOC and the radial microtubule arrays that it organizes are essential for nuclear positioning and for secretion of basement membrane components via retrograde dynein-dependent endosomal trafficking that restricts plasma membrane growth. Together, this study identifies a perinuclear ncMTOC with unique architecture that regulates microtubules, serving vital functions.


Subject(s)
Basement Membrane/metabolism , Cell Nucleus , Microtubule-Organizing Center/physiology , Actins/physiology , Animals , Cell Membrane , Cell Nucleus/ultrastructure , Centrosome , Drosophila/metabolism , Drosophila/ultrastructure , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Dyneins/physiology , Endosomes/metabolism , Fat Body/metabolism , Fat Body/ultrastructure , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/physiology , Microtubule-Organizing Center/ultrastructure , Microtubules/physiology , Muscle Proteins/metabolism , Tubulin/physiology
9.
Cell ; 180(1): 165-175.e16, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31862189

ABSTRACT

The γ-tubulin ring complex (γ-TuRC) is an essential regulator of centrosomal and acentrosomal microtubule formation, yet its structure is not known. Here, we present a cryo-EM reconstruction of the native human γ-TuRC at ∼3.8 Å resolution, revealing an asymmetric, cone-shaped structure. Pseudo-atomic models indicate that GCP4, GCP5, and GCP6 form distinct Y-shaped assemblies that structurally mimic GCP2/GCP3 subcomplexes distal to the γ-TuRC "seam." We also identify an unanticipated structural bridge that includes an actin-like protein and spans the γ-TuRC lumen. Despite its asymmetric architecture, the γ-TuRC arranges γ-tubulins into a helical geometry poised to nucleate microtubules. Diversity in the γ-TuRC subunits introduces large (>100,000 Å2) surfaces in the complex that allow for interactions with different regulatory factors. The observed compositional complexity of the γ-TuRC could self-regulate its assembly into a cone-shaped structure to control microtubule formation across diverse contexts, e.g., within biological condensates or alongside existing filaments.


Subject(s)
Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/ultrastructure , Tubulin/ultrastructure , Actins/metabolism , Cryoelectron Microscopy/methods , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/ultrastructure , Microtubules/metabolism , Tubulin/metabolism
10.
Curr Opin Plant Biol ; 46: 1-7, 2018 12.
Article in English | MEDLINE | ID: mdl-29981930

ABSTRACT

Centrosomes play various critical roles in animal cells such as microtubule nucleation and stabilization, mitotic spindle morphogenesis, and spindle orientation. Land plants have lost centrosomes and yet must execute many of these functions. Recent studies have revealed the crucial roles played by morphologically distinct cytoplasmic microtubule-organizing centers (MTOCs) in initiating spindle bipolarity and maintaining spindle orientation robustness. These MTOCs resemble centrosomes in many aspects, implying an evolutionary divergence of MT-organizing structures in plants. However, their functions rely on conserved nucleation and amplification mechanisms, indicating a similarity in MT network establishment between animals and plants. Moreover, recent characterization of a plant-specific MT minus-end tracking protein suggests that plants have developed functionally equivalent modules to stabilize and organize MTs at minus ends. These findings support the theory that plants overcome centrosome loss by utilizing modified but substantially conserved mechanisms to organize MT networks.


Subject(s)
Centrosome/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Plant Cells/physiology , Centrosome/ultrastructure , Microtubule-Organizing Center/ultrastructure , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
11.
Nat Commun ; 8(1): 2236, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269729

ABSTRACT

Apicomplexan parasites are typified by an apical complex that contains a unique microtubule-organizing center (MTOC) that organizes the cytoskeleton. In apicomplexan parasites such as Toxoplasma gondii, the apical complex includes a spiral cap of tubulin-rich fibers called the conoid. Although described ultrastructurally, the composition and functions of the conoid are largely unknown. Here, we localize 11 previously undescribed apical proteins in T. gondii and identify an essential component named conoid protein hub 1 (CPH1), which is conserved in apicomplexan parasites. CPH1 contains ankyrin repeats that are required for structural integrity of the conoid, parasite motility, and host cell invasion. Proximity labeling and protein interaction network analysis reveal that CPH1 functions as a hub linking key motor and structural proteins that contain intrinsically disordered regions and coiled coil domains. Our findings highlight the importance of essential protein hubs in controlling biological networks of MTOCs in early-branching protozoan parasites.


Subject(s)
Microtubule-Organizing Center/metabolism , Movement , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Ankyrin Repeat , Apicomplexa/genetics , Apicomplexa/metabolism , Cytoskeleton/metabolism , Microtubule-Organizing Center/ultrastructure , Proteome/metabolism , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/pathogenicity , Toxoplasma/ultrastructure , Tubulin/metabolism
12.
J Biol Chem ; 292(50): 20394-20409, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29054927

ABSTRACT

Microtubules in animal cells assemble (nucleate) from both the centrosome and the cis-Golgi cisternae. A-kinase anchor protein 350 kDa (AKAP350A, also called AKAP450/CG-NAP/AKAP9) is a large scaffolding protein located at both the centrosome and Golgi apparatus. Previous findings have suggested that AKAP350 is important for microtubule dynamics at both locations, but how this scaffolding protein assembles microtubule nucleation machinery is unclear. Here, we found that overexpression of the C-terminal third of AKAP350A, enhanced GFP-AKAP350A(2691-3907), induces the formation of multiple microtubule-nucleation centers (MTNCs). Nevertheless, these induced MTNCs lacked "true" centriole proteins, such as Cep135. Mapping analysis with AKAP350A truncations demonstrated that AKAP350A contains discrete regions responsible for promoting or inhibiting the formation of multiple MTNCs. Moreover, GFP-AKAP350A(2691-3907) recruited several pericentriolar proteins to MTNCs, including γ-tubulin, pericentrin, Cep68, Cep170, and Cdk5RAP2. Proteomic analysis indicated that Cdk5RAP2 and Cep170 both interact with the microtubule nucleation-promoting region of AKAP350A, whereas Cep68 interacts with the distal C-terminal AKAP350A region. Yeast two-hybrid assays established a direct interaction of Cep170 with AKAP350A. Super-resolution and deconvolution microscopy analyses were performed to define the association of AKAP350A with centrosomes, and these studies disclosed that AKAP350A spans the bridge between centrioles, co-localizing with rootletin and Cep68 in the linker region. siRNA-mediated depletion of AKAP350A caused displacement of both Cep68 and Cep170 from the centrosome. These results suggest that AKAP350A acts as a scaffold for factors involved in microtubule nucleation at the centrosome and coordinates the assembly of protein complexes associating with the intercentriolar bridge.


Subject(s)
A Kinase Anchor Proteins/metabolism , Centrosome/metabolism , Cytoskeletal Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Models, Molecular , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , A Kinase Anchor Proteins/antagonists & inhibitors , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/genetics , Biomarkers/metabolism , Cell Cycle Proteins , Cell Line , Centrosome/ultrastructure , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Humans , Imaging, Three-Dimensional , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Organizing Center/ultrastructure , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Multimerization , Proteomics/methods , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Two-Hybrid System Techniques
13.
Dev Cell ; 39(4): 424-437, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27818179

ABSTRACT

Vertebrate cells can initiate ciliogenesis from centrioles at the cell center, near the Golgi, forming primary cilia confined or submerged in a deep narrow pit created by membrane invagination. How or why cells maintain submerged cilia is unclear. Here, by characterizing centriole subdistal appendages (sDAP) in cells exclusively growing submerged cilia, we found that a group of sDAP components localize to the centriole proximal end through the cohesion factor C-Nap1 and that sDAP function redundantly with C-Nap1 for submerged cilia maintenance. Loss of sDAP and C-Nap1 has no effect on cilia assembly, but it disrupts stable Golgi-cilia association and allows normally submerged cilia to fully surface, losing the deep membrane invagination. Intriguingly, unlike submerged cilia (stationary), surfaced cilia actively respond to mechanical stimuli with motions and can ectopically recruit Hedgehog signaling components in the absence of agonist. We propose that spatial control of ciliogenesis uncouples or specifies sensory properties of cilia.


Subject(s)
Cilia/metabolism , Morphogenesis , Autoantigens/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Centrioles/metabolism , Centrioles/ultrastructure , Centrosome/metabolism , Centrosome/ultrastructure , Cilia/ultrastructure , Gene Knockout Techniques , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Hedgehog Proteins/metabolism , Humans , Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/ultrastructure , Microtubules/metabolism , Microtubules/ultrastructure , Motion , Mutation/genetics , Rheology , Sensation , Signal Transduction
14.
Eur J Protistol ; 54: 74-82, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27161993

ABSTRACT

In this detailed electron microscopic study of Paradermamoeba levis Smirnov et Goodkov 1994 (phylum Amoebozoa, class Discosea, subclass Longamoebia, order Dermamoebida, family Dermamoebidae) based on numerous fixations and studies of a large number of cells we provide the first comprehensive description of the ultrastructure of this species. P. levis possesses cytoplasmic microtubule-organizing centres (MTOCs) associated with dictyosomes of the Golgi complex. This finding adds evidence to our earlier suggestion that the presence of cytoplasmic MTOCs is a synapomorphy of the phylogenetic lineages forming the subclass Longamoebia. The so-called "supernumerary nucleus" of P. levis noted in the initial description was found to be not an individual structure but an outgrowth of the cell nucleus containing its own nucleolus. Enigmatic trichocyst-like bodies were noted in all studied strains, originating from different geographic locations. This proves that these bodies are integral parts of the cell structure, not an occasional property of the type strain. P. levis is now reliably recorded from several European locations (North-Western Russia, Croatia, Switzerland, UK) and Far East of Russia.


Subject(s)
Amoebozoa/ultrastructure , Microtubule-Organizing Center/ultrastructure , Cytoplasm/ultrastructure , Golgi Apparatus/ultrastructure , Microscopy, Electron, Transmission , Species Specificity
15.
Cell Rep ; 15(1): 54-60, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27052165

ABSTRACT

Errors during cell division in oocytes and early embryos are linked to birth defects in mammals. Bipolar spindle assembly in early mouse embryos is unique in that three or more acentriolar microtubule-organizing centers (MTOCs) are initially formed and are then clustered into two spindle poles. Using a knockout mouse and live imaging of spindles in embryos, we demonstrate that MTOC clustering during the blastocyst stage requires augmin, a critical complex for MT-dependent MT nucleation within the spindle. Functional analyses in cultured cells with artificially increased numbers of centrosomes indicate that the lack of intra-spindle MT nucleation, but not loss of augmin per se or overall reduction of spindle MTs, is the cause of clustering failure. These data suggest that onset of mitosis with three or more MTOCs is turned into a typical bipolar division through augmin-dependent intra-spindle MT assembly.


Subject(s)
Blastocyst/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Animals , Blastocyst/ultrastructure , Cells, Cultured , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Organizing Center/ultrastructure , Mitosis
16.
Tissue Cell ; 48(2): 104-13, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26899558

ABSTRACT

The ancestral eukaryotes presumably had an MTOC (microtubule organizing center) which late gave origin to the centriole and the flagellar axoneme. The centrosome of insect early spermatids is in general composed of two components: a single centriole and a cloud of electron-dense pericentriolar material (PCM). During spermiogenesis, the centriole changes its structure and gives rise to a flagellar axoneme, while the proteins of PCM, gamma tubulin in particular, are involved in the production of microtubules for the elongation and shaping of spermatid components. At the end of spermiogenesis, in many insects, additional material is deposited beneath the nucleus to form the centriole adjunct (ca). This material can also extend along the flagellum in two accessory bodies (ab) flanking the axoneme. Among Homoptera Sternorrhyncha, a progressive modification of their sperm flagella until complete disappearance has been verified. In the Archaeococcidae Matsucoccus feytaudi, however, a motile sperm flagellum-like structure is formed by an MTOC activity. This finding gives support to the hypothesis that an evolutionary reversal has occurred in the group and that the cell, when a non-functional centriole is present, activates an ancestral structure, an MTOC, to form a polarized motile bundle of microtubules restoring sperm motility. The presence and extension of the centriole adjunct in the different insect orders is also enlisted.


Subject(s)
Biological Evolution , Sperm Tail/ultrastructure , Spermatids/ultrastructure , Spermatogenesis/genetics , Animals , Axoneme/genetics , Axoneme/ultrastructure , Centrioles/genetics , Centrioles/ultrastructure , Centrosome/ultrastructure , Hemiptera/genetics , Hemiptera/ultrastructure , Male , Microscopy, Electron, Transmission , Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/ultrastructure
17.
Cell Cycle ; 14(22): 3566-79, 2015.
Article in English | MEDLINE | ID: mdl-26654596

ABSTRACT

Polo-like kinase 1 (Plk1) is pivotal for proper mitotic progression, its targeting activity is regulated by precise subcellular positioning and phosphorylation. Here we assessed the protein expression, subcellular localization and possible functions of phosphorylated Plk1 (pPlk1(Ser137) and pPlk1(Thr210)) in mouse oocytes during meiotic division. Western blot analysis revealed a peptide of pPlk1(Ser137) with high and stable expression from germinal vesicle (GV) until metaphase II (MII), while pPlk1(Thr210) was detected as one large single band at GV stage and 2 small bands after germinal vesicle breakdown (GVBD), which maintained stable up to MII. Immunofluorescence analysis showed pPlk1(Ser137) was colocalized with microtubule organizing center (MTOC) proteins, γ-tubulin and pericentrin, on spindle poles, concomitantly with persistent concentration at centromeres and dynamic aggregation between chromosome arms. Differently, pPlk1(Thr210) was persistently distributed across the whole body of chromosomes after meiotic resumption. The specific Plk1 inhibitor, BI2536, repressed pPlk1(Ser137) accumulation at MTOCs and between chromosome arms, consequently disturbed γ-tubulin and pericentrin recruiting to MTOCs, destroyed meiotic spindle formation, and delayed REC8 cleavage, therefore arresting oocytes at metaphase I (MI) with chromosome misalignment. BI2536 completely reversed the premature degradation of REC8 and precocious segregation of chromosomes induced with okadaic acid (OA), an inhibitor to protein phosphatase 2A. Additionally, the protein levels of pPlk1(Ser137) and pPlk1(Thr210), as well as the subcellular distribution of pPlk1(Thr210), were not affected by BI2536. Taken together, our results demonstrate that Plk1 activity is required for meiotic spindle assembly and REC8 cleavage, with pPlk1(Ser137) is the action executor, in mouse oocytes during meiotic division.


Subject(s)
Cell Cycle Proteins/genetics , Meiosis , Nuclear Proteins/genetics , Oocytes/metabolism , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , Antigens/genetics , Antigens/metabolism , Antimitotic Agents/pharmacology , Cell Cycle Proteins/metabolism , Chromosome Segregation , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Cumulus Cells/ultrastructure , Cytokinesis/genetics , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/ultrastructure , Nuclear Proteins/metabolism , Okadaic Acid/pharmacology , Oocytes/drug effects , Oocytes/ultrastructure , Phosphoproteins/metabolism , Phosphorylation , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Proto-Oncogene Proteins/metabolism , Pteridines/pharmacology , Signal Transduction , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Tubulin/genetics , Tubulin/metabolism , Polo-Like Kinase 1
18.
Arthropod Struct Dev ; 44(3): 237-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25805602

ABSTRACT

A microtubule organizing centre (MTOC) has been described in the spermatid of the hemipteran Matsucoccus feytaudi (Coccoidea). This structure, revealed as a fluorescent ring by treatment with γ-tubulin antibody, gives rise to a bundle of microtubules which surrounds the elongated cylindrical nucleus. This microtubule bundle has been considered an atypical sperm flagellum provided with sperm motility. A comparison of the M. feytaudi MTOC with the material associated with the centriole of Drosophila melanogaster spermatids confirms the great similarity between the two structures, both involved in the nucleation of microtubules. Like the D. melanogaster material associated with the centriole, the M. feytaudi MTOC is a transient structure which disappears or degenerates at the end of spermiogenesis and is no longer visible in the mature sperm.


Subject(s)
Drosophila melanogaster/physiology , Hemiptera/physiology , Microtubule-Organizing Center/ultrastructure , Sperm Tail/ultrastructure , Spermatogenesis , Animals , Drosophila melanogaster/growth & development , Drosophila melanogaster/ultrastructure , Fluorescent Antibody Technique , Hemiptera/growth & development , Hemiptera/ultrastructure , Male , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nymph/growth & development , Nymph/physiology , Nymph/ultrastructure , Pupa/growth & development , Pupa/physiology , Pupa/ultrastructure , Tubulin
19.
Cell Cycle ; 14(6): 880-93, 2015.
Article in English | MEDLINE | ID: mdl-25714331

ABSTRACT

Cyclin-dependent kinases (CDKs) are central regulators of eukaryotic cell cycle progression. In contrast to interphase CDKs, the mitotic phase CDK1 is the only CDK capable of driving the entire cell cycle and it can do so from yeast to mammals. Interestingly, plants and the marine chordate, Oikopleura dioica, possess paralogs of the highly conserved CDK1 regulator. However, whereas in plants the 2 CDK1 paralogs replace interphase CDK functions, O. dioica has a full complement of interphase CDKs in addition to its 5 odCDK1 paralogs. Here we show specific sub-functionalization of odCDK1 paralogs during oogenesis. Differential spatiotemporal dynamics of the odCDK1a, d and e paralogs and the meiotic polo-like kinase 1 (Plk1) and aurora kinase determine the subset of meiotic nuclei in prophase I arrest that will seed growing oocytes and complete meiosis. Whereas we find odCDK1e to be non-essential, knockdown of the odCDK1a paralog resulted in the spawning of non-viable oocytes of reduced size. Knockdown of odCDK1d also resulted in the spawning of non-viable oocytes. In this case, the oocytes were of normal size, but were unable to extrude polar bodies upon exposure to sperm, because they were unable to resume meiosis from prophase I arrest, a classical function of the sole CDK1 during meiosis in other organisms. Thus, we reveal specific sub-functionalization of CDK1 paralogs, during the meiotic oogenic program.


Subject(s)
CDC2 Protein Kinase/metabolism , Chordata/metabolism , Meiosis , Oogenesis , Sequence Homology, Amino Acid , Animals , Gene Knockdown Techniques , Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/ultrastructure , Nuclear Envelope/metabolism , Phenotype , RNA, Double-Stranded/metabolism
20.
Nat Commun ; 5: 4888, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25215410

ABSTRACT

Coordination of ciliary beating is essential to ensure mucus clearance in the airway tract. The orientation and synchronization of ciliary motion responds in part to the organization of the underlying cytoskeletal networks. Using electron tomography on mouse trachea, we show that basal bodies are collectively hooked at the cortex by a regular microtubule array composed of 4-5 microtubules. Removal of galectin-3, one of basal-body components, provokes misrecruitment of γ-tubulin, disorganization of this microtubule framework emanating from the basal-foot cap, together with loss of basal-body alignment and cilium orientation, defects in cilium organization and reduced fluid flow in the tracheal lumen. We conclude that galectin-3 plays a crucial role in the maintenance of the microtubule-organizing centre of the cilium and the 'pillar' microtubules, and that this network is instrumental for the coordinated orientation and stabilization of motile cilia.


Subject(s)
Cilia/ultrastructure , Galectin 3/genetics , Microtubule-Organizing Center/ultrastructure , Microtubules/ultrastructure , Respiratory Mucosa/ultrastructure , Trachea/ultrastructure , Animals , Cilia/metabolism , Galectin 3/deficiency , Gene Expression , Male , Mice , Mice, Knockout , Microscopy, Electron , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Respiratory Mucosa/metabolism , Rheology , Trachea/metabolism , Tubulin/genetics , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL