Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Curr Biol ; 33(19): 4202-4216.e9, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37729913

ABSTRACT

Proper centrosome number and function relies on the accurate assembly of centrioles, barrel-shaped structures that form the core duplicating elements of the organelle. The growth of centrioles is regulated in a cell cycle-dependent manner; while new daughter centrioles elongate during the S/G2/M phase, mature mother centrioles maintain their length throughout the cell cycle. Centriole length is controlled by the synchronized growth of the microtubules that ensheathe the centriole barrel. Although proteins exist that target the growing distal tips of centrioles, such as CP110 and Cep97, these proteins are generally thought to suppress centriolar microtubule growth, suggesting that distal tips may also contain unidentified counteracting factors that facilitate microtubule polymerization. Currently, a mechanistic understanding of how distal tip proteins balance microtubule growth and shrinkage to either promote daughter centriole elongation or maintain centriole length is lacking. Using a proximity-labeling screen in Drosophila cells, we identified Cep104 as a novel component of a group of evolutionarily conserved proteins that we collectively refer to as the distal tip complex (DTC). We found that Cep104 regulates centriole growth and promotes centriole elongation through its microtubule-binding TOG domain. Furthermore, analysis of Cep104 null flies revealed that Cep104 and Cep97 cooperate during spermiogenesis to align spermatids and coordinate individualization. Lastly, we mapped the complete DTC interactome and showed that Cep97 is the central scaffolding unit required to recruit DTC components to the distal tip of centrioles.


Subject(s)
Centrioles , Microtubule-Associated Proteins , Male , Animals , Centrioles/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Drosophila/metabolism , Centrosome/metabolism , Spermatogenesis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
2.
Curr Biol ; 33(14): 3031-3040.e6, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37379844

ABSTRACT

Centrosomes are multi-protein organelles that function as microtubule (MT) organizing centers (MTOCs), ensuring spindle formation and chromosome segregation during cell division.1,2,3 Centrosome structure includes core centrioles that recruit pericentriolar material (PCM) that anchors γ-tubulin to nucleate MTs.1,2 In Drosophila melanogaster, PCM organization depends on proper regulation of proteins like Spd-2, which dynamically localizes to centrosomes and is required for PCM, γ-tubulin, and MTOC activity in brain neuroblast (NB) mitosis and male spermatocyte (SC) meiosis.4,5,6,7,8 Some cells have distinct requirements for MTOC activity due to differences in characteristics like cell size9,10 or whether they are mitotic or meiotic.11,12 How centrosome proteins achieve cell-type-specific functional differences is poorly understood. Previous work identified alternative splicing13 and binding partners14 as contributors to cell-type-specific differences in centrosome function. Gene duplication, which can generate paralogs with specialized functions,15,16 is also implicated in centrosome gene evolution,17 including cell-type-specific centrosome genes.18,19 To gain insight into cell-type-specific differences in centrosome protein function and regulation, we investigated a duplication of Spd-2 in Drosophila willistoni, which has Spd-2A (ancestral) and Spd-2B (derived). We find that Spd-2A functions in NB mitosis, whereas Spd-2B functions in SC meiosis. Ectopically expressed Spd-2B accumulates and functions in mitotic NBs, but ectopically expressed Spd-2A failed to accumulate in meiotic SCs, suggesting cell-type-specific differences in translation or protein stability. We mapped this failure to accumulate and function in meiosis to the C-terminal tail domain of Spd-2A, revealing a novel regulatory mechanism that can potentially achieve differences in PCM function across cell types.


Subject(s)
Cytoskeletal Proteins , Drosophila Proteins , Drosophila , Gene Duplication , Tubulin , Animals , Male , Centrioles/genetics , Centrioles/metabolism , Centrosome/metabolism , Drosophila/genetics , Drosophila/metabolism , Meiosis , Mitosis , Tubulin/metabolism , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics
3.
Mol Biol Cell ; 34(9): br15, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37342879

ABSTRACT

Centrosomes are essential parts of diverse cellular processes, and precise regulation of the levels of their constituent proteins is critical for their function. One such protein is Pericentrin (PCNT) in humans and Pericentrin-like protein (PLP) in Drosophila. Increased PCNT expression and its protein accumulation are linked to clinical conditions including cancer, mental disorders, and ciliopathies. However, the mechanisms by which PCNT levels are regulated remain underexplored. Our previous study demonstrated that PLP levels are sharply down-regulated during early spermatogenesis and this regulation is essential to spatially position PLP on the proximal end of centrioles. We hypothesized that the sharp drop in PLP protein was a result of rapid protein degradation during the male germ line premeiotic G2 phase. Here, we show that PLP is subject to ubiquitin-mediated degradation and identify multiple proteins that promote the reduction of PLP levels in spermatocytes, including the UBR box containing E3 ligase Poe (UBR4), which we show binds to PLP. Although protein sequences governing posttranslational regulation of PLP are not restricted to a single region of the protein, we identify a region that is required for Poe-mediated degradation. Experimentally stabilizing PLP, via internal PLP deletions or loss of Poe, leads to PLP accumulation in spermatocytes, its mispositioning along centrioles, and defects in centriole docking in spermatids.


Subject(s)
Centrioles , Ubiquitin-Protein Ligases , Male , Humans , Ubiquitin-Protein Ligases/metabolism , Centrioles/metabolism , Centrosome/metabolism , Antigens/metabolism
4.
J Cell Biol ; 221(9)2022 09 05.
Article in English | MEDLINE | ID: mdl-35929834

ABSTRACT

Centrosome positioning is essential for their function. Typically, centrosomes are transported to various cellular locations through the interaction of centrosomal microtubules (MTs) with motor proteins anchored at the cortex or the nuclear surface. However, it remains unknown how centrioles migrate in cellular contexts in which they do not nucleate MTs. Here, we demonstrate that during interphase, inactive centrioles move directly along the interphase MT network as Kinesin-1 cargo. We identify Pericentrin-Like-Protein (PLP) as a novel Kinesin-1 interacting molecule essential for centriole motility. In vitro assays show that PLP directly interacts with the cargo binding domain of Kinesin-1, allowing PLP to migrate on MTs. Binding assays using purified proteins revealed that relief of Kinesin-1 autoinhibition is critical for its interaction with PLP. Finally, our studies of neural stem cell asymmetric divisions in the Drosophila brain show that the PLP-Kinesin-1 interaction is essential for the timely separation of centrioles, the asymmetry of centrosome activity, and the age-dependent centrosome inheritance.


Subject(s)
Antigens , Centrioles , Kinesins , Animals , Antigens/metabolism , Calmodulin-Binding Proteins/metabolism , Centrioles/metabolism , Centrosome/metabolism , Drosophila , Drosophila Proteins/metabolism , Kinesins/metabolism , Microtubules/metabolism , Neural Stem Cells , Protein Transport
5.
Dev Cell ; 53(1): 86-101.e7, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32169161

ABSTRACT

The centriole, or basal body, is the center of attachment between the sperm head and tail. While the distal end of the centriole templates the cilia, the proximal end associates with the nucleus. Using Drosophila, we identify a centriole-centric mechanism that ensures proper proximal end docking to the nucleus. This mechanism relies on the restriction of pericentrin-like protein (PLP) and the pericentriolar material (PCM) to the proximal end of the centriole. PLP is restricted proximally by limiting its mRNA and protein to the earliest stages of centriole elongation. Ectopic positioning of PLP to more distal portions of the centriole is sufficient to redistribute PCM and microtubules along the entire centriole length. This results in erroneous, lateral centriole docking to the nucleus, leading to spermatid decapitation as a result of a failure to form a stable head-tail linkage.


Subject(s)
Centrioles/metabolism , Centrosome/metabolism , Microtubules/metabolism , Sperm Head/metabolism , Sperm Tail/metabolism , Animals , Basal Bodies/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Male
6.
J Cell Biol ; 219(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31841145

ABSTRACT

During centriole duplication, a preprocentriole forms at a single site on the mother centriole through a process that includes the hierarchical recruitment of a conserved set of proteins, including the Polo-like kinase 4 (Plk4), Ana2/STIL, and the cartwheel protein Sas6. Ana2/STIL is critical for procentriole assembly, and its recruitment is controlled by the kinase activity of Plk4, but how this works remains poorly understood. A structural motif called the G-box in the centriole outer wall protein Sas4 interacts with a short region in the N terminus of Ana2/STIL. Here, we show that binding of Ana2 to the Sas4 G-box enables hyperphosphorylation of the Ana2 N terminus by Plk4. Hyperphosphorylation increases the affinity of the Ana2-G-box interaction, and, consequently, promotes the accumulation of Ana2 at the procentriole to induce daughter centriole formation.


Subject(s)
Cell Cycle Proteins/genetics , Centrioles/genetics , Drosophila Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Cell Cycle/genetics , Cell Line , Drosophila melanogaster/genetics , Intracellular Signaling Peptides and Proteins/genetics , Microtubule-Associated Proteins/genetics , Phosphorylation/genetics , Protein Binding/genetics
7.
Mol Biol Cell ; 30(8): 992-1007, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30726162

ABSTRACT

Cell division is critical for development, organ growth, and tissue repair. The later stages of cell division include the formation of the microtubule (MT)-rich central spindle in anaphase, which is required to properly define the cell equator, guide the assembly of the acto-myosin contractile ring and ultimately ensure complete separation and isolation of the two daughter cells via abscission. Much is known about the molecular machinery that forms the central spindle, including proteins needed to generate the antiparallel overlapping interzonal MTs. One critical protein that has garnered great attention is the protein regulator of cytokinesis 1, or Fascetto (Feo) in Drosophila, which forms a homodimer to cross-link interzonal MTs, ensuring proper central spindle formation and cytokinesis. Here, we report on a new direct protein interactor and regulator of Feo we named Feo interacting protein (FIP). Loss of FIP results in a reduction in Feo localization, rapid disassembly of interzonal MTs, and several defects related to cytokinesis failure, including polyploidization of neural stem cells. Simultaneous reduction in Feo and FIP results in very large, tumorlike DNA-filled masses in the brain that contain hundreds of centrosomes. In aggregate, our data show that FIP acts directly on Feo to ensure fully accurate cell division.


Subject(s)
Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/metabolism , Anaphase/physiology , Animals , Cell Division/physiology , Centrosome/metabolism , Cytokinesis , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Embryonic Development , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , Myosins/metabolism , Protein Interaction Domains and Motifs/physiology , Spindle Apparatus/metabolism
8.
PLoS One ; 13(1): e0190530, 2018.
Article in English | MEDLINE | ID: mdl-29315319

ABSTRACT

The centrosome serves as the main microtubule-organizing center in metazoan cells, yet despite its functional importance, little is known mechanistically about the structure and organizational principles that dictate protein organization in the centrosome. In particular, the protein-protein interactions that allow for the massive structural transition between the tightly organized interphase centrosome and the highly expanded matrix-like arrangement of the mitotic centrosome have been largely uncharacterized. Among the proteins that undergo a major transition is the Drosophila melanogaster protein centrosomin that contains a conserved carboxyl terminus motif, CM2. Recent crystal structures have shown this motif to be dimeric and capable of forming an intramolecular interaction with a central region of centrosomin. Here we use a combination of in-cell microscopy and in vitro oligomer assessment to show that dimerization is not necessary for CM2 recruitment to the centrosome and that CM2 alone undergoes significant cell cycle dependent rearrangement. We use NMR binding assays to confirm this intramolecular interaction and show that residues involved in solution are consistent with the published crystal structure and identify L1137 as critical for binding. Additionally, we show for the first time an in vitro interaction of CM2 with the Drosophila pericentrin-like-protein that exploits the same set of residues as the intramolecular interaction. Furthermore, NMR experiments reveal a calcium sensitive interaction between CM2 and calmodulin. Although unexpected because of sequence divergence, this suggests that centrosomin-mediated assemblies, like the mammalian pericentrin, may be calcium regulated. From these results, we suggest an expanded model where during interphase CM2 interacts with pericentrin-like-protein to form a layer of centrosomin around the centriole wall and that at the onset of mitosis this population acts as a nucleation site of intramolecular centrosomin interactions that support the expansion into the metaphase matrix.


Subject(s)
Cell Cycle/physiology , Drosophila Proteins/metabolism , Homeodomain Proteins/metabolism , Animals , Binding Sites , Drosophila Proteins/physiology , Drosophila melanogaster , Homeodomain Proteins/physiology , Nuclear Magnetic Resonance, Biomolecular , Polymerase Chain Reaction , Protein Binding , Two-Hybrid System Techniques
9.
Nat Commun ; 7: 12476, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27558293

ABSTRACT

The centrosome is the major microtubule-organizing centre of many cells, best known for its role in mitotic spindle organization. How the proteins of the centrosome are accurately assembled to carry out its many functions remains poorly understood. The non-membrane-bound nature of the centrosome dictates that protein-protein interactions drive its assembly and functions. To investigate this massive macromolecular organelle, we generated a 'domain-level' centrosome interactome using direct protein-protein interaction data from a focused yeast two-hybrid screen. We then used biochemistry, cell biology and the model organism Drosophila to provide insight into the protein organization and kinase regulatory machinery required for centrosome assembly. Finally, we identified a novel role for Plk4, the master regulator of centriole duplication. We show that Plk4 phosphorylates Cep135 to properly position the essential centriole component Asterless. This interaction landscape affords a critical framework for research of normal and aberrant centrosomes.


Subject(s)
Centrosome/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Duplication , Organelles/metabolism , Protein Interaction Maps , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Phosphorylation , Protein Binding , Protein Multimerization , Substrate Specificity
10.
J Cell Biol ; 213(4): 435-50, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27185836

ABSTRACT

Centrioles are the foundation of two organelles, centrosomes and cilia. Centriole numbers and functions are tightly controlled, and mutations in centriole proteins are linked to a variety of diseases, including microcephaly. Loss of the centriole protein Asterless (Asl), the Drosophila melanogaster orthologue of Cep152, prevents centriole duplication, which has limited the study of its nonduplication functions. Here, we identify populations of cells with Asl-free centrioles in developing Drosophila tissues, allowing us to assess its duplication-independent function. We show a role for Asl in controlling centriole length in germline and somatic tissue, functioning via the centriole protein Cep97. We also find that Asl is not essential for pericentriolar material recruitment or centrosome function in organizing mitotic spindles. Lastly, we show that Asl is required for proper basal body function and spermatid axoneme formation. Insights into the role of Asl/Cep152 beyond centriole duplication could help shed light on how Cep152 mutations lead to the development of microcephaly.


Subject(s)
Centrioles/metabolism , Centrioles/physiology , Drosophila Proteins/metabolism , Spermatozoa/growth & development , Spermatozoa/metabolism , Animals , Axoneme/metabolism , Axoneme/physiology , Basal Bodies/metabolism , Basal Bodies/physiology , Cell Cycle Proteins/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Male , Mitosis/physiology , Spermatozoa/physiology
11.
J Cell Biol ; 211(5): 987-98, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26620907

ABSTRACT

The interaction between centrosomes and mitotic spindle poles is important for efficient spindle formation, orientation, and cell polarity. However, our understanding of the dynamics of this relationship and implications for tissue homeostasis remains poorly understood. Here we report that Drosophila melanogaster calmodulin (CaM) regulates the ability of the microcephaly-associated protein, abnormal spindle (Asp), to cross-link spindle microtubules. Both proteins colocalize on spindles and move toward spindle poles, suggesting that they form a complex. Our binding and structure-function analysis support this hypothesis. Disruption of the Asp-CaM interaction alone leads to unfocused spindle poles and centrosome detachment. This behavior leads to randomly inherited centrosomes after neuroblast division. We further show that spindle polarity is maintained in neuroblasts despite centrosome detachment, with the poles remaining stably associated with the cell cortex. Finally, we provide evidence that CaM is required for Asp's spindle function; however, it is completely dispensable for Asp's role in microcephaly suppression.


Subject(s)
Calmodulin/metabolism , Centrosome/metabolism , Drosophila Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/cytology , Animals , Brain/pathology , Cell Division , Cell Line , Cell Polarity , Clustered Regularly Interspaced Short Palindromic Repeats , Drosophila melanogaster/metabolism , Exons , Genotype , Green Fluorescent Proteins/metabolism , Microtubules/metabolism , Mutation , Phenotype , Protein Binding , RNA Interference , Spindle Apparatus/metabolism
12.
J Cell Biol ; 210(1): 79-97, 2015 Jul 06.
Article in English | MEDLINE | ID: mdl-26150390

ABSTRACT

Pericentriolar material (PCM) mediates the microtubule (MT) nucleation and anchoring activity of centrosomes. A scaffold organized by Centrosomin (Cnn) serves to ensure proper PCM architecture and functional changes in centrosome activity with each cell cycle. Here, we investigate the mechanisms that spatially restrict and temporally coordinate centrosome scaffold formation. Focusing on the mitotic-to-interphase transition in Drosophila melanogaster embryos, we show that the elaboration of the interphase Cnn scaffold defines a major structural rearrangement of the centrosome. We identify an unprecedented role for Pericentrin-like protein (PLP), which localizes to the tips of extended Cnn flares, to maintain robust interphase centrosome activity and promote the formation of interphase MT asters required for normal nuclear spacing, centrosome segregation, and compartmentalization of the syncytial embryo. Our data reveal that Cnn and PLP directly interact at two defined sites to coordinate the cell cycle-dependent rearrangement and scaffolding activity of the centrosome to permit normal centrosome organization, cell division, and embryonic viability.


Subject(s)
Centrosome/metabolism , Drosophila Proteins/metabolism , Homeodomain Proteins/metabolism , Interphase , Amino Acid Sequence , Animals , Calmodulin-Binding Proteins , Chromosome Segregation , Drosophila melanogaster , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Genomic Instability , Microtubules/metabolism , Mitosis , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Transport
13.
Mol Biol Cell ; 25(18): 2682-94, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25031429

ABSTRACT

Pericentrin is a critical centrosomal protein required for organizing pericentriolar material (PCM) in mitosis. Mutations in pericentrin cause the human genetic disorder Majewski/microcephalic osteodysplastic primordial dwarfism type II, making a detailed understanding of its regulation extremely important. Germaine to pericentrin's function in organizing PCM is its ability to localize to the centrosome through the conserved C-terminal PACT domain. Here we use Drosophila pericentrin-like-protein (PLP) to understand how the PACT domain is regulated. We show that the interaction of PLP with calmodulin (CaM) at two highly conserved CaM-binding sites in the PACT domain controls the proper targeting of PLP to the centrosome. Disrupting the PLP-CaM interaction with single point mutations renders PLP inefficient in localizing to centrioles in cultured S2 cells and Drosophila neuroblasts. Although levels of PCM are unaffected, it is highly disorganized. We also demonstrate that basal body formation in the male testes and the production of functional sperm does not rely on the PLP-CaM interaction, whereas production of functional mechanosensory neurons does.


Subject(s)
Basal Bodies/metabolism , Calmodulin/metabolism , Drosophila Proteins/metabolism , Neurons/physiology , Spermatozoa/physiology , Amino Acid Sequence , Animals , Calmodulin-Binding Proteins , Cell Line , Centrioles/physiology , Drosophila melanogaster , Male , Mechanotransduction, Cellular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Transport , Sperm Motility , Spermatogenesis
14.
Mol Biol Cell ; 23(7): 1254-66, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22337772

ABSTRACT

Kinesin-5 is an essential mitotic motor. However, how its spatial-temporal distribution is regulated in mitosis remains poorly understood. We expressed localization and affinity purification-tagged Eg5 from a mouse bacterial artificial chromosome (this construct was called mEg5) and found its distribution to be tightly regulated throughout mitosis. Fluorescence recovery after photobleaching analysis showed rapid Eg5 turnover throughout mitosis, which cannot be accounted for by microtubule turnover. Total internal reflection fluorescence microscopy and high-resolution, single-particle tracking revealed that mEg5 punctae on both astral and midzone microtubules rapidly bind and unbind. mEg5 punctae on midzone microtubules moved transiently both toward and away from spindle poles. In contrast, mEg5 punctae on astral microtubules moved transiently toward microtubule minus ends during early mitosis but switched to plus end-directed motion during anaphase. These observations explain the poleward accumulation of Eg5 in early mitosis and its redistribution in anaphase. Inhibition of dynein blocked mEg5 movement on astral microtubules, whereas depletion of the Eg5-binding protein TPX2 resulted in plus end-directed mEg5 movement. However, motion of Eg5 on midzone microtubules was not altered. Our results reveal differential and precise spatial and temporal regulation of Eg5 in the spindle mediated by dynein and TPX2.


Subject(s)
Cell Cycle Proteins/metabolism , Dyneins/metabolism , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis/physiology , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism , Animals , Base Sequence , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Fluorescence Recovery After Photobleaching , Kinesins/genetics , LLC-PK1 Cells , Mice , Microscopy, Fluorescence , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Swine
15.
Mol Biol Cell ; 21(6): 979-88, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20110350

ABSTRACT

TPX2 is a Ran-regulated spindle assembly factor that is required for kinetochore fiber formation and activation of the mitotic kinase Aurora A. TPX2 is enriched near spindle poles and is required near kinetochores, suggesting that it undergoes dynamic relocalization throughout mitosis. Using photoactivation, we measured the movement of PA-GFP-TPX2 in the mitotic spindle. TPX2 moves poleward in the half-spindle and is static in the interzone and near spindle poles. Poleward transport of TPX2 is sensitive to inhibition of dynein or Eg5 and to suppression of microtubule flux with nocodazole or antibodies to Kif2a. Poleward transport requires the C terminus of TPX2, a domain that interacts with Eg5. Overexpression of TPX2 lacking this domain induced excessive microtubule formation near kinetochores, defects in spindle assembly and blocked mitotic progression. Our data support a model in which poleward transport of TPX2 down-regulates its microtubule nucleating activity near kinetochores and links microtubules generated at kinetochores to dynein for incorporation into the spindle.


Subject(s)
Cell Cycle Proteins/metabolism , Dyneins/metabolism , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line , Dynactin Complex , Kinesins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/ultrastructure , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spindle Apparatus/ultrastructure , Swine
16.
Curr Biol ; 19(21): 1833-8, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19836236

ABSTRACT

Mitotic spindle assembly requires the combined activity of various molecular motor proteins, including Eg5 and dynein. Together, these motors generate antagonistic forces during mammalian bipolar spindle assembly; what remains unknown, however, is how these motors are functionally coordinated such that antagonism is possible. Given that Eg5 generates an outward force by crosslinking and sliding apart antiparallel microtubules (MTs), we explored the possibility that dynein generates an inward force by likewise sliding antiparallel MTs. We reasoned that antiparallel overlap, and therefore the magnitude of a dynein-mediated force, would be inversely proportional to the initial distance between centrosomes. To capitalize on this relationship, we utilized a nocodazole washout assay to mimic spindle assembly. We found that Eg5 inhibition led to either monopolar or bipolar spindle formation, depending on whether centrosomes were initially separated by less than or greater than 5.5 microm, respectively. Mathematical modeling predicted this same spindle bistability in the absence of functional Eg5 and required dynein acting on antiparallel MTs to do so. Our results suggest that dynein functionally coordinates with Eg5 by crosslinking and sliding antiparallel MTs, a novel role for dynein within the framework of spindle assembly.


Subject(s)
Dyneins/physiology , Kinesins/antagonists & inhibitors , Microtubules/metabolism , Animals , Cell Line , Cell Polarity , Centrosome/metabolism , Dyneins/metabolism , Microtubules/ultrastructure , Models, Biological , Nocodazole/pharmacology , Spindle Apparatus/metabolism , Swine
17.
J Cell Sci ; 120(Pt 16): 2819-27, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17652157

ABSTRACT

The mitotic spindle of vascular plants is assembled and maintained by processes that remain poorly explored at a molecular level. Here, we report that AtKRP125c, one of four kinesin-5 motor proteins in arabidopsis, decorates microtubules throughout the cell cycle and appears to function in both interphase and mitosis. In a temperature-sensitive mutant, interphase cortical microtubules are disorganized at the restrictive temperature and mitotic spindles are massively disrupted, consistent with a defect in the stabilization of anti-parallel microtubules in the spindle midzone, as previously described in kinesin-5 mutants from animals and yeast. AtKRP125c introduced into mammalian epithelial cells by transfection decorates microtubules throughout the cell cycle but is unable to complement the loss of the endogenous kinesin-5 motor (Eg5). These results are among the first reports of any motor with a major role in anastral spindle structure in plants and demonstrate that the conservation of kinesin-5 motor function throughout eukaryotes extends to vascular plants.


Subject(s)
Arabidopsis/cytology , Arabidopsis/metabolism , Kinesins/metabolism , Mitosis , Animals , Arabidopsis/genetics , Base Sequence , Cell Survival , Chromosome Mapping , Cytokinesis , Genes, Plant , Interphase , Microscopy, Confocal , Microtubules/metabolism , Molecular Sequence Data , Mutation/genetics , Plant Roots/cytology , Prophase , Protein Transport , Spindle Apparatus/metabolism , Swine
18.
Curr Biol ; 16(5): 536-41, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16527751

ABSTRACT

In centrosome-containing cells, microtubules nucleated at centrosomes are thought to play a major role in spindle assembly. In addition, microtubule formation at kinetochores has also been observed, most recently under physiological conditions in live cells. The relative contributions of microtubule formation at kinetochores and centrosomes to spindle assembly, and their molecular requirements, remain incompletely understood. Using mammalian cells released from nocodazole-induced disassembly, we observed microtubule formation at centrosomes and at Bub1-positive sites on chromosomes. Kinetochore-associated microtubules rapidly coalesced into pole-like structures in a dynein-dependent manner. Microinjection of excess importin-beta or depletion of the Ran-dependent spindle assembly factor, TPX2, blocked kinetochore-associated microtubule formation, enhanced centrosome-associated microtubule formation, but did not prevent chromosome capture by centrosomal microtubules. Depletion of the chromosome passenger protein, survivin, reduced microtubule formation at kinetochores in an MCAK-dependent manner. Microtubule formation in cells depleted of Bub1 or Nuf2 was indistinguishable from that in controls. Our data demonstrate that microtubule assembly at centrosomes and kinetochores is kinetically distinct and differentially regulated. The presence of microtubules at kinetochores provides a mechanism to reconcile the time required for spindle assembly in vivo with that observed in computer simulations of search and capture.


Subject(s)
Kinetochores/metabolism , Microtubules/metabolism , Animals , Cell Cycle Proteins/physiology , Cells, Cultured , Dyneins/physiology , Kinetochores/ultrastructure , Microtubule-Associated Proteins/physiology , Microtubules/ultrastructure , Models, Biological , Nuclear Proteins/physiology , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Tubulin/metabolism , ran GTP-Binding Protein/physiology
20.
J Cell Sci ; 118(Pt 18): 4113-22, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16118246

ABSTRACT

Mammalian cells develop a polarized morphology and migrate directionally into a wound in a monolayer culture. To understand how microtubules contribute to these processes, we used GFP-tubulin to measure dynamic instability and GFP-EB1, a protein that marks microtubule plus-ends, to measure microtubule growth events at the centrosome and cell periphery. Growth events at the centrosome, or nucleation, do not show directional bias, but are equivalent toward and away from the wound. Cells with two centrosomes nucleated approximately twice as many microtubules/minute as cells with one centrosome. The average number of growing microtubules per microm2 at the cell periphery is similar for leading and trailing edges and for cells containing one or two centrosomes. In contrast to microtubule growth, measurement of the parameters of microtubule dynamic instability demonstrate that microtubules in the trailing edge are more dynamic than those in the leading edge. Inhibition of Rho with C3 transferase had no detectable effect on microtubule dynamics in the leading edge, but stimulated microtubule turnover in the trailing edge. Our data demonstrate that in wound-edge cells, microtubule nucleation is non-polarized, in contrast to microtubule dynamic instability, which is highly polarized, and that factors in addition to Rho contribute to microtubule stabilization.


Subject(s)
Microtubules/physiology , Wound Healing/physiology , Animals , CHO Cells , Cell Growth Processes/physiology , Cell Polarity , Cells, Cultured , Centrosome/physiology , Centrosome/ultrastructure , Cricetinae , Cytoskeleton/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Swine , Transfection , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...