44 research outputs found

    Analysis of a nuclear complex of proteins that form a spindle matrix during mitosis in Drosophila

    Get PDF
    A spindle matrix is a macromolecular structure hypothesized to provide a stationary support or strut for the anchorage of motor proteins during force generation, microtubule sliding, and chromosome segregation in mitosis. Previous work in Drosophila has shown Skeletor, as the first putative molecular candidate of spindle matrix structure since it possesses many features consistent with the proposed spindle matrix structure.;We have identified two more putative spindle matrix candidate proteins, Chromator and Megator, using yeast two-hybrid interaction assay, cross-immunoprecipitation and immunocytochemistry. Chromator is a novel and essential chromodomain containing protein that interacts directly with Skeletor and extensively colocalizes with it throughout the cell cycle. During interphase Chromator colocalizes with Skeletor on the chromosomes, and then redistributes on to the Skeletor defined spindle during metaphase and anaphase. Furthermore, RNAi depletion of Chromator in S2 cells leads to abnormal microtubule spindle morphology and to chromosome segregation defects, thus suggesting that Chromator plays a role in proper spindle dynamics during mitosis. We have generated new Chromator mutant alleles to further aid in the functional analysis of its roles in force production and chromosome segregation during mitosis.;Megator (Bx34 antigen) is a Drosophila Tpr ortholog and has an extended N-terminal coiled-coil domain. During interphase Megator is localized to the nuclear rim and occupies the intranuclear space surrounding the chromosomes. However, during mitosis Megator reorganizes and aligns together with Skeletor and Chromator into a fusiform spindle structure. The Megator metaphase spindle persists in the absence of microtubule spindles, thus strongly implying that the existence of the Megator-defined spindle does not require polymerized microtubules. Furthermore, deletion construct analysis in S2 cells revealed that the NH2-terminal coiled-coil region of Megator self assembles into large spheres indicating its ability to form polymers and serve as the structural basis for the putative spindle matrix complex

    A nuclear-derived proteinaceous matrix embeds the microtubule spindle apparatus during mitosis

    Get PDF
    The concept of a spindle matrix has long been proposed. Whether such a structure exists, however, and what its molecular and structural composition are have remained controversial. In this study, using a live-imaging approach in Drosophilasyncytial embryos, we demonstrate that nuclear proteins reorganize during mitosis to form a highly dynamic, viscous spindle matrix that embeds the microtubule spindle apparatus, stretching from pole to pole. We show that this “internal” matrix is a distinct structure from the microtubule spindle and from a lamin B–containing spindle envelope. By injection of 2000-kDa dextran, we show that the disassembling nuclear envelope does not present a diffusion barrier. Furthermore, when microtubules are depolymerized with colchicine just before metaphase the spindle matrix contracts and coalesces around the chromosomes, suggesting that microtubules act as “struts” stretching the spindle matrix. In addition, we demonstrate that the spindle matrix protein Megator requires its coiled-coil amino-terminal domain for spindle matrix localization, suggesting that specific interactions between spindle matrix molecules are necessary for them to form a complex confined to the spindle region. The demonstration of an embedding spindle matrix lays the groundwork for a more complete understanding of microtubule dynamics and of the viscoelastic properties of the spindle during cell division

    Digitor/dASCIZ Has Multiple Roles in Drosophila Development

    Get PDF
    In this study we provide evidence that the spindle matrix protein Skeletor in Drosophila interacts with the human ASCIZ (also known as ATMIN and ZNF822) ortholog, Digitor/dASCIZ. This interaction was first detected in a yeast two-hybrid screen and subsequently confirmed by pull-down assays. We also confirm a previously documented function of Digitor/dASCIZ as a regulator of Dynein light chain/Cut up expression. Using transgenic expression of a mCitrine-labeled Digitor construct, we show that Digitor/dASCIZ is a nuclear protein that is localized to interband and developmental puff chromosomal regions during interphase but redistributes to the spindle region during mitosis. Its mitotic localization and physical interaction with Skeletor suggest the possibility that Digitor/dASCIZ plays a direct role in mitotic progression as a member of the spindle matrix complex. Furthermore, we have characterized a P-element insertion that is likely to be a true null Digitor/dASCIZ allele resulting in complete pupal lethality when homozygous, indicating that Digitor/dASCIZ is an essential gene. Phenotypic analysis of the mutant provided evidence that Digitor/dASCIZ plays critical roles in regulation of metamorphosis and organogenesis as well as in the DNA damage response. In the Digitor/dASCIZ null mutant larvae there was greatly elevated levels of ÎłH2Av, indicating accumulation of DNA double-strand breaks. Furthermore, reduced levels of Digitor/dASCIZ decreased the resistance to paraquat-induced oxidative stress resulting in increased mortality in a stress test paradigm. We show that an early developmental consequence of the absence of Digitor/dASCIZ is reduced third instar larval brain size although overall larval development appeared otherwise normal at this stage. While Digitor/dASCIZ mutant larvae initiate pupation, all mutant pupae failed to eclose and exhibited various defects in metamorphosis such as impaired differentiation, incomplete disc eversion, and faulty apoptosis. Altogether we provide evidence that Digitor/dASCIZ is a nuclear protein that performs multiple roles in Drosophilalarval and pupal development

    The chromodomain protein, Chromator, interacts with JIL-1 kinase and regulates the structure of Drosophila polytene chromosomes

    Get PDF
    In this study we have generated two new hypomorphic Chro alleles and analyzed the consequences of reduced Chromator protein function on polytene chromosome structure. We show that in Chro71/Chro612 mutants the polytene chromosome arms were coiled and compacted with a disruption and misalignment of band and interband regions and with numerous ectopic contacts connecting non-homologous regions. Furthermore, we demonstrate that Chromator co-localizes with the JIL-1 kinase at polytene interband regions and that the two proteins interact within the same protein complex. That both proteins are necessary and may function together is supported by the finding that a concomitant reduction in JIL-1 and Chromator function synergistically reduces viability during development. Overlay assays and deletion construct analysis suggested that the interaction between JIL-1 and Chromator is direct and that it is mediated by sequences in the C-terminal domain of Chromator and by the acidic region within the C-terminal domain of JIL-1. Taken together these findings indicate that Chromator and JIL-1 interact in an interband-specific complex that functions to establish or maintain polytene chromosome structure in Drosophila

    Human Cep192 Is Required for Mitotic Centrosome and Spindle Assembly

    Get PDF
    SummaryAs cells enter mitosis, centrosomes dramatically increase in size and ability to nucleate microtubules. This process, termed centrosome maturation, is driven by the accumulation and activation of γ-tubulin and other proteins that form the pericentriolar material on centrosomes during G2/prophase. Here, we show that the human centrosomal protein, Cep192 (centrosomal protein of 192 kDa), is an essential component of the maturation machinery. Specifically, we have found that siRNA depletion of Cep192 results in a complete loss of functional centrosomes in mitotic but not interphase cells. In mitotic cells lacking Cep192, microtubules become organized around chromosomes but rarely acquire stable bipolar configurations. These cells contain normal numbers of centrioles but cannot assemble γ-tubulin, pericentrin, or other pericentriolar proteins into an organized PCM. Alternatively, overexpression of Cep192 results in the formation of multiple, extracentriolar foci of γ-tubulin and pericentrin. Together, our findings support the hypothesis that Cep192 stimulates the formation of the scaffolding upon which γ-tubulin ring complexes and other proteins involved in microtubule nucleation and spindle assembly become functional during mitosis

    Motor domain phosphorylation and regulation of the Drosophila kinesin 13, KLP10A

    Get PDF
    Microtubule (MT)-destabilizing kinesin 13s perform fundamental roles throughout the cell cycle. In this study, we show that the Drosophila melanogaster kinesin 13, KLP10A, is phosphorylated in vivo at a conserved serine (S573) positioned within the α-helix 5 of the motor domain. In vitro, a phosphomimic KLP10A S573E mutant displays a reduced capacity to depolymerize MTs but normal affinity for the MT lattice. In cells, replacement of endogenous KLP10A with KLP10A S573E dampens MT plus end dynamics throughout the cell cycle, whereas a nonphosphorylatable S573A mutant apparently enhances activity during mitosis. Electron microscopy suggests that KLP10A S573 phosphorylation alters its association with the MT lattice, whereas molecular dynamics simulations reveal how KLP10A phosphorylation can alter the kinesin–MT interface without changing important structural features within the motor’s core. Finally, we identify casein kinase 1α as a possible candidate for KLP10A phosphorylation. We propose a model in which phosphorylation of the KLP10A motor domain provides a regulatory switch controlling the time and place of MT depolymerization

    Drosophila katanin is a microtubule depolymerase that regulates cortical-microtubule plus-end interactions and cell migration

    Get PDF
    Regulation of microtubule dynamics at the cell cortex is important for cell motility, morphogenesis and division. Here we show that the Drosophila Katanin, Dm-Kat60, functions to generate a dynamic cortical-microtubule interface in interphase cells. In S2 cells, Dm-Kat60 concentrates at the interphase cell cortex where it suppresses the polymerization of microtubule plus-ends thereby preventing the formation of aberrantly dense cortical arrays. Dm-Kat60 also localizes to the leading edge migratory D17 cells and negatively regulates multiple parameters of their motility. Finally, in vitro, Dm-Kat60 severs and depolymerizes MTs from their ends. Based on these data, we propose that Dm-Kat60 removes tubulin from microtubule ends or lattice that contact specific cortical sites to preventing stable and/or lateral attachments. The asymmetric distribution of such an activity could help generate regional variations in MT behaviors involved in cell migration

    Analysis of a nuclear complex of proteins that form a spindle matrix during mitosis in Drosophila

    No full text
    A spindle matrix is a macromolecular structure hypothesized to provide a stationary support or strut for the anchorage of motor proteins during force generation, microtubule sliding, and chromosome segregation in mitosis. Previous work in Drosophila has shown Skeletor, as the first putative molecular candidate of spindle matrix structure since it possesses many features consistent with the proposed spindle matrix structure.;We have identified two more putative spindle matrix candidate proteins, Chromator and Megator, using yeast two-hybrid interaction assay, cross-immunoprecipitation and immunocytochemistry. Chromator is a novel and essential chromodomain containing protein that interacts directly with Skeletor and extensively colocalizes with it throughout the cell cycle. During interphase Chromator colocalizes with Skeletor on the chromosomes, and then redistributes on to the Skeletor defined spindle during metaphase and anaphase. Furthermore, RNAi depletion of Chromator in S2 cells leads to abnormal microtubule spindle morphology and to chromosome segregation defects, thus suggesting that Chromator plays a role in proper spindle dynamics during mitosis. We have generated new Chromator mutant alleles to further aid in the functional analysis of its roles in force production and chromosome segregation during mitosis.;Megator (Bx34 antigen) is a Drosophila Tpr ortholog and has an extended N-terminal coiled-coil domain. During interphase Megator is localized to the nuclear rim and occupies the intranuclear space surrounding the chromosomes. However, during mitosis Megator reorganizes and aligns together with Skeletor and Chromator into a fusiform spindle structure. The Megator metaphase spindle persists in the absence of microtubule spindles, thus strongly implying that the existence of the Megator-defined spindle does not require polymerized microtubules. Furthermore, deletion construct analysis in S2 cells revealed that the NH2-terminal coiled-coil region of Megator self assembles into large spheres indicating its ability to form polymers and serve as the structural basis for the putative spindle matrix complex.</p
    corecore