Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
PLoS One ; 11(11): e0166829, 2016.
Article in English | MEDLINE | ID: mdl-27861562

ABSTRACT

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 γH2Av, 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 Drosophila larval and pupal development.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , DNA Damage , Drosophila/growth & development , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Gene Expression , Genotype , Heat-Shock Response , Humans , Interphase , Metamorphosis, Biological , Mitosis , Mutation , Nuclear Matrix-Associated Proteins/chemistry , Nuclear Matrix-Associated Proteins/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Signal Transduction , Stress, Physiological , Transcription Factors/chemistry , Transcription, Genetic
2.
PLoS One ; 9(7): e103855, 2014.
Article in English | MEDLINE | ID: mdl-25072297

ABSTRACT

The chromodomain protein, Chromator, is localized to chromosomes during interphase; however, during cell division together with other nuclear proteins Chromator redistributes to form a macro molecular spindle matrix complex that embeds the microtubule spindle apparatus. It has been demonstrated that the CTD of Chromator is sufficient for localization to the spindle matrix and that expression of this domain alone could partially rescue Chro mutant microtubule spindle defects. Furthermore, the presence of frayed and unstable microtubule spindles during mitosis after Chromator RNAi depletion in S2 cells indicated that Chromator may interact with microtubules. In this study using a variety of biochemical assays we have tested this hypothesis and show that Chromator not only has binding activity to microtubules with a Kd of 0.23 µM but also to free tubulin. Furthermore, we have mapped the interaction with microtubules to a relatively small stretch of 139 amino acids in the carboxy-terminal region of Chromator. This sequence is likely to contain a novel microtubule binding interface since database searches did not find any sequence matches with known microtubule binding motifs.


Subject(s)
Drosophila Proteins/metabolism , Microtubules/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Tubulin/metabolism , Animals , Animals, Genetically Modified/metabolism , Antibodies/immunology , Drosophila Proteins/chemistry , Drosophila Proteins/immunology , Drosophila melanogaster/metabolism , Immunoprecipitation , Microtubules/chemistry , Nuclear Matrix-Associated Proteins/chemistry , Nuclear Matrix-Associated Proteins/immunology , Protein Binding , Protein Structure, Tertiary , Tubulin/chemistry , Tubulin/immunology
3.
Mol Biol Cell ; 23(18): 3532-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22855526

ABSTRACT

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 Drosophila syncytial 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.


Subject(s)
Drosophila Proteins/metabolism , Microtubules/metabolism , Mitosis , Nuclear Matrix-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Animals , Animals, Genetically Modified , Chromosomes, Insect/metabolism , Dextrans/chemistry , Dextrans/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lamin Type B/genetics , Lamin Type B/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Matrix-Associated Proteins/genetics , Time-Lapse Imaging , Tubulin/genetics , Tubulin/metabolism , Red Fluorescent Protein
4.
Cell Cycle ; 11(12): 2359-66, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22672901

ABSTRACT

Fidgetin is a member of the AAA protein superfamily with important roles in mammalian development. Here we show that human Fidgetin is a potent microtubule severing and depolymerizing the enzyme used to regulate mitotic spindle architecture, dynamics and anaphase A. In vitro, recombinant human Fidgetin severs taxol-stabilized microtubules along their length and promotes depolymerization, primarily from their minus-ends. In cells, human Fidgetin targets to centrosomes, and its depletion with siRNA significantly reduces the velocity of poleward tubulin flux and anaphase A chromatid-to-pole motion. In addition, the loss of Fidgetin induces a microtubule-dependent enlargement of mitotic centrosomes and an increase in the number and length of astral microtubules. Based on these data, we propose that human Fidgetin actively suppresses microtubule growth from and attachment to centrosomes.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Adenosine Triphosphatases/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitosis , ATPases Associated with Diverse Cellular Activities/antagonists & inhibitors , ATPases Associated with Diverse Cellular Activities/genetics , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Anaphase , Cell Line, Tumor , Centrosome/metabolism , Humans , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism
5.
Chromosome Res ; 19(3): 423-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21461696

ABSTRACT

The mechanisms that move chromatids poleward during anaphase A have fascinated researchers for decades. There is now growing evidence that this movement is tightly linked to the active depolymerization of both ends of kinetochore-associated microtubules, a mechanism we refer to as "Pacman-Flux." Contemporary data suggest that this is catalyzed by the integration of multiple enzymatic activities including (1) microtubule-end depolymerases housed at the pole or kinetochore, (2) microtubule-severing enzymes used to uncap the ends of kinetochore-associated microtubules, and (3) molecular motors which drive tubulins towards the pole or into kinetochores.


Subject(s)
Anaphase/physiology , Anaphase/genetics , Animals , Chromatids/metabolism , Humans , Kinetochores/metabolism , Microtubules/metabolism
6.
Nat Cell Biol ; 13(4): 361-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21378981

ABSTRACT

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. Dm-Kat60 concentrates at the cell cortex of S2 Drosophila cells during interphase, where it suppresses the polymerization of microtubule plus-ends, thereby preventing the formation of aberrantly dense cortical arrays. Dm-Kat60 also localizes at the leading edge of migratory D17 Drosophila cells and negatively regulates multiple parameters of their motility. Finally, in vitro, Dm-Kat60 severs and depolymerizes microtubules from their ends. On the basis of these data, we propose that Dm-Kat60 removes tubulin from microtubule lattice or microtubule ends that contact specific cortical sites to prevent stable and/or lateral attachments. The asymmetric distribution of such an activity could help generate regional variations in microtubule behaviours involved in cell migration.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Movement/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Microtubules/metabolism , Adenosine Triphosphatases/genetics , Animals , Cell Cycle/physiology , Cell Line , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Cytoskeleton/metabolism , Drosophila Proteins/genetics , Humans , Katanin , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/ultrastructure , RNA Interference , Tubulin/metabolism
7.
Cell Cycle ; 9(12): 2403-11, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20519956

ABSTRACT

Katanin is a microtubule severing enzyme with demonstrated roles in a variety of cellular activities including mitosis. Here we identify the closely related, but relatively uncharacterized human protein, Katanin-like 1 (KL1), as a novel mitotic regulator. Over expression of KL1 in tissue culture cells results in the complete disassembly of cellular microtubules strongly suggesting that it is an active microtubule severing protein. During mitosis, the localization of KL1 is restricted to spindle poles and is notably absent from centrosomes. This is in clear contrast to conventional Katanin whose localization extends from centrosomes onto poles. Consistent with its localization, siRNA depletion of KL1 from U2OS cells results in a specific and significant reduction in the density of microtubules at spindle poles and significantly increases spindle length. Depletion of KL1 also alters the distribution of gamma-tubulin at centrosomes/spindle poles. Despite its impact on spindle morphology, we could find no evidence that KL1 influences anaphase chromosome motility. Based on our findings, we propose that KL1-mediated microtubule severing is utilized to generate microtubule seeds within the poles and that loss of this activity alters the normal balance of motor-generated forces that determine spindle length.


Subject(s)
Endopeptidases/metabolism , Microtubules/enzymology , Mitosis , Spindle Apparatus/enzymology , Cell Line , Centrosome , Chromosomes, Human , Endopeptidases/genetics , Fluorescent Antibody Technique , Gene Expression , Humans , Katanin , RNA, Small Interfering , Tubulin/metabolism
8.
Mol Biol Cell ; 20(22): 4696-705, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19793918

ABSTRACT

Chromosome movements are linked to the active depolymerization of spindle microtubule (MT) ends. Here we identify the kinesin-13 family member, KLP59D, as a novel and uniquely important regulator of spindle MT dynamics and chromosome motility in Drosophila somatic cells. During prometaphase and metaphase, depletion of KLP59D, which targets to centrosomes and outer kinetochores, suppresses the depolymerization of spindle pole-associated MT minus ends, thereby inhibiting poleward tubulin Flux. Subsequently, during anaphase, loss of KLP59D strongly attenuates chromatid-to-pole motion by suppressing the depolymerization of both minus and plus ends of kinetochore-associated MTs. The mechanism of KLP59D's impact on spindle MT plus and minus ends appears to differ. Our data support a model in which KLP59D directly depolymerizes kinetochore-associated plus ends during anaphase, but influences minus ends indirectly by localizing the pole-associated MT depolymerase KLP10A. Finally, electron microscopy indicates that, unlike the other Drosophila kinesin-13s, KLP59D is largely incapable of oligomerizing into MT-associated rings in vitro, suggesting that such structures are not a requisite feature of kinetochore-based MT disassembly and chromosome movements.


Subject(s)
Chromosomes/metabolism , Drosophila Proteins/metabolism , Exoribonucleases/metabolism , Kinesins/metabolism , Animals , Cell Cycle/physiology , Chromosomes/ultrastructure , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/physiology , Exoribonucleases/genetics , Kinesins/genetics , Kinetochores/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , RNA Interference , Spindle Apparatus/metabolism
9.
Curr Biol ; 19(19): R899-901, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19825352

ABSTRACT

Kinesin-5s help assemble the bipolar spindle by crosslinking and sliding apart antiparallel microtubules. A recent study has uncovered a novel pathway for the phospho-regulation of these motors.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/physiology , Microtubule-Associated Proteins/metabolism , Mitosis/physiology , Models, Biological , Molecular Motor Proteins/metabolism , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Spindle Apparatus/physiology , Animals , Drosophila/metabolism
10.
J Cell Biol ; 186(4): 481-90, 2009 Aug 24.
Article in English | MEDLINE | ID: mdl-19687256

ABSTRACT

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 alpha-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 1alpha 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.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Kinesins/metabolism , Microtubules/metabolism , Amino Acid Sequence , Animals , Casein Kinase Ialpha/genetics , Casein Kinase Ialpha/metabolism , Cell Line , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Kinesins/chemistry , Kinesins/genetics , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Tubulin/chemistry , Tubulin/genetics , Tubulin/metabolism
11.
Dev Biol ; 334(1): 253-63, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19632217

ABSTRACT

The chromodomain protein, Chromator, has been shown to have multiple functions that include regulation of chromatin structure as well as coordination of muscle remodeling during metamorphosis depending on the developmental context. In this study we show that mitotic neuroblasts from brain squash preparations from larvae heteroallelic for the two Chromator loss-of-function alleles Chro(71) and Chro(612) have severe microtubule spindle and chromosome segregation defects that were associated with a reduction in brain size. The microtubule spindles formed were incomplete, unfocused, and/or without clear spindle poles and at anaphase chromosomes were lagging and scattered. Time-lapse analysis of mitosis in S2 cells depleted of Chromator by RNAi treatment suggested that the lagging and scattered chromosome phenotypes were caused by incomplete alignment of chromosomes at the metaphase plate, possibly due to a defective spindle-assembly checkpoint, as well as of frayed and unstable microtubule spindles during anaphase. Expression of full-length Chromator transgenes under endogenous promoter control restored both microtubule spindle morphology as well as brain size strongly indicating that the observed mutant defects were directly attributable to lack of Chromator function.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/metabolism , Mitosis , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Spindle Apparatus/physiology , Animals , Drosophila/genetics , Immunohistochemistry , Larva/metabolism , Microtubules/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
12.
Curr Biol ; 17(22): 1960-6, 2007 Nov 20.
Article in English | MEDLINE | ID: mdl-17980596

ABSTRACT

As 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 gamma-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 gamma-tubulin, pericentrin, or other pericentriolar proteins into an organized PCM. Alternatively, overexpression of Cep192 results in the formation of multiple, extracentriolar foci of gamma-tubulin and pericentrin. Together, our findings support the hypothesis that Cep192 stimulates the formation of the scaffolding upon which gamma-tubulin ring complexes and other proteins involved in microtubule nucleation and spindle assembly become functional during mitosis.


Subject(s)
Centrosome/metabolism , Chromosomal Proteins, Non-Histone/physiology , Microtubule Proteins/physiology , Mitosis/physiology , Spindle Apparatus/metabolism , Cell Differentiation/physiology , HeLa Cells , Humans , Microtubules/metabolism , Tubulin/physiology
13.
J Cell Sci ; 119(Pt 11): 2332-41, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16723739

ABSTRACT

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 Chro(71)/Chro(612) 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.


Subject(s)
Chromosomes/ultrastructure , Drosophila Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Alleles , Animals , Chromosomes/genetics , Chromosomes/metabolism , Drosophila , Drosophila Proteins/genetics , Mutation , Nuclear Matrix-Associated Proteins/genetics , Protein Serine-Threonine Kinases/genetics
14.
J Cell Biochem ; 95(6): 1284-91, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15962301

ABSTRACT

We have used immunocytochemistry to demonstrate that the EAST protein in Drosophila, which forms an expandable nuclear endoskeleton at interphase, redistributes during mitosis to colocalize with the spindle matrix proteins, Megator and Skeletor. EAST and Megator also colocalize to the intranuclear space surrounding the chromosomes at interphase. EAST is a novel protein that does not have any previously characterized motifs or functional domains. However, we show by immunoprecipitation experiments that EAST is likely to molecularly interact with Megator which has a large NH2-terminal coiled-coil domain with the capacity for self assembly. On the basis of these findings, we propose that Megator and EAST interact to form a nuclear endoskeleton and as well are important components of the putative spindle matrix complex during mitosis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Mitosis , Nuclear Matrix-Associated Proteins/metabolism , Phosphoproteins/metabolism , Spindle Apparatus/metabolism , Animals , Cell Cycle Proteins , Protein Binding , Tubulin/metabolism
15.
J Neurobiol ; 64(2): 157-69, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15818553

ABSTRACT

By database searches of the Drosophila genome project we have identified D-hil as the fly member of a novel family of W180-domain containing proteins. Immunocytochemistry demonstrated that D-hil is localized to the neuropil of the embryonic CNS, to the cellular cortex of dividing neuroblasts from larval brains, and that it is up-regulated in the cleavage furrow of S2 cells. We show that D-hil distribution overlaps extensively with that of the septin family member Pnut. Cross-immunoprecipitation experiments further indicated that the two proteins may be members of the same protein complex. Analysis of a severe hypomorphic P-element mutation in the D-hil locus suggested that D-hil is a nonessential protein. However, by creating double mutant flies we show that the D-hil locus acts as a modulator of Pnut function by increasing the level of polyploidy of neuroblasts in Pnut(KG00478)/Pnut(KG00478) larval brains. Based on these results we propose that D-hil may function as a regulator of septin function during cytokinesis in the developing nervous system.


Subject(s)
Brain/metabolism , Cytokinesis/physiology , Drosophila Proteins/physiology , Microfilament Proteins/metabolism , Nerve Tissue Proteins/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Blotting, Western/methods , Brain/embryology , Cell Line , Drosophila , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Genomic Imprinting , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Immunoprecipitation/methods , Larva , Molecular Sequence Data , Mutagenesis/physiology , Phylogeny , Protein Biosynthesis , Sequence Alignment , Sequence Homology, Amino Acid , Time Factors , Transfection/methods
16.
Mol Biol Cell ; 15(11): 4854-65, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15356261

ABSTRACT

We have used immunocytochemistry and cross-immunoprecipitation analysis to demonstrate that Megator (Bx34 antigen), a Tpr ortholog in Drosophila with an extended coiled-coil domain, colocalizes with the putative spindle matrix proteins Skeletor and Chromator during mitosis. Analysis of P-element mutations in the Megator locus showed that Megator is an essential protein. 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, strongly implying that the existence of the Megator-defined spindle does not require polymerized microtubules. Deletion construct analysis in S2 cells indicates that the COOH-terminal part of Megator without the coiled-coil region was sufficient for both nuclear as well as spindle localization. In contrast, the NH2-terminal coiled-coil region remains in the cytoplasm; however, we show that it is capable of assembling into spherical structures. On the basis of these findings we propose that the COOH-terminal domain of Megator functions as a targeting and localization domain, whereas the NH2-terminal domain is responsible for forming polymers that may serve as a structural basis for the putative spindle matrix complex.


Subject(s)
Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Nuclear Matrix-Associated Proteins/biosynthesis , Nuclear Matrix-Associated Proteins/genetics , Spindle Apparatus , Animals , Cell Cycle , Cell Line , Cell Nucleus/metabolism , Cell Survival , Chromosomal Proteins, Non-Histone/biosynthesis , Cytoplasm/metabolism , Drosophila melanogaster/metabolism , Electrophoresis, Polyacrylamide Gel , Gene Deletion , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Interphase , Microscopy, Fluorescence , Microtubules/chemistry , Mitosis , Models, Genetic , Nocodazole/pharmacology , Protein Structure, Tertiary , RNA Interference , Time Factors , Transfection
17.
J Cell Biochem ; 93(5): 1033-47, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15389869

ABSTRACT

We have used a yeast two-hybrid interaction assay to identify Chromator, a novel chromodomain containing protein that interacts directly with the putative spindle matrix protein Skeletor. Immunocytochemistry demonstrated that Chromator and Skeletor show extensive co-localization throughout the cell cycle. During interphase Chromator is localized on chromosomes to interband chromatin regions in a pattern that overlaps that of Skeletor. However, during mitosis both Chromator and Skeletor detach from the chromosomes and align together in a spindle-like structure. Deletion construct analysis in S2 cells showed that the COOH-terminal half of Chromator without the chromodomain was sufficient for both nuclear as well as spindle localization. Analysis of P-element mutations in the Chromator locus shows that Chromator is an essential protein. Furthermore, RNAi depletion of Chromator in S2 cells leads to abnormal microtubule spindle morphology and to chromosome segregation defects. These findings suggest that Chromator is a nuclear protein that plays a role in proper spindle dynamics during mitosis.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Amino Acid Sequence , Animals , Cell Cycle/physiology , Cell Line , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Molecular Sequence Data , Nuclear Matrix-Associated Proteins/genetics , Protein Binding , RNA Interference , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...