Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 660
Filter
1.
EMBO J ; 42(17): e109738, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37401899

ABSTRACT

The centrosome linker joins the two interphase centrosomes of a cell into one microtubule organizing center. Despite increasing knowledge on linker components, linker diversity in different cell types and their role in cells with supernumerary centrosomes remained unexplored. Here, we identified Ninein as a C-Nap1-anchored centrosome linker component that provides linker function in RPE1 cells while in HCT116 and U2OS cells, Ninein and Rootletin link centrosomes together. In interphase, overamplified centrosomes use the linker for centrosome clustering, where Rootletin gains centrosome linker function in RPE1 cells. Surprisingly, in cells with centrosome overamplification, C-Nap1 loss prolongs metaphase through persistent activation of the spindle assembly checkpoint indicated by BUB1 and MAD1 accumulation at kinetochores. In cells lacking C-Nap1, the reduction of microtubule nucleation at centrosomes and the delay in nuclear envelop rupture in prophase probably cause mitotic defects like multipolar spindle formation and chromosome mis-segregation. These defects are enhanced when the kinesin HSET, which normally clusters multiple centrosomes in mitosis, is partially inhibited indicating a functional interplay between C-Nap1 and centrosome clustering in mitosis.


Subject(s)
Cell Cycle Proteins , Centrosome , Centrosome/metabolism , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Interphase/physiology , Mitosis , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
2.
Protein Sci ; 31(6): e4324, 2022 06.
Article in English | MEDLINE | ID: mdl-35634770

ABSTRACT

Proper protein destruction by the ubiquitin (Ub)-proteasome system is vital for a faithful cell cycle. Hence, the activity of Ub ligases is tightly controlled. The Anaphase-Promoting Complex/Cyclosome (APC/C) is a 1.2 MDa Ub ligase responsible for mitotic progression and G1 maintenance. At the G1/S transition, the APC/C is inhibited by EMI1 to prevent APC/C-dependent polyubiquitination of cell cycle effectors. EMI1 uses several interaction motifs to block the recruitment of APC/C substrates as well as the APC/C-associated E2s, UBE2C, and UBE2S. Paradoxically, EMI1 is also an APC/C substrate during G1. Using a comprehensive set of enzyme assays, we determined the context-dependent involvement of the EMI1 motifs in APC/C-dependent ubiquitination of EMI1 and other substrates. Furthermore, we demonstrated that an isolated C-terminal peptide fragment of EMI1 activates APC/C-dependent substrate priming by UBE2C. Together, these findings reveal the multiple roles of the EMI1 C-terminus for G1 maintenance and the G1/S transition.


Subject(s)
F-Box Proteins , Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Interphase/physiology , Ubiquitin/metabolism
3.
Dev Cell ; 57(4): 466-479.e6, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35231427

ABSTRACT

The cytoplasm is a crowded, visco-elastic environment whose physical properties change according to physiological or developmental states. How the physical properties of the cytoplasm impact cellular functions in vivo remains poorly understood. Here, we probe the effects of cytoplasmic concentration on microtubules by applying osmotic shifts to fission yeast, moss, and mammalian cells. We show that the rates of both microtubule polymerization and depolymerization scale linearly and inversely with cytoplasmic concentration; an increase in cytoplasmic concentration decreases the rates of microtubule polymerization and depolymerization proportionally, whereas a decrease in cytoplasmic concentration leads to the opposite. Numerous lines of evidence indicate that these effects are due to changes in cytoplasmic viscosity rather than cellular stress responses or macromolecular crowding per se. We reconstituted these effects on microtubules in vitro by tuning viscosity. Our findings indicate that, even in normal conditions, the viscosity of the cytoplasm modulates the reactions that underlie microtubule dynamic behaviors.


Subject(s)
Cytoplasm/metabolism , Microtubules/metabolism , Polymerization , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Cell Nucleus/metabolism , Interphase/physiology , Spindle Apparatus/metabolism
4.
Dev Biol ; 482: 101-113, 2022 02.
Article in English | MEDLINE | ID: mdl-34906546

ABSTRACT

During metazoan early embryogenesis, the intracellular properties of proteins and organelles change dynamically through rapid cleavage. In particular, a change in the nucleus size is known to contribute to embryonic development-dependent cell cycle and gene expression regulation. Here, we compared the nuclear sizes of various blastomeres from developing Xenopus embryos and analyzed the mechanisms that control the nuclear expansion dynamics by manipulating the amount of intracellular components in a cell-free system. Nuclear expansion was slower in blastomeres from vegetal hemispheres during a longer interphase than in those from animal hemispheres. Furthermore, upon recapitulating interphase events by manipulating the concentration of yolk platelets, which are originally rich in the vegetal blastomeres, in cell-free cytoplasmic extracts, nuclear expansion and DNA replication became slower than that in normal yolk-free conditions. Under these conditions, the supplemented yolk platelets accumulated around the nucleus in a microtubule-dependent manner and impeded the organization of the endoplasmic reticulum network. Overall, we propose that yolk platelets around the nucleus reduce membrane supply from the endoplasmic reticulum to the nucleus, resulting in slower nuclear expansion and cell cycle progression in the yolk-rich vegetal blastomeres.


Subject(s)
Blastomeres/physiology , Cell Membrane/physiology , Cell Nucleus/physiology , Endoplasmic Reticulum/physiology , Xenopus laevis/embryology , Animals , Cell Size , Embryo, Nonmammalian/cytology , Embryonic Development/physiology , Interphase/physiology
5.
Elife ; 102021 12 01.
Article in English | MEDLINE | ID: mdl-34850681

ABSTRACT

Dramatic change in chromosomal DNA morphology between interphase and mitosis is a defining features of the eukaryotic cell cycle. Two types of enzymes, namely cohesin and condensin confer the topology of chromosomal DNA by extruding DNA loops. While condensin normally configures chromosomes exclusively during mitosis, cohesin does so during interphase. The processivity of cohesin's loop extrusion during interphase is limited by a regulatory factor called WAPL, which induces cohesin to dissociate from chromosomes via a mechanism that requires dissociation of its kleisin from the neck of SMC3. We show here that a related mechanism may be responsible for blocking condensin II from acting during interphase. Cells derived from patients affected by microcephaly caused by mutations in the MCPH1 gene undergo premature chromosome condensation. We show that deletion of Mcph1 in mouse embryonic stem cells unleashes an activity of condensin II that triggers formation of compact chromosomes in G1 and G2 phases, accompanied by enhanced mixing of A and B chromatin compartments, and this occurs even in the absence of CDK1 activity. Crucially, inhibition of condensin II by MCPH1 depends on the binding of a short linear motif within MCPH1 to condensin II's NCAPG2 subunit. MCPH1's ability to block condensin II's association with chromatin is abrogated by the fusion of SMC2 with NCAPH2, hence may work by a mechanism similar to cohesin. Remarkably, in the absence of both WAPL and MCPH1, cohesin and condensin II transform chromosomal DNAs of G2 cells into chromosomes with a solenoidal axis.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Embryonic Stem Cells/drug effects , Interphase/genetics , Interphase/physiology , Animals , Gene Expression Regulation , Metabolic Networks and Pathways , Mice
6.
Sci Rep ; 11(1): 19278, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34588507

ABSTRACT

The cell nucleus is a tightly regulated organelle and its architectural structure is dynamically orchestrated to maintain normal cell function. Indeed, fluctuations in nuclear size and shape are known to occur during the cell cycle and alterations in nuclear morphology are also hallmarks of many diseases including cancer. Regrettably, automated reliable tools for cell cycle staging at single cell level using in situ images are still limited. It is therefore urgent to establish accurate strategies combining bioimaging with high-content image analysis for a bona fide classification. In this study we developed a supervised machine learning method for interphase cell cycle staging of individual adherent cells using in situ fluorescence images of nuclei stained with DAPI. A Support Vector Machine (SVM) classifier operated over normalized nuclear features using more than 3500 DAPI stained nuclei. Molecular ground truth labels were obtained by automatic image processing using fluorescent ubiquitination-based cell cycle indicator (Fucci) technology. An average F1-Score of 87.7% was achieved with this framework. Furthermore, the method was validated on distinct cell types reaching recall values higher than 89%. Our method is a robust approach to identify cells in G1 or S/G2 at the individual level, with implications in research and clinical applications.


Subject(s)
Cell Nucleus/physiology , Image Processing, Computer-Assisted , Interphase/physiology , Single-Cell Analysis/methods , Support Vector Machine , Animals , Cell Line , Datasets as Topic , Humans , Intravital Microscopy/methods , Mice , Microscopy, Fluorescence/methods
7.
Biomolecules ; 11(7)2021 07 20.
Article in English | MEDLINE | ID: mdl-34356684

ABSTRACT

The maintenance of genome integrity in the cell is an essential process for the accurate transmission of the genetic material. BRCA2 participates in this process at several levels, including DNA repair by homologous recombination, protection of stalled replication forks, and cell division. These activities are regulated and coordinated via cell-cycle dependent modifications. Pathogenic variants in BRCA2 cause genome instability and are associated with breast and/or ovarian cancers. BRCA2 is a very large protein of 3418 amino acids. Most well-characterized variants causing a strong predisposition to cancer are mutated in the C-terminal 700 residues DNA binding domain of BRCA2. The rest of the BRCA2 protein is predicted to be disordered. Interactions involving intrinsically disordered regions (IDRs) remain difficult to identify both using bioinformatics tools and performing experimental assays. However, the lack of well-structured binding sites provides unique functional opportunities for BRCA2 to bind to a large set of partners in a tightly regulated manner. We here summarize the predictive and experimental arguments that support the presence of disorder in BRCA2. We describe how BRCA2 IDRs mediate self-assembly and binding to partners during DNA double-strand break repair, mitosis, and meiosis. We highlight how phosphorylation by DNA repair and cell-cycle kinases regulate these interactions. We finally discuss the impact of cancer-associated variants on the function of BRCA2 IDRs and more generally on genome stability and cancer risk.


Subject(s)
BRCA2 Protein/chemistry , BRCA2 Protein/metabolism , DNA Repair/physiology , BRCA2 Protein/genetics , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , Female , Humans , Interphase/physiology , Magnetic Resonance Spectroscopy , Mitosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
8.
ACS Chem Biol ; 16(10): 1917-1923, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34161081

ABSTRACT

Mammalian cell cycle is a central process for tissue growth and maintenance. Protein O-linked ß-N-acetylglucosamine (O-GlcNAc) modification has been found to occur on several important cell cycle regulators. However, the O-GlcNAcylated proteome has not been extensively profiled during cell cycle progression. Herein, we report a quantitative profiling of protein O-GlcNAcylation sites in cell proliferation, by using an O-GlcNAc chemoproteomic strategy. In HeLa cells, a total of 902, 439, and 872 high-confidence O-GlcNAcylation sites distributed on 414, 265, and 425 proteins are identified in the interphase, early mitosis, and mitotic exit stages, respectively. The identified O-GlcNAcylation events occur on a variety of important regulators, which are involved in the processes of cell division, DNA repair, and cell death. Furthermore, we show that O-GlcNAcylation is dynamically regulated in a cell cycle stage-dependent manner. Our results provide a valuable resource for investigating the functional roles of O-GlcNAc in the mammalian cell cycle.


Subject(s)
Acetylglucosamine/analysis , Cell Cycle/physiology , Glycoproteins/analysis , Glycoproteins/metabolism , Proteome/analysis , Proteome/metabolism , Anaphase/physiology , Glycoproteins/chemistry , Glycosylation , HeLa Cells , Humans , Interphase/physiology , Protein Processing, Post-Translational , Proteome/chemistry , Proteomics
9.
Ann Clin Lab Sci ; 51(1): 61-72, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33653782

ABSTRACT

OBJECTIVE: Conventional karyotyping of multiple myeloma (MM) is hampered by the low mitotic index of plasma cells (PCs), and low proportion of PCs in some specimens may lead to false negative results in fluorescence in situ hybridisation (FISH) detection. METHODS: Bone marrow cells were cultured in an ordinary medium for 24 h or in a medium containing 10 ng/mL IL-6 and 40 ng/mL GM-CSF for 6 d. Fluorescence immunophenotyping and interphase cytogenetics as a tool for the investigation of neoplasms (FICTION) was also conducted, combining CD138 fluorescent immunophenotype and FISH. RESULTS: Under modified culture conditions, the successful rate of culture and abnormality detection rate during karyotype analysis increased to 86.4% and 40.9%, respectively. The abnormality detection rate of FICTION (89.5%) was significantly higher than that of FISH (60.0%). The genetic abnormality detection rate increased to 92.3% when FICTION and karyotyping were conducted under modified culture conditions. CONCLUSION: The established modified culture system could improve karyotyping quality in MM. Due to its obvious advantages compared with FISH, FICTION is recommended for detecting genetic abnormalities in MM.


Subject(s)
Immunophenotyping/methods , Karyotyping/methods , Multiple Myeloma/diagnosis , Adult , Aged , Aged, 80 and over , Bone Marrow Cells/pathology , China , Chromosome Aberrations , Cytogenetic Analysis/methods , Cytogenetics/methods , Female , Humans , In Situ Hybridization, Fluorescence/methods , Interphase/physiology , Male , Middle Aged , Multiple Myeloma/immunology , Plasma Cells/pathology
10.
Mol Biol Cell ; 32(9): 869-879, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33439671

ABSTRACT

Microtubule plus-end depolymerization rate is a potentially important target of physiological regulation, but it has been challenging to measure, so its role in spatial organization is poorly understood. Here we apply a method for tracking plus ends based on time difference imaging to measure depolymerization rates in large interphase asters growing in Xenopus egg extract. We observed strong spatial regulation of depolymerization rates, which were higher in the aster interior compared with the periphery, and much less regulation of polymerization or catastrophe rates. We interpret these data in terms of a limiting component model, where aster growth results in lower levels of soluble tubulin and microtubule-associated proteins (MAPs) in the interior cytosol compared with that at the periphery. The steady-state polymer fraction of tubulin was ∼30%, so tubulin is not strongly depleted in the aster interior. We propose that the limiting component for microtubule assembly is a MAP that inhibits depolymerization, and that egg asters are tuned to low microtubule density.


Subject(s)
Actin Depolymerizing Factors/metabolism , Microtubules/physiology , Actin Depolymerizing Factors/physiology , Actins/metabolism , Animals , Interphase/physiology , Kinesins/metabolism , Microscopy, Fluorescence/methods , Microtubules/metabolism , Spatio-Temporal Analysis , Spindle Apparatus/metabolism , Tubulin/metabolism , Xenopus laevis
11.
Mol Biol Cell ; 31(24): 2646-2656, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32966175

ABSTRACT

The centriole duplication cycle normally ensures that centriole number is maintained at two centrioles per G1 cell. However, some circumstances can result in an aberrant increase in centriole number-a phenotype that is particularly prevalent in several types of cancer. Following an artificial increase in centriole number without tetraploidization due to transient overexpression of the kinase PLK4, human cells return to a normal centriole number during the proliferation of the population. We examine the mechanisms responsible for this return to normal centriole number at the population level in human retinal pigment epithelial cells. We find that the return to normal centriole number in the population of induced cells cannot be explained by limited duplication of centrioles, instability of extra centrioles, or by grossly asymmetric segregation of extra centrioles in mitosis. However, cells with extra centrioles display heterogenous phenotypes including extended cell cycle arrest, longer interphase durations, and death, which overall results in a proliferative disadvantage relative to normal cells in the population. Although about half of cells with extra centrioles in a population were able to divide, the extent of the disadvantages conferred by other fates is sufficient to account for the observed rate of return to normal centriole number. These results suggest that only under conditions of positive selection for cells with extra centrioles, continuous generation of such centrioles, or alleviation of the disadvantageous growth phenotypes would they be maintained in a population.


Subject(s)
Centrioles/metabolism , Centrioles/physiology , Protein Serine-Threonine Kinases/metabolism , Cell Cycle/physiology , Cell Cycle Checkpoints/physiology , Cell Cycle Proteins/metabolism , Cell Line , Cell Proliferation/physiology , Centrosome/metabolism , Homeostasis , Humans , Interphase/physiology , Mitosis , Protein Serine-Threonine Kinases/physiology , Retinal Pigment Epithelium/metabolism
12.
J Cell Biol ; 219(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32342107

ABSTRACT

Nuclear envelope herniations (blebs) containing FG-nucleoporins and ubiquitin are the phenotypic hallmark of Torsin ATPase manipulation. Both the dynamics of blebbing and the connection to nuclear pore biogenesis remain poorly understood. We employ a proteomics-based approach to identify myeloid leukemia factor 2 (MLF2) as a luminal component of the bleb. Using an MLF2-based live-cell imaging platform, we demonstrate that nuclear envelope blebbing occurs rapidly and synchronously immediately after nuclear envelope reformation during mitosis. Bleb formation is independent of ubiquitin conjugation within the bleb, but strictly dependent on POM121, a transmembrane nucleoporin essential for interphase nuclear pore biogenesis. Nup358, a late marker for interphase nuclear pore complex (NPC) biogenesis, is underrepresented relative to FG-nucleoporins in nuclear envelopes of Torsin-deficient cells. The kinetics of bleb formation, its dependence on POM121, and a reduction of mature NPCs in Torsin-deficient cells lead us to conclude that the hallmark phenotype of Torsin manipulation represents aberrant NPC intermediates.


Subject(s)
Molecular Chaperones/metabolism , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Nuclear Proteins/metabolism , Gene Knockout Techniques , HeLa Cells , Humans , Interphase/genetics , Interphase/physiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Microscopy, Electron, Transmission , Mitosis/genetics , Mitosis/physiology , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Nuclear Envelope/genetics , Nuclear Envelope/ultrastructure , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , RNA, Small Interfering , Ubiquitin/chemistry , Ubiquitin/metabolism
13.
J Cell Biol ; 219(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32348467

ABSTRACT

Primary cilia play critical roles in development and disease. Their assembly and disassembly are tightly coupled to cell cycle progression. Here, we present data identifying KIF14 as a regulator of cilia formation and Hedgehog (HH) signaling. We show that RNAi depletion of KIF14 specifically leads to defects in ciliogenesis and basal body (BB) biogenesis, as its absence hampers the efficiency of primary cilium formation and the dynamics of primary cilium elongation, and disrupts the localization of the distal appendage proteins SCLT1 and FBF1 and components of the IFT-B complex. We identify deregulated Aurora A activity as a mechanism contributing to the primary cilium and BB formation defects seen after KIF14 depletion. In addition, we show that primary cilia in KIF14-depleted cells are defective in response to HH pathway activation, independently of the effects of Aurora A. In sum, our data point to KIF14 as a critical node connecting cell cycle machinery, effective ciliogenesis, and HH signaling.


Subject(s)
Aurora Kinase A/metabolism , Cell Cycle/genetics , Cilia/metabolism , Hedgehog Proteins/metabolism , Kinesins/metabolism , Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Basal Bodies/metabolism , Chromatography, Liquid , Cilia/genetics , Cilia/pathology , HEK293 Cells , Humans , Interphase/physiology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kinesins/genetics , Mitosis/genetics , Oncogene Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Signal Transduction/genetics , Sodium Channels/metabolism , Tandem Mass Spectrometry
14.
Cell Rep ; 31(3): 107533, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32320646

ABSTRACT

The cohesin- and condensin-related SMC5/6 complex has largely been studied in the context of DNA repair. Nevertheless, SMC5/6 has an undefined essential function even in the absence of cellular stress. Through the use of an auxin-inducible degradation system for rapidly depleting subunits of the SMC5/6 complex, we show that SMC5/6 is essential for viability in cancer-derived and normal human cells. Impairment of SMC5/6 function is associated with spontaneous induction of DNA damage, p53 activation, cell-cycle arrest, and senescence, as well as an increased frequency of various mitotic chromosome segregation abnormalities. However, we show that this chromosome missegregation is apparent only when SMC5/6 function is impaired during the preceding S and G2 phases. In contrast, degradation of SMC5/6 immediately prior to mitotic entry has little or no impact on the fidelity of chromosome segregation, highlighting the importance of the complex during interphase in order to ensure faithful sister chromatid disjunction.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cell Cycle Proteins/genetics , Cell Line , Cell Proliferation/physiology , Cell Survival/physiology , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Genomic Instability , HCT116 Cells , Humans , Interphase/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Dev Cell ; 52(2): 210-222.e7, 2020 01 27.
Article in English | MEDLINE | ID: mdl-31928973

ABSTRACT

Most metazoan cells entering mitosis undergo characteristic rounding, which is important for accurate spindle positioning and chromosome separation. Rounding is driven by contractile tension generated by myosin motors in the sub-membranous actin cortex. Recent studies highlight that alongside myosin activity, cortical actin organization is a key regulator of cortex tension. Yet, how mitotic actin organization is controlled remains poorly understood. To address this, we characterized the F-actin interactome in spread interphase and round mitotic cells. Using super-resolution microscopy, we then screened for regulators of cortex architecture and identified the intermediate filament vimentin and the actin-vimentin linker plectin as unexpected candidates. We found that vimentin is recruited to the mitotic cortex in a plectin-dependent manner. We then showed that cortical vimentin controls actin network organization and mechanics in mitosis and is required for successful cell division in confinement. Together, our study highlights crucial interactions between cytoskeletal networks during cell division.


Subject(s)
Actin Cytoskeleton/physiology , Actins/metabolism , Cell Physiological Phenomena , Intermediate Filaments/physiology , Interphase/physiology , Mitosis , Vimentin/metabolism , Chromosome Segregation , HeLa Cells , Humans
16.
Biochem Biophys Res Commun ; 523(2): 293-298, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31864705

ABSTRACT

During mitosis, transcription is ceased, chromatin becomes condensed, many chromatin features are lost, and most transcription factors (TFs) are excluded from chromosomes. The mechanism on how daughter cells maintain cell identity after exiting mitosis remains unclear. A subset of multiple lineage-specific and general TFs remains bound to mitotic chromosomes during mitosis, thereby suggesting a potential mechanism termed mitotic bookmarking. Here, genome-wide binding analysis of TF ZNF143 in human A549 lung epithelial cells reveals that ZNF143 remains partially associated with its interphase-specific genomic regions during mitosis. Genome distribution analysis shows that 80% of these regions preferentially localize to promoters. In addition, ZNF143 in mitosis may could recruit other relative TFs when the cells re-enter into G1 phase and rapidly initiates gene transcription. These results suggest that the dynamic binding of ZNF143 during cell cycle has a potential mitotic bookmarking role in maintaining cell fate and identity.


Subject(s)
Trans-Activators/metabolism , A549 Cells , Binding Sites/genetics , Chromatin Immunoprecipitation Sequencing , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Humans , Interphase/genetics , Interphase/physiology , Mitosis/genetics , Mitosis/physiology , Molecular Sequence Annotation , Promoter Regions, Genetic , Protein Binding , Trans-Activators/chemistry , Trans-Activators/genetics
17.
Biochem J ; 476(15): 2141-2156, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31383821

ABSTRACT

The spatial configuration of chromatin is fundamental to ensure any given cell can fulfil its functional duties, from gene expression to specialised cellular division. Significant technological innovations have facilitated further insights into the structure, function and regulation of three-dimensional chromatin organisation. To date, the vast majority of investigations into chromatin organisation have been conducted in interphase and mitotic cells leaving meiotic chromatin relatively unexplored. In combination, cytological and genome-wide contact frequency analyses in mammalian germ cells have recently demonstrated that large-scale chromatin structures in meiotic prophase I are reminiscent of the sequential loop arrays found in mitotic cells, although interphase-like segmentation of transcriptionally active and inactive regions are also evident along the length of chromosomes. Here, we discuss the similarities and differences of such large-scale chromatin architecture, between interphase, mitotic and meiotic cells, as well as their functional relevance and the proposed modulatory mechanisms which underlie them.


Subject(s)
Chromatin/metabolism , Germ Cells/metabolism , Interphase/physiology , Meiosis/physiology , Mitosis/physiology , Animals , Germ Cells/cytology , Humans
18.
Cells ; 8(5)2019 05 17.
Article in English | MEDLINE | ID: mdl-31108912

ABSTRACT

Cytokinesis D is known as the midwife mechanism in which neighboring cells facilitate cell division by crossing the cleavage furrow of dividing cells. Cytokinesis D is thought to be mediated by chemotaxis, where midwife cells migrate toward dividing cells by sensing an unknown chemoattractant secreted from the cleavage furrow. In this study, to validate this chemotaxis model, we aspirated the fluid from the vicinity of the cleavage furrow of a dividing Dictyostelium cell and discharged it onto a neighboring cell using a microcapillary. However, the neighboring cells did not show any chemotaxis toward the fluid. In addition, the cells did not manifest an increase in the levels of intracellular Ca2+, cAMP, or cGMP, which are expected to rise in chemotaxing cells. From several lines of our experiments, including these findings, we concluded that chemotaxis does not contribute to cytokinesis D. As an alternative, we propose a cortical-flow model, where a migrating cell attaches to a dividing cell by chance and is guided toward the furrow by the cortical flow on the dividing cell, and then physically assists the separation of the daughter cells.


Subject(s)
Chemotaxis/physiology , Cytokinesis/physiology , Dictyostelium/cytology , Dictyostelium/physiology , Actin Cytoskeleton/metabolism , Actins/metabolism , Calcium/metabolism , Cell Movement , Cell Tracking/methods , Cells, Cultured , Chemotactic Factors/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cytoplasm/metabolism , Green Fluorescent Proteins/metabolism , Interphase/physiology , Microscopy, Phase-Contrast , Mitosis/physiology , Models, Biological , Pleckstrin Homology Domains/physiology
19.
BMC Mol Cell Biol ; 20(1): 1, 2019 01 24.
Article in English | MEDLINE | ID: mdl-31041892

ABSTRACT

BACKGROUND: The conserved NDR-family kinase Sid2p localizes to the contractile ring during fission yeast cytokinesis to promote ring constriction, septation, and completion of cell division. Previous studies have found that the Type 2 interphase node proteins Blt1p and Gef2p contribute to localization of Sid2p and its regulatory protein Mob1p at the division site. However, their relative contributions and whether they operate in the same or parallel pathways has been unclear. In this study, we quantify the respective roles of Blt1p and Gef2p in Sid2p/Mob1p recruitment and characterize the effect of single and double deletion mutants on contractile ring dynamics and completion of cell division. RESULTS: Using quantitative confocal fluorescence microscopy, we measured Sid2p and Mob1p recruitment to the division site in blt1∆, gef2∆, and blt1∆/gef2∆ mutant cells. We observed an equivalent decrease in Sid2p/Mob1p localization for both single and double mutants. Though assembly of the contractile ring is normal in these mutants, the reduction in Sid2p/Mob1p at the division site delayed the onset of contractile ring constriction and completion of division. We quantified localization of Blt1p and Gef2p at the medial cortex throughout the cell cycle and found that Blt1p localization to interphase nodes and the contractile ring is independent of Gef2p. However, Gef2p localization to the contractile ring is decreased in blt1∆ mutants. CONCLUSIONS: Blt1p and Gef2p work in the same pathway, rather than in parallel, to localize the NDR-family kinase Sid2p and its regulatory partner Mob1p to the division site, thereby promoting timely completion of cell division. Future studies are necessary to understand how additional fission yeast cytokinesis proteins work with these Type 2 interphase node components to promote Sid2p/Mob1p recruitment.


Subject(s)
Cell Cycle Proteins/metabolism , Cytokinesis/physiology , Guanine Nucleotide Exchange Factors/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/physiology , Cell Cycle Proteins/genetics , Cell Survival/genetics , Guanine Nucleotide Exchange Factors/genetics , Interphase/physiology , Microscopy, Fluorescence , Mitosis/physiology , Mutation , Phosphoproteins/metabolism , Protein Kinases/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces pombe Proteins/genetics , Signal Transduction
20.
J Mol Cell Biol ; 11(11): 944-955, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31087092

ABSTRACT

Microtubules grow not only from the centrosome but also from various noncentrosomal microtubule-organizing centers (MTOCs), including the nuclear envelope (NE) and pre-existing microtubules. The evolutionarily conserved proteins Mto1/CDK5RAP2 and Alp14/TOG/XMAP215 have been shown to be involved in promoting microtubule nucleation. However, it has remained elusive as to how the microtubule nucleation promoting factors are specified to various noncentrosomal MTOCs, particularly the NE, and how these proteins coordinate to organize microtubule assembly. Here, we demonstrate that in the fission yeast Schizosaccharomyces pombe, efficient interphase microtubule growth from the NE requires Alp7/TACC, Alp14/TOG/XMAP215, and Mto1/CDK5RAP2. The absence of Alp7, Alp14, or Mto1 compromises microtubule regrowth on the NE in cells undergoing microtubule repolymerization. We further demonstrate that Alp7 and Mto1 interdependently localize to the NE in cells without microtubules and that Alp14 localizes to the NE in an Alp7 and Mto1-dependent manner. Tethering Mto1 to the NE in cells lacking Alp7 partially restores microtubule number and the efficiency of microtubule generation from the NE. Hence, our study delineates that Alp7, Alp14, and Mto1 work in concert to regulate interphase microtubule regrowth on the NE.


Subject(s)
Carrier Proteins/metabolism , Interphase/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nuclear Envelope/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/physiology , Models, Biological , Protein Binding , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...