Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3880, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719804

ABSTRACT

Correlative evidence has suggested that the methyl-CpG-binding protein MeCP2 contributes to the formation of heterochromatin condensates via liquid-liquid phase separation. This interpretation has been reinforced by the observation that heterochromatin, DNA methylation and MeCP2 co-localise within prominent foci in mouse cells. The findings presented here revise this view. MeCP2 localisation is independent of heterochromatin as MeCP2 foci persist even when heterochromatin organisation is disrupted. Additionally, MeCP2 foci fail to show hallmarks of phase separation in live cells. Importantly, we find that mouse cellular models are highly atypical as MeCP2 distribution is diffuse in most mammalian species, including humans. Notably, MeCP2 foci are absent in Mus spretus which is a mouse subspecies lacking methylated satellite DNA repeats. We conclude that MeCP2 has no intrinsic tendency to form condensates and its localisation is independent of heterochromatin. Instead, the distribution of MeCP2 in the nucleus is primarily determined by global DNA methylation patterns.


Subject(s)
DNA Methylation , Heterochromatin , Methyl-CpG-Binding Protein 2 , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Heterochromatin/metabolism , Animals , Mice , Humans , Cell Nucleus/metabolism , Protein Binding , DNA/metabolism , DNA, Satellite/metabolism , DNA, Satellite/genetics , Phase Separation
2.
EMBO J ; 42(21): e113647, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37592895

ABSTRACT

During mitosis, spindle architecture alters as chromosomes segregate into daughter cells. The microtubule crosslinker protein regulator of cytokinesis 1 (PRC1) is essential for spindle stability, chromosome segregation and completion of cytokinesis, but how it recruits motors to the central spindle to coordinate the segregation of chromosomes is unknown. Here, we combine structural and cell biology approaches to show that the human CENP-E motor, which is essential for chromosome capture and alignment by microtubules, binds to PRC1 through a conserved hydrophobic motif. This binding mechanism is also used by Kinesin-4 Kif4A:PRC1. Using in vitro reconstitution, we demonstrate that CENP-E slides antiparallel PRC1-crosslinked microtubules. We find that the regulation of CENP-E -PRC1 interaction is spatially and temporally coupled with relocalization to overlapping microtubules in anaphase. Finally, we demonstrate that the PRC1-microtubule motor interaction is essential in anaphase to control chromosome partitioning, retain central spindle integrity and ensure cytokinesis. Taken together our findings reveal the molecular basis for the cell cycle regulation of motor-PRC1 complexes to couple chromosome segregation and cytokinesis.


Subject(s)
Cytokinesis , Kinesins , Humans , Cytokinesis/physiology , Kinesins/genetics , Kinesins/metabolism , Phosphorylation , Spindle Apparatus/metabolism , Mitosis , Cell Cycle Proteins/metabolism , Microtubules/metabolism
3.
J Cell Sci ; 136(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-35502670

ABSTRACT

The precise regulation of microtubule length during mitosis is essential to assemble and position the mitotic spindle and segregate chromosomes. The kinesin-13 Kif2C or MCAK acts as a potent microtubule depolymerase that diffuses short distances on microtubules, whereas the kinesin-8 Kif18b is a processive motor with weak depolymerase activity. However, the individual activities of these factors cannot explain the dramatic increase in microtubule dynamics in mitosis. Using in vitro reconstitution and single-molecule imaging, we demonstrate that Kif18b, MCAK and the plus-end tracking protein EB3 (also known as MAPRE3) act in an integrated manner to potently promote microtubule depolymerization at very low concentrations. We find that Kif18b can transport EB3 and MCAK and promotes their accumulation to microtubule plus ends through multivalent weak interactions. Together, our work defines the mechanistic basis for a cooperative Kif18b-MCAK-EB network at microtubule plus ends, that acts to efficiently shorten and regulate microtubules in mitosis, essential for correct chromosome segregation.


Subject(s)
Kinesins , Microtubules , Chromosome Segregation , Kinesins/genetics , Microtubules/metabolism , Mitosis , Spindle Apparatus/metabolism
4.
J Cell Biol ; 221(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35776132

ABSTRACT

Centromere association of the chromosomal passenger complex (CPC; Borealin-Survivin-INCENP-Aurora B) and Sgo1 is crucial for chromosome biorientation, a process essential for error-free chromosome segregation. Phosphorylated histone H3 Thr3 (H3T3ph; directly recognized by Survivin) and histone H2A Thr120 (H2AT120ph; indirectly recognized via Sgo1), together with CPC's intrinsic nucleosome-binding ability, facilitate CPC centromere recruitment. However, the molecular basis for CPC-Sgo1 binding and how their physical interaction influences CPC centromere localization are lacking. Here, using an integrative structure-function approach, we show that the "histone H3-like" Sgo1 N-terminal tail-Survivin BIR domain interaction acts as a hotspot essential for CPC-Sgo1 assembly, while downstream Sgo1 residues and Borealin contribute for high-affinity binding. Disrupting Sgo1-Survivin interaction abolished CPC-Sgo1 assembly and perturbed CPC centromere localization and function. Our findings reveal that Sgo1 and H3T3ph use the same surface on Survivin to bind CPC. Hence, it is likely that these interactions take place in a spatiotemporally restricted manner, providing a rationale for the Sgo1-mediated "kinetochore-proximal" CPC centromere pool.


Subject(s)
Cell Cycle Proteins , Centromere , Histones , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centromere/metabolism , Histones/genetics , Histones/metabolism , Kinetochores/metabolism , Phosphorylation , Survivin/genetics , Survivin/metabolism
5.
Curr Biol ; 32(14): 3121-3136.e6, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35830853

ABSTRACT

The establishment of centromere-specific CENP-A chromatin is influenced by epigenetic and genetic processes. Central domain sequences from fission yeast centromeres are preferred substrates for CENP-ACnp1 incorporation, but their use is context dependent, requiring adjacent heterochromatin. CENP-ACnp1 overexpression bypasses heterochromatin dependency, suggesting that heterochromatin ensures exposure to conditions or locations permissive for CENP-ACnp1 assembly. Centromeres cluster around spindle-pole bodies (SPBs). We show that heterochromatin-bearing minichromosomes localize close to SPBs, consistent with this location promoting CENP-ACnp1 incorporation. We demonstrate that heterochromatin-independent de novo CENP-ACnp1 chromatin assembly occurs when central domain DNA is placed near, but not far from, endogenous centromeres or neocentromeres. Moreover, direct tethering of central domain DNA at SPBs permits CENP-ACnp1 assembly, suggesting that the nuclear compartment surrounding SPBs is permissive for CENP-ACnp1 incorporation because target sequences are exposed to high levels of CENP-ACnp1 and associated assembly factors. Thus, nuclear spatial organization is a key epigenetic factor that influences centromere identity.


Subject(s)
Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Centromere/metabolism , Centromere Protein A/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
6.
J Cell Sci ; 132(4)2019 01 14.
Article in English | MEDLINE | ID: mdl-30578316

ABSTRACT

Kinesin-13 motors regulate precise microtubule dynamics and limit microtubule length throughout metazoans by depolymerizing microtubule ends. Recently, the kinesin-13 motor family member MCAK (also known Kif2C) has been proposed to undergo large conformational changes during its catalytic cycle, as it switches from being in solution to being bound to microtubules. Here, we reveal that MCAK has a compact conformation in solution through crosslinking and electron microscopy experiments. When MCAK is bound to the microtubule ends, it adopts an extended conformation with the N-terminus and neck region of MCAK interacting with the microtubule. Interestingly, the region of MCAK that interacts with the microtubule is the region phosphorylated by Aurora B and contains an end binding (EB) protein-binding motif. The level of phosphorylation of the N-terminus results in a graded microtubule depolymerase activity. Here, we show that the N-terminus of MCAK forms a platform to integrate Aurora B kinase downstream signals and in response fine-tunes its depolymerase activity during mitosis. We propose that this allosteric control mechanism allows decoupling of the N-terminus from the motor domain of MCAK to allow MCAK depolymerase activity at kinetochores.


Subject(s)
Aurora Kinase B/chemistry , Kinesins/chemistry , Kinetochores/metabolism , Microtubules/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinesins/genetics , Kinesins/metabolism , Kinetochores/ultrastructure , Microtubules/ultrastructure , Mitosis , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera
7.
Mol Biol Cell ; 29(13): 1743-1752, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29771628

ABSTRACT

Kif15 is a kinesin-12 that contributes critically to bipolar spindle assembly in humans. Here we use force-ramp experiments in an optical trap to probe the mechanics of single Kif15 molecules under hindering or assisting loads and in a variety of nucleotide states. While unloaded Kif15 is established to be highly processive, we find that under hindering loads, Kif15 takes <∼10 steps. As hindering load is increased, Kif15 forestep:backstep ratio decreases exponentially, with stall occurring at 6 pN. In contrast, under assisting loads, Kif15 detaches readily and rapidly, even from its AMPPNP state. Kif15 mechanics thus depend markedly on the loading direction. Kif15 interacts with a binding partner, Tpx2, and we show that Tpx2 locks Kif15 to microtubules under both hindering and assisting loads. Overall, our data predict that Kif15 in the central spindle will act as a mechanical ratchet, supporting spindle extension but resisting spindle compression.


Subject(s)
Kinesins/metabolism , Animals , Antibodies/metabolism , Biomechanical Phenomena , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Models, Biological , Protein Binding , Protein Multimerization
8.
J Cell Biol ; 217(7): 2403-2416, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29661912

ABSTRACT

Mitotic spindle positioning specifies the plane of cell division during anaphase. Spindle orientation and positioning are therefore critical to ensure symmetric division in mitosis and asymmetric division during development. The control of astral microtubule length plays an essential role in positioning the spindle. In this study, using gene knockout, we show that the kinesin-8 Kif18b controls microtubule length to center the mitotic spindle at metaphase. Using in vitro reconstitution, we reveal that Kif18b is a highly processive plus end-directed motor that uses a C-terminal nonmotor microtubule-binding region to accumulate at growing microtubule plus ends. This region is regulated by phosphorylation to spatially control Kif18b accumulation at plus ends and is essential for Kif18b-dependent spindle positioning and regulation of microtubule length. Finally, we demonstrate that Kif18b shortens microtubules by increasing the catastrophe rate of dynamic microtubules. Overall, our work reveals that Kif18b uses its motile properties to reach microtubule ends, where it regulates astral microtubule length to ensure spindle centering.


Subject(s)
Kinesins/genetics , Microtubule-Associated Proteins/genetics , Mitosis/genetics , Anaphase/genetics , CRISPR-Cas Systems/genetics , Cell Division/genetics , HeLa Cells , Humans , Microtubules/genetics , Microtubules/metabolism , Protein Binding/genetics , Spindle Apparatus/genetics
9.
J Cell Biol ; 216(4): 855-857, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28320823

ABSTRACT

Dynein removes the checkpoint proteins from kinetochores once chromosomes are bioriented. In this issue, Gama et al. (2017. J. Cell Biol. https://doi.org/10.1083/jcb.201610108) and Mosalaganti et al. (2017. J. Cell Biol. https://doi.org/10.1083/jcb.201611060) reveal the molecular basis for how dynein and its adaptor protein Spindly are recruited to the ROD-Zw10-Zwilch complex in the fibrous corona of unattached kinetochores.


Subject(s)
Dyneins/metabolism , Kinetochores/metabolism , Cell Cycle Proteins/metabolism , Chromosomes/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism
10.
Elife ; 3: e01724, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24668168

ABSTRACT

Kinesin-12 motors are a little studied branch of the kinesin superfamily with the human protein (Kif15) implicated in spindle mechanics and chromosome movement. In this study, we reconstitute full-length hKif15 and its microtubule-targeting factor hTpx2 in vitro to gain insight into the motors mode of operation. We reveal that hKif15 is a plus-end-directed processive homotetramer that can step against loads of up to 3.5 pN. We further show that hKif15 is the first kinesin that effectively switches microtubule tracks at intersections, enabling it to navigate microtubule networks, such as the spindle. hKif15 tetramers are also capable of cross-linking microtubules, but unexpectedly, this does not depend on hTpx2. Instead, we find that hTpx2 inhibits hKif15 stepping when microtubule-bound. Our data reveal that hKif15 is a second tetrameric spindle motor in addition to the kinesin-5 Eg5 and provides insight into the mechanisms by which hKif15 and its inhibitor hTpx2 modulate spindle microtubule architecture. DOI: http://dx.doi.org/10.7554/eLife.01724.001.


Subject(s)
Kinesins/metabolism , Protein Multimerization , Cell Cycle Proteins/metabolism , Cell Line , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nuclear Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...