Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 385
Filter
1.
Nat Struct Mol Biol ; 31(6): 861-873, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459128

ABSTRACT

Biorientation of chromosomes during cell division is necessary for precise dispatching of a mother cell's chromosomes into its two daughters. Kinetochores, large layered structures built on specialized chromosome loci named centromeres, promote biorientation by binding and sensing spindle microtubules. One of the outer layer main components is a ten-subunit assembly comprising Knl1C, Mis12C and Ndc80C (KMN) subcomplexes. The KMN is highly elongated and docks on kinetochores and microtubules through interfaces at its opposite extremes. Here, we combine cryogenic electron microscopy reconstructions and AlphaFold2 predictions to generate a model of the human KMN that reveals all intra-KMN interfaces. We identify and functionally validate two interaction interfaces that link Mis12C to Ndc80C and Knl1C. Through targeted interference experiments, we demonstrate that this mutual organization strongly stabilizes the KMN assembly. Our work thus reports a comprehensive structural and functional analysis of this part of the kinetochore microtubule-binding machinery and elucidates the path of connections from the chromatin-bound components to the force-generating components.


Subject(s)
Cryoelectron Microscopy , Kinetochores , Microtubule-Associated Proteins , Models, Molecular , Nuclear Proteins , Humans , Kinetochores/metabolism , Kinetochores/ultrastructure , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/chemistry , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/chemistry , Microtubules/metabolism , Microtubules/ultrastructure , Protein Binding , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/chemistry , HeLa Cells
2.
J Cell Biol ; 222(4)2023 04 03.
Article in English | MEDLINE | ID: mdl-36705601

ABSTRACT

Proper chromosome segregation is crucial for cell division. In eukaryotes, this is achieved by the kinetochore, an evolutionarily conserved multiprotein complex that physically links the DNA to spindle microtubules and takes an active role in monitoring and correcting erroneous spindle-chromosome attachments. Our mechanistic understanding of these functions and how they ensure an error-free outcome of mitosis is still limited, partly because we lack a complete understanding of the kinetochore structure in the cell. In this study, we use single-molecule localization microscopy to visualize individual kinetochore complexes in situ in budding yeast. For major kinetochore proteins, we measured their abundance and position within the metaphase kinetochore. Based on this comprehensive dataset, we propose a quantitative model of the budding yeast kinetochore. While confirming many aspects of previous reports based on bulk imaging, our results present a unifying nanoscale model of the kinetochore in budding yeast.


Subject(s)
Kinetochores , Saccharomyces cerevisiae , Chromosome Segregation , Kinetochores/ultrastructure , Microtubules/genetics , Microtubules/metabolism , Mitosis , Spindle Apparatus/genetics , Saccharomyces cerevisiae/genetics
3.
J Cell Biol ; 222(4)2023 04 03.
Article in English | MEDLINE | ID: mdl-36705602

ABSTRACT

The key to ensuring proper chromosome segregation during mitosis is the kinetochore (KT), a tightly regulated multiprotein complex that links the centromeric chromatin to the spindle microtubules and as such leads the segregation process. Understanding its architecture, function, and regulation is therefore essential. However, due to its complexity and dynamics, only its individual subcomplexes could be studied in structural detail so far. In this study, we construct a nanometer-precise in situ map of the human-like regional KT of Schizosaccharomyces pombe using multi-color single-molecule localization microscopy. We measure each protein of interest (POI) in conjunction with two references, cnp1CENP-A at the centromere and sad1 at the spindle pole. This allows us to determine cell cycle and mitotic plane, and to visualize individual centromere regions separately. We determine protein distances within the complex using Bayesian inference, establish the stoichiometry of each POI and, consequently, build an in situ KT model with unprecedented precision, providing new insights into the architecture.


Subject(s)
Kinetochores , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Bayes Theorem , Centromere/metabolism , Chromosome Segregation , Kinetochores/ultrastructure , Mitosis , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Spindle Apparatus/metabolism
4.
Nat Commun ; 12(1): 5931, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34635673

ABSTRACT

The chromatin remodeler RSF1 enriched at mitotic centromeres is essential for proper chromosome alignment and segregation and underlying mechanisms remain to be disclosed. We here show that PLK1 recruitment by RSF1 at centromeres creates an activating phosphorylation on Thr236 in the activation loop of Aurora B and this is indispensable for the Aurora B activation. In structural modeling the phosphorylated Thr236 enhances the base catalysis by Asp200 nearby, facilitating the Thr232 autophosphorylation. Accordingly, RSF1-PLK1 is central for Aurora B-mediated microtubule destabilization in error correction. However, under full microtubule-kinetochore attachment RSF1-PLK1 positions at kinetochores, halts activating Aurora B and phosphorylates BubR1, regardless of tension. Spatial movement of RSF1-PLK1 to kinetochores is triggered by Aurora B-mediated phosphorylation of centromeric histone H3 on Ser28. We propose a regulatory RSF1-PLK1 axis that spatiotemporally controls on/off switch on Aurora B. This feedback circuit among RSF1-PLK1-Aurora B may coordinate dynamic microtubule-kinetochore attachment in early mitosis when full tension yet to be generated.


Subject(s)
Aurora Kinase B/genetics , Cell Cycle Proteins/genetics , Chromosome Segregation , Mitosis , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , Trans-Activators/genetics , Aspartic Acid/metabolism , Aurora Kinase B/metabolism , Cell Cycle Proteins/metabolism , Chromatin/chemistry , Chromatin/metabolism , Feedback, Physiological , Gene Expression Regulation , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Kinetochores/metabolism , Kinetochores/ultrastructure , Microtubules/metabolism , Microtubules/ultrastructure , Nuclear Proteins/deficiency , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Serine/metabolism , Trans-Activators/deficiency , Polo-Like Kinase 1
5.
Open Biol ; 11(9): 210131, 2021 09.
Article in English | MEDLINE | ID: mdl-34465213

ABSTRACT

Expansion microscopy (ExM) has become a powerful super-resolution method in cell biology. It is a simple, yet robust approach, which does not require any instrumentation or reagents beyond those present in a standard microscopy facility. In this study, we used kinetoplastid parasites Trypanosoma brucei and Leishmania major, which possess a complex, yet well-defined microtubule-based cytoskeleton, to demonstrate that this method recapitulates faithfully morphology of structures as previously revealed by a combination of sophisticated electron microscopy (EM) approaches. Importantly, we also show that due to the rapidness of image acquisition and three-dimensional reconstruction of cellular volumes ExM is capable of complementing EM approaches by providing more quantitative data. This is demonstrated on examples of less well-appreciated microtubule structures, such as the neck microtubule of T. brucei or the pocket, cytosolic and multivesicular tubule-associated microtubules of L. major. We further demonstrate that ExM enables identifying cell types rare in a population, such as cells in mitosis and cytokinesis. Three-dimensional reconstruction of an entire volume of these cells provided details on the morphology of the mitotic spindle and the cleavage furrow. Finally, we show that established antibody markers of major cytoskeletal structures function well in ExM, which together with the ability to visualize proteins tagged with small epitope tags will facilitate studies of the kinetoplastid cytoskeleton.


Subject(s)
Kinetochores/metabolism , Kinetoplastida/metabolism , Leishmania major/metabolism , Microscopy, Electron/methods , Microtubules/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/metabolism , Kinetochores/ultrastructure , Kinetoplastida/ultrastructure , Leishmania major/ultrastructure , Microtubules/ultrastructure , Trypanosoma brucei brucei/ultrastructure
6.
Curr Genet ; 67(4): 511-518, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33745061

ABSTRACT

This review describes the current models for how the subunit abundance of the Ndc80 complex, a key kinetochore component, is regulated in budding yeast and metazoan meiosis. The past decades of kinetochore research have established the Ndc80 complex to be a key microtubule interactor and a central hub for regulating chromosome segregation. Recent studies further demonstrate that Ndc80 is the limiting kinetochore subunit that dictates the timing of kinetochore activation in budding yeast meiosis. Here, we discuss the molecular circuits that regulate Ndc80 protein synthesis and degradation in budding yeast meiosis and compare the findings with those from metazoans. We envision the regulatory principles discovered in budding yeast to be conserved in metazoans, thereby providing guidance into future investigations on kinetochore regulation in human health and disease.


Subject(s)
Chromosome Segregation/genetics , Cytoskeletal Proteins/ultrastructure , Meiosis/genetics , Nuclear Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Cytoskeletal Proteins/genetics , Humans , Kinetochores/ultrastructure , Microtubules/genetics , Nuclear Proteins/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/ultrastructure
7.
EMBO J ; 40(5): e105671, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33463726

ABSTRACT

The CENP-A nucleosome is a key structure for kinetochore assembly. Once the CENP-A nucleosome is established in the centromere, additional proteins recognize the CENP-A nucleosome to form a kinetochore. CENP-C and CENP-N are CENP-A binding proteins. We previously demonstrated that vertebrate CENP-C binding to the CENP-A nucleosome is regulated by CDK1-mediated CENP-C phosphorylation. However, it is still unknown how the phosphorylation of CENP-C regulates its binding to CENP-A. It is also not completely understood how and whether CENP-C and CENP-N act together on the CENP-A nucleosome. Here, using cryo-electron microscopy (cryo-EM) in combination with biochemical approaches, we reveal a stable CENP-A nucleosome-binding mode of CENP-C through unique regions. The chicken CENP-C structure bound to the CENP-A nucleosome is stabilized by an intramolecular link through the phosphorylated CENP-C residue. The stable CENP-A-CENP-C complex excludes CENP-N from the CENP-A nucleosome. These findings provide mechanistic insights into the dynamic kinetochore assembly regulated by CDK1-mediated CENP-C phosphorylation.


Subject(s)
Centromere Protein A/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cryoelectron Microscopy/methods , Kinetochores/metabolism , Nucleosomes/metabolism , Animals , Centromere/ultrastructure , Centromere Protein A/ultrastructure , Chickens , Chromosomal Proteins, Non-Histone/ultrastructure , Kinetochores/ultrastructure , Models, Molecular , Nucleosomes/ultrastructure , Phosphorylation , Protein Conformation
8.
Curr Biol ; 30(24): 4869-4881.e5, 2020 12 21.
Article in English | MEDLINE | ID: mdl-33035484

ABSTRACT

The nanoscale protein architecture of the kinetochore plays an integral role in specifying the mechanisms underlying its functions in chromosome segregation. However, defining this architecture in human cells remains challenging because of the large size and compositional complexity of the kinetochore. Here, we use Förster resonance energy transfer to reveal the architecture of individual kinetochore-microtubule attachments in human cells. We find that the microtubule-binding domains of the Ndc80 complex cluster at the microtubule plus end. This clustering occurs only after microtubule attachment, and it increases proportionally with centromeric tension. Surprisingly, Ndc80 complex clustering is independent of the organization and number of its centromeric receptors. Moreover, this clustering is similar in yeast and human kinetochores despite significant differences in their centromeric organizations. These and other data suggest that the microtubule-binding interface of the human kinetochore behaves like a flexible "lawn" despite being nucleated by repeating biochemical subunits.


Subject(s)
Centromere/metabolism , Chromosome Segregation , Kinetochores/ultrastructure , Microtubules/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/isolation & purification , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/ultrastructure , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/isolation & purification , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/ultrastructure , Fluorescence Resonance Energy Transfer , Gene Knockdown Techniques , Genes, Reporter/genetics , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Kinetochores/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Saccharomyces cerevisiae
9.
PLoS Genet ; 16(7): e1008918, 2020 07.
Article in English | MEDLINE | ID: mdl-32730246

ABSTRACT

Holocentric chromosomes possess multiple kinetochores along their length rather than the single centromere typical of other chromosomes [1]. They have been described for the first time in cytogenetic experiments dating from 1935 and, since this first observation, the term holocentric chromosome has referred to chromosomes that: i. lack the primary constriction corresponding to centromere observed in monocentric chromosomes [2]; ii. possess multiple kinetochores dispersed along the chromosomal axis so that microtubules bind to chromosomes along their entire length and move broadside to the pole from the metaphase plate [3]. These chromosomes are also termed holokinetic, because, during cell division, chromatids move apart in parallel and do not form the classical V-shaped figures typical of monocentric chromosomes [4-6]. Holocentric chromosomes evolved several times during both animal and plant evolution and are currently reported in about eight hundred diverse species, including plants, insects, arachnids and nematodes [7,8]. As a consequence of their diffuse kinetochores, holocentric chromosomes may stabilize chromosomal fragments favouring karyotype rearrangements [9,10]. However, holocentric chromosome may also present limitations to crossing over causing a restriction of the number of chiasma in bivalents [11] and may cause a restructuring of meiotic divisions resulting in an inverted meiosis [12].


Subject(s)
Caenorhabditis elegans/genetics , Chromosomes/genetics , Kinetochores/ultrastructure , Meiosis/genetics , Animals , Caenorhabditis elegans/cytology , Centromere/genetics , Centromere/ultrastructure , Chromatids/genetics , Chromatids/ultrastructure , Chromosome Segregation/genetics , Chromosomes/ultrastructure , Karyotype , Plants/genetics
10.
Nature ; 574(7777): 278-282, 2019 10.
Article in English | MEDLINE | ID: mdl-31578520

ABSTRACT

In eukaryotes, accurate chromosome segregation in mitosis and meiosis maintains genome stability and prevents aneuploidy. Kinetochores are large protein complexes that, by assembling onto specialized Cenp-A nucleosomes1,2, function to connect centromeric chromatin to microtubules of the mitotic spindle3,4. Whereas the centromeres of vertebrate chromosomes comprise millions of DNA base pairs and attach to multiple microtubules, the simple point centromeres of budding yeast are connected to individual microtubules5,6. All 16 budding yeast chromosomes assemble complete kinetochores using a single Cenp-A nucleosome (Cenp-ANuc), each of which is perfectly centred on its cognate centromere7-9. The inner and outer kinetochore modules are responsible for interacting with centromeric chromatin and microtubules, respectively. Here we describe the cryo-electron microscopy structure of the Saccharomyces cerevisiae inner kinetochore module, the constitutive centromere associated network (CCAN) complex, assembled onto a Cenp-A nucleosome (CCAN-Cenp-ANuc). The structure explains the interdependency of the constituent subcomplexes of CCAN and shows how the Y-shaped opening of CCAN accommodates Cenp-ANuc to enable specific CCAN subunits to contact the nucleosomal DNA and histone subunits. Interactions with the unwrapped DNA duplex at the two termini of Cenp-ANuc are mediated predominantly by a DNA-binding groove in the Cenp-L-Cenp-N subcomplex. Disruption of these interactions impairs assembly of CCAN onto Cenp-ANuc. Our data indicate a mechanism of Cenp-A nucleosome recognition by CCAN and how CCAN acts as a platform for assembly of the outer kinetochore to link centromeres to the mitotic spindle for chromosome segregation.


Subject(s)
Centromere Protein A/metabolism , Kinetochores/chemistry , Kinetochores/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nucleosomes/chemistry , Nucleosomes/metabolism , Centromere Protein A/chemistry , Centromere Protein A/ultrastructure , Cryoelectron Microscopy , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , Kinetochores/ultrastructure , Models, Molecular , Multiprotein Complexes/ultrastructure , Nucleosomes/ultrastructure , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure
11.
FEBS Lett ; 593(20): 2889-2907, 2019 10.
Article in English | MEDLINE | ID: mdl-31469407

ABSTRACT

In mitosis, the spindle assembly checkpoint (SAC) monitors the formation of microtubule-kinetochore attachments during capture of chromosomes by the mitotic spindle. Spindle assembly is complete once there are no longer any unattached kinetochores. Here, we will discuss the mechanism and key components of spindle checkpoint signalling. Unattached kinetochores bind the principal spindle checkpoint kinase monopolar spindle 1 (MPS1). MPS1 triggers the recruitment of other spindle checkpoint proteins and the formation of a soluble inhibitor of anaphase, thus preventing exit from mitosis. On microtubule attachment, kinetochores become checkpoint silent due to the actions of PP2A-B56 and PP1. This SAC responsive period has to be coordinated with mitotic spindle formation to ensure timely mitotic exit and accurate chromosome segregation. We focus on the molecular mechanisms by which the SAC permissive state is created, describing a central role for CDK1-cyclin B1 and its counteracting phosphatase PP2A-B55. Furthermore, we discuss how CDK1-cyclin B1, through its interaction with MAD1, acts as an integral component of the SAC, and actively orchestrates checkpoint signalling and thus contributes to the faithful execution of mitosis.


Subject(s)
CDC2 Protein Kinase/genetics , Cyclin B1/genetics , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Microtubules/metabolism , Spindle Apparatus/metabolism , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Segregation , Chromosomes, Human/chemistry , Chromosomes, Human/metabolism , Cyclin B1/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Kinetochores/ultrastructure , Microtubules/ultrastructure , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Signal Transduction , Spindle Apparatus/ultrastructure
12.
J Mol Biol ; 431(22): 4444-4454, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31425683

ABSTRACT

Kinetochores are the multiprotein complexes that link chromosomal centromeres to mitotic-spindle microtubules. Budding yeast centromeres comprise three sequential "centromere-determining elements", CDEI, II, and III. CDEI (8 bp) and CDEIII (∼25 bp) are conserved between Kluyveromyces lactis and Saccharomyces cerevisiae, but CDEII in the former is twice as long (160 bp) as CDEII in the latter (80 bp). The CBF3 complex recognizes CDEIII and is required for assembly of a centromeric nucleosome, which in turn recruits other kinetochore components. To understand differences in centromeric nucleosome assembly between K. lactis and S. cerevisiae, we determined the structure of a K. lactis CBF3 complex by electron cryomicroscopy at ∼4 Å resolution and compared it with published structures of S. cerevisiae CBF3. We show differences in the pose of Ndc10 and discuss potential models of the K. lactis centromeric nucleosome that account for the extended CDEII length.


Subject(s)
Centromere/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Kluyveromyces/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Centromere/ultrastructure , Cryoelectron Microscopy , Kinetochores/metabolism , Kinetochores/ultrastructure , Kluyveromyces/ultrastructure , Saccharomyces cerevisiae/ultrastructure
13.
J Biol Chem ; 294(38): 14119-14134, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31366733

ABSTRACT

The successful assembly and regulation of the kinetochore are critical for the equal and accurate segregation of genetic material during the cell cycle. CENP-C (centromere protein C), a conserved inner kinetochore component, has been broadly characterized as a scaffolding protein and is required for the recruitment of multiple kinetochore proteins to the centromere. At its C terminus, CENP-C harbors a conserved cupin domain that has an established role in protein dimerization. Although the crystal structure of the Saccharomyces cerevisiae Mif2CENP-C cupin domain has been determined, centromeric organization and kinetochore composition vary greatly between S. cerevisiae (point centromere) and other eukaryotes (regional centromere). Therefore, whether the structural and functional role of the cupin domain is conserved throughout evolution requires investigation. Here, we report the crystal structures of the Schizosaccharomyces pombe and Drosophila melanogaster CENP-C cupin domains at 2.52 and 1.81 Å resolutions, respectively. Although the central jelly roll architecture is conserved among the three determined CENP-C cupin domain structures, the cupin domains from organisms with regional centromeres contain additional structural features that aid in dimerization. Moreover, we found that the S. pombe Cnp3CENP-C jelly roll fold harbors an inner binding pocket that is used to recruit the meiosis-specific protein Moa1. In summary, our results unveil the evolutionarily conserved and unique features of the CENP-C cupin domain and uncover the mechanism by which it functions as a recruitment factor.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/ultrastructure , Animals , Cell Cycle Proteins/metabolism , Centromere/metabolism , Centromere Protein A/metabolism , Crystallography, X-Ray/methods , DNA-Binding Proteins/metabolism , Dimerization , Drosophila Proteins/metabolism , Drosophila Proteins/ultrastructure , Drosophila melanogaster/metabolism , Histones/metabolism , Kinetochores/metabolism , Kinetochores/ultrastructure , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism
14.
EMBO Rep ; 20(8): e47905, 2019 08.
Article in English | MEDLINE | ID: mdl-31290587

ABSTRACT

The accuracy of the two sequential meiotic divisions in oocytes is essential for creating a haploid gamete with a normal chromosomal content. Here, we have analysed the 3D dynamics of chromosomes during the second meiotic division in live mouse oocytes. We find that chromosomes form stable kinetochore-microtubule attachments at the end of prometaphase II stage that are retained until anaphase II onset. Remarkably, we observe that more than 20% of the kinetochore-microtubule attachments at the metaphase II stage are merotelic or lateral. However, < 1% of all chromosomes at onset of anaphase II are found to lag at the spindle equator and < 10% of the laggards missegregate and give rise to aneuploid gametes. Our results demonstrate that aberrant kinetochore-microtubule attachments are not corrected at the metaphase stage of the second meiotic division. Thus, the accuracy of the chromosome segregation process in mouse oocytes during meiosis II is ensured by an efficient correction process acting at the anaphase stage.


Subject(s)
Anaphase , Kinetochores/ultrastructure , Metaphase , Microtubules/ultrastructure , Oocytes/ultrastructure , Amino Acid Sequence , Animals , Chromatids/metabolism , Chromatids/ultrastructure , Chromosome Segregation , Female , Humans , Kinetochores/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Oocytes/metabolism , Spermatocytes/metabolism , Spermatocytes/ultrastructure , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Time-Lapse Imaging
15.
Elife ; 82019 06 13.
Article in English | MEDLINE | ID: mdl-31194673

ABSTRACT

Kinetochores are the chromosomal attachment points for spindle microtubules. They are also signaling hubs that control major cell cycle transitions and coordinate chromosome folding. Most well-studied eukaryotes rely on a conserved set of factors, which are divided among two loosely-defined groups, for these functions. Outer kinetochore proteins contact microtubules or regulate this contact directly. Inner kinetochore proteins designate the kinetochore assembly site by recognizing a specialized nucleosome containing the H3 variant Cse4/CENP-A. We previously determined the structure, resolved by cryo-electron microscopy (cryo-EM), of the yeast Ctf19 complex (Ctf19c, homologous to the vertebrate CCAN), providing a high-resolution view of inner kinetochore architecture (Hinshaw and Harrison, 2019). We now extend these observations by reporting a near-atomic model of the Ctf3 complex, the outermost Ctf19c sub-assembly seen in our original cryo-EM density. The model is sufficiently well-determined by the new data to enable molecular interpretation of Ctf3 recruitment and function.


Subject(s)
Kinetochores/chemistry , Kinetochores/metabolism , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Kinetochores/ultrastructure , Models, Molecular , Protein Binding , Saccharomyces cerevisiae Proteins/ultrastructure
16.
J Cell Biol ; 218(2): 455-473, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30504246

ABSTRACT

In dividing cells, depolymerizing spindle microtubules move chromosomes by pulling at their kinetochores. While kinetochore subcomplexes have been studied extensively in vitro, little is known about their in vivo structure and interactions with microtubules or their response to spindle damage. Here we combine electron cryotomography of serial cryosections with genetic and pharmacological perturbation to study the yeast chromosome segregation machinery in vivo. Each kinetochore microtubule has one (rarely, two) Dam1C/DASH outer kinetochore assemblies. Dam1C/DASH contacts the microtubule walls and does so with its flexible "bridges"; there are no contacts with the protofilaments' curved tips. In metaphase, ∼40% of the Dam1C/DASH assemblies are complete rings; the rest are partial rings. Ring completeness and binding position along the microtubule are sensitive to kinetochore attachment and tension, respectively. Our study and those of others support a model in which each kinetochore must undergo cycles of conformational change to couple microtubule depolymerization to chromosome movement.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosome Segregation/physiology , Chromosomes, Fungal , Kinetochores , Microtubule-Associated Proteins/metabolism , Models, Biological , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , Spindle Apparatus , Chromosomes, Fungal/metabolism , Chromosomes, Fungal/ultrastructure , Cryoelectron Microscopy , Kinetochores/metabolism , Kinetochores/ultrastructure , Microtubules/metabolism , Microtubules/ultrastructure , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
17.
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
18.
Elife ; 72018 12 14.
Article in English | MEDLINE | ID: mdl-30547880

ABSTRACT

Accurate chromosome segregation relies on bioriented amphitelic attachments of chromosomes to microtubules of the mitotic spindle, in which sister chromatids are connected to opposite spindle poles. BUB-1 is a protein of the Spindle Assembly Checkpoint (SAC) that coordinates chromosome attachment with anaphase onset. BUB-1 is also required for accurate sister chromatid segregation independently of its SAC function, but the underlying mechanism remains unclear. Here we show that, in Caenorhabditis elegans embryos, BUB-1 accelerates the establishment of non-merotelic end-on kinetochore-microtubule attachments by recruiting the RZZ complex and its downstream partner dynein-dynactin at the kinetochore. In parallel, BUB-1 limits attachment maturation by the SKA complex. This activity opposes kinetochore-microtubule attachment stabilisation promoted by CLS-2CLASP-dependent kinetochore-microtubule assembly. BUB-1 is therefore a SAC component that coordinates the function of multiple downstream kinetochore-associated proteins to ensure accurate chromosome segregation.


Subject(s)
Anaphase , Caenorhabditis elegans Proteins/genetics , Chromosome Segregation , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Dynactin Complex/genetics , Dynactin Complex/metabolism , Dyneins/genetics , Dyneins/metabolism , Embryo, Nonmammalian , Gene Expression Regulation , Kinetochores/ultrastructure , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Spindle Apparatus/ultrastructure
19.
Elife ; 72018 11 26.
Article in English | MEDLINE | ID: mdl-30475206

ABSTRACT

Several studies have shown that RNAi-mediated depletion of splicing factors (SFs) results in mitotic abnormalities. However, it is currently unclear whether these abnormalities reflect defective splicing of specific pre-mRNAs or a direct role of the SFs in mitosis. Here, we show that two highly conserved SFs, Sf3A2 and Prp31, are required for chromosome segregation in both Drosophila and human cells. Injections of anti-Sf3A2 and anti-Prp31 antibodies into Drosophila embryos disrupt mitotic division within 1 min, arguing strongly against a splicing-related mitotic function of these factors. We demonstrate that both SFs bind spindle microtubules (MTs) and the Ndc80 complex, which in Sf3A2- and Prp31-depleted cells is not tightly associated with the kinetochores; in HeLa cells the Ndc80/HEC1-SF interaction is restricted to the M phase. These results indicate that Sf3A2 and Prp31 directly regulate interactions among kinetochores, spindle microtubules and the Ndc80 complex in both Drosophila and human cells.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eye Proteins/genetics , Mitosis , Nuclear Proteins/genetics , RNA Splicing Factors/genetics , Animals , Antibodies, Neutralizing/pharmacology , Chromosome Segregation/drug effects , Conserved Sequence , Cytoskeletal Proteins , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Eye Proteins/antagonists & inhibitors , Eye Proteins/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Kinetochores/drug effects , Kinetochores/metabolism , Kinetochores/ultrastructure , Microtubules/drug effects , Microtubules/metabolism , Microtubules/ultrastructure , Mitosis/drug effects , Nuclear Proteins/metabolism , Protein Binding , RNA Splicing Factors/antagonists & inhibitors , RNA Splicing Factors/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
20.
Exp Cell Res ; 371(2): 435-443, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30195030

ABSTRACT

Nucleoporins (Nups) are a large and diverse family of proteins that mediate nucleocytoplasmic transport at interphase of vertebrate cells. Nups also function in mitosis progression. However, whether Nups are involved in oocyte meiosis progression is still rarely known. In this study, we delineated the roles and regulatory mechanisms of Nucleoporin35 (Nup35) during oocyte meiotic maturation. The immunofluorescent signal of Nup35 was localized in the nuclear membrane at germinal vesicle (GV) stage, the microtubules and spindle at pro-metaphase I (pro-MI), metaphase I (MI), and metaphase II (MII), but to the spindle poles at anaphase I (AI) and telophase I (TI). The dynamic localization pattern of Nup35 during oocyte meiotic maturation implied its specific roles. We also found that Nup35 existed as a putatively phosphorylated form after resumption of meiosis (GVBD), but not at GV stage, implying its functional switch from nuclear membrane to meiotic progression. Further study uncovered that knockdown of Nup35 by specific siRNA significantly compromised the extrusion of first polar body (PBE), but not GVBD, with defects of spindle assembly and chromosome alignment and dissociated some localization signal of p-ERK1/2 from spindle poles to cytoplasm. A defective kinetochore - microtubule attachment (K-MT) was also identified in oocytes after knockdown of Nup35, which activates spindle assembly checkpoint. In conclusion, our results suggest that Nup35 is putatively phosphorylated and released to the cytoplasm after resumption of meiosis, and regulates spindle assembly and chromosome alignment.


Subject(s)
Kinetochores/metabolism , Meiosis , Microtubules/metabolism , Nuclear Pore Complex Proteins/genetics , Oocytes/metabolism , Spindle Apparatus/metabolism , Animals , Female , Gene Expression Regulation, Developmental , Kinetochores/ultrastructure , Mice , Microtubules/ultrastructure , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Nuclear Pore Complex Proteins/antagonists & inhibitors , Nuclear Pore Complex Proteins/metabolism , Oocytes/ultrastructure , Phosphorylation , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spindle Apparatus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...