Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Elife ; 132024 Mar 08.
Article in English | MEDLINE | ID: mdl-38456462

ABSTRACT

The physical basis of phase separation is thought to consist of the same types of bonds that specify conventional macromolecular interactions yet is unsatisfyingly often referred to as 'fuzzy'. Gaining clarity on the biogenesis of membraneless cellular compartments is one of the most demanding challenges in biology. Here, we focus on the chromosome passenger complex (CPC), that forms a chromatin body that regulates chromosome segregation in mitosis. Within the three regulatory subunits of the CPC implicated in phase separation - a heterotrimer of INCENP, Survivin, and Borealin - we identify the contact regions formed upon droplet formation using hydrogen/deuterium exchange mass spectrometry (HXMS). These contact regions correspond to some of the interfaces seen between individual heterotrimers within the crystal lattice they form. A major contribution comes from specific electrostatic interactions that can be broken and reversed through initial and compensatory mutagenesis, respectively. Our findings reveal structural insight for interactions driving liquid-liquid demixing of the CPC. Moreover, we establish HXMS as an approach to define the structural basis for phase separation.


Subject(s)
Cell Cycle Proteins , Phase Separation , Cell Cycle Proteins/genetics , Chromosomes , Mitosis , Cytoskeleton , Chromosome Segregation , Aurora Kinase B/genetics
2.
J Biol Chem ; 300(3): 105669, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38272221

ABSTRACT

The mitotic spindle contains many bundles of microtubules (MTs) including midzones and kinetochore fibers, but little is known about how bundled structures are formed. Here, we show that the chromosomal passenger complex (CPC) purified from Escherichia coli undergoes liquid-liquid demixing in vitro. An emergent property of the resultant condensates is to generate parallel MT bundles when incubated with free tubulin and GTP in vitro. We demonstrate that MT bundles emerge from CPC droplets with protruding minus ends that then grow into long and tapered MT structures. During this growth, we found that the CPC in these condensates apparently reorganize to coat and bundle the resulting MT structures. CPC mutants attenuated for liquid-liquid demixing or MT binding prevented the generation of parallel MT bundles in vitro and reduced the number of MTs present at spindle midzones in HeLa cells. Our data demonstrate that an in vitro biochemical activity to produce MT bundles emerges after the concentration of the CPC and provides models for how cells generate parallel-bundled MT structures that are important for the assembly of the mitotic spindle. Moreover, these data suggest that cells contain MT-organizing centers that generate MT bundles that emerge with the opposite polarity from centrosomes.


Subject(s)
Chromosomes , Microtubules , Spindle Apparatus , Humans , HeLa Cells , Kinetochores/metabolism , Microtubules/metabolism , Mitosis , Spindle Apparatus/metabolism , Tubulin/genetics , Tubulin/metabolism , Animals , Xenopus laevis
3.
Cell Rep ; 42(11): 113380, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37950869

ABSTRACT

Coronary artery disease (CAD) is characterized by atherosclerotic plaque formation in the arterial wall. CAD progression involves complex interactions and phenotypic plasticity among vascular and immune cell lineages. Single-cell RNA-seq (scRNA-seq) studies have highlighted lineage-specific transcriptomic signatures, but human cell phenotypes remain controversial. Here, we perform an integrated meta-analysis of 22 scRNA-seq libraries to generate a comprehensive map of human atherosclerosis with 118,578 cells. Besides characterizing granular cell-type diversity and communication, we leverage this atlas to provide insights into smooth muscle cell (SMC) modulation. We integrate genome-wide association study data and uncover a critical role for modulated SMC phenotypes in CAD, myocardial infarction, and coronary calcification. Finally, we identify fibromyocyte/fibrochondrogenic SMC markers (LTBP1 and CRTAC1) as proxies of atherosclerosis progression and validate these through omics and spatial imaging analyses. Altogether, we create a unified atlas of human atherosclerosis informing cell state-specific mechanistic and translational studies of cardiovascular diseases.


Subject(s)
Atherosclerosis , Coronary Artery Disease , Myocardial Infarction , Plaque, Atherosclerotic , Humans , Genome-Wide Association Study , Atherosclerosis/genetics , Coronary Artery Disease/genetics , Myocytes, Smooth Muscle , Calcium-Binding Proteins/genetics
4.
bioRxiv ; 2023 Jul 23.
Article in English | MEDLINE | ID: mdl-37292983

ABSTRACT

The physical basis of phase separation is thought to consist of the same types of bonds that specify conventional macromolecular interactions yet is unsatisfyingly often referred to as 'fuzzy'. Gaining clarity on the biogenesis of membraneless cellular compartments is one of the most demanding challenges in biology. Here, we focus on the chromosome passenger complex (CPC), that forms a chromatin body that regulates chromosome segregation in mitosis. Within the three regulatory subunits of the CPC implicated in phase separation - a heterotrimer of INCENP, Survivin, and Borealin - we identify the contact regions formed upon droplet formation using hydrogen/deuterium-exchange mass spectrometry (HXMS). These contact regions correspond to some of the interfaces seen between individual heterotrimers within the crystal lattice they form. A major contribution comes from specific electrostatic interactions that can be broken and reversed through initial and compensatory mutagenesis, respectively. Our findings reveal structural insight for interactions driving liquid-liquid demixing of the CPC. Moreover, we establish HXMS as an approach to define the structural basis for phase separation.

5.
Front Cell Dev Biol ; 11: 1139367, 2023.
Article in English | MEDLINE | ID: mdl-36994100

ABSTRACT

Cell division events require regulatory systems to ensure that events happen in a distinct order. The classic view of temporal control of the cell cycle posits that cells order events by linking them to changes in Cyclin Dependent Kinase (CDK) activities. However, a new paradigm is emerging from studies of anaphase where chromatids separate at the central metaphase plate and then move to opposite poles of the cell. These studies suggest that distinct events are ordered depending upon the location of each chromosome along its journey from the central metaphase plate to the elongated spindle poles. This system is dependent upon a gradient of Aurora B kinase activity that emerges during anaphase and acts as a spatial beacon to control numerous anaphase/telophase events and cytokinesis. Recent studies also suggest that Aurora A kinase activity specifies proximity of chromosomes or proteins to spindle poles during prometaphase. Together these studies argue that a key role for Aurora kinases is to provide spatial information that controls events depending upon the location of chromosomes or proteins along the mitotic spindle.

6.
Mol Biol Cell ; 34(1): br1, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36350697

ABSTRACT

Dynein inactivates the spindle assembly checkpoint (SAC) by transporting checkpoint proteins away from kinetochores toward spindle poles in a process known as "stripping." We find that inhibition of Aurora A kinase, which is localized to spindle poles, enables the accumulation of the spindle checkpoint activator Mad1 at poles where it is normally absent. Aurora kinases phosphorylate the dynein activator NudE neurodevelopment protein 1 like 1 (Ndel1) on Ser285 and Mad1 accumulates at poles when Ndel1 is replaced by a nonphosphorylatable mutant in human cells. The pole focusing protein NuMA, transported to poles by dynein, also accumulates at poles in cells harboring a mutant Ndel1. Phosphorylation of Ndel1 on Ser285 is required for robust spindle checkpoint activity and regulates the poles of asters in Xenopus extracts. Our data suggest that dynein/SAC complexes that are generated at kinetochores and then transported directionally toward poles on microtubules are inhibited by Aurora A before they reach spindle poles. These data suggest that Aurora A generates a spatial signal at spindle poles that controls dynein transport and spindle function.


Subject(s)
Dyneins , Spindle Apparatus , Humans , Dyneins/metabolism , Spindle Apparatus/metabolism , Aurora Kinase A/metabolism , Kinetochores/metabolism , Cell Cycle Proteins/metabolism , Spindle Poles/metabolism , Microtubules/metabolism , Carrier Proteins/metabolism
7.
Mol Biol Cell ; 33(9): br15, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35653296

ABSTRACT

The Chromosome Passenger Complex (CPC) generates chromosome autonomous signals that regulate mitotic events critical for genome stability. Tip60 is a lysine acetyltransferase that is a tumor suppressor and is targeted for proteasomal degradation by oncogenic papilloma viruses. Mitotic regulation requires the localization of the CPC to inner centromeres, which is driven by the Haspin kinase phosphorylating histone H3 on threonine 3 (H3T3ph). Here we describe how Tip60 acetylates histone H3 at lysine 4 (H3K4ac) to block both the H3T3ph writer and the reader to ensure that this mitotic signaling cannot begin before prophase. Specifically, H3K4ac inhibits Haspin phosphorylation of H3T3 and prevents binding of the Survivin subunit to H3T3ph. Tip60 acetylates H3K4 during S/G2 at centromeres. Inhibition of Tip60 allows the CPC to bind centromeres in G2 cells, and targeting of Tip60 to centromeres prevents CPC localization in mitosis. The H3K4ac mark is removed in prophase by HDAC3 to initiate the CPC localization cascade. Together, our results suggest that Tip60 and HDAC3 temporally control H3K4 acetylation to precisely time the targeting of the CPC to inner centromeres.


Subject(s)
Histones , Protein Serine-Threonine Kinases , Acetylation , Centromere/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitosis , Phosphorylation , Threonine/genetics , Threonine/metabolism
8.
BMC Cancer ; 21(1): 186, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33622270

ABSTRACT

BACKGROUND: Targeting Protein for Xenopus Kinesin Like Protein 2 (TPX2) is a microtubule associated protein that functions in mitotic spindle assembly. TPX2 also localizes to the nucleus where it functions in DNA damage repair during S-phase. We and others have previously shown that TPX2 RNA levels are strongly associated with chromosomal instability (CIN) in breast and other cancers, and TPX2 RNA levels have been demonstrated to correlate with aggressive behavior and poor clinical outcome across a range of solid malignancies, including breast cancer. METHODS: We perform TPX2 IHC on a cohort of 253 primary breast cancers and adopt a clinically amenable scoring system to separate tumors into low, intermediate, or high TPX2 expression. We then correlate TPX2 expression against diverse pathologic parameters and important measures of clinical outcome, including disease-specific and overall survival. We link TPX2 expression to TP53 mutation and evaluate whether TPX2 is an independent predictor of chromosomal instability (CIN). RESULTS: We find that TPX2 nuclear expression strongly correlates with high grade morphology, elevated clinical stage, negative ER and PR status, and both disease-specific and overall survival. We also show that increased TPX2 nuclear expression correlates with elevated ploidy, supernumerary centrosomes, and TP53 mutation. TPX2 nuclear expression correlates with CIN via univariate analyses but is not independently predictive when compared to ploidy, Ki67, TP53 mutational status, centrosome number, and patient age. CONCLUSIONS: Our findings demonstrate a strong correlation between TPX2 nuclear expression and aggressive tumor behavior, and show that TPX2 overexpression frequently occurs in the setting of TP53 mutation and elevated ploidy. However, TPX2 expression is not an independent predictor of CIN where it fails to outperform existing clinical and pathologic metrics.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle Proteins/physiology , Cell Nucleus/chemistry , Chromosomal Instability , Microtubule-Associated Proteins/physiology , Mutation , Tumor Suppressor Protein p53/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cell Proliferation , Cohort Studies , Female , Humans , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/genetics , Middle Aged , RNA, Messenger/analysis
9.
Trends Cell Biol ; 30(9): 676-687, 2020 09.
Article in English | MEDLINE | ID: mdl-32684321

ABSTRACT

The inner centromere is a region on the mitotic chromosome that serves as a platform for mitotic signaling and possesses unique biophysical properties that enable it to withstand relatively large pulling forces that are generated by kinetochores (KTs) during chromosome segregation. The chromosomal passenger complex (CPC) localizes to and is the key regulator of inner centromere organization and function during mitosis. Recently, we demonstrated that in addition to its kinase and histone code-reading activities, the CPC also can undergo liquid-liquid phase separation (LLPS) and proposed that the inner centromere is a membraneless organelle scaffolded by the CPC. In this perspective, we explore mechanisms that can allow the formation and dissolution of this membraneless body. The cell-cycle-regulated spatially defined assembly and disassembly of the CPC condensate at the inner centromere can reveal general principles about how histone modifications control chromatin-bound membraneless organelles. We further explore how the ability of the CPC to undergo LLPS may contribute to the organization and function of the inner centromere during mitosis.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Animals , Humans , Kinetochores/metabolism , Mitosis , Models, Biological , Phase Transition
10.
Front Oncol ; 10: 585551, 2020.
Article in English | MEDLINE | ID: mdl-33489883

ABSTRACT

It has long been recognized that defects in cell cycle checkpoint and DNA repair pathways give rise to genomic instability, tumor heterogeneity, and metastasis. Despite this knowledge, the transcription factor-mediated gene expression programs that enable survival and proliferation in the face of enormous replication stress and DNA damage have remained elusive. Using robust omics data from two independent studies, we provide evidence that a large cohort of lung adenocarcinomas exhibit significant genome instability and overexpress the DNA damage responsive transcription factor MYB proto-oncogene like 2 (MYBL2). Across two studies, elevated MYBL2 expression was a robust marker of poor overall survival and disease-free survival outcomes, regardless of disease stage. Clinically, elevated MYBL2 expression identified patients with aggressive early onset disease, increased lymph node involvement, and increased incidence of distant metastases. Analysis of genomic sequencing data demonstrated that MYBL2 High lung adenocarcinomas had elevated somatic mutation burden, widespread chromosomal alterations, and alterations in single-strand DNA break repair pathways. In this study, we provide evidence that impaired single-strand break repair, combined with a loss of cell cycle regulators TP53 and RB1, give rise to MYBL2-mediated transcriptional programs. Omics data supports a model wherein tumors with significant genomic instability upregulate MYBL2 to drive genes that control replication stress responses, promote error-prone DNA repair, and antagonize faithful homologous recombination repair. Our study supports the use of checkpoint kinase 1 (CHK1) pharmacological inhibitors, in targeted MYBL2 High patient cohorts, as a future therapy to improve lung adenocarcinoma patient outcomes.

11.
Nat Cell Biol ; 21(9): 1127-1137, 2019 09.
Article in English | MEDLINE | ID: mdl-31481798

ABSTRACT

The inner centromere is a region on every mitotic chromosome that enables specific biochemical reactions that underlie properties, such as the maintenance of cohesion, the regulation of kinetochores and the assembly of specialized chromatin, that can resist microtubule pulling forces. The chromosomal passenger complex (CPC) is abundantly localized to the inner centromeres and it is unclear whether it is involved in non-kinase activities that contribute to the generation of these unique chromatin properties. We find that the borealin subunit of the CPC drives phase separation of the CPC in vitro at concentrations that are below those found on the inner centromere. We also provide strong evidence that the CPC exists in a phase-separated state at the inner centromere. CPC phase separation is required for its inner-centromere localization and function during mitosis. We suggest that the CPC combines phase separation, kinase and histone code-reading activities to enable the formation of a chromatin body with unique biochemical activities at the inner centromere.


Subject(s)
Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Cell Cycle Proteins/metabolism , Chromosome Segregation/physiology , Cytoskeleton/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mitosis
12.
Nat Commun ; 10(1): 682, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737408

ABSTRACT

Proper chromosome segregation depends upon kinetochore phosphorylation by the Chromosome Passenger Complex (CPC). Current models suggest the activity of the CPC decreases in response to the inter-kinetochore stretch that accompanies the formation of bi-oriented microtubule attachments, however little is known about tension-independent CPC phosphoregulation. Microtubule bundles initially lie in close proximity to inner centromeres and become depleted by metaphase. Here we find these microtubules control kinetochore phosphorylation by the CPC in a tension independent manner via a microtubule-binding site on the Borealin subunit. Disruption of Borealin-microtubule interactions generates reduced phosphorylation of prometaphase kinetochores, improper kinetochore-microtubule attachments and weakened spindle checkpoint signals. Experimental and modeling evidence suggests that kinetochore phosphorylation is greatly stimulated when the CPC binds microtubules that lie near the inner centromere, even if kinetochores have high inter-kinetochore stretch. We propose the CPC senses its local environment through microtubule structures to control phosphorylation of kinetochores.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomes/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Cell Cycle Proteins/genetics , Humans , Microtubules/genetics , Mitosis/physiology , Phosphorylation , Protein Binding
13.
Nat Commun ; 9(1): 4275, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323222

ABSTRACT

Predicting the response and identifying additional targets that will improve the efficacy of chemotherapy is a major goal in cancer research. Through large-scale in vivo and in vitro CRISPR knockout screens in pancreatic ductal adenocarcinoma cells, we identified genes whose genetic deletion or pharmacologic inhibition synergistically increase the cytotoxicity of MEK signaling inhibitors. Furthermore, we show that CRISPR viability scores combined with basal gene expression levels could model global cellular responses to the drug treatment. We develop drug response evaluation by in vivo CRISPR screening (DREBIC) method and validated its efficacy using large-scale experimental data from independent experiments. Comparative analyses demonstrate that DREBIC predicts drug response in cancer cells from a wide range of tissues with high accuracy and identifies therapeutic vulnerabilities of cancer-causing mutations to MEK inhibitors in various cancer types.


Subject(s)
Antineoplastic Agents/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Combinatorial Chemistry Techniques , Drug Delivery Systems , Gene Knockout Techniques , Genetic Testing , Models, Biological , Pancreatic Neoplasms/genetics , Animals , Cell Cycle Checkpoints , Cell Death , Cell Line, Tumor , Drug Synergism , Humans , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Reproducibility of Results
14.
Cell Rep ; 23(9): 2758-2769, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29847804

ABSTRACT

Although aneuploidy is found in the majority of tumors, the degree of aneuploidy varies widely. It is unclear how cancer cells become aneuploid or how highly aneuploid tumors are different from those of more normal ploidy. We developed a simple computational method that measures the degree of aneuploidy or structural rearrangements of large chromosome regions of 522 human breast tumors from The Cancer Genome Atlas (TCGA). Highly aneuploid tumors overexpress activators of mitotic transcription and the genes encoding proteins that segregate chromosomes. Overexpression of three mitotic transcriptional regulators, E2F1, MYBL2, and FOXM1, is sufficient to increase the rate of lagging anaphase chromosomes in a non-transformed vertebrate tissue, demonstrating that this event can initiate aneuploidy. Highly aneuploid human breast tumors are also enriched in TP53 mutations. TP53 mutations co-associate with the overexpression of mitotic transcriptional activators, suggesting that these events work together to provide fitness to breast tumors.


Subject(s)
Aneuploidy , Breast Neoplasms/genetics , Anaphase/genetics , Animals , Breast Neoplasms/pathology , Chromosomal Instability , Chromosomes, Human/genetics , Embryo, Nonmammalian/metabolism , Female , Gene Frequency/genetics , Humans , Mitosis/genetics , Models, Genetic , Mutation/genetics , Phenotype , Transcription Factors/metabolism , Transcription, Genetic , Xenopus/embryology
15.
Chromosoma ; 127(3): 375-386, 2018 09.
Article in English | MEDLINE | ID: mdl-29656322

ABSTRACT

Assembly of the mitotic spindle is essential for proper chromosome segregation during mitosis. Maintenance of spindle poles requires precise regulation of kinesin- and dynein-generated forces, and improper regulation of these forces disrupts pole integrity leading to pole fragmentation. The formation and function of the mitotic spindle are regulated by many proteins, including Aurora A kinase and the motor proteins Kif2a and Eg5. Here, we characterize a surprising role for the RhoA GTPase-activating protein, p190RhoGAP, in regulating the mitotic spindle. We show that cells depleted of p190RhoGAP arrest for long periods in mitosis during which cells go through multiple transitions between having bipolar and multipolar spindles. Most of the p190RhoGAP-depleted cells finally achieve a stable bipolar attachment and proceed through anaphase. The multipolar spindle phenotype can be rescued by low doses of an Eg5 inhibitor. Moreover, we show that p190RhoGAP-depleted multipolar cells localize Aurora A to all the poles, but the kinase is only activated at the two centriolar poles. Overall, our data identify an unappreciated connection between p190RhoGAP and the proteins that control spindle poles including Aurora A kinase and Eg5 that is required to prevent or correct spindle pole fragmentation.


Subject(s)
Aurora Kinase A/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Mitosis , Repressor Proteins/metabolism , Spindle Apparatus/metabolism , Animals , Cell Cycle Checkpoints , Cell Line , Centrosome , Humans , Kinesins/metabolism
16.
Dev Cell ; 41(4): 438-449.e4, 2017 05 22.
Article in English | MEDLINE | ID: mdl-28535377

ABSTRACT

Yeast use the ring-shaped Dam1 complex to slide down depolymerizing microtubules to move chromosomes, but current models suggest that other eukaryotes do not have a sliding ring. We visualized Ndc80 and Ska complexes on microtubules by electron microscopic tomography to identify the structure of the human kinetochore-microtubule attachment. Ndc80 recruits the Ska complex so that the V shape of the Ska dimer interacts along protofilaments. We identify a mutant of the Ndc80 tail that is deficient in Ska recruitment to kinetochores and in orienting Ska along protofilaments in vitro. This mutant Ndc80 binds microtubules with normal affinity but is deficient in clustering along protofilaments. We propose that Ska is recruited to kinetochores by clusters of Ndc80 proteins and that our structure of Ndc80 and Ska complexes on microtubules suggests a mechanism for metazoan kinetochores to couple the depolymerization of microtubules to power the movement of chromosomes.


Subject(s)
Kinetochores/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Aurora Kinases/metabolism , Cytoskeletal Proteins , HeLa Cells , Humans , Kinetochores/ultrastructure , Metaphase , Microtubules/metabolism , Microtubules/ultrastructure , Models, Biological , Models, Molecular , Nuclear Proteins/chemistry , Point Mutation/genetics , Protein Domains , Spindle Apparatus/metabolism
17.
Mol Biol Cell ; 28(1): 54-64, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27807043

ABSTRACT

Centromeric chromatin is required for kinetochore assembly during mitosis and accurate chromosome segregation. A unique nucleosome containing the histone H3-specific variant CENP-A is the defining feature of centromeric chromatin. In humans, CENP-A nucleosome deposition occurs in early G1 just after mitotic exit at the time when the CENP-A deposition machinery localizes to centromeres. The mechanism by which CENP-A is deposited onto an existing, condensed chromatin template is not understood. Here we identify the selective association of the CENP-A chaperone HJURP with the condensin II complex and not condensin I. We show CAPH2 is present at centromeres during early G1 at the time when CENP-A deposition is occurring. CAPH2 localization to early G1 centromeres is dependent on HJURP. The CENP-A chaperone and assembly factor HJURP induces decondensation of a noncentromeric LacO array, and this decondensation is modulated by the condensin II complex. We show that condensin II function at the centromere is required for new CENP-A deposition in human cells. These data demonstrate that HJURP selectively recruits the condensin II chromatin-remodeling complex to facilitate CENP-A deposition in human cells.


Subject(s)
Autoantigens/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Adenosine Triphosphatases/metabolism , Centromere Protein A , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Chromosome Segregation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , G1 Phase , Histones/metabolism , Humans , Mitosis , Multiprotein Complexes/metabolism , Nucleosomes , Protein Binding/physiology , Serine Endopeptidases/metabolism , Serine Endopeptidases/physiology , Succinate Dehydrogenase/metabolism
18.
Elife ; 52016 Mar 16.
Article in English | MEDLINE | ID: mdl-26981768

ABSTRACT

The spindle- and kinetochore-associated (Ska) complex is essential for normal anaphase onset in mitosis. The C-terminal domain (CTD) of Ska1 binds microtubules and was proposed to facilitate kinetochore movement on depolymerizing spindle microtubules. Here, we show that Ska complex recruits protein phosphatase 1 (PP1) to kinetochores. This recruitment requires the Ska1 CTD, which binds PP1 in vitro and in human HeLa cells. Ska1 lacking its CTD fused to a PP1-binding peptide or fused directly to PP1 rescues mitotic defects caused by Ska1 depletion. Ska1 fusion to catalytically dead PP1 mutant does not rescue and shows dominant negative effects. Thus, the Ska complex, specifically the Ska1 CTD, recruits PP1 to kinetochores to oppose spindle checkpoint signaling kinases and promote anaphase onset. Microtubule binding by Ska, rather than acting in force production for chromosome movement, may instead serve to promote PP1 recruitment to kinetochores fully attached to spindle microtubules at metaphase.


Subject(s)
Anaphase , Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Metaphase , Protein Phosphatase 1/metabolism , HeLa Cells , Humans , Protein Domains
19.
Trends Biochem Sci ; 41(2): 160-174, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26705896

ABSTRACT

There is increasing evidence that regulators of the spindle checkpoint, kinetochore-microtubule attachments, and sister chromatid cohesion are part of an interconnected mitotic regulatory circuit with two positive feedback loops and the chromosome passenger complex (CPC) at its center. If true, this conceptual breakthrough needs to be integrated into models of mitosis. In this review, we describe this circuit and point out how the double feedback loops could provide insights into the self-organization of some mitotic processes and the autonomy of every chromosome on the mitotic spindle. We also provide working models for how mitotic events may be coordinated by this circuit.


Subject(s)
Centromere/metabolism , Mitosis , Signal Transduction
20.
Dev Biol ; 408(2): 345-57, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26391338

ABSTRACT

Functional characterisation of proteins and large-scale, systems-level studies are enabled by extensive sets of cloned open reading frames (ORFs) in an easily-accessible format that enables many different applications. Here we report the release of the first stage of the Xenopus ORFeome, which contains 8673 ORFs from the Xenopus Gene Collection (XGC) for Xenopus laevis, cloned into a Gateway® donor vector enabling rapid in-frame transfer of the ORFs to expression vectors. This resource represents an estimated 7871 unique genes, approximately 40% of the non-redundant X. laevis gene complement, and includes 2724 genes where the human ortholog has an association with disease. Transfer into the Gateway system was validated by 5' and 3' end sequencing of the entire collection and protein expression of a set of test clones. In a parallel process, the underlying ORF predictions from the original XGC collection were re-analysed to verify quality and full-length status, identifying those proteins likely to exhibit truncations when translated. These data are integrated into Xenbase, the Xenopus community database, which associates genomic, expression, function and human disease model metadata to each ORF, enabling end-users to search for ORFeome clones with links to commercial distributors of the collection. When coupled with the experimental advantages of Xenopus eggs and embryos, the ORFeome collection represents a valuable resource for functional genomics and disease modelling.


Subject(s)
Open Reading Frames , Xenopus/genetics , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Databases, Genetic , Disease/genetics , Genomics , Humans , Models, Genetic , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Species Specificity , Xenopus Proteins/genetics , Xenopus laevis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL