Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
PLoS Genet ; 17(5): e1009592, 2021 05.
Article in English | MEDLINE | ID: mdl-34033659

ABSTRACT

The spindle assembly checkpoint (SAC) prevents anaphase onset in response to chromosome attachment defects, and SAC silencing is essential for anaphase onset. Following anaphase onset, activated Cdc14 phosphatase dephosphorylates the substrates of cyclin-dependent kinase to facilitate anaphase progression and mitotic exit. In budding yeast, Cdc14 dephosphorylates Fin1, a regulatory subunit of protein phosphatase 1 (PP1), to enable kinetochore localization of Fin1-PP1. We previously showed that kinetochore-localized Fin1-PP1 promotes the removal of the SAC protein Bub1 from the kinetochore during anaphase. We report here that Fin1-PP1 also promotes kinetochore removal of Bub3, the Bub1 partner, but has no effect on another SAC protein Mad1. Moreover, the kinetochore localization of Bub1-Bub3 during anaphase requires Aurora B/Ipl1 kinase activity. We further showed that Fin1-PP1 facilitates the dephosphorylation of kinetochore protein Ndc80, a known Ipl1 substrate. This dephosphorylation reduces kinetochore association of Bub1-Bub3 during anaphase. In addition, we found that untimely Ndc80 dephosphorylation causes viability loss in response to tensionless chromosome attachments. These results suggest that timely localization of Fin1-PP1 to the kinetochore controls the functional window of SAC and is therefore critical for faithful chromosome segregation.


Subject(s)
Anaphase , Aurora Kinases/metabolism , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Kinetochores/metabolism , Nuclear Proteins/metabolism , Protein Phosphatase 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , Chromosome Segregation , Kinetochores/chemistry , Kinetochores/drug effects , Microbial Viability/genetics , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphorylation , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Spindle Apparatus/drug effects , Time Factors
2.
Cell Rep ; 32(5): 107987, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32755581

ABSTRACT

Advanced maternal age is highly associated with a decline in oocyte quality, but effective approaches to improve it have still not been fully determined. Here, we report that in vivo supplementation of nicotinamide mononucleotide (NMN) efficaciously improves the quality of oocytes from naturally aged mice by recovering nicotinamide adenine dinucleotide (NAD+) levels. NMN supplementation not only increases ovulation of aged oocytes but also enhances their meiotic competency and fertilization ability by maintaining the normal spindle/chromosome structure and the dynamics of the cortical granule component ovastacin. Moreover, single-cell transcriptome analysis shows that the beneficial effect of NMN on aged oocytes is mediated by restoration of mitochondrial function, eliminating the accumulated ROS to suppress apoptosis. Collectively, our data reveal that NMN supplementation is a feasible approach to protect oocytes from advanced maternal age-related deterioration, contributing to the improvement of reproductive outcome of aged women and assisted reproductive technology.


Subject(s)
Aging/physiology , Cellular Senescence , Nicotinamide Mononucleotide/pharmacology , Oocytes/cytology , Animals , Apoptosis/drug effects , Cellular Senescence/drug effects , Chromosomes, Mammalian/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , DNA Damage , Dietary Supplements , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Fertilization/drug effects , Kinetochores/drug effects , Kinetochores/metabolism , Male , Meiosis/drug effects , Metalloproteases/metabolism , Mice, Inbred ICR , Microtubules/drug effects , Microtubules/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Oocytes/drug effects , Reactive Oxygen Species/metabolism , Spermatozoa/drug effects , Spermatozoa/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Transcriptome/genetics
3.
Open Biol ; 10(7): 200101, 2020 07.
Article in English | MEDLINE | ID: mdl-32634373

ABSTRACT

The distance between fluorescent spots formed by various kinetochore proteins (delta) is commonly interpreted as a manifestation of intrakinetochore tension (IKT) caused by microtubule-mediated forces. However, large-scale changes of the kinetochore architecture (such as its shape or dimensions) may also contribute to the value of delta. To assess contributions of these non-elastic changes, we compare behaviour of delta values in human kinetochores with small yet mechanically malleable kinetochores against compound kinetochores in Indian muntjac (IM) cells whose architecture remains constant. Due to the micrometre-scale length of kinetochore plates in IM, their shape and orientation are discernible in conventional light microscopy, which enables precise measurements of IKT independent of contributions from changes in overall architecture of the organelle. We find that delta in IM kinetochores remains relatively constant when microtubule-mediated forces are suppressed by Taxol, but it prominently decreases upon detachment of microtubules. By contrast, large decreases of delta observed in Taxol-treated human cells coincide with prominent changes in length and curvature of the kinetochore plate. These observations, supported by computational modelling, suggest that at least 50% of the decrease in delta in human cells reflects malleable reorganization of kinetochore architecture rather than elastic recoil due to IKT.


Subject(s)
Chromosomes/drug effects , Kinetochores/drug effects , Mitosis/genetics , Nuclear Proteins/genetics , Animals , Centromere Protein A/genetics , Chromosome Segregation/drug effects , Chromosome Segregation/genetics , Chromosomes/genetics , Cytoskeletal Proteins/genetics , Humans , Metaphase/genetics , Microtubules/drug effects , Microtubules/genetics , Mitosis/drug effects , Muntjacs/genetics , Nuclear Proteins/antagonists & inhibitors , Paclitaxel/pharmacology , Spindle Apparatus/drug effects , Spindle Apparatus/genetics
4.
Methods Cell Biol ; 158: 91-116, 2020.
Article in English | MEDLINE | ID: mdl-32423652

ABSTRACT

During mitosis, spindle microtubules dynamically attach to and detach from kinetochores in a precise and regulated fashion. To ensure mitotic fidelity, kinetochore-microtubule (k-MT) attachments must be stable enough to satisfy the spindle assembly checkpoint (SAC), but sufficiently unstable to facilitate the correction of maloriented attachments. Different methods are available to assess k-MT stability in both live and fixed cells, but a comparative survey of these methods has not yet been reported. Here, we evaluate several quantitative and semiquantitative methods for determining k-MT stability and apply each technique to illustrate changes in spindle microtubule dynamics upon perturbation with physiologically relevant concentrations of microtubule stabilizing (Taxol) and destabilizing (UMK57 and nocodazole) compounds. We discuss the utility of each technique for defining specific features of spindle microtubule dynamics and k-MT attachment stability.


Subject(s)
Cytological Techniques/methods , Kinetochores/metabolism , Microtubules/metabolism , Calcium/pharmacology , Cell Line , Cold-Shock Response/drug effects , Humans , Imaging, Three-Dimensional , Kinetochores/drug effects , Light , Microtubules/drug effects , Nocodazole/pharmacology , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
5.
J Cell Physiol ; 235(10): 7030-7042, 2020 10.
Article in English | MEDLINE | ID: mdl-32017059

ABSTRACT

Histone deacetylase 6 (HDAC6) participates in mouse oocyte maturation by deacetylating α-tubulin. However, how HDAC6 expression is regulated in oocytes remains unknown. In the present study, we discovered that mouse oocytes had a high level of HDAC6 expression and a low level of DNA methylation status in their promoter region. Then, a selective HDAC6 inhibitor, tubastatin A (Tub-A) was chosen to investigate the role of HDAC6 in oocyte maturation. Our results revealed that inhibition of HDAC6 caused meiotic progression arrest, disturbed spindle/chromosome organization, and kinetochore-microtubule attachments without impairing spindle assembly checkpoint function. Moreover, inhibition of HDAC6 not only increased the acetylation of α-tubulin but also elevated the acetylation status of H4K16 and decreased the phosphorylation level of H3T3 and H3S10. Conversely, depressed H3T3 phosphorylation by its kinase inhibitor increased the acetylation level of H4K16. Finally, single cell RNA-seq analysis revealed that the cell cycle-related genes CCNB1, CDK2, SMAD3, YWHAZ and the methylation-related genes DNMT1 and DNMT3B were strongly repressed in Tub-A treated oocytes. Taken together, our results indicate that HDAC6 plays important roles in chromosome condensation and kinetochore function via regulating several key histone modifications and messenger RNA transcription during oocyte meiosis.


Subject(s)
Histone Deacetylase 6/antagonists & inhibitors , Histones/metabolism , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Meiosis/drug effects , Oocytes/drug effects , RNA, Messenger/metabolism , Acetylation/drug effects , Animals , Chromosome Segregation/drug effects , Female , Histone Deacetylase Inhibitors/pharmacology , Kinetochores/drug effects , Kinetochores/metabolism , Mice , Mice, Inbred ICR , Microtubules/drug effects , Oocytes/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Tubulin/metabolism
6.
Int J Biol Sci ; 15(11): 2408-2418, 2019.
Article in English | MEDLINE | ID: mdl-31595158

ABSTRACT

Meiotic maturation of oocyte is an important process for successful fertilization, in which cytoskeletal integrality takes a significant role. The p-21 activated kinases (PAKs) belong to serine/threonine kinases that affect wide range of processes that are crucial for cell motility, survival, cell cycle, and proliferation. In this study, we used a highly selective inhibitor of PAK4, PF-3758309, to investigate the functions of PAK4 during meiotic maturation of mouse oocytes. We found that PAK4 inhibition resulted in meiotic arrest by inducing abnormal microfilament and microtubule dynamics. PAK4 inhibition impaired the microtubule stability and led to the defective kinetochore-microtubule (K-M) attachment which inevitably resulted in aneuploidy. Also, PAK4 inhibition induced abnormal acentriolar centrosome assembly during meiotic maturation. In conclusion, all these combined results suggest that PAK4 is necessary for the oocyte meiosis maturation as a regulator of cytoskeleton.


Subject(s)
Actins/metabolism , Meiosis/drug effects , Microtubules/drug effects , Microtubules/metabolism , p21-Activated Kinases/metabolism , Animals , Centrosome/drug effects , Centrosome/metabolism , Chromosome Segregation/drug effects , Female , Kinetochores/drug effects , Kinetochores/metabolism , Mice , Oocytes/drug effects , Oocytes/metabolism , Pyrazoles/pharmacology , Pyrroles/pharmacology , p21-Activated Kinases/antagonists & inhibitors
7.
PLoS One ; 14(5): e0217828, 2019.
Article in English | MEDLINE | ID: mdl-31150492

ABSTRACT

The discovery of 20 unconventional kinetochore proteins in Trypanosoma brucei has opened a new and interesting area of evolutionary research to study a biological process previously thought to be highly conserved in all eukaryotes. In addition, the discovery of novel proteins involved in a critical cellular process provides an opportunity to exploit differences between kinetoplastid and human kinetochore proteins to develop therapeutics for diseases caused by kinetoplastid parasites. Consequently, we identified two of the unconventional kinetochore proteins as key targets (the highly related kinases KKT10 and KKT19). Recombinant T. brucei KKT19 (TbKKT19) protein was produced, a peptide substrate phosphorylated by TbKKT19 identified (KKLRRTLSVA), Michaelis constants for KKLRRTLSVA and ATP were determined (179 µM and 102 µM respectively) and a robust high-throughput compatible biochemical assay developed. This biochemical assay was validated pharmacologically with inhibition by staurosporine and hypothemycin (IC50 values of 288 nM and 65 nM respectively). Surprisingly, a subsequent high-throughput screen of a kinase-relevant compound library (6,624 compounds) yielded few hits (8 hits; final hit rate 0.12%). The low hit rate observed was unusual for a kinase target, particularly when screened against a compound library enriched with kinase hinge binding scaffolds. In an attempt to understand the low hit rate a TbKKT19 homology model, based on human cdc2-like kinase 1 (CLK1), was generated. Analysis of the TbKKT19 sequence and structure revealed no obvious features that could explain the low hit rates. Further work will therefore be necessary to explore this unique kinetochore kinase as well as to assess whether the few hits identified can be developed into tool molecules or new drugs.


Subject(s)
Peptides/antagonists & inhibitors , Phosphotransferases/antagonists & inhibitors , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/diet therapy , Animals , Drug Discovery , High-Throughput Screening Assays , Humans , Kinetochores/drug effects , Kinetochores/enzymology , Peptides/chemistry , Phosphotransferases/chemistry , Phosphotransferases/genetics , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Staurosporine/pharmacology , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/parasitology , Zearalenone/analogs & derivatives , Zearalenone/pharmacology
8.
Cell Cycle ; 18(12): 1349-1363, 2019 06.
Article in English | MEDLINE | ID: mdl-31122175

ABSTRACT

During mitosis, Aurora B kinase is required for forming proper bi-oriented kinetochore-microtubule attachments. Current models suggest that tension exerted between a pair of sister-kinetochores (inter-kinetochore stretch) produces a spatial separation of Aurora B kinase from kinetochore-associated microtubule binding substrates, such as the Knl1-Mis12-Ndc80 (KMN) network, resulting in a decrease of phosphorylation and, thus, an increase of affinity for microtubules. Using Single-Molecule High-Resolution Colocalization (SHREC) microscopy analysis of the kinetochore-associated motor CENP-E, we now show that CENP-E undergoes structural rearrangements prior to and after tension generation at the kinetochore, and displays a bi-modal Gaussian distribution on a pair of bi-oriented sister kinetochores. The conformational change of CENP-E depends on its microtubule-stimulated motor motility and the highly flexible coiled-coil between its motor and kinetochore-binding tail domains. Chemical inhibition of the motor motility or perturbations of the coiled-coil domain of CENP-E increases Aurora B-mediated Ndc80 phosphorylation in a tension-independent manner. Metaphase chromosome misalignment caused by CENP-E inhibition can be rescued by chemical inhibition of Aurora B kinase. Furthermore, a pair of monotelic sister-kinetochores shows asymmetric levels of Aurora B-mediated phosphorylation in mono-polar spindles depending on CENP-E motor activity. These results collectively suggest a tension-independent mechanism to reduce Aurora B-mediated phosphorylation of outer kinetochore components in response to microtubule capture by CENP-E.


Subject(s)
Aurora Kinase B/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Aurora Kinase B/antagonists & inhibitors , Biomechanical Phenomena , Chromosomal Proteins, Non-Histone/chemistry , HeLa Cells , Humans , Kinetochores/drug effects , Microtubules/drug effects , Phosphorylation/drug effects , Protein Domains , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
9.
Article in English | MEDLINE | ID: mdl-31138411

ABSTRACT

Topoisomerase II (topo II) inhibitors are commonly used as chemotherapy to treat multiple types of cancer, though their use is also associated with the development of therapy related acute leukemias. While the chromosome-damaging effects of etoposide, a topo II poison, have been proposed to act through a threshold mechanism, little is known about the chromosome damaging effects and dose responses for the catalytic inhibitors of the enzyme. The current study was designed to further investigate the potencies and concentration-response relationships of several topoisomerase II inhibitors, including the topoisomerase II poison etoposide, as well as catalytic inhibitors aclarubicin, merbarone, ICRF-154 and ICRF-187 using both a traditional in vitro micronucleus assay as well as a flow-cytometry based version of the assay. Benchmark dose (BMD) analysis was used to identify models that best fit the data and estimate a BMD, in this case the concentration at which a one standard deviation increase above the control frequency would be expected. All of the agents tested were potent in inducing micronuclei in human lymphoblastoid TK6 cells, with significant increases seen at low micromolar, and in the cases of aclarubicin and etoposide, at low nanomolar concentrations. Use of the anti-kinetochore CREST antibody with the microscopy-based assay demonstrated that the vast majority of the micronuclei originated from chromosome breakage. In comparing the two versions of the micronucleus assay, significant increases in micronucleated cells were observed at similar or lower concentrations using the traditional microscopy-based assay. BMD modeling of the data exhibited several advantages and proved to be a valuable alternative for concentration-response analysis, producing points of departure comparable to those derived using traditional no-observed or lowest-observed genotoxic effect level (NOGEL or LOGEL) approaches.


Subject(s)
Chromosome Aberrations/drug effects , DNA Topoisomerases, Type II/metabolism , Topoisomerase II Inhibitors/pharmacology , Cell Line , Etoposide/pharmacology , Humans , Kinetochores/drug effects , Micronucleus Tests/methods
10.
Toxicol In Vitro ; 59: 115-125, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30980863

ABSTRACT

Bisphenol A [BPA, 2,2-bis-(4-hydroxyphenyl)propane] is one of the most prevalent synthetic environmental estrogens; as an endocrine disruptor, it is associated with endocrine-related cancers including breast, ovarian, and prostate. However, the mechanisms by which BPA contributes to carcinogenesis are unclear. This study aims to clarify its toxic effects on mitotic cells and investigate the molecular mechanism. In vitro effects of BPA on mitotic progression were examined by performing experiments on HeLa cells. Proteins involved in mitotic processes were detected by Western blot, live cell imaging, and immunofluorescence staining. The results showed that BPA increased chromosomal instability by perturbing mitotic processes such as bipolar spindle formation and spindle microtubule attachment to the kinetochore. BPA prolonged mitotic progression by disturbing spindle attachment and concomitant activating spindle assembly checkpoint (SAC). Mechanistically, BPA interfered proper localization of HURP to the proximal ends of spindle microtubules, Kif2a to the minus ends of spindle microtubules, and TPX2 on the mitotic spindle. This mislocalization of microtubule associated proteins (MAPs) is postulated to lead to spindle attachment failure. Furthermore, BPA caused multipolar spindle by inducing centriole overduplication and premature disengagement. Although BPA acts as an estrogen receptor (ER) agonist, mitotic defects caused by BPA occurred in an ER-independent manner. Our findings indicate that BPA may stimulate carcinogenesis not only by acting as an endocrine disruptor but also by increasing chromosomal instability during mitosis.


Subject(s)
Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Mitosis/drug effects , Phenols/toxicity , Carcinogenesis/chemically induced , Centrioles/drug effects , Chromosomal Instability/drug effects , HeLa Cells , Humans , Kinetochores/drug effects , MCF-7 Cells , Neoplasm Proteins/metabolism
11.
Environ Mol Mutagen ; 60(3): 227-242, 2019 04.
Article in English | MEDLINE | ID: mdl-30561837

ABSTRACT

The in vitro micronucleus test according to OECD Test Guideline 487 (TG 487) is widely used to investigate the genotoxic potential of drugs. Besides the identification of in vitro genotoxicants, the assay can be complemented with kinetochore staining for the differentiation between clastogens and aneugens. This differentiation constitutes a major contribution to risk assessment as especially aneugens show a threshold response. Thus, a novel method for automated MN plus kinetochore (k+) scoring by image analysis was developed based on the OECD TG 487. Compound-induced increases in MN frequency can be detected using the cytokinesis-block (cytochalasin B) method in V79 cells after 24 h in a 96-well format. Nuclei, MN, and kinetochores were labeled with nuclear counterstain and anti-kinetochore antibodies, respectively, to score MN in binuclear or multinuclear cells and to differentiate compound-induced MN by the presence of kinetochores. First, a reference data set was created by manual scoring using two clastogens and aneugens. After developing the automated scoring process, a set of 14 reference genotoxicants were studied. The automated image analysis yielded the expected results: 5/5 clastogens and 6/6 aneugens (sensitivity: 100%) as well as 3/3 non-genotoxicants (specificity: 100%) were correctly identified. Further, a threshold was determined for identifying aneugens. Based on the data for our internally characterized reference compounds, unknown compounds that induce ≥53.8% k+ MN are classified as aneugens. The current data demonstrate excellent specificity and sensitivity and the methodology is superior to manual microscopic analysis in terms of speed and throughput as well as the absence of human bias. Environ. Mol. Mutagen. 60:227-242, 2019. © 2018 Wiley Periodicals, Inc.


Subject(s)
Aneugens/pharmacology , Image Processing, Computer-Assisted/methods , Kinetochores/drug effects , Micronuclei, Chromosome-Defective/chemically induced , Micronucleus Tests/methods , Mutagens/pharmacology , Animals , Cell Line , Cricetinae , DNA Damage/drug effects , Dose-Response Relationship, Drug , Staining and Labeling/methods
12.
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
13.
Mol Biol Cell ; 29(11): 1332-1345, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29851559

ABSTRACT

During metaphase, sister chromatids are connected to microtubules extending from the opposite spindle poles via kinetochores to protein complexes on the chromosome. Kinetochores congress to the equatorial plane of the spindle and oscillate around it, with kinesin-8 motors restricting these movements. Yet, the physical mechanism underlying kinetochore movements is unclear. We show that kinetochore movements in the fission yeast Schizosaccharomyces pombe are regulated by kinesin-8-promoted microtubule catastrophe, force-induced rescue, and microtubule dynamic instability. A candidate screen showed that among the selected motors only kinesin-8 motors Klp5/Klp6 are required for kinetochore centering. Kinesin-8 accumulates at the end of microtubules, where it promotes catastrophe. Laser ablation of the spindle resulted in kinetochore movement toward the intact spindle pole in wild-type and klp5Δ cells, suggesting that kinetochore movement is driven by pulling forces. Our theoretical model with Langevin description of microtubule dynamic instability shows that kinesin-8 motors are required for kinetochore centering, whereas sensitivity of rescue to force is necessary for the generation of oscillations. We found that irregular kinetochore movements occur for a broader range of parameters than regular oscillations. Thus, our work provides an explanation for how regulation of microtubule dynamic instability contributes to kinetochore congression and the accompanying movements around the spindle center.


Subject(s)
Kinesins/metabolism , Kinetochores/metabolism , Metaphase , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/metabolism , Chromosomes, Fungal/metabolism , Hydroxyurea/pharmacology , Kinetochores/drug effects , Microtubules/drug effects , Models, Biological , Movement , Mutation/genetics , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
14.
Cell Cycle ; 17(9): 1087-1091, 2018.
Article in English | MEDLINE | ID: mdl-29895228

ABSTRACT

The spindle assembly checkpoint (SAC) ensures accurate chromosome segregation by delaying anaphase onset in response to unattached kinetochores. Anaphase is delayed by the generation of the mitotic checkpoint complex (MCC) composed of the checkpoint proteins Mad2 and BubR1/Bub3 bound to the protein Cdc20. Current models assume that MCC production is catalyzed at unattached kinetochores and that the Mad1/Mad2 complex is instrumental in the conversion of Mad2 from an open form (O-Mad2) to a closed form (C-Mad2) that can bind to Cdc20. Importantly the levels of Mad2 at kinetochores correlate with SAC activity but whether C-Mad2 at kinetochores exclusively represents its complex with Mad1 is not fully established. Here we use a recently established C-Mad2 specific monoclonal antibody to show that Cdc20 and C-Mad2 levels correlate at kinetochores and that depletion of Cdc20 reduces Mad2 but not Mad1 kinetochore levels. Importantly reintroducing wild type Cdc20 but not Cdc20 R132A, a mutant form that cannot bind Mad2, restores Mad2 levels. In agreement with this live cell imaging of fluorescent tagged Mad2 reveals that Cdc20 depletion strongly reduces Mad2 localization to kinetochores. These results support the presence of Mad2-Cdc20 complexes at kinetochores in agreement with current models of the SAC but also argue that Mad2 levels at kinetochores cannot be used as a direct readout of Mad1 levels.


Subject(s)
Cdc20 Proteins/metabolism , Cell Cycle Proteins/metabolism , Kinetochores/metabolism , Mad2 Proteins/metabolism , Anaphase , Antibodies, Monoclonal , Binding Sites , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/physiology , HeLa Cells , Humans , Kinetochores/drug effects , M Phase Cell Cycle Checkpoints/physiology , Mad2 Proteins/immunology , Nocodazole/pharmacology , Protein Folding , Quinolines/pharmacology , Thiazoles/pharmacology
15.
J Cell Sci ; 131(7)2018 04 12.
Article in English | MEDLINE | ID: mdl-29555820

ABSTRACT

During the prometaphase stage of mitosis, the cell builds a bipolar spindle of microtubules that mechanically segregates sister chromatids between two daughter cells in anaphase. The spindle assembly checkpoint (SAC) is a quality control mechanism that monitors proper attachment of microtubules to chromosome kinetochores during prometaphase. Segregation occurs only when each chromosome is bi-oriented with each kinetochore pair attached to microtubules emanating from opposite spindle poles. Overexpression of the protein kinase Aurora A is a feature of various cancers and is thought to enable tumour cells to bypass the SAC, leading to aneuploidy. Here, we took advantage of a chemical and chemical-genetic approach to specifically inhibit Aurora A kinase activity in late prometaphase. We observed that a loss of Aurora A activity directly affects SAC function, that Aurora A is essential for maintaining the checkpoint protein Mad2 on unattached kinetochores and that inhibition of Aurora A leads to loss of the SAC, even in the presence of nocodazole or Taxol. This is a new finding that should affect the way Aurora A inhibitors are used in cancer treatments.This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Aurora Kinase A/genetics , M Phase Cell Cycle Checkpoints/genetics , Mad2 Proteins/genetics , Prometaphase/genetics , Anaphase/genetics , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Cell Line, Tumor , Chromatids/genetics , Chromosome Segregation/drug effects , Gene Expression Regulation, Enzymologic/genetics , Humans , Kinetochores/drug effects , Microtubules/drug effects , Mitosis/drug effects , Mitosis/genetics , Nocodazole/pharmacology , Paclitaxel/pharmacology , Prometaphase/drug effects , Pyrimidines/pharmacology , Spindle Apparatus/genetics
16.
FASEB J ; 32(1): 342-352, 2018 01.
Article in English | MEDLINE | ID: mdl-28904021

ABSTRACT

Benzo[a]pyrene (BaP) is a ubiquitous environmental pollutant and carcinogen that is frequently found in particulate matter, with a diameter of ≤2.5 µm (PM2.5). It has been reported to interrupt the normal reproductive system, but the exact molecular basis has not been clearly defined. To understand the underlying mechanisms regarding how BaP exposure disrupts female fertility, we evaluated oocyte quality by assessing the critical regulators and events during oocyte meiotic maturation and fertilization. We found that BaP exposure compromised the mouse oocyte meiotic progression by disrupting normal spindle assembly, chromosome alignment, and kinetochore-microtubule attachment, consequently leading to the generation of aneuploid eggs. In addition, BaP administration significantly decreased the fertilization rate of mouse eggs by reducing the number of sperm binding to the zona pellucida, which was consistent with the premature cleavage of N terminus of zona pellucida sperm-binding protein 2 and precocious exocytosis of ovastacin. Furthermore, BaP exposure interfered with the gamete fusion process by perturbing the localization and protein level of Juno. Notably, we found that BaP exposure induced oxidative stress with an increased level of reactive oxygen species and apoptosis in oocytes and thereby led to the deterioration of critical regulators and events during oocyte meiotic progression and fertilization. Our data document that BaP exposure reduces female fertility via impairing oocyte maturation and fertilization ability induced by oxidative stress and early apoptosis in murine models.-Zhang, M., Miao, Y., Chen, Q., Cai, M., Dong, W., Dai, X., Lu, Y., Zhou, C., Cui, Z., Xiong, B. BaP exposure causes oocyte meiotic arrest and fertilization failure to weaken female fertility.


Subject(s)
Benzo(a)pyrene/toxicity , Fertilization/drug effects , Infertility, Female/chemically induced , Oocytes/drug effects , Oocytes/pathology , Aneugens/toxicity , Animals , Apoptosis/drug effects , Environmental Pollutants/toxicity , Female , Infertility, Female/pathology , Kinetochores/drug effects , Male , Meiosis/drug effects , Mice , Mice, Inbred ICR , Microtubules/drug effects , Oocytes/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sperm-Ovum Interactions/drug effects
17.
Curr Biol ; 28(1): 130-139.e3, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29276128

ABSTRACT

Cell division with partitioning of the genetic material should take place only when paired chromosomes named bivalents (meiosis I) or sister chromatids (mitosis and meiosis II) are correctly attached to the bipolar spindle in a tension-generating manner. For this to happen, the spindle assembly checkpoint (SAC) checks whether unattached kinetochores are present, in which case anaphase onset is delayed to permit further establishment of attachments. Additionally, microtubules are stabilized when they are attached and under tension. In mitosis, attachments not under tension activate the so-named error correction pathway depending on Aurora B kinase substrate phosphorylation. This leads to microtubule detachments, which in turn activates the SAC [1-3]. Meiotic divisions in mammalian oocytes are highly error prone, with severe consequences for fertility and health of the offspring [4, 5]. Correct attachment of chromosomes in meiosis I leads to the generation of stretched bivalents, but-unlike mitosis-not to tension between sister kinetochores, which co-orient. Here, we set out to address whether reduction of tension applied by the spindle on bioriented bivalents activates error correction and, as a consequence, the SAC. Treatment of oocytes in late prometaphase I with Eg5 kinesin inhibitor affects spindle tension, but not attachments, as we show here using an optimized protocol for confocal imaging. After Eg5 inhibition, bivalents are correctly aligned but less stretched, and as a result, Aurora-B/C-dependent error correction with microtubule detachment takes place. This loss of attachments leads to SAC activation. Crucially, SAC activation itself does not require Aurora B/C kinase activity in oocytes.


Subject(s)
Kinetochores/physiology , M Phase Cell Cycle Checkpoints/physiology , Meiosis/physiology , Oocytes/physiology , Animals , Cell Division/drug effects , Cell Division/physiology , Cysteine/analogs & derivatives , Cysteine/pharmacology , Female , Kinesins/antagonists & inhibitors , Kinetochores/drug effects , M Phase Cell Cycle Checkpoints/drug effects , Mice , Oocytes/drug effects , Paclitaxel/pharmacology , Pyrimidines/pharmacology , Thiones/pharmacology , Tubulin Modulators/pharmacology
18.
J Cell Biol ; 216(12): 3949-3957, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28978643

ABSTRACT

The spindle assembly checkpoint (SAC) prevents chromosome missegregation by coupling anaphase onset with correct chromosome attachment and tension to microtubules. It does this by generating a diffusible signal from free kinetochores into the cytoplasm, inhibiting the anaphase-promoting complex (APC). The volume in which this signal remains effective is unknown. This raises the possibility that cell volume may be the reason the SAC is weak, and chromosome segregation error-prone, in mammalian oocytes. Here, by a process of serial bisection, we analyzed the influence of oocyte volume on the ability of the SAC to inhibit bivalent segregation in meiosis I. We were able to generate oocytes with cytoplasmic volumes reduced by 86% and observed changes in APC activity consistent with increased SAC control. However, bivalent biorientation remained uncoupled from APC activity, leading to error-prone chromosome segregation. We conclude that volume is one factor contributing to SAC weakness in oocytes. However, additional factors likely uncouple chromosome biorientation with APC activity.


Subject(s)
Cell Size , Chromosome Segregation , Microtubules/metabolism , Oocytes/metabolism , Spindle Apparatus/metabolism , Anaphase-Promoting Complex-Cyclosome/genetics , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , Female , Gene Expression Regulation , Gonadotropins, Equine/pharmacology , Kinetochores/drug effects , Kinetochores/metabolism , Kinetochores/ultrastructure , Meiosis/genetics , Mice , Mice, Inbred C57BL , Microinjections , Microtubules/drug effects , Microtubules/ultrastructure , Milrinone/pharmacology , Nocodazole/pharmacology , Oocytes/drug effects , Oocytes/ultrastructure , RNA, Complementary/genetics , RNA, Complementary/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/ultrastructure
19.
Development ; 144(19): 3475-3486, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28851706

ABSTRACT

Mouse oocytes carrying DNA damage arrest in meiosis I, thereby preventing creation of embryos with deleterious mutations. The arrest is dependent on activation of the spindle assembly checkpoint, which results in anaphase-promoting complex (APC) inhibition. However, little is understood about how this checkpoint is engaged following DNA damage. Here, we find that within minutes of DNA damage checkpoint proteins are assembled at the kinetochore, not at damage sites along chromosome arms, such that the APC is fully inhibited within 30 min. Despite this robust response, there is no measurable loss in k-fibres, or tension across the bivalent. Through pharmacological inhibition we observed that the response is dependent on Mps1 kinase, aurora kinase and Haspin. Using oocyte-specific knockouts we find the response does not require the DNA damage response kinases ATM or ATR. Furthermore, checkpoint activation does not occur in response to DNA damage in fully mature eggs during meiosis II, despite the divisions being separated by just a few hours. Therefore, mouse oocytes have a unique ability to sense DNA damage rapidly by activating the checkpoint at their kinetochores.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Meiosis , Oocytes/cytology , Oocytes/metabolism , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , Aurora Kinases/metabolism , Centromere/drug effects , Centromere/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kinetochores/drug effects , M Phase Cell Cycle Checkpoints/drug effects , Meiosis/drug effects , Mice , Models, Biological , Oocytes/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism
20.
Cancer Lett ; 403: 74-85, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28602975

ABSTRACT

Neuroblastoma is a biologically and clinically heterogeneous pediatric malignancy that includes a high-risk subset for which new therapeutic agents are urgently required. As well as MYCN amplification, activating point mutations of ALK and NRAS are associated with high-risk and relapsing neuroblastoma. As both ALK and RAS signal through the MEK/ERK pathway, we sought to evaluate two previously reported inhibitors of ETS-related transcription factors, which are transcriptional mediators of the Ras-MEK/ERK pathway in other cancers. Here we show that YK-4-279 suppressed growth and triggered apoptosis in nine neuroblastoma cell lines, while BRD32048, another ETV1 inhibitor, was ineffective. These results suggest that YK-4-279 acts independently of ETS-related transcription factors. Further analysis reveals that YK-4-279 induces mitotic arrest in prometaphase, resulting in subsequent cell death. Mechanistically, we show that YK-4-279 inhibits the formation of kinetochore microtubules, with treated cells showing a broad range of abnormalities including multipolar, fragmented and unseparated spindles, together leading to disrupted progression through mitosis. Notably, YK-4-279 does not affect microtubule acetylation, unlike the conventional mitotic poisons paclitaxel and vincristine. Consistent with this, we demonstrate that YK-4-279 overcomes vincristine-induced resistance in two neuroblastoma cell-line models. Furthermore, combinations of YK-4-279 with vincristine, paclitaxel or the Aurora kinase A inhibitor MLN8237/Alisertib show strong synergy, particularly at low doses. Thus, YK-4-279 could potentially be used as a single-agent or in combination therapies for the treatment of high-risk and relapsing neuroblastoma, as well as other cancers.


Subject(s)
Antimitotic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Indoles/pharmacology , Mitosis/drug effects , Neuroblastoma/drug therapy , Apoptosis/drug effects , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/metabolism , Azepines/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Humans , Inhibitory Concentration 50 , Kinetochores/drug effects , Kinetochores/pathology , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Paclitaxel/pharmacology , Prometaphase/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA Interference , Signal Transduction/drug effects , Spindle Apparatus/drug effects , Spindle Apparatus/pathology , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vincristine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...