Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.471
Filter
1.
Elife ; 112022 07 27.
Article in English | MEDLINE | ID: mdl-35894209

ABSTRACT

During cell division, kinetochore microtubules (KMTs) provide a physical linkage between the chromosomes and the rest of the spindle. KMTs in mammalian cells are organized into bundles, so-called kinetochore-fibers (k-fibers), but the ultrastructure of these fibers is currently not well characterized. Here, we show by large-scale electron tomography that each k-fiber in HeLa cells in metaphase is composed of approximately nine KMTs, only half of which reach the spindle pole. Our comprehensive reconstructions allowed us to analyze the three-dimensional (3D) morphology of k-fibers and their surrounding MTs in detail. We found that k-fibers exhibit remarkable variation in circumference and KMT density along their length, with the pole-proximal side showing a broadening. Extending our structural analysis then to other MTs in the spindle, we further observed that the association of KMTs with non-KMTs predominantly occurs in the spindle pole regions. Our 3D reconstructions have implications for KMT growth and k-fiber self-organization models as covered in a parallel publication applying complementary live-cell imaging in combination with biophysical modeling (Conway et al., 2022). Finally, we also introduce a new visualization tool allowing an interactive display of our 3D spindle data that will serve as a resource for further structural studies on mitosis in human cells.


Subject(s)
Kinetochores , Spindle Apparatus , Animals , Chromosomes , HeLa Cells , Humans , Mammals , Metaphase , Microtubules/ultrastructure , Spindle Apparatus/ultrastructure
2.
Theriogenology ; 186: 60-69, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35430549

ABSTRACT

Sirtuins play an important role in female mammalian reproductive function, participating in folliculogenesis and oocyte maturation. Studies exploring the consequences of inhibition/deletion of a specific sirtuin (SIRT) have demonstrated a deleterious effect on follicular growth, oocyte maturation, fertilization rates and embryo development, suggesting that sirtuins must have a relevant role in these processes. However, the exact mechanisms behind sirtuin function are still unclear. Most of the knowledge currently available derives from mouse studies and the literature is scarce in other species. So far, there is insufficient information about the subcellular localization of sirtuins during bovine meiosis, which would contribute to understanding the role and participation of sirtuins in the process of oocyte maturation, due to the close relation between location and function. Using in vitro maturation (IVM) of bovine oocytes we comprehensively documented and illustrated the subcellular localization pattern and distribution of SIRT1, 2 and 3 during meiotic progression. Moreover, we also detailed and quantified the colocalization of those sirtuins with the meiotic spindle, from the germinal-vesicle (GV)-stage until the Metaphase-II (MII)-stage. Our study demonstrated an increase in the expression of SIRT1, 2 and 3 during in vitro oocyte maturation and, for the first time, colocalization of SIRT1, 2 and 3 with both metaphase-I and metaphase-II spindles. These findings suggest that all three sirtuins may have a role in meiotic spindle assembly and microtubule dynamics in the bovine model. In addition, we have demonstrated the nuclear presence of SIRT1 and SIRT2 in the GV-stage. The apparent perinucleolar location of SIRT2 suggests that SIRT2 may shuttle into the nucleus at the GV-stage to regulate heterochromatin. This study reinforces the value of sirtuins during in vitro bovine meiotic progression and indicates potential molecular targets to improve maturation rates and embryo development.


Subject(s)
Sirtuin 1 , Sirtuin 2 , Animals , Cattle , Female , In Vitro Oocyte Maturation Techniques/veterinary , Mammals , Meiosis , Oocytes/physiology , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuin 2/genetics , Sirtuin 2/metabolism , Sirtuin 3/metabolism , Spindle Apparatus/ultrastructure
3.
Science ; 375(6581): eabj3944, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35143306

ABSTRACT

Human oocytes are prone to assembling meiotic spindles with unstable poles, which can favor aneuploidy in human eggs. The underlying causes of spindle instability are unknown. We found that NUMA (nuclear mitotic apparatus protein)-mediated clustering of microtubule minus ends focused the spindle poles in human, bovine, and porcine oocytes and in mouse oocytes depleted of acentriolar microtubule-organizing centers (aMTOCs). However, unlike human oocytes, bovine, porcine, and aMTOC-free mouse oocytes have stable spindles. We identified the molecular motor KIFC1 (kinesin superfamily protein C1) as a spindle-stabilizing protein that is deficient in human oocytes. Depletion of KIFC1 recapitulated spindle instability in bovine and aMTOC-free mouse oocytes, and the introduction of exogenous KIFC1 rescued spindle instability in human oocytes. Thus, the deficiency of KIFC1 contributes to spindle instability in human oocytes.


Subject(s)
Cell Cycle Proteins/metabolism , Kinesins/deficiency , Oocytes/physiology , Oocytes/ultrastructure , Spindle Apparatus/physiology , Spindle Poles/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Cattle , Dynactin Complex/metabolism , Dyneins/metabolism , Female , Humans , Kinesins/genetics , Kinesins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/physiology , Microtubule-Organizing Center/ultrastructure , Microtubules/metabolism , Recombinant Proteins/metabolism , Spindle Apparatus/ultrastructure , Spindle Poles/ultrastructure , Swine
4.
Cells ; 10(10)2021 10 05.
Article in English | MEDLINE | ID: mdl-34685637

ABSTRACT

The centrosome of Dictyostelium amoebae contains no centrioles and consists of a cylindrical layered core structure surrounded by a corona harboring microtubule-nucleating γ-tubulin complexes. It is the major centrosomal model beyond animals and yeasts. Proteomics, protein interaction studies by BioID and superresolution microscopy methods led to considerable progress in our understanding of the composition, structure and function of this centrosome type. We discuss all currently known components of the Dictyostelium centrosome in comparison to other centrosomes of animals and yeasts.


Subject(s)
Centrosome/metabolism , Dictyostelium/metabolism , Cell Nucleus/metabolism , Centrosome/ultrastructure , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
5.
Elife ; 102021 08 18.
Article in English | MEDLINE | ID: mdl-34406118

ABSTRACT

DNA loop extrusion by condensins and decatenation by DNA topoisomerase II (topo II) are thought to drive mitotic chromosome compaction and individualization. Here, we reveal that the linker histone H1.8 antagonizes condensins and topo II to shape mitotic chromosome organization. In vitro chromatin reconstitution experiments demonstrate that H1.8 inhibits binding of condensins and topo II to nucleosome arrays. Accordingly, H1.8 depletion in Xenopus egg extracts increased condensins and topo II levels on mitotic chromatin. Chromosome morphology and Hi-C analyses suggest that H1.8 depletion makes chromosomes thinner and longer through shortening the average loop size and reducing the DNA amount in each layer of mitotic loops. Furthermore, excess loading of condensins and topo II to chromosomes by H1.8 depletion causes hyper-chromosome individualization and dispersion. We propose that condensins and topo II are essential for chromosome individualization, but their functions are tuned by the linker histone to keep chromosomes together until anaphase.


Subject(s)
Chromatin/metabolism , Chromosomes/genetics , DNA Topoisomerases, Type II/genetics , Histones/genetics , Adenosine Triphosphatases/metabolism , Animals , Cell Extracts/chemistry , Chromosomes/ultrastructure , DNA-Binding Proteins/metabolism , Female , Models, Biological , Multiprotein Complexes/metabolism , Oocytes/chemistry , Oocytes/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/pathology , Spindle Apparatus/ultrastructure , Xenopus laevis
6.
J Cell Biol ; 220(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34287649

ABSTRACT

Aurora A is a serine/threonine kinase essential for mitotic entry and spindle assembly. Recent molecular studies have revealed the existence of multiple, distinct mechanisms of Aurora A activation, each occurring at specific subcellular locations, optimized for cellular context, and primed by signaling events including phosphorylation and oxidation.


Subject(s)
Aurora Kinase A/genetics , Cell Cycle Proteins/genetics , Microtubule-Associated Proteins/genetics , Mitosis , Protein Processing, Post-Translational , Allosteric Regulation , Animals , Aurora Kinase A/metabolism , Cell Cycle Proteins/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/enzymology , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , Oxidation-Reduction , Phosphorylation , Protein Binding , Signal Transduction , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
7.
Mol Reprod Dev ; 88(5): 349-361, 2021 05.
Article in English | MEDLINE | ID: mdl-33843103

ABSTRACT

This study investigated the effect of the antioxidant dieckol, a component of Ecklonia cava, on maturation and developmental competence of porcine oocytes exposed to oxidative stress in vitro. Oocytes were matured in in vitro maturation (IVM) medium containing various concentrations of dieckol. The blastocyst formation rate was highest in the 0.5 µM dieckol-treated (0.5 DEK) group. The reactive oxygen species level was decreased, and the level of glutathione and expression of antioxidant genes (NFE2L, SOD1, and SOD2) at metaphase II were increased in the 0.5 DEK group. Abnormal spindle organization and chromosome misalignment were prevented in the 0.5 DEK group. Expression of maternal markers (CCNB1 and MOS) and activity of p44/42 mitogen-activated protein kinase were increased in the 0.5 DEK group. After parthenogenetic activation, the total number of cells per blastocyst was increased and the percentage of apoptotic cells was decreased in the 0.5 DEK group. Expression of development-related genes (CX45, CDX2, POU5F1, and NANOG), antiapoptotic genes (BCL2L1 and BIRC5), and a proapoptotic gene (CASP3) were altered in the 0.5 DEK group. These results indicate that the antioxidant dieckol improves IVM and subsequent development of porcine oocytes and can be used to improve the quality of oocytes under peroxidation experimental conditions.


Subject(s)
Antioxidants/pharmacology , Benzofurans/pharmacology , Embryonic Development/drug effects , Oocytes/drug effects , Oxidative Stress/drug effects , Parthenogenesis/drug effects , Animals , Antioxidants/administration & dosage , Benzofurans/administration & dosage , Blastocyst/cytology , Chromosome Positioning/drug effects , Dose-Response Relationship, Drug , Embryo Culture Techniques , Female , Gene Expression Regulation, Developmental/drug effects , Glutathione/metabolism , In Vitro Oocyte Maturation Techniques , MAP Kinase Signaling System/drug effects , Meiosis , Oocytes/metabolism , Phaeophyceae/chemistry , Reactive Oxygen Species/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/ultrastructure , Swine
8.
Nucleic Acids Res ; 49(4): 2027-2043, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33476374

ABSTRACT

Dysfunction of splicing factors often result in abnormal cell differentiation and apoptosis, especially in neural tissues. Mutations in pre-mRNAs processing factor 31 (PRPF31) cause autosomal dominant retinitis pigmentosa, a progressive retinal degeneration disease. The transcriptome-wide splicing events specifically regulated by PRPF31 and their biological roles in the development and maintenance of retina are still unclear. Here, we showed that the differentiation and viability of retinal progenitor cells (RPCs) are severely perturbed in prpf31 knockout zebrafish when compared with other tissues at an early embryonic stage. At the cellular level, significant mitotic arrest and DNA damage were observed. These defects could be rescued by the wild-type human PRPF31 rather than the disease-associated mutants. Further bioinformatic analysis and experimental verification uncovered that Prpf31 deletion predominantly causes the skipping of exons with a weak 5' splicing site. Moreover, genes necessary for DNA repair and mitotic progression are most enriched among the differentially spliced events, which may explain the cellular and tissular defects in prpf31 mutant retinas. This is the first time that Prpf31 is demonstrated to be essential for the survival and differentiation of RPCs during retinal neurogenesis by specifically modulating the alternative splicing of genes involved in DNA repair and mitosis.


Subject(s)
Alternative Splicing , Neural Stem Cells/metabolism , Neurogenesis/genetics , Retina/embryology , Zebrafish Proteins/physiology , Animals , Apoptosis , CRISPR-Cas Systems , Cell Survival , DNA Damage , DNA Repair , Exons , Gene Knockout Techniques , M Phase Cell Cycle Checkpoints , Neural Stem Cells/cytology , Retinal Neurons/cytology , Retinal Neurons/metabolism , Spindle Apparatus/ultrastructure , Tumor Suppressor Protein p53/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Folia Biol (Praha) ; 67(5-6): 208-212, 2021.
Article in English | MEDLINE | ID: mdl-35439854

ABSTRACT

The effects of prostaglandin F2α on the cytoskeleton and membrane organelles of oocytes was investigated by culturing ovulated mouse oocytes in its presence (50 or 100 ng/ml) for 3 h. Tubulin, fibrillar actin, membranes and chromatin were visualized by specific antibodies, phalloidin, lipophilic dye DiOC6 and Hoechst 33342, respectively. Control oocytes were characterized by a meiotic spindle with chromosomes aligned at its equator, and a cortical layer of microfilaments with an actin cap. Intracellular membranes were localized mostly in the central region in metaphase I and in a broader volume, but still excluding the cell periphery, in metaphase II, and were slightly concentrated around the chromosomes. In oocytes treated with 50 ng/ml prostaglandin, cortical actin staining was diminished, the membrane distribution was clustered, and chromosomes showed signs of misalignment despite the apparently preserved spindle. In cells treated with 100 ng/ml prostaglandin, both the spindle and the actin cortex had degenerated or disappeared as microscopic objects. Metaphase plates were on average broader and more disorganized than in the 50 ng/ml group, and the distribution of membrane organelles had become uniform. These effects, to our knowledge observed for the first time, did not require presence of the cumulus during the incubation. They could be regarded as acceleration of the oocyte postovulatory aging, in which cytoskeletal deterioration seemed to have a leading role.


Subject(s)
Actins , Dinoprost , Actins/metabolism , Animals , Dinoprost/metabolism , Meiosis , Metaphase , Mice , Oocytes/metabolism , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
10.
Semin Cell Dev Biol ; 107: 91-102, 2020 11.
Article in English | MEDLINE | ID: mdl-32747191

ABSTRACT

During mitosis microtubules self-organize to form a bipolar mitotic spindle structure, which positions the sister chromatids on the spindle mid-plane and separates them afterwards. Previous studies have identified many spindle associated proteins. Yet, we do not fully understand how these nanoscopic proteins lead to force generation through interactions of individual microtubules, motor proteins and chromosomes, and how a large number of these local interactions ultimately determine the structure and mechanics of the spindle in micron scale. Here we review the current understanding and open questions related to the structure and mechanics of the mitotic spindle. We then discuss how a combination of electron microscopy and computational modeling can be used to tackle some of these open questions.


Subject(s)
Spindle Apparatus/metabolism , Animals , Biomechanical Phenomena , Humans , Models, Biological , Polymerization , Rheology , Spindle Apparatus/ultrastructure
11.
Biosci Biotechnol Biochem ; 84(8): 1667-1680, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32441227

ABSTRACT

The cAMP-dependent protein kinase Pka1 is known as a regulator of glycogenesis, transition into meiosis, proper chromosome segregation, and stress responses in Schizosaccharomyces pombe. We demonstrated that both the cAMP/PKA pathway and glucose limitation play roles in appropriate spindle formation. Overexpression of Mal3 (1-308), an EB1 family protein, caused growth defects, increased 4C DNA content, and induced monopolar spindle formation. Overproduction of a high-affinity microtubule binding mutant (Q89R) and a recombinant protein possessing the CH and EB1 domains (1-241) both resulted in more severe phenotypes than Mal3 (1-308). Loss of functional Pka1 and glucose limitation rescued the phenotypes of Mal3-overexpressing cells, whereas deletion of Tor1 or Ssp2 did not. Growth defects and monopolar spindle formation in a kinesin-5 mutant, cut7-446, was partially rescued by pka1 deletion or glucose limitation. These findings suggest that Pka1 and glucose limitation regulate proper spindle formation in Mal3-overexpressing cells and the cut7-446 mutant.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Gene Expression Regulation, Fungal , Glucose/deficiency , Kinesins/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Spindle Apparatus/metabolism , Amino Acid Substitution , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/deficiency , DNA, Fungal/genetics , DNA, Fungal/metabolism , Gene Deletion , Glucose/pharmacology , Kinesins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitosis/drug effects , Mutation , Phenotype , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Schizosaccharomyces/drug effects , Schizosaccharomyces/growth & development , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/ultrastructure
12.
J Cell Biol ; 219(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32356865

ABSTRACT

The Aurora B chromosomal passenger complex (CPC) is a conserved regulator of mitosis. Its functions require localization first to the chromosome arms and then centromeres in mitosis and subsequently the central spindle in anaphase. Here, we analyze the requirements for core CPC subunits, survivin and INCENP, and the mitotic kinesin-like protein 2 (MKLP2) in targeting to these distinct localizations. Centromere recruitment of the CPC requires interaction of survivin with histone H3 phosphorylated at threonine 3, and we provide a complete structure of this assembly. Furthermore, we show that the INCENP RRKKRR-motif is required for both centromeric localization of the CPC in metaphase and MKLP2-dependent transport in anaphase. MKLP2 and DNA bind competitively to this motif, and INCENP T59 phosphorylation acts as a switch preventing MKLP2 binding in metaphase. In anaphase, CPC binding promotes the microtubule-dependent ATPase activity of MKLP2. These results explain how centromere targeting of the CPC in mitosis is coupled to its movement to the central spindle in anaphase.


Subject(s)
Anaphase , Aurora Kinase B/metabolism , Chromatin/metabolism , Histones/metabolism , Kinesins/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Aurora Kinase B/chemistry , Aurora Kinase B/genetics , Binding, Competitive , Centromere/metabolism , Centromere/ultrastructure , Chromatin/ultrastructure , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , HeLa Cells , Histones/chemistry , Histones/genetics , Humans , Kinesins/chemistry , Kinesins/genetics , Metaphase , Microtubules/metabolism , Microtubules/ultrastructure , Models, Molecular , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Transport , Sequence Alignment , Sequence Homology, Amino Acid , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Survivin/chemistry , Survivin/genetics , Survivin/metabolism
13.
Nat Cell Biol ; 22(3): 321-331, 2020 03.
Article in English | MEDLINE | ID: mdl-32123335

ABSTRACT

CRISPR-Cas9 technology has revolutionized genome editing and is applicable to the organoid field. However, precise integration of exogenous DNA sequences into human organoids is lacking robust knock-in approaches. Here, we describe CRISPR-Cas9-mediated homology-independent organoid transgenesis (CRISPR-HOT), which enables efficient generation of knock-in human organoids representing different tissues. CRISPR-HOT avoids extensive cloning and outperforms homology directed repair (HDR) in achieving precise integration of exogenous DNA sequences into desired loci, without the necessity to inactivate TP53 in untransformed cells, which was previously used to increase HDR-mediated knock-in. CRISPR-HOT was used to fluorescently tag and visualize subcellular structural molecules and to generate reporter lines for rare intestinal cell types. A double reporter-in which the mitotic spindle was labelled by endogenously tagged tubulin and the cell membrane by endogenously tagged E-cadherin-uncovered modes of human hepatocyte division. Combining tubulin tagging with TP53 knock-out revealed that TP53 is involved in controlling hepatocyte ploidy and mitotic spindle fidelity. CRISPR-HOT simplifies genome editing in human organoids.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Gene Knock-In Techniques/methods , Organoids/cytology , Hepatocytes/cytology , Hepatocytes/ultrastructure , Humans , Intestines/cytology , Liver/cytology , Organoids/ultrastructure , Spindle Apparatus/ultrastructure , Tumor Suppressor Protein p53/physiology
14.
Wiley Interdiscip Rev Dev Biol ; 9(5): e376, 2020 09.
Article in English | MEDLINE | ID: mdl-32003549

ABSTRACT

Cell division without growth results in progressive cell size reductions during early embryonic development. How do the sizes of intracellular structures and organelles scale with cell size and what are the functional implications of such scaling relationships? Model organisms, in particular Caenorhabditis elegans worms, Drosophila melanogaster flies, Xenopus laevis frogs, and Mus musculus mice, have provided insights into developmental size scaling of the nucleus, mitotic spindle, and chromosomes. Nuclear size is regulated by nucleocytoplasmic transport, nuclear envelope proteins, and the cytoskeleton. Regulators of microtubule dynamics and chromatin compaction modulate spindle and mitotic chromosome size scaling, respectively. Developmental scaling relationships for membrane-bound organelles, like the endoplasmic reticulum, Golgi, mitochondria, and lysosomes, have been less studied, although new imaging approaches promise to rectify this deficiency. While models that invoke limiting components and dynamic regulation of assembly and disassembly can account for some size scaling relationships in early embryos, it will be exciting to investigate the contribution of newer concepts in cell biology such as phase separation and interorganellar contacts. With a growing understanding of the underlying mechanisms of organelle size scaling, future studies promise to uncover the significance of proper scaling for cell function and embryonic development, as well as how aberrant scaling contributes to disease. This article is categorized under: Establishment of Spatial and Temporal Patterns > Regulation of Size, Proportion, and Timing Early Embryonic Development > Fertilization to Gastrulation Comparative Development and Evolution > Model Systems.


Subject(s)
Embryonic Development , Organelle Size , Animals , Cell Membrane Structures/metabolism , Cell Membrane Structures/ultrastructure , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
15.
Fertil Steril ; 113(1): 105-113.e1, 2020 01.
Article in English | MEDLINE | ID: mdl-31739977

ABSTRACT

OBJECTIVE: To evaluate oocyte meiotic spindle (OMS) morphology at intracytoplasmic sperm injection (ICSI) as a predictor of blastocyst ploidy and whether OMS morphology could aid standard morphology-based blastocyst selection. DESIGN: Prospective cohort study. SETTING: In vitro fertilization clinic. PATIENT(S): Patients undergoing ICSI cycles with an intention to perform preimplantation genetic testing for aneuploidy (PGT-A) from October 2014 to December 2017. INTERVENTION(S): The OMS was visualized with the use of polarized light microscopy at the time of ICSI and the morphology classified as normal, dysmorphic, translucent, not visible, or in telophase. Blastocyst biopsy for PGT-A was performed on embryos with suitable development. MAIN OUTCOME MEASURE(S): The association of OMS morphology with the resulting blastocyst ploidy was evaluated on an "intention-to-treat" (ITT) and an "as-treated analysis" (ATA) basis. RESULT(S): The morphology of 2,056 OMSs were classified. A strong association of OMS morphology with fertilization, cleavage to at least 6 cells on day 3, and good/top-quality blastocyst formation was present. Normal OMS was positively associated with blastocyst euploidy compared with all other OMS types combined, per either ITT or ATA. Even after controlling for female age, blastocyst quality, and developmental stage, the presence of a normal OMS was strongly associated with the probability of blastocyst euploidy. CONCLUSION(S): OMS morphology is a predictive marker of blastocyst ploidy and can potentially aid standard morphology-based blastocyst selection.


Subject(s)
Blastocyst/physiology , Oocytes/physiology , Ploidies , Spindle Apparatus/physiology , Adult , Blastocyst/ultrastructure , Cohort Studies , Female , Humans , Oocytes/ultrastructure , Predictive Value of Tests , Pregnancy , Prospective Studies , Sperm Injections, Intracytoplasmic/methods , Spindle Apparatus/ultrastructure
16.
J Mol Cell Biol ; 12(8): 654-665, 2020 08 01.
Article in English | MEDLINE | ID: mdl-31174204

ABSTRACT

Error-free cell division depends on the accurate assembly of the spindle midzone from dynamic spindle microtubules to ensure chromatid segregation during metaphase-anaphase transition. However, the mechanism underlying the key transition from the mitotic spindle to central spindle before anaphase onset remains elusive. Given the prevalence of chromosome instability phenotype in gastric tumorigenesis, we developed a strategy to model context-dependent cell division using a combination of light sheet microscope and 3D gastric organoids. Light sheet microscopic image analyses of 3D organoids showed that CENP-E inhibited cells undergoing aberrant metaphase-anaphase transition and exhibiting chromosome segregation errors during mitosis. High-resolution real-time imaging analyses of 2D cell culture revealed that CENP-E inhibited cells undergoing central spindle splitting and chromosome instability phenotype. Using biotinylated syntelin as an affinity matrix, we found that CENP-E forms a complex with PRC1 in mitotic cells. Chemical inhibition of CENP-E in metaphase by syntelin prevented accurate central spindle assembly by perturbing temporal assembly of PRC1 to the midzone. Thus, CENP-E-mediated PRC1 assembly to the central spindle constitutes a temporal switch to organize dynamic kinetochore microtubules into stable midzone arrays. These findings reveal a previously uncharacterized role of CENP-E in temporal control of central spindle assembly. Since CENP-E is absent from yeast, we reasoned that metazoans evolved an elaborate central spindle organization machinery to ensure accurate sister chromatid segregation during anaphase and cytokinesis.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Mitosis , Spindle Apparatus/metabolism , Anaphase , HEK293 Cells , HeLa Cells , Humans , Models, Biological , Organoids/metabolism , Spindle Apparatus/ultrastructure , Stomach/cytology , Time Factors
17.
Elife ; 82019 12 13.
Article in English | MEDLINE | ID: mdl-31833472

ABSTRACT

During organogenesis, precise control of spindle orientation balances proliferation and differentiation. In the developing murine epidermis, planar and perpendicular divisions yield symmetric and asymmetric fate outcomes, respectively. Classically, division axis specification involves centrosome migration and spindle rotation, events occurring early in mitosis. Here, we identify a novel orientation mechanism which corrects erroneous anaphase orientations during telophase. The directionality of reorientation correlates with the maintenance or loss of basal contact by the apical daughter. While the scaffolding protein LGN is known to determine initial spindle positioning, we show that LGN also functions during telophase to reorient oblique divisions toward perpendicular. The fidelity of telophase correction also relies on the tension-sensitive adherens junction proteins vinculin, α-E-catenin, and afadin. Failure of this corrective mechanism impacts tissue architecture, as persistent oblique divisions induce precocious, sustained differentiation. The division orientation plasticity provided by telophase correction may enable progenitors to adapt to local tissue needs.


Subject(s)
Epidermal Cells/cytology , Epithelial Cells/cytology , Telophase/physiology , Actomyosin/physiology , Anaphase , Animals , Cell Self Renewal , Cell Shape , Cytoskeleton/ultrastructure , Epidermis/embryology , Female , Genes, Reporter , Intravital Microscopy , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Protein Conformation , RNA Interference , Spindle Apparatus/ultrastructure , Vinculin/genetics , Vinculin/physiology , alpha Catenin/genetics , alpha Catenin/physiology
18.
Proc Natl Acad Sci U S A ; 116(47): 23691-23697, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685623

ABSTRACT

Predictive biomarkers for tumor response to neoadjuvant chemotherapy are needed in breast cancer. This study investigates the predictive value of 280 genes encoding proteins that regulate microtubule assembly and function. By analyzing 3 independent multicenter randomized cohorts of breast cancer patients, we identified 17 genes that are differentially regulated in tumors achieving pathological complete response (pCR) to neoadjuvant chemotherapy. We focused on the MTUS1 gene, whose major product, ATIP3, is a microtubule-associated protein down-regulated in aggressive breast tumors. We show here that low levels of ATIP3 are associated with an increased pCR rate, pointing to ATIP3 as a predictive biomarker of breast tumor chemosensitivity. Using preclinical models of patient-derived xenografts and 3-dimensional models of breast cancer cell lines, we show that low ATIP3 levels sensitize tumors to the effects of taxanes but not DNA-damaging agents. ATIP3 silencing improves the proapoptotic effects of paclitaxel and induces mitotic abnormalities, including centrosome amplification and multipolar spindle formation, which results in chromosome missegregation leading to aneuploidy. As shown by time-lapse video microscopy, ATIP3 depletion exacerbates cytokinesis failure and mitotic death induced by low doses of paclitaxel. Our results favor a mechanism by which the combination of ATIP3 deficiency and paclitaxel treatment induces excessive aneuploidy, which in turn results in elevated cell death. Together, these studies highlight ATIP3 as an important regulator of mitotic integrity and a useful predictive biomarker for a population of chemoresistant breast cancer patients.


Subject(s)
Aneuploidy , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Neoplasm Proteins/physiology , Paclitaxel/pharmacology , Tumor Suppressor Proteins/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cytokinesis/drug effects , DNA, Neoplasm/drug effects , Gene Expression Profiling , Heterografts , Humans , Microtubules/drug effects , Microtubules/physiology , Multicenter Studies as Topic/statistics & numerical data , Neoadjuvant Therapy , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , RNA Interference , Randomized Controlled Trials as Topic/statistics & numerical data , Spindle Apparatus/drug effects , Spindle Apparatus/ultrastructure , Taxoids/pharmacology , Time-Lapse Imaging , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
19.
Nat Commun ; 10(1): 4651, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604948

ABSTRACT

Mammalian oocytes assemble a bipolar acentriolar microtubule spindle to segregate chromosomes during asymmetric division. There is increasing evidence that actin in the spindle interior not only participates in spindle migration and positioning but also protects oocytes from chromosome segregation errors leading to aneuploidy. Here we show that actin is an integral component of the meiotic machinery that closely interacts with microtubules during all major events of human oocyte maturation from the time point of spindle assembly till polar body extrusion and metaphase arrest. With the aid of drugs selectively affecting cytoskeleton dynamics and transiently disturbing the integrity of the two cytoskeleton systems, we identify interdependent structural rearrangements indicative of a close communication between actin and microtubules as fundamental feature of human oocytes. Our data support a model of actin-microtubule interplay that is essential for bipolar spindle assembly and correct partitioning of the nuclear genome in human oocyte meiosis.


Subject(s)
Actins/physiology , Chromosome Segregation/physiology , Oocytes/cytology , Spindle Apparatus/metabolism , Female , Humans , Meiosis , Microtubules/physiology , Oocytes/ultrastructure , Polar Bodies/cytology , Polar Bodies/metabolism , Polar Bodies/ultrastructure , Spindle Apparatus/ultrastructure , Tubulin/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...