Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
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
2.
NPJ Syst Biol Appl ; 7(1): 13, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637746

ABSTRACT

The GTPase Cdc42 is the master regulator of eukaryotic cell polarisation. During this process, the active form of Cdc42 is accumulated at a particular site on the cell membrane called the pole. It is believed that the accumulation of the active Cdc42 resulting in a pole is driven by a combination of activation-inactivation reactions and diffusion. It has been proposed using mathematical modelling that this is the result of diffusion-driven instability, originally proposed by Alan Turing. In this study, we developed, analysed and validated a 3D bulk-surface model of the dynamics of Cdc42. We show that the model can undergo both classic and non-classic Turing instability by deriving necessary conditions for which this occurs and conclude that the non-classic case can be viewed as a limit case of the classic case of diffusion-driven instability. Using three-dimensional Spatio-temporal simulation we predicted pole size and time to polarisation, suggesting that cell polarisation is mainly driven by the reaction strength parameter and that the size of the pole is determined by the relative diffusion.


Subject(s)
Cell Polarity/physiology , Spindle Poles/physiology , cdc42 GTP-Binding Protein/metabolism , Biological Phenomena , Computer Simulation , Diffusion , Linear Models , Models, Biological , Models, Theoretical , cdc42 GTP-Binding Protein/physiology
3.
Curr Genet ; 66(4): 719-727, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32266430

ABSTRACT

Although cell division is usually portrayed as an equitable process by which a progenitor cell originates two identical daughter cells, there are multiple examples of asymmetric divisions that generate two cells that differ in their content, morphology and/or proliferative potential. The capacity of the cells to generate asymmetry during their division is of paramount biological relevance, playing essential roles during embryonic development, cellular regeneration and tissue morphogenesis. Problems with the proper establishment of asymmetry and polarity during cell division can give rise to cancer and neurodevelopmental disorders, as well as to also accelerate cellular aging. Interestingly, the microtubule organizing centers that orchestrate the formation of the mitotic spindle have been described among the cellular structures that can be differentially allocated during asymmetric cell divisions. This mini-review focuses on recent research from our group and others uncovering a role for the non-random distribution of the spindle-associated microtubule organizing centers in the differential distribution of aging factors during asymmetric mitoses and therefore in the maintenance of the replicative lifespan of the cells.


Subject(s)
Asymmetric Cell Division , Neurodegenerative Diseases/pathology , Spindle Poles/physiology , Aging , Animals , Carcinogenesis , Centrosome , Female , Humans , Male , Mitosis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Spindle Apparatus , Stem Cells/cytology , Stem Cells/physiology
5.
Curr Biol ; 29(22): 3825-3837.e3, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31679937

ABSTRACT

Separation of duplicated spindle poles is the first step in forming the mitotic spindle. Kinesin-5 crosslinks and slides anti-parallel microtubules (MTs), but it is unclear how these two activities contribute to the first steps in spindle formation. In this study, we report that in monopolar spindles, the duplicated spindle poles snap apart in a fast and irreversible step that produces a nascent bipolar spindle. Using mutations in Kinesin-5 that inhibit microtubule sliding, we show that the fast, irreversible pole separation is primarily driven by microtubule crosslinking. Electron tomography revealed microtubule pairs in monopolar spindles have short overlaps that intersect at high angles and are unsuited for ensemble Kinesin-5 sliding. However, maximal extension of a subset of anti-parallel microtubule pairs approaches the length of nascent bipolar spindles and is consistent with a Kinesin-5 crosslinking-driven transition. Nonetheless, microtubule sliding by Kinesin-5 contributes to stabilizing the nascent spindle and setting its stereotyped equilibrium length.


Subject(s)
Kinesins/genetics , Kinesins/metabolism , Spindle Apparatus/physiology , Cell Cycle/genetics , Microtubules/metabolism , Microtubules/physiology , Mitosis/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Spindle Poles/genetics , Spindle Poles/physiology
6.
Mol Biol Cell ; 30(22): 2802-2813, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31532702

ABSTRACT

Bipolar spindles are organized by motor proteins that generate microtubule--dependent forces to separate the two spindle poles. The fission yeast Cut7 (kinesin-5) is a plus-end-directed motor that generates the outward force to separate the two spindle poles, whereas the minus-end-directed motor Pkl1 (kinesin-14) generates the inward force. Balanced forces by these antagonizing kinesins are essential for bipolar spindle organization in mitosis. Here, we demonstrate that chromosomes generate another outward force that contributes to the bipolar spindle assembly. First, it was noted that the cut7 pkl1 double knockout failed to separate spindle poles in meiosis I, although the mutant is known to succeed it in mitosis. It was assumed that this might be because meiotic kinetochores of bivalent chromosomes joined by cross-overs generate weaker tensions in meiosis I than the strong tensions in mitosis generated by tightly tethered sister kinetochores. In line with this idea, when meiotic mono-oriented kinetochores were artificially converted to a mitotic bioriented layout, the cut7 pkl1 mutant successfully separated spindle poles in meiosis I. Therefore, we propose that spindle pole separation is promoted by outward forces transmitted from kinetochores to spindle poles through microtubules.


Subject(s)
Kinetochores/metabolism , Spindle Pole Bodies/metabolism , Spindle Poles/metabolism , Chromosome Segregation , Dyneins/metabolism , Kinesins/metabolism , Kinetochores/physiology , Meiosis/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitosis/physiology , Nuclear Proteins/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Spindle Apparatus/metabolism , Spindle Poles/physiology
7.
Curr Biol ; 28(19): R1131-R1135, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30300593

ABSTRACT

During the cell cycle it is critical that the duplicated DNA faithfully segregates to give rise to two genetically identical daughter cells. An even distribution of the genome during mitosis is mediated by mitotic spindle microtubules, assisted by, among others, motor proteins of the kinesin superfamily. Chromokinesins are members of the kinesin superfamily that harbour a specific DNA-binding domain. The best characterized chromokinesins belong to the kinesin-4/Kif4 and kinesin-10/Kif22 families, respectively. Functional analysis of chromokinesins in several model systems revealed their involvement in chromosome arm orientation and oscillations. This is consistent with their originally proposed role in the generation of polar ejection forces that assist chromosome congression to the spindle equator. Kinesin-12/Kif15 members comprise a third family of chromokinesins, but their role remains less understood. Noteworthy, all chromokinesins exhibit chromosome-independent localization on spindle microtubules, and recent works have significantly extended the portfolio of mitotic processes in which chromokinesins play a role, from error correction and DNA compaction, to the regulation of spindle microtubule dynamics.


Subject(s)
DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Kinesins/metabolism , Kinesins/physiology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Cell Division/physiology , Chromosome Segregation , Chromosomes/physiology , Dyneins/metabolism , Humans , Kinetochores/metabolism , Microtubules/metabolism , Mitosis , Spindle Apparatus/metabolism , Spindle Poles/physiology
8.
Math Biosci ; 303: 46-51, 2018 09.
Article in English | MEDLINE | ID: mdl-29792897

ABSTRACT

The mitotic spindle orientation and position is crucial for the fidelity of chromosome segregation during asymmetric cell division to generate daughter cells with different sizes or fates. This mechanism is best understood in the budding yeast Saccharomyces cerevisiae, named the spindle position checkpoint (SPOC). The SPOC inhibits cells from exiting mitosis until the mitotic spindle is properly oriented along the mother-daughter polarity axis. Despite many experimental studies, the mechanisms underlying SPOC regulation remains elusive and unexplored theoretically. Here, a minimal mathematical is developed to describe SPOC activation and silencing having autocatalytic feedback-loop. Numerical simulations of the nonlinear ordinary differential equations (ODEs) model accurately reproduce the phenotype of SPOC mechanism. Bifurcation analysis of the nonlinear ODEs reveals the orientation dependency on spindle pole bodies, and how this dependence is altered by parameter values. Partial differential equation (PDEs) model as well as linear stability analysis indicate that diffusion play no major role using experimental high diffusion values. These results provide for systems understanding on the molecular organization of spindle orientation system via mathematical modeling. The presented mathematical model is easy to understand and, within the above mentioned context, can be used as a base for further development of quantitative models in asymmetric cell-division.


Subject(s)
Models, Biological , Spindle Apparatus/physiology , Spindle Apparatus/ultrastructure , Cell Division/physiology , Cell Polarity/physiology , Computer Simulation , Mathematical Concepts , Mitosis/physiology , Nonlinear Dynamics , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/physiology , Spindle Poles/physiology , Spindle Poles/ultrastructure
9.
Biomolecules ; 9(1)2018 12 28.
Article in English | MEDLINE | ID: mdl-30597919

ABSTRACT

For appropriate chromosome segregation, kinetochores on sister chromatids have to attach to microtubules from opposite spindle poles (bi-orientation). Chromosome alignment at the spindle equator, referred to as congression, can occur through the attachment of kinetochores to the lateral surface of spindle microtubules, facilitating bi-orientation establishment. However, the contribution of this phenomenon to mitotic fidelity has not been clarified yet. Here, we addressed whether delayed chromosome alignment to the spindle equator increases the rate of chromosome missegregation. Cancer cell lines depleted of Kid, a chromokinesin involved in chromosome congression, showed chromosome alignment with a slight delay, and increased frequency of lagging chromosomes. Delayed chromosome alignment concomitant with an increased rate of lagging chromosomes was also seen in cells depleted of kinesin family member 4A (KIF4A), another chromokinesin. Cells that underwent chromosome missegregation took relatively longer time to align chromosomes in both control and Kid/KIF4A-depleted cells. Tracking of late-aligning chromosomes showed that they exhibit a higher rate of lagging chromosomes. Intriguingly, the metaphase of cells that underwent chromosome missegregation was shortened, and delaying anaphase onset ameliorated the increased chromosome missegregation. These data suggest that late-aligning chromosomes do not have sufficient time to establish bi-orientation, leading to chromosome missegregation. Our data imply that delayed chromosome alignment is not only a consequence, but also a cause of defective bi-orientation establishment, which can lead to chromosomal instability in cells without severe mitotic defects.


Subject(s)
Chromosome Segregation/physiology , Kinesins/metabolism , Spindle Poles/physiology , Cell Line, Tumor , Chromosomal Instability , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Kinesins/antagonists & inhibitors , Kinesins/genetics , Microscopy, Fluorescence , Mitosis , Prometaphase , RNA Interference , RNA, Small Interfering/metabolism , Statistics, Nonparametric
10.
Mol Cell Biol ; 38(2)2018 01 15.
Article in English | MEDLINE | ID: mdl-29061732

ABSTRACT

The migration of chromosomes during mitosis is mediated primarily by kinesins that bind to the chromosomes and move along the microtubules, exerting pulling and pushing forces on the centrosomes. We report that a DNA replication protein, Sld5, localizes to the centrosomes, resisting the microtubular pulling forces experienced during chromosome congression. In the absence of Sld5, centriolar satellites, which normally cluster around the centrosomes, are dissipated throughout the cytoplasm, resulting in the loss of their known function of recruiting the centrosomal protein, pericentrin. We observed that Sld5-deficient centrosomes lacking pericentrin were unable to endure the CENP-E- and Kid-mediated microtubular forces that converge on the centrosomes during chromosome congression, resulting in monocentriolar and acentriolar spindle poles. The minus-end-directed kinesin-14 motor protein, HSET, sustains the traction forces that mediate centrosomal fragmentation in Sld5-depleted cells. Thus, we report that a DNA replication protein has an as yet unknown function of ensuring spindle pole resistance to traction forces exerted during chromosome congression.


Subject(s)
Centrioles/metabolism , Centrosome/physiology , Chromosomal Proteins, Non-Histone/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 3/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 3/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrioles/genetics , Centrosome/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human/metabolism , DNA Damage , HeLa Cells , Humans , Interphase/physiology , Microtubules/chemistry , Microtubules/physiology , Mitosis , Spindle Poles/physiology , Spindle Poles/ultrastructure , Time-Lapse Imaging
11.
Mol Biol Cell ; 28(15): 2035-2041, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28539402

ABSTRACT

Aneuploidy, a condition that results from unequal partitioning of chromosomes during mitosis, is a hallmark of many cancers, including those caused by human papillomaviruses (HPVs). E6 and E7 are the primary transforming proteins in HPV that drive tumor progression. In this study, we stably expressed E6 and E7 in noncancerous RPE1 cells and analyzed the specific mitotic defects that contribute to aneuploidy in each cell line. We find that E6 expression results in multiple chromosomes associated with one or both spindle poles, causing a significant mitotic delay. In most cells, the misaligned chromosomes eventually migrated to the spindle equator, leading to mitotic exit. In some cells, however, mitotic exit occurred in the presence of pole-associated chromosomes. We determined that this premature mitotic exit is due to defects in spindle assembly checkpoint (SAC) signaling, such that cells are unable to maintain a prolonged mitotic arrest in the presence of unaligned chromosomes. This SAC defect is caused in part by a loss of kinetochore-associated Mad2 in E6-expressing cells. Our results demonstrate that E6-expressing cells exhibit previously unappreciated mitotic defects that likely contribute to HPV-mediated cancer progression.


Subject(s)
Cell Transformation, Viral , Chromatids/metabolism , Human papillomavirus 16/metabolism , M Phase Cell Cycle Checkpoints/physiology , Oncogene Proteins, Viral/metabolism , Repressor Proteins/metabolism , Spindle Poles/metabolism , Aneuploidy , Cell Cycle Proteins/metabolism , Chromosome Segregation , Humans , Kinetochores/metabolism , Mitosis , Papillomavirus E7 Proteins/metabolism , Signal Transduction , Spindle Apparatus/metabolism , Spindle Poles/physiology
12.
Oncogene ; 36(33): 4750-4766, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28394342

ABSTRACT

Centrosomes together with the mitotic spindle ensure the faithful distribution of chromosomes between daughter cells, and spindle orientation is a major determinant of cell fate during tissue regeneration. Spindle defects are not only an impetus of chromosome instability but are also a cause of developmental disorders involving defective asymmetric cell division. In this work, we demonstrate BCCIP, especially BCCIPα, as a previously unidentified component of the mitotic spindle pole and the centrosome. We demonstrate that BCCIP localizes proximal to the mother centriole and participates in microtubule organization and then redistributes to the spindle pole to ensure faithful spindle architecture. We find that BCCIP depletion leads to morphological defects, disoriented mitotic spindles, chromosome congression defects and delayed mitotic progression. Our study identifies BCCIP as a novel factor critical for microtubule regulation and explicates a mechanism utilized by BCCIP in tumor suppression.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Centrosome/physiology , Microtubules/physiology , Mitosis/physiology , Nuclear Proteins/metabolism , Spindle Poles/physiology , Animals , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Chromosome Segregation/physiology , Dynactin Complex/metabolism , Dyneins/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Microtubules/drug effects , Nocodazole/pharmacology , Nuclear Proteins/genetics , Paclitaxel/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tubulin Modulators/pharmacology
13.
Results Probl Cell Differ ; 61: 49-82, 2017.
Article in English | MEDLINE | ID: mdl-28409300

ABSTRACT

The budding yeast S. cerevisiae is a powerful model to understand the multiple layers of control driving an asymmetric cell division. In budding yeast, asymmetric targeting of the spindle poles to the mother and bud cell compartments respectively orients the mitotic spindle along the mother-bud axis. This program exploits an intrinsic functional asymmetry arising from the age distinction between the spindle poles-one inherited from the preceding division and the other newly assembled. Extrinsic mechanisms convert this age distinction into differential fate. Execution of this program couples spindle orientation with the segregation of the older spindle pole to the bud. Remarkably, similar stereotyped patterns of inheritance occur in self-renewing stem cell divisions underscoring the general importance of studying spindle polarity and differential fate in yeast. Here, we review the mechanisms accounting for this pivotal interplay between intrinsic and extrinsic asymmetries that translate spindle pole age into differential fate.


Subject(s)
Asymmetric Cell Division/physiology , Cell Polarity/physiology , Saccharomyces cerevisiae/physiology , Spindle Poles/physiology , Saccharomyces cerevisiae Proteins , Spindle Apparatus/physiology
14.
Mol Biol Cell ; 27(20): 3122-3131, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27559133

ABSTRACT

Although centrosomes contribute to spindle formation in most cell types, oocytes of many species are acentrosomal and must organize spindles in their absence. Here we investigate this process in Caenorhabditis elegans, detailing how acentrosomal spindles form and revealing mechanisms required to establish bipolarity. Using high-resolution imaging, we find that in meiosis I, microtubules initially form a "cage-like" structure inside the disassembling nuclear envelope. This structure reorganizes so that minus ends are sorted to the periphery of the array, forming multiple nascent poles that then coalesce until bipolarity is achieved. In meiosis II, microtubules nucleate in the vicinity of chromosomes but then undergo similar sorting and pole formation events. We further show that KLP-18/kinesin-12 and MESP-1, previously shown to be required for spindle bipolarity, likely contribute to bipolarity by sorting microtubules. After their depletion, minus ends are not sorted outward at the early stages of spindle assembly and instead converge. These proteins colocalize on microtubules, are interdependent for localization, and can interact, suggesting that they work together. We propose that KLP-18/kinesin-12 and MESP-1 form a complex that functions to sort microtubules of mixed polarity into a configuration in which minus ends are away from the chromosomes, enabling formation of nascent poles.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Kinesins/metabolism , Spindle Apparatus/metabolism , Animals , Caenorhabditis elegans/metabolism , Cell Polarity/physiology , Centrosome/metabolism , Chromosomes/metabolism , Meiosis/physiology , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Oocytes/metabolism , Spindle Poles/metabolism , Spindle Poles/physiology
15.
Cell Rep ; 14(12): 2975-87, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26997271

ABSTRACT

CPAP is required for centriole elongation during S/G2 phase, but the role of CPAP in mitosis is incompletely understood. Here, we show that CPAP maintains spindle pole integrity through its phosphorylation by Aurora-A during mitosis. Depletion of CPAP induced a prolonged delay in mitosis, pericentriolar material (PCM) dispersion, and multiple mitotic abnormalities. Further studies demonstrated that CPAP directly interacts with and is phosphorylated by Aurora-A at serine 467 during mitosis. Interestingly, the dispersal of the PCM was effectively rescued by ectopic expression of wild-type CPAP or a phospho-mimic CPAP-S467D mutant, but not a non-phosphorylated CPAP-S467A mutant. Finally, we found that CPAP-S467D has a low affinity for microtubule binding but a high affinity for PCM proteins. Together, our results support a model wherein CPAP is required for proper mitotic progression, and phosphorylation of CPAP by Aurora-A is essential for maintaining spindle pole integrity.


Subject(s)
Aurora Kinase A/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis , Spindle Poles/physiology , Antigens/genetics , Antigens/metabolism , Aurora Kinase A/genetics , Cell Cycle Proteins , Centrosome/physiology , HeLa Cells , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Microscopy, Confocal , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Time-Lapse Imaging , Tubulin/genetics , Tubulin/metabolism
16.
Nat Commun ; 5: 5496, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25400221

ABSTRACT

BRCA1-a breast and ovarian cancer suppressor gene-promotes genome integrity. To study the functionality of BRCA1 in the heterozygous state, we established a collection of primary human BRCA1(+/+) and BRCA1(mut/+) mammary epithelial cells and fibroblasts. Here we report that all BRCA1(mut/+) cells exhibited multiple normal BRCA1 functions, including the support of homologous recombination- type double-strand break repair (HR-DSBR), checkpoint functions, centrosome number control, spindle pole formation, Slug expression and satellite RNA suppression. In contrast, the same cells were defective in stalled replication fork repair and/or suppression of fork collapse, that is, replication stress. These defects were rescued by reconstituting BRCA1(mut/+) cells with wt BRCA1. In addition, we observed 'conditional' haploinsufficiency for HR-DSBR in BRCA1(mut/+) cells in the face of replication stress. Given the importance of replication stress in epithelial cancer development and of an HR defect in breast cancer pathogenesis, both defects are candidate contributors to tumorigenesis in BRCA1-deficient mammary tissue.


Subject(s)
DNA Replication/physiology , Genes, BRCA1/physiology , Haploinsufficiency/physiology , Animals , Breast/cytology , Cells, Cultured , Centrosome/physiology , DNA Replication/genetics , Female , Haploinsufficiency/genetics , Heterozygote , Humans , Mice , RNA, Satellite/genetics , RNA, Satellite/physiology , Rad51 Recombinase/genetics , Rad51 Recombinase/physiology , Recombinational DNA Repair/genetics , Recombinational DNA Repair/physiology , Spindle Poles/genetics , Spindle Poles/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...