Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.652
Filter
1.
Nat Commun ; 15(1): 5381, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918406

ABSTRACT

During human embryonic development, early cleavage-stage embryos are more susceptible to errors. Studies have shown that many problems occur during the first mitosis, such as direct cleavage, chromosome segregation errors, and multinucleation. However, the mechanisms whereby these errors occur during the first mitosis in human embryos remain unknown. To clarify this aspect, in the present study, we image discarded living human two-pronuclear stage zygotes using fluorescent labeling and confocal microscopy without microinjection of DNA or mRNA and investigate the association between spindle shape and nuclear abnormality during the first mitosis. We observe that the first mitotic spindles vary, and low-aspect-ratio-shaped spindles tend to lead to the formation of multiple nuclei at the 2-cell stage. Moreover, we observe defocusing poles in many of the first mitotic spindles, which are strongly associated with multinucleation. Additionally, we show that differences in the positions of the centrosomes cause spindle abnormality in the first mitosis. Furthermore, many multinuclei are modified to form mononuclei after the second mitosis because the occurrence of pole defocusing is firmly reduced. Our study will contribute markedly to research on the occurrence of mitotic errors during the early cleavage of human embryos.


Subject(s)
Cell Nucleus , Mitosis , Spindle Apparatus , Humans , Spindle Apparatus/metabolism , Cell Nucleus/metabolism , Zygote/cytology , Zygote/metabolism , Embryo, Mammalian/cytology , Microscopy, Confocal , Centrosome/metabolism , Embryonic Development/physiology , Female
2.
Proc Natl Acad Sci U S A ; 121(25): e2323009121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38875144

ABSTRACT

Error correction is central to many biological systems and is critical for protein function and cell health. During mitosis, error correction is required for the faithful inheritance of genetic material. When functioning properly, the mitotic spindle segregates an equal number of chromosomes to daughter cells with high fidelity. Over the course of spindle assembly, many initially erroneous attachments between kinetochores and microtubules are fixed through the process of error correction. Despite the importance of chromosome segregation errors in cancer and other diseases, there is a lack of methods to characterize the dynamics of error correction and how it can go wrong. Here, we present an experimental method and analysis framework to quantify chromosome segregation error correction in human tissue culture cells with live cell confocal imaging, timed premature anaphase, and automated counting of kinetochores after cell division. We find that errors decrease exponentially over time during spindle assembly. A coarse-grained model, in which errors are corrected in a chromosome-autonomous manner at a constant rate, can quantitatively explain both the measured error correction dynamics and the distribution of anaphase onset times. We further validated our model using perturbations that destabilized microtubules and changed the initial configuration of chromosomal attachments. Taken together, this work provides a quantitative framework for understanding the dynamics of mitotic error correction.


Subject(s)
Chromosome Segregation , Kinetochores , Microtubules , Mitosis , Spindle Apparatus , Humans , Kinetochores/metabolism , Spindle Apparatus/metabolism , Microtubules/metabolism , Anaphase , Models, Biological , HeLa Cells
3.
PLoS Genet ; 20(6): e1011302, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38829899

ABSTRACT

Cryptococcus neoformans is an opportunistic, human fungal pathogen which undergoes fascinating switches in cell cycle control and ploidy when it encounters stressful environments such as the human lung. Here we carry out a mechanistic analysis of the spindle checkpoint which regulates the metaphase to anaphase transition, focusing on Mps1 kinase and the downstream checkpoint components Mad1 and Mad2. We demonstrate that Cryptococcus mad1Δ or mad2Δ strains are unable to respond to microtubule perturbations, continuing to re-bud and divide, and die as a consequence. Fluorescent tagging of Chromosome 3, using a lacO array and mNeonGreen-lacI fusion protein, demonstrates that mad mutants are unable to maintain sister-chromatid cohesion in the absence of microtubule polymers. Thus, the classic checkpoint functions of the SAC are conserved in Cryptococcus. In interphase, GFP-Mad1 is enriched at the nuclear periphery, and it is recruited to unattached kinetochores in mitosis. Purification of GFP-Mad1 followed by mass spectrometric analysis of associated proteins show that it forms a complex with Mad2 and that it interacts with other checkpoint signalling components (Bub1) and effectors (Cdc20 and APC/C sub-units) in mitosis. We also demonstrate that overexpression of Mps1 kinase is sufficient to arrest Cryptococcus cells in mitosis, and show that this arrest is dependent on both Mad1 and Mad2. We find that a C-terminal fragment of Mad1 is an effective in vitro substrate for Mps1 kinase and map several Mad1 phosphorylation sites. Some sites are highly conserved within the C-terminal Mad1 structure and we demonstrate that mutation of threonine 667 (T667A) leads to loss of checkpoint signalling and abrogation of the GAL-MPS1 arrest. Thus Mps1-dependent phosphorylation of C-terminal Mad1 residues is a critical step in Cryptococcus spindle checkpoint signalling. We conclude that CnMps1 protein kinase, Mad1 and Mad2 proteins have all conserved their important, spindle checkpoint signalling roles helping ensure high fidelity chromosome segregation.


Subject(s)
Cell Cycle Proteins , Cryptococcus neoformans , Mad2 Proteins , Spindle Apparatus , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Mad2 Proteins/metabolism , Mad2 Proteins/genetics , Spindle Apparatus/metabolism , Spindle Apparatus/genetics , Signal Transduction , Fungal Proteins/metabolism , Fungal Proteins/genetics , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , M Phase Cell Cycle Checkpoints/genetics , Mitosis/genetics , Kinetochores/metabolism , Chromosome Segregation/genetics , Microtubules/metabolism , Microtubules/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
4.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38869473

ABSTRACT

At each cell division, nanometer-scale motors and microtubules give rise to the micron-scale spindle. Many mitotic motors step helically around microtubules in vitro, and most are predicted to twist the spindle in a left-handed direction. However, the human spindle exhibits only slight global twist, raising the question of how these molecular torques are balanced. Here, we find that anaphase spindles in the epithelial cell line MCF10A have a high baseline twist, and we identify factors that both increase and decrease this twist. The midzone motors KIF4A and MKLP1 are together required for left-handed twist at anaphase, and we show that KIF4A generates left-handed torque in vitro. The actin cytoskeleton also contributes to left-handed twist, but dynein and its cortical recruitment factor LGN counteract it. Together, our work demonstrates that force generators regulate twist in opposite directions from both within and outside the spindle, preventing strong spindle twist during chromosome segregation.


Subject(s)
Anaphase , Kinesins , Microtubules , Spindle Apparatus , Humans , Spindle Apparatus/metabolism , Kinesins/metabolism , Kinesins/genetics , Microtubules/metabolism , Dyneins/metabolism , Dyneins/genetics , Torque , Chromosome Segregation , Actin Cytoskeleton/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 121(25): e2318838121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38870057

ABSTRACT

Hertwig's rule states that cells divide along their longest axis, usually driven by forces acting on the mitotic spindle. Here, we show that in contrast to this rule, microtubule-based pulling forces in early Caenorhabditis elegans embryos align the spindle with the short axis of the cell. We combine theory with experiments to reveal that in order to correct this misalignment, inward forces generated by the constricting cytokinetic ring rotate the entire cell until the spindle is aligned with the cell's long axis. Experiments with slightly compressed mouse zygotes indicate that this cytokinetic ring-driven mechanism of ensuring Hertwig's rule is general for cells capable of rotating inside a confining shell, a scenario that applies to early cell divisions of many systems.


Subject(s)
Caenorhabditis elegans , Spindle Apparatus , Animals , Caenorhabditis elegans/embryology , Mice , Spindle Apparatus/metabolism , Microtubules/metabolism , Cytokinesis/physiology , Rotation , Zygote/metabolism , Zygote/cytology , Zygote/growth & development , Embryo, Nonmammalian/cytology , Embryonic Development/physiology , Models, Biological
7.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38928036

ABSTRACT

Paclitaxel induces multipolar spindles at clinically relevant doses but does not substantially increase mitotic indices. Paclitaxel's anti-cancer effects are hypothesized to occur by promoting chromosome mis-segregation on multipolar spindles leading to apoptosis, necrosis and cyclic-GMP-AMP Synthase-Stimulator of Interferon Genes (cGAS-STING) pathway activation in daughter cells, leading to secretion of type I interferon (IFN) and immunogenic cell death. Eribulin and vinorelbine have also been reported to cause increases in multipolar spindles in cancer cells. Recently, suppression of Anaphase-Promoting Complex/Cyclosome-Cell Division Cycle 20 (APC/C-CDC20) activity using CRISPR/Cas9 mutagenesis has been reported to increase sensitivity to Kinesin Family 18a (KIF18a) inhibition, which functions to suppress multipolar mitotic spindles in cancer cells. We propose that a way to enhance the effectiveness of anti-cancer agents that increase multipolar spindles is by suppressing the APC/C-CDC20 to delay, but not block, anaphase entry. Delaying anaphase entry in genomically unstable cells may enhance multipolar spindle-induced cell death. In genomically stable healthy human cells, delayed anaphase entry may suppress the level of multipolar spindles induced by anti-cancer drugs and lower mitotic cytotoxicity. We outline specific combinations of molecules to investigate that may achieve the goal of enhancing the effectiveness of anti-cancer agents.


Subject(s)
Anaphase-Promoting Complex-Cyclosome , Antineoplastic Agents , Spindle Apparatus , Humans , Anaphase-Promoting Complex-Cyclosome/metabolism , Antineoplastic Agents/pharmacology , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Cdc20 Proteins/metabolism , Cdc20 Proteins/genetics , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Mitosis/drug effects
8.
Yi Chuan ; 46(6): 502-508, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38886153

ABSTRACT

Ssu72 is a component of the yeast cleavage/polyadenylation factor (CPF) complex, which catalyzes the dephosphorylation of the C-terminal domain (CTD) of RNA polymerase II at S5-P and S7-P. It has been shown that Ssu72 phosphatase is involved in regulating chromosome cohesion during mitosis. To further clarify whether Ssu72 phosphatase affects chromosome separation during meiotic division in Schizosaccharomyces pombe, we utilized green fluorescent protein (GFP) to label centromeres and red fluorescent protein to label microtubule protein Atb2. The entire meiotic chromosome separation process of ssu72∆ cells was observed in real-time under fluorescence microscope. It was found that two spindles of ssu72∆ cells crossed during the metaphase and anaphase of the second meiotic division, and this spindle crossing led to a new type of spore defect distribution pattern. The results of this study can provide important reference significance for studying the roles of phosphatase Ssu72 in higher organisms.


Subject(s)
Meiosis , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Spindle Apparatus , Schizosaccharomyces/genetics , Schizosaccharomyces/enzymology , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Chromosome Segregation
9.
FASEB J ; 38(13): e23750, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38888878

ABSTRACT

Kif16A, a member of the kinesin-3 family of motor proteins, has been shown to play crucial roles in inducing mitotic arrest, apoptosis, and mitotic cell death. However, its roles during oocyte meiotic maturation have not been fully defined. In this study, we report that Kif16A exhibits unique accumulation on the spindle apparatus and colocalizes with microtubule fibers during mouse oocyte meiotic maturation. Targeted depletion of Kif16A using gene-targeting siRNA disrupts the progression of the meiotic cell cycle. Furthermore, Kif16A depletion leads to aberrant spindle assembly and chromosome misalignment in oocytes. Our findings also indicate that Kif16A depletion reduces tubulin acetylation levels and compromises microtubule resistance to depolymerizing drugs, suggesting its crucial role in microtubule stability maintenance. Notably, we find that the depletion of Kif16A results in a notably elevated incidence of defective kinetochore-microtubule attachments and the absence of BubR1 localization at kinetochores, suggesting a critical role for Kif16A in the activation of the spindle assembly checkpoint (SAC) activity. Additionally, we observe that Kif16A is indispensable for proper actin filament distribution, thereby impacting spindle migration. In summary, our findings demonstrate that Kif16A plays a pivotal role in regulating microtubule and actin dynamics crucial for ensuring both spindle assembly and migration during mouse oocyte meiotic maturation.


Subject(s)
Kinesins , Meiosis , Microtubules , Oocytes , Spindle Apparatus , Animals , Kinesins/metabolism , Kinesins/genetics , Meiosis/physiology , Oocytes/metabolism , Microtubules/metabolism , Mice , Spindle Apparatus/metabolism , Female , Actins/metabolism , Kinetochores/metabolism
10.
FASEB J ; 38(13): e23739, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38884157

ABSTRACT

Arf6 is a member of ADP-ribosylation factor (Arf) family, which is widely implicated in the regulation of multiple physiological processes including endocytic recycling, cytoskeletal organization, and membrane trafficking during mitosis. In this study, we investigated the potential relationship between Arf6 and aging-related oocyte quality, and its roles on organelle rearrangement and cytoskeleton dynamics in porcine oocytes. Arf6 expressed in porcine oocytes throughout meiotic maturation, and it decreased in aged oocytes. Disruption of Arf6 led to the failure of cumulus expansion and polar body extrusion. Further analysis indicated that Arf6 modulated ac-tubulin for meiotic spindle organization and microtubule stability. Besides, Arf6 regulated cofilin phosphorylation and fascin for actin assembly, which further affected spindle migration, indicating the roles of Arf6 on cytoskeleton dynamics. Moreover, the lack of Arf6 activity caused the dysfunction of Golgi and ER for protein synthesis and signal transduction. Mitochondrial dysfunction was also observed in Arf6-deficient porcine oocytes, which was supported by the increased ROS level and abnormal membrane potential. In conclusion, our results reported that insufficient Arf6 was related to aging-induced oocyte quality decline through spindle organization, actin assembly, and organelle rearrangement in porcine oocytes.


Subject(s)
ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors , Oocytes , Animals , Oocytes/metabolism , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , Swine , Female , Meiosis/physiology , Spindle Apparatus/metabolism , Aging/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
11.
EMBO Rep ; 25(6): 2722-2742, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38773322

ABSTRACT

Alpha, beta, and gamma tubulins are essential building blocks for all eukaryotic cells. The functions of the non-canonical tubulins, delta, epsilon, and zeta, however, remain poorly understood and their requirement in mammalian development untested. Herein we have used a spermatogenesis model to define epsilon tubulin (TUBE1) function in mice. We show that TUBE1 is essential for the function of multiple complex microtubule arrays, including the meiotic spindle, axoneme and manchette and in its absence, there is a dramatic loss of germ cells and male sterility. Moreover, we provide evidence for the interplay between TUBE1 and katanin-mediated microtubule severing, and for the sub-specialization of individual katanin paralogs in the regulation of specific microtubule arrays.


Subject(s)
Katanin , Microtubules , Spermatogenesis , Tubulin , Animals , Male , Microtubules/metabolism , Tubulin/metabolism , Mice , Katanin/metabolism , Katanin/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Germ Cells/metabolism , Spindle Apparatus/metabolism , Spermatozoa/metabolism , Infertility, Male/metabolism , Infertility, Male/genetics , Mice, Knockout , Axoneme/metabolism
12.
J Cell Sci ; 137(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38770570

ABSTRACT

Dinoflagellates are marine organisms that undergo seasonal proliferation events known as algal blooms. Vegetative cell proliferation is a main contributing factor in these events. However, mechanistical understanding of mitosis and cytokinesis in dinoflagellates remains rudimentary. Using an optimized immunofluorescence protocol, we analysed changes in microtubule organization occurring during the mitotic cycle of the toxic dinoflagellate Ostreopsis cf. ovata. We find that the flagella and the cortical microtubule array persist throughout the mitotic cycle. Two cytoplasmic microtubule bundles originate from the ventral area, where the basal bodies are located - a cortical bundle and a cytoplasmic bundle. The latter associates with the nucleus in the cell centre before mitosis and with the acentrosomal extranuclear spindle during mitosis. Analysis of tubulin post-translational modifications identifies two populations of spindle microtubules - polar acetylated microtubules, whose length is constant, and central tyrosinated microtubules, which elongate during chromosome segregation. During cell division a microtubule-rich structure forms along the dorsal-ventral axis, associated with the site of cytokinesis, consistent with a cytokinetic mechanism that is independent of the actomyosin ring typical of animal and yeast cells.


Subject(s)
Dinoflagellida , Microtubules , Mitosis , Microtubules/metabolism , Dinoflagellida/metabolism , Dinoflagellida/cytology , Cytokinesis , Spindle Apparatus/metabolism , Cell Division , Tubulin/metabolism
14.
Nat Commun ; 15(1): 3779, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710684

ABSTRACT

The α-Aurora kinase is a crucial regulator of spindle microtubule organization during mitosis in plants. Here, we report a post-mitotic role for α-Aurora in reorganizing the phragmoplast microtubule array. In Arabidopsis thaliana, α-Aurora relocated from spindle poles to the phragmoplast midzone, where it interacted with the microtubule cross-linker MAP65-3. In a hypomorphic α-Aurora mutant, MAP65-3 was detected on spindle microtubules, followed by a diffuse association pattern across the phragmoplast midzone. Simultaneously, phragmoplast microtubules remained belatedly in a solid disk array before transitioning to a ring shape. Microtubules at the leading edge of the matured phragmoplast were often disengaged, accompanied by conspicuous retentions of MAP65-3 at the phragmoplast interior edge. Specifically, α-Aurora phosphorylated two residues towards the C-terminus of MAP65-3. Mutation of these residues to alanines resulted in an increased association of MAP65-3 with microtubules within the phragmoplast. Consequently, the expansion of the phragmoplast was notably slower compared to wild-type cells or cells expressing a phospho-mimetic variant of MAP65-3. Moreover, mimicking phosphorylation reinstated disrupted MAP65-3 behaviors in plants with compromised α-Aurora function. Overall, our findings reveal a mechanism in which α-Aurora facilitates cytokinesis progression through phosphorylation-dependent restriction of MAP65-3 associating with microtubules at the phragmoplast midzone.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cytokinesis , Microtubule-Associated Proteins , Microtubules , Arabidopsis/metabolism , Arabidopsis/genetics , Microtubules/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Phosphorylation , Mutation , Spindle Apparatus/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Plants, Genetically Modified , Mitosis
15.
PLoS Genet ; 20(5): e1011272, 2024 May.
Article in English | MEDLINE | ID: mdl-38768219

ABSTRACT

The position of the nucleus before it divides during mitosis is variable in different budding yeasts. Studies in the pathogenic intron-rich fungus Cryptococcus neoformans reveal that the nucleus moves entirely into the daughter bud before its division. Here, we report functions of a zinc finger motif containing spliceosome protein C. neoformans Slu7 (CnSlu7) in cell cycle progression. The budding yeast and fission yeast homologs of Slu7 have predominant roles for intron 3' splice site definition during pre-mRNA splicing. Using a conditional knockdown strategy, we show CnSlu7 is an essential factor for viability and is required for efficient cell cycle progression with major role during mitosis. Aberrant nuclear migration, including improper positioning of the nucleus as well as the spindle, were frequently observed in cells depleted of CnSlu7. However, cell cycle delays observed due to Slu7 depletion did not activate the Mad2-dependent spindle assembly checkpoint (SAC). Mining of the global transcriptome changes in the Slu7 knockdown strain identified downregulation of transcripts encoding several cell cycle regulators and cytoskeletal factors for nuclear migration, and the splicing of specific introns of these genes was CnSlu7 dependent. To test the importance of splicing activity of CnSlu7 on nuclear migration, we complemented Slu7 knockdown cells with an intron less PAC1 minigene and demonstrated that the nuclear migration defects were significantly rescued. These findings show that CnSlu7 regulates the functions of diverse cell cycle regulators and cytoskeletal components, ensuring timely cell cycle transitions and nuclear division during mitosis.


Subject(s)
Cell Nucleus , Cryptococcus neoformans , Fungal Proteins , Mitosis , RNA Splicing , Spliceosomes , Mitosis/genetics , Cryptococcus neoformans/genetics , RNA Splicing/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Spindle Apparatus/metabolism , Spindle Apparatus/genetics , Gene Expression Regulation, Fungal , Cell Cycle/genetics
16.
Open Biol ; 14(5): 230460, 2024 May.
Article in English | MEDLINE | ID: mdl-38806145

ABSTRACT

The precise spatial and temporal control of histone phosphorylations is important for the ordered progression through the different phases of mitosis. The phosphorylation of H2B at S6 (H2B S6ph), which is crucial for chromosome segregation, reaches its maximum level during metaphase and is limited to the inner centromere. We discovered that the temporal and spatial regulation of this modification, as well as its intensity, are governed by the scaffold protein RepoMan and its associated catalytically active phosphatases, PP1α and PP1γ. Phosphatase activity is inhibited at the area of maximal H2B S6 phosphorylation at the inner centromere by site-specific Aurora B-mediated inactivation of the PP1/RepoMan complex. The motor protein Mklp2 contributes to the relocalization of Aurora B from chromatin to the mitotic spindle during anaphase, thus alleviating Aurora B-dependent repression of the PP1/RepoMan complex and enabling dephosphorylation of H2B S6. Accordingly, dysregulation of Mklp2 levels, as commonly observed in tumour cells, leads to the lack of H2B S6 dephosphorylation during early anaphase, which might contribute to chromosomal instability.


Subject(s)
Aurora Kinase B , Cell Cycle Proteins , Histones , Mitosis , Protein Phosphatase 1 , Aurora Kinase B/metabolism , Phosphorylation , Humans , Histones/metabolism , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , HeLa Cells , Spindle Apparatus/metabolism , Centromere/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
17.
Cell Mol Biol Lett ; 29(1): 68, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730334

ABSTRACT

BACKGROUND: Members of the nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing (NLRP) family regulate various physiological and pathological processes. However, none have been shown to regulate actin cap formation or spindle translocation during the asymmetric division of oocyte meiosis I. NLRP4E has been reported as a candidate protein in female fertility, but its function is unknown. METHODS: Immunofluorescence, reverse transcription polymerase chain reaction (RT-PCR), and western blotting were employed to examine the localization and expression levels of NLRP4E and related proteins in mouse oocytes. small interfering RNA (siRNA) and antibody transfection were used to knock down NLRP4E and other proteins. Immunoprecipitation (IP)-mass spectrometry was used to identify the potential proteins interacting with NLRP4E. Coimmunoprecipitation (Co-IP) was used to verify the protein interactions. Wild type (WT) or mutant NLRP4E messenger RNA (mRNA) was injected into oocytes for rescue experiments. In vitro phosphorylation was employed to examine the activation of steroid receptor coactivator (SRC) by NLRP4E. RESULTS: NLRP4E was more predominant within oocytes compared with other NLRP4 members. NLRP4E knockdown significantly inhibited actin cap formation and spindle translocation toward the cap region, resulting in the failure of polar body extrusion at the end of meiosis I. Mechanistically, GRIN1, and GANO1 activated NLRP4E by phosphorylation at Ser429 and Thr430; p-NLRP4E is translocated and is accumulated in the actin cap region during spindle translocation. Next, we found that p-NLRP4E directly phosphorylated SRC at Tyr418, while p-SRC negatively regulated p-CDC42-S71, an inactive form of CDC42 that promotes actin cap formation and spindle translocation in the GTP-bound form. CONCLUSIONS: NLRP4E activated by GRIN1 and GANO1 regulates actin cap formation and spindle translocation toward the cap region through upregulation of p-SRC-Tyr418 and downregulation of p-CDC42-S71 during meiosis I.


Subject(s)
Actins , Meiosis , Oocytes , cdc42 GTP-Binding Protein , Animals , Oocytes/metabolism , Mice , Female , Actins/metabolism , Actins/genetics , cdc42 GTP-Binding Protein/metabolism , cdc42 GTP-Binding Protein/genetics , Phosphorylation , Spindle Apparatus/metabolism
18.
Cell ; 187(12): 3006-3023.e26, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38744280

ABSTRACT

Centromeres are scaffolds for the assembly of kinetochores that ensure chromosome segregation during cell division. How vertebrate centromeres obtain a three-dimensional structure to accomplish their primary function is unclear. Using super-resolution imaging, capture-C, and polymer modeling, we show that vertebrate centromeres are partitioned by condensins into two subdomains during mitosis. The bipartite structure is found in human, mouse, and chicken cells and is therefore a fundamental feature of vertebrate centromeres. Super-resolution imaging and electron tomography reveal that bipartite centromeres assemble bipartite kinetochores, with each subdomain binding a distinct microtubule bundle. Cohesin links the centromere subdomains, limiting their separation in response to spindle forces and avoiding merotelic kinetochore-spindle attachments. Lagging chromosomes during cancer cell divisions frequently have merotelic attachments in which the centromere subdomains are separated and bioriented. Our work reveals a fundamental aspect of vertebrate centromere biology with implications for understanding the mechanisms that guarantee faithful chromosome segregation.


Subject(s)
Centromere , Cohesins , Kinetochores , Mitosis , Animals , Humans , Mice , Cell Cycle Proteins/metabolism , Centromere/metabolism , Chickens , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosome Segregation , Kinetochores/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism
19.
J Cell Biol ; 223(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38727808

ABSTRACT

Accurate chromosome segregation requires sister kinetochores to biorient, attaching to opposite spindle poles. To this end, the mammalian kinetochore destabilizes incorrect attachments and stabilizes correct ones, but how it discriminates between these is not yet clear. Here, we test the model that kinetochore tension is the stabilizing cue and ask how chromosome size impacts that model. We live image PtK2 cells, with just 14 chromosomes, widely ranging in size, and find that long chromosomes align at the metaphase plate later than short chromosomes. Enriching for errors and imaging error correction live, we show that long chromosomes exhibit a specific delay in correcting attachments. Using chromokinesin overexpression and laser ablation to perturb polar ejection forces, we find that chromosome size and force on arms determine alignment order. Thus, we propose a model where increased force on long chromosomes can falsely stabilize incorrect attachments, delaying their biorientation. As such, long chromosomes may require compensatory mechanisms for correcting errors to avoid chromosomal instability.


Subject(s)
Chromosome Segregation , Chromosomes, Mammalian , Kinetochores , Mitosis , Animals , Cell Line , Chromosomes, Mammalian/chemistry , Chromosomes, Mammalian/metabolism , Kinetochores/metabolism , Spindle Apparatus/metabolism , Potoroidae
20.
Elife ; 122024 May 15.
Article in English | MEDLINE | ID: mdl-38747713

ABSTRACT

During mammalian oocyte meiosis, spindle migration and asymmetric cytokinesis are unique steps for the successful polar body extrusion. The asymmetry defects of oocytes will lead to the failure of fertilization and embryo implantation. In present study, we reported that an actin nucleating factor Formin-like 2 (FMNL2) played critical roles in the regulation of spindle migration and organelle distribution in mouse and porcine oocytes. Our results showed that FMNL2 mainly localized at the oocyte cortex and periphery of spindle. Depletion of FMNL2 led to the failure of polar body extrusion and large polar bodies in oocytes. Live-cell imaging revealed that the spindle failed to migrate to the oocyte cortex, which caused polar body formation defects, and this might be due to the decreased polymerization of cytoplasmic actin by FMNL2 depletion in the oocytes of both mice and pigs. Furthermore, mass spectrometry analysis indicated that FMNL2 was associated with mitochondria and endoplasmic reticulum (ER)-related proteins, and FMNL2 depletion disrupted the function and distribution of mitochondria and ER, showing with decreased mitochondrial membrane potential and the occurrence of ER stress. Microinjecting Fmnl2-EGFP mRNA into FMNL2-depleted oocytes significantly rescued these defects. Thus, our results indicate that FMNL2 is essential for the actin assembly, which further involves into meiotic spindle migration and ER/mitochondria functions in mammalian oocytes.


Subject(s)
Actins , Endoplasmic Reticulum , Formins , Meiosis , Mitochondria , Oocytes , Animals , Endoplasmic Reticulum/metabolism , Oocytes/metabolism , Formins/metabolism , Formins/genetics , Mitochondria/metabolism , Mice , Actins/metabolism , Swine , Female , Spindle Apparatus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...