Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
EMBO J ; 42(17): e114415, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37427462

ABSTRACT

Cell fragmentation is commonly observed in human preimplantation embryos and is associated with poor prognosis during assisted reproductive technology (ART) procedures. However, the mechanisms leading to cell fragmentation remain largely unknown. Here, light sheet microscopy imaging of mouse embryos reveals that inefficient chromosome separation due to spindle defects, caused by dysfunctional molecular motors Myo1c or dynein, leads to fragmentation during mitosis. Extended exposure of the cell cortex to chromosomes locally triggers actomyosin contractility and pinches off cell fragments. This process is reminiscent of meiosis, during which small GTPase-mediated signals from chromosomes coordinate polar body extrusion (PBE) by actomyosin contraction. By interfering with the signals driving PBE, we find that this meiotic signaling pathway remains active during cleavage stages and is both required and sufficient to trigger fragmentation. Together, we find that fragmentation happens in mitosis after ectopic activation of actomyosin contractility by signals emanating from DNA, similar to those observed during meiosis. Our study uncovers the mechanisms underlying fragmentation in preimplantation embryos and, more generally, offers insight into the regulation of mitosis during the maternal-zygotic transition.


Subject(s)
Actomyosin , Polar Bodies , Humans , Animals , Mice , Polar Bodies/metabolism , Actomyosin/metabolism , Blastocyst , Chromosomes , Meiosis , Oocytes/metabolism , Spindle Apparatus/genetics , Myosin Type I/genetics , Myosin Type I/metabolism
2.
Bull Math Biol ; 84(10): 114, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36058957

ABSTRACT

The intrinsic polarity of migrating cells is regulated by spatial distributions of protein activity. Those proteins (Rho-family GTPases, such as Rac and Rho) redistribute in response to stimuli, determining the cell front and back. Reaction-diffusion equations with mass conservation and positive feedback have been used to explain initial polarization of a cell. However, the sensitivity of a polar cell to a reversal stimulus has not yet been fully understood. We carry out a PDE bifurcation analysis of two polarity models to investigate routes to repolarization: (1) a single-GTPase ("wave-pinning") model and (2) a mutually antagonistic Rac-Rho model. We find distinct routes to reversal in (1) vs. (2). We show numerical simulations of full PDE solutions for the RD equations, demonstrating agreement with predictions of the bifurcation results. Finally, we show that simulations of the polarity models in deforming 1D model cells are consistent with biological experiments.


Subject(s)
Cell Polarity , Polar Bodies , Cell Polarity/physiology , Mathematical Concepts , Models, Biological , Polar Bodies/metabolism , rho GTP-Binding Proteins/metabolism
3.
Cell Cycle ; 21(21): 2239-2254, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35775922

ABSTRACT

Polar body emission is a special form of cytokinesis in oocyte meiosis that ensures the correct number of chromosomes in reproduction-competent eggs. The molecular mechanism of the last step, polar body abscission, is poorly understood. While it has been proposed that Ca2+ signaling plays important roles in embryonic cytokinesis, to date transient increases in intracellular free Ca2+ have been difficult to document in oocyte meiosis except for the global Ca2+ wave induced by sperm at fertilization. Here, we find that microinjection of the calcium chelator dibromo-BAPTA inhibits polar body abscission in Xenopus laevis oocytes. Using a novel, microtubule-targeted ratio-metric calcium sensor, we detected a calcium transient that is focused at the contractile ring-associated plasma membrane and which occurred after anaphase and constriction of the contractile ring but prior to abscission. This calcium transient was confirmed by mobile calcium probes. Further, the Ca2+-sensitive protein kinase Cß C2 domain transiently translocated to the contractile ring-associated membrane simultaneously with the calcium transient. Collectively, these results demonstrate that a calcium transient, apparently originating at the contractile ring-associated plasma membrane, promotes polar body abscission.


Subject(s)
Calcium , Polar Bodies , Animals , Calcium/metabolism , Calcium Chelating Agents/metabolism , Male , Meiosis , Oocytes/metabolism , Polar Bodies/metabolism , Protein Kinases/metabolism , Semen/metabolism
4.
Fertil Steril ; 117(1): 213-220, 2022 01.
Article in English | MEDLINE | ID: mdl-34548166

ABSTRACT

OBJECTIVE: To examine the cause of monopronucleated zygote (1PN) formation that includes both maternal and paternal genomes. DESIGN: Retrospective cohort study. SETTING: Private fertility clinic. PATIENT(S): A total of 44 1PN and 726 2-pronuclear zygotes from 702 patients were observed using 2 different time-lapse observation systems. INTERVENTION(S): Previously recorded time lapse data were reviewed to examine the mechanism of 1PN formation. MAIN OUTCOME MEASURE(S): The distance between the position of the second polar body extrusion and the fertilization cone or epicenter/starting position of the cytoplasmic wave was measured, and the consequent data were analyzed. Cytoplasmic waves were confirmed using vector analysis software. RESULT(S): The cut-off value for the difference in the distance between the position of the second polar body extrusion and the fertilization cone or the epicenter/starting position of the cytoplasmic wave was 17 µm (AUC: 0.987, 95% CI: 0.976-0.999) for the Embryo Scope and 18 µm (AUC: 0.972, 95% CI: 0.955-0.988) for the iBIS time-lapse observation systems. CONCLUSION(S): In this study, it was found with a high degree of accuracy that a monopronucleus is formed when the fusion of the sperm takes place within 18 µm from the point of the second polar body extrusion. The theoretical chance of 1PN occurrence after in vitro fertilization is 2.7% when the sperm is considered to be fused anywhere in the plasma membrane of an oocyte.


Subject(s)
Fertilization in Vitro , Genome, Human , Zygote/physiology , Adult , Cell Nucleus/genetics , Cohort Studies , Cytoplasm/genetics , Cytoplasm/metabolism , Embryonic Development/genetics , Female , Humans , Male , Polar Bodies/metabolism , Retrospective Studies , Sex Chromosomes/genetics , Sperm Injections, Intracytoplasmic , Time-Lapse Imaging , Zygote/cytology
5.
Int J Mol Sci ; 22(9)2021 May 03.
Article in English | MEDLINE | ID: mdl-34063622

ABSTRACT

Ribonucleic acid export 1 (Rae1) is an important nucleoporin that participates in mRNA export during the interphase of higher eukaryotes and regulates the mitotic cell cycle. In this study, small RNA interference technology was used to knockdown Rae1, and immunofluorescence, immunoblotting, and chromosome spreading were used to study the role of Rae1 in mouse oocyte meiotic maturation. We found that Rae1 is a crucial regulator of meiotic maturation of mouse oocytes. After the resumption of meiosis (GVBD), Rae1 was concentrated on the kinetochore structure. The knockdown of Rae1 by a specific siRNA inhibited GVBD progression at 2 h, finally leading to a decreased 14 h polar body extrusion (PBE) rate. However, a comparable 14 h PBE rate was found in the control, and the Rae1 knockdown groups that had already undergone GVBD. Furthermore, we found elevated PBE after 9.5 h in the Rae1 knockdown oocytes. Further analysis revealed that Rae1 depletion significantly decreased the protein level of securin. In addition, we detected weakened kinetochore-microtubule (K-MT) attachments, misaligned chromosomes, and an increased incidence of aneuploidy in the Rae1 knockdown oocytes. Collectively, we propose that Rae1 modulates securin protein levels, which contribute to chromosome alignment, K-MT attachments, and aneuploidy in meiosis.


Subject(s)
Meiosis/genetics , Microtubule-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Oocytes/metabolism , Animals , Gene Knockdown Techniques , In Vitro Oocyte Maturation Techniques , Kinetochores/metabolism , Mice , Oocytes/growth & development , Polar Bodies/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics
6.
J Cell Physiol ; 236(11): 7725-7733, 2021 11.
Article in English | MEDLINE | ID: mdl-34018605

ABSTRACT

During mouse oocyte meiotic maturation, actin filaments play multiple roles in meiosis such as spindle migration and cytokinesis. FASCIN is shown to be an actin-binding and bundling protein, making actin filaments tightly packed and parallel-aligned, and FASCIN is involved in several cellular processes like adhesion and migration. FASCIN is also a potential prognostic biomarker and therapeutic target for the treatment of metastatic disease. However, little is known about the functions of FASCIN in oocyte meiosis. In the present study, we knocked down the expression of FASCIN, and our results showed that FASCIN was essential for oocyte maturation. FASCIN was all expressed in the different stages of oocyte meiosis, and it mainly localized at the cortex of oocytes from the GV stage to the MII stage and showed a similar localization pattern with actin and DAAM1. Depletion of FASCIN affected the extrusion of the first polar body, and we also observed that some oocytes extruded from the large polar bodies. This might have resulted from the defects of actin assembly, which further affected the meiotic spindle positioning. In addition, we showed that inhibition of PKC activity decreased FASCIN expression, indicating that FASCIN might be regulated by PKC. Taken together, our results provided evidence for the important role of FASCIN on actin filaments for spindle migration and polar body extrusion in mouse oocyte meiosis.


Subject(s)
Actin Cytoskeleton/metabolism , Carrier Proteins/metabolism , Meiosis , Microfilament Proteins/metabolism , Oocytes/metabolism , Polar Bodies/metabolism , Spindle Apparatus/metabolism , Actin Cytoskeleton/genetics , Animals , Carrier Proteins/genetics , Cells, Cultured , Female , Mice, Inbred ICR , Microfilament Proteins/genetics , Protein Kinase C/metabolism , Spindle Apparatus/genetics , rho GTP-Binding Proteins/metabolism
7.
Biochim Biophys Acta Mol Cell Res ; 1868(7): 119044, 2021 06.
Article in English | MEDLINE | ID: mdl-33865884

ABSTRACT

Cyclin D-CDK4/6 complex mediates the transition from the G1 to S phase in mammalian somatic cells. Meiotic oocytes pass through the G2/M transition and complete the first meiosis to reach maturation at the metaphase of meiosis II without intervening S phase, while Cyclin D-CDK4/6 complex is found to express during meiotic progression. Whether Cyclin D-CDK4/6 complex regulates meiotic cell cycle progression is not known. Here, we found its different role in oocyte meiosis: Cyclin D-CDK4/6 complex served as a regulator of spindle assembly checkpoint (SAC) to prevent aneuploidy in meiosis I. Inhibition of CDK4/6 kinases disrupted spindle assembly, chromosome alignment and kinetochore-microtubule attachments, but unexpectedly accelerated meiotic progression by inactivating SAC, consequently resulting in production of aneuploid oocytes. Further studies showed that the MPF activity decrease before first polar body extrusion was accelerated probably by inactivation of the SAC to promote ubiquitin-mediated cyclin B1 degradation. Taken together, these data reveal a novel role of Cyclin D-CDK4/6 complex in mediating control of the SAC in female meiosis I.


Subject(s)
Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , M Phase Cell Cycle Checkpoints/physiology , Aneuploidy , Animals , Cell Cycle Proteins/metabolism , Chromosome Segregation/physiology , Cyclin B1/metabolism , Female , Meiosis/physiology , Mesothelin , Metaphase/physiology , Mice , Mice, Inbred ICR , Oocytes/metabolism , Polar Bodies/metabolism , Spindle Apparatus/metabolism
8.
J Assist Reprod Genet ; 38(5): 1061-1068, 2021 May.
Article in English | MEDLINE | ID: mdl-33619678

ABSTRACT

PURPOSE: When rescue artificial oocyte activation (ROA) is performed on the day after intracytoplasmic sperm injection (ICSI) or later, embryonic development is poor and seldom results in live births. The efficacy of an early ROA after ICSI is unclear. Is early ROA effective in rescuing unfertilized oocytes that have not undergone second polar body extrusion several hours after ICSI? METHODS: We performed retrospective cohort study between October 2016 and September 2019, targeting 2891 oocytes in 843 cycles when ICSI was performed. We performed ROA with calcium ionophore on 395 of the 475 oocytes with no second polar extrusion 2.5-6 h after ICSI. RESULTS: The normal fertilization rate of ROA oocytes was significantly higher than non-ROA oocytes (65.8% vs 6.7%, P < 0.001). The blastocyst development rate in ROA oocytes was significantly lower than spontaneously activated oocytes (48.9% vs 67.2%, P < 0.001). The ROA oocyte implantation rate did not significantly differ from the spontaneously activated oocytes (36.0% vs 41.2%). We observed no differences in the implantation rates and blastocyst development rates over the 2.5-6 h from ICSI until ROA. CONCLUSION: Early ROA is effective, and the optimal timing appears to be 2.5-6 h after ICSI.


Subject(s)
Embryonic Development/genetics , Fertilization in Vitro , Live Birth/epidemiology , Oocytes/growth & development , Blastocyst/drug effects , Calcium Ionophores/pharmacology , Embryo Implantation/genetics , Embryo Transfer/trends , Embryonic Development/drug effects , Female , Humans , Male , Oocytes/drug effects , Polar Bodies/drug effects , Polar Bodies/metabolism , Sperm Injections, Intracytoplasmic/trends
9.
Methods Mol Biol ; 2218: 157-167, 2021.
Article in English | MEDLINE | ID: mdl-33606230

ABSTRACT

The polar body, with haploid DNA, is a small cell produced during the meiosis of an oocyte. Here, we describe the detailed procedures for the detection of the second polar body in zebrafish (Danio rerio) embryos after 10 min post fertilization. A polar body can be easily distinguished as a small dot with a DAPI-stained nucleus surrounded by Phalloidin-labeled F-actin in each fertilized zebrafish embryo.


Subject(s)
Fertilization/physiology , Polar Bodies/physiology , Actin Cytoskeleton/metabolism , Animals , Cell Nucleus/metabolism , Cell Nucleus/physiology , Female , Fertilization in Vitro/methods , Male , Meiosis/physiology , Oocytes/metabolism , Oocytes/physiology , Polar Bodies/metabolism , Zebrafish/metabolism , Zebrafish/physiology
10.
J Assist Reprod Genet ; 38(2): 531-537, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33405007

ABSTRACT

OBJECTIVE: Assisted oocyte activation (AOA) can restore fertilization rates after IVF/ICSI cycles with fertilization failure. AOA is an experimental technique, and its downstream effects remain poorly characterized. Clarifying the relationship between AOA and embryo, morphokinetics could offer complementary insights into the quality and viability of the embryos obtained with this technique. The aim of this study is to compare the preimplantation morphokinetic development of embryos derived from ICSI-AOA (experimental group) vs. ICSI cycles (control group). METHODS: A retrospective cohort study was carried out with 141 embryos from fresh oocyte donation cycles performed between 2013 and 2017; 41 embryos were derived from 7 ICSI-AOA cycles and 100 embryos from 18 ICSI cycles. Morphokinetic development of all embryos was followed using a time-lapse system. RESULTS: We show that embryos from both groups develop similarly for most milestones, with the exception of the time of second polar body extrusion (tPB2) and the time to second cell division (t3). CONCLUSIONS: We conclude that ionomycin mediated AOA does not seem to affect the morphokinetic pattern of preimplantation embryo development, despite the alterations found in tPB2 and t3, which could directly reflect the use of a Ca2+ ionophore as a transient and quick non-physiologic increase of free intracytoplasmic Ca2+.


Subject(s)
Embryo Transfer , Embryonic Development/genetics , Oocytes/metabolism , Reproductive Techniques, Assisted , Adult , Female , Fertilization in Vitro , Humans , Oocyte Donation , Oocytes/growth & development , Polar Bodies/metabolism , Pregnancy , Pregnancy Rate , Sperm Injections, Intracytoplasmic , Time-Lapse Imaging
11.
Genes (Basel) ; 11(12)2020 11 24.
Article in English | MEDLINE | ID: mdl-33255457

ABSTRACT

Polar body (PB) formation is an extreme form of unequal cell division that occurs in oocytes due to the eccentric position of the small meiotic spindle near the oocyte cortex. Prior to PB formation, a chromatin-centered process causes the cortex overlying the meiotic chromosomes to become polarized. This polarized cortical subdomain marks the site where a cortical protrusion or outpocket forms at the oocyte surface creating the future PBs. Using ascidians, we observed that PB1 becomes tethered to the fertilized egg via PB2, indicating that the site of PB1 cytokinesis directed the precise site for PB2 emission. We therefore studied whether the midbody remnant left behind following PB1 emission was involved, together with the egg chromatin, in defining the precise cortical site for PB2 emission. During outpocketing of PB2 in ascidians, we discovered that a small structure around 1 µm in diameter protruded from the cortical outpocket that will form the future PB2, which we define as the "polar corps". As emission of PB2 progressed, this small polar corps became localized between PB2 and PB1 and appeared to link PB2 to PB1. We tested the hypothesis that this small polar corps on the surface of the forming PB2 outpocket was the midbody remnant from the previous round of PB1 cytokinesis. We had previously discovered that Plk1::Ven labeled midbody remnants in ascidian embryos. We therefore used Plk1::Ven to follow the dynamics of the PB1 midbody remnant during meiosis II. Plk1::Ven strongly labeled the small polar corps that formed on the surface of the cortical outpocket that created PB2. Following emission of PB2, this polar corps was rich in Plk1::Ven and linked PB2 to PB1. By labelling actin (with TRITC-Phalloidin) we also demonstrated that actin accumulates at the midbody remnant and also forms a cortical cap around the midbody remnant in meiosis II that prefigured the precise site of cortical outpocketing during PB2 emission. Phalloidin staining of actin and immunolabelling of anti-phospho aPKC during meiosis II in fertilized eggs that had PB1 removed suggested that the midbody remnant remained within the fertilized egg following emission of PB1. Dynamic imaging of microtubules labelled with Ens::3GFP, MAP7::GFP or EB3::3GFP showed that one pole of the second meiotic spindle was located near the midbody remnant while the other pole rotated away from the cortex during outpocketing. Finally, we report that failure of the second meiotic spindle to rotate can lead to the formation of two cortical outpockets at anaphase II, one above each set of chromatids. It is not known whether the midbody remnant of PB1 is involved in directing the precise location of PB2 since our data are correlative in ascidians. However, a review of the literature indicates that PB1 is tethered to the egg surface via PB2 in several species including members of the cnidarians, lophotrochozoa and echinoids, suggesting that the midbody remnant formed during PB1 emission may be involved in directing the precise site of PB2 emission throughout the invertebrates.


Subject(s)
Meiosis/physiology , Polar Bodies/physiology , Actins/metabolism , Animals , Bivalvia/metabolism , Bivalvia/physiology , Chromatin/metabolism , Chromatin/physiology , Chromosomes/metabolism , Chromosomes/physiology , Cytokinesis/physiology , Oocytes/metabolism , Oocytes/physiology , Polar Bodies/metabolism , Spindle Apparatus/metabolism , Spindle Apparatus/physiology , Urochordata/metabolism , Urochordata/physiology , Zygote/metabolism , Zygote/physiology
12.
Syst Biol Reprod Med ; 66(6): 378-386, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32972244

ABSTRACT

The Origin Replication Complex subunit 4 (ORC4) is one in six subunits of the Origin Replication Complexes (ORCs) which is essential for initiating licensing at DNA replication origins and recruiting adaptor molecules necessary for various cellular processes. Previously, we reported that ORC4 also plays a vital role in polar body extrusion (PBE) during oogenesis in which half the chromosomes are extruded from the oocyte. We hypothesized that ORC4 might play a broader role in chromatin elimination. We tested its role in enucleation during the development of erythrocytes. Murine erythroleukemia (MEL) cells can be propagated in culture indefinitely and can be induced to enucleate their DNA by treatment with Vacuolin-1, thereby mimicking normal erythrocyte enucleation. We found that ORC4 appeared around the nuclei of the MEL cells with Vacuolin-1 treatment, gradually increasing in thickness before enucleation. We then tested whether ORC4 was required for MEL enucleation by down regulating ORC4 with siRNA-ORC4 during Vacuolin-1 treatment and found that this prevented MEL enucleation. These data are consistent with the model that ORC4 is required for erythroblast enucleation just as it is for oocyte PBE. They suggest a new model in which ORC4 expression is a marker for the initiation to the enucleation pathway.


Subject(s)
Cell Nucleus/metabolism , Erythroblasts/metabolism , Leukemia, Erythroblastic, Acute/metabolism , Oogenesis , Origin Recognition Complex/metabolism , Polar Bodies/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/pathology , DNA Replication , Erythroblasts/drug effects , Erythroblasts/pathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/pathology , Mice , Origin Recognition Complex/genetics
13.
Sci Rep ; 10(1): 6556, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32300113

ABSTRACT

To study the functional differences between maternal and paternal genomes in mammalian development, embryos with only one parental genome are often used. Androgenetic embryos are produced by the removal of maternal chromosomes before or after fertilization by techniques that require specialized skills and are associated with high risk of cellular damage. Here, we developed a novel method for producing androgenetic mouse embryos without the invasive enucleation process. We found that during in vitro fertilization in the presence of low-dose nocodazole, a microtubule destabilizing drug, whole oocyte chromosomes were extruded into the second polar body resulting in the production of androgenetic embryos. We further demonstrated that low-dose nocodazole decreased the spindle size and prevented chromosome segregation but did not compromise oocyte meiotic resumption. This led to the formation of a protrusion around the chromosomes, accumulation of protein regulator of cytokinesis 1 (PRC1) to the microtubules around the chromosomes, and assembly of a contractile ring at the neck region of the protrusion. Our method uses the intrinsic cytokinetic mechanism to exclude maternal chromatin from zygotes and may be applicable to other mammals.


Subject(s)
Embryo, Mammalian/metabolism , Spindle Apparatus/metabolism , Animals , Chromatin/metabolism , Chromosomes, Mammalian/metabolism , Cytokinesis/drug effects , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Female , Fertilization in Vitro , Genome , Male , Mice, Inbred C57BL , Models, Biological , Nocodazole/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Polar Bodies/drug effects , Polar Bodies/metabolism , Spindle Apparatus/drug effects , Zygote/drug effects , Zygote/metabolism
14.
Zygote ; 28(3): 217-222, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32156320

ABSTRACT

The aim of this study was to provide a more comprehensive understanding of 1PN intracytoplasmic sperm injection (ICSI) zygotes. To achieve this objective, we assessed whether all 1PN-derived embryos showed a similar morphokinetic pattern, and if the morphokinetic behaviour of 1PN-derived embryos was comparable with that of 2PN-derived embryos. In total, 149 1PN ICSI zygotes (study group) and 195 2PN ICSI zygotes (control group) were included in the study. Embryo development potential was evaluated in terms of blastocyst rate. Morphokinetic parameters, including the pronucleus diameter and kinetics of in vitro development, were also analyzed. Embryos derived from 1PN ICSI zygotes showed impaired development compared with 2PN-derived embryos, with blastocyst rates of 28.9% and 67.2%, respectively. The diameter of the pronucleus of 1PN zygotes was larger than that of 2PN zygotes. When compared with 2PN-derived embryos, those derived from 1PN zygotes had a visible pronucleus for a shorter time, in addition to a longer syngamy time and slower kinetic behaviour from two to nine cells. When 1PN-derived blastocysts and 2PN-derived blastocysts were compared, the developmental kinetics were similar in both groups, except for a delayed and longer duration of the compaction phase in 1PN-derived embryos. In conclusion, monopronucleated ICSI zygotes present differences in developmental capacity and morphokinetic behaviour compared with 2PN ICSI zygotes, showing particular morphokinetic parameters related to pronucleus formation. Only the 1PN ICSI-derived embryos that reached the blastocyst stage have similar morphokinetic development to blastocysts from 2PN zygotes.


Subject(s)
Blastocyst/cytology , Embryo Transfer/methods , Embryonic Development , Fertilization in Vitro/methods , Sperm Injections, Intracytoplasmic/methods , Zygote/cytology , Adult , Animals , Blastocyst/metabolism , Cell Nucleus/metabolism , Female , Humans , Male , Polar Bodies/metabolism , Pregnancy , Pregnancy Rate , Retrospective Studies , Time-Lapse Imaging/methods , Zygote/metabolism
15.
Gene ; 741: 144495, 2020 May 30.
Article in English | MEDLINE | ID: mdl-32088244

ABSTRACT

As a member of the chromosomal passenger complex, CDCA8 (cell division cycle associated 8) plays an important role in human mitosis, but its roles in human meiosis are unknown. Here, we show that CDCA8 expression is increased and its encoded protein has dynamic localization in human oocytes from germinal vesicle breakdown (GVBD) to metaphase Ⅱ (MⅡ), and that there are multipolar spindles, disordered chromosomes, and that microtubule assembly is affected after CDCA8 RNA interference (RNAi) in GV-stage oocytes. The GVBD and polar body extrusion (PBE) rates were not affected following CDCA8 depletion, but the PBE time was extended. There was no statistical difference between CDCA8 expression of oocytes from older and younger women, but the first polar body from older women was prone to chromosome abnormalities, and oocytes with such abnormalities had lower CDCA8 expression than oocytes with normal polar bodies. These results indicate that CDCA8 is associated with bipolar spindle formation, chromosome segregation, PBE during human oocyte meiosis, and that it may affect the incidence of aneuploidy embryos in older women.


Subject(s)
Cell Cycle Proteins/genetics , Meiosis/genetics , Oocytes/growth & development , Spindle Apparatus/genetics , Adult , Chromosome Segregation , Female , Gene Expression Regulation, Developmental , Humans , Mitosis/genetics , Oocytes/metabolism , Polar Bodies/metabolism , RNA Interference
16.
J Cell Biochem ; 121(7): 3547-3559, 2020 07.
Article in English | MEDLINE | ID: mdl-31898356

ABSTRACT

Oocyte apoptosis can be used as an indicator of oocyte quality and development competency. Phospholipase C (PLC) is a critical enzyme that participates in phosphoinositide metabolic regulation and performs many functions, including the regulation of reproduction. In this study, we aimed to explore whether PLC participates in the regulation of apoptosis in porcine oocytes and investigated its possible mechanism. In porcine oocytes, 0.5 µM U73122 (the PLC inhibitor) was considered to be the best concentration to facilitate maturation, and 0.5 µM m-3M3FBS (the PLC activator) was regarded as the most appropriate concentration to inhibit maturation. The percentage of cleavage and blastocysts treated with 0.5 µM U73122 was lower than that of the control group. Furthermore, the percentage of cleavage and blastocysts treated with 0.5 µM m-3M3FBS was higher than that of the control group. The relative PLC messenger RNA (mRNA) expression tested by a quantitative real-time polymerase chain reaction was found to be inhibited by 0.5 µM U73122 or activated by 0.5 µM m-3M3FBS. The relative mRNA abundance of BAK, BAX, CASP3, CASP8, and TP53 and protein abundance of Bak, cleaved caspase-3, caspase-8, and P53 was activated by U73122 or inhibited by m-3M3FBS, while the relative mRNA and protein level of BCL6 showed the opposite trend. The intracellular Ca2+ concentration increased and the expression of PLCB1 protein also increased in porcine oocytes when they were cultured with 0.5 µM m-3M3FBS for 44 hours. The abundance of proteins PKCß and CAMKIIα and the expression of several downstream genes (CDC42, NFATc1, NFATc2, NFκB, and NLK) were activated by m-3M3FBS or inhibited by U73122. Our findings indicate that PLC inhibits apoptosis and maturation in porcine oocytes. The intracellular Ca2+ concentration, two Ca2+ -sensitive proteins, and several downstream genes were positively regulated by PLC.


Subject(s)
Apoptosis/drug effects , Gene Expression Regulation, Developmental , Oocytes/drug effects , Phospholipase C beta/pharmacology , Animals , Blastocyst/cytology , Calcium/metabolism , Cell Nucleus/metabolism , Estrenes/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , In Vitro Techniques , Oocytes/metabolism , Ovary/metabolism , Polar Bodies/metabolism , Pyrrolidinones/pharmacology , RNA, Messenger/metabolism , Signal Transduction , Swine
17.
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
18.
Mol Reprod Dev ; 86(9): 1116-1125, 2019 09.
Article in English | MEDLINE | ID: mdl-31347225

ABSTRACT

Allicin, a chemical component of garlic, has strong antioxidant activity and is thought to exert antiaging effects in vitro. We investigated whether allicin treatment would protect porcine oocytes and embryos from postovulatory aging mediated by apoptosis and autophagy. The rates of oocyte survival and polar body extrusion in samples treated with 1 µM allicin (1 AL) were significantly higher than in untreated samples (0 AL). In addition, 1 AL prevented defects in spindle formation and chromosome alignment, as well as decreases in the expression of maturation markers, during in vitro aging. In this study, we considered allicin to be a regulator of autophagy rather than an antioxidant or antiapoptotic agent. At the embryo level, although the cleavage rate after parthenogenetic activation was similar in all groups, the blastocyst formation rate was higher in the 1 AL group than in the 0 AL group. Our findings demonstrate that allicin effectively prevents the deterioration of porcine oocytes during aging in vitro, and could therefore be used to improve the quality of aged oocytes used in in vitro experiments.


Subject(s)
Apoptosis/drug effects , Autophagic Cell Death/drug effects , Blastocyst/metabolism , Cellular Senescence/drug effects , Parthenogenesis/drug effects , Polar Bodies/metabolism , Sulfinic Acids/pharmacology , Animals , Disulfides , Swine
19.
Cell Reprogram ; 21(3): 129-140, 2019 06.
Article in English | MEDLINE | ID: mdl-31084435

ABSTRACT

The present study was designed to investigate the comprehensive function of maternal factors of primordial germ cell 7 (PGC7) and POU5F1-POU class 5 homeobox 1 (OCT4), as well as the epigenetic modification roles on the mitosis for the extrusion of first polar body (PB1) in pig maturated oocytes. First, the common distribution of histone modifications, including H3K4me2, H3K27me3, H3K9me2, and H4K12ac and DNA methylation, were detected at the high level in the nucleus. However, only one part of the chromosome was higher methylated or acetylated when the mitosis happened to extrude the PB1. When the mitosis was inhibited by the cytochalasin B (CB) treatment, the expression of PGC7, OCT4, DNA methyltransferase1 (DNMT1), DNA methyltransferase3b (DNMT3b), tet methylcytosine dioxygenase 1 (TET1), tet methylcytosine dioxygenase 2 (TET2), and tet methylcytosine dioxygenase 3 (TET3) could be inhibited (p < 0.01), and no concentrated expression of the PGC7 and OCT4 was observed on the chromosome, but the levels of H3K9me2 and H4K12ac were higher. In addition, when the trichostatin A was performed on the in vitro maturation, the extrusion of the PB1 was inhibited too. And the histone methylation (H3K9me2 and H3K27me3) could be detected all the time with relative higher level and no demethylation could be observed. However, the expression of PGC7 and OCT4 was lower in the chromosome. It might indicate that the maternal factor of PGC7 and histone modification that included H4K12ac and H3K9me2 could regulate the extrusion of the PB1 and play an important role in the maturation of pig oocytes.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Octamer Transcription Factor-3/metabolism , Polar Bodies/metabolism , Sus scrofa/genetics , Animals , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Oocytes/metabolism , Promoter Regions, Genetic , DNA Methyltransferase 3B
20.
Proc Natl Acad Sci U S A ; 116(20): 9883-9892, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31010926

ABSTRACT

Mammalian oocytes carry specific nongenetic information, including DNA methylation to the next generation, which is important for development and disease. However, evaluation and manipulation of specific methylation for both functional analysis and therapeutic purposes remains challenging. Here, we demonstrate evaluation of specific methylation in single oocytes from its sibling first polar body (PB1) and manipulation of specific methylation in single oocytes by microinjection-mediated dCas9-based targeted methylation editing. We optimized a single-cell bisulfite sequencing approach with high efficiency and demonstrate that the PB1 carries similar methylation profiles at specific regions to its sibling oocyte. By bisulfite sequencing of a single PB1, the methylation information regarding agouti viable yellow (Avy )-related coat color, as well as imprinting linked parthenogenetic development competency, in a single oocyte can be efficiently evaluated. Microinjection-based dCas9-Tet/Dnmt-mediated methylation editing allows targeted manipulation of specific methylation in single oocytes. By targeted methylation editing, we were able to reverse Avy -related coat color, generate full-term development of bimaternal mice, and correct familial Angelman syndrome in a mouse model. Our work will facilitate the investigation of specific methylation events in oocytes and provides a strategy for prevention and correction of maternally transmitted nongenetic disease or disorders.


Subject(s)
DNA Methylation , Genetic Engineering/methods , Polar Bodies/metabolism , Animals , Female , Mice, Inbred C57BL , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...