Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.524
Filter
1.
Nature ; 607(7920): 756-761, 2022 07.
Article in English | MEDLINE | ID: mdl-35859172

ABSTRACT

Oocytes form before birth and remain viable for several decades before fertilization1. Although poor oocyte quality accounts for most female fertility problems, little is known about how oocytes maintain cellular fitness, or why their quality eventually declines with age2. Reactive oxygen species (ROS) produced as by-products of mitochondrial activity are associated with lower rates of fertilization and embryo survival3-5. Yet, how healthy oocytes balance essential mitochondrial activity with the production of ROS is unknown. Here we show that oocytes evade ROS by remodelling the mitochondrial electron transport chain through elimination of complex I. Combining live-cell imaging and proteomics in human and Xenopus oocytes, we find that early oocytes exhibit greatly reduced levels of complex I. This is accompanied by a highly active mitochondrial unfolded protein response, which is indicative of an imbalanced electron transport chain. Biochemical and functional assays confirm that complex I is neither assembled nor active in early oocytes. Thus, we report a physiological cell type without complex I in animals. Our findings also clarify why patients with complex-I-related hereditary mitochondrial diseases do not experience subfertility. Complex I suppression represents an evolutionarily conserved strategy that allows longevity while maintaining biological activity in long-lived oocytes.


Subject(s)
Electron Transport Complex I , Mitochondria , Oocytes , Reactive Oxygen Species , Animals , Electron Transport , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Female , Humans , Mitochondria/metabolism , Oocytes/cytology , Oocytes/enzymology , Oocytes/metabolism , Proteomics , Unfolded Protein Response , Xenopus laevis
2.
J Mol Histol ; 53(1): 63-74, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34741214

ABSTRACT

The effects of culture media on DNA methylation process, which is one of the epigenetic mechanisms, have not been clearly elucidated although it is known that in vitro culture conditions alter epigenetic mechanisms. This study was designed to address the question: does embryo culture media approach, sequential or single step, differentially affect DNA methylating enzymes and global DNA methylation. Mouse zygotes were cultured either in single step or sequential culture media until the blastocyst stage and in vivo developed blastocyst were utilized as control. Similarly, GV stage oocytes were in vitro matured either in single step or first step of sequential culture media. In vivo matured MII oocytes were used as control. The expression levels and cellular localization of Dnmt1 and 3a enzymes were analyzed by immunofluorescence and western blot analysis while global DNA methylation was evaluated by immunofluorescence. We found that signal intensities of Dnmt1 and Dnmt3a enzymes were significantly low in embryos or oocytes cultured in sequential media compared to single step media and control, which were comparable amongst themself. Similarly, global DNA methylation level in single step media and control groups was comparable but both was higher than the sequential media. This study demonstrated that composition of culture media may differentially affect DNA methylation levels in mouse embryos and oocytes. Since abnormal DNA methylation may cause aberrant oocyte or embryo development, we think that further studies are needed to test human embryos and oocyte, and to explain molecular mechanisms.


Subject(s)
Culture Media/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , DNA Methyltransferase 3A/metabolism , Embryo Culture Techniques/methods , Embryo, Mammalian/enzymology , Oocytes/enzymology , Animals , Blastocyst/cytology , Blastocyst/enzymology , Blotting, Western , Embryonic Development/physiology , Female , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred BALB C , Oocytes/cytology , Pregnancy
3.
Mol Hum Reprod ; 27(11)2021 11 02.
Article in English | MEDLINE | ID: mdl-34590701

ABSTRACT

PIWI-interacting small RNAs (piRNAs) maintain genome stability in animal germ cells, with a predominant role in silencing transposable elements. Mutations in the piRNA pathway in the mouse uniformly lead to failed spermatogenesis and male sterility. By contrast, mutant females are fertile. In keeping with this paradigm, we previously reported male sterility and female fertility associated with loss of the enzyme HENMT1, which is responsible for stabilising piRNAs through the catalysation of 3'-terminal 2'-O-methylation. However, the Henmt1 mutant females were poor breeders, suggesting they could be subfertile. Therefore, we investigated oogenesis and female fertility in these mice in greater detail. Here, we show that mutant females indeed have a 3- to 4-fold reduction in follicle number and reduced litter sizes. In addition, meiosis-II mutant oocytes display various spindle abnormalities and have a dramatically altered transcriptome which includes a down-regulation of transcripts required for microtubule function. This down-regulation could explain the spindle defects observed with consequent reductions in litter size. We suggest these various effects on oogenesis could be exacerbated by asynapsis, an apparently universal feature of piRNA mutants of both sexes. Our findings reveal that loss of the piRNA pathway in females has significant functional consequences.


Subject(s)
Fertility , Infertility, Female/enzymology , Meiosis , Methyltransferases/metabolism , Oocytes/enzymology , Oogenesis , RNA, Small Interfering/metabolism , Animals , Female , Gene Expression Regulation, Developmental , Infertility, Female/genetics , Infertility, Female/physiopathology , Methyltransferases/genetics , Mice , RNA, Small Interfering/genetics , Transcriptome
4.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068371

ABSTRACT

This study aimed to describe glutathione peroxidase 4 (GPx4) in rat oocytes, preimplantation embryos, and female genital organs. After copulation, Sprague Dawley female rats were euthanized with anesthetic on the first (D1), third (D3), and fifth days of pregnancy (D5). Ovaries, oviducts, and uterine horns were removed, and oocytes and preimplantation embryos were obtained. Immunohistochemical, immunofluorescent, and Western blot methods were employed. Using immunofluorescence, we detected GPx4 in both the oocytes and preimplantation embryos. Whereas in the oocytes, GPx4 was homogeneously diffused, in the blastomeres, granules were formed, and in the blastocysts, even clusters were present mainly around the cell nuclei. Employing immunohistochemistry, we detected GPx4 inside the ovary in the corpus luteum, stroma, follicles, and blood vessels. In the oviduct, the enzyme was present in the epithelium, stroma, blood vessels, and smooth muscles. In the uterus, GPx4 was found in the endometrium, myometrium, blood vessels, and stroma. Moreover, we observed GPx4 positive granules in the uterine gland epithelium on D1 and D3 and cytoplasm of fibroblasts forming in the decidua on D5. Western blot showed the highest GPx4 levels in the uterus and the lowest levels in the ovary. Our results show that the GPx4 is necessary as early as in the preimplantation development of a new individual because we detected it in an unfertilized oocyte in a blastocyst and not only after implantation, as was previously thought.


Subject(s)
Blastocyst/enzymology , Embryo Implantation , Embryonic Development , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Oocytes/enzymology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Animals , Blastocyst/cytology , Endometrium/enzymology , Female , Male , Oocytes/cytology , Ovary/enzymology , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Uterus/enzymology
5.
PLoS Genet ; 17(5): e1009567, 2021 05.
Article in English | MEDLINE | ID: mdl-34014923

ABSTRACT

The widely conserved kinase Aurora B regulates important events during cell division. Surprisingly, recent work has uncovered a few functions of Aurora-family kinases that do not require kinase activity. Thus, understanding this important class of cell cycle regulators will require strategies to distinguish kinase-dependent from independent functions. Here, we address this need in C. elegans by combining germline-specific, auxin-induced Aurora B (AIR-2) degradation with the transgenic expression of kinase-inactive AIR-2. Through this approach, we find that kinase activity is essential for AIR-2's major meiotic functions and also for mitotic chromosome segregation. Moreover, our analysis revealed insight into the assembly of the ring complex (RC), a structure that is essential for chromosome congression in C. elegans oocytes. AIR-2 localizes to chromosomes and recruits other components to form the RC. However, we found that while kinase-dead AIR-2 could load onto chromosomes, other components were not recruited. This failure in RC assembly appeared to be due to a loss of RC SUMOylation, suggesting that there is crosstalk between SUMOylation and phosphorylation in building the RC and implicating AIR-2 in regulating the SUMO pathway in oocytes. Similar conditional depletion approaches may reveal new insights into other cell cycle regulators.


Subject(s)
Aurora Kinase B/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Chromosome Segregation , Oocytes/enzymology , Animals , Caenorhabditis elegans/genetics , Chromosomes/metabolism , Meiosis/genetics , Mitosis/genetics , Oocytes/cytology , Phosphorylation , Reproducibility of Results , Spindle Apparatus/enzymology , Sumoylation
6.
Pol J Vet Sci ; 24(1): 93-99, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33847105

ABSTRACT

It is known that the basic variable in the cellular environment is temperature and low temperature decreases cellular metabolism rate. Also, low cellular metabolic activity reduces oxidative stress, resulting in low ROS production. The aim of this study was therefore to investigate the effect of 36.5°C (low) and 38.5°C (conventional) incubation temperatures during IVM on glutathione peroxidase activity of oocytes and blastocysts following fertilization. Bovine oocytes were matured in medium-199 for 22 hours at either 36.5°C or 38.5°C and they were subjected to in vitro fertilization (IVF). Putative zygotes were then transferred randomly into SOFaa embryo culture media with or without antioxidant (a mixture of GSH and SOD) until development to the blastocyst stage. Glutathione peroxidase enzyme (GSH-Px) activity was lower (p⟨0.05) in oocytes matured at low temperature than those of conventional temperature. Similarly, GSH-Px activity was lower (p⟨0.05) in blastocysts, which were obtained from oocytes matured at low temperature and cultured in antioxidants-supplemented embryo media. The GSH-Px activity of blastocysts, obtained from oocytes matured in low temperature, cultured in antioxidants-free embryo media was similar to blastocysts obtained from oocytes matured in conventional temperature, cultured in antioxidants-supplemented embryo media. The results of the present study show that decreasing the in vitro maturation temperature decreases antioxidant enzyme activity in both oocyte and blastocyst. Additionally, maturation of bovine oocytes at 36.5°C incubation temperature may provide an optimal thermal condition for the enzymatic antioxidant system of both oocytes and blastocyst.


Subject(s)
Cattle/embryology , Embryo Culture Techniques/veterinary , Glutathione Peroxidase/metabolism , In Vitro Oocyte Maturation Techniques/veterinary , Oocytes/enzymology , Oocytes/metabolism , Animals , Cattle/physiology , Temperature
7.
Reprod Sci ; 28(9): 2630-2640, 2021 09.
Article in English | MEDLINE | ID: mdl-33725312

ABSTRACT

It remained unknown whether HDAC6 affected the histone deacetylation of in vitro maturation oocytes and the reprogramming of nuclear transplantation in pig. Our results indicated that HDAC6 specific inhibition did not affect overall HDAC activity and meiosis process, which increased histone H3K9/K14 and H4K8 acetylation of porcine in vitro maturation oocytes and pseudo-pronucleus embryos. HDAC6 inhibition also significantly enhanced the cleavage and blastocyst of nuclear transfer embryos (0.81 ± 0.12 vs. 0.68 ± 0.12 and 0.46 ± 0.19; 0.73 ± 0.13 vs. 0.63 ± 0.18 and 0.40 ± 0.16, P<0.05). The inhibition of HDAC6 significantly enhanced histone H3K9/K14 and H4K8 acetylation, and upregulated the OCT4 and CDX2 expressions (1.83 ± 0.16 vs. 1.00 ± 0.00 %; 2.07 ± 0.09 vs. 1.00 ± 0.00; P<0.05) in porcine SCNT blastocysts. Interestingly, HDAC6 inhibition significantly increased the pseudo-pronucleus volume during somatic cell reprogramming. Thus, HDAC6 was required for porcine histone deacetylation during the in vitro maturation and pseudo-pronucleus stages. HDAC6 inhibition improved the in vitro development of nuclear transfer embryos. HDAC6 may restrict the reprogramming of somatic nuclear transfer by regulating pseudo-pronucleus expansion. We need further research to confirm this in the future.


Subject(s)
Blastocyst/enzymology , Cellular Reprogramming , Histone Deacetylase 6/metabolism , Histones/metabolism , In Vitro Oocyte Maturation Techniques , Nuclear Transfer Techniques , Oocytes/enzymology , Acetylation , Animals , Blastocyst/drug effects , Bufexamac/pharmacology , Cells, Cultured , Embryo Culture Techniques , Embryonic Development , Female , Gene Expression Regulation, Developmental , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Oocytes/drug effects , Protein Processing, Post-Translational , Sus scrofa
8.
Nature ; 587(7832): 139-144, 2020 11.
Article in English | MEDLINE | ID: mdl-33116310

ABSTRACT

Zygotic genome activation (ZGA) is the first transcription event in life1. However, it is unclear how RNA polymerase is engaged in initiating ZGA in mammals. Here, by developing small-scale Tn5-assisted chromatin cleavage with sequencing (Stacc-seq), we investigated the landscapes of RNA polymerase II (Pol II) binding in mouse embryos. We found that Pol II undergoes 'loading', 'pre-configuration', and 'production' during the transition from minor ZGA to major ZGA. After fertilization, Pol II is preferentially loaded to CG-rich promoters and accessible distal regions in one-cell embryos (loading), in part shaped by the inherited parental epigenome. Pol II then initiates relocation to future gene targets before genome activation (pre-configuration), where it later engages in full transcription elongation upon major ZGA (production). Pol II also maintains low poising at inactive promoters after major ZGA until the blastocyst stage, coinciding with the loss of promoter epigenetic silencing factors. Notably, inhibition of minor ZGA impairs the Pol II pre-configuration and embryonic development, accompanied by aberrant retention of Pol II and ectopic expression of one-cell targets upon major ZGA. Hence, stepwise transition of Pol II occurs when mammalian life begins, and minor ZGA has a key role in the pre-configuration of transcription machinery and chromatin for genome activation.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Genome/genetics , RNA Polymerase II/metabolism , Zygote/metabolism , Alleles , Animals , Chromatin/genetics , Chromatin/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Embryo, Mammalian/metabolism , Epigenome/genetics , Female , Male , Maternal Inheritance/genetics , Mice , Mice, Inbred C57BL , Oocytes/enzymology , Oocytes/metabolism , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , Zygote/cytology , Zygote/enzymology
9.
J Reprod Immunol ; 142: 103191, 2020 11.
Article in English | MEDLINE | ID: mdl-32937223

ABSTRACT

OBJECTIVE: Follicular development can be disturbed due to many factors, including having polycystic ovaries. Aberrant expression of genes involved in steroidogenesis pathway could lead to aberrant oocyte development. In this study, the gene expression levels of a number of genes that is functioning in steroidogenesis pathway were investigated. MATERIALS AND METHODS: The spare oocytes were collected from NEU Hospital IVF Center following controlled ovarian stimulation cycle. RNA was extracted using RNA/DNA Purification Kit (Norgen, Canada) and reverse transcription was performed using TruScript First Strand cDNA Synthesis Kit (Norgen, Canada). Real time PCR was conducted using LightCycler® 480 SYBR Green I Master (Roche, UK). RESULTS AND CONCLUSION: The expression levels of CYP11, CYP17, CYP19, HSD17B1, HSD3B2 and ACTB were detected in human MII stage oocytes obtained from oocyte donors aged between 18-30 years. The number of follicles and oocytes collected from the patients with polycystic ovaries were slightly higher compared to the control group. The expression level of CYP11A1 was shown to be statistically different in the oocytes obtained from the patients who do not have polycystic ovaries (p < 0.05), whereas statistically significant expression levels were observed for CYP17 in the oocytes obtained from patients with polycystic ovaries (p < 0.05). The expression level of HSD17B1 was also shown to be statistically different in the oocytes (p < 0.05). The extrapolation of the results indicates that the genes involved in steroidogenesis pathway are altered in cases of polycystic ovaries. Thus, it may have a role in the development of polycystic ovaries.


Subject(s)
Androgens/biosynthesis , Hyperandrogenism/pathology , Oocytes/enzymology , Polycystic Ovary Syndrome/complications , Adolescent , Adult , Case-Control Studies , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Estradiol Dehydrogenases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Humans , Oocytes/growth & development , Oocytes/pathology , Oogenesis , Ovarian Follicle/pathology , Polycystic Ovary Syndrome/pathology , Steroid 17-alpha-Hydroxylase/metabolism , Young Adult
10.
J Assist Reprod Genet ; 37(8): 1807-1814, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32767205

ABSTRACT

PURPOSE: Reduction in methylenetetrahydrofolate reductase (MTHFR) activity due to genetic variations in the MTHFR gene has been controversially implicated in subfertility in human in vitro fertilization. However, there is no direct gene-knockdown study of embryonic MTHFR to assess its involvement in mammalian preimplantation development. The purpose of this study is to investigate expression profiles and functional roles of MTHFR in bovine preimplantation development. METHODS: Reverse transcription-quantitative PCR (RT-qPCR) and analysis of publicly available RNA-seq data were performed to reveal expression levels of MTHFR during bovine preimplantation development. We knocked down MTHFR by siRNA-mediated RNA interference from the 8- to 16-cell stage and assessed the effects on preimplantation development. RESULTS: The RT-qPCR analysis showed relatively high MTHFR expression at the GV oocyte stage, which was decreased toward the 8- to 16-cell stage and then slightly restored at the blastocyst stage. Public data-based analysis also showed the similar pattern of expression with substantial embryonic expression at the blastocyst stage. MTHFR knockdown reduced the blastocyst rate (P < 0.01) and the numbers of total (P < 0.0001), trophectoderm (P < 0.0001), and inner cell mass (P < 0.001) cells. CONCLUSION: The results indicate that embryonic MTHFR is indispensable for normal blastocyst development. The findings provide insight into the debatable roles of MTHFR in fertility and may be applicable for the improvement of care for early embryos via modulation of surrounding folate-related nutritional conditions in vitro and/or in utero, depending on the parental and embryonic MTHFR genotype.


Subject(s)
Blastocyst/enzymology , Embryonic Development/genetics , Fertility/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Animals , Blastocyst/metabolism , Blastocyst/ultrastructure , Cattle , Female , Fertility/physiology , Gene Expression Regulation, Developmental/genetics , Humans , Oocytes/enzymology , Oocytes/growth & development , Oocytes/ultrastructure , RNA, Small Interfering
11.
Cell Death Dis ; 11(6): 490, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32606310

ABSTRACT

HFM1 (helicase for meiosis 1) is widely recognized as an ATP-dependent DNA helicase and is expressed mainly in germ-line cells. HFM1 is a candidate gene of premature ovarian failure (POF), hence it is also known as POF9. However, the roles of HFM1 in mammalian oocytes remain uncertain. To investigate the functions of HFM1, we established a conditional knockout (cKO) mouse model. Specific knockout of Hfm1 in mouse oocytes from the primordial follicle stage resulted in depletion of ovarian follicular reserve and subfertility of mice. In particular, abnormal spindle, misaligned chromosomes, loss of cortical actin cap, and failing polar body extrusion were readily observed in Hfm1-cKO oocytes. Further studies indicated that in addition to its cytoplasmic distribution, Hfm1 accumulated at the spindle poles, colocalized with the Golgi marker protein, GM130. Generally, GM130 signals overlapped with p-Mapk at the two spindle poles to regulate meiotic spindle assembly and asymmetric division. In this research, centrosome associated proteins, such as GM130 and p-Mapk, detached from the spindle poles in Hfm1-cKO oocytes. In conclusion, our data suggest that Hfm1 participates in Golgi-associated spindle assembly and division in mouse oocyte meiosis. These findings provide clues for pathogenesis of POF.


Subject(s)
DNA Helicases/metabolism , Golgi Apparatus/metabolism , Meiosis , Oocytes/cytology , Oocytes/metabolism , Spindle Apparatus/metabolism , Animals , Fertility , MAP Kinase Signaling System , Mice, Inbred C57BL , Mice, Knockout , Oocytes/enzymology , Organ Specificity
12.
Eur J Pharmacol ; 883: 173293, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32663542

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDEs) are group of enzymes responsible for the hydrolysis of cyclic adenosine 3', 5' monophosphate (cAMP) and cyclic guanosine 3', 5' monophosphate (cGMP) levels in wide variety of cell types. These PDEs are detected in encircling granulosa cells or in oocyte with in follicular microenvironment and responsible for the decrease of cAMP and cGMP levels in mammalian oocytes. A transient decrease of cAMP level initiates downstream pathways to cause spontaneous meiotic resumption from diplotene arrest and induces oocyte maturation. The nonspecific PDE inhibitors (caffeine, pentoxifylline, theophylline, IBMX etc.) as well as specific PDE inhibitors (cilostamide, milrinone, org 9935, cilostazol etc.) have been used to elevate cAMP level and inhibit meiotic resumption from diplotene arrest and oocyte maturation, ovulation, fertilization and pregnancy rates both in vivo as well as under in vitro culture conditions. The PDEs inhibitors are used as powerful experimental tools to demonstrate cyclic nucleotide mediated changes in ovarian functions and thereby fertility. Indeed, non-hormonal nature and reversible effects of nonspecific as well as specific PDE inhibitors hold promise for the development of novel therapeutic drugs for female fertility regulation.


Subject(s)
Fertility Agents, Female/therapeutic use , Fertility/drug effects , Infertility, Female/drug therapy , Oocytes/drug effects , Ovary/drug effects , Phosphodiesterase Inhibitors/therapeutic use , Animals , Female , Humans , Infertility, Female/enzymology , Infertility, Female/physiopathology , Oocytes/enzymology , Ovary/enzymology , Ovary/physiopathology , Ovulation/drug effects , Pregnancy
13.
Reprod Fertil Dev ; 32(7): 690-696, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32317093

ABSTRACT

We aimed to analyse the histone acetylation status and expression profile of genes involved in histone acetylation (histone acetyltransferase 1 (HAT1), lysine acetyltransferase 2A (KAT2A), histone deacetylase 1(HDAC1), HDAC2 and HDAC3) in bovine oocytes of different competences during invitro maturation (IVM). Cumulus-oocyte complexes were recovered from two groups of follicles: minor follicles (1.0-3.0mm in diameter), classified as low competence (LC) and large follicles (6.0-8.0mm in diameter) classified as high competence (HC). Oocytes were submitted to IVM for 0, 8 and 24h and stored for analysis. Acetylation status of histone H4 on lysine K5, K6, K12 and K16 was assessed by immunohistochemistry. For gene expression, mRNA levels were determined by real-time quantitative polymerase chain reaction. All oocytes, regardless of their competence, showed a gradual decrease (P<0.05) in acetylation signals during IVM. From 0 to 8h of maturation, an increase (P<0.05) in the relative abundance of HAT1 mRNA was observed only in the HC oocytes. In this group, higher (P<0.05) mRNA levels of HDAC1 at 8h of maturation were also observed. In conclusion, in the present study, LC oocytes were shown to have adequate acetylation levels for the resumption and progression of meiosis; however, these oocytes do not have the capacity to synthesise RNA during IVM as the HC oocytes do.


Subject(s)
Cattle , Histone Acetyltransferases/metabolism , In Vitro Oocyte Maturation Techniques/veterinary , Oocytes/enzymology , Acetylation , Animals , Cumulus Cells/physiology , Female , Histone Acetyltransferases/genetics , Histones/metabolism , Lysine/metabolism , Oocytes/growth & development , Oogenesis/genetics , Oogenesis/physiology , RNA, Messenger/analysis
14.
Cell Death Differ ; 27(8): 2468-2483, 2020 08.
Article in English | MEDLINE | ID: mdl-32094512

ABSTRACT

KIAA1429 (also known as vir-like m6A methyltransferase-associated protein (VIRMA)), a newly identified component of the RNA m6A methyltransferase complex, plays critical roles in guiding region-selective m6A deposition. However, in mammals, whether KIAA1429 mediates RNA m6A regulatory pathway functions in vivo remains unknown. Here, we show that the Kiaa1429-specific deficiency in oocytes resulted in female infertility with defective follicular development and fully grown germinal vesicle (GV) oocytes failing to undergo germinal vesicle breakdown (GVBD) and consequently losing the ability to resume meiosis. The oocyte growth is accompanied by the accumulation of abundant RNAs and posttranscriptional regulation. We found that the loss of Kiaa1429 could also lead to abnormal RNA metabolism in GV oocytes. RNA-seq profiling revealed that Kiaa1429 deletion altered the expression pattern of the oocyte-derived factors essential for follicular development. In addition, our data show that the conditional depletion of Kiaa1429 decreased the m6A levels in oocytes and mainly affected the alternative splicing of genes associated with oogenesis. In summary, the m6A methyltransferase KIAA1429-mediated RNA metabolism plays critical roles in folliculogenesis and the maintenance of oocyte competence.


Subject(s)
Methyltransferases/metabolism , Oocytes/cytology , Oocytes/enzymology , Ovarian Follicle/embryology , Ovarian Follicle/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Alternative Splicing/genetics , Animals , Cell Nucleus/metabolism , Cell Proliferation , Female , Fertility , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Methyltransferases/genetics , Mice, Inbred C57BL , Models, Biological , Organogenesis/genetics , Ovarian Follicle/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors/metabolism
15.
Cells ; 9(2)2020 02 07.
Article in English | MEDLINE | ID: mdl-32046180

ABSTRACT

Meiosis is required to reduce to haploid the diploid genome content of a cell, generating gametes-oocytes and sperm-with the correct number of chromosomes. To achieve this goal, two specialized cell divisions without intermediate S-phase are executed in a time-controlled manner. In mammalian female meiosis, these divisions are error-prone. Human oocytes have an exceptionally high error rate that further increases with age, with significant consequences for human fertility. To understand why errors in chromosome segregation occur at such high rates in oocytes, it is essential to understand the molecular players at work controlling these divisions. In this review, we look at the interplay of kinase and phosphatase activities at the transition from metaphase-to-anaphase for correct segregation of chromosomes. We focus on the activity of PP2A-B56, a key phosphatase for anaphase onset in both mitosis and meiosis. We start by introducing multiple roles PP2A-B56 occupies for progression through mitosis, before laying out whether or not the same principles may apply to the first meiotic division in oocytes, and describing the known meiosis-specific roles of PP2A-B56 and discrepancies with mitotic cell cycle regulation.


Subject(s)
Meiosis , Oocytes/cytology , Oocytes/enzymology , Protein Phosphatase 2/metabolism , Animals , Kinetochores/metabolism , Mice , Microtubules/metabolism , Spindle Apparatus/metabolism
16.
Cells ; 9(1)2020 01 17.
Article in English | MEDLINE | ID: mdl-31963573

ABSTRACT

The role of hydrogen sulfide (H2S) is addressed in Xenopuslaevis oocytes. Three enzymes involved in H2S metabolism, cystathionine ß-synthase, cystathionine γ-lyase, and 3-mercaptopyruvate sulfurtransferase, were detected in prophase I and metaphase II-arrested oocytes and drove an acceleration of oocyte meiosis resumption when inhibited. Moreover, meiosis resumption is associated with a significant decrease in endogenous H2S. On another hand, a dose-dependent inhibition was obtained using the H2S donor, NaHS (1 and 5 mM). NaHS impaired translation. NaHS did not induce the dissociation of the components of the M-phase promoting factor (MPF), cyclin B and Cdk1, nor directly impacted the MPF activity. However, the M-phase entry induced by microinjection of metaphase II MPF-containing cytoplasm was diminished, suggesting upstream components of the MPF auto-amplification loop were sensitive to H2S. Superoxide dismutase and catalase hindered the effects of NaHS, and this sensitivity was partially dependent on the production of reactive oxygen species (ROS). In contrast to other species, no apoptosis was promoted. These results suggest a contribution of H2S signaling in the timing of amphibian oocytes meiosis resumption.


Subject(s)
Hydrogen Sulfide/metabolism , Maturation-Promoting Factor/metabolism , Meiosis/drug effects , Oocytes/metabolism , Sulfides/pharmacology , Animals , Apoptosis/drug effects , Catalase/metabolism , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Cyclin B/metabolism , Cystathionine beta-Synthase/antagonists & inhibitors , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/metabolism , Cytoplasm/metabolism , Female , Meiotic Prophase I/drug effects , Metaphase/drug effects , Oocytes/chemistry , Oocytes/enzymology , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sulfides/metabolism , Sulfurtransferases/antagonists & inhibitors , Sulfurtransferases/metabolism , Superoxide Dismutase/metabolism , Xenopus Proteins/metabolism , Xenopus laevis , cdc25 Phosphatases/metabolism
17.
Reprod Biol ; 20(1): 48-62, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31889629

ABSTRACT

The present study evaluated the effects of the blockade of meiosis in bovine oocytes by the cyclin-dependent kinase inhibitors roscovitine (ROS) and butyrolactone-I (BL-I) on nuclear maturation and extracellular signal-regulated kinase 1/2 (ERK1/2), cyclin B1 and p34cdc2 protein expression and localization. We also evaluated ultrastructural changes in oocytes. Immature oocytes were obtained from slaughtered bovines and divided into: (1) control (oocytes for in vitro maturation only in tissue culture medium-199 for 24 h), (2) oocytes that were treated with 12.5µMROS for 6 h, (3) oocytes that were treated with 50µMBL-I for 6 h and (4) oocytes that were treated with 6.25 µMROS+25 µMBL-I for 6 h. Incubation with inhibitors was followed by the reversal of blockade for 18 h. Oocytes then underwent immunohistochemical analysis to visualize chromatin and assess ERK1/2, cyclin B1 and p34cdc2 localization/expression, followed by preparation of the cells for ultrastructure analysis by electron microscopy. The groups at 6 h of maturation and before IVM exhibited the lowest number of oocytes in metaphase I. ROS group had the highest number of degenerating oocytes (p < 0.05). After maturation, majority of oocytes were in metaphaseII with no differences among groups (p> 0.05). ERK1/2, cyclin B1 and p34cdc2 expression differed throughout inhibition and oocyte maturation (p < 0.05). No difference was observed in the localization of these proteins in the ooplasm. No ultrastructural changes in oocytes were observed between treatments, with the exception of treatment with drugs that augmented lipid metabolism (p < 0.05). Results indicate that the effects of CDK1 inhibitors are reversible in bovine oocytes, indicated by nuclear, cytoplasmic, and molecular maturation parameters.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Oocytes/enzymology , 4-Butyrolactone/analogs & derivatives , Animals , CDC2 Protein Kinase/metabolism , Cattle , Cyclin B1/metabolism , Female , Meiosis , Mitogen-Activated Protein Kinase 3/metabolism , Oocytes/ultrastructure , Roscovitine
18.
Cell Cycle ; 19(3): 354-362, 2020 02.
Article in English | MEDLINE | ID: mdl-31910069

ABSTRACT

The smallest histone deacetylase (HDAC) and the solely member of class IV, HDAC11, is reported to regulate mitosis process and tumorigenesis, yet its roles in meiosis process remain unknown. In the present study, we first analyzed the expression of HDAC11 in mouse oocytes. HDAC11 showed gradual lower expression from GV (Germinal Vesicle) to MII (Metaphase II) stage oocytes. Then, the specific inhibitor of HDAC11, JB3-22 was used to explore the role of HDAC11 during mouse oocytes maturation. We found that inhibition of HDAC11 significantly interrupted mouse oocytes meiosis progress, caused abnormal spindle organization and misaligned chromosomes, impaired kinetochore-microtubule attachment and spindle assembly checkpoint (SAC) function. Moreover, HDAC11 inhibition significantly increased the acetylation level of α-tubulin that is associated with microtubule stability, and increased acetylation level of H4K16 that is important for kinetochore function. In conclusion, our study indicates that HDAC11 is an essential factor for oocytes maturation and it promotes meiotic process most likely though decreasing acetylation status of α-tubulin and H4K16.


Subject(s)
Histone Deacetylases/metabolism , Histones/metabolism , Meiosis/genetics , Oocytes/metabolism , Oogenesis/genetics , Tubulin/metabolism , Acetylation , Animals , Chromosome Segregation/genetics , Female , Gene Expression Regulation/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Kinetochores/metabolism , Kinetochores/pathology , Lysine/metabolism , M Phase Cell Cycle Checkpoints/genetics , Metaphase , Mice , Microtubules/metabolism , Oocytes/enzymology , Oocytes/growth & development , Protein Processing, Post-Translational , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Spindle Apparatus/pathology
19.
Theriogenology ; 142: 62-66, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31574402

ABSTRACT

In this study, we evaluated DNase activity of rainbow trout oocyte using an in vitro and in vivo study. First, synthetic single strand and natural double strand DNA from Eukaryotic and prokaryotic sources as well as naked DNA were in vitro incubated with the oocyte cytoplasm. Results showed that the DNase activity of rainbow trout oocyte is strong enough to degrade any type of DNA at the onset of the incubation. Then, we evaluated if similar to the mammalian species, dead spermatozoa from rainbow trout can protect exogenous DNA from oocyte DNases. A series of dead spermatozoa was incubated with pDB2, carrying EGFP transgene, for 30 min followed by the ooplasm treatment for an additional 30 min. Not only did oocyte DNases completely degrade the exogenous DNA, but also it degraded the compact genome of spermatozoa. In conclusion, in vitro results clearly showed that strong DNase activity of ooplasm could degrade any types of foreign DNAs including oligonucleotides and intensively compact sperm genome. The strong DNase activity of rainbow trout ooplasm could be a stumbling block for genetic modification using plasmids in salmonids.


Subject(s)
DNA/metabolism , Deoxyribonucleases/metabolism , Oncorhynchus mykiss , Oocytes/enzymology , Animals , Male , Oocytes/metabolism , Plasmids , Spermatozoa , Transfection
20.
Annu Rev Biochem ; 89: 135-158, 2020 06 20.
Article in English | MEDLINE | ID: mdl-31815535

ABSTRACT

DNA methylation at the 5-position of cytosine (5mC) plays vital roles in mammalian development. DNA methylation is catalyzed by DNA methyltransferases (DNMTs), and the two DNMT families, DNMT3 and DNMT1, are responsible for methylation establishment and maintenance, respectively. Since their discovery, biochemical and structural studies have revealed the key mechanisms underlying how DNMTs catalyze de novo and maintenance DNA methylation. In particular, recent development of low-input genomic and epigenomic technologies has deepened our understanding of DNA methylation regulation in germ lines and early stage embryos. In this review, we first describe the methylation machinery including the DNMTs and their essential cofactors. We then discuss how DNMTs are recruited to or excluded from certain genomic elements. Lastly, we summarize recent understanding of the regulation of DNA methylation dynamics in mammalian germ lines and early embryos with a focus on both mice and humans.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA/genetics , Gene Expression Regulation, Developmental , Genome , Animals , Coenzymes/chemistry , Coenzymes/metabolism , CpG Islands , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Embryo, Mammalian , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Oocytes/cytology , Oocytes/enzymology , Oocytes/growth & development , Signal Transduction , Spermatozoa/cytology , Spermatozoa/enzymology , Spermatozoa/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...