Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 51(22): 12076-12091, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37950888

ABSTRACT

Translation is critical for development as transcription in the oocyte and early embryo is silenced. To illustrate the translational changes during meiosis and consecutive two mitoses of the oocyte and early embryo, we performed a genome-wide translatome analysis. Acquired data showed significant and uniform activation of key translational initiation and elongation axes specific to M-phases. Although global protein synthesis decreases in M-phases, translation initiation and elongation activity increases in a uniformly fluctuating manner, leading to qualitative changes in translation regulation via the mTOR1/4F/eEF2 axis. Overall, we have uncovered a highly dynamic and oscillatory pattern of translational reprogramming that contributes to the translational regulation of specific mRNAs with different modes of polysomal occupancy/translation that are important for oocyte and embryo developmental competence. Our results provide new insights into the regulation of gene expression during oocyte meiosis as well as the first two embryonic mitoses and show how temporal translation can be optimized. This study is the first step towards a comprehensive analysis of the molecular mechanisms that not only control translation during early development, but also regulate translation-related networks employed in the oocyte-to-embryo transition and embryonic genome activation.


Subject(s)
Embryonic Development , Oocytes , Protein Biosynthesis , Gene Expression Regulation, Developmental , Meiosis , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Mice
2.
Eur J Cell Biol ; 101(2): 151210, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35240557

ABSTRACT

In mammalian females, oocytes are stored in the ovary and meiosis is arrested at the diplotene stage of prophase I. When females reach puberty oocytes are selectively recruited in cycles to grow, overcome the meiotic arrest, complete the first meiotic division and become mature (ready for fertilization). At a molecular level, the master regulator of prophase I arrest and meiotic resumption is the maturation-promoting factor (MPF) complex, formed by the active form of cyclin dependent kinase 1 (CDK1) and Cyclin B1. However, we still do not have complete information regarding the factors implicated in MPF activation. In this study we document that out of three mammalian serum-glucocorticoid kinase proteins (SGK1, SGK2, SGK3), mouse oocytes express only SGK1 with a phosphorylated (active) form dominantly localized in the nucleoplasm. Further, suppression of SGK1 activity in oocytes results in decreased CDK1 activation via the phosphatase cell division cycle 25B (CDC25B), consequently delaying or inhibiting nuclear envelope breakdown. Expression of exogenous constitutively active CDK1 can rescue the phenotype induced by SGK1 inhibition. These findings bring new insights into the molecular pathways acting upstream of MPF and a better understanding of meiotic resumption control by presenting a new key player SGK1 in mammalian oocytes.


Subject(s)
Immediate-Early Proteins , Maturation-Promoting Factor , Animals , Cell Cycle Checkpoints , Female , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mammals/metabolism , Maturation-Promoting Factor/metabolism , Meiosis , Meiotic Prophase I , Mice , Oocytes/metabolism , Protein Serine-Threonine Kinases/genetics
3.
Aging Cell ; 19(10): e13231, 2020 10.
Article in English | MEDLINE | ID: mdl-32951297

ABSTRACT

Increasing maternal age in mammals is associated with poorer oocyte quality, involving higher aneuploidy rates and decreased developmental competence. Prior to resumption of meiosis, fully developed mammalian oocytes become transcriptionally silent until the onset of zygotic genome activation. Therefore, meiotic progression and early embryogenesis are driven largely by translational utilization of previously synthesized mRNAs. We report that genome-wide translatome profiling reveals considerable numbers of transcripts that are differentially translated in oocytes obtained from aged compared to young females. Additionally, we show that a number of aberrantly translated mRNAs in oocytes from aged females are associated with cell cycle. Indeed, we demonstrate that four specific maternal age-related transcripts (Sgk1, Castor1, Aire and Eg5) with differential translation rates encode factors that are associated with the newly forming meiotic spindle. Moreover, we report substantial defects in chromosome alignment and cytokinesis in the oocytes of young females, in which candidate CASTOR1 and SGK1 protein levels or activity are experimentally altered. Our findings indicate that improper translation of specific proteins at the onset of meiosis contributes to increased chromosome segregation problems associated with female ageing.


Subject(s)
Oocytes/metabolism , Age Factors , Animals , Female , Humans , Mammals
4.
Sci Rep ; 10(1): 7455, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366979

ABSTRACT

In several species, including Xenopus, mouse and human, two members of cyclin A family were identified. Cyclin A2, which is ubiquitously expressed in dividing cells and plays role in DNA replication, entry into mitosis and spindle assembly, and cyclin A1, whose function is less clear and which is expressed in spermatocytes, leukemia cells and in postmitotic multiciliated cells. Deletion of the gene showed that cyclin A1 is essential for male meiosis, but nonessential for female meiosis. Our results revealed, that the cyclin A1 is not only dispensable in oocytes, we show here that its expression is in fact undesirable in these cells. Our data demonstrate that the APC/C and proteasome in oocytes are unable to target sufficiently cyclin A1 before anaphase, which leads into anaphase arrest and direct inhibition of separase. The cyclin A1-induced cell cycle arrest is oocyte-specific and the presence of cyclin A1 in early embryos has no effect on cell cycle progression or chromosome division. Cyclin A1 is therefore not only an important cell cycle regulator with biased expression in germline, being essential for male and damaging for female meiosis, its persistent expression during anaphase in oocytes shows fundamental differences between APC/C function in oocytes and in early embryos.


Subject(s)
Anaphase , Chromosome Segregation , Cyclin A1/physiology , Oocytes/cytology , Animals , Cyclin A2/physiology , Female , Male , Meiosis , Metaphase , Mice , Microinjections , Microscopy, Fluorescence , Proteasome Endopeptidase Complex/physiology
5.
Int J Mol Sci ; 21(4)2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32070012

ABSTRACT

Meiotic maturation of oocyte relies on pre-synthesised maternal mRNA, the translation of which is highly coordinated in space and time. Here, we provide a detailed polysome profiling protocol that demonstrates a combination of the sucrose gradient ultracentrifugation in small SW55Ti tubes with the qRT-PCR-based quantification of 18S and 28S rRNAs in fractionated polysome profile. This newly optimised method, named Scarce Sample Polysome Profiling (SSP-profiling), is suitable for both scarce and conventional sample sizes and is compatible with downstream RNA-seq to identify polysome associated transcripts. Utilising SSP-profiling we have assayed the translatome of mouse oocytes at the onset of nuclear envelope breakdown (NEBD)-a developmental point, the study of which is important for furthering our understanding of the molecular mechanisms leading to oocyte aneuploidy. Our analyses identified 1847 transcripts with moderate to strong polysome occupancy, including abundantly represented mRNAs encoding mitochondrial and ribosomal proteins, proteasomal components, glycolytic and amino acids synthetic enzymes, proteins involved in cytoskeleton organization plus RNA-binding and translation initiation factors. In addition to transcripts encoding known players of meiotic progression, we also identified several mRNAs encoding proteins of unknown function. Polysome profiles generated using SSP-profiling were more than comparable to those developed using existing conventional approaches, being demonstrably superior in their resolution, reproducibility, versatility, speed of derivation and downstream protocol applicability.


Subject(s)
Nuclear Envelope/genetics , Oocytes/growth & development , Polyribosomes/genetics , RNA-Binding Proteins/genetics , Animals , Female , Gene Expression Regulation, Developmental/genetics , Meiosis/genetics , Mice , Nuclear Envelope/metabolism , Oocytes/metabolism , RNA, Messenger, Stored/genetics , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 28S/genetics , RNA-Seq
6.
Biol Reprod ; 101(4): 719-732, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31290535

ABSTRACT

Culture media used in assisted reproduction are commonly supplemented with gonadotropin hormones to support the nuclear and cytoplasmic maturation of in vitro matured oocytes. However, the effect of gonadotropins on protein synthesis in oocytes is yet to be fully understood. As published data have previously documented a positive in vitro effect of follicle-stimulating hormone (FSH) on cytoplasmic maturation, we exposed mouse denuded oocytes to FSH in order to evaluate the changes in global protein synthesis. We found that dose-dependent administration of FSH resulted in a decrease of methionine incorporation into de novo synthesized proteins in denuded mouse oocytes and oocytes cultured in cumulus-oocyte complexes. Similarly, FSH influenced methionine incorporation in additional mammalian species including human. Furthermore, we showed the expression of FSH-receptor protein in oocytes. We found that major translational regulators were not affected by FSH treatment; however, the amino acid uptake became impaired. We propose that the effect of FSH treatment on amino acid uptake is influenced by FSH receptor with the effect on oocyte metabolism and physiology.


Subject(s)
Amino Acids/metabolism , Follicle Stimulating Hormone/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Animals , Cattle , Cells, Cultured , Cleavage Stage, Ovum/drug effects , Cleavage Stage, Ovum/metabolism , Culture Media/chemistry , Culture Media/pharmacology , Female , Humans , In Vitro Oocyte Maturation Techniques/methods , Mammals , Mice , Swine
7.
Mol Cell Biol ; 39(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30988159

ABSTRACT

Cell division cycle (Cdc) kinase subunit (CKS) proteins bind cyclin-dependent kinases (CDKs) and play important roles in cell division control and development, though their precise molecular functions are not fully understood. Mammals express two closely related paralogs called CKS1 and CKS2, but only CKS2 is expressed in the germ line, indicating that it is solely responsible for regulating CDK functions in meiosis. Using cks2-/- knockout mice, we show that CKS2 is a crucial regulator of maturation-promoting factor (MPF; CDK1-cyclin A/B) activity in meiosis. cks2-/- oocytes display reduced and delayed MPF activity during meiotic progression, leading to defects in germinal vesicle breakdown (GVBD), anaphase-promoting complex/cyclosome (APC/C) activation, and meiotic spindle assembly. cks2-/- germ cells express significantly reduced levels of the MPF components CDK1 and cyclins A1/B1. Additionally, injection of MPF plus CKS2, but not MPF alone, restored normal GVBD in cks2-/- oocytes, demonstrating that GVBD is driven by a CKS2-dependent function of MPF. Moreover, we generated cks2cks1/cks1 knock-in mice and found that CKS1 can compensate for CKS2 in meiosis in vivo, but homozygous embryos arrested development at the 2- to 5-cell stage. Collectively, our results show that CKS2 is a crucial regulator of MPF functions in meiosis and that its paralog, CKS1, must be excluded from the germ line for proper embryonic development.


Subject(s)
CDC2-CDC28 Kinases/genetics , Cell Cycle Proteins/genetics , Embryonic Development , Oocytes/cytology , Animals , CDC2-CDC28 Kinases/metabolism , Cell Cycle Proteins/metabolism , Female , Gene Knock-In Techniques , Male , Maturation-Promoting Factor/metabolism , Meiosis , Mesothelin , Mice , Mice, Knockout , Oocytes/metabolism
8.
Int J Mol Sci ; 19(10)2018 Oct 12.
Article in English | MEDLINE | ID: mdl-30322039

ABSTRACT

Oocyte meiotic maturation and embryogenesis are some of the most important physiological processes that occur in organisms, playing crucial roles in the preservation of life in all species. The post-transcriptional regulation of maternal messenger ribonucleic acids (mRNAs) and the post-translational regulation of proteins are critical in the control of oocyte maturation and early embryogenesis. Translational control affects the basic mechanism of protein synthesis, thus, knowledge of the key components included in this machinery is required in order to understand its regulation. Cytoplasmic polyadenylation element binding proteins (CPEBs) bind to the 3'-end of mRNAs to regulate their localization and translation and are necessary for proper development. In this study we examined the expression pattern of cytoplasmic polyadenylation element binding protein 2 (CPEB2) both on the mRNA (by real-time quantitative reverse transcription polymerase chain reaction, qRT-PCR) and protein (by Western blotting, WB) level, as well as its localization during the meiotic maturation of porcine oocytes and early embryonic development by immunocytochemistry (ICC). For the elucidation of its functions, CPEB2 knockdown by double-strand RNA (dsRNA) was used. We discovered that CPEB2 is expressed during all stages of porcine meiotic maturation and embryonic development. Moreover, we found that it is necessary to enable a high percentage of oocytes to reach the metaphase II (MII) stage, as well as for the production of good-quality parthenogenetic blastocysts.


Subject(s)
Meiosis , Oocytes/cytology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Animals , Embryonic Development , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Oocytes/metabolism , Parthenogenesis , Pregnancy , RNA, Messenger/metabolism , Swine
9.
Int J Mol Sci ; 19(9)2018 Sep 19.
Article in English | MEDLINE | ID: mdl-30235877

ABSTRACT

The rate of chromosome segregation errors that emerge during meiosis I in the mammalian female germ line are known to increase with maternal age; however, little is known about the underlying molecular mechanism. The objective of this study was to analyze meiotic progression of mouse oocytes in relation to maternal age. Using the mouse as a model system, we analyzed the timing of nuclear envelope breakdown and the morphology of the nuclear lamina of oocytes obtained from young (2 months old) and aged females (12 months old). Oocytes obtained from older females display a significantly faster progression through meiosis I compared to the ones obtained from younger females. Furthermore, in oocytes from aged females, lamin A/C structures exhibit rapid phosphorylation and dissociation. Additionally, we also found an increased abundance of MPF components and increased translation of factors controlling translational activity in the oocytes of aged females. In conclusion, the elevated MPF activity observed in aged female oocytes affects precocious meiotic processes that can multifactorially contribute to chromosomal errors in meiosis I.


Subject(s)
Aging/metabolism , Maturation-Promoting Factor/metabolism , Meiosis , Oocytes/metabolism , Aging/genetics , Animals , Female , Maturation-Promoting Factor/genetics , Mesothelin , Mice , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Oocytes/cytology , Phosphorylation , Protein Processing, Post-Translational
10.
Int J Mol Sci ; 19(3)2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29494492

ABSTRACT

Although the involvement of the extracellular signal-regulated kinases 1 and 2 (ERK1/2) pathway in the regulation of cytostatic factor (CSF) activity; as well as in microtubules organization during meiotic maturation of oocytes; has already been described in detail; rather less attention has been paid to the role of ERK1/2 in the regulation of mRNA translation. However; important data on the role of ERK1/2 in translation during oocyte meiosis have been documented. This review focuses on recent findings regarding the regulation of translation and the role of ERK1/2 in this process in the meiotic cycle of mammalian oocytes. The specific role of ERK1/2 in the regulation of mammalian target of rapamycin (mTOR); eukaryotic translation initiation factor 4E (eIF4E) and cytoplasmic polyadenylation element binding protein 1 (CPEB1) activity is addressed along with additional focus on the other key players involved in protein translation.


Subject(s)
Meiosis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oocytes/metabolism , Protein Biosynthesis , Animals , Cytoplasm/genetics , Cytoplasm/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Developmental , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Polyadenylation , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism
11.
PLoS One ; 13(3): e0192544, 2018.
Article in English | MEDLINE | ID: mdl-29529035

ABSTRACT

The tight correlation between mRNA distribution and subsequent protein localization and function indicate a major role for mRNA localization within the cell. RNA localization, followed by local translation, presents a mechanism for spatial and temporal gene expression regulation utilized by various cell types. However, little is known about mRNA localization and translation in the mammalian oocyte and early embryo. Importantly, fully-grown oocyte becomes transcriptionally inactive and only utilizes transcripts previously synthesized and stored during earlier development. We discovered an abundant RNA population in the oocyte and early embryo nucleus together with RNA binding proteins. We also characterized specific ribosomal proteins, which contribute to translation in the oocyte and embryo. By applying selected markers to mouse and human oocytes, we found that there might be a similar mechanism of RNA metabolism in both species. In conclusion, we visualized the localization of RNAs and translation machinery in the oocyte, that could shed light on this terra incognita of these unique cell types in mouse and human.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Oocytes/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Animals , Cells, Cultured , Embryo, Mammalian/ultrastructure , Female , Humans , Mice , Oocytes/ultrastructure , RNA, Messenger/analysis , RNA-Binding Proteins/analysis , RNA-Binding Proteins/genetics , Transcriptome
12.
Results Probl Cell Differ ; 63: 257-295, 2017.
Article in English | MEDLINE | ID: mdl-28779322

ABSTRACT

Fully grown oocytes arrest meiosis at prophase I and deposit maternal RNAs. A subset of maternal transcripts is stored in a dormant state in the oocyte, and the timely driven translation of specific mRNAs guides meiotic progression, the oocyte-embryo transition, and early embryo development. In the absence of transcription, the regulation of gene expression in oocytes is controlled almost exclusively at the level of transcriptome and proteome stabilization and at the level of protein synthesis.This chapter focuses on the recent findings on RNA distribution related to the temporal and spatial translational control of the meiotic cycle progression in mammalian oocytes. We discuss the most relevant mechanisms involved in the organization of the oocyte's maternal transcriptome storage and localization, and the regulation of translation, in correlation with the regulation of oocyte meiotic progression.


Subject(s)
Gene Expression Regulation , Maternal Inheritance/genetics , Meiosis/genetics , Oocytes/metabolism , Protein Biosynthesis , RNA, Messenger, Stored/genetics , Animals , Female , Oocytes/cytology , RNA, Messenger, Stored/analysis , Transcriptome/genetics
13.
Cell Cycle ; 16(10): 927-939, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28272965

ABSTRACT

Fully grown mammalian oocytes utilize transcripts synthetized and stored during earlier development. RNA localization followed by a local translation is a mechanism responsible for the regulation of spatial and temporal gene expression. Here we show that the mouse oocyte contains 3 forms of cap-dependent translational repressor expressed on the mRNA level: 4E-BP1, 4E-BP2 and 4E-BP3. However, only 4E-BP1 is present as a protein in oocytes, it becomes inactivated by phosphorylation after nuclear envelope breakdown and as such it promotes cap-dependent translation after NEBD. Phosphorylation of 4E-BP1 can be seen in the oocytes after resumption of meiosis but it is not detected in the surrounding cumulus cells, indicating that 4E-BP1 promotes translation at a specific cell cycle stage. Our immunofluorescence analyses of 4E-BP1 in oocytes during meiosis I showed an even localization of global 4E-BP1, as well as of its 4E-BP1 (Thr37/46) phosphorylated form. On the other hand, 4E-BP1 phosphorylated on Ser65 is localized at the spindle poles, and 4E-BP1 phosphorylated on Thr70 localizes on the spindle. We further show that the main positive regulators of 4E-BP1 phosphorylation after NEBD are mTOR and CDK1 kinases, but not PLK1 kinase. CDK1 exerts its activity toward 4E-BP1 phosphorylation via phosphorylation and activation of mTOR. Moreover, both CDK1 and phosphorylated mTOR co-localize with 4E-BP1 phosphorylated on Thr70 on the spindle at the onset of meiotic resumption. Expression of the dominant negative 4E-BP1 mutant adversely affects translation and results in spindle abnormality. Taken together, our results show that the phosphorylation of 4E-BP1 promotes translation at the onset of meiosis to support the spindle assembly and suggest an important role of CDK1 and mTOR kinases in this process. We also show that the mTOR regulatory pathway is present in human oocytes and is likely to function in a similar way as in mouse oocytes.


Subject(s)
CDC2 Protein Kinase/genetics , Carrier Proteins/genetics , Oocytes/growth & development , Phosphoproteins/genetics , TOR Serine-Threonine Kinases/genetics , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle/genetics , Cell Cycle Proteins , Eukaryotic Initiation Factors , Gene Expression Regulation, Developmental , Humans , Mice , Oocytes/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Biosynthesis , Spindle Apparatus/genetics
14.
Cell Tissue Res ; 363(1): 69-84, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26340983

ABSTRACT

A hallmark of oocyte development in mammals is the dependence on the translation and utilization of stored RNA and proteins rather than the de novo transcription of genes in order to sustain meiotic progression and early embryo development. In the absence of transcription, the completion of meiosis and early embryo development in mammals relies significantly on maternally synthesized RNAs. Post-transcriptional control of gene expression at the translational level has emerged as an important cellular function in normal development. Therefore, the regulation of gene expression in oocytes is controlled almost exclusively at the level of mRNA and protein stabilization and protein synthesis. This current review is focused on the recently emerged findings on RNA distribution related to the temporal and spatial translational control of the meiotic progression of the mammalian oocyte.


Subject(s)
Oocytes/cytology , Oocytes/metabolism , Protein Biosynthesis , Animals , Gene Expression Regulation, Developmental , Humans , Meiosis , Oogenesis , RNA/analysis , RNA/genetics , Transcriptome
15.
Nat Commun ; 6: 6078, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25629602

ABSTRACT

The fully grown mammalian oocyte is transcriptionally quiescent and utilizes only transcripts synthesized and stored during early development. However, we find that an abundant RNA population is retained in the oocyte nucleus and contains specific mRNAs important for meiotic progression. Here we show that during the first meiotic division, shortly after nuclear envelope breakdown, translational hotspots develop in the chromosomal area and in a region that was previously surrounded the nucleus. These distinct translational hotspots are separated by endoplasmic reticulum and Lamin, and disappear following polar body extrusion. Chromosomal translational hotspots are controlled by the activity of the mTOR-eIF4F pathway. Here we reveal a mechanism that-following the resumption of meiosis-controls the temporal and spatial translation of a specific set of transcripts required for normal spindle assembly, chromosome alignment and segregation.


Subject(s)
Eukaryotic Initiation Factor-4F/metabolism , Mammals/metabolism , Oocytes/metabolism , Protein Biosynthesis , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Chromosomes, Mammalian/metabolism , Down-Regulation , Fertilization , Genomic Instability , Humans , Meiosis , Mice , Nuclear Envelope/metabolism , RNA Caps/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
16.
PLoS One ; 9(7): e101222, 2014.
Article in English | MEDLINE | ID: mdl-24983972

ABSTRACT

Regulation of mRNA translation by cytoplasmic polyadenylation is known to be important for oocyte maturation and further development. This process is generally controlled by phosphorylation of cytoplasmic polyadenylation element binding protein 1 (CPEB1). The aim of this study is to determine the role of Aurora kinase A in CPEB1 phosphorylation and the consequent CPEB1-dependent polyadenylation of maternal mRNAs during mammalian oocyte meiosis. For this purpose, we specifically inhibited Aurora kinase A with MLN8237 during meiotic maturation of porcine oocytes. Using poly(A)-test PCR method, we monitored the effect of Aurora kinase A inhibition on poly(A)-tail extension of long and short cyclin B1 encoding mRNAs as markers of CPEB1-dependent cytoplasmic polyadenylation. Our results show that inhibition of Aurora kinase A activity impairs neither cyclin B1 mRNA polyadenylation nor its translation and that Aurora kinase A is unlikely to be involved in CPEB1 activating phosphorylation.


Subject(s)
Aurora Kinase A/metabolism , Cyclin B1/genetics , Meiosis , Oocytes/metabolism , RNA, Messenger/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Female , Oocytes/enzymology , Phosphorylation , Polyadenylation , Sus scrofa/metabolism , mRNA Cleavage and Polyadenylation Factors/chemistry
17.
Cell Cycle ; 11(16): 3011-8, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22871737

ABSTRACT

Chromosome segregation errors are highly frequent in mammalian female meiosis, and their incidence gradually increases with maternal age. The fate of aneuploid eggs is obviously dependent on the stringency of mechanisms for detecting unattached or repairing incorrectly attached kinetochores. In case of their failure, the newly formed embryo will inherit the impaired set of chromosomes, which will have severe consequences for its further development. Whether spindle assembly checkpoint (SAC) in oocytes is capable of arresting cell cycle progression in response to unaligned kinetochores was discussed for a long time. It is known that abolishing SAC increases frequency of chromosome segregation errors and causes precocious entry into anaphase; SAC, therefore, seems to be essential for normal chromosome segregation in meiosis I. However, it was also reported that for anaphase-promoting complex (APC) activation, which is a prerequisite for entering anaphase; alignment of only a critical mass of kinetochores on equatorial plane is sufficient. This indicates that the function of SAC and of cooperating chromosome attachment correction mechanisms in oocytes is different from somatic cells. To analyze this phenomenon, we used live cell confocal microscopy to monitor chromosome movements, spindle formation, APC activation and polar body extrusion (PBE) simultaneously in individual oocytes at various time points during first meiotic division. Our results, using oocytes from aged animals and interspecific crosses, demonstrate that multiple unaligned kinetochores and severe congression defects are tolerated at the metaphase to anaphase transition, although such cells retain sensitivity to nocodazole. This indicates that checkpoint mechanisms, operating in oocytes at this point, are essential for accurate timing of APC activation in meiosis I, but they are insufficient in detection or correction of unaligned chromosomes, preparing thus conditions for propagation of the aneuploidy to the embryo.


Subject(s)
Chromosome Pairing , Chromosome Segregation , Chromosomes, Mammalian/metabolism , Oocytes/cytology , Anaphase , Anaphase-Promoting Complex-Cyclosome , Aneuploidy , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromosomes, Mammalian/genetics , Female , Histones/genetics , Histones/metabolism , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Male , Mammals , Metaphase , Mice , Mice, Inbred C57BL , Microinjections , Microscopy, Confocal/methods , Oocytes/metabolism , Proteolysis , Securin , Time-Lapse Imaging/methods , Tubulin/genetics , Tubulin/metabolism , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligase Complexes/metabolism
18.
Cell Tissue Res ; 346(1): 1-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21969023

ABSTRACT

Gametogenesis and fertilization are the key events in sexual reproduction. In the female, meiosis results in a large oocyte that is competent for fertilization and fundamental for the success of early embryonic development. Progression through meiosis is monitored by fine regulatory mechanisms. In this review, we focus on one of the most well-known regulatory elements, the E3 ligase APC/C, which mediates proteolytic degradation of a number of important substrates via the ubiquitin proteasome pathway (UPP). The UPP also indirectly regulates protein synthesis by affecting proteins involved in RNA metabolism, a process that is paramount for the transcriptionally silent oocyte. During the past few years, more evidence has accumulated to suggest that the UPP has an important role in zona pellucida penetration and gamete fusion in mammals. This review focuses on the function of the UPP in regulating oocyte meiotic maturation in mammals, with special attention to its role in chromosome segregation and polar body extrusion, its role in the acquisition of meiotic/developmental competence and recent advances in our understanding of the UPP role in fertilization.


Subject(s)
Meiosis/physiology , Oocytes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Sperm-Ovum Interactions/physiology , Ubiquitinated Proteins/metabolism , Animals , Female , Humans , Male , Mammals , Oocytes/cytology , RNA , Ubiquitin-Protein Ligases/metabolism , Zona Pellucida/metabolism
19.
PLoS One ; 6(4): e18892, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21556143

ABSTRACT

It is generally accepted that mammalian oocytes are frequently suffering from chromosome segregation errors during meiosis I, which have severe consequences, including pregnancy loss, developmental disorders and mental retardation. In a search for physiologically more relevant model than rodent oocytes to study this phenomenon, we have employed comparative genomic hybridization (CGH), combined with whole genome amplification (WGA), to study the frequency of aneuploidy in porcine oocytes, including rare cells obtained from aged animals. Using this method, we were able to analyze segregation pattern of each individual chromosome during meiosis I. In contrast to the previous reports where conventional methods, such as chromosome spreads or FISH, were used to estimate frequency of aneuploidy, our results presented here show, that the frequency of this phenomenon was overestimated in porcine oocytes. Surprisingly, despite the results from human and mouse showing an increase in the frequency of aneuploidy with advanced maternal age, our results obtained by the most accurate method currently available for scoring the aneuploidy in oocytes indicated no increase in the frequency of aneuploidy even in oocytes from animals, whose age was close to the life expectancy of the breed.


Subject(s)
Aging/genetics , Aneuploidy , Oocytes/cytology , Animals , Comparative Genomic Hybridization , Swine
20.
Reprod Fertil Dev ; 22(4): 644-52, 2010.
Article in English | MEDLINE | ID: mdl-20353724

ABSTRACT

The multiprotein complexes known as condensins (I and II) are major players in chromosome dynamics in mitotic and meiotic cells. Here, we report for the first time the detection of different condensin subunits from both complexes in mammalian oocytes. Using immunoblotting analysis we examined expression levels of condensin subunits during meiotic maturation of porcine oocytes. The expression of the core subunit structural maintenance of chromosomes 2 (SMC2), identical in both condensin complexes, did not change significantly during maturation. Similarly, there was no significant change in the expression of the chromosome associated protein (CAP)-H and CAP-H2 subunits, components of condensin I and II, respectively. Conversely, the expression profiles of CAP-G, CAP-D2 (condensin I) and CAP-D3 (condensin II) were more interesting. At least two isoforms of the CAP-D2 subunit were detected, along with three isoforms of the CAP-D3 and CAP-G subunits. We suggest that this diverse migration of subunit isoforms is due to post-translational modification. Earlier, it was reported that non-SMC proteins are phosphorylated by cyclin-dependent kinase 1. In the present study, we analysed the phosphorylation status of the three subunits in oocyte extracts using alkaline phosphatase treatment and we found that at least the fastest migrating form of CAP-D3 was likely to be phosphorylated in maturing porcine oocytes. In addition, the localisation of CAP-H and CAP-H2 subunits was examined using immunofluorescence staining with specific antibodies, as well as following microinjection of their enhanced green fluorescent protein-tagged mRNA into germinal vesicle-stage oocytes. CAP-H was found in the cytoplasm, whereas CAP-H2 was localised within the nucleus.


Subject(s)
Adenosine Triphosphatases/physiology , DNA-Binding Proteins/physiology , Meiosis/physiology , Multiprotein Complexes/physiology , Oocytes/physiology , Swine/physiology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/ultrastructure , Animals , Chromatin/physiology , Chromosomes/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Female , Immunoblotting/veterinary , Microscopy, Confocal/veterinary , Microscopy, Fluorescence/veterinary , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Processing, Post-Translational , Protein Subunits
SELECTION OF CITATIONS
SEARCH DETAIL
...