Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 450(3): 1175-81, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24721426

ABSTRACT

Like most metazoans, eggs of echinoderms and tunicates (marine deuterostomes, there is no data for the cephalochordates) arrest awaiting fertilization due to the activity of the Mos/MEK/MAPK cascade and are released from this cell cycle arrest by sperm-triggered Ca2+ signals. Invertebrate deuterostome eggs display mainly three distinct types of cell cycle arrest before fertilization mediated by potentially different cytostatic factors (CSF): one CSF causes arrest during meiotic metaphase I (MI-CSF in tunicates and some starfishes), another CSF likely causes arrest during meiotic metaphase II (amphioxus), and yet another form of CSF causes arrest to occur after meiotic exit during G1 of the first mitotic cycle (G1-CSF). In tunicates and echinoderms these different CSF activities have been shown to rely on the Mos//MAPK pathway for establishment and on Ca2+ signals for their inactivation. Despite these molecular similarities, release of MI-CSF arrest is caused by APC/C activation (to destroy cyclin B) whereas release from G1-CSF is caused by stimulating S phase and the synthesis of cyclins. Further research is needed to understand how both the Mos//MAPK cascade and Ca2+ achieve these tasks in different marine invertebrate deuterostomes. Another conserved feature of eggs is that protein synthesis of specific mRNAs is necessary to proceed through oocyte maturation and to maintain CSF-induced cell cycle arrest. Then activation of development at fertilization is accompanied by an increase in the rate of protein synthesis but the mechanisms involved are still largely unknown in most of the marine deuterostomes. How the sperm-triggered Ca2+ signals cause an increase in protein synthesis has been studied mainly in sea urchin eggs. Here we review these conserved features of eggs (arrest, activation and protein synthesis) focusing on the non-vertebrate deuterostomes.


Subject(s)
Cell Cycle Checkpoints/physiology , Echinodermata/cytology , Echinodermata/growth & development , Urochordata/cytology , Urochordata/growth & development , Animals , Calcium Signaling/physiology , Echinodermata/physiology , Female , Fertilization/physiology , MAP Kinase Signaling System/physiology , Male , Oocytes/cytology , Oocytes/growth & development , Oocytes/physiology , Protein Biosynthesis/physiology , Proto-Oncogene Proteins c-mos/physiology , Urochordata/physiology , Zygote/cytology , Zygote/growth & development , Zygote/physiology
2.
Biol Reprod ; 86(4): 114, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22302686

ABSTRACT

Precise coordination of meiotic progression is a critical determinant of an egg's capacity to be fertilized successfully, and zinc has emerged as a key regulatory element in this process. An early manifestation of a regulatory role for this transition metal is the significant increase in total intracellular zinc. This accumulation is essential for meiotic progression beyond telophase I and the establishment of meiotic arrest at metaphase II. The subsequent developmental event, fertilization, induces a rapid expulsion of labile zinc that is a hallmark event in meiotic resumption. In the present study, we show that the zinc fluxes work, in part, by altering the activity of the cytostatic factor (CSF), the cellular activity required for the establishment and maintenance of metaphase II arrest in the mature, unfertilized egg. We propose a model in which zinc exerts concentration-dependent regulation of meiosis through the CSF component EMI2, a zinc-binding protein. Together, the data support the conclusion that zinc itself, through its interaction with EMI2, is a central component of the CSF.


Subject(s)
Cell Cycle Checkpoints/physiology , F-Box Proteins/physiology , Meiosis/physiology , Oocytes/cytology , Proto-Oncogene Proteins c-mos/physiology , Zinc/physiology , Animals , Cell Cycle Checkpoints/drug effects , Chelating Agents/pharmacology , Ethylenediamines/pharmacology , F-Box Proteins/drug effects , Female , Meiosis/drug effects , Mice , Oocytes/chemistry , Oocytes/drug effects , Proto-Oncogene Proteins c-mos/drug effects , Zinc/deficiency
3.
Development ; 138(5): 885-95, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21303846

ABSTRACT

Mos kinase is a universal mediator of oocyte meiotic maturation and is produced during oogenesis and destroyed after fertilization. The hallmark of maternal meiosis is that two successive M phases (meiosis I and II) drive two rounds of asymmetric cell division (ACD). However, how the egg limits the number of meioses to just two, thereby preventing gross aneuploidy, is poorly characterized. Here, in urochordate eggs, we show that loss of Mos/MAPK activity is necessary to prevent entry into meiosis III. Remarkably, maintaining the Mos/MAPK pathway active after fertilization at near physiological levels induces additional rounds of meiotic M phase (meiosis III, IV and V). During these additional rounds of meiosis, the spindle is positioned asymmetrically resulting in further rounds of ACD. In addition, inhibiting meiotic exit with Mos prevents pronuclear formation, cyclin A accumulation and maintains sperm-triggered Ca(2+) oscillations, all of which are hallmarks of the meiotic cell cycle in ascidians. It will be interesting to determine whether Mos availability in mammals can also control the number of meioses as it does in the urochordates. Our results demonstrate the power of urochordate eggs as a model to dissect the egg-to-embryo transition.


Subject(s)
Meiosis , Ovum/cytology , Proto-Oncogene Proteins c-mos/physiology , Urochordata/cytology , Animals , Cell Division , Ciona intestinalis , Embryo, Nonmammalian , MAP Kinase Signaling System , Urochordata/embryology , Zygote
4.
Biol Reprod ; 84(3): 526-36, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21076080

ABSTRACT

Zinc is essential for many biological processes, including proper functioning of gametes. We recently reported that zinc levels rise by over 50% during oocyte maturation and that attenuation of zinc availability during this period could be achieved using the membrane-permeable heavy metal chelator N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN). This zinc insufficiency resulted in formation of large polar bodies, failure to establish metaphase II arrest, and impaired establishment of cortical polarity. As these phenotypes resemble those of MOS null oocytes, we examined the impact of zinc insufficiency on the MOS-MAPK pathway. Reduced levels of both MOS protein and phosphorylation of MAP2K1/2 are observed in zinc-insufficient oocytes; however, these differences appear only after completion of the first meiotic division. In addition, activation of the downstream effector of the MOS pathway, MAPK3/1, is not affected by zinc insufficiency, and reduced MOS levels are observed only with the presence of TPEN after the first polar body extrusion. These data are inconsistent with the hypothesis that reduced MOS mediates the observed phenotype. Finally, MOS overexpression does not rescue the phenotype of zinc-insufficient oocytes, confirming that the observed disruption of asymmetric division and spindle abnormalities cannot be attributed to impaired MOS signaling. Zinc-insufficient oocytes do not increase maturation promoting factor (MPF) activity following the first meiotic division, and increasing MPF activity through expression of nondegradable cyclin B1 partially rescues the ability of zinc-insufficient oocytes to enter metaphase II. Although we have shown that zinc has a novel role in the meiotic cell cycle, it is not mediated through the MOS-MAPK pathway.


Subject(s)
Cell Division , MAP Kinase Signaling System/physiology , Meiosis , Oocytes/cytology , Proto-Oncogene Proteins c-mos/physiology , Zinc/physiology , Actin Capping Proteins/metabolism , Animals , Cell Division/drug effects , Cell Division/genetics , Cell Division/physiology , Chromatin/physiology , Female , MAP Kinase Signaling System/drug effects , Meiosis/drug effects , Meiosis/genetics , Meiosis/physiology , Mesothelin , Mice , Models, Biological , Oocytes/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Oogenesis/physiology , Phosphorylation , Proto-Oncogene Proteins c-mos/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Zinc/deficiency , Zinc/metabolism , Zinc/pharmacology
5.
Mol Biol Cell ; 19(8): 3536-43, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550795

ABSTRACT

The transition of oocytes from meiosis I (MI) to meiosis II (MII) requires partial cyclin B degradation to allow MI exit without S phase entry. Rapid reaccumulation of cyclin B allows direct progression into MII, producing a cytostatic factor (CSF)-arrested egg. It has been reported that dampened translation of the anaphase-promoting complex (APC) inhibitor Emi2 at MI allows partial APC activation and MI exit. We have detected active Emi2 translation at MI and show that Emi2 levels in MI are mainly controlled by regulated degradation. Emi2 degradation in MI depends not on Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), but on Cdc2-mediated phosphorylation of multiple sites within Emi2. As in MII, this phosphorylation is antagonized by Mos-mediated recruitment of PP2A to Emi2. Higher Cdc2 kinase activity in MI than MII allows sufficient Emi2 phosphorylation to destabilize Emi2 in MI. At MI anaphase, APC-mediated degradation of cyclin B decreases Cdc2 activity, enabling Cdc2-mediated Emi2 phosphorylation to be successfully antagonized by Mos-mediated PP2A recruitment. These data suggest a model of APC autoinhibition mediated by stabilization of Emi2; Emi2 proteins accumulate at MI exit and inhibit APC activity sufficiently to prevent complete degradation of cyclin B, allowing MI exit while preventing interphase before MII entry.


Subject(s)
Cyclin B/physiology , F-Box Proteins/physiology , Gene Expression Regulation , Meiosis , Proto-Oncogene Proteins c-mos/physiology , Animals , CDC2 Protein Kinase , Cell Movement , Cyclin B/metabolism , Cyclin-Dependent Kinases , Endocytosis , HL-60 Cells , Humans , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Neutrophils/metabolism , Proto-Oncogene Proteins c-mos/metabolism
7.
Cell Cycle ; 6(6): 732-8, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17361104

ABSTRACT

Vertebrate eggs arrest at metaphase of meiosis II due to an activity known as cytostatic factor (CSF). CSF antagonizes the ubiquitin ligase activity of the anaphase-promoting complex/cyclosome (APC/C), preventing cyclin B destruction and meiotic exit until fertilization occurs. A puzzling feature of CSF arrest is that APC/C inhibition is leaky. Ongoing cyclin B synthesis is counterbalanced by a limited amount of APC/C-mediated cyclin B destruction; thus, cyclin B/Cdc2 activity remains at steady state. How the APC/C can be slightly active toward cyclin B, and yet restrained from ubiquitinating cyclin B altogether, is unknown. Emi2/XErp1 is the critical CSF component directly responsible for APC/C inhibition during CSF arrest. Fertilization triggers the Ca2+-dependent destruction of Emi2, releasing the APC/C to ubiquitinate the full pool of cyclin B and initiate completion of meiosis. Previously, we showed that a phosphatase maintains Emi2's APC/C-inhibitory activity in CSF-arrested Xenopus egg extracts. Here, we demonstrate that phosphatase inhibition permits Emi2 phosphorylation at thr-545 and -551, which inactivates Emi2. Furthermore, we provide evidence that adding excess cyclin B to CSF extracts stimulates Cdc2 phosphorylation of these same residues, antagonizing Emi2-APC/C association. Our findings suggest a model wherein the pool of Emi2 acts analogously to a rheostat by integrating Cdc2 and phosphatase activities to prevent cyclin B overaccumulation and Cdc2 hyperactivity during the indefinite period of time between arrival at metaphase II and eventual fertilization. Finally, we propose that inactivation of Emi2 by Cdc2 permits mitotic progression during early embryonic cleavage cycles.


Subject(s)
F-Box Proteins/physiology , Maturation-Promoting Factor/physiology , Proto-Oncogene Proteins c-mos/physiology , Xenopus Proteins/physiology , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Animals , Cell Division/physiology , F-Box Proteins/antagonists & inhibitors , F-Box Proteins/metabolism , Female , Maturation-Promoting Factor/metabolism , Mesothelin , Mice , Molecular Sequence Data , Oocytes , Proto-Oncogene Proteins c-mos/metabolism , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligase Complexes/physiology , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/metabolism
8.
Cell Cycle ; 6(6): 725-31, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17361107

ABSTRACT

Cytostatic factor (CSF) arrests unfertilized vertebrate eggs in metaphase of meiosis II by inhibiting the anaphase-promoting complex/cyclosome (APC/C) from mediating cyclin destruction. The APC/C inhibitor Emi2/XErp1 satisfies a number of historical criteria for the molecular identification of CSF, but the mechanism by which CSF is activated selectively in meiosis II is the remaining unexplained criterion. Here we provide an explanation by showing that Emi2 is expressed specifically in meiosis II through translational de-repression or "unmasking" of its mRNA. We find that Emi2 protein is undetectable in immature, G2/prophase-arrested Xenopus oocytes and accumulates approximately 90 minutes after germinal vesicle breakdown. The 3' untranslated region of Emi2 mRNA contains cytoplasmic polyadenylation elements that directly bind the CPEB protein and confer temporal regulation of Emi2 polyadenylation and translation. Our results demonstrate that cytoplasmic polyadenylation and translational unmasking of Emi2 directs meiosis II-specific CSF arrest.


Subject(s)
F-Box Proteins/genetics , F-Box Proteins/metabolism , Meiosis/genetics , Protein Biosynthesis/physiology , Proto-Oncogene Proteins c-mos/antagonists & inhibitors , Proto-Oncogene Proteins c-mos/physiology , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/physiology , Animals , F-Box Proteins/physiology , Female , Oocytes/cytology , Oocytes/metabolism , Rabbits , Xenopus , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
9.
Oncogene ; 26(9): 1286-9, 2007 Feb 26.
Article in English | MEDLINE | ID: mdl-17322913

ABSTRACT

Since the discovery of cytostatic factor (CSF) 35 years ago, significant progress has been made in identifying molecular components of CSF activity and the mechanism of CSF-induced metaphase II arrest (CSF arrest). This short review focuses on recent discoveries in the field and discusses the implication of these results for a general picture of CSF establishment and release. One recent focus is on the cyclin E/Cdk2 pathway. The discovery of a downstream target for cyclin E/Cdk2, the spindle checkpoint protein Mps1, provides insight into how cyclin E/Cdk2 contributes to CSF arrest. The anaphase promoting complex/cyclosome (APC/C) inhibitor Emi2 is another recent focus of work in the field. It is now clear that not only is degradation of Emi2 critical for CSF release, but its abrupt accumulation during meiosis II (M II) is also required for the establishment of CSF arrest. Thus, by discrete pathways of APC/C inhibition operative during CSF arrest, the stability of cell cycle arrest in the egg appears to be reinforced by multiple mechanisms.


Subject(s)
Metaphase/physiology , Proto-Oncogene Proteins c-mos/physiology , Calcium/physiology , Humans , Proto-Oncogene Proteins c-mos/antagonists & inhibitors
10.
Fertil Steril ; 87(3): 677-90, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17126334

ABSTRACT

OBJECTIVE: To identify genes that are expressed differently during final oocyte maturation and early embryonic development in humans. DESIGN: Comparison of gene expression profiles of human germinal vesicle oocytes (hGVO), human embryonic stem cells (hESC) and human foreskin fibroblasts. SETTING: Research centers and a fertility unit in a university hospital. PATIENT(S): Fifty-five healthy women donated 76 hGVO. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Gene expression profiles were analyzed and compared with the use of microarray and reverse-transcription polymerase chain reaction. RESULT(S): Altogether, 10,183 genes were expressed in hGVO, and 45% of these genes were unclassified by biologic function. Four oocyte-specific genes (MATER, ZAR1, NPM2 and FIGLA) were detected in hGVO for the first time. Known components of 4 signaling pathways (MOS-MPF, transforming growth factor-beta, WNT, and NOTCH) were also found expressed in hGVO, with some components detected in hGVO for the first time. Distinct sets of genes that were revealed by comparison of expression profiles between hGVO, hESC, and human foreskin fibroblasts appear to be involved in oocyte maturation and early embryonic development. CONCLUSION(S): We obtained, for the first time, a large amount of information on gene expression of hGVO as compared with hESC. These data, from a unique research material-human oocytes, can now be used to understand the molecular mechanisms of early human development.


Subject(s)
Embryonic Stem Cells/physiology , Oocytes/physiology , Adult , Female , Foreskin/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genomic Imprinting , Humans , Male , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-mos/physiology , Receptors, Notch/physiology , Reproducibility of Results , Signal Transduction , Transforming Growth Factor beta/physiology , Wnt Proteins/physiology
11.
Curr Biol ; 16(19): 1968-73, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-17027495

ABSTRACT

Cytostatic factor (CSF) arrests vertebrate eggs in metaphase of meiosis II through several pathways that inhibit activation of the anaphase-promoting complex/cyclosome (APC/C). In Xenopus, the Mos-MEK1-MAPK-p90(Rsk) cascade utilizes spindle-assembly-checkpoint components to effect metaphase arrest. Another pathway involves cyclin E-Cdk2, and sustained cyclin E-Cdk2 activity in egg extracts causes metaphase arrest in the absence of Mos; this latter finding suggests that an independent pathway contributes to CSF arrest. Here, we demonstrate that metaphase arrest with cyclin E-Cdk2, but not with Mos, requires the spindle-checkpoint kinase monopolar spindles 1 (Mps1), a cyclin E-Cdk2 target that is also implicated in centrosome duplication. xMps1 is synthesized and activated during oocyte maturation and inactivated upon CSF release. In egg extracts, CSF release by calcium was inhibited by constitutively active cyclin E-Cdk2 and delayed by wild-type xMps1. Ablation of cyclin E by antisense oligonucleotides blocked accumulation of xMps1, suggesting that cyclin E-Cdk2 controls Mps1 levels. During meiosis II, activated cyclin E-Cdk2 significantly inhibited the APC/C even in the absence of the Mos-MAPK pathway, but this inhibition was not sufficient to suppress S phase between meiosis I and II. These results uniquely place xMps1 downstream of cyclin E-Cdk2 in mediating a pathway of APC/C inhibition and metaphase arrest.


Subject(s)
Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Metaphase/physiology , Protein Serine-Threonine Kinases/physiology , Xenopus Proteins/physiology , Animals , Cyclin E/antagonists & inhibitors , Cyclin E/genetics , MAP Kinase Signaling System , Oligonucleotides, Antisense , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-mos/metabolism , Proto-Oncogene Proteins c-mos/physiology , Xenopus , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
12.
Cell Cycle ; 5(11): 1152-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16760654

ABSTRACT

The development of an immature oocyte into a fertilizable gamete is a process known as meiotic maturation. In vertebrates, it corresponds to the transition from the prophase arrest of the first meiotic division (usually considered as a late G(2) phase) to the metaphase arrest of the second meiotic division. This transition is controlled by modulating the activity of the cyclin B-Cdc2 complex, MPF (M-phase promoting factor), the universal regulator of the G(2)/M transition. Meiotic maturation of frog oocytes is triggered by steroid hormones through a rapid, necessary and sufficient suppression of PKA and requires ongoing protein synthesis. A long-standing question has been to identify key protein(s) required to trigger the activation of MPF in response to the hormonal signal. Here we will discuss data supporting the view that steroids bring about meiotic maturation through functionally redundant pathways involving synthesis of Mos or of cyclin proteins, reinforcing the robustness of the system.


Subject(s)
Cyclins/physiology , Meiosis , Oocytes/physiology , Proto-Oncogene Proteins c-mos/physiology , Amphibian Proteins , Amphibians , Animals , Germ Cells/cytology , Signal Transduction , Steroids/pharmacology
13.
BMC Cancer ; 6: 6, 2006 Jan 09.
Article in English | MEDLINE | ID: mdl-16401344

ABSTRACT

BACKGROUND: We have previously reported that p53 mutated radioresistant lymphoma cell lines undergo mitotic catastrophe after irradiation, resulting in metaphase arrest and the generation of endopolyploid cells. A proportion of these endopolyploid cells then undergo a process of de-polyploidisation, stages of which are partially reminiscent of meiotic prophase. Furthermore, expression of meiosis-specific proteins of the cancer/testis antigens group of genes has previously been reported in tumours. We therefore investigated whether expression of meiosis-specific genes was associated with the polyploidy response in our tumour model. METHODS: Three lymphoma cell lines, Namalwa, WI-L2-NS and TK6, of varying p53 status were exposed to a single 10 Gy dose of gamma radiation and their responses assessed over an extended time course. DNA flow cytometry and mitotic counts were used to assess the kinetics and extent of polyploidisation and mitotic progression. Expression of meiotic genes was analysed using RT-PCR and western blotting. In addition, localisation of the meiotic cohesin REC8 and its relation to centromeres was analysed by immunofluorescence. RESULTS: The principal meiotic regulator MOS was found to be significantly post-transcriptionally up-regulated after irradiation in p53 mutated but not p53 wild-type lymphoma cells. The maximum expression of MOS coincided with the maximal fraction of metaphase arrested cells and was directly proportional to both the extent of the arrest and the number of endopolyploid cells that subsequently emerged. The meiotic cohesin REC8 was also found to be up-regulated after irradiation, linking sister chromatid centromeres in the metaphase-arrested and subsequent giant cells. Finally, RT-PCR revealed expression of the meiosis-prophase genes, DMC1, STAG3, SYCP3 and SYCP1. CONCLUSION: We conclude that multiple meiotic genes are aberrantly activated during mitotic catastrophe in p53 mutated lymphoma cells after irradiation. Furthermore, we suggest that the coordinated expression of MOS and REC8 regulate the extent of arrested mitoses and polyploidy.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma/genetics , Meiosis/genetics , Meiosis/radiation effects , Blotting, Western , Cell Cycle , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/physiology , DNA Damage , Flow Cytometry , Gene Expression Profiling , Genes, p53 , Humans , Lymphoma/pathology , Mitosis/genetics , Mitosis/radiation effects , Polyploidy , Proto-Oncogene Proteins c-mos/biosynthesis , Proto-Oncogene Proteins c-mos/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Up-Regulation
14.
Reproduction ; 130(6): 801-11, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322540

ABSTRACT

During meiotic maturation of mammalian oocytes, two successive divisions occur without an intermediate phase of DNA replication, so that haploid gametes are produced. Moreover, these two divisions are asymmetric, to ensure that most of the maternal stores are retained within the oocyte. This leads to the formation of daughter cells with different sizes: the large oocyte and the small polar bodies. All these events are dependent upon the dynamic changes in the organization of the oocyte cytoskeleton (microtubules and microfilaments) and are highly regulated in time and space. We review here the current knowledge of the interplay between the cytoskeleton and the cell cycle machinery in mouse oocytes, with an emphasis on the two major activities that control meiotic maturation in vertebrates, MPF (Maturation promoting factor) and CSF (Cytostatic factor).


Subject(s)
Cell Cycle/physiology , Cytoskeleton/ultrastructure , Meiosis/physiology , Oocytes/cytology , Oogenesis/physiology , Animals , Chromosomes/physiology , Cyclin B/metabolism , Female , Maturation-Promoting Factor/physiology , Mesothelin , Mice , Oocytes/ultrastructure , Proto-Oncogene Proteins c-mos/physiology , Spindle Apparatus/ultrastructure
16.
J Cell Biol ; 167(6): 1037-50, 2004 Dec 20.
Article in English | MEDLINE | ID: mdl-15611331

ABSTRACT

In Xenopus oocytes, the spindle assembly checkpoint (SAC) kinase Bub1 is required for cytostatic factor (CSF)-induced metaphase arrest in meiosis II. To investigate whether matured mouse oocytes are kept in metaphase by a SAC-mediated inhibition of the anaphase-promoting complex/cyclosome (APC/C) complex, we injected a dominant-negative Bub1 mutant (Bub1dn) into mouse oocytes undergoing meiosis in vitro. Passage through meiosis I was accelerated, but even though the SAC was disrupted, injected oocytes still arrested at metaphase II. Bub1dn-injected oocytes released from CSF and treated with nocodazole to disrupt the second meiotic spindle proceeded into interphase, whereas noninjected control oocytes remained arrested at metaphase. Similar results were obtained using dominant-negative forms of Mad2 and BubR1, as well as checkpoint resistant dominant APC/C activating forms of Cdc20. Thus, SAC proteins are required for checkpoint functions in meiosis I and II, but, in contrast to frog eggs, the SAC is not required for establishing or maintaining the CSF arrest in mouse oocytes.


Subject(s)
Oocytes/cytology , Oocytes/metabolism , Proto-Oncogene Proteins c-mos/physiology , Spindle Apparatus/physiology , Anaphase-Promoting Complex-Cyclosome , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Female , Mad2 Proteins , Meiosis , Mice , Mice, Inbred Strains , Mutation , Nocodazole/pharmacology , Nuclear Proteins , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-mos/metabolism , Spindle Apparatus/chemistry , Spindle Apparatus/genetics , Strontium/pharmacology , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/physiology
17.
EMBO J ; 23(23): 4649-59, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15526037

ABSTRACT

Vertebrate eggs arrest at metaphase of the second meiotic division before fertilization under the effect of a cytostatic factor (CSF). This arrest is established during oocyte maturation by the MAPK kinase module, comprised of Mos, MEK, MAPKs and p90Rsk. Maintenance of CSF arrest at metaphase requires inhibitors of the anaphase-promoting complex (APC) like Emi1, which sequesters the APC activator Cdc20. Although it was proposed that the Mos pathway and Emi1 act independently, neither one alone is sufficient to entirely reproduce CSF arrest. Herein we demonstrate that p90Rsk2 associates with and phosphorylates Emi1 upstream of the binding region for Cdc20, thus stabilizing their interaction. Experiments in transfected cells and two-cell embryos indicate that Emi1 and p90Rsk2 cooperate to induce the metaphase arrest. Moreover, oocyte maturation was impaired by interfering with the interaction between p90Rsk2 and Emi1 or by RNA interference of Emi1. Our results indicate that p90Rsk2 and Emi1 functionally interact during oocyte maturation and that the Mos pathway establishes CSF activity through stabilization of an APC-inhibitory complex composed by Emi1 and Cdc20 before fertilization.


Subject(s)
Cell Cycle Proteins/metabolism , MAP Kinase Signaling System/physiology , Metaphase , Oocytes/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Animals , Blastomeres/cytology , Blastomeres/metabolism , Cdc20 Proteins , Cells, Cultured , Female , Humans , Mice , Molecular Sequence Data , Oocytes/cytology , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-mos/metabolism , Proto-Oncogene Proteins c-mos/physiology , RNA Interference , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction , Ubiquitin-Protein Ligase Complexes/metabolism
19.
EMBO J ; 21(15): 4026-36, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12145203

ABSTRACT

In Xenopus oocytes, the c-mos proto-oncogene product has been proposed to act downstream of progesterone to control the entry into meiosis I, the transition from meiosis I to meiosis II, which is characterized by the absence of S phase, and the metaphase II arrest seen prior to fertilization. Here, we report that inhibition of Mos synthesis by morpholino antisense oligonucleotides does not prevent the progesterone-induced initiation of Xenopus oocyte meiotic maturation, as previously thought. Mos-depleted oocytes complete meiosis I but fail to arrest at metaphase II, entering a series of embryonic-like cell cycles accompanied by oscillations of Cdc2 activity and DNA replication. We propose that the unique and conserved role of Mos is to prevent mitotic cell cycles of the female gamete until the fertilization in Xenopus, starfish and mouse oocytes.


Subject(s)
Egg Proteins/physiology , Meiosis/physiology , Oocytes/cytology , Oogenesis/physiology , Proto-Oncogene Proteins c-mos/physiology , Xenopus Proteins/physiology , Xenopus laevis/physiology , Animals , CDC2 Protein Kinase/metabolism , Cyclin B/metabolism , Cyclin B2 , DNA Replication , Egg Proteins/genetics , Enzyme Activation , Female , Genetic Complementation Test , MAP Kinase Signaling System , Meiosis/drug effects , Meiosis/genetics , Morpholines/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Oogenesis/genetics , Protein Biosynthesis/drug effects , Protein Kinases/metabolism , Proto-Oncogene Proteins c-mos/deficiency , Proto-Oncogene Proteins c-mos/genetics , Recombinant Fusion Proteins/physiology , Ribosomal Protein S6 Kinases , Species Specificity , Xenopus Proteins/deficiency , Xenopus Proteins/genetics , Xenopus laevis/genetics
20.
Curr Biol ; 12(12): 1027-33, 2002 Jun 25.
Article in English | MEDLINE | ID: mdl-12123578

ABSTRACT

In vertebrate unfertilized eggs, metaphase arrest in Meiosis II is mediated by an activity known as cytostatic factor (CSF). CSF arrest is dependent upon Mos-dependent activation of the MAPK/Rsk pathway, and Rsk activates the spindle checkpoint kinase Bub1, leading to inhibition of the anaphase-promoting complex (APC), an E3 ubiquitin ligase required for the metaphase/anaphase transition. However, it is not known whether Bub1 is required for the establishment of CSF arrest or whether other pathways also contribute. Here, we show that immunodepletion of Bub1 from egg extracts blocks the ability of Mos to establish CSF arrest, and arrest can be restored by the addition of wild-type, but not kinase-dead, Bub1. The appearance of CSF arrest at Meiosis II may result from coexpression of cyclin E/Cdk2 with the MAPK/Bub1 pathway. Cyclin E/Cdk2 was able to cause metaphase arrest in egg extracts even in the absence of Mos and could also inhibit cyclin B degradation in oocytes when expressed at anaphase of Meiosis I. Once it has been established, metaphase arrest can be maintained in the absence of MAPK, Bub1, or cyclin E/Cdk2 activity. Both pathways are independent of each other, but each appears to block activation of the APC, which is required for cyclin B degradation and the metaphase/anaphase transition.


Subject(s)
CDC2-CDC28 Kinases , Cyclin E/physiology , Cyclin-Dependent Kinases/physiology , Meiosis/physiology , Protein Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-mos/physiology , Animals , Cell Line , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Xenopus , Xenopus Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...