Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 7243, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37945563

ABSTRACT

Histone modifications influence the recruitment of reader proteins to chromosomes to regulate events including transcription and cell division. The idea of a histone code, where combinations of modifications specify unique downstream functions, is widely accepted and can be demonstrated in vitro. For example, on synthetic peptides, phosphorylation of Histone H3 at threonine-3 (H3T3ph) prevents the binding of reader proteins that recognize trimethylation of the adjacent lysine-4 (H3K4me3), including the TAF3 component of TFIID. To study these combinatorial effects in cells, we analyzed the genome-wide distribution of H3T3ph and H3K4me2/3 during mitosis. We find that H3T3ph anti-correlates with adjacent H3K4me2/3 in cells, and that the PHD domain of TAF3 can bind H3K4me2/3 in isolated mitotic chromatin despite the presence of H3T3ph. Unlike in vitro, H3K4 readers are still displaced from chromosomes in mitosis in Haspin-depleted cells lacking H3T3ph. H3T3ph is therefore unlikely to be responsible for transcriptional downregulation during cell division.


Subject(s)
Histones , Transcription Factors , Histones/metabolism , Phosphorylation , Transcription Factors/metabolism , Reading , Chromosomes/genetics , Chromosomes/metabolism , Mitosis/genetics
2.
Cell Rep ; 37(6): 109818, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758321

ABSTRACT

Kinetochores assemble on chromosomes in mitosis to allow microtubules to attach and bring about accurate chromosome segregation. The kinases Cyclin B-Cdk1 and Aurora B are crucial for the formation of stable kinetochores. However, the activity of these two kinases appears to decline dramatically at centromeres during anaphase onset, precisely when microtubule attachments are required to move chromosomes toward opposite poles of the dividing cell. We find that, although Aurora B leaves centromeres at anaphase, a gradient of Aurora B activity centered on the central spindle is still able to phosphorylate kinetochore substrates such as Dsn1 to modulate kinetochore stability in anaphase and to regulate kinetochore disassembly as cells enter telophase. We provide a model to explain how Aurora B co-operates with Cyclin B-Cdk1 to maintain kinetochore function in anaphase.


Subject(s)
Anaphase , Aurora Kinase B/metabolism , Chromosome Segregation , Kinetochores/enzymology , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cyclin B/genetics , Cyclin B/metabolism , Female , HeLa Cells , Humans , Phosphorylation , Protein Binding , Time Factors
3.
Nat Commun ; 12(1): 4322, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262048

ABSTRACT

Successful cell division relies on the timely removal of key cell cycle proteins such as securin. Securin inhibits separase, which cleaves the cohesin rings holding chromosomes together. Securin must be depleted before anaphase to ensure chromosome segregation occurs with anaphase. Here we find that in meiosis I, mouse oocytes contain an excess of securin over separase. We reveal a mechanism that promotes excess securin destruction in prometaphase I. Importantly, this mechanism relies on two phenylalanine residues within the separase-interacting segment (SIS) of securin that are only exposed when securin is not bound to separase. We suggest that these residues facilitate the removal of non-separase-bound securin ahead of metaphase, as inhibiting this period of destruction by mutating both residues causes the majority of oocytes to arrest in meiosis I. We further propose that cellular securin levels exceed the amount an oocyte is capable of removing in metaphase alone, such that the prometaphase destruction mechanism identified here is essential for correct meiotic progression in mouse oocytes.


Subject(s)
Meiosis , Oocytes/cytology , Securin/metabolism , Amino Acid Motifs , Animals , Chromosome Segregation , Mice , Mutation , Oocytes/metabolism , Phenylalanine/genetics , Phenylalanine/metabolism , Prometaphase , Protein Binding , Securin/chemistry , Securin/genetics , Separase/metabolism
4.
Dev Cell ; 48(5): 672-684.e5, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30745144

ABSTRACT

Successful mitosis requires that cyclin B1:CDK1 kinase activity remains high until chromosomes are correctly aligned on the mitotic spindle. It has therefore been unclear why, in mammalian oocyte meiosis, cyclin B1 destruction begins before chromosome alignment is complete. Here, we resolve this paradox and show that mouse oocytes exploit an imbalance in the ratio of cyclin B1 to CDK1 to control CDK1 activity; early cyclin B1 destruction reflects the loss of an excess of non-CDK1-bound cyclin B1 in late prometaphase, while CDK1-bound cyclin B1 is destroyed only during metaphase. The ordered destruction of the two forms of cyclin B1 is brought about by a previously unidentified motif that is accessible in free cyclin B1 but masked when cyclin B1 is in complex with CDK1. This protects the CDK1-bound fraction from destruction in prometaphase, ensuring a period of prolonged CDK1 activity sufficient to achieve optimal chromosome alignment and prevent aneuploidy.


Subject(s)
Aneuploidy , CDC2 Protein Kinase/metabolism , Cyclin B1/genetics , Oocytes/metabolism , Animals , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Female , Meiosis/physiology , Mice , Mitosis/physiology , Spindle Apparatus/metabolism
5.
Development ; 140(22): 4583-93, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24194472

ABSTRACT

The fertilising sperm triggers a transient Ca(2+) increase that releases eggs from cell cycle arrest in the vast majority of animal eggs. In vertebrate eggs, Erp1, an APC/C(cdc20) inhibitor, links release from metaphase II arrest with the Ca(2+) transient and its degradation is triggered by the Ca(2+)-induced activation of CaMKII. By contrast, many invertebrate groups have mature eggs that arrest at metaphase I, and these species do not possess the CaMKII target Erp1 in their genomes. As a consequence, it is unknown exactly how cell cycle arrest at metaphase I is achieved and how the fertilisation Ca(2+) transient overcomes the arrest in the vast majority of animal species. Using live-cell imaging with a novel cyclin reporter to study cell cycle arrest and its release in urochordate ascidians, the closest living invertebrate group to the vertebrates, we have identified a new signalling pathway for cell cycle resumption in which CaMKII plays no part. Instead, we find that the Ca(2+)-activated phosphatase calcineurin (CN) is required for egg activation. Moreover, we demonstrate that parthenogenetic activation of metaphase I-arrested eggs by MEK inhibition, independent of a Ca(2+) increase, requires the activity of a second egg phosphatase: PP2A. Furthermore, PP2A activity, together with CN, is required for normal egg activation during fertilisation. As ascidians are a sister group of the vertebrates, we discuss these findings in relation to cell cycle arrest and egg activation in chordates.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Cycle Checkpoints , Meiosis , Ovum/cytology , Phosphoprotein Phosphatases/metabolism , Urochordata/cytology , Urochordata/enzymology , Anaphase-Promoting Complex-Cyclosome/antagonists & inhibitors , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , Antigens, Polyomavirus Transforming/metabolism , Calcineurin/metabolism , Calcineurin Inhibitors , Calcium/pharmacology , Calcium Signaling/drug effects , Cell Cycle Checkpoints/drug effects , Cyclin B/metabolism , Enzyme Activation/drug effects , Fertilization/drug effects , Mammals/metabolism , Meiosis/drug effects , Metaphase/drug effects , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Ovum/enzymology , Protein Phosphatase 2/metabolism , Rats , Substrate Specificity/drug effects , Urochordata/drug effects
6.
Methods Mol Biol ; 957: 121-34, 2013.
Article in English | MEDLINE | ID: mdl-23138948

ABSTRACT

Fluorescently tagged proteins have become a crucial weapon in the armory of a successful cell biology laboratory. This chapter describes how to produce cRNA coding for a fluorescently tagged protein of choice, such that it is suitable for microinjection and subsequent expression studies in live oocytes.


Subject(s)
Green Fluorescent Proteins/genetics , Luminescent Proteins/genetics , Microinjections/methods , Molecular Imaging/methods , Oocytes/cytology , Oocytes/metabolism , RNA, Complementary/genetics , Animals , Cell Survival , Escherichia coli/genetics , Gene Expression , Genetic Vectors/genetics , Plasmids/genetics , Polymerase Chain Reaction , RNA, Complementary/administration & dosage , Transcription, Genetic , Transformation, Genetic , Red Fluorescent Protein
7.
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
8.
Dev Biol ; 321(2): 379-86, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18639540

ABSTRACT

Mammalian eggs remain arrested at metaphase of the second meiotic division (metII) for an indeterminate time before fertilization. During this period, which can last several hours, the continued attachment of sister chromatids is thought to be achieved by inhibition of the protease separase. Separase is known to be inhibited by binding either securin or Maturation (M-Phase)-Promoting Factor, a heterodimer of CDK1/cyclin B1. However, the relative contribution of securin and CDK/cyclin B1 to sister chromatid attachment during metII arrest has not been assessed. Although there are conditions in which either CDK1/cyclinB1 activity or securin can prevent sister chromatid disjunction, principally by overexpression of non-degradable cyclin B1 or securin, we find here that separase activity is primarily regulated by securin and not CDK1/cyclin B1. Thus the CDK1 inhibitor roscovitine and an antibody we designed to block the interaction of CDK1/cyclin B1 with separase, both failed to induce sister disjunction. In contrast, securin morpholino knockdown specifically induced loss of sister attachment, that could be restored by securin cRNA rescue. During metII arrest separase appears primarily regulated by securin binding, not CDK1/cyclin B1.


Subject(s)
Carrier Proteins/metabolism , Chromatids/metabolism , Meiosis/physiology , Ovum/cytology , Animals , Blotting, Western , Cell Cycle Proteins/metabolism , Cytogenetic Analysis , Endopeptidases/metabolism , Mice , Oligonucleotides , Securin , Separase
9.
Nat Cell Biol ; 9(10): 1192-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17891138

ABSTRACT

The first female meiotic division (meiosis I, MI) is uniquely prone to chromosome segregation errors through non-disjunction, resulting in trisomies and early pregnancy loss. Here, we show a fundamental difference in the control of mammalian meiosis that may underlie such susceptibility. It involves a reversal in the well-established timing of activation of the anaphase-promoting complex (APC) by its co-activators cdc20 and cdh1. APC(cdh1) was active first, during prometaphase I, and was needed in order to allow homologue congression, as loss of cdh1 speeded up MI, leading to premature chromosome segregation and a non-disjunction phenotype. APC(cdh1) targeted cdc20 for degradation, but did not target securin or cyclin B1. These were degraded later in MI through APC(cdc20), making cdc20 re-synthesis essential for successful meiotic progression. The switch from APC(cdh1) to APC(cdc20) activity was controlled by increasing CDK1 and cdh1 loss. These findings demonstrate a fundamentally different mechanism of control for the first meiotic division in mammalian oocytes that is not observed in meioses of other species.


Subject(s)
Oocytes/metabolism , Prometaphase/physiology , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cdc20 Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin B/genetics , Cyclin B/metabolism , Cyclin B1 , Female , Meiosis/genetics , Meiosis/physiology , Mice , Microscopy, Fluorescence , Prometaphase/genetics , Securin , Time Factors , Ubiquitin-Protein Ligase Complexes/genetics
10.
J Cell Sci ; 120(Pt 10): 1763-71, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17502483

ABSTRACT

Fertilisation in ascidians triggers a series of periodic rises in cytosolic Ca(2+) that are essential for release from metaphase I arrest and progression through meiosis II. These sperm-triggered Ca(2+) oscillations are switched off at exit from meiosis II. Ascidian zygotes provided the first demonstration of the positive feedback loop whereby elevated Cdk1 activity maintained these Ca(2+) oscillations. Since then it has been reported that Cdk1 sensitises the type I inositol trisphosphate [Ins(1,4,5)P(3)] receptor in somatic cells, and that sperm-triggered Ca(2+) oscillations in mouse zygotes stop because the forming pronuclei sequester phospholipase C zeta that was delivered to the egg by the fertilising sperm. Here, using enucleation, we demonstrate in ascidian eggs that Ca(2+) spiking stops at the correct time in the absence of pronuclei. Sequestration of sperm factor is therefore not involved in terminating Ca(2+) spiking for these eggs. Instead we found that microinjection of the Cdk1 inhibitor p21 blocked Ca(2+) spiking induced by ascidian sperm extract (ASE). However, such eggs were still capable of releasing Ca(2+) in response to Ins(1,4,5)P(3) receptor agonists, indicating that ASE-triggered Ca(2+) oscillations can stop even though the response to Ins(1,4,5)P(3) remained elevated. These data suggest that Cdk1 activity promotes Ins(1,4,5)P(3) production in the presence of the sperm factor, rather than sensitising the Ca(2+) releasing machinery to Ins(1,4,5)P(3). These findings suggest a new link between this cell cycle kinase and the Ins(1,4,5)P(3) pathway.


Subject(s)
Biological Clocks/physiology , CDC2 Protein Kinase/metabolism , Calcium Signaling/physiology , Meiosis/physiology , Oocytes/metabolism , Urochordata/metabolism , Animals , Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Female , Fertilization/physiology , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Oocytes/cytology , Signal Transduction/physiology , Sperm-Ovum Interactions/physiology , Spermatozoa/metabolism , Urochordata/cytology
11.
J Cell Biol ; 174(6): 791-801, 2006 Sep 11.
Article in English | MEDLINE | ID: mdl-16966421

ABSTRACT

During interkinesis, a metaphase II (MetII) spindle is built immediately after the completion of meiosis I. Oocytes then remain MetII arrested until fertilization. In mouse, we find that early mitotic inhibitor 2 (Emi2), which is an anaphase-promoting complex inhibitor, is involved in both the establishment and the maintenance of MetII arrest. In MetII oocytes, Emi2 needs to be degraded for oocytes to exit meiosis, and such degradation, as visualized by fluorescent protein tagging, occurred tens of minutes ahead of cyclin B1. Emi2 antisense morpholino knockdown during oocyte maturation did not affect polar body (PB) extrusion. However, in interkinesis the central spindle microtubules from meiosis I persisted for a short time, and a MetII spindle failed to assemble. The chromatin in the oocyte quickly decondensed and a nucleus formed. All of these effects were caused by the essential role of Emi2 in stabilizing cyclin B1 after the first PB extrusion because in Emi2 knockdown oocytes a MetII spindle was recovered by Emi2 rescue or by expression of nondegradable cyclin B1 after meiosis I.


Subject(s)
Cyclin B/metabolism , Cytokinesis/physiology , F-Box Proteins/metabolism , Meiosis/physiology , Oocytes/metabolism , Oogenesis/physiology , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cyclin B1 , Down-Regulation/physiology , F-Box Proteins/antagonists & inhibitors , F-Box Proteins/genetics , Female , Metaphase/physiology , Mice , Microtubules/metabolism , Microtubules/ultrastructure , Oligonucleotides, Antisense/pharmacology , Oocytes/ultrastructure , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
12.
EMBO Rep ; 7(10): 1040-5, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16878123

ABSTRACT

Cdc20 and cdh1 are coactivators of the anaphase-promoting complex (APC). APC(cdc20) is necessary for the metaphase-anaphase transition and, at the end of mitosis, vertebrate cdc20 itself becomes a target for degradation through KEN-box-dependent APC(cdh1) activity. By studying the degradation of fluorescent protein chimaeras in mammalian oocytes and early embryos, we found that cdc20 was degraded through two independent degradation signals (degrons), the KEN box and a newly described CRY box. In both oocytes and G1-stage embryos, the rate of degradation through the CRY box was greater than through the KEN box, although both were mediated by APC(cdh1). Thus, mammalian oocytes and embryos have the capacity to recognize two degrons in cdc20.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cdc20 Proteins , Cell Cycle Proteins/genetics , Embryo, Mammalian/metabolism , Female , G1 Phase , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Meiotic Prophase I , Mice , Oocytes/metabolism , Protein Denaturation , Protein Sorting Signals , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Nat Cell Biol ; 8(5): 539-40, 2006 May.
Article in English | MEDLINE | ID: mdl-16715549

ABSTRACT

Fully grown mammalian oocytes maintain a prophase I germinal-vesicle stage arrest in the ovary for extended periods before a luteinizing hormone surge induces entry into the first meiotic division. Cdh1 is an activator of the anaphase-promoting complex (APC) and APCcdh1 is normally restricted to late M to early G1 phases of the cell cycle. Here, we find that APCcdh1 is active in mouse oocytes and is necessary to maintain prophase arrest.


Subject(s)
Meiotic Prophase I/physiology , Oocytes/cytology , Oocytes/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Mice , RNA, Complementary/genetics , Time Factors
14.
Reproduction ; 130(6): 829-43, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322543

ABSTRACT

Mad2 is a pivotal component of the spindle assembly checkpoint (SAC) which inhibits anaphase promoting complex/cyclo-some (APC/C) activity by sequestering Cdc20 thereby regulating the destruction of securin and cyclin B. During mitosis, spindle depolymerisation induces a robust Mad2-dependent arrest due to inhibition of securin and cyclin B destruction. In contrast to mitosis, the molecular details underpinning the meiosis I arrest experienced by mouse oocytes exposed to spindle depolymerisation remain incompletely characterised. Notably, the role of Mad2 and the fate of the anaphase-marker, securin, are unexplored. As shown previously, we find that spindle depolymerisation by nocodazole inhibits first polar body extrusion (PBE) and stabilises cyclin B and cyclin-dependent kinase 1 activity in mouse oocytes. Here we show that stabilisation of cyclin B in nocodazole can be sustained for several hours and is associated with stabilisation of securin. These effects are SAC-mediated as, in oocytes depleted of the majority of Mad2 by morpholino antisense, securin and cyclin B are destabilised and 15% of oocytes undergo PBE. This reflects premature APC/C activation as a mutant form of cyclin B lacking its APC/C degradation signal is stable in Mad2-depleted oocytes. Moreover, homologues do not disjoin during the prolonged meiosis I arrest (> 18 h) induced by nocodaozole indicating that a non-cleavage mechanism is insufficient on its own for resolution of arm cohesion in mammalian oocytes. In conclusion, when all kinetochores lack attachment and tension, mouse oocytes mount a robust Mad2-dependent meiosis I arrest which inhibits the destruction of securin and cyclin B.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclin B/metabolism , Meiosis/physiology , Oocytes/metabolism , Spindle Apparatus/metabolism , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cells, Cultured , Cyclin B1 , Female , Fluorescent Antibody Technique , Histones/metabolism , Immunoblotting/methods , Mad2 Proteins , Mice , Mice, Inbred Strains , Microtubules/drug effects , Microtubules/metabolism , Myelin Basic Protein/metabolism , Nocodazole/pharmacology , Oligonucleotides, Antisense/pharmacology , Oocytes/cytology , Securin , Time Factors
15.
J Cell Sci ; 118(Pt 17): 3849-59, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16091425

ABSTRACT

Mouse eggs arrest at metaphase II following ovulation and are only triggered to complete meiosis when fertilized. Sperm break the cell-cycle arrest by a long-lasting series of Ca2+ spikes that lead to an activation of the anaphase-promoting complex/cyclosome. The signal transduction pathway is not fully resolved but both protein kinase C (PKC) and calmodulin-dependent protein kinase II (CamKII) activities increase at fertilization and previous pharmacological studies have implicated both in cell-cycle resumption. We have used a combination of pharmacological inhibitors and constitutively active cRNA constructs of PKCalpha and CamKIIalpha microinjected into mouse eggs to show that it is CamKII and not PKC that is the sufficient trigger for cell-cycle resumption from metaphase II arrest. Constitutively active PKC constructs had no effect on the resumption of meiosis but caused an immediate and persistent elevation in intracellular Ca2+ when store-operated Ca2+ entry was stimulated. With respect to resumption of meiosis, the effects of constitutively active CamKII on eggs were the same as sperm. Eggs underwent second polar body extrusion and pronucleus formation with normal timings; while both securin and cyclin B1 destruction, visualised by coupling to fluorescent protein tags, were complete by the time of polar body extrusion. Induction of a spindle checkpoint by overexpression of Mad2 or by spindle poisons blocked CamKII-induced resumption of meiosis, but the Ca2+ chelator BAPTA did not. Furthermore direct measurement of Ca2+ levels showed that CamKII did not induce exit from metaphase II arrest by raising Ca2+. Therefore, we conclude that PKCs may play an important role in maintaining Ca2+ spiking at fertilization by promoting store-operated Ca2+ entry, while CamKII transduces cell-cycle resumption, and lies downstream of sperm-induced Ca2+ release but upstream of a spindle checkpoint. These data, combined with the knowledge that CamKII activity increase at fertilization, suggest that mouse eggs undergo cell-cycle resumption through stimulation of CamKII.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Meiosis/physiology , Oocytes/physiology , Protein Kinase C/metabolism , Animals , Benzylamines/metabolism , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Carrier Proteins/metabolism , Cyclin B/metabolism , Cyclin B1 , Female , Fertilization , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Oocytes/cytology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase Inhibitors/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Securin , Signal Transduction/physiology , Sulfonamides/metabolism
16.
Mol Hum Reprod ; 11(6): 397-404, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15908454

ABSTRACT

Although female meiosis I errors account for the majority of human aneuploidy, their molecular basis is largely unknown. By elucidating gene function, gene knockdown using RNA interference (RNAi) could shed light on this enigmatic process. In practice, however, the extreme paucity of immature human oocytes makes the evaluation of gene-targeting tools difficult. Here, we undertake RNAi in human oocytes and describe an approach employing mouse oocytes which could overcome the problem of limited biological material. We designed a short interfering RNA (siRNA) designated si539 to target the human mitotic arrest deficient 2 (hMad2) spindle checkpoint component. In human oocytes microinjected with si539, the hMad2 signal detected by Western blotting was 85-92% less intense than in oocytes injected with control siRNA indicating efficient silencing. Further examination of si539's targeting efficiency was undertaken using a green fluorescent protein (GFP)-tagged hMad2 mRNA construct in mouse oocytes. Consistent with Western blot analysis, si539 reduced hMad2-GFP expression in mouse oocytes by approximately 94% and relieved the meiosis I arrest otherwise induced by hMad2-GFP in mouse oocytes. By facilitating the investigation of candidate genes involved in regulating human female meiosis I, this approach can bring us closer to understanding the origins of aneuploidies such as Down's syndrome.


Subject(s)
Aneuploidy , Calcium-Binding Proteins/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Meiosis/genetics , Oocytes/cytology , RNA Interference , Repressor Proteins/antagonists & inhibitors , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Female , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Mad2 Proteins , Mice , Oocytes/drug effects , Oocytes/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Repressor Proteins/genetics , Repressor Proteins/metabolism
17.
Cell Cycle ; 4(5): 650-3, 2005 May.
Article in English | MEDLINE | ID: mdl-15846076

ABSTRACT

In mammalian somatic cells, the spindle assembly checkpoint (SAC) is indispensable for ensuring the fidelity of chromosome segregation by delaying cell-cycle progression in the face of even a single misaligned chromosome. In contrast, the role of the SAC in unperturbed mammalian oocytes is less well defined as progression through meiosis I is unaltered in mouse oocytes in the presence of one or a few misaligned chromosomes. Furthermore, attempts to disable the function of the SAC protein, Mad2, in mouse oocytes have produced conflicting results. To gain further insight into SAC function during female mammalian meiosis I, we recently utilised a morpholino-based antisense approach to deplete the majority of Mad2 in mouse oocytes. Our results define a clear role for Mad2 in ensuring the proper timing of meiosis I events and ultimately, in ensuring the fidelity of homologue disjunction. We discuss the implications of these results for the regulation of meiosis I in mammalian oocytes and for the genesis of human aneuploidy.


Subject(s)
Cell Cycle Proteins/physiology , Meiotic Prophase I/physiology , Oogenesis/physiology , Spindle Apparatus/physiology , Anaphase-Promoting Complex-Cyclosome , Aneuploidy , Animals , Carrier Proteins/analysis , Carrier Proteins/physiology , Cell Cycle Proteins/analysis , Cyclin B/analysis , Cyclin B/physiology , Female , Humans , Mad2 Proteins , Mice , Mitosis/physiology , Nuclear Proteins/analysis , Nuclear Proteins/physiology , Oocytes/chemistry , Oocytes/cytology , Oocytes/physiology , Securin , Ubiquitin-Protein Ligase Complexes/analysis , Ubiquitin-Protein Ligase Complexes/physiology
18.
Genes Dev ; 19(2): 202-7, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15655110

ABSTRACT

In mitosis, the spindle checkpoint protein Mad2 averts aneuploidy by delaying anaphase onset until chromosomes align. Here we show that depletion of Mad2 in meiosis I mouse oocytes induced an increased incidence of aneuploidy. Proteolysis of cyclin B and securin commenced earlier in Mad2-depleted oocytes, resulting in a shortened duration of meiosis I. Furthermore, overexpression of Mad2 inhibited homolog disjunction. We conclude that Mad2 delays the onset of cyclin B and securin degradation and averts aneuploidy during meiosis I in mammalian oocytes. The data suggest a link between trisomies such as Down syndrome and defective oocyte spindle checkpoint function.


Subject(s)
Aneuploidy , Carrier Proteins/metabolism , Cyclin B/metabolism , Meiotic Prophase I/physiology , Oocytes/physiology , Anaphase/physiology , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Down Syndrome/genetics , Down Syndrome/metabolism , Mad2 Proteins , Meiotic Prophase I/genetics , Mice , Mitosis/genetics , Mitosis/physiology , Nuclear Proteins , Oocytes/cytology , Securin
19.
J Cell Sci ; 117(Pt 26): 6289-96, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15561765

ABSTRACT

Metaphase II-arrested mouse eggs are stimulated to complete meiosis by sperm-induced Ca2+ spiking. The Ca2+ signal causes activation of the E3 ligase anaphase-promoting complex/cyclosome (APC), leading to the destruction of key proteins necessary for meiotic exit. We show, using western blots of mouse eggs, the presence of both APC activators cdc20 and cdh1, which target D-box and D-box/KEN-box substrates, respectively, for proteolysis. We decided to examine the temporal activation of APCcdc20 and APCcdh1 by coupling APC substrates to GFP and examining their destruction in real-time following release from second meiotic division arrest. D-box substrates were degraded quickly after the initiation of sperm-induced Ca2+ spiking, such that their degradation was complete by the time of second polar body extrusion. By contrast, KEN-box-containing substrates were degraded when CDK1 activity was low, during the period between polar body extrusion and pronucleus formation. This observation of apparent APCcdh1 activity in meiosis II based on destruction of exogenous GFP-coupled substrates was then confirmed by observing destruction of endogenous APCcdh1 substrates. These data are consistent with a model of initial APCcdc20 activation on sperm-induced activation, followed by APCcdh1 activation after second polar body extrusion. Interestingly, therefore, we propose that mammalian eggs undergo meiosis II with both APCcdc20 and APCcdh1, whereas eggs of other species so far described have APCcdc20 activity only.


Subject(s)
Cell Cycle Proteins/metabolism , Meiosis , Oocytes , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Animals, Outbred Strains , Blotting, Western , Calcium/metabolism , Calcium Signaling , Carrier Proteins/metabolism , Cdc20 Proteins , Cyclin B/metabolism , Cyclin B1 , Enzyme Activation , Female , Green Fluorescent Proteins/metabolism , Kinetics , Mice , Microinjections , Securin , Tritium , Ubiquitin-Protein Ligases/metabolism
20.
Dev Biol ; 275(1): 68-81, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15464573

ABSTRACT

Mammalian eggs naturally arrest at metaphase of the second meiotic division, until sperm triggers a series of Ca(2+) spikes that result in activation of the anaphase-promoting complex/cyclosome (APC/C). APC/C activation at metaphase targets destruction-box containing substrates, such as cyclin B1 and securin, for degradation, and as such eggs complete the second meiotic division. Cyclin B1 degradation reduces maturation (M-phase)-promoting factor (MPF) activity and securin degradation allows sister chromatid separation. Here we examined the second meiotic division in mouse eggs following expression of a cyclin B1 construct with an N-terminal 90 amino acid deletion (Delta 90 cyclin B1) that was visualized by coupling to EGFP. This cyclin construct was not an APC/C substrate, and so following fertilization, sperm were incapable of stimulating Delta 90 cyclin B1 degradation. In these eggs, chromatin remained condensed and no pronuclei formed. As a consequence of the lack of pronucleus formation, sperm-triggered Ca(2+) spiking continued indefinitely, consistent with a current model in which the sperm-activating factor is localized to the nucleus. Because Ca(2+) spiking was not inhibited by Delta 90 cyclin B1, the degradation timing of securin, visualized by coupling it to EGFP, was unaffected. However, despite rapid securin degradation, sister chromatids remained attached. This was a direct consequence of MPF activity because separation was induced following application of the MPF inhibitor roscovitine. Similar observations regarding the ability of MPF to prevent sister chromatid separation have recently been made in Xenopus egg extracts and in HeLa cells. The results presented here show this mechanism can also occur in intact mammalian eggs and further that this mechanism appears conserved among vertebrates. We present a model in which metaphase II arrest is maintained primarily by MPF levels only.


Subject(s)
Chromatids/physiology , Cyclin B/genetics , Maturation-Promoting Factor/physiology , Ovum/physiology , Animals , Calcium/metabolism , Cell Nucleus/physiology , Chromatids/drug effects , Chromosome Pairing/physiology , Cyclin B/metabolism , Cyclin B1 , Female , Fertilization/physiology , Growth Inhibitors/pharmacology , Mesothelin , Mice , Purines/pharmacology , Roscovitine , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...