Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Biochem J ; 479(19): 2115-2130, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36240068

ABSTRACT

Claspin is an adaptor protein required for ATR-dependent phosphorylation of CHK1 during S-phase following DNA replication stress. Claspin expression is highly variable in cancer, with low levels frequently correlating with poor patient survival. To learn more about the biological consequences of reduced Claspin expression and its effects on tumorigenesis, we investigated mice with a heterozygous knockout of the Clspn gene. Claspin haploinsufficiency resulted in reduced female fertility and a maternally inherited defect in oocyte meiosis I cell cycle progression. Furthermore, aged Clspn+/- mice developed spontaneous lymphoid hyperplasia and increased susceptibility to non-alcoholic fatty liver disease. Importantly, we demonstrate a tumour suppressor role for Claspin. Reduced Claspin levels result in increased liver damage and tumourigenesis in the DEN model of hepatocellular carcinoma. These data reveal that Clspn haploinsufficiency has widespread unanticipated biological effects and establishes the importance of Claspin as a regulatory node controlling tumorigenesis and multiple disease aetiologies.


Subject(s)
DNA Replication , Haploinsufficiency , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1 , Female , Fertility/genetics , Hyperplasia , Mice , Phosphorylation
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.
Nat Struct Mol Biol ; 25(7): 557-569, 2018 07.
Article in English | MEDLINE | ID: mdl-29915389

ABSTRACT

Meiotic chromosomes adopt unique structures in which linear arrays of chromatin loops are bound together in homologous chromosome pairs by a supramolecular protein assembly, the synaptonemal complex. This three-dimensional scaffold provides the essential structural framework for genetic exchange by crossing over and subsequent homolog segregation. The core architecture of the synaptonemal complex is provided by SYCP1. Here we report the structure and self-assembly mechanism of human SYCP1 through X-ray crystallographic and biophysical studies. SYCP1 has an obligate tetrameric structure in which an N-terminal four-helical bundle bifurcates into two elongated C-terminal dimeric coiled-coils. This building block assembles into a zipper-like lattice through two self-assembly sites. N-terminal sites undergo cooperative head-to-head assembly in the midline, while C-terminal sites interact back to back on the chromosome axis. Our work reveals the underlying molecular structure of the synaptonemal complex in which SYCP1 self-assembly generates a supramolecular lattice that mediates meiotic chromosome synapsis.


Subject(s)
Chromosome Pairing/physiology , Nuclear Proteins/chemistry , Biophysical Phenomena , Crystallography, X-Ray , DNA-Binding Proteins , Humans , Meiosis/physiology , Models, Molecular , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Static Electricity , Synaptonemal Complex/chemistry
6.
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
7.
Cell Div ; 2: 4, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17257429

ABSTRACT

Oocytes from higher chordates, including man and nearly all mammals, arrest at metaphase of the second meiotic division before fertilization. This arrest is due to an activity that has been termed 'Cytostatic Factor'. Cytostatic Factor maintains arrest through preventing loss in Maturation-Promoting Factor (MPF; CDK1/cyclin B). Physiologically, Cytostatic Factor - induced metaphase arrest is only broken by a Ca2+ rise initiated by the fertilizing sperm and results in degradation of cyclin B, the regulatory subunit of MPF through the Anaphase-Promoting Complex/Cyclosome (APC/C). Arrest at metaphase II may therefore be viewed as being maintained by inhibition of the APC/C, and Cytostatic Factor as being one or more pathways, one of which inhibits the APC/C, consorting in the preservation of MPF activity. Many studies over several years have implicated the c-Mos/MEK/MAPK pathway in the metaphase arrest of the two most widely studied vertebrates, frog and mouse. Murine downstream components of this cascade are not known but in frog involve members of the spindle assembly checkpoint, which act to inhibit the APC/C. Interesting these downstream components appear not to be involved in the arrest of mouse eggs, suggesting a lack of conservation with respect to c-Mos targets. However, the recent discovery of Emi2 as an egg specific APC/C inhibitor whose degradation is Ca2+ dependent has greatly increased our understanding of MetII arrest. Emi2 is involved in both the establishment and maintenance of metaphase II arrest in frog and mouse suggesting a conservation of metaphase II arrest. Its identity as the physiologically relevant APC/C inhibitor involved in Cytostatic Factor arrest prompted us to re-evaluate the role of the c-Mos pathway in metaphase II arrest. This review presents a model of Cytostatic Factor arrest, which is primarily induced by Emi2 mediated APC/C inhibition but which also requires the c-Mos pathway to set MPF levels within physiological limits, not too high to induce an arrest that cannot be broken, or too low to induce parthenogenesis.

8.
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
9.
Nat Cell Biol ; 8(9): 1035-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16906143

ABSTRACT

Separase not only triggers anaphase of meiosis I by proteolytic cleavage of cohesin on chromosome arms, but in vitro vertebrate separase also acts as a direct inhibitor of cyclin-dependent kinase 1 (Cdk1) on liberation from the inhibitory protein, securin. Blocking separase-Cdk1 complex formation by microinjection of anti-separase antibodies prevents polar-body extrusion in vertebrate oocytes. Importantly, proper meiotic maturation is rescued by chemical inhibition of Cdk1 or expression of Cdk1-binding separase fragments lacking cohesin-cleaving activity.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Endopeptidases/metabolism , Meiosis/physiology , Oocytes/physiology , Animals , CDC2 Protein Kinase/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line , Endopeptidases/genetics , Female , Histones/metabolism , Humans , Mice , Neoplasm Proteins/metabolism , Oocytes/metabolism , Securin , Separase , Xenopus
10.
Dev Biol ; 296(2): 388-95, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16824507

ABSTRACT

Fertilization in mammalian eggs is accompanied by oscillatory changes in intracellular Ca(2+) concentration, which are critical for initiating and completing egg activation events and the developmental program. Ca(2+)/Camodulin-dependent protein kinase II (CaMKII) is a multifunctional enzyme that is postulated to be the downstream transducer of the Ca(2+) signal in many cell types. We tested the hypothesis that CaMKII is the major integrator of Ca(2+)-induced egg activation events and embryo development by microinjecting a cRNA that encodes a constitutively active (Ca(2+)-independent) mutant form of CaMKII (CA-CaMKII) into mouse eggs. Expression of this cRNA, which does not increase intracellular Ca(2+), induced a sustained rise in CaMKII activity and triggered egg activation events, including cell cycle resumption, and degradation and recruitment of maternal mRNAs; cortical granule exocytosis, however, did not occur normally. Furthermore, when mouse eggs were injected with sperm devoid of Ca(2+)-releasing activity and activated with either CA-CaMKII cRNA or by SrCl(2), similar rates and incidence of development to the blastocyst stage were observed. These results strongly suggest that CaMKII is a major integrator of the Ca(2+) changes that occur following fertilization.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Calcium/metabolism , Embryonic Development/physiology , Ovum/enzymology , Animals , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Female , Fertilization/physiology , Male , Mice , Mice, Inbred C57BL , Ovum/metabolism , Sperm Injections, Intracytoplasmic , Sperm-Ovum Interactions/physiology , Spermatozoa/physiology , Tissue Culture Techniques
11.
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
12.
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
...