Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Biol Chem ; 289(25): 17480-96, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24825897

ABSTRACT

The mitotic kinase Aurora A (AurA) is regulated by a complex network of factors that includes co-activator binding, autophosphorylation, and dephosphorylation. Dephosphorylation of AurA by PP2A (human, Ser-51; Xenopus, Ser-53) destabilizes the protein, whereas mitotic dephosphorylation of its T-loop (human, Thr-288; Xenopus, Thr-295) by PP6 represses AurA activity. However, AurA(Thr-295) phosphorylation is restricted throughout the early embryonic cell cycle, not just during M-phase, and how Thr-295 is kept dephosphorylated during interphase and whether or not this mechanism impacts the cell cycle oscillator were unknown. Titration of okadaic acid (OA) or fostriecin into Xenopus early embryonic extract revealed that phosphatase activity other than PP1 continuously suppresses AurA(Thr-295) phosphorylation during the early embryonic cell cycle. Unexpectedly, we observed that inhibiting a phosphatase activity highly sensitive to OA caused an abnormal increase in AurA(Thr-295) phosphorylation late during interphase that corresponded with delayed cyclin-dependent kinase 1 (CDK1) activation. AurA(Thr-295) phosphorylation indeed influenced this timing, because AurA isoforms retaining an intact Thr-295 residue further delayed M-phase entry. Using mathematical modeling, we determined that one phosphatase would be insufficient to restrict AurA phosphorylation and regulate CDK1 activation, whereas a dual phosphatase topology best recapitulated our experimental observations. We propose that two phosphatases target Thr-295 of AurA to prevent premature AurA activation during interphase and that phosphorylated AurA(Thr-295) acts as a competitor substrate with a CDK1-activating phosphatase in late interphase. These results suggest a novel relationship between AurA and protein phosphatases during progression throughout the early embryonic cell cycle and shed new light on potential defects caused by AurA overexpression.


Subject(s)
Aurora Kinase A/metabolism , Cell Cycle/physiology , Dual-Specificity Phosphatases/metabolism , Embryo, Nonmammalian/enzymology , Xenopus Proteins/metabolism , Animals , Aurora Kinase A/genetics , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Dual-Specificity Phosphatases/genetics , Embryo, Nonmammalian/cytology , Enzyme Activation/physiology , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Phosphorylation/physiology , Threonine/genetics , Threonine/metabolism , Xenopus Proteins/genetics , Xenopus laevis
2.
Phys Biol ; 8(4): 046005, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21566270

ABSTRACT

Cell signaling is achieved predominantly by reversible phosphorylation-dephosphorylation reaction cascades. Up until now, circuits conferring adaptation have all required the presence of a cascade with some type of closed topology: negative-feedback loop with a buffering node, or incoherent feed-forward loop with a proportioner node. In this paper--using Goldbeter and Koshland-type expressions--we propose a differential equation model to describe a generic, open signaling cascade that elicits an adaptation response. This is accomplished by coupling N phosphorylation-dephosphorylation cycles unidirectionally, without any explicit feedback loops. Using this model, we show that as the length of the cascade grows, the steady states of the downstream cycles reach a limiting value. In other words, our model indicates that there are a minimum number of cycles required to achieve a maximum in sensitivity and amplitude in the response of a signaling cascade. We also describe for the first time that the phenomenon of ultrasensitivity can be further subdivided into three sub-regimes, separated by sharp stimulus threshold values: OFF, OFF-ON-OFF, and ON. In the OFF-ON-OFF regime, an interesting property emerges. In the presence of a basal amount of activity, the temporal evolution of early cycles yields damped peak responses. On the other hand, the downstream cycles switch rapidly to a higher activity state for an extended period of time, prior to settling to an OFF state (OFF-ON-OFF). This response arises from the changing dynamics between a feed-forward activation module and dephosphorylation reactions. In conclusion, our model gives the new perspective that open signaling cascades embedded in complex biochemical circuits may possess the ability to show a switch-like adaptation response, without the need for any explicit feedback circuitry.


Subject(s)
Adaptation, Physiological , Models, Biological , Proteins/metabolism , Signal Transduction , Animals , Computer Simulation , Humans , Kinetics , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation
3.
Nat Cell Biol ; 5(4): 346-51, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12629549

ABSTRACT

In the early embryonic cell cycle, Cdc2-cyclin B functions like an autonomous oscillator, whose robust biochemical rhythm continues even when DNA replication or mitosis is blocked. At the core of the oscillator is a negative feedback loop; cyclins accumulate and produce active mitotic Cdc2-cyclin B; Cdc2 activates the anaphase-promoting complex (APC); the APC then promotes cyclin degradation and resets Cdc2 to its inactive, interphase state. Cdc2 regulation also involves positive feedback, with active Cdc2-cyclin B stimulating its activator Cdc25 (refs 5-7) and inactivating its inhibitors Wee1 and Myt1 (refs 8-11). Under the correct circumstances, these positive feedback loops could function as a bistable trigger for mitosis, and oscillators with bistable triggers may be particularly relevant to biological applications such as cell cycle regulation. Therefore, we examined whether Cdc2 activation is bistable. We confirm that the response of Cdc2 to non-degradable cyclin B is temporally abrupt and switch-like, as would be expected if Cdc2 activation were bistable. We also show that Cdc2 activation exhibits hysteresis, a property of bistable systems with particular relevance to biochemical oscillators. These findings help establish the basic systems-level logic of the mitotic oscillator.


Subject(s)
Biological Clocks/genetics , CDC2-CDC28 Kinases , Cell Cycle/genetics , Cyclin B/genetics , Cyclin B/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Eukaryotic Cells/enzymology , Feedback, Physiological/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Biological Clocks/drug effects , Cell Cycle/drug effects , Cell Extracts/pharmacology , Cyclin-Dependent Kinase 2 , Eukaryotic Cells/drug effects , Feedback, Physiological/drug effects , Female , Mitosis/drug effects , Mitosis/genetics , Models, Biological , Oocytes , Reaction Time/drug effects , Reaction Time/genetics , Xenopus Proteins , Xenopus laevis
4.
Curr Biol ; 17(1): 85-91, 2007 Jan 09.
Article in English | MEDLINE | ID: mdl-17208191

ABSTRACT

Mitosis is thought to be triggered by the activation of Cdk-cyclin complexes. Here we have used RNA interference (RNAi) to assess the roles of three mitotic cyclins, cyclins A2, B1, and B2, in the regulation of centrosome separation and nuclear-envelope breakdown (NEB) in HeLa cells. We found that the timing of NEB was affected very little by knocking down cyclins B1 and B2 alone or in combination. However, knocking down cyclin A2 markedly delayed NEB, and knocking down both cyclins A2 and B1 delayed NEB further. The timing of cyclin B1-Cdk1 activation was normal in cyclin A2 knockdown cells, and there was no delay in centrosome separation, an event apparently controlled by the activation of cytoplasmic cyclin B1-Cdk1. However, nuclear accumulation of cyclin B1-Cdk1 was markedly delayed in cyclin A2 knockdown cells. Finally, a constitutively nuclear cyclin B1, but not wild-type cyclin B1, restored normal NEB timing in cyclin A2 knockdown cells. These findings show that cyclin A2 is required for timely NEB, whereas cyclins B1 and B2 are not. Nevertheless cyclin B1 translocates to the nucleus just prior to NEB in a cyclin A2-dependent fashion and is capable of supporting NEB if rendered constitutively nuclear.


Subject(s)
Centrosome/metabolism , Cyclin A/physiology , Cyclin B/physiology , Mitosis/physiology , Nuclear Envelope/metabolism , Cell Nucleus/metabolism , Cyclin A2 , Cyclin B/metabolism , Cyclin B1 , Cyclin B2 , Cyclin-Dependent Kinases/metabolism , HeLa Cells , Humans
5.
Curr Opin Biotechnol ; 19(4): 381-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18634875

ABSTRACT

As the systems biology era progresses, theoreticians and experimentalists continue uncovering the molecular mechanisms that underlie the regulation of complex cellular phenomena, including those governing proliferation, differentiation, and death. The discovery of bistability in cellular responses and their signaling pathways has become a recurring theme, and prompted strong interest in understanding both the design and function of these networks. Modeling these systems has been crucial in assisting experimentalists to better understand how this and other types of behavior can emerge from a subset of regulators, and also to analyze and identify systems-level characteristics that would otherwise be difficult to intuit. In this review, recent advances in both theoretical and experimental work investigating the mechanistic as well as biological basis for bistability will be presented. These will include the role of positive feedback loops, the potential function of dual phosphorylation cycles, and substrate competition as a means of generating ultrasensitivity.


Subject(s)
Systems Biology , Cell Division , Phosphorylation
6.
Free Radic Biol Med ; 89: 263-73, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26393423

ABSTRACT

We investigated whether altering Warburg metabolism (aerobic glycolysis) by treatment with the metabolic agent dichloroacetate (DCA) could increase the X-ray-induced cell killing of the radiation-resistant human non-small-cell lung cancer (NSCLC) cell lines A549 and H1299. Treatment with 50mM DCA decreased lactate production and glucose consumption in both A549 and H1299, clear indications of attenuated aerobic glycolysis. In addition, we found that DCA treatment also slowed cell growth, increased population-doubling time, and altered cell cycle distribution. Furthermore, we report that treatment with 50mM DCA significantly increased single and fractionated X-ray-induced cell killing of A549 and H1299 cells. Assay of DNA double-strand break repair by neutral comet assays demonstrated that DCA inhibited both the fast and the slow kinetics of X-ray-induced DSB repair in both A549 and H1299 NSCL cancer cells. Taken together the data suggest a correlation between an attenuated aerobic glycolysis and enhanced cytotoxicity and radiation-induced cell killing in radiation-resistant NSCLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Dichloroacetic Acid/pharmacology , Glycolysis/physiology , Lung Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Comet Assay , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Glycolysis/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Tumor Cells, Cultured , X-Rays
7.
Mol Biol Cell ; 25(4): 441-56, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24356446

ABSTRACT

Cdh1, a coactivator of the anaphase-promoting complex (APC), is a potential tumor suppressor. Cdh1 ablation promotes precocious S-phase entry, but it was unclear how this affects DNA replication dynamics while contributing to genomic instability and tumorigenesis. We find that Cdh1 depletion causes early S-phase onset in conjunction with increase in Rb/E2F1-mediated cyclin E1 expression, but reduced levels of cyclin E1 protein promote this transition. We hypothesize that this is due to a weakened cyclin-dependent kinase inhibitor (CKI)-cyclin-dependent kinase 2 positive-feedback loop, normally generated by APC-Cdh1-mediated proteolysis of Skp2. Indeed, Cdh1 depletion increases Skp2 abundance while diminishing levels of the CKI p27. This lowers the level of cyclin E1 needed for S-phase entry and delays cyclin E1 proteolysis during S-phase progression while corresponding to slowed replication fork movement and reduced frequency of termination events. In summary, using both experimental and computational approaches, we show that APC-Cdh1 establishes a stimulus-response relationship that promotes S phase by ensuring that proper levels of p27 accumulate during G1 phase, and defects in its activation accelerate the timing of S-phase onset while prolonging its progression.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/genetics , Cadherins/genetics , Cell Transformation, Neoplastic/genetics , Feedback, Physiological , S Phase , Anaphase-Promoting Complex-Cyclosome/metabolism , Antigens, CD , Cadherins/antagonists & inhibitors , Cadherins/metabolism , Cell Transformation, Neoplastic/pathology , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA Replication , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , G1 Phase , Gene Expression Regulation , HeLa Cells , Humans , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Time Factors
8.
Cell Cycle ; 17(1): 9-10, 2018.
Article in English | MEDLINE | ID: mdl-29108455

Subject(s)
Cyclins , Cell Cycle
9.
Mol Biol Cell ; 23(2): 284-96, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22130797

ABSTRACT

Cyclin B activates cyclin-dependent kinase 1 (CDK1) at mitosis, but conflicting views have emerged on the dynamics of its synthesis during embryonic cycles, ranging from continuous translation to rapid synthesis during mitosis. Here we show that a CDK1-mediated negative-feedback loop attenuates cyclin production before mitosis. Cyclin B plateaus before peak CDK1 activation, and proteasome inhibition caused minimal accumulation during mitosis. Inhibiting CDK1 permitted continual cyclin B synthesis, whereas adding nondegradable cyclin stalled it. Cycloheximide treatment before mitosis affected neither cyclin levels nor mitotic entry, corroborating this repression. Attenuated cyclin production collaborates with its destruction, since excess cyclin B1 mRNA accelerated cyclin synthesis and caused incomplete proteolysis and mitotic arrest. This repression involved neither adenylation nor the 3' untranslated region, but it corresponded with a shift in cyclin B1 mRNA from polysome to nonpolysome fractions. A pulse-driven CDK1-anaphase-promoting complex (APC) model corroborated these results, revealing reduced cyclin levels during an oscillation and permitting more effective removal. This design also increased the robustness of the oscillator, with lessened sensitivity to changes in cyclin synthesis rate. Taken together, the results of this study underscore that attenuating cyclin synthesis late in interphase improves both the efficiency and robustness of the CDK1-APC oscillator.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle , Cyclin B/biosynthesis , Embryo, Nonmammalian/cytology , Mitosis , Adenosine Monophosphate/metabolism , Animals , Cell Division , Embryo, Nonmammalian/metabolism , Feedback, Physiological , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Xenopus laevis
10.
PLoS One ; 7(3): e33835, 2012.
Article in English | MEDLINE | ID: mdl-22479455

ABSTRACT

Mitotic progression in eukaryotic cells depends upon the activation of cyclin-dependent kinase 1 (CDK1), followed by its inactivation through the anaphase-promoting complex (APC)/cyclosome-mediated degradation of M-phase cyclins. Previous work revealed that expression of a constitutively active CDK1 (CDK1AF) in HeLa cells permitted their division, but yielded G1 daughter cells that underwent premature S-phase and early mitotic events. While CDK1AF was found to impede the sustained activity of APC-Cdh1, it was unknown if this defect improperly stabilized mitotic substrates and contributed to the occurrence of these premature M phases. Here, we show that CDK1AF expression in HeLa cells improperly stabilized APC-Cdh1 substrates in G1-phase daughter cells, including mitotic kinases and the APC adaptor, Cdc20. Division of CDK1AF-expressing cells produced G1 daughters with an accelerated S-phase onset, interrupted by the formation of premature bipolar spindles capable of spindle assembly checkpoint function. Further characterization of these phenotypes induced by CDK1AF expression revealed that this early spindle formation depended upon premature CDK1 and Aurora B activities, and their inhibition induced rapid spindle disassembly. Following its normal M-phase degradation, we found that the absence of Wee1 in these prematurely cycling daughter cells permitted the endogenous CDK1 to contribute to these premature mitotic events, since expression of a non-degradable Wee1 reduced the number of cells that exhibited premature cyclin B1oscillations. Lastly, we discovered that Cdh1-ablated cells could not be forced into a premature M phase, despite cyclin B1 overexpression and proteasome inhibition. Together, these results demonstrate that expression of constitutively active CDK1AF hampers the destruction of critical APC-Cdh1 targets, and that this type of condition could prevent newly divided cells from properly maintaining a prolonged interphase state. We propose that this more subtle type of defect in activity of the APC-driven negative-feedback loop may have implications for triggering genome instability and tumorigenesis.


Subject(s)
CDC2 Protein Kinase/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , Spindle Apparatus/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Aurora Kinase B , Aurora Kinases , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly , Cyclin B1/genetics , Cyclin B1/metabolism , DNA Replication , Enzyme Stability , Gene Expression , Gene Silencing , Genomic Instability , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints , Mad2 Proteins , Mitosis , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proteasome Inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
11.
FEBS Lett ; 583(21): 3388-96, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19818353

ABSTRACT

A positive-feedback loop is a simple motif that is ubiquitous to the modules and networks that comprise cellular signaling systems. Signaling behaviors that are synonymous with positive feedback include amplification and rapid switching, maintenance, and the coherence of outputs. Recent advances have been made towards understanding how positive-feedback loops function, as well as their mechanistic basis in controlling eukaryotic cell cycle progression. Some of these advances will be reviewed here, including: how cyclin controls passage through Start and maintains coherence of G1/S regulon expression in yeast; how Polo-like kinase 1 activation is driven by Bora and Aurora A, and its expression is stimulated by Forkhead Box M1 in mammalian cells; and how some of the various dynamic behaviors of spindle assembly and anaphase onset can be produced.


Subject(s)
Cell Cycle , Feedback, Physiological , Animals , Cell Cycle/genetics , Feedback, Physiological/genetics , Gene Expression Regulation , Humans , Mitosis , Regulon/genetics
12.
FEBS Lett ; 583(24): 3999-4005, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-19878681

ABSTRACT

Here we review some of our work over the last decade on Xenopus oocyte maturation, a cell fate switch, and the Xenopus embryonic cell cycle, a highly dynamical process. Our approach has been to start with wiring diagrams for the regulatory networks that underpin the processes; carry out quantitative experiments to describe the response functions for individual legs of the networks; and then construct simple analytical models based on chemical kinetic theory and the graphical rate-balance formalism. These studies support the view that the all-or-none, irreversible nature of oocyte maturation arises from a saddle-node bifurcation in the regulatory system that drives the process, and that the clock-like oscillations of the embryo are built upon a hysteretic switch with two saddle-node bifurcations. We believe that this type of reductionistic systems biology holds great promise for understanding complicated biochemical processes in simpler terms.


Subject(s)
Feedback, Physiological , G1 Phase , Metabolic Networks and Pathways , Models, Animal , Oocytes/physiology , Xenopus laevis , Animals , CDC2 Protein Kinase/biosynthesis , Embryo, Nonmammalian , Enzyme Activation
13.
Mol Biol Cell ; 19(8): 3426-41, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18480403

ABSTRACT

In Xenopus embryos, the cell cycle is driven by an autonomous biochemical oscillator that controls the periodic activation and inactivation of cyclin B1-CDK1. The oscillator circuit includes a system of three interlinked positive and double-negative feedback loops (CDK1 -> Cdc25 -> CDK1; CDK1 -/ Wee1 -/ CDK1; and CDK1 -/ Myt1 -/ CDK1) that collectively function as a bistable trigger. Previous work established that this bistable trigger is essential for CDK1 oscillations in the early embryonic cell cycle. Here, we assess the importance of the trigger in the somatic cell cycle, where checkpoints and additional regulatory mechanisms could render it dispensable. Our approach was to express the phosphorylation site mutant CDK1AF, which short-circuits the feedback loops, in HeLa cells, and to monitor cell cycle progression by live cell fluorescence microscopy. We found that CDK1AF-expressing cells carry out a relatively normal first mitosis, but then undergo rapid cycles of cyclin B1 accumulation and destruction at intervals of 3-6 h. During these cycles, the cells enter and exit M phase-like states without carrying out cytokinesis or karyokinesis. Phenotypically similar rapid cycles were seen in Wee1 knockdown cells. These findings show that the interplay between CDK1, Wee1/Myt1, and Cdc25 is required for the establishment of G1 phase, for the normal approximately 20-h cell cycle period, and for the switch-like oscillations in cyclin B1 abundance characteristic of the somatic cell cycle. We propose that the HeLa cell cycle is built upon an unreliable negative feedback oscillator and that the normal high reliability, slow pace and switch-like character of the cycle is imposed by a bistable CDK1/Wee1/Myt1/Cdc25 system.


Subject(s)
CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , G1 Phase , Gene Expression Regulation, Developmental , Animals , Cell Cycle , Cell Cycle Proteins/metabolism , Cyclin B/metabolism , Cyclin B1 , Gene Expression Regulation , HeLa Cells , Humans , Mitosis , Models, Biological , Mutation , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Time Factors , Xenopus
14.
Science ; 321(5885): 126-9, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18599789

ABSTRACT

A simple negative feedback loop of interacting genes or proteins has the potential to generate sustained oscillations. However, many biological oscillators also have a positive feedback loop, raising the question of what advantages the extra loop imparts. Through computational studies, we show that it is generally difficult to adjust a negative feedback oscillator's frequency without compromising its amplitude, whereas with positive-plus-negative feedback, one can achieve a widely tunable frequency and near-constant amplitude. This tunability makes the latter design suitable for biological rhythms like heartbeats and cell cycles that need to provide a constant output over a range of frequencies. Positive-plus-negative oscillators also appear to be more robust and easier to evolve, rationalizing why they are found in contexts where an adjustable frequency is unimportant.


Subject(s)
Biological Clocks , CDC2 Protein Kinase/metabolism , Cell Cycle , Feedback, Physiological , Algorithms , Anaphase-Promoting Complex-Cyclosome , Animals , Biological Evolution , Cell Division , Circadian Rhythm , Cyclin B/biosynthesis , Cyclin B/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Interphase , Models, Biological , Monte Carlo Method , Ubiquitin-Protein Ligase Complexes/metabolism , Xenopus Proteins/metabolism , Xenopus laevis
15.
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
16.
Cell ; 122(4): 565-78, 2005 Aug 26.
Article in English | MEDLINE | ID: mdl-16122424

ABSTRACT

The cell-cycle oscillator includes an essential negative-feedback loop: Cdc2 activates the anaphase-promoting complex (APC), which leads to cyclin destruction and Cdc2 inactivation. Under some circumstances, a negative-feedback loop is sufficient to generate sustained oscillations. However, the Cdc2/APC system also includes positive-feedback loops, whose functional importance we now assess. We show that short-circuiting positive feedback makes the oscillations in Cdc2 activity faster, less temporally abrupt, and damped. This compromises the activation of cyclin destruction and interferes with mitotic exit and DNA replication. This work demonstrates a systems-level role for positive-feedback loops in the embryonic cell cycle and provides an example of how oscillations can emerge out of combinations of subcircuits whose individual behaviors are not oscillatory. This work also underscores the fundamental similarity of cell-cycle oscillations in embryos to repetitive action potentials in pacemaker neurons, with both systems relying on a combination of negative and positive-feedback loops.


Subject(s)
Biological Clocks/physiology , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Feedback, Physiological/physiology , Ubiquitin-Protein Ligase Complexes/physiology , Anaphase-Promoting Complex-Cyclosome , Animals , CDC2 Protein Kinase/metabolism , Cyclins/metabolism , DNA Replication/physiology , Embryonic Development/physiology , Female , Mitosis/physiology , Models, Biological , Oocytes , Stem Cells/physiology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL