Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Dev Cell ; 47(4): 494-508.e4, 2018 11 19.
Article En | MEDLINE | ID: mdl-30473004

Cell-cell heterogeneity can facilitate lineage choice during embryonic development because it primes cells to respond to differentiation cues. However, remarkably little is known about the origin of heterogeneity or whether intrinsic and extrinsic variation can be controlled to generate reproducible cell type proportioning seen in vivo. Here, we use experimentation and modeling in D. discoideum to demonstrate that population-level cell cycle heterogeneity can be optimized to generate robust cell fate proportioning. First, cell cycle position is quantitatively linked to responsiveness to differentiation-inducing signals. Second, intrinsic variation in cell cycle length ensures cells are randomly distributed throughout the cell cycle at the onset of multicellular development. Finally, extrinsic perturbation of optimal cell cycle heterogeneity is buffered by compensatory changes in global signal responsiveness. These studies thus illustrate key regulatory principles underlying cell-cell heterogeneity optimization and the generation of robust and reproducible fate choice in development.


Cell Cycle/physiology , Cell Differentiation/physiology , Cell Division/physiology , Dictyostelium/metabolism , Animals , Cell Lineage/physiology , Spores, Fungal/metabolism
2.
Cell Cycle ; 13(15): 2370-8, 2014.
Article En | MEDLINE | ID: mdl-25483188

Sister chromatid separation creates a sudden loss of tension on kinetochores, which could, in principle, re-activate the spindle checkpoint in anaphase. This so-called "anaphase problem" is probably avoided by timely inactivation of cyclin B1-Cdk1, which may prevent the spindle tension sensing Aurora B kinase from destabilizing kinetochore-microtubule interactions as they lose tension in anaphase. However, exactly how spindle checkpoint re-activation is prevented remains unclear. Here, we investigated how different degrees of cyclin B1 stabilization affected the spindle checkpoint in metaphase and anaphase. Cells expressing a strongly stabilized (R42A) mutant of cyclin B1 degraded APC/C(Cdc20) substrates normally, showing that checkpoint release was not inhibited by high cyclin B1-Cdk1 activity. However, after this initial wave of APC/C(Cdc20) activity, the spindle checkpoint returned in cells with uncohesed sister chromatids. Expression of a lysine mutant of cyclin B1 that is degraded only slightly inefficiently allowed a normal metaphase-to-anaphase transition. Strikingly, however, the spindle checkpoint returned in cells that had not degraded the cyclin B1 mutant 10-15 min after anaphase onset. When cyclin B1 remained in late anaphase, cytokinesis stalled, and translocation of INCENP from separated sister chromatids to the spindle midzone was blocked. This late anaphase arrest required the activity of Aurora B and Mps1. In conclusion, our results reveal that complete removal of cyclin B1 is essential to prevent the return of the spindle checkpoint following sister chromatid disjunction. Speculatively, increasing activity of APC/C(Cdc20) in late anaphase helps to keep cyclin B1 levels low.


Cyclin B1/metabolism , M Phase Cell Cycle Checkpoints/physiology , Proteolysis , Sister Chromatid Exchange/physiology , Aurora Kinase B/metabolism , CDC2 Protein Kinase , Cdc20 Proteins/metabolism , Cell Line, Tumor , Cyclin B1/genetics , Cyclin-Dependent Kinases/metabolism , Humans , Lysine/metabolism , Merozoite Surface Protein 1/metabolism , Mutation
3.
J Cell Biol ; 190(4): 587-602, 2010 Aug 23.
Article En | MEDLINE | ID: mdl-20733055

The ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C) is activated at prometaphase by mitotic phosphorylation and binding of its activator, Cdc20. This initiates cyclin A degradation, whereas cyclin B1 is stabilized by the spindle checkpoint. Upon checkpoint release, the RXXL destruction box (D box) was proposed to direct cyclin B1 to core APC/C or Cdc20. In this study, we report that endogenous cyclin B1-Cdk1 is recruited to checkpoint-inhibited, phosphorylated APC/C in prometaphase independently of Cdc20 or the cyclin B1 D box. Like cyclin A, cyclin B1 binds the APC/C by the Cdk cofactor Cks and the APC3 subunit. Prior binding to APC/C(Cdc20) makes cyclin B1 a better APC/C substrate in metaphase, driving mitotic exit and cytokinesis. We conclude that in prometaphase, the phosphorylated APC/C can recruit both cyclin A and cyclin B1 in a Cks-dependent manner. This suggests that the spindle checkpoint blocks D box recognition of APC/C-bound cyclin B1, whereas distinctive complexes between the N terminus of cyclin A and Cdc20 evade checkpoint control.


CDC2 Protein Kinase/metabolism , Carrier Proteins/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinases/metabolism , Mitosis/physiology , Prometaphase/physiology , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , CDC2 Protein Kinase/genetics , CDC2-CDC28 Kinases , Carrier Proteins/genetics , Cdc20 Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cyclin A/genetics , Cyclin A/metabolism , Cyclin B1/genetics , Cyclin-Dependent Kinases/genetics , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nocodazole/metabolism , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/metabolism , Purines/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Roscovitine , Securin , Tubulin Modulators/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligase Complexes/genetics
4.
Biochem Soc Trans ; 38(Pt 1): 72-7, 2010 Feb.
Article En | MEDLINE | ID: mdl-20074038

Active cyclin B1-Cdk1 (cyclin-dependent kinase 1) keeps cells in mitosis, allowing time for spindle microtubules to capture the chromosomes and for incorrect chromosome-spindle attachments to be repaired. Meanwhile, securin, an inhibitor of separase, secures cohesion between sister chromatids, preventing anaphase onset. The spindle checkpoint is a signalling pathway emerging from improperly attached chromosomes that inhibits Cdc20, the mitotic activator of the APC/C (anaphase-promoting complex/cyclosome) ubiquitin ligase. Blocking Cdc20 stabilizes cyclin B1 and securin to delay mitotic exit and anaphase until all chromosomes reach bipolar spindle attachments. Cells entering mitosis in the absence of a functional spindle checkpoint degrade cyclin B1 and securin right after nuclear-envelope breakdown, in prometaphase. Interestingly, two APC/C substrates, cyclin A and Nek2A, are normally degraded at nuclear-envelope breakdown, even when the spindle checkpoint is active. This indicates that the APC/C is activated early in mitosis, whereas cyclin B1 and securin are protected as long as the spindle checkpoint inhibits Cdc20. Remarkably, destruction of cyclin A and Nek2A also depends on Cdc20. The paradox of Cdc20 being both active and inhibited in prometaphase could be explained if cyclin A and Nek2A are either exceptionally efficient Cdc20 substrates, or if they are equipped with 'stealth' mechanisms to effectively escape detection by the spindle checkpoint. In the present paper, we discuss recently emerging models for spindle-checkpoint-independent APC/C-Cdc20 activity, which might even have implications for cancer therapy.


Cyclin A/metabolism , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cell Cycle Proteins/metabolism , Cell Division/physiology , Fungal Proteins/metabolism , Genes, cdc , Humans , Signal Transduction/physiology
5.
J Cell Sci ; 123(Pt 3): 321-30, 2010 Feb 01.
Article En | MEDLINE | ID: mdl-20053638

Progression through mitosis and cytokinesis requires the sequential proteolysis of several cell-cycle regulators. This proteolysis is mediated by the ubiquitin-proteasome system, with the E3 ligase being the anaphase-promoting complex, also known as the cyclosome (APC/C). The APC/C is regulated by two activators, namely Cdc20 and Cdh1. The current view is that prior to anaphase, the APC/C is activated by Cdc20, but that following anaphase, APC/C switches to Cdh1-dependent activation. However, here we present an analysis of the kinetochore protein Cenp-F that is inconsistent with this notion. Although it has long been appreciated that Cenp-F is degraded sometime during or after mitosis, exactly when and how has not been clear. Here we show that degradation of Cenp-F initiates about six minutes after anaphase, and that this is dependent on a C-terminal KEN-box. Although these two observations are consistent with Cenp-F being a substrate of Cdh1-activated APC/C, Cenp-F is degraded normally in Cdh1-null cells. By contrast, RNAi-mediated repression of APC/C subunits or Cdc20 does inhibit Cenp-F degradation. These findings therefore suggest that the APC/C does not simply 'switch' upon anaphase onset; rather, our observations indicate that Cdc20 also contributes to post-anaphase activation of the APC/C. We also show that the post-anaphase, KEN-box-dependent degradation of Cenp-F requires it to be farnesylated, a post-translational modification usually linked to membrane association. Because so many of the behaviours of Cenp-F are farnesylation-dependent, we suggest that this modification plays a more global role in Cenp-F function.


Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Microfilament Proteins/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Antigens, CD , Cadherins/genetics , Cadherins/metabolism , Cdc20 Proteins , Cdh1 Proteins , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cells, Cultured , Humans , Kinetochores/metabolism , Mice , Microscopy , Prenylation , RNA Interference , Ubiquitin-Protein Ligase Complexes/metabolism
6.
PLoS One ; 4(9): e6936, 2009 Sep 07.
Article En | MEDLINE | ID: mdl-19738907

Cohesion between sister chromatids is essential for faithful chromosome segregation. In budding yeast, the acetyltransferase Eco1/Ctf7 establishes cohesion during DNA replication in S phase and in response to DNA double strand breaks in G2/M phase. In humans two Eco1 orthologs exist: ESCO1 and ESCO2. Both proteins are required for proper sister chromatid cohesion, but their exact function is unclear at present. Since ESCO2 has been identified as the gene defective in the rare autosomal recessive cohesinopathy Roberts syndrome (RBS), cells from RBS patients can be used to elucidate the role of ESCO2. We investigated for the first time RBS cells in comparison to isogenic controls that stably express V5- or GFP-tagged ESCO2. We show that the sister chromatid cohesion defect in the transfected cell lines is rescued and suggest that ESCO2 is regulated by proteasomal degradation in a cell cycle-dependent manner. In comparison to the corrected cells RBS cells were hypersensitive to the DNA-damaging agents mitomycin C, camptothecin and etoposide, while no particular sensitivity to UV, ionizing radiation, hydroxyurea or aphidicolin was found. The cohesion defect of RBS cells and their hypersensitivity to DNA-damaging agents were not corrected by a patient-derived ESCO2 acetyltransferase mutant (W539G), indicating that the acetyltransferase activity of ESCO2 is essential for its function. In contrast to a previous study on cells from patients with Cornelia de Lange syndrome, another cohesinopathy, RBS cells failed to exhibit excessive chromosome aberrations after irradiation in G2 phase of the cell cycle. Our results point at an S phase-specific role for ESCO2 in the maintenance of genome stability.


Acetyltransferases/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Fibroblasts/metabolism , Growth Disorders/diagnosis , Camptothecin/pharmacology , Cell Cycle Proteins/metabolism , Chromosome Aberrations , Chromosome Segregation , Congenital Abnormalities/diagnosis , Congenital Abnormalities/genetics , DNA Damage , Etoposide/pharmacology , Growth Disorders/genetics , Humans , Infant , Male , Mitomycin/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Sister Chromatid Exchange , Syndrome , Cohesins
7.
PLoS One ; 4(4): e5282, 2009.
Article En | MEDLINE | ID: mdl-19390576

Polo-like kinase-1 (Plk1) is activated before mitosis by Aurora A and its cofactor Bora. In mitosis, Bora is degraded in a manner dependent on Plk1 kinase activity and the E3 ubiquitin ligase SCF-betaTrCP. Here, we show that Plk1 is also required for the timely destruction of its activator Aurora A in late anaphase. It has been shown that Aurora A destruction is controlled by the auxiliary subunit Cdh1 of the Anaphase-Promoting Complex/Cyclosome (APC/C). Remarkably, we found that Plk1-depletion prevented the efficient dephosphorylation of Cdh1 during mitotic exit. Plk1 mediated its effect on Cdh1, at least in part, through direct phosphorylation of the human phosphatase Cdc14A, controlling the phosphorylation state of Cdh1. We conclude that Plk1 facilitates efficient Aurora A degradation through APC/C-Cdh1 activation after mitosis, with a potential role for hCdc14A.


Cadherins/metabolism , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase , Anaphase-Promoting Complex-Cyclosome , Aurora Kinases , Cell Line, Tumor , Flow Cytometry , Humans , Mitosis , Phosphoric Monoester Hydrolases/metabolism , Polo-Like Kinase 1
8.
Mol Cell ; 30(3): 290-302, 2008 May 09.
Article En | MEDLINE | ID: mdl-18471975

Successful mitosis requires the right protein be degraded at the right time. Central to this is the spindle checkpoint that prevents the destruction of securin and cyclin B1 when there are improperly attached chromosomes. The principal target of the checkpoint is Cdc20, which activates the anaphase-promoting complex/cyclosome (APC/C). A Drosophila Cdc20/fizzy mutant arrests in mitosis with high levels of cyclins A and B, but paradoxically the spindle checkpoint does not stabilize cyclin A. Here, we investigated this paradox and found that Cdc20 is rate limiting for cyclin A destruction. Indeed, Cdc20 binds efficiently to cyclin A before and in mitosis, and this complex has little associated Mad2. Furthermore, the cyclin A complex must bind to a Cks protein to be degraded independently of the checkpoint. Thus, we identify a crucial role for the Cks proteins in mitosis and one mechanism by which the APC/C can target substrates independently of the spindle checkpoint.


Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Cyclin A/metabolism , Cyclin-Dependent Kinases/metabolism , Spindle Apparatus/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , CDC2-CDC28 Kinases , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cdc20 Proteins , Cell Cycle Proteins/genetics , Cell Line , Cyclin A/genetics , Cyclin A2 , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Mitosis/physiology , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
PLoS Biol ; 5(5): e123, 2007 May.
Article En | MEDLINE | ID: mdl-17472438

Activation of cyclin B1-cyclin-dependent kinase 1 (Cdk1), triggered by a positive feedback loop at the end of G2, is the key event that initiates mitotic entry. In metaphase, anaphase-promoting complex/cyclosome-dependent destruction of cyclin B1 inactivates Cdk1 again, allowing mitotic exit and cell division. Several models describe Cdk1 activation kinetics in mitosis, but experimental data on how the activation proceeds in mitotic cells have largely been lacking. We use a novel approach to determine the temporal development of cyclin B1-Cdk1 activity in single cells. By quantifying both dephosphorylation of Cdk1 and phosphorylation of the Cdk1 target anaphase-promoting complex/cyclosome 3, we disclose how cyclin B1-Cdk1 continues to be activated after centrosome separation. Importantly, we discovered that cytoplasmic cyclin B1-Cdk1 activity can be maintained even when cyclin B1 translocates to the nucleus in prophase. These experimental data are fitted into a model describing cyclin B1-Cdk1 activation in human cells, revealing a striking resemblance to a bistable circuit. In line with the observed kinetics, cyclin B1-Cdk1 levels required to enter mitosis are lower than the amount of cyclin B1-Cdk1 needed for mitotic progression. We propose that gradually increasing cyclin B1-Cdk1 activity after centrosome separation is critical to coordinate mitotic progression.


CDC2 Protein Kinase/metabolism , Centrosome/physiology , Cyclin B/metabolism , Mitosis/physiology , Models, Biological , Anaphase-Promoting Complex-Cyclosome , Cyclin B1 , Enzyme Activation/physiology , HeLa Cells , Humans , Phosphorylation , Ubiquitin-Protein Ligase Complexes/metabolism
...