Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
NPJ Breast Cancer ; 10(1): 19, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38438376

ABSTRACT

CDK4/6 inhibitors are effective at treating advanced HR+ /HER2- breast cancer, however biomarkers that can predict response are urgently needed. We demonstrate here that previous large-scale screens designed to identify which tumour types or genotypes are most sensitive to CDK4/6 inhibitors have misrepresented the responsive cell lines because of a reliance on metabolic proliferation assays. CDK4/6-inhibited cells arrest in G1 but continue to grow in size, thereby producing more mitochondria. We show that this growth obscures the arrest using ATP-based proliferation assays but not if DNA-based assays are used instead. Furthermore, lymphoma lines, previously identified as the most sensitive, simply appear to respond the best using ATP-based assays because they fail to overgrow during the G1 arrest. Similarly, the CDK4/6 inhibitor abemaciclib appears to inhibit proliferation better than palbociclib because it also restricts cellular overgrowth through off-target effects. DepMap analysis of screening data using reliable assay types, demonstrates that palbociclib-sensitive cell types are also sensitive to Cyclin D1, CDK4 and CDK6 knockout/knockdown, whereas the palbociclib-resistant lines are sensitive to Cyclin E1, CDK2 and SKP2 knockout/knockdown. Potential biomarkers of palbociclib-sensitive cells are increased expression of CCND1 and RB1, and reduced expression of CCNE1 and CDKN2A. Probing DepMap with similar data from metabolic assays fails to reveal these associations. Together, this demonstrates why CDK4/6 inhibitors, and any other anti-cancer drugs that arrest the cell cycle but permit continued cell growth, must now be re-screened against a wide-range of cell types using an appropriate proliferation assay. This would help to better inform clinical trials and to identify much needed biomarkers of response.

2.
Mol Cell ; 83(22): 4047-4061.e6, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977117

ABSTRACT

CDK4/6 inhibitors are remarkable anti-cancer drugs that can arrest tumor cells in G1 and induce their senescence while causing only relatively mild toxicities in healthy tissues. How they achieve this mechanistically is unclear. We show here that tumor cells are specifically vulnerable to CDK4/6 inhibition because during the G1 arrest, oncogenic signals drive toxic cell overgrowth. This overgrowth causes permanent cell cycle withdrawal by either preventing progression from G1 or inducing genotoxic damage during the subsequent S-phase and mitosis. Inhibiting or reverting oncogenic signals that converge onto mTOR can rescue this excessive growth, DNA damage, and cell cycle exit in cancer cells. Conversely, inducing oncogenic signals in non-transformed cells can drive these toxic phenotypes and sensitize the cells to CDK4/6 inhibition. Together, this demonstrates that cell cycle arrest and oncogenic cell growth is a synthetic lethal combination that is exploited by CDK4/6 inhibitors to induce tumor-specific toxicity.


Subject(s)
Antineoplastic Agents , Neoplasms , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase Cell Cycle Checkpoints , Tumor Suppressor Protein p53/genetics , Cell Cycle , Cell Cycle Proteins/metabolism , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics
3.
Mol Cell ; 83(22): 4062-4077.e5, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977118

ABSTRACT

Abnormal increases in cell size are associated with senescence and cell cycle exit. The mechanisms by which overgrowth primes cells to withdraw from the cell cycle remain unknown. We address this question using CDK4/6 inhibitors, which arrest cells in G0/G1 and are licensed to treat advanced HR+/HER2- breast cancer. We demonstrate that CDK4/6-inhibited cells overgrow during G0/G1, causing p38/p53/p21-dependent cell cycle withdrawal. Cell cycle withdrawal is triggered by biphasic p21 induction. The first p21 wave is caused by osmotic stress, leading to p38- and size-dependent accumulation of p21. CDK4/6 inhibitor washout results in some cells entering S-phase. Overgrown cells experience replication stress, resulting in a second p21 wave that promotes cell cycle withdrawal from G2 or the subsequent G1. We propose that the levels of p21 integrate signals from overgrowth-triggered stresses to determine cell fate. This model explains how hypertrophy can drive senescence and why CDK4/6 inhibitors have long-lasting effects in patients.


Subject(s)
Tumor Suppressor Protein p53 , Humans , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cell Cycle , Cell Division , Tumor Suppressor Protein p53/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism
4.
EMBO J ; 42(20): e112630, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37712330

ABSTRACT

Two major mechanisms safeguard genome stability during mitosis: the mitotic checkpoint delays mitosis until all chromosomes have attached to microtubules, and the kinetochore-microtubule error-correction pathway keeps this attachment process free from errors. We demonstrate here that the optimal strength and dynamics of these processes are set by a kinase-phosphatase pair (PLK1-PP2A) that engage in negative feedback from adjacent phospho-binding motifs on the BUB complex. Uncoupling this feedback to skew the balance towards PLK1 produces a strong checkpoint, hypostable microtubule attachments and mitotic delays. Conversely, skewing the balance towards PP2A causes a weak checkpoint, hyperstable microtubule attachments and chromosome segregation errors. These phenotypes are associated with altered BUB complex recruitment to KNL1-MELT motifs, implicating PLK1-PP2A in controlling auto-amplification of MELT phosphorylation. In support, KNL1-BUB disassembly becomes contingent on PLK1 inhibition when KNL1 is engineered to contain excess MELT motifs. This elevates BUB-PLK1/PP2A complex levels on metaphase kinetochores, stabilises kinetochore-microtubule attachments, induces chromosome segregation defects and prevents KNL1-BUB disassembly at anaphase. Together, these data demonstrate how a bifunctional PLK1/PP2A module has evolved together with the MELT motifs to optimise BUB complex dynamics and ensure accurate chromosome segregation.


Subject(s)
Kinetochores , M Phase Cell Cycle Checkpoints , Humans , Kinetochores/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Segregation , Phosphorylation , Microtubules/metabolism , Mitosis , HeLa Cells
5.
EMBO J ; 41(6): e108599, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35037284

ABSTRACT

CDK4/6 inhibitors arrest the cell cycle in G1-phase. They are approved to treat breast cancer and are also undergoing clinical trials against a range of other tumour types. To facilitate these efforts, it is important to understand why a cytostatic arrest in G1 causes long-lasting effects on tumour growth. Here, we demonstrate that a prolonged G1 arrest following CDK4/6 inhibition downregulates replisome components and impairs origin licencing. Upon release from that arrest, many cells fail to complete DNA replication and exit the cell cycle in a p53-dependent manner. If cells fail to withdraw from the cell cycle following DNA replication problems, they enter mitosis and missegregate chromosomes causing excessive DNA damage, which further limits their proliferative potential. These effects are observed in a range of tumour types, including breast cancer, implying that genotoxic stress is a common outcome of CDK4/6 inhibition. This unanticipated ability of CDK4/6 inhibitors to induce DNA damage now provides a rationale to better predict responsive tumour types and effective combination therapies, as demonstrated by the fact that CDK4/6 inhibition induces sensitivity to chemotherapeutics that also cause replication stress.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle , Cell Division , Cell Line, Tumor , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Female , G1 Phase , Humans
6.
J Cell Biol ; 219(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-33125045

ABSTRACT

Local phosphatase regulation is needed at kinetochores to silence the mitotic checkpoint (a.k.a. spindle assembly checkpoint [SAC]). A key event in this regard is the dephosphorylation of MELT repeats on KNL1, which removes SAC proteins from the kinetochore, including the BUB complex. We show here that PP1 and PP2A-B56 phosphatases are primarily required to remove Polo-like kinase 1 (PLK1) from the BUB complex, which can otherwise maintain MELT phosphorylation in an autocatalytic manner. This appears to be their principal role in the SAC because both phosphatases become redundant if PLK1 is inhibited or BUB-PLK1 interaction is prevented. Surprisingly, MELT dephosphorylation can occur normally under these conditions even when the levels or activities of PP1 and PP2A are strongly inhibited at kinetochores. Therefore, these data imply that kinetochore phosphatase regulation is critical for the SAC, but primarily to restrain and extinguish autonomous PLK1 activity. This is likely a conserved feature of the metazoan SAC, since the relevant PLK1 and PP2A-B56 binding motifs have coevolved in the same region on MADBUB homologues.


Subject(s)
Cell Cycle Proteins/metabolism , Kinetochores/enzymology , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Spindle Apparatus/enzymology , Cell Cycle Proteins/genetics , HeLa Cells , Humans , Phosphorylation/genetics , Protein Phosphatase 1/genetics , Protein Phosphatase 2/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Spindle Apparatus/genetics , Polo-Like Kinase 1
7.
EMBO J ; 39(12): e103180, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32202322

ABSTRACT

Cyclin B:CDK1 is the master kinase regulator of mitosis. We show here that, in addition to its kinase functions, mammalian Cyclin B also scaffolds a localised signalling pathway to help preserve genome stability. Cyclin B1 localises to an expanded region of the outer kinetochore, known as the corona, where it scaffolds the spindle assembly checkpoint (SAC) machinery by binding directly to MAD1. In vitro reconstitutions map the key binding interface to a few acidic residues in the N-terminal region of MAD1, and point mutations in this sequence abolish MAD1 corona localisation and weaken the SAC. Therefore, Cyclin B1 is the long-sought-after scaffold that links MAD1 to the corona, and this specific pool of MAD1 is needed to generate a robust SAC response. Robustness arises because Cyclin B1:MAD1 localisation loses dependence on MPS1 kinase after the corona has been established, ensuring that corona-localised MAD1 can still be phosphorylated when MPS1 activity is low. Therefore, this study explains how corona-MAD1 generates a robust SAC signal, and it reveals a scaffolding role for the key mitotic kinase, Cyclin B1:CDK1, which ultimately helps to inhibit its own degradation.


Subject(s)
Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Cyclin B1/metabolism , Kinetochores/metabolism , Mitosis , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cyclin B1/genetics , HeLa Cells , Humans , Point Mutation , Protein Domains
8.
Cell Rep ; 28(8): 2206-2219.e8, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433993

ABSTRACT

PP1 and PP2A-B56 are major serine/threonine phosphatase families that achieve specificity by colocalizing with substrates. At the kinetochore, however, both phosphatases localize to an almost identical molecular space and yet they still manage to regulate unique pathways and processes. By switching or modulating the positions of PP1/PP2A-B56 at kinetochores, we show that their unique downstream effects are not due to either the identity of the phosphatase or its precise location. Instead, these phosphatases signal differently because their kinetochore recruitment can be either inhibited (PP1) or enhanced (PP2A) by phosphorylation inputs. Mathematical modeling explains how these inverse phospho-dependencies elicit unique forms of cross-regulation and feedback, which allows otherwise indistinguishable phosphatases to produce distinct network behaviors and control different mitotic processes. Furthermore, our genome-wide analysis suggests that these major phosphatase families may have evolved to respond to phosphorylation inputs in opposite ways because many other PP1 and PP2A-B56-binding motifs are also phospho-regulated.


Subject(s)
Kinetochores/metabolism , Protein Phosphatase 2/metabolism , Receptors, Neuropeptide Y/metabolism , Amino Acid Motifs , Amino Acid Sequence , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Models, Biological , Phenotype , Phosphorylation , Protein Phosphatase 2/chemistry , Receptors, Neuropeptide Y/chemistry , Signal Transduction
9.
Elife ; 82019 03 04.
Article in English | MEDLINE | ID: mdl-30829571

ABSTRACT

PP2A-B56 is a serine/threonine phosphatase complex that regulates several major mitotic processes, including sister chromatid cohesion, kinetochore-microtubule attachment and the spindle assembly checkpoint. We show here that these key functions are divided between different B56 isoforms that localise to either the centromere or kinetochore. The centromeric isoforms rely on a specific interaction with Sgo2, whereas the kinetochore isoforms bind preferentially to BubR1 and other proteins containing an LxxIxE motif. In addition to these selective binding partners, Sgo1 helps to anchor PP2A-B56 at both locations: it collaborates with BubR1 to maintain B56 at the kinetochore and it helps to preserve the Sgo2/B56 complex at the centromere. A series of chimaeras were generated to map the critical region in B56 down to a small C-terminal loop that regulates the key interactions and defines B56 localisation. Together, this study describes how different PP2A-B56 complexes utilise isoform-specific interactions to control distinct processes during mitosis.


Subject(s)
Centromere/enzymology , Kinetochores/enzymology , Mitosis , Multiprotein Complexes/metabolism , Protein Isoforms/metabolism , Protein Phosphatase 2/metabolism , Cell Cycle Proteins/metabolism , HeLa Cells , Humans , Protein Binding , Protein Multimerization , Protein Serine-Threonine Kinases/metabolism
10.
Front Cell Dev Biol ; 6: 62, 2018.
Article in English | MEDLINE | ID: mdl-29971233

ABSTRACT

Multiple kinases and phosphatases act on the kinetochore to control chromosome segregation: Aurora B, Mps1, Bub1, Plk1, Cdk1, PP1, and PP2A-B56, have all been shown to regulate both kinetochore-microtubule attachments and the spindle assembly checkpoint. Given that so many kinases and phosphatases converge onto two key mitotic processes, it is perhaps not surprising to learn that they are, quite literally, entangled in cross-talk. Inhibition of any one of these enzymes produces secondary effects on all the others, which results in a complicated picture that is very difficult to interpret. This review aims to clarify this picture by first collating the direct effects of each enzyme into one overarching schematic of regulation at the Knl1/Mis12/Ndc80 (KMN) network (a major signaling hub at the outer kinetochore). This schematic will then be used to discuss the implications of the cross-talk that connects these enzymes; both in terms of why it may be needed to produce the right type of kinetochore signals and why it nevertheless complicates our interpretations about which enzymes control what processes. Finally, some general experimental approaches will be discussed that could help to characterize kinetochore signaling by dissociating the direct from indirect effect of kinase or phosphatase inhibition in vivo. Together, this review should provide a framework to help understand how a network of kinases and phosphatases cooperate to regulate two key mitotic processes.

11.
Sci Rep ; 8(1): 7898, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29785044

ABSTRACT

SiR-Hoechst (SiR-DNA) is a far-red fluorescent DNA probe being used widely for time-lapse imaging of living cells that is reported to be minimally toxic at concentrations as high as 10-25 µM. However, measuring nuclear import of Cyclin B1, inhibition of mitotic entry, and the induction of γH2AX foci in cultured human cells reveals that SiR-Hoechst induces DNA damage responses and G2 arrest at concentrations well below 1 µM. SiR-Hoechst is useful for live cell imaging, but it should be used with caution and at the lowest practicable concentration.


Subject(s)
Cell Cycle , DNA Damage , DNA/chemistry , Fluorescent Dyes/toxicity , Osteosarcoma/pathology , Retinal Pigment Epithelium/pathology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cells, Cultured , Cyclin B1/genetics , Cyclin B1/metabolism , Fluorescent Dyes/chemistry , Humans , Microscopy, Fluorescence , Mitosis , Molecular Imaging , Osteosarcoma/genetics , Retinal Pigment Epithelium/metabolism , Staining and Labeling
12.
Cell Rep ; 23(3): 852-865, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29669289

ABSTRACT

Faithful chromosome segregation during mitosis depends on the spindle assembly checkpoint (SAC), which delays progression through mitosis until every chromosome has stably attached to spindle microtubules via the kinetochore. We show here that the deubiquitinase USP9X strengthens the SAC by antagonizing the turnover of the mitotic checkpoint complex produced at unattached kinetochores. USP9X thereby opposes activation of anaphase-promoting complex/cyclosome (APC/C) and specifically inhibits the mitotic degradation of SAC-controlled APC/C substrates. We demonstrate that depletion or loss of USP9X reduces the effectiveness of the SAC, elevates chromosome segregation defects, and enhances chromosomal instability (CIN). These findings provide a rationale to explain why loss of USP9X could be either pro- or anti-tumorigenic depending on the existing level of CIN.


Subject(s)
Mitosis , Spindle Apparatus/metabolism , Ubiquitin Thiolesterase/metabolism , Anaphase-Promoting Complex-Cyclosome/metabolism , Apc11 Subunit, Anaphase-Promoting Complex-Cyclosome/antagonists & inhibitors , Apc11 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc11 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Cdc20 Proteins/metabolism , Chromosomal Instability , Chromosome Segregation , Cyclin B/metabolism , HeLa Cells , Humans , Karyotype , Kinesins/metabolism , Kinetochores/metabolism , Mitosis/drug effects , NIMA-Related Kinases/metabolism , Nocodazole/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics
13.
Dev Cell ; 44(6): 659-663, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29587141

ABSTRACT

Protein phosphorylation is a dynamic post-translational modification critical for biological responses. At the level of individual molecules, phosphorylation dynamics can have important functional implications, but this information is rarely quantified. We discuss how rapid phosphorylation-dephosphorylation cycles could underlie important signaling properties, including the ability to rapidly bind and release proteins.


Subject(s)
Models, Biological , Protein Processing, Post-Translational , Proteins/metabolism , Animals , Humans , Phosphorylation , Signal Transduction
14.
Trends Cell Biol ; 28(1): 6-21, 2018 01.
Article in English | MEDLINE | ID: mdl-29089159

ABSTRACT

Kinases and phosphatases work antagonistically to control the behaviour of individual substrate molecules. This can be incorrectly extrapolated to imply that they also work antagonistically on the signals or processes that these molecules control. In fact, in many situations kinases and phosphatases work together to positively drive signal responses. We explain how this 'cooperativity' is critical for setting the amplitude, localisation, timing, and shape of phosphorylation signals. We use mitosis to illustrate why these properties are important for controlling mitotic entry, sister chromatid cohesion, kinetochore-microtubule attachments, the spindle assembly checkpoint, mitotic spindle elongation, and mitotic exit. These examples provide a rationale to explain how complex signalling behaviour could rely on similar types of integration within many other biological processes.


Subject(s)
Cell Cycle Proteins/physiology , Mitosis/physiology , Phosphoric Monoester Hydrolases/physiology , Protein Kinases/physiology , Animals , Cell Cycle Proteins/genetics , Chromosome Segregation/genetics , Humans , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Kinases/genetics
15.
Sci Rep ; 7(1): 16115, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170437

ABSTRACT

Polo-like kinase-1 (PLK1) plays a major role in driving mitotic events, including centrosome disjunction and separation, and is frequently over-expressed in human cancers. PLK1 inhibition is a promising therapeutic strategy and works by arresting cells in mitosis due to monopolar spindles. The p53 tumour suppressor protein is a short-lived transcription factor that can inhibit the growth, or stimulate the death, of developing cancer cells. Curiously, although p53 normally acts in an anti-cancer capacity, it can offer significant protection against inhibitors of PLK1, but the events underpinning this effect are not known. Here, we show that functional p53 reduces the sensitivity to PLK1 inhibitors by permitting centrosome separation to occur, allowing cells to traverse mitosis and re-enter cycle with a normal complement of 2N chromosomes. Protection entails the activation of p53 through the DNA damage-response enzymes, ATM and ATR, and requires the phosphorylation of p53 at the key regulatory site, Ser15. These data highlight a previously unrecognised link between p53, PLK1 and centrosome separation that has therapeutic implications for the use of PLK1 inhibitors in the clinic.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Benzimidazoles/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Survival/drug effects , Centrosome/drug effects , Fluorescent Antibody Technique , Gene Silencing , HCT116 Cells , Humans , Mitosis/drug effects , Mitosis/genetics , Mitosis/physiology , Morpholines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrazines/pharmacology , Pyrones/pharmacology , Sulfones/pharmacology , Thiophenes/pharmacology , Tumor Suppressor Protein p53/genetics , Polo-Like Kinase 1
16.
Methods Mol Biol ; 1413: 333-47, 2016.
Article in English | MEDLINE | ID: mdl-27193859

ABSTRACT

Mitotic kinetochores are signaling network hubs that regulate chromosome movements, attachment error-correction, and the spindle assembly checkpoint. Key switches in these networks are kinases and phosphatases that enable rapid responses to changing conditions. Describing the mechanisms and dynamics of their localized activation and deactivation is therefore instrumental for understanding the spatiotemporal control of chromosome segregation.


Subject(s)
Kinetochores/metabolism , Phosphotransferases/metabolism , Enzyme Activation , Mitosis , Molecular Imaging/methods , Phosphorylation , Phosphotransferases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Transport , Signal Transduction , Spindle Apparatus/metabolism , Time-Lapse Imaging
17.
J Cell Sci ; 128(22): 4035-8, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26574504

ABSTRACT

The Company of Biologists Workshop 'Getting Into and Out of Mitosis' was held 10-13 May 2015 at Wiston House in West Sussex, UK. The workshop brought together researchers from wide-ranging disciplines and provided a forum to discuss their latest work on the control of cell division from mitotic entry to exit. This report highlights the main topics and summarises the discussion around the key themes and questions that emerged from the meeting.


Subject(s)
Mitosis/physiology
18.
Cell Cycle ; 14(6): 795-6, 2015.
Article in English | MEDLINE | ID: mdl-25714119

ABSTRACT

Kinases and phosphatases, two sides of the same coin; are they opposing forces that switch signals on and off or enzymes that work together to give the right type of response at the right time? It depends on how close you stand when you view the big picture. Up close and detailed, and you'll see individual phosphorylation sites as binary switches - lights being toggled on/off by antagonistic forces. Take a step back and multiple copies of the same light are being toggled, perhaps leading to a range of intensities, or a flickering pattern, lights flashing in unison or at random. It depends what the signal requires. Stand even further back, let the story unfold, and you'll see a dazzling multicolour array of different lights. A coordinated sequence of color that appears to burst into life at different times in different places, with a pace that is both frantic and serene. This is a vision of mitosis and what a true spectacle it is.


Subject(s)
Feedback, Physiological , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints/genetics , Spindle Apparatus/genetics , Humans
19.
Cell Cycle ; 13(17): 2733-43, 2014.
Article in English | MEDLINE | ID: mdl-25486360

ABSTRACT

Upon DNA damage, cell cycle progression is temporally blocked to avoid propagation of mutations. While transformed cells largely maintain the competence to recover from a cell cycle arrest, untransformed cells past the G1/S transition lose mitotic inducers, and thus the ability to resume cell division. This permanent cell cycle exit depends on p21, p53, and APC/C(Cdh1). However, when and how permanent cell cycle exit occurs remains unclear. Here, we have investigated the cell cycle response to DNA damage in single cells that express Cyclin B1 fused to eYFP at the endogenous locus. We find that upon DNA damage Cyclin B1-eYFP continues to accumulate up to a threshold level, which is reached only in G2 phase. Above this threshold, a p21 and p53-dependent nuclear translocation required for APC/C(Cdh1)-mediated Cyclin B1-eYFP degradation is initiated. Thus, cell cycle exit is decoupled from activation of the DNA damage response in a manner that correlates to Cyclin B1 levels, suggesting that G2 activities directly feed into the decision for cell cycle exit. Once Cyclin B1-eYFP nuclear translocation occurs, checkpoint inhibition can no longer promote mitotic entry or re-expression of mitotic inducers, suggesting that nuclear translocation of Cyclin B1 marks the restriction point for permanent cell cycle exit in G2 phase.


Subject(s)
Cell Cycle Checkpoints , Cell Nucleus/metabolism , Cyclin B1/metabolism , G2 Phase , Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Gene Targeting , Humans , Protein Transport , Proteolysis , Tumor Suppressor Protein p53/metabolism
20.
Nat Cell Biol ; 16(12): 1257-64, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25402682

ABSTRACT

Kinetochores are specialized multi-protein complexes that play a crucial role in maintaining genome stability. They bridge attachments between chromosomes and microtubules during mitosis and they activate the spindle assembly checkpoint (SAC) to arrest division until all chromosomes are attached. Kinetochores are able to efficiently integrate these two processes because they can rapidly respond to changes in microtubule occupancy by switching localized SAC signalling ON or OFF. We show that this responsiveness arises because the SAC primes kinetochore phosphatases to induce negative feedback and silence its own signal. Active SAC signalling recruits PP2A-B56 to kinetochores where it antagonizes Aurora B to promote PP1 recruitment. PP1 in turn silences the SAC and delocalizes PP2A-B56. Preventing or bypassing key regulatory steps demonstrates that this spatiotemporal control of phosphatase feedback underlies rapid signal switching at the kinetochore by: allowing the SAC to quickly transition to the ON state in the absence of antagonizing phosphatase activity; and ensuring phosphatases are then primed to rapidly switch the SAC signal OFF when kinetochore kinase activities are diminished by force-producing microtubule attachments.


Subject(s)
Feedback, Physiological , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints/genetics , Spindle Apparatus/genetics , Amino Acid Motifs/genetics , Aurora Kinase B/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Chromosome Segregation , HeLa Cells , Humans , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nocodazole/pharmacology , Phosphorylation , Protein Binding , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction , Smad2 Protein/genetics , Tubulin Modulators/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...