Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.015
Filter
Add more filters

Publication year range
1.
Cell ; 181(3): 702-715.e20, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32315619

ABSTRACT

Protein phosphatase 2A (PP2A) enzymes can suppress tumors, but they are often inactivated in human cancers overexpressing inhibitory proteins. Here, we identify a class of small-molecule iHAPs (improved heterocyclic activators of PP2A) that kill leukemia cells by allosterically assembling a specific heterotrimeric PP2A holoenzyme consisting of PPP2R1A (scaffold), PPP2R5E (B56ε, regulatory), and PPP2CA (catalytic) subunits. One compound, iHAP1, activates this complex but does not inhibit dopamine receptor D2, a mediator of neurologic toxicity induced by perphenazine and related neuroleptics. The PP2A complex activated by iHAP1 dephosphorylates the MYBL2 transcription factor on Ser241, causing irreversible arrest of leukemia and other cancer cells in prometaphase. In contrast, SMAPs, a separate class of compounds, activate PP2A holoenzymes containing a different regulatory subunit, do not dephosphorylate MYBL2, and arrest tumor cells in G1 phase. Our findings demonstrate that small molecules can serve as allosteric switches to activate distinct PP2A complexes with unique substrates.


Subject(s)
Protein Phosphatase 2/metabolism , Apoptosis , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Enzyme Activators/metabolism , G1 Phase , Humans , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , Phenothiazines/pharmacology , Phosphorylation , Protein Phosphatase 2/physiology , Protein Subunits/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism
2.
Cell ; 173(6): 1481-1494.e13, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29706543

ABSTRACT

Global profiling of protein expression through the cell cycle has revealed subsets of periodically expressed proteins. However, expression levels alone only give a partial view of the biochemical processes determining cellular events. Using a proteome-wide implementation of the cellular thermal shift assay (CETSA) to study specific cell-cycle phases, we uncover changes of interaction states for more than 750 proteins during the cell cycle. Notably, many protein complexes are modulated in specific cell-cycle phases, reflecting their roles in processes such as DNA replication, chromatin remodeling, transcription, translation, and disintegration of the nuclear envelope. Surprisingly, only small differences in the interaction states were seen between the G1 and the G2 phase, suggesting similar hardwiring of biochemical processes in these two phases. The present work reveals novel molecular details of the cell cycle and establishes proteome-wide CETSA as a new strategy to study modulation of protein-interaction states in intact cells.


Subject(s)
Cell Cycle , Protein Interaction Mapping , Cell Division , Chromatin/chemistry , Cluster Analysis , DNA Replication , G1 Phase , G2 Phase , Humans , K562 Cells , Nuclear Envelope , Proteome , Proteomics/methods
3.
Mol Cell ; 83(22): 3946-3947, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977113

ABSTRACT

In this issue of Molecular Cell, Crozier et al.,1 Foy et al.,2 Manohar et al.,3 and Wilson et al.4 show how excessive cell growth caused by a temporary G1 arrest leads to permanent cell cycle exit at different stages of the cell cycle.


Subject(s)
Cellular Senescence , Cell Cycle , Cell Division , G1 Phase , Cell Proliferation
4.
Nature ; 631(8020): 424-431, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926571

ABSTRACT

Tissue repair, immune defence and cancer progression rely on a vital cellular decision between quiescence and proliferation1,2. Mammalian cells proliferate by triggering a positive feedback mechanism3,4. The transcription factor E2F activates cyclin-dependent kinase 2 (CDK2), which in turn phosphorylates and inactivates the E2F inhibitor protein retinoblastoma (Rb). This action further increases E2F activity to express genes needed for proliferation. Given that positive feedback can inadvertently amplify small signals, understanding how cells keep this positive feedback in check remains a puzzle. Here we measured E2F and CDK2 signal changes in single cells and found that the positive feedback mechanism engages only late in G1 phase. Cells spend variable and often extended times in a reversible state of intermediate E2F activity before committing to proliferate. This intermediate E2F activity is proportional to the amount of phosphorylation of a conserved T373 residue in Rb that is mediated by CDK2 or CDK4/CDK6. Such T373-phosphorylated Rb remains bound on chromatin but dissociates from it once Rb is hyperphosphorylated at many sites, which fully activates E2F. The preferential initial phosphorylation of T373 can be explained by its relatively slower rate of dephosphorylation. Together, our study identifies a primed state of intermediate E2F activation whereby cells sense external and internal signals and decide whether to reverse and exit to quiescence or trigger the positive feedback mechanism that initiates cell proliferation.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase 2 , E2F Transcription Factors , Retinoblastoma Protein , Phosphorylation , Cyclin-Dependent Kinase 2/metabolism , Retinoblastoma Protein/metabolism , E2F Transcription Factors/metabolism , Humans , Animals , Mice , Single-Cell Analysis , Chromatin/metabolism , G1 Phase , Feedback, Physiological , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cell Line
5.
Nature ; 623(7985): 183-192, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37853125

ABSTRACT

The DNA damage response is essential to safeguard genome integrity. Although the contribution of chromatin in DNA repair has been investigated1,2, the contribution of chromosome folding to these processes remains unclear3. Here we report that, after the production of double-stranded breaks (DSBs) in mammalian cells, ATM drives the formation of a new chromatin compartment (D compartment) through the clustering of damaged topologically associating domains, decorated with γH2AX and 53BP1. This compartment forms by a mechanism that is consistent with polymer-polymer phase separation rather than liquid-liquid phase separation. The D compartment arises mostly in G1 phase, is independent of cohesin and is enhanced after pharmacological inhibition of DNA-dependent protein kinase (DNA-PK) or R-loop accumulation. Importantly, R-loop-enriched DNA-damage-responsive genes physically localize to the D compartment, and this contributes to their optimal activation, providing a function for DSB clustering in the DNA damage response. However, DSB-induced chromosome reorganization comes at the expense of an increased rate of translocations, also observed in cancer genomes. Overall, we characterize how DSB-induced compartmentalization orchestrates the DNA damage response and highlight the critical impact of chromosome architecture in genomic instability.


Subject(s)
Cell Compartmentation , Chromatin , DNA Damage , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Chromatin/genetics , Chromatin/metabolism , DNA Breaks, Double-Stranded , DNA Repair , DNA-Activated Protein Kinase/metabolism , G1 Phase , Histones/metabolism , Neoplasms/genetics , R-Loop Structures , Tumor Suppressor p53-Binding Protein 1/metabolism
6.
Nature ; 619(7969): 363-370, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37407814

ABSTRACT

In mammalian cells, the decision to proliferate is thought to be irreversibly made at the restriction point of the cell cycle1,2, when mitogen signalling engages a positive feedback loop between cyclin A2/cyclin-dependent kinase 2 (CDK2) and the retinoblastoma protein3-5. Contrary to this textbook model, here we show that the decision to proliferate is actually fully reversible. Instead, we find that all cycling cells will exit the cell cycle in the absence of mitogens unless they make it to mitosis and divide first. This temporal competition between two fates, mitosis and cell cycle exit, arises because cyclin A2/CDK2 activity depends upon CDK4/6 activity throughout the cell cycle, not just in G1 phase. Without mitogens, mitosis is only observed when the half-life of cyclin A2 protein is long enough to sustain CDK2 activity throughout G2/M. Thus, cells are dependent on mitogens and CDK4/6 activity to maintain CDK2 activity and retinoblastoma protein phosphorylation throughout interphase. Consequently, even a 2-h delay in a cell's progression towards mitosis can induce cell cycle exit if mitogen signalling is lost. Our results uncover the molecular mechanism underlying the restriction point phenomenon, reveal an unexpected role for CDK4/6 activity in S and G2 phases and explain the behaviour of all cells following loss of mitogen signalling.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , G2 Phase , S Phase , Animals , Cyclin A2/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/deficiency , Cyclin-Dependent Kinase 4/metabolism , Mitogens/deficiency , Mitogens/metabolism , Mitosis , Phosphorylation , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Cyclin-Dependent Kinase 6/deficiency , Cyclin-Dependent Kinase 6/metabolism , G1 Phase
7.
Mol Cell ; 81(24): 5007-5024.e9, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34767771

ABSTRACT

As cells enter mitosis, chromatin compacts to facilitate chromosome segregation yet remains transcribed. Transcription supercoils DNA to levels that can impede further progression of RNA polymerase II (RNAPII) unless it is removed by DNA topoisomerase 1 (TOP1). Using ChIP-seq on mitotic cells, we found that TOP1 is required for RNAPII translocation along genes. The stimulation of TOP1 activity by RNAPII during elongation allowed RNAPII clearance from genes in prometaphase and enabled chromosomal segregation. Disruption of the TOP1-RNAPII interaction impaired RNAPII spiking at promoters and triggered defects in the post-mitotic transcription program. This program includes factors necessary for cell growth, and cells with impaired TOP1-RNAPII interaction are more sensitive to inhibitors of mTOR signaling. We conclude that TOP1 is necessary for assisting transcription during mitosis with consequences for growth and gene expression long after mitosis is completed. In this sense, TOP1 ensures that cellular memory is preserved in subsequent generations.


Subject(s)
Cell Proliferation , Chromatin Assembly and Disassembly , Colorectal Neoplasms/enzymology , DNA Topoisomerases, Type I/metabolism , G1 Phase , Mitosis , RNA Polymerase II/metabolism , Transcription, Genetic , Cell Proliferation/drug effects , Chromatin Immunoprecipitation Sequencing , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Topoisomerases, Type I/genetics , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , MTOR Inhibitors/pharmacology , Mitosis/drug effects , RNA Polymerase II/genetics
8.
Mol Cell ; 81(12): 2596-2610.e7, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33961796

ABSTRACT

p53-binding protein 1 (53BP1) regulates both the DNA damage response and p53 signaling. Although 53BP1's function is well established in DNA double-strand break repair, how its role in p53 signaling is modulated remains poorly understood. Here, we identify the scaffolding protein AHNAK as a G1 phase-enriched interactor of 53BP1. We demonstrate that AHNAK binds to the 53BP1 oligomerization domain and controls its multimerization potential. Loss of AHNAK results in hyper-accumulation of 53BP1 on chromatin and enhanced phase separation, culminating in an elevated p53 response, compromising cell survival in cancer cells but leading to senescence in non-transformed cells. Cancer transcriptome analyses indicate that AHNAK-53BP1 cooperation contributes to the suppression of p53 target gene networks in tumors and that loss of AHNAK sensitizes cells to combinatorial cancer treatments. These findings highlight AHNAK as a rheostat of 53BP1 function, which surveys cell proliferation by preventing an excessive p53 response.


Subject(s)
Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Cell Line, Tumor , Chromatin/metabolism , DNA/genetics , DNA Breaks, Double-Stranded , DNA Repair , G1 Phase/physiology , Histones/metabolism , Humans , MCF-7 Cells , Membrane Proteins/genetics , Membrane Proteins/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/physiology
9.
Mol Cell ; 81(9): 1951-1969.e6, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33761311

ABSTRACT

The initiation of DNA replication involves cell cycle-dependent assembly and disassembly of protein complexes, including the origin recognition complex (ORC) and CDC6 AAA+ ATPases. We report that multiple short linear protein motifs (SLiMs) within intrinsically disordered regions (IDRs) in ORC1 and CDC6 mediate cyclin-CDK-dependent and independent protein-protein interactions, conditional on the cell cycle phase. A domain within the ORC1 IDR is required for interaction between the ORC1 and CDC6 AAA+ domains in G1, whereas the same domain prevents CDC6-ORC1 interaction during mitosis. Then, during late G1, this domain facilitates ORC1 destruction by a SKP2-cyclin A-CDK2-dependent mechanism. During G1, the CDC6 Cy motif cooperates with cyclin E-CDK2 to promote ORC1-CDC6 interactions. The CDC6 IDR regulates self-interaction by ORC1, thereby controlling ORC1 protein levels. Protein phosphatase 1 binds directly to a SLiM in the ORC1 IDR, causing ORC1 de-phosphorylation upon mitotic exit, increasing ORC1 protein, and promoting pre-RC assembly.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , DNA Replication , Intrinsically Disordered Proteins/metabolism , Mitosis , Nuclear Proteins/metabolism , Origin Recognition Complex/metabolism , AAA Domain , ATPases Associated with Diverse Cellular Activities/genetics , Cell Cycle Proteins/genetics , Cell Nucleus/genetics , Cyclin A/genetics , Cyclin A/metabolism , Cyclin E/genetics , Cyclin E/metabolism , G1 Phase , HeLa Cells , Humans , Intrinsically Disordered Proteins/genetics , Nuclear Proteins/genetics , Origin Recognition Complex/genetics , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Stability , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism
10.
Cell ; 155(1): 135-47, 2013 09 26.
Article in English | MEDLINE | ID: mdl-24074866

ABSTRACT

Self-renewal and differentiation of stem cells are fundamentally associated with cell-cycle progression to enable tissue specification, organ homeostasis, and potentially tumorigenesis. However, technical challenges have impaired the study of the molecular interactions coordinating cell fate choice and cell-cycle progression. Here, we bypass these limitations by using the FUCCI reporter system in human pluripotent stem cells and show that their capacity of differentiation varies during the progression of their cell cycle. These mechanisms are governed by the cell-cycle regulators cyclin D1-3 that control differentiation signals such as the TGF-ß-Smad2/3 pathway. Conversely, cell-cycle manipulation using a small molecule directs differentiation of hPSCs and provides an approach to generate cell types with a clinical interest. Our results demonstrate that cell fate decisions are tightly associated with the cell-cycle machinery and reveal insights in the mechanisms synchronizing differentiation and proliferation in developing tissues.


Subject(s)
Cell Cycle , Cell Differentiation , Cyclin D/metabolism , Embryonic Stem Cells/cytology , Signal Transduction , Cyclin D/genetics , Embryonic Stem Cells/metabolism , G1 Phase , Gene Knockdown Techniques , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Smad2 Protein/genetics , Smad3 Protein/genetics
11.
Nature ; 605(7909): 357-365, 2022 05.
Article in English | MEDLINE | ID: mdl-35508654

ABSTRACT

The entry of mammalian cells into the DNA synthesis phase (S phase) represents a key event in cell division1. According to current models of the cell cycle, the kinase CDC7 constitutes an essential and rate-limiting trigger of DNA replication, acting together with the cyclin-dependent kinase CDK2. Here we show that CDC7 is dispensable for cell division of many different cell types, as determined using chemical genetic systems that enable acute shutdown of CDC7 in cultured cells and in live mice. We demonstrate that another cell cycle kinase, CDK1, is also active during G1/S transition both in cycling cells and in cells exiting quiescence. We show that CDC7 and CDK1 perform functionally redundant roles during G1/S transition, and at least one of these kinases must be present to allow S-phase entry. These observations revise our understanding of cell cycle progression by demonstrating that CDK1 physiologically regulates two distinct transitions during cell division cycle, whereas CDC7 has a redundant function in DNA replication.


Subject(s)
Cell Cycle Proteins , G1 Phase , Protein Serine-Threonine Kinases , Proteolysis , S Phase , Animals , Cell Cycle Proteins/metabolism , DNA Replication , Mice , Protein Serine-Threonine Kinases/metabolism
12.
Nature ; 606(7912): 197-203, 2022 06.
Article in English | MEDLINE | ID: mdl-35585235

ABSTRACT

Eukaryotic genomes are compacted into loops and topologically associating domains (TADs)1-3, which contribute to transcription, recombination and genomic stability4,5. Cohesin extrudes DNA into loops that are thought to lengthen until CTCF boundaries are encountered6-12. Little is known about whether loop extrusion is impeded by DNA-bound machines. Here we show that the minichromosome maintenance (MCM) complex is a barrier that restricts loop extrusion in G1 phase. Single-nucleus Hi-C (high-resolution chromosome conformation capture) of mouse zygotes reveals that MCM loading reduces CTCF-anchored loops and decreases TAD boundary insulation, which suggests that loop extrusion is impeded before reaching CTCF. This effect extends to HCT116 cells, in which MCMs affect the number of CTCF-anchored loops and gene expression. Simulations suggest that MCMs are abundant, randomly positioned and partially permeable barriers. Single-molecule imaging shows that MCMs are physical barriers that frequently constrain cohesin translocation in vitro. Notably, chimeric yeast MCMs that contain a cohesin-interaction motif from human MCM3 induce cohesin pausing, indicating that MCMs are 'active' barriers with binding sites. These findings raise the possibility that cohesin can arrive by loop extrusion at MCMs, which determine the genomic sites at which sister chromatid cohesion is established. On the basis of in vivo, in silico and in vitro data, we conclude that distinct loop extrusion barriers shape the three-dimensional genome.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , DNA , Minichromosome Maintenance Proteins , Animals , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Chromatids/chemistry , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA/chemistry , DNA/metabolism , G1 Phase , HCT116 Cells , Humans , Mice , Minichromosome Maintenance Complex Component 3/chemistry , Minichromosome Maintenance Complex Component 3/metabolism , Minichromosome Maintenance Proteins/chemistry , Minichromosome Maintenance Proteins/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Nucleic Acid Conformation , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Cohesins
13.
Mol Cell ; 80(2): 183-192, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32946743

ABSTRACT

The Cdk-Rb-E2F pathway integrates external and internal signals to control progression at the G1/S transition of the mammalian cell cycle. Alterations in this pathway are found in most human cancers, and specific cyclin-dependent kinase Cdk4/6 inhibitors are approved or in clinical trials for the treatment of diverse cancers. In the long-standing paradigm for G1/S control, Cdks inactivate the retinoblastoma tumor suppressor protein (Rb) through phosphorylation, which releases E2F transcription factors to drive cell-cycle progression from G1 to S. However, recent observations in the laboratory and clinic challenge central tenets of the current paradigm and demonstrate that our understanding of the Rb pathway and G1/S control is still incomplete. Here, we integrate these new findings with the previous paradigm to synthesize a current molecular and cellular view of the mammalian G1/S transition. A more complete and accurate understanding of G1/S control will lead to improved therapeutic strategies targeting the cell cycle in cancer.


Subject(s)
G1 Phase , S Phase , Animals , Cell Proliferation , Cyclin-Dependent Kinases/metabolism , Humans , Models, Biological , Retinoblastoma Protein/metabolism
14.
Mol Cell ; 78(2): 359-370.e6, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32246903

ABSTRACT

Yeast cells must grow to a critical size before committing to division. It is unknown how size is measured. We find that as cells grow, mRNAs for some cell-cycle activators scale faster than size, increasing in concentration, while mRNAs for some inhibitors scale slower than size, decreasing in concentration. Size-scaled gene expression could cause an increasing ratio of activators to inhibitors with size, triggering cell-cycle entry. Consistent with this, expression of the CLN2 activator from the promoter of the WHI5 inhibitor, or vice versa, interfered with cell size homeostasis, yielding a broader distribution of cell sizes. We suggest that size homeostasis comes from differential scaling of gene expression with size. Differential regulation of gene expression as a function of cell size could affect many cellular processes.


Subject(s)
Cell Division/genetics , Cell Size , Cyclins/genetics , Saccharomyces cerevisiae Proteins/genetics , Cell Cycle/genetics , G1 Phase/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Fungal/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development
15.
Mol Cell ; 78(1): 127-140.e7, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32035037

ABSTRACT

As cells enter mitosis, the genome is restructured to facilitate chromosome segregation, accompanied by dramatic changes in gene expression. However, the mechanisms that underlie mitotic transcriptional regulation are unclear. In contrast to transcribed genes, centromere regions retain transcriptionally active RNA polymerase II (Pol II) in mitosis. Here, we demonstrate that chromatin-bound cohesin is necessary to retain elongating Pol II at centromeres. We find that WAPL-mediated removal of cohesin from chromosome arms during prophase is required for the dissociation of Pol II and nascent transcripts, and failure of this process dramatically alters mitotic gene expression. Removal of cohesin/Pol II from chromosome arms in prophase is important for accurate chromosome segregation and normal activation of gene expression in G1. We propose that prophase cohesin removal is a key step in reprogramming gene expression as cells transition from G2 through mitosis to G1.


Subject(s)
Cell Cycle Proteins/physiology , Chromosomal Proteins, Non-Histone/physiology , Gene Expression Regulation , Mitosis/genetics , Transcription, Genetic , Anaphase/genetics , Animals , Aurora Kinase B/analysis , Cell Cycle , Cell Cycle Proteins/analysis , Cell Line , Centromere/enzymology , Chromosome Segregation , G1 Phase/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Humans , Metaphase/genetics , Prophase , RNA Polymerase II/metabolism , Xenopus laevis , Cohesins
16.
EMBO J ; 42(2): e110321, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36420556

ABSTRACT

Eukaryotic cells decide in late G1 phase of the cell cycle whether to commit to another round of division. This point of cell cycle commitment is termed "Restriction Point" in mammals and "Start" in the budding yeast Saccharomyces cerevisiae. At Start, yeast cells integrate multiple signals such as pheromones and nutrients, and will not pass Start if nutrients are lacking. However, how cells respond to nutrient depletion after the Start decision remains poorly understood. Here, we analyze how post-Start cells respond to nutrient depletion, by monitoring Whi5, the cell cycle inhibitor whose export from the nucleus determines Start. Surprisingly, we find that cells that have passed Start can re-import Whi5 into the nucleus. In these cells, the positive feedback loop activating G1/S transcription is interrupted, and the Whi5 repressor re-binds DNA. Cells which re-import Whi5 become again sensitive to mating pheromone, like pre-Start cells, and CDK activation can occur a second time upon replenishment of nutrients. These results demonstrate that upon starvation, the commitment decision at Start can be reversed. We therefore propose that cell cycle commitment in yeast is a multi-step process, similar to what has been suggested for mammalian cells.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomycetales , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Cell Cycle , Cell Division , G1 Phase , Saccharomycetales/metabolism
17.
Nat Rev Mol Cell Biol ; 16(6): 360-74, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25999062

ABSTRACT

DNA replication begins with the assembly of pre-replication complexes (pre-RCs) at thousands of DNA replication origins during the G1 phase of the cell cycle. At the G1-S-phase transition, pre-RCs are converted into pre-initiation complexes, in which the replicative helicase is activated, leading to DNA unwinding and initiation of DNA synthesis. However, only a subset of origins are activated during any S phase. Recent insights into the mechanisms underlying this choice reveal how flexibility in origin usage and temporal activation are linked to chromosome structure and organization, cell growth and differentiation, and replication stress.


Subject(s)
DNA Replication/physiology , DNA/biosynthesis , G1 Phase/physiology , Replication Origin/physiology , S Phase/physiology , Animals , Cell Differentiation/physiology , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , DNA/genetics , Humans
18.
Cell ; 148(5): 908-21, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22341456

ABSTRACT

The extent to which the three-dimensional organization of the genome contributes to chromosomal translocations is an important question in cancer genomics. We generated a high-resolution Hi-C spatial organization map of the G1-arrested mouse pro-B cell genome and used high-throughput genome-wide translocation sequencing to map translocations from target DNA double-strand breaks (DSBs) within it. RAG endonuclease-cleaved antigen-receptor loci are dominant translocation partners for target DSBs regardless of genomic position, reflecting high-frequency DSBs at these loci and their colocalization in a fraction of cells. To directly assess spatial proximity contributions, we normalized genomic DSBs via ionizing radiation. Under these conditions, translocations were highly enriched in cis along single chromosomes containing target DSBs and within other chromosomes and subchromosomal domains in a manner directly related to pre-existing spatial proximity. By combining two high-throughput genomic methods in a genetically tractable system, we provide a new lens for viewing cancer genomes.


Subject(s)
Genome , Neoplasms/genetics , Translocation, Genetic , Animals , DNA Breaks, Double-Stranded/radiation effects , G1 Phase , High-Throughput Nucleotide Sequencing , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Precursor Cells, B-Lymphoid/cytology , Receptors, Antigen/genetics
19.
Cell ; 148(1-2): 99-111, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22265405

ABSTRACT

The replication of eukaryotic chromosomes is organized temporally and spatially within the nucleus through epigenetic regulation of replication origin function. The characteristic initiation timing of specific origins is thought to reflect their chromatin environment or sub-nuclear positioning, however the mechanism remains obscure. Here we show that the yeast Forkhead transcription factors, Fkh1 and Fkh2, are global determinants of replication origin timing. Forkhead regulation of origin timing is independent of local levels or changes of transcription. Instead, we show that Fkh1 and Fkh2 are required for the clustering of early origins and their association with the key initiation factor Cdc45 in G1 phase, suggesting that Fkh1 and Fkh2 selectively recruit origins to emergent replication factories. Fkh1 and Fkh2 bind Fkh-activated origins, and interact physically with ORC, providing a plausible mechanism to cluster origins. These findings add a new dimension to our understanding of the epigenetic basis for differential origin regulation and its connection to chromosomal domain organization.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Transcription Factors/metabolism , Replication Origin , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DNA-Binding Proteins/metabolism , G1 Phase , Nuclear Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Transcription, Genetic
20.
Mol Cell ; 74(3): 571-583.e8, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30898438

ABSTRACT

In mitosis, cells inactivate DNA double-strand break (DSB) repair pathways to preserve genome stability. However, some early signaling events still occur, such as recruitment of the scaffold protein MDC1 to phosphorylated histone H2AX at DSBs. Yet, it remains unclear whether these events are important for maintaining genome stability during mitosis. Here, we identify a highly conserved protein-interaction surface in MDC1 that is phosphorylated by CK2 and recognized by the DNA-damage response mediator protein TOPBP1. Disruption of MDC1-TOPBP1 binding causes a specific loss of TOPBP1 recruitment to DSBs in mitotic but not interphase cells, accompanied by mitotic radiosensitivity, increased micronuclei, and chromosomal instability. Mechanistically, we find that TOPBP1 forms filamentous structures capable of bridging MDC1 foci in mitosis, indicating that MDC1-TOPBP1 complexes tether DSBs until repair is reactivated in the following G1 phase. Thus, we reveal an important, hitherto-unnoticed cooperation between MDC1 and TOPBP1 in maintaining genome stability during cell division.


Subject(s)
Carrier Proteins/genetics , Chromosomal Instability/genetics , DNA-Binding Proteins/genetics , Mitosis/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA Repair/genetics , G1 Phase/genetics , Genome, Human/genetics , Genomic Instability/genetics , Histones , Humans , Phosphorylation , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL