Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
NAR Cancer ; 5(3): zcad029, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37325550

ABSTRACT

Ovarian cancer is driven by genetic alterations that necessitate protective DNA damage and replication stress responses through cell cycle control and genome maintenance. This creates specific vulnerabilities that may be exploited therapeutically. WEE1 kinase is a key cell cycle control kinase, and it has emerged as a promising cancer therapy target. However, adverse effects have limited its clinical progress, especially when tested in combination with chemotherapies. A strong genetic interaction between WEE1 and PKMYT1 led us to hypothesize that a multiple low-dose approach utilizing joint WEE1 and PKMYT1 inhibition would allow exploitation of the synthetic lethality. We found that the combination of WEE1 and PKMYT1 inhibition exhibited synergistic effects in eradicating ovarian cancer cells and organoid models at a low dose. The WEE1 and PKMYT1 inhibition synergistically promoted CDK activation. Furthermore, the combined treatment exacerbated DNA replication stress and replication catastrophe, leading to increase of the genomic instability and inflammatory STAT1 signalling activation. These findings suggest a new multiple low-dose approach to harness the potency of WEE1 inhibition through the synthetic lethal interaction with PKMYT1 that may contribute to the development of new treatments for ovarian cancer.

2.
Trends Cell Biol ; 33(10): 850-859, 2023 10.
Article in English | MEDLINE | ID: mdl-36997393

ABSTRACT

Self-inflicted DNA strand breaks are canonically linked with cell death pathways and the establishment of genetic diversity in immune and germline cells. Moreover, this form of DNA damage is an established source of genome instability in cancer development. However, recent studies indicate that nonlethal self-inflicted DNA strand breaks play an indispensable but underappreciated role in a variety of cell processes, including differentiation and cancer therapy responses. Mechanistically, these physiological DNA breaks originate from the activation of nucleases, which are best characterized for inducing DNA fragmentation in apoptotic cell death. In this review, we outline the emerging biology of one critical nuclease, caspase-activated DNase (CAD), and how directed activation or deployment of this enzyme can lead to divergent cell fate outcomes.


Subject(s)
Apoptosis , Neoplasms , Humans , DNA/metabolism , DNA Damage , Neoplasms/genetics , Cell Differentiation , DNA Breaks
3.
iScience ; 26(1): 105806, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36632060

ABSTRACT

Wee1-like protein kinase (WEE1) restrains activities of cyclin-dependent kinases (CDKs) in S and G2 phase. Inhibition of WEE1 evokes drastic increase in CDK activity, which perturbs replication dynamics and compromises cell cycle checkpoints. Notably, WEE1 inhibitors such as adavosertib are tested in cancer treatment trials; however, WEE1-regulated phosphoproteomes and their dynamics have not been systematically investigated. In this study, we identified acute time-resolved alterations in the cellular phosphoproteome following WEE1 inhibition with adavosertib. These treatments acutely elevated CDK activities with distinct phosphorylation dynamics revealing more than 600 potential uncharacterized CDK sites. Moreover, we identified a major role for WEE1 in controlling CDK-dependent phosphorylation of multiple clustered sites in the key DNA repair factors MDC1, 53BP1, and RIF1. Functional analysis revealed that WEE1 fine-tunes CDK activities to permit recruitment of 53BP1 to chromatin. Thus, our findings uncover WEE1-controlled targets and pathways with translational potential for the clinical application of WEE1 inhibitors.

4.
Mol Cell ; 82(18): 3350-3365.e7, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36049481

ABSTRACT

It has been proposed that ATR kinase senses the completion of DNA replication to initiate the S/G2 transition. In contrast to this model, we show here that the TRESLIN-MTBP complex prevents a premature entry into G2 from early S-phase independently of ATR/CHK1 kinases. TRESLIN-MTBP acts transiently at pre-replication complexes (preRCs) to initiate origin firing and is released after the subsequent recruitment of CDC45. This dynamic behavior of TRESLIN-MTBP implements a monitoring system that checks the activation of replication forks and senses the rate of origin firing to prevent the entry into G2. This system detects the decline in the number of origins of replication that naturally occurs in very late S, which is the signature that cells use to determine the completion of DNA replication and permit the S/G2 transition. Our work introduces TRESLIN-MTBP as a key player in cell-cycle control independent of canonical checkpoints.


Subject(s)
Cell Cycle Proteins , DNA Replication , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/genetics , DNA-Binding Proteins/genetics
5.
Science ; 376(6592): 476-483, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35482866

ABSTRACT

Genotoxic therapy such as radiation serves as a frontline cancer treatment, yet acquired resistance that leads to tumor reoccurrence is frequent. We found that cancer cells maintain viability during irradiation by reversibly increasing genome-wide DNA breaks, thereby limiting premature mitotic progression. We identify caspase-activated DNase (CAD) as the nuclease inflicting these de novo DNA lesions at defined loci, which are in proximity to chromatin-modifying CCCTC-binding factor (CTCF) sites. CAD nuclease activity is governed through phosphorylation by DNA damage response kinases, independent of caspase activity. In turn, loss of CAD activity impairs cell fate decisions, rendering cancer cells vulnerable to radiation-induced DNA double-strand breaks. Our observations highlight a cancer-selective survival adaptation, whereby tumor cells deploy regulated DNA breaks to delimit the detrimental effects of therapy-evoked DNA damage.


Subject(s)
DNA Damage , Neoplasms , Chromatin , DNA/radiation effects , DNA Breaks, Double-Stranded , DNA Repair , Neoplasms/genetics
6.
Cell Rep ; 38(3): 110261, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045293

ABSTRACT

Cellular feedback systems ensure genome maintenance during DNA replication. When replication forks stall, newly replicated DNA is protected by pathways that limit excessive DNA nuclease attacks. Here we show that WEE1 activity guards against nascent DNA degradation at stalled forks. Furthermore, we identify WEE1-dependent suppression of cyclin-dependent kinase 2 (CDK2) as a major activity counteracting fork degradation. We establish DNA2 as the nuclease responsible for excessive fork degradation in WEE1-inhibited cells. In addition, WEE1 appears to be unique among CDK activity suppressors in S phase because neither CHK1 nor p21 promote fork protection as WEE1 does. Our results identify a key role of WEE1 in protecting stalled forks, which is separate from its established role in safeguarding DNA replication initiation. Our findings highlight how WEE1 inhibition evokes massive genome challenges during DNA replication, and this knowledge may improve therapeutic strategies to specifically eradicate cancer cells that frequently harbor elevated DNA replication stress.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 2/metabolism , DNA Replication/physiology , Protein-Tyrosine Kinases/metabolism , Cell Line , Genomic Instability/physiology , Humans
7.
Breast Cancer Res Treat ; 191(2): 431-441, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34755241

ABSTRACT

PURPOSE: Decades of research have identified multiple genetic variants associated with breast cancer etiology. However, there is no database that archives breast cancer genes and variants responsible for predisposition. We set out to build a dynamic repository of curated breast cancer genes. METHODS: A comprehensive literature search was performed in PubMed and Google Scholar, followed by data extraction and harmonization for downstream analysis. RESULTS: Using a subset of 345 studies, we cataloged 652 breast cancer-associated loci across the genome. A majority of these were present in the non-coding region (i.e., intergenic (101) and intronic (345)), whereas only 158 were located within an exon. Using the odds ratio, we identified 429 loci to increase the disease risk and 198 to confer protection against breast cancer, whereas 25 were identified to both increase disease risk and confer protection against breast cancer. Chromosomal ideogram analysis indicated that chromosomes 17 and 19 have the highest density of breast cancer loci. We manually annotated and collated breast cancer genes in which a previous association between rare-monogenic variant and breast cancer has been documented. Finally, network and functional enrichment analysis revealed that steroid metabolism and DNA repair pathways were predominant among breast cancer genes and variants. CONCLUSIONS: We have built an online interactive catalog of curated breast cancer genes ( https://cbcg.dk ). This will expedite clinical diagnostics and support the ongoing efforts in managing breast cancer etiology. Moreover, the database will serve as an essential repository when designing new breast cancer multigene panels.


Subject(s)
Breast Neoplasms , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Female , Genetic Predisposition to Disease , Genotype , Humans , Polymorphism, Single Nucleotide
8.
Cell Rep ; 30(7): 2416-2429.e7, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075739

ABSTRACT

It has been long assumed that normally leading strand synthesis must proceed coordinated with the lagging strand to prevent strand uncoupling and the pathological accumulation of single-stranded DNA (ssDNA) in the cell, a dogma recently challenged by in vitro studies in prokaryotes. Here, we report that human DNA polymerases can function independently at each strand in vivo and that the resulting strand uncoupling is supported physiologically by a cellular tolerance to ssDNA. Active forks rapidly accumulate ssDNA at the lagging strand when POLA1 is inhibited without triggering a stress response, despite ssDNA formation being considered a hallmark of replication stress. Acute POLA1 inhibition causes a lethal RPA exhaustion, but cells can duplicate their DNA with limited POLA1 activity and exacerbated strand uncoupling as long as RPA molecules suffice to protect the elevated ssDNA. Although robust, this uncoupled mode of DNA replication is also an in-built weakness that can be targeted for cancer treatment.


Subject(s)
DNA Replication/genetics , DNA, Single-Stranded/genetics , Protein Binding/genetics , Humans
9.
Cancer Cell ; 35(6): 821-822, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31185206

ABSTRACT

Combined inhibitions of PARP and DNA damage checkpoint have the potential for high anti-cancer efficacy, but concurrent inhibitions have been hampered by intolerable side effects. In this issue of Cancer Cell, Fang and colleagues (Fang et al., 2019) propose that sequential inhibitions of PARP and DNA damage checkpoint considerably widen the therapeutic window.


Subject(s)
Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Cell Cycle Checkpoints , Cell Cycle Proteins , Humans , Nuclear Proteins , Protein-Tyrosine Kinases
10.
EMBO J ; 36(14): 2161-2176, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28607002

ABSTRACT

After DNA damage, the cell cycle is arrested to avoid propagation of mutations. Arrest in G2 phase is initiated by ATM-/ATR-dependent signaling that inhibits mitosis-promoting kinases such as Plk1. At the same time, Plk1 can counteract ATR-dependent signaling and is required for eventual resumption of the cell cycle. However, what determines when Plk1 activity can resume remains unclear. Here, we use FRET-based reporters to show that a global spread of ATM activity on chromatin and phosphorylation of ATM targets including KAP1 control Plk1 re-activation. These phosphorylations are rapidly counteracted by the chromatin-bound phosphatase Wip1, allowing cell cycle restart despite persistent ATM activity present at DNA lesions. Combining experimental data and mathematical modeling, we propose a model for how the minimal duration of cell cycle arrest is controlled. Our model shows how cell cycle restart can occur before completion of DNA repair and suggests a mechanism for checkpoint adaptation in human cells.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromatin/metabolism , G2 Phase Cell Cycle Checkpoints , Protein Phosphatase 2C/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Cell Line , Fluorescence Resonance Energy Transfer , Humans , Models, Biological , Models, Theoretical , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , Repressor Proteins/metabolism , Tripartite Motif-Containing Protein 28 , Polo-Like Kinase 1
11.
Oncotarget ; 7(12): 14458-75, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26883108

ABSTRACT

PP2C family serine/threonine phosphatase WIP1 acts as a negative regulator of the tumor suppressor p53 and is implicated in silencing of cellular responses to genotoxic stress. Chromosomal locus 17q23 carrying the PPM1D (coding for WIP1) is commonly amplified in breast carcinomas and WIP1 was proposed as potential pharmacological target. Here we employed a cellular model with knocked out PPM1D to validate the specificity and efficiency of GSK2830371, novel small molecule inhibitor of WIP1. We have found that GSK2830371 increased activation of the DNA damage response pathway to a comparable level as the loss of PPM1D. In addition, GSK2830371 did not affect proliferation of cells lacking PPM1D but significantly supressed proliferation of breast cancer cells with amplified PPM1D. Over time cells treated with GSK2830371 accumulated in G1 and G2 phases of the cell cycle in a p21-dependent manner and were prone to induction of senescence by a low dose of MDM2 antagonist nutlin-3. In addition, combined treatment with GSK2830371 and doxorubicin or nutlin-3 potentiated cell death through a strong induction of p53 pathway and activation of caspase 9. We conclude that efficient inhibition of WIP1 by GSK2830371 sensitizes breast cancer cells with amplified PPM1D and wild type p53 to chemotherapy.


Subject(s)
Breast Neoplasms/drug therapy , Cell Cycle/drug effects , DNA Damage/drug effects , Drug Resistance, Neoplasm , Imidazoles/pharmacology , Piperazines/pharmacology , Protein Phosphatase 2C/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Aminopyridines/pharmacology , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Dipeptides/pharmacology , Female , Humans , Protein Phosphatase 2C/genetics , Protein Phosphatase 2C/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
12.
Biomolecules ; 5(3): 1912-37, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26295265

ABSTRACT

Cancer treatments such as radiotherapy and most of the chemotherapies act by damaging DNA of cancer cells. Upon DNA damage, cells stop proliferation at cell cycle checkpoints, which provides them time for DNA repair. Inhibiting the checkpoint allows entry to mitosis despite the presence of DNA damage and can lead to cell death. Importantly, as cancer cells exhibit increased levels of endogenous DNA damage due to an excessive replication stress, inhibiting the checkpoint kinases alone could act as a directed anti-cancer therapy. Here, we review the current status of inhibitors targeted towards the checkpoint effectors and discuss mechanisms of their actions in killing of cancer cells.


Subject(s)
Cell Cycle Checkpoints/drug effects , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Cell Death/drug effects , Humans , Neoplasms/enzymology , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Tumor Suppressor Protein p53/metabolism
13.
Cell Cycle ; 14(2): 219-31, 2015.
Article in English | MEDLINE | ID: mdl-25607646

ABSTRACT

In response to genotoxic stress, cells protect their genome integrity by activation of a conserved DNA damage response (DDR) pathway that coordinates DNA repair and progression through the cell cycle. Extensive modification of the chromatin flanking the DNA lesion by ATM kinase and RNF8/RNF168 ubiquitin ligases enables recruitment of various repair factors. Among them BRCA1 and 53BP1 are required for homologous recombination and non-homologous end joining, respectively. Whereas mechanisms of DDR are relatively well understood in interphase cells, comparatively less is known about organization of DDR during mitosis. Although ATM can be activated in mitotic cells, 53BP1 is not recruited to the chromatin until cells exit mitosis. Here we report mitotic phosphorylation of 53BP1 by Plk1 and Cdk1 that impairs the ability of 53BP1 to bind the ubiquitinated H2A and to properly localize to the sites of DNA damage. Phosphorylation of 53BP1 at S1618 occurs at kinetochores and in cytosol and is restricted to mitotic cells. Interaction between 53BP1 and Plk1 depends on the activity of Cdk1. We propose that activity of Cdk1 and Plk1 allows spatiotemporally controlled suppression of 53BP1 function during mitosis.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Repair , Mitosis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA Damage/radiation effects , Gamma Rays , HeLa Cells , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Kinetochores/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor p53-Binding Protein 1 , Ubiquitination , Polo-Like Kinase 1
14.
J Cell Biol ; 201(4): 511-21, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23649806

ABSTRACT

The DNA damage response (DDR) pathway and its core component tumor suppressor p53 block cell cycle progression after genotoxic stress and represent an intrinsic barrier preventing cancer development. The serine/threonine phosphatase PPM1D/Wip1 inactivates p53 and promotes termination of the DDR pathway. Wip1 has been suggested to act as an oncogene in a subset of tumors that retain wild-type p53. In this paper, we have identified novel gain-of-function mutations in exon 6 of PPM1D that result in expression of C-terminally truncated Wip1. Remarkably, mutations in PPM1D are present not only in the tumors but also in other tissues of breast and colorectal cancer patients, indicating that they arise early in development or affect the germline. We show that mutations in PPM1D affect the DDR pathway and propose that they could predispose to cancer.


Subject(s)
G1 Phase , Gene Expression Regulation, Neoplastic , Mutation , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/physiology , Tumor Suppressor Protein p53/genetics , Cell Cycle , Cell Line, Tumor , DNA Damage , Genetic Predisposition to Disease , HeLa Cells , Humans , MCF-7 Cells , Neoplasms/metabolism , Protein Phosphatase 2C
15.
Cell Cycle ; 12(2): 251-62, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23255129

ABSTRACT

Cells are constantly challenged by DNA damage and protect their genome integrity by activation of an evolutionary conserved DNA damage response pathway (DDR). A central core of DDR is composed of a spatiotemporally ordered net of post-translational modifications, among which protein phosphorylation plays a major role. Activation of checkpoint kinases ATM/ATR and Chk1/2 leads to a temporal arrest in cell cycle progression (checkpoint) and allows time for DNA repair. Following DNA repair, cells re-enter the cell cycle by checkpoint recovery. Wip1 phosphatase (also called PPM1D) dephosphorylates multiple proteins involved in DDR and is essential for timely termination of the DDR. Here we have investigated how Wip1 is regulated in the context of the cell cycle. We found that Wip1 activity is downregulated by several mechanisms during mitosis. Wip1 protein abundance increases from G(1) phase to G(2) and declines in mitosis. Decreased abundance of Wip1 during mitosis is caused by proteasomal degradation. In addition, Wip1 is phosphorylated at multiple residues during mitosis, and this leads to inhibition of its enzymatic activity. Importantly, ectopic expression of Wip1 reduced γH2AX staining in mitotic cells and decreased the number of 53BP1 nuclear bodies in G(1) cells. We propose that the combined decrease and inhibition of Wip1 in mitosis decreases the threshold necessary for DDR activation and enables cells to react adequately even to modest levels of DNA damage encountered during unperturbed mitotic progression.


Subject(s)
DNA Damage , Gene Expression Regulation/physiology , M Phase Cell Cycle Checkpoints/physiology , Mitosis/physiology , Phosphoprotein Phosphatases/metabolism , Signal Transduction/physiology , Cell Line, Tumor , DNA Primers/genetics , Fluorescent Antibody Technique , Humans , Mass Spectrometry , Phosphorylation , Protein Phosphatase 2C , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...