Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 173, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38597967

RESUMEN

Heterozygous mutations in any of three major genes, BRCA1, BRCA2 and PALB2, are associated with high-risk hereditary breast cancer susceptibility frequently seen as familial disease clustering. PALB2 is a key interaction partner and regulator of several vital cellular activities of BRCA1 and BRCA2, and is thus required for DNA damage repair and alleviation of replicative and oxidative stress. Little is however known about how PALB2-deficiency affects cell function beyond that, especially in the three-dimensional setting, and also about its role during early steps of malignancy development. To answer these questions, we have generated biologically relevant MCF10A mammary epithelial cell lines with mutations that are comparable to certain clinically important PALB2 defects. We show in a non-cancerous background how both mono- and biallelically PALB2-mutated cells exhibit gross spontaneous DNA damage and mitotic aberrations. Furthermore, PALB2-deficiency disturbs three-dimensional spheroid morphology, increases the migrational capacity and invasiveness of the cells, and broadly alters their transcriptome profiles. TGFß signaling and KRT14 expression are enhanced in PALB2-mutated cells and their inhibition and knock down, respectively, lead to partial restoration of cell functions. KRT14-positive cells are also more abundant with DNA damage than KRT14-negative cells. The obtained results indicate comprehensive cellular changes upon PALB2 mutations, even in the presence of half dosage of wild type PALB2 and demonstrate how PALB2 mutations may predispose their carriers to malignancy.


Asunto(s)
Neoplasias , Transducción de Señal , Humanos , Reparación del ADN , Células Epiteliales , Mama , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética
2.
Nucleic Acids Res ; 50(17): 9948-9965, 2022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36099415

RESUMEN

Mutations in the lamin A/C gene (LMNA) cause laminopathies such as the premature aging Hutchinson Gilford progeria syndrome (HGPS) and altered lamin A/C levels are found in diverse malignancies. The underlying lamin-associated mechanisms remain poorly understood. Here we report that lamin A/C-null mouse embryo fibroblasts (Lmna-/- MEFs) and human progerin-expressing HGPS fibroblasts both display reduced NAD+ levels, unstable mitochondrial DNA and attenuated bioenergetics. This mitochondrial dysfunction is associated with reduced chromatin recruitment (Lmna-/- MEFs) or low levels (HGPS) of PGC1α, the key transcription factor for mitochondrial homeostasis. Lmna-/- MEFs showed reduced expression of the NAD+-biosynthesis enzyme NAMPT and attenuated activity of the NAD+-dependent deacetylase SIRT1. We find high PARylation in lamin A/C-aberrant cells, further decreasing the NAD+ pool and consistent with impaired DNA base excision repair in both cell models, a condition that fuels DNA damage-induced PARylation under oxidative stress. Further, ATAC-sequencing revealed a substantially altered chromatin landscape in Lmna-/- MEFs, including aberrantly reduced accessibility at the Nampt gene promoter. Thus, we identified a new role of lamin A/C as a key modulator of mitochondrial function through impairments of PGC1α and the NAMPT-NAD+ pathway, with broader implications for the aging process.


Asunto(s)
Lamina Tipo A/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Progeria , Animales , Cromatina/metabolismo , ADN Mitocondrial/metabolismo , Fibroblastos/metabolismo , Humanos , Lamina Tipo A/genética , Ratones , Mitocondrias/metabolismo , NAD/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Progeria/metabolismo , Sirtuina 1/genética
3.
EMBO J ; 38(20): e103421, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31566762

RESUMEN

Marked cyclin protein oscillations over the cell cycle ensure tight regulation of all cell cycle transitions. Despite expression patterns closely mirroring those of cyclin A, cyclin F has long been regarded as an odd outlier within the cyclin family. Constituting part of an E3 ubiquitin ligase, its main role was seen as highly restricted to timely degradation of very few key substrates to ensure termination of one error-free round of replication. Now, a recent series of studies suggests that cyclin F has very similar roles as its closest relatives, merely mediated through a very different mechanism.


Asunto(s)
Proteínas de Ciclo Celular , Ciclinas , Ciclo Celular , Factores de Transcripción E2F , Proteolisis , Mutaciones Letales Sintéticas , Factores de Transcripción
4.
Breast Cancer Res Treat ; 191(2): 431-441, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34755241

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/genética , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Polimorfismo de Nucleótido Simple
5.
Cell Mol Life Sci ; 77(4): 735-749, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31297568

RESUMEN

During S phase, replication forks can encounter several obstacles that lead to fork stalling, which if persistent might result in fork collapse. To avoid this collapse and to preserve the competence to restart, cells have developed mechanisms that maintain fork stability upon replication stress. In this study, we aimed to understand the mechanisms involved in fork stability maintenance in non-transformed human cells by performing an isolation of proteins on nascent DNA-mass spectrometry analysis in hTERT-RPE cells under different replication stress conditions. Our results show that acute hydroxyurea-induced replication blockade causes the accumulation of large amounts of single-stranded DNA at the fork. Remarkably, this results in the disengagement of replisome components from nascent DNA without compromising fork restart. Notably, Cdc45-MCM-GINS helicase maintains its integrity and replisome components remain associated with chromatin upon acute hydroxyurea treatment, whereas replisome stability is lost upon a sustained replication stress that compromises the competence to restart.


Asunto(s)
Replicación del ADN/efectos de los fármacos , Hidroxiurea/farmacología , Línea Celular , ADN de Cadena Simple/genética , Humanos , Fase S/efectos de los fármacos
6.
Nucleic Acids Res ; 46(22): e135, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30215776

RESUMEN

Analysis of large-scale interphase genome positioning with reference to a nuclear landmark has recently been studied using sequencing-based single cell approaches. However, these approaches are dependent upon technically challenging, time consuming and costly high throughput sequencing technologies, requiring specialized bioinformatics tools and expertise. Here, we propose a novel, affordable and robust microscopy-based single cell approach, termed Topokaryotyping, to analyze and reconstruct the interphase positioning of genomic loci relative to a given nuclear landmark, detectable as banding pattern on mitotic chromosomes. This is accomplished by proximity-dependent histone labeling, where biotin ligase BirA fused to nuclear envelope marker Emerin was coexpressed together with Biotin Acceptor Peptide (BAP)-histone fusion followed by (i) biotin labeling, (ii) generation of mitotic spreads, (iii) detection of the biotin label on mitotic chromosomes and (iv) their identification by karyotyping. Using Topokaryotyping, we identified both cooperativity and stochasticity in the positioning of emerin-associated chromatin domains in individual cells. Furthermore, the chromosome-banding pattern showed dynamic changes in emerin-associated domains upon physical and radiological stress. In summary, Topokaryotyping is a sensitive and reliable technique to quantitatively analyze spatial positioning of genomic regions interacting with a given nuclear landmark at the single cell level in various experimental conditions.


Asunto(s)
Cariotipificación/métodos , Mitosis , Membrana Nuclear/metabolismo , Análisis de la Célula Individual/métodos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Células HEK293 , Células HeLa , Histonas/metabolismo , Humanos , Hibridación Fluorescente in Situ , Interfase , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Membrana Nuclear/genética , Proteínas Nucleares/metabolismo , Reproducibilidad de los Resultados
7.
Nucleic Acids Res ; 44(7): 3176-89, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26792895

RESUMEN

Successful and accurate completion of the replication of damage-containing DNA requires mainly recombination and RAD18-dependent DNA damage tolerance pathways. RAD18 governs at least two distinct mechanisms: translesion synthesis (TLS) and template switching (TS)-dependent pathways. Whereas TS is mainly error-free, TLS can work in an error-prone manner and, as such, the regulation of these pathways requires tight control to prevent DNA errors and potentially oncogenic transformation and tumorigenesis. In humans, the PCNA-associated recombination inhibitor (PARI) protein has recently been shown to inhibit homologous recombination (HR) events. Here, we describe a biochemical mechanism in which PARI functions as an HR regulator after replication fork stalling and during double-strand break repair. In our reconstituted biochemical system, we show that PARI inhibits DNA repair synthesis during recombination events in a PCNA interaction-dependent way but independently of its UvrD-like helicase domain. In accordance, we demonstrate that PARI inhibits HR in vivo, and its knockdown suppresses the UV sensitivity of RAD18-depleted cells. Our data reveal a novel human regulatory mechanism that limits the extent of HR and represents a new potential target for anticancer therapy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Reparación del ADN por Recombinación , Secuencias de Aminoácidos , ADN/biosíntesis , ADN Polimerasa III/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/fisiología , Células HEK293 , Humanos , Ubiquitina-Proteína Ligasas/fisiología , Rayos Ultravioleta
8.
Semin Cancer Biol ; 37-38: 16-25, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26805514

RESUMEN

A dividing cell has to duplicate its DNA precisely once during the cell cycle to preserve genome integrity avoiding the accumulation of genetic aberrations that promote diseases such as cancer. A large number of endogenous impacts can challenge DNA replication and cells harbor a battery of pathways to promote genome integrity during DNA replication. This includes suppressing new replication origin firing, stabilization of replicating forks, and the safe restart of forks to prevent any loss of genetic information. Here, we describe mechanisms by which oncogenes can interfere with DNA replication thereby causing DNA replication stress and genome instability. Further, we describe cellular and systemic responses to these insults with a focus on DNA replication restart pathways. Finally, we discuss the therapeutic potential of exploiting intrinsic replicative stress in cancer cells for targeted therapy.


Asunto(s)
Replicación del ADN , Neoplasias/genética , Medicina de Precisión/métodos , Origen de Réplica , Daño del ADN , Inestabilidad Genómica , Humanos , Neoplasias/terapia , Oncogenes
9.
Int J Mol Sci ; 18(7)2017 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-28698521

RESUMEN

Genomic DNA is compacted into chromatin through packaging with histone and non-histone proteins. Importantly, DNA accessibility is dynamically regulated to ensure genome stability. This is exemplified in the response to DNA damage where chromatin relaxation near genomic lesions serves to promote access of relevant enzymes to specific DNA regions for signaling and repair. Furthermore, recent data highlight genome maintenance roles of chromatin through the regulation of endogenous DNA-templated processes including transcription and replication. Here, we review research that shows the importance of chromatin structure regulation in maintaining genome integrity by multiple mechanisms including facilitating DNA repair and directly suppressing endogenous DNA damage.


Asunto(s)
Daño del ADN/fisiología , Reparación del ADN/fisiología , Animales , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Daño del ADN/genética , Reparación del ADN/genética , Replicación del ADN/genética , Replicación del ADN/fisiología , Inestabilidad Genómica/genética , Inestabilidad Genómica/fisiología , Humanos
10.
Proc Natl Acad Sci U S A ; 110(42): 16856-61, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082115

RESUMEN

DNA damage can obstruct replication forks, resulting in replicative stress. By siRNA screening, we identified kinases involved in the accumulation of phosphohistone 2AX (γH2AX) upon UV irradiation-induced replication stress. Surprisingly, the strongest reduction of phosphohistone 2AX followed knockdown of the MAP kinase-activated protein kinase 2 (MK2), a kinase currently implicated in p38 stress signaling and G2 arrest. Depletion or inhibition of MK2 also protected cells from DNA damage-induced cell death, and mice deficient for MK2 displayed decreased apoptosis in the skin upon UV irradiation. Moreover, MK2 activity was required for damage response, accumulation of ssDNA, and decreased survival when cells were treated with the nucleoside analogue gemcitabine or when the checkpoint kinase Chk1 was antagonized. By using DNA fiber assays, we found that MK2 inhibition or knockdown rescued DNA replication impaired by gemcitabine or by Chk1 inhibition. This rescue strictly depended on translesion DNA polymerases. In conclusion, instead of being an unavoidable consequence of DNA damage, alterations of replication speed and origin firing depend on MK2-mediated signaling.


Asunto(s)
Replicación del ADN , Puntos de Control de la Fase G2 del Ciclo Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Daño del ADN , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Técnicas de Silenciamiento del Gen , Histonas/genética , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Rayos Ultravioleta , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Gemcitabina
11.
Nucleic Acids Res ; 41(5): 2797-806, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23345616

RESUMEN

Maintenance of genomic integrity is essential to ensure normal organismal development and to prevent diseases such as cancer. Nuclear DNA is packaged into chromatin, and thus genome maintenance can be influenced by distinct chromatin environments. In particular, post-translational modifications of histones have emerged as key regulators of genomic integrity. Intense research during the past few years has revealed histone H4 lysine 20 methylation (H4K20me) as critically important for the biological processes that ensure genome integrity, such as DNA damage repair, DNA replication and chromatin compaction. The distinct H4K20 methylation states are mediated by SET8/PR-Set7 that catalyses monomethylation of H4K20, whereas SUV4-20H1 and SUV4-20H2 enzymes mediate further H4K20 methylation to H4K20me2 and H4K20me3. Disruption of these H4K20-specific histone methyltransferases leads to genomic instability, demonstrating the important functions of H4K20 methylation in genome maintenance. In this review, we explain molecular mechanisms underlying these defects and discuss novel ideas for furthering our understanding of genome maintenance in higher eukaryotes.


Asunto(s)
Epigénesis Genética , Inestabilidad Genómica , Histonas/metabolismo , Lisina/metabolismo , Animales , Ciclo Celular , Cromatina/metabolismo , Daño del ADN , Replicación del ADN , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metilación , Proteína 1 de Unión al Supresor Tumoral P53
12.
Nucleic Acids Res ; 40(2): 477-86, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21937510

RESUMEN

Mechanisms that preserve genome integrity are highly important during the normal life cycle of human cells. Loss of genome protective mechanisms can lead to the development of diseases such as cancer. Checkpoint kinases function in the cellular surveillance pathways that help cells to cope with DNA damage. Importantly, the checkpoint kinases ATR, CHK1 and WEE1 are not only activated in response to exogenous DNA damaging agents, but are active during normal S phase progression. Here, we review recent evidence that these checkpoint kinases are critical to avoid deleterious DNA breakage during DNA replication in normal, unperturbed cell cycle. Possible mechanisms how loss of these checkpoint kinases may cause DNA damage in S phase are discussed. We propose that the majority of DNA damage is induced as a consequence of deregulated CDK activity that forces unscheduled initiation of DNA replication. This could generate structures that are cleaved by DNA endonucleases leading to the formation of DNA double-strand breaks. Finally, we discuss how these S phase effects may impact on our understanding of cancer development following disruption of these checkpoint kinases, as well as on the potential of these kinases as targets for cancer treatment.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Replicación del ADN , Proteínas Nucleares/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Transformación Celular Neoplásica/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Daño del ADN , Genoma , Humanos , Ratones
13.
EMBO Rep ; 12(7): 705-12, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21637299

RESUMEN

To identify key connections between DNA-damage repair and checkpoint pathways, we performed RNA interference screens for regulators of the ionizing radiation-induced G2 checkpoint, and we identified the breast cancer gene BRCA2. The checkpoint was also abrogated following depletion of PALB2, an interaction partner of BRCA2. BRCA2 and PALB2 depletion led to premature checkpoint abrogation and earlier activation of the AURORA A-PLK1 checkpoint-recovery pathway. These results indicate that the breast cancer tumour suppressors and homologous recombination repair proteins BRCA2 and PALB2 are main regulators of G2 checkpoint maintenance following DNA-damage.


Asunto(s)
Proteína BRCA2/metabolismo , Fase G2/fisiología , Ensayos Analíticos de Alto Rendimiento , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína BRCA2/genética , Línea Celular , Daño del ADN , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Fase G2/genética , Biblioteca de Genes , Células HCT116 , Células HeLa , Humanos , Proteínas Nucleares/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Recombinación Genética , Transducción de Señal/genética , Proteínas Supresoras de Tumor/genética
14.
iScience ; 26(1): 105806, 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36632060

RESUMEN

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.

15.
NAR Cancer ; 5(3): zcad029, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37325550

RESUMEN

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.

16.
Cancer Gene Ther ; 30(10): 1369-1381, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37495855

RESUMEN

Radiotherapy is one of the most common cancer treatments, yet, some patients require high doses to respond. Therefore, the development of new strategies leans toward personalizing therapy to avoid unnecessary burden on cancer patients. This approach prevents the administration of ineffective treatments or uses combination strategies to increase the sensitivity of cancer cells. ADAM12 has been shown to be upregulated in many cancers and correlate with poor survival and chemoresistance, thus making it a potential candidate responsible for radioresistance. Here, we show that ADAM12 expression is upregulated in response to irradiation in both mouse and human cancer cells in vitro, as well as in tumor tissues from rectal cancer patients. Interestingly, the expression of ADAM12 following radiotherapy correlates with the initial disease stage and predicts the response of rectal cancer patients to the treatment. While we found no cell-autonomous effects of ADAM12 on the response of colon cancer cells to irradiation in vitro, depletion of ADAM12 expression markedly reduced the tumor growth of irradiated cancer cells when subcutaneously transplanted in syngeneic mice. Interestingly, loss of cancer cell-derived ADAM12 expression increased the number of CD31+FAP- cells in murine tumors. Moreover, conditioned medium from ADAM12-/- colon cancer cells led to increased tube formation when added to endothelial cell cultures. Thus, it is tempting to speculate that altered tumor vascularity may be implicated in the observed effect of ADAM12 on response to radiotherapy in rectal cancer. We conclude that ADAM12 represents a promising prognostic factor for stratification of rectal cancer patients receiving radiotherapy and suggest that targeting ADAM12 in combination with radiotherapy could potentially improve the treatment response.


Asunto(s)
Neoplasias del Colon , Neoplasias del Recto , Animales , Humanos , Ratones , Proteína ADAM12/genética , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/radioterapia , Regulación Neoplásica de la Expresión Génica , Pronóstico , Neoplasias del Recto/genética , Neoplasias del Recto/radioterapia
17.
Nat Metab ; 5(4): 642-659, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37012496

RESUMEN

Cancer cells fuel their increased need for nucleotide supply by upregulating one-carbon (1C) metabolism, including the enzymes methylenetetrahydrofolate dehydrogenase-cyclohydrolase 1 and 2 (MTHFD1 and MTHFD2). TH9619 is a potent inhibitor of dehydrogenase and cyclohydrolase activities in both MTHFD1 and MTHFD2, and selectively kills cancer cells. Here, we reveal that, in cells, TH9619 targets nuclear MTHFD2 but does not inhibit mitochondrial MTHFD2. Hence, overflow of formate from mitochondria continues in the presence of TH9619. TH9619 inhibits the activity of MTHFD1 occurring downstream of mitochondrial formate release, leading to the accumulation of 10-formyl-tetrahydrofolate, which we term a 'folate trap'. This results in thymidylate depletion and death of MTHFD2-expressing cancer cells. This previously uncharacterized folate trapping mechanism is exacerbated by physiological hypoxanthine levels that block the de novo purine synthesis pathway, and additionally prevent 10-formyl-tetrahydrofolate consumption for purine synthesis. The folate trapping mechanism described here for TH9619 differs from other MTHFD1/2 inhibitors and antifolates. Thus, our findings uncover an approach to attack cancer and reveal a regulatory mechanism in 1C metabolism.


Asunto(s)
Metilenotetrahidrofolato Deshidrogenasa (NADP) , Neoplasias , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Ácido Fólico/metabolismo , Formiatos , Purinas , Tetrahidrofolatos
18.
Nat Cell Biol ; 7(2): 195-201, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15665856

RESUMEN

The essential checkpoint kinase Chk1 is required for cell-cycle delays after DNA damage or blocked DNA replication. However, it is unclear whether Chk1 is involved in the repair of damaged DNA. Here we establish that Chk1 is a key regulator of genome maintenance by the homologous recombination repair (HRR) system. Abrogation of Chk1 function with small interfering RNA or chemical antagonists inhibits HRR, leading to persistent unrepaired DNA double-strand breaks (DSBs) and cell death after replication inhibition with hydroxyurea or DNA-damage caused by camptothecin. After hydroxyurea treatment, the essential recombination repair protein RAD51 is recruited to DNA repair foci performing a vital role in correct HRR. We demonstrate that Chk1 interacts with RAD51, and that RAD51 is phosphorylated on Thr 309 in a Chk1-dependent manner. Consistent with a functional interplay between Chk1 and RAD51, Chk1-depleted cells failed to form RAD51 nuclear foci after exposure to hydroxyurea, and cells expressing a phosphorylation-deficient mutant RAD51(T309A) were hypersensitive to hydroxyurea. These results highlight a crucial role for the Chk1 signalling pathway in protecting cells against lethal DNA lesions through regulation of HRR.


Asunto(s)
Reparación del ADN , Proteínas Quinasas/fisiología , Recombinación Genética , Animales , Camptotecina/farmacología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Cricetinae , ADN , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Hidroxiurea/farmacología , Recombinasa Rad51 , Transducción de Señal
19.
Cancer Cell ; 3(3): 247-58, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12676583

RESUMEN

Chk1 kinase coordinates cell cycle progression and preserves genome integrity. Here, we show that chemical or genetic ablation of human Chk1 triggered supraphysiological accumulation of the S phase-promoting Cdc25A phosphatase, prevented ionizing radiation (IR)-induced degradation of Cdc25A, and caused radioresistant DNA synthesis (RDS). The basal turnover of Cdc25A operating in unperturbed S phase required Chk1-dependent phosphorylation of serines 123, 178, 278, and 292. IR-induced acceleration of Cdc25A proteolysis correlated with increased phosphate incorporation into these residues generated by a combined action of Chk1 and Chk2 kinases. Finally, phosphorylation of Chk1 by ATM was required to fully accelerate the IR-induced degradation of Cdc25A. Our results provide evidence that the mammalian S phase checkpoint functions via amplification of physiologically operating, Chk1-dependent mechanisms.


Asunto(s)
Ciclo Celular/fisiología , Proteínas Quinasas/metabolismo , Fosfatasas cdc25/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de la radiación , Proteínas de Ciclo Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Replicación del ADN/efectos de la radiación , Proteínas de Unión al ADN , Activación Enzimática , Células HeLa , Humanos , Cinética , Modelos Biológicos , Fosforilación , Proteínas Serina-Treonina Quinasas/fisiología , Radiación Ionizante , Fase S/efectos de la radiación , Serina/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Fosfatasas cdc25/efectos de la radiación
20.
Cell Rep ; 38(3): 110261, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35045293

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Replicación del ADN/fisiología , Proteínas Tirosina Quinasas/metabolismo , Línea Celular , Inestabilidad Genómica/fisiología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA