Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Cycle ; 17(7): 833-843, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29683380

RESUMEN

Mutagenesis is a hallmark and enabling characteristic of cancer cells. The E3 ubiquitin ligase RAD18 and its downstream effectors, the 'Y-family' Trans-Lesion Synthesis (TLS) DNA polymerases, confer DNA damage tolerance at the expense of DNA replication fidelity. Thus, RAD18 and TLS polymerases are attractive candidate mediators of mutagenesis and carcinogenesis. The skin cancer-propensity disorder xeroderma pigmentosum-variant (XPV) is caused by defects in the Y-family DNA polymerase Pol eta (Polη). However it is unknown whether TLS dysfunction contributes more generally to other human cancers. Recent analyses of cancer genomes suggest that TLS polymerases generate many of the mutational signatures present in diverse cancers. Moreover biochemical studies suggest that the TLS pathway is often reprogrammed in cancer cells and that TLS facilitates tolerance of oncogene-induced DNA damage. Here we review recent evidence supporting widespread participation of RAD18 and the Y-family DNA polymerases in the different phases of multi-step carcinogenesis.


Asunto(s)
Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Neoplasias/genética , Ubiquitina-Proteína Ligasas/genética , Xerodermia Pigmentosa/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Daño del ADN , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Genoma Humano , Humanos , Familia de Multigenes , Mutagénesis , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Xerodermia Pigmentosa/metabolismo , Xerodermia Pigmentosa/patología
2.
DNA Repair (Amst) ; 58: 52-61, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28866241

RESUMEN

Acylpeptide hydrolase (APEH) deacetylates N-alpha-acetylated peptides and selectively degrades oxidised proteins, but the biochemical pathways that are regulated by this protease are unknown. Here, we identify APEH as a component of the cellular response to DNA damage. Although APEH is primarily localised in the cytoplasm, we show that a sub-fraction of this enzyme is sequestered at sites of nuclear damage following UVA irradiation or following oxidative stress. We show that localization of APEH at sites of nuclear damage is mediated by direct interaction with XRCC1, a scaffold protein that accelerates the repair of DNA single-strand breaks. We show that APEH interacts with the amino-terminal domain of XRCC1, and that APEH facilitates both single-strand break repair and cell survival following exposure to H2O2 in human cells. These data identify APEH as a novel proteolytic component of the DNA damage response.


Asunto(s)
Roturas del ADN de Cadena Simple , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Péptido Hidrolasas/metabolismo , ADN/efectos de los fármacos , ADN/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
3.
J Cell Biol ; 216(10): 3097-3115, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28835467

RESUMEN

The mechanisms by which neoplastic cells tolerate oncogene-induced DNA replication stress are poorly understood. Cyclin-dependent kinase 2 (CDK2) is a major mediator of oncogenic DNA replication stress. In this study, we show that CDK2-inducing stimuli (including Cyclin E overexpression, oncogenic RAS, and WEE1 inhibition) activate the DNA repair protein RAD18. CDK2-induced RAD18 activation required initiation of DNA synthesis and was repressed by p53. RAD18 and its effector, DNA polymerase κ (Polκ), sustained ongoing DNA synthesis in cells harboring elevated CDK2 activity. RAD18-deficient cells aberrantly accumulated single-stranded DNA (ssDNA) after CDK2 activation. In RAD18-depleted cells, the G2/M checkpoint was necessary to prevent mitotic entry with persistent ssDNA. Rad18-/- and Polκ-/- cells were highly sensitive to the WEE1 inhibitor MK-1775 (which simultaneously activates CDK2 and abrogates the G2/M checkpoint). Collectively, our results show that the RAD18-Polκ signaling axis allows tolerance of CDK2-mediated oncogenic stress and may allow neoplastic cells to breach tumorigenic barriers.


Asunto(s)
Roturas del ADN de Cadena Simple , ADN de Neoplasias/biosíntesis , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Resistencia a Antineoplásicos , Neoplasias/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Células A549 , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , ADN de Neoplasias/genética , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/genética , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Pirimidinonas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética
4.
Blood ; 130(2): 156-166, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28495793

RESUMEN

The role of deubiquitylase ubiquitin-specific protease 7 (USP7) in the regulation of the p53-dependent DNA damage response (DDR) pathway is well established. Whereas previous studies have mostly focused on the mechanisms underlying how USP7 directly controls p53 stability, we recently showed that USP7 modulates the stability of the DNA damage responsive E3 ubiquitin ligase RAD18. This suggests that targeting USP7 may have therapeutic potential even in tumors with defective p53 or ibrutinib resistance. To test this hypothesis, we studied the effect of USP7 inhibition in chronic lymphocytic leukemia (CLL) where the ataxia telangiectasia mutated (ATM)-p53 pathway is inactivated with relatively high frequency, leading to treatment resistance and poor clinical outcome. We demonstrate that USP7 is upregulated in CLL cells, and its loss or inhibition disrupts homologous recombination repair (HRR). Consequently, USP7 inhibition induces significant tumor-cell killing independently of ATM and p53 through the accumulation of genotoxic levels of DNA damage. Moreover, USP7 inhibition sensitized p53-defective, chemotherapy-resistant CLL cells to clinically achievable doses of HRR-inducing chemotherapeutic agents in vitro and in vivo in a murine xenograft model. Together, these results identify USP7 as a promising therapeutic target for the treatment of hematological malignancies with DDR defects, where ATM/p53-dependent apoptosis is compromised.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Leucemia Linfocítica Crónica de Células B/genética , Reparación del ADN por Recombinación/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Proteasas Ubiquitina-Específicas/genética , Adenina/análogos & derivados , Animales , Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Daño del ADN , Resistencia a Antineoplásicos/genética , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Ratones , Ratones Endogámicos NOD , Piperidinas , Cultivo Primario de Células , Pirazoles/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Peptidasa Específica de Ubiquitina 7 , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Proteasas Ubiquitina-Específicas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nat Genet ; 49(4): 537-549, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28191891

RESUMEN

To ensure efficient genome duplication, cells have evolved numerous factors that promote unperturbed DNA replication and protect, repair and restart damaged forks. Here we identify downstream neighbor of SON (DONSON) as a novel fork protection factor and report biallelic DONSON mutations in 29 individuals with microcephalic dwarfism. We demonstrate that DONSON is a replisome component that stabilizes forks during genome replication. Loss of DONSON leads to severe replication-associated DNA damage arising from nucleolytic cleavage of stalled replication forks. Furthermore, ATM- and Rad3-related (ATR)-dependent signaling in response to replication stress is impaired in DONSON-deficient cells, resulting in decreased checkpoint activity and the potentiation of chromosomal instability. Hypomorphic mutations in DONSON substantially reduce DONSON protein levels and impair fork stability in cells from patients, consistent with defective DNA replication underlying the disease phenotype. In summary, we have identified mutations in DONSON as a common cause of microcephalic dwarfism and established DONSON as a critical replication fork protein required for mammalian DNA replication and genome stability.


Asunto(s)
Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Enanismo/genética , Inestabilidad Genómica/genética , Microcefalia/genética , Mutación/genética , Línea Celular , Daño del ADN/genética , Femenino , Humanos , Masculino
6.
Genes (Basel) ; 8(2)2017 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-28208741

RESUMEN

Accurate DNA replication is crucial for cell survival and the maintenance of genome stability. Cells have developed mechanisms to cope with the frequent genotoxic injuries that arise from both endogenous and environmental sources. Lesions encountered during DNA replication are often tolerated by post-replication repair mechanisms that prevent replication fork collapse and avert the formation of DNA double strand breaks. There are two predominant post-replication repair pathways, trans-lesion synthesis (TLS) and template switching (TS). TLS is a DNA damage-tolerant and low-fidelity mode of DNA synthesis that utilizes specialized 'Y-family' DNA polymerases to replicate damaged templates. TS, however, is an error-free 'DNA damage avoidance' mode of DNA synthesis that uses a newly synthesized sister chromatid as a template in lieu of the damaged parent strand. Both TLS and TS pathways are tightly controlled signaling cascades that integrate DNA synthesis with the overall DNA damage response and are thus crucial for genome stability. This review will cover the current knowledge of the primary mediators of post-replication repair and how they are regulated in the cell.

7.
Mol Cell Oncol ; 3(1): e1063571, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27308573

RESUMEN

RAD18 functions to promote DNA damage tolerance (DTT), a process that ensures faithful genome duplication. Protein ubiquitylation/deubiquitylation is a critical regulatory mechanism controlling DTT. Recently, we have identified the deubiquitylating enzyme USP7 as a component of the DTT machinery that acts to protect RAD18 from proteasome-dependent degradation.

8.
Nat Genet ; 48(1): 36-43, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26595769

RESUMEN

DNA lesions encountered by replicative polymerases threaten genome stability and cell cycle progression. Here we report the identification of mutations in TRAIP, encoding an E3 RING ubiquitin ligase, in patients with microcephalic primordial dwarfism. We establish that TRAIP relocalizes to sites of DNA damage, where it is required for optimal phosphorylation of H2AX and RPA2 during S-phase in response to ultraviolet (UV) irradiation, as well as fork progression through UV-induced DNA lesions. TRAIP is necessary for efficient cell cycle progression and mutations in TRAIP therefore limit cellular proliferation, providing a potential mechanism for microcephaly and dwarfism phenotypes. Human genetics thus identifies TRAIP as a component of the DNA damage response to replication-blocking DNA lesions.


Asunto(s)
Daño del ADN , Enanismo/genética , Mutación , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Proliferación Celular/genética , Preescolar , Daño del ADN/efectos de la radiación , Facies , Histonas/genética , Histonas/metabolismo , Humanos , Microcefalia/genética , Datos de Secuencia Molecular , Fosforilación , Proteína de Replicación A/metabolismo , Fase S/efectos de la radiación , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Ubiquitina-Proteína Ligasas/genética , Rayos Ultravioleta
9.
Mol Cell ; 59(3): 462-77, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26166705

RESUMEN

Recognition and repair of damaged replication forks are essential to maintain genome stability and are coordinated by the combined action of the Fanconi anemia and homologous recombination pathways. These pathways are vital to protect stalled replication forks from uncontrolled nucleolytic activity, which otherwise causes irreparable genomic damage. Here, we identify BOD1L as a component of this fork protection pathway, which safeguards genome stability after replication stress. Loss of BOD1L confers exquisite cellular sensitivity to replication stress and uncontrolled resection of damaged replication forks, due to a failure to stabilize RAD51 at these forks. Blocking DNA2-dependent resection, or downregulation of the helicases BLM and FBH1, suppresses both catastrophic fork processing and the accumulation of chromosomal damage in BOD1L-deficient cells. Thus, our work implicates BOD1L as a critical regulator of genome integrity that restrains nucleolytic degradation of damaged replication forks.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Línea Celular , Supervivencia Celular , Daño del ADN , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Genoma Humano , Inestabilidad Genómica , Células HeLa , Humanos , RecQ Helicasas/metabolismo
10.
EMBO J ; 32(12): 1778-92, 2013 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-23708797

RESUMEN

Limiting the levels of homologous recombination (HR) that occur at sites of DNA damage is a major role of BLM helicase. However, very little is known about the mechanisms dictating its relocalization to these sites. Here, we demonstrate that the ubiquitin/SUMO-dependent DNA damage response (UbS-DDR), controlled by the E3 ligases RNF8/RNF168, triggers BLM recruitment to sites of replication fork stalling via ubiquitylation in the N-terminal region of BLM and subsequent BLM binding to the ubiquitin-interacting motifs of RAP80. Furthermore, we show that this mechanism of BLM relocalization is essential for BLM's ability to suppress excessive/uncontrolled HR at stalled replication forks. Unexpectedly, we also uncovered a requirement for RNF8-dependent ubiquitylation of BLM and PML for maintaining the integrity of PML-associated nuclear bodies and as a consequence the localization of BLM to these structures. Lastly, we identified a novel role for RAP80 in preventing proteasomal degradation of BLM in unstressed cells. Taken together, these data highlight an important biochemical link between the UbS-DDR and BLM-dependent pathways involved in maintaining genome stability.


Asunto(s)
Daño del ADN , Inestabilidad Genómica/fisiología , Recombinación Homóloga/fisiología , Proteolisis , RecQ Helicasas/metabolismo , Ubiquitinación/fisiología , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , RecQ Helicasas/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
11.
Mol Cell ; 43(4): 649-62, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21855803

RESUMEN

Posttranslational modification of PCNA by ubiquitin plays an important role in coordinating the processes of DNA damage tolerance during DNA replication. The monoubiquitination of PCNA was shown to facilitate the switch between the replicative DNA polymerase with the low-fidelity polymerase eta (η) to bypass UV-induced DNA lesions during replication. Here, we show that in response to oxidative stress, PCNA becomes transiently monoubiquitinated in an S phase- and USP1-independent manner. Moreover, Polη interacts with mUb-PCNA at sites of oxidative DNA damage via its PCNA-binding and ubiquitin-binding motifs. Strikingly, while functional base excision repair is not required for this modification of PCNA or Polη recruitment to chromatin, the presence of hMsh2-hMsh6 is indispensable. Our findings highlight an alternative pathway in response to oxidative DNA damage that may coordinate the removal of oxidatively induced clustered DNA lesions and could explain the high levels of oxidized DNA lesions in MSH2-deficient cells.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/fisiología , ADN Polimerasa Dirigida por ADN/fisiología , Proteína 2 Homóloga a MutS/fisiología , Estrés Oxidativo , Antígeno Nuclear de Célula en Proliferación/fisiología , Proteínas de Arabidopsis , Línea Celular , Cromatina/metabolismo , ADN Polimerasa beta/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Endopeptidasas/metabolismo , Humanos , Proteína 2 Homóloga a MutS/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteasas Ubiquitina-Específicas , Ubiquitinación , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
12.
DNA Repair (Amst) ; 9(5): 588-92, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20197240

RESUMEN

Through the action of multiple sensors, mediators, and effectors, the DNA damage response (DDR) orchestrates the repair of DNA damage to ensure maintenance of genomic integrity. Recently, in addition to phosphorylation, other post-translational modifications such as ubiquitylation and SUMOylation have emerged as important regulators of the DDR network. Two recent papers highlight the importance of SUMO modifications of proteins that execute the response to DNA damage.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteína SUMO-1/metabolismo , Animales , Humanos
13.
EMBO J ; 27(21): 2883-95, 2008 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-18923427

RESUMEN

Human DNA polymerase iota (poliota) is a unique member of the Y-family of specialised polymerases that displays a 5'deoxyribose phosphate (dRP) lyase activity. Although poliota is well conserved in higher eukaryotes, its role in mammalian cells remains unclear. To investigate the biological importance of poliota in human cells, we generated fibroblasts stably downregulating poliota (MRC5-pol iota(KD)) and examined their response to several types of DNA-damaging agents. We show that cell lines downregulating poliota exhibit hypersensitivity to DNA damage induced by hydrogen peroxide (H(2)O(2)) or menadione but not to ethylmethane sulphonate (EMS), UVC or UVA. Interestingly, extracts from cells downregulating poliota show reduced base excision repair (BER) activity. In addition, poliota binds to chromatin after treatment of cells with H(2)O(2) and interacts with the BER factor XRCC1. Finally, green fluorescent protein-tagged poliota accumulates at the sites of oxidative DNA damage in living cells. This recruitment is partially mediated by its dRP lyase domain and ubiquitin-binding domains. These data reveal a novel role of human poliota in protecting cells from oxidative damage.


Asunto(s)
Citoprotección , ADN Polimerasa Dirigida por ADN/metabolismo , Fibroblastos/citología , Fibroblastos/enzimología , Estrés Oxidativo , Alquilantes/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Extractos Celulares , Línea Celular , Cromatina/enzimología , Citoprotección/efectos de los fármacos , Citoprotección/efectos de la radiación , Daño del ADN , ADN Polimerasa beta/metabolismo , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/deficiencia , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Fase G1/efectos de los fármacos , Fase G1/efectos de la radiación , Humanos , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Unión Proteica/efectos de los fármacos , Unión Proteica/efectos de la radiación , Estructura Terciaria de Proteína , Rayos Ultravioleta , Uracilo/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X , ADN Polimerasa iota
14.
Cell ; 117(1): 17-28, 2004 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-15066279

RESUMEN

CK2 was the first protein kinase identified and is required for the proliferation and survival of mammalian cells. Here, we have identified an unanticipated role for CK2. We show that this essential protein kinase phosphorylates the scaffold protein XRCC1 and thereby enables the assembly and activity of DNA single-strand break repair protein complexes in vitro and at sites of chromosomal breakage. Moreover, we show that inhibiting XRCC1 phosphorylation by mutation of the CK2 phosphorylation sites or preventing CK2 activity using a highly specific inhibitor ablates the rapid repair of cellular DNA single-strand breaks by XRCC1. These data identify a direct role for CK2 in the repair of chromosomal DNA strand breaks and in maintaining genetic integrity.


Asunto(s)
Cromosomas/enzimología , Daño del ADN/genética , Reparación del ADN/genética , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Sitios de Unión/genética , Células CHO , Quinasa de la Caseína II , Cromosomas/genética , Cricetinae , Reparación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Mutación/genética , Fosforilación/efectos de los fármacos , Polinucleótido 5'-Hidroxil-Quinasa/genética , Polinucleótido 5'-Hidroxil-Quinasa/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína/genética , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X , Levaduras/enzimología , Levaduras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA