Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Med Rep ; 16(2): 1885-1899, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28627616

RESUMEN

Repair of DNA interstrand crosslinks (ICLs) predominantly involves the Fanconi anemia (FA) pathway and homologous recombination (HR). The HR repair system eliminates DNA double strand breaks (DSBs) that emerge during ICLs removal. The current study presents a novel cell line, CL­V8B, representing a new complementation group of Chinese hamster cell mutants hypersensitive to DNA crosslinking factors. CL­V8B exhibits increased sensitivity to various DNA­damaging agents, including compounds leading to DSBs formation (bleomycin and 6­thioguanine), and is extremely sensitive to poly (ADP-ribose) polymerase inhibitor (>400­fold), which is typical for HR­defective cells. In addition, this cell line exhibits a reduced number of spontaneous and induced sister chromatid exchanges, which suggests likely impairment of HR in CL­V8B cells. However, in contrast to other known HR mutants, CL­V8B cells do not show defects in Rad51 foci induction, but only slight alterations in the focus formation kinetics. CL­V8B is additionally characterized by a considerable chromosomal instability, as indicated by a high number of spontaneous and MMC­induced chromosomal aberrations, and a twice as large proportion of cells with abnormal centrosomes than that in the wild type cell line. The molecular defect present in CL­V8B does not affect the efficiency and stabilization of replication forks. However, stalling of the forks in response to replication stress is observed relatively rarely, which suggests an impairment of a signaling mechanism. Exposure of CL­V8B to crosslinking agents results in S­phase arrest (as in the wild type cells), but also in larger proportion of G2/M­phase cells and apoptotic cells. CL­V8B exhibits similarities to HR­ and/or FA­defective Chinese hamster mutants sensitive to DNA crosslinking agents. However, the unique phenotype of this new mutant implies that it carries a defect of a yet unidentified gene involved in the repair of ICLs.


Asunto(s)
Daño del ADN , Reparación del ADN , Recombinación Homóloga , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Centrosoma/metabolismo , Aberraciones Cromosómicas , Células Clonales , Cricetinae , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Recombinación Homóloga/efectos de los fármacos , Cinética , Mitomicina/toxicidad , Mutágenos/toxicidad , Fenotipo , Recombinasa Rad51/metabolismo , Intercambio de Cromátides Hermanas/efectos de los fármacos
2.
Postepy Hig Med Dosw (Online) ; 68: 459-72, 2014 May 08.
Artículo en Polaco | MEDLINE | ID: mdl-24864098

RESUMEN

The Fanconi anemia (FA) pathway is one of the DNA repair systems involved in removal of DNA crosslinks. Proteins which belong to this pathway are crucial to the protection of genetic information, whereas disturbances in their function have serious implications for the whole organism. Biallelic mutations in FA genes are the cause of Fanconi anemia - a genetic disease which manifests itself through numerous congenital abnormalities, chromosomal instability and increased predisposition to cancer. The FA pathway is composed of fifteen proteins. Eight of them, in the presence of DNA interstrand crosslinks (ICLs), form a nuclear core complex responsible for monoubiquitination of FANCD2 and FANCI, which is a key step of ICL repair. FA proteins which are not involved in the monoubiquitination step participate in repair of DNA double strand breaks via homologous recombination. Some of the FA proteins, besides having a direct role in the repair of DNA damage, are engaged in replication, cell cycle control and mitosis. The unperturbed course of those processes determines the maintenance of genome stability.


Asunto(s)
Reparación del ADN/fisiología , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Inestabilidad Genómica/fisiología , Inestabilidad Cromosómica , Roturas del ADN de Doble Cadena , Daño del ADN , Replicación del ADN , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Humanos , Mutación
3.
Postepy Hig Med Dosw (Online) ; 66: 372-84, 2012 Jun 15.
Artículo en Polaco | MEDLINE | ID: mdl-22706123

RESUMEN

 A majority of currently used anticancer drugs belong to a group of chemical agents that damage DNA. The efficiency of the treatment is limited by effective DNA repair systems functioning in cancer cells. Many chemotherapeutic compounds cause strong systemic toxicity. Therefore, there is still a need for new anticancer agents which are less toxic for nontransformed cells and selectively kill cancer cells. One of the most promising molecular targets in cancer therapy is poly(ADP-ribose) polymerases (PARP). PARP play an essential role in repairing DNA strand breaks. Small molecule inhibitors of these enzymes have been developed and have proved to be extremely toxic for cancer cells that lack the functional BRCA1 and BRCA2 proteins that are involved in homologous recombination, a complex repair mechanism of DNA double strand breaks. Mutations in BRCA1/2 genes are associated with genetically inherited breast and ovarian cancers. Therefore PARP inhibitors may prove to be very effective and selective in the treatment of these cancer types. This review is focused on the function of BRCA1/2 proteins and poly(ADP-ribose) polymerases in DNA repair systems, especially in the homologous recombination process. A short history of the studies that led to synthesis of high specificity small molecule PARP inhibitors is also presented, as well as the results of clinical trials concerning the most effective PARP inhibitors in view of their potential application in oncological treatment, particularly breast cancers.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Genes BRCA1/efectos de los fármacos , Genes BRCA2/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Antineoplásicos/uso terapéutico , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos
4.
Int J Cancer ; 131(10): 2433-44, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22377908

RESUMEN

An apurinic/apyrimidinic (AP) site is an obligatory cytotoxic intermediate in DNA Base Excision Repair (BER) that is processed by human AP endonuclease 1 (APE1). APE1 is essential for BER and an emerging drug target in cancer. We have isolated novel small molecule inhibitors of APE1. In this study, we have investigated the ability of APE1 inhibitors to induce synthetic lethality (SL) in a panel of DNA double-strand break (DSB) repair deficient and proficient cells; i) Chinese hamster (CH) cells: BRCA2 deficient (V-C8), ATM deficient (V-E5), wild type (V79) and BRCA2 revertant [V-C8(Rev1)]. ii) Human cancer cells: BRCA1 deficient (MDA-MB-436), BRCA1 proficient (MCF-7), BRCA2 deficient (CAPAN-1 and HeLa SilenciX cells), BRCA2 proficient (PANC1 and control SilenciX cells). We also tested SL in CH ovary cells expressing a dominant-negative form of APE1 (E8 cells) using ATM inhibitors and DNA-PKcs inhibitors (DSB inhibitors). APE1 inhibitors are synthetically lethal in BRCA and ATM deficient cells. APE1 inhibition resulted in accumulation of DNA DSBs and G2/M cell cycle arrest. SL was also demonstrated in CH cells expressing a dominant-negative form of APE1 treated with ATM or DNA-PKcs inhibitors. We conclude that APE1 is a promising SL target in cancer.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Animales , Proteína BRCA1/deficiencia , Proteína BRCA2/deficiencia , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cricetinae , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/toxicidad , Humanos
5.
PLoS One ; 6(5): e19659, 2011 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-21573016

RESUMEN

Nimustine (ACNU) and temozolomide (TMZ) are DNA alkylating agents which are commonly used in chemotherapy for glioblastomas. ACNU is a DNA cross-linking agent and TMZ is a methylating agent. The therapeutic efficacy of these agents is limited by the development of resistance. In this work, the role of the Fanconi anemia (FA) repair pathway for DNA damage induced by ACNU or TMZ was examined. Cultured mouse embryonic fibroblasts were used: FANCA(-/-), FANCC(-/-), FANCA(-/-)C(-/-), FANCD2(-/-) cells and their parental cells, and Chinese hamster ovary and lung fibroblast cells were used: FANCD1/BRCA2mt, FANCG(-/-) and their parental cells. Cell survival was examined after a 3 h ACNU or TMZ treatment by using colony formation assays. All FA repair pathways were involved in ACNU-induced DNA damage. However, FANCG and FANCD1/BRCA2 played notably important roles in the repair of TMZ-induced DNA damage. The most effective molecular target correlating with cellular sensitivity to both ACNU and TMZ was FANCD1/BRCA2. In addition, it was found that FANCD1/BRCA2 small interference RNA efficiently enhanced cellular sensitivity toward ACNU and TMZ in human glioblastoma A172 cells. These findings suggest that the down-regulation of FANCD1/BRCA2 might be an effective strategy to increase cellular chemo-sensitization towards ACNU and TMZ.


Asunto(s)
Proteína BRCA2/metabolismo , Daño del ADN , Reparación del ADN/efectos de los fármacos , Dacarbazina/análogos & derivados , Nimustina/farmacología , Animales , Línea Celular , Daño del ADN/genética , Reparación del ADN/genética , Dacarbazina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Anemia de Fanconi/genética , Silenciador del Gen/efectos de los fármacos , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Modelos Biológicos , ARN Interferente Pequeño/metabolismo , Recombinasa Rad51/metabolismo , Recombinación Genética/efectos de los fármacos , Recombinación Genética/genética , Temozolomida
6.
Biochem Biophys Res Commun ; 404(1): 206-10, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21111709

RESUMEN

The role of the Fanconi anemia (FA) repair pathway for DNA damage induced by formaldehyde was examined in the work described here. The following cell types were used: mouse embryonic fibroblast cell lines FANCA(-/-), FANCC(-/-), FANCA(-/-)C(-/-), FANCD2(-/-) and their parental cells, the Chinese hamster cell lines FANCD1 mutant (mt), FANCGmt, their revertant cells, and the corresponding wild-type (wt) cells. Cell survival rates were determined with colony formation assays after formaldehyde treatment. DNA double strand breaks (DSBs) were detected with an immunocytochemical γH2AX-staining assay. Although the sensitivity of FANCA(-/-), FANCC(-/-) and FANCA(-/-)C(-/-) cells to formaldehyde was comparable to that of proficient cells, FANCD1mt, FANCGmt and FANCD2(-/-) cells were more sensitive to formaldehyde than the corresponding proficient cells. It was found that homologous recombination (HR) repair was induced by formaldehyde. In addition, γH2AX foci in FANCD1mt cells persisted for longer times than in FANCD1wt cells. These findings suggest that formaldehyde-induced DSBs are repaired by HR through the FA repair pathway which is independent of the FA nuclear core complex.


Asunto(s)
Daño del ADN , Reparación del ADN/genética , ADN Recombinante , Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Animales , Proteína BRCA2/fisiología , Células CHO , Cricetinae , Cricetulus , Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Proteína del Grupo de Complementación C de la Anemia de Fanconi/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Formaldehído/toxicidad , Histonas/metabolismo , Ratones
7.
Mutat Res ; 689(1-2): 50-8, 2010 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-20471405

RESUMEN

Homologous recombination is essential for repair of DNA interstrand cross-links and double-strand breaks. The Rad51C protein is one of the five Rad51 paralogs in vertebrates implicated in homologous recombination. A previously described hamster cell mutant defective in Rad51C (CL-V4B) showed increased sensitivity to DNA damaging agents and displayed genomic instability. Here, we identified a splice donor mutation at position +5 of intron 5 of the Rad51C gene in this mutant, and generated mice harboring an analogous base pair alteration. Rad51C(splice) heterozygous animals are viable and do not display any phenotypic abnormalities, however homozygous Rad51C(splice) embryos die during early development (E8.5). Detailed analysis of two CL-V4B revertants, V4B-MR1 and V4B-MR2, that have reduced levels of full-length Rad51C transcript when compared to wild type hamster cells, showed increased sensitivity to mitomycin C (MMC) in clonogenic survival, suggesting haploinsufficiency of Rad51C. Similarly, mouse Rad51C(splice/neo) heterozygous ES cells also displayed increased MMC sensitivity. Moreover, in both hamster revertants, Rad51C haploinsufficiency gives rise to increased frequencies of spontaneous and MMC-induced chromosomal aberrations, impaired sister chromatid cohesion and reduced cloning efficiency. These results imply that adequate expression of Rad51C in mammalian cells is essential for maintaining genomic stability and sister chromatid cohesion to prevent malignant transformation.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Inestabilidad Genómica , Animales , Aberraciones Cromosómicas , Cricetinae , Cricetulus , Femenino , Haploidia , Ratones , Ratones Endogámicos C57BL , Mitomicina/farmacología , Mutación , Embarazo , Intercambio de Cromátides Hermanas
8.
Am J Hum Genet ; 86(2): 262-6, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20137776

RESUMEN

The iron-sulfur-containing DNA helicases XPD, FANCJ, DDX11, and RTEL represent a small subclass of superfamily 2 helicases. XPD and FANCJ have been connected to the genetic instability syndromes xeroderma pigmentosum and Fanconi anemia. Here, we report a human individual with biallelic mutations in DDX11. Defective DDX11 is associated with a unique cellular phenotype in which features of Fanconi anemia (drug-induced chromosomal breakage) and Roberts syndrome (sister chromatid cohesion defects) coexist. The DDX11-deficient patient represents another cohesinopathy, besides Cornelia de Lange syndrome and Roberts syndrome, and shows that DDX11 functions at the interface between DNA repair and sister chromatid cohesion.


Asunto(s)
Anomalías Múltiples/enzimología , Anomalías Múltiples/genética , Rotura Cromosómica , ARN Helicasas DEAD-box/genética , ADN Helicasas/genética , Mutación/genética , Intercambio de Cromátides Hermanas/genética , Xerodermia Pigmentosa/genética , Adolescente , Secuencia de Bases , Preescolar , ARN Helicasas DEAD-box/deficiencia , ADN Helicasas/deficiencia , Análisis Mutacional de ADN , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Neoplasias/genética , Linaje , Fenotipo , Polonia , Embarazo , Síndrome
9.
DNA Repair (Amst) ; 9(4): 365-73, 2010 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-20079696

RESUMEN

V(D)J rearrangement in lymphoid cells involves repair of double-strand breaks (DSBs) through non-homologous end joining (NHEJ). Defects in this process lead to increased radiosensitivity and severe combined immunodeficiency (RS-SCID). Here, a SCID patient, M3, is described with a T(-)B(+)NK(+) phenotype but without causative mutations in CD3delta, epsilon, zeta or IL7Ralpha, genes specifically involved in T cell development. Clonogenic survival of M3 fibroblasts showed an increased sensitivity to the DSB-inducing agents ionizing radiation and bleomycin, as well as the crosslinking compound, mitomycin C. We did not observe inactivating mutations in known NHEJ genes and results of various DSB-repair assays in G(1) M3 cells were indistinguishable from those obtained with normal cells. However, we found increased chromosomal radiosensitivity at the G(2) phase of the cell cycle. Checkpoint analysis indicated functional G(1)/S and intra-S checkpoints after irradiation but impaired activation of the "early" G(2)/M checkpoint. Together these results indicate a novel class of RS-SCID patients characterized by the specific absence of T lymphocytes and associated with defects in G(2)-specific DSB repair. The pronounced G(2)/M radiosensitivity of the RS-SCID patient described here, suggests a defect in a putative novel and uncharacterized factor involved in cellular DNA damage responses and T cell development.


Asunto(s)
División Celular/efectos de la radiación , Fase G2/efectos de la radiación , Tolerancia a Radiación/genética , Inmunodeficiencia Combinada Grave/genética , Línea Celular , Daño del ADN , Reordenamiento Génico , Humanos , Linfocitos T/metabolismo , VDJ Recombinasas/genética , VDJ Recombinasas/metabolismo
10.
DNA Repair (Amst) ; 8(1): 72-86, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18840549

RESUMEN

O(6)-methylguanine (O(6)MeG) is a highly critical DNA adduct induced by methylating carcinogens and anticancer drugs such as temozolomide, streptozotocine, procarbazine and dacarbazine. Induction of cell death by O(6)MeG lesions requires mismatch repair (MMR) and cell proliferation and is thought to be dependent on the formation of DNA double-strand breaks (DSBs) or, according to an alternative hypothesis, direct signaling by the MMR complex. Given a role for DSBs in this process, either homologous recombination (HR) or non-homologous end joining (NHEJ) or both might protect against O(6)MeG. Here, we compared the response of cells mutated in HR and NHEJ proteins to temozolomide and N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). The data show that cells defective in HR (Xrcc2 and Brca2 mutants) are extremely sensitive to cell death by apoptosis and chromosomal aberration formation and less sensitive to sister-chromatid exchange (SCE) induction than the corresponding wild-type. Cells defective in NHEJ were not (Ku80 mutant), or only slightly more sensitive (DNA-PK(cs) mutant) to cell death and showed similar aberration and SCE frequencies than the corresponding wild-type. Transfection of O(6)-methylguanine-DNA methyltransferase (MGMT) in all of the mutants almost completely abrogated the genotoxic effects in both HR and NHEJ defective cells, indicating the mutant-specific hypersensitivity was due to O(6)MeG lesions. MNNG provoked H2AX phosphorylation 24-48h after methylation both in wild-type and HR mutants, which was not found in MGMT transfected cells. The gammaH2AX foci formed in response to O(6)MeG declined later in wild-type but not in HR-defective cells. The data support a model where DSBs are formed in response to O(6)MeG in the post-treatment cell cycle, which are repaired by HR, but not NHEJ, in a process that leads to SCEs. Therefore, HR can be considered as a mechanism that causes tolerance of O(6)MeG adducts. The data implicate that down-regulation or inhibition of HR might be a powerful strategy in improving cancer therapy with methylating agents.


Asunto(s)
Apoptosis , Proteína BRCA2/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/genética , Guanina/análogos & derivados , Intercambio de Cromátides Hermanas/genética , Animales , Células CHO , Muerte Celular , Aberraciones Cromosómicas , Cricetinae , Cricetulus , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente , Guanina/metabolismo , Ratones , Mutación , O(6)-Metilguanina-ADN Metiltransferasa/genética , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Recombinación Genética , Temozolomida , Transfección
11.
J Clin Invest ; 119(1): 91-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19075392

RESUMEN

Radiosensitive T-B- severe combined immunodeficiency (RS-SCID) is caused by defects in the nonhomologous end-joining (NHEJ) DNA repair pathway, which results in failure of functional V(D)J recombination. Here we have identified the first human RS-SCID patient to our knowledge with a DNA-PKcs missense mutation (L3062R). The causative mutation did not affect the kinase activity or DNA end-binding capacity of DNA-PKcs itself; rather, the presence of long P-nucleotide stretches in the immunoglobulin coding joints indicated that it caused insufficient Artemis activation, something that is dependent on Artemis interaction with autophosphorylated DNA-PKcs. Moreover, overall end-joining activity was hampered, suggesting that Artemis-independent DNA-PKcs functions were also inhibited. This study demonstrates that the presence of DNA-PKcs kinase activity is not sufficient to rule out a defect in this gene during diagnosis and treatment of RS-SCID patients. Further, the data suggest that residual DNA-PKcs activity is indispensable in humans.


Asunto(s)
Proteína Quinasa Activada por ADN/genética , Mutación Missense , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Tolerancia a Radiación , Recombinación Genética , Inmunodeficiencia Combinada Grave/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Preescolar , ADN Ligasa (ATP) , ADN Ligasas/genética , ADN Ligasas/metabolismo , Análisis Mutacional de ADN , Reparación del ADN , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN , Endonucleasas , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Fibroblastos/efectos de la radiación , Genotipo , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Linaje , Alineación de Secuencia , Inmunodeficiencia Combinada Grave/diagnóstico
12.
Cancer Res ; 68(11): 4347-51, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519695

RESUMEN

Artesunate is a semisynthetic derivative from artemisinin, a natural product from the Chinese herb Artemisia annua L. It exerts antimalarial activity, and, additionally, artemisinin and its derivatives are active against cancer cells. The active moiety is an endoperoxide bridge. Its cleavage leads to the formation of reactive oxygen species and carbon-centered radicals. These highly reactive molecules target several proteins in Plasmodia, which is thought to result in killing of the microorganism. DNA damage induced by artemisinins has not yet been described. Here, we show that artesunate induces apoptosis and necrosis. It also induces DNA breakage in a dose-dependent manner as shown by single-cell gel electrophoresis. This genotoxic effect was confirmed by measuring the level of gamma-H2AX, which is considered to be an indication of DNA double-strand breaks (DSB). Polymerase beta-deficient cells were more sensitive than the wild-type to artesunate, indicating that the drug induces DNA damage that is repaired by base excision repair. irs1 and VC8 cells defective in homologous recombination (HR) due to inactivation of XRCC2 and BRCA2, respectively, were more sensitive to artesunate than the corresponding wild-type. This was also true for XR-V15B cells defective in nonhomologous end-joining (NHEJ) due to inactivation of Ku80. The data indicate that DSBs induced by artesunate are repaired by the HR and NHEJ pathways. They suggest that DNA damage induced by artesunate contributes to its therapeutic effect against cancer cells.


Asunto(s)
Artemisininas/farmacología , Daño del ADN , Reparación del ADN , ADN/efectos de los fármacos , Medicina Tradicional China , Animales , Apoptosis , Artesunato , Western Blotting , Línea Celular , Ensayo Cometa , Cricetinae , Cricetulus , Técnica del Anticuerpo Fluorescente
13.
J Cell Sci ; 121(Pt 2): 162-6, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18089650

RESUMEN

Faithful genome transmission requires a network of pathways coordinating DNA replication to DNA repair and recombination. Here, we used molecular combing to measure the impact of homologous recombination (HR) on the velocity of DNA replication forks. We used three hamster cell lines defective in HR either by overexpression of a RAD51 dominant-negative form, or by a defect in the RAD51 paralogue XRCC2 or the breast tumor suppressor BRCA2. Irrespectively of the type or extent of HR alteration, all three cell lines exhibited a similar reduction in the rate of replication-fork progression, associated with an increase in the density of replication forks. Importantly, this phenotype was completely reversed in complemented derivatives of Xrcc2 and Brca2 mutants. These data reveal a novel role for HR, different from the reactivation of stalled replication forks, which may play an important role in genome stability and thus in tumor protection.


Asunto(s)
Reparación del ADN , Replicación del ADN , Recombinación Genética , Animales , Proteína BRCA2/biosíntesis , Línea Celular , Línea Celular Tumoral , Separación Celular , Cricetinae , Proteínas de Unión al ADN/metabolismo , Citometría de Flujo , Humanos , Modelos Biológicos , Modelos Genéticos , Recombinasa Rad51/biosíntesis
14.
Mutat Res ; 615(1-2): 111-24, 2007 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-17169382

RESUMEN

We analyzed the phenotype of cells derived from SCID patients with different mutations in the Artemis gene. Using clonogenic survival assay an increased sensitivity was found to X-rays (2-3-fold) and bleomycin (2-fold), as well as to etoposide, camptothecin and methylmethane sulphonate (up to 1.5-fold). In contrast, we did not find increased sensitivity to cross-linking agents mitomycin C and cis-platinum. The kinetics of DSB repair assessed by pulsed-field gel electrophoresis and gammaH2AX foci formation after ionizing irradiation, indicate that 15-20% of DSB are not repaired in Artemis-deficient cells. In order to get a better understanding of the repair defect in Artemis-deficient cells, we studied chromosomal damage at different stages of the cell cycle. In contrast to AT cells, Artemis-deficient cells appear to have a normal G(1)/S-block that resulted in a similar frequency of dicentrics and translocations, however, frequency of acentrics fragments was found to be 2-4-fold higher compared to normal fibroblasts. Irradiation in G(2) resulted in a higher frequency of chromatid-type aberrations (1.5-3-fold) than in normal cells, indicating that a fraction of DSB requires Artemis for proper repair. Our data are consistent with a function of Artemis protein in processing of a subset of complex DSB, without G(1) cell cycle checkpoint defects. This type of DSB can be induced in high proportion and persist through S-phase and in part might be responsible for the formation of chromatid-type exchanges in G(1)-irradiated Artemis-deficient cells. Among different human radiosensitive fibroblasts studied for endogenous (in untreated samples) as well as X-ray-induced DNA damage, the ranking order on the basis of higher incidence of spontaneously occurring chromosomal alterations and induced ones was: ligase 4> or =AT>Artemis. This observation implicates that in human fibroblasts following exposure to ionizing radiation a lower risk might be created when cells are devoid of endogenous damage.


Asunto(s)
Reparación del ADN/genética , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Línea Celular , Células Cultivadas , Inestabilidad Cromosómica/efectos de los fármacos , Inestabilidad Cromosómica/efectos de la radiación , Aberraciones Cromosómicas , Ensayo de Unidades Formadoras de Colonias , Roturas del ADN de Doble Cadena , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN , Endonucleasas , Humanos , Hibridación Fluorescente in Situ , Mutágenos/toxicidad , Fenotipo , Tolerancia a Radiación , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/metabolismo , Inmunodeficiencia Combinada Grave/patología
15.
Cancer Res ; 66(16): 8109-15, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16912188

RESUMEN

Deficiency in either of the breast cancer susceptibility proteins BRCA1 or BRCA2 induces profound cellular sensitivity to the inhibition of poly(ADP-ribose) polymerase (PARP) activity. We hypothesized that the critical role of BRCA1 and BRCA2 in the repair of double-strand breaks by homologous recombination (HR) was the underlying reason for this sensitivity. Here, we examine the effects of deficiency of several proteins involved in HR on sensitivity to PARP inhibition. We show that deficiency of RAD51, RAD54, DSS1, RPA1, NBS1, ATR, ATM, CHK1, CHK2, FANCD2, FANCA, or FANCC induces such sensitivity. This suggests that BRCA-deficient cells are, at least in part, sensitive to PARP inhibition because of HR deficiency. These results indicate that PARP inhibition might be a useful therapeutic strategy not only for the treatment of BRCA mutation-associated tumors but also for the treatment of a wider range of tumors bearing a variety of deficiencies in the HR pathway or displaying properties of 'BRCAness.'


Asunto(s)
Daño del ADN , Reparación del ADN , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Recombinación Genética , Animales , Proteína BRCA1/genética , Proteína BRCA2/genética , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Fluorobencenos/farmacología , Ratones , Ratones Noqueados , Ftalazinas/farmacología , Células Madre/citología
16.
Mutat Res ; 601(1-2): 191-201, 2006 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16920162

RESUMEN

Fanconi anemia (FA) is an inherited cancer-susceptibility disorder, characterized by genomic instability and hypersensitivity to DNA cross-linking agents. The discovery of biallelic BRCA2 mutations in the FA-D1 complementation group allows for the first time to study the characteristics of primary BRCA2-deficient human cells. FANCD1/BRCA2-deficient fibroblasts appeared hypersensitive to mitomycin C (MMC), slightly sensitive to methyl methane sulfonate (MMS), and like cells derived from other FA complementation groups, not sensitive to X-ray irradiation. However, unlike other FA cells, FA-D1 cells were slightly sensitive to UV irradiation. Despite the observed lack of X-ray sensitivity in cell survival, significant radioresistant DNA synthesis (RDS) was observed in the BRCA2-deficient fibroblasts but also in the FANCA-deficient fibroblasts, suggesting an impaired S-phase checkpoint. FA-D1/BRCA2 cells displayed greatly enhanced levels of spontaneous as well as MMC-induced chromosomal aberrations (CA), similar to cells deficient in homologous recombination (HR) and non-D1 FA cells. In contrast to Brca2-deficient rodent cells, FA-D1/BRCA2 cells showed normal sister chromatid exchange (SCE) levels, both spontaneous as well as after MMC treatment. Hence, these data indicate that human cells with biallelic BRCA2 mutations display typical features of both FA- and HR-deficient cells, which suggests that FANCD1/BRCA2 is part of the integrated FA/BRCA DNA damage response pathway but also controls other functions outside the FA pathway.


Asunto(s)
Proteína BRCA2/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Fibroblastos/metabolismo , Bleomicina/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Aberraciones Cromosómicas/efectos de los fármacos , Aberraciones Cromosómicas/efectos de la radiación , Daño del ADN/genética , Reparación del ADN/genética , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Humanos , Metilmetanosulfonato/farmacología , Mitomicina/farmacología , Intercambio de Cromátides Hermanas/efectos de los fármacos , Intercambio de Cromátides Hermanas/efectos de la radiación
17.
Proc Natl Acad Sci U S A ; 103(23): 8768-73, 2006 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-16731627

RESUMEN

The BRCA2 tumor suppressor plays an important role in the repair of DNA damage by homologous recombination, also termed homology-directed repair (HDR). Human BRCA2 is 3,418 aa and is composed of several domains. The central part of the protein contains multiple copies of a motif that binds the Rad51 recombinase (the BRC repeat), and the C terminus contains domains that have structural similarity to domains in the ssDNA-binding protein replication protein A (RPA). To gain insight into the role of BRCA2 in the repair of DNA damage, we fused a single (BRC3, BRC4) or multiple BRC motifs to the large RPA subunit. Expression of any of these protein fusions in Brca2 mutant cells substantially improved HDR while suppressing mutagenic repair. A fusion containing a Rad52 ssDNA-binding domain also was active in HDR. Mutations that reduced ssDNA or Rad51 binding impaired the ability of the fusion proteins to function in HDR. The high level of spontaneous chromosomal aberrations in Brca2 mutant cells was largely suppressed by the BRC-RPA fusion proteins, supporting the notion that the primary role of BRCA2 in maintaining genomic integrity is in HDR, specifically to deliver Rad51 to ssDNA. The fusion proteins also restored Rad51 focus formation and cellular survival in response to DNA damaging agents. Because as little as 2% of BRCA2 fused to RPA is sufficient to suppress cellular defects found in Brca2-mutant mammalian cells, these results provide insight into the recently discovered diversity of BRCA2 domain structures in different organisms.


Asunto(s)
Proteína BRCA2/deficiencia , Proteína BRCA2/metabolismo , Reparación del ADN , Proteínas Recombinantes de Fusión/metabolismo , Animales , Proteína BRCA2/química , Aberraciones Cromosómicas , Cricetinae , Daño del ADN/genética , ADN de Cadena Simple/metabolismo , Expresión Génica , Humanos , Ratones , Fenotipo , Unión Proteica , Recombinasa Rad51/metabolismo , Recombinación Genética , Proteína de Replicación A/metabolismo
18.
Methods Mol Biol ; 314: 1-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16673869

RESUMEN

Mutant rodent cell lines hypersensitive to DNA-damaging agents have provided a useful tool for the characterization of DNA repair pathways and have contributed to a better understanding of the mechanisms involved in the cellular responses to mutagenic treatment. Here we present a detailed description of how to isolate mutagen-sensitive mutants from hamster "wild-type" cell lines. First, cells are treated with ethyl nitrosourea, and then the mutagenized cell populations are screened for cells with an increased sensitivity to various mutagens using a replica-plating method. Mutagen-sensitive clones are identified and then characterized by assessing their stability, degree of sensitivity to various mutagens, and by genetic complementation analysis.


Asunto(s)
Células CHO , Pruebas de Mutagenicidad , Animales , Cricetinae , Cricetulus , Resistencia a Medicamentos/genética , Mutagénesis , Mutágenos/toxicidad , Mutación , Tolerancia a Radiación/genética , Radiación Ionizante
19.
Methods Mol Biol ; 314: 51-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16673873

RESUMEN

Inhibition of replicative DNA synthesis by ionizing radiation is partly caused by an active, signal-mediated response termed the "S-phase checkpoint." Defects in this checkpoint were first discovered in the human inherited disorder ataxia-telangiectasia (AT). gamma-Irradiated cells from AT patients consistently display a diminished inhibition of DNA synthesis, a feature called "radioresistant DNA synthesis" (RDS). RDS has been widely used as a diagnostic marker for AT, in postnatal as well as prenatal material. The regulation and control of the S-phase checkpoint is complex and multifaceted; it is not restricted to ionizing radiation, but can occur after many genotoxic stressors. Defects in both upstream control functions, such as ATM, NBS1, and MRE11, as well as downstream modulators can provoke an RDS phenotype. Here a simple, accurate and highly reproducible experimental protocol is presented for the generation of DNA synthesis inhibition curves from cells in culture.


Asunto(s)
Ataxia Telangiectasia/diagnóstico , Replicación del ADN/efectos de la radiación , Rayos gamma , Síndrome de Nijmegen/diagnóstico , Fase S/efectos de la radiación , Animales , Biomarcadores/análisis , Células Cultivadas , Humanos , Marcaje Isotópico , Timidina/análisis , Timidina/metabolismo
20.
Mutat Res ; 600(1-2): 79-88, 2006 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-16643964

RESUMEN

The previously described Chinese hamster cell mutant V-C8 that is defective in Brca2 shows a very complex phenotype, including increased sensitivity towards a wide variety of DNA damaging agents, chromosomal instability, abnormal centrosomes and impaired formation of Rad51 foci in response to DNA damage. Here, we demonstrate that V-C8 cells display biallelic nonsense mutations in Brca2, one in exon 15 and the other in exon 16, both resulting in truncated Brca2 proteins. We generated several independent mitomycin C (MMC)-resistant clones from V-C8 cells that had acquired an additional mutation leading to the restoration of the open reading frame of one of the Brca2 alleles. In two of these revertants, V-C8-Rev 1 and V-C8-Rev 6, the reversions lead to the wild-type Brca2 sequence. The V-C8 revertants did not gain the entire wild-type phenotype and still show a 2.5-fold increased sensitivity to mitomycin C (MMC), higher levels of spontaneous and MMC-induced chromosomal aberrations, as well as abnormal centrosomes when compared to wild-type cells. Our results suggest that Brca2 heterozygosity in hamster cells primarily gives rise to sensitivity to DNA cross-linking agents, especially chromosomal instability, a feature that might also be displayed in BRCA2 heterozygous mutation carriers.


Asunto(s)
Línea Celular , Inestabilidad Cromosómica , Codón sin Sentido , Cricetulus/genética , Genes BRCA2 , Alelos , Secuencia de Aminoácidos , Animales , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Centrosoma/metabolismo , Aberraciones Cromosómicas/efectos de los fármacos , Codón de Terminación , Cricetinae , Reactivos de Enlaces Cruzados/farmacología , Femenino , Heterocigoto , Modelos Genéticos , Datos de Secuencia Molecular , Fenotipo , Recombinasa Rad51/metabolismo , Intercambio de Cromátides Hermanas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...