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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 61(4): 535-546, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26833090

RESUMEN

XPG is a structure-specific endonuclease required for nucleotide excision repair, and incision-defective XPG mutations cause the skin cancer-prone syndrome xeroderma pigmentosum. Truncating mutations instead cause the neurodevelopmental progeroid disorder Cockayne syndrome, but little is known about how XPG loss results in this devastating disease. We identify XPG as a partner of BRCA1 and BRCA2 in maintaining genomic stability through homologous recombination (HRR). XPG depletion causes DNA double-strand breaks, chromosomal abnormalities, cell-cycle delays, defective HRR, inability to overcome replication fork stalling, and replication stress. XPG directly interacts with BRCA2, RAD51, and PALB2, and XPG depletion reduces their chromatin binding and subsequent RAD51 foci formation. Upstream in HRR, XPG interacts directly with BRCA1. Its depletion causes BRCA1 hyper-phosphorylation and persistent chromatin binding. These unexpected findings establish XPG as an HRR protein with important roles in genome stability and suggest how XPG defects produce severe clinical consequences including cancer and accelerated aging.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Síndrome de Cockayne/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Inestabilidad Genómica , Recombinación Homóloga , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Línea Celular Tumoral , Síndrome de Cockayne/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Genoma Humano , Células HeLa , Humanos , Ratones , Proteínas Nucleares/metabolismo , Fosforilación , Recombinasa Rad51/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
2.
Hum Mol Genet ; 28(24): 4148-4160, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31630195

RESUMEN

Whilst heterozygous germline mutations in the ABRAXAS1 gene have been associated with a hereditary predisposition to breast cancer, their effect on promoting tumourigenesis at the cellular level has not been explored. Here, we demonstrate in patient-derived cells that the Finnish ABRAXAS1 founder mutation (c.1082G > A, Arg361Gln), even in the heterozygous state leads to decreased BRCA1 protein levels as well as reduced nuclear localization and foci formation of BRCA1 and CtIP. This causes disturbances in basal BRCA1-A complex localization, which is reflected by a restraint in error-prone DNA double-strand break repair pathway usage, attenuated DNA damage response and deregulated G2-M checkpoint control. The current study clearly demonstrates how the Finnish ABRAXAS1 founder mutation acts in a dominant-negative manner on BRCA1 to promote genome destabilization in heterozygous carrier cells.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Neoplasias de la Mama/genética , Proteínas Portadoras/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Mutación de Línea Germinal , Adulto , Puntos de Control del Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Femenino , Genes BRCA1 , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Proteínas Supresoras de Tumor/genética
3.
Mol Ther ; 24(1): 117-24, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26265251

RESUMEN

Over 90% of patients with Nijmegen breakage syndrome (NBS), a hereditary cancer disorder, are homoallelic for a 5 bp deletion in the NBN gene involved in the cellular response to DNA damage. This hypomorphic mutation leads to a carboxy-terminal protein fragment, p70-nibrin, with some residual function. Average age at malignancy, typically lymphoma, is 9.7 years. NBS patients are hypersensitive to chemotherapeutic and radiotherapeutic treatments, thus prevention of cancer development is of particular importance. Expression of an internally deleted NBN protein, p80-nibrin, has been previously shown to be associated with a milder cellular phenotype and absence of cancer in a 62-year-old NBS patient. Here we show that cells from this patient, unlike other NBS patients, have DNA replication and origin firing rates comparable to control cells. We used here antisense oligonucleotides to enforce alternative splicing in NBS patient cells and efficiently generate the same internally deleted p80-nibrin protein. Injecting the same antisense sequences as morpholino oligomers (VivoMorpholinos) into the tail vein of a humanized NBS murine mouse model also led to efficient alternative splicing in vivo. Thus, proof of principle for the use of antisense oligonucleotides as a potential cancer prophylaxis has been demonstrated.


Asunto(s)
Empalme Alternativo , Proteínas de Ciclo Celular/genética , Síndrome de Nijmegen/terapia , Proteínas Nucleares/genética , Oligonucleótidos Antisentido/administración & dosificación , Eliminación de Secuencia , Empalme Alternativo/efectos de los fármacos , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular , Niño , Replicación del ADN , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Persona de Mediana Edad , Síndrome de Nijmegen/genética , Proteínas Nucleares/antagonistas & inhibidores , Oligonucleótidos Antisentido/farmacología
4.
Nucleic Acids Res ; 43(20): 9817-34, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26323318

RESUMEN

NUCKS1 (nuclear casein kinase and cyclin-dependent kinase substrate 1) is a 27 kD chromosomal, vertebrate-specific protein, for which limited functional data exist. Here, we demonstrate that NUCKS1 shares extensive sequence homology with RAD51AP1 (RAD51 associated protein 1), suggesting that these two proteins are paralogs. Similar to the phenotypic effects of RAD51AP1 knockdown, we find that depletion of NUCKS1 in human cells impairs DNA repair by homologous recombination (HR) and chromosome stability. Depletion of NUCKS1 also results in greatly increased cellular sensitivity to mitomycin C (MMC), and in increased levels of spontaneous and MMC-induced chromatid breaks. NUCKS1 is critical to maintaining wild type HR capacity, and, as observed for a number of proteins involved in the HR pathway, functional loss of NUCKS1 leads to a slow down in DNA replication fork progression with a concomitant increase in the utilization of new replication origins. Interestingly, recombinant NUCKS1 shares the same DNA binding preference as RAD51AP1, but binds to DNA with reduced affinity when compared to RAD51AP1. Our results show that NUCKS1 is a chromatin-associated protein with a role in the DNA damage response and in HR, a DNA repair pathway critical for tumor suppression.


Asunto(s)
Inestabilidad Genómica , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Reparación del ADN por Recombinación , Línea Celular , Cromatina/metabolismo , Aberraciones Cromosómicas , ADN/metabolismo , Daño del ADN , Replicación del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/fisiología , Células HeLa/fisiología , Humanos , Mitomicina/farmacología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/efectos de la radiación , Proteínas de Unión al ARN , Recombinasa Rad51/metabolismo , Fase S/efectos de la radiación , Homología de Secuencia de Aminoácido , Rayos X
5.
Recent Results Cancer Res ; 198: 1-24, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27318679

RESUMEN

Cellular chromosomal DNA is the principal target through which ionising radiation exerts it diverse biological effects. This chapter summarises the relevant DNA damage signalling and repair pathways used by normal and tumour cells in response to irradiation. Strategies for tumour radiosensitisation are reviewed which exploit tumour-specific DNA repair deficiencies or signalling pathway addictions, with a special focus on growth factor signalling, PARP, cancer stem cells, cell cycle checkpoints and DNA replication. This chapter concludes with a discussion of DNA repair-related candidate biomarkers of tumour response which are of crucial importance for implementing precision medicine in radiation oncology.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , Daño del ADN , Reparación del ADN , Neoplasias/radioterapia , Replicación del ADN/genética , Replicación del ADN/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Redes Reguladoras de Genes/efectos de la radiación , Humanos , Modelos Genéticos , Neoplasias/genética , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación
6.
Cancers (Basel) ; 14(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35804930

RESUMEN

The standard of care for advanced head and neck cancers (HNSCCs) is radiochemotherapy, including cisplatin. This treatment results in a cure rate of approximately 85% for oropharyngeal HPV-positive HNSCCs, in contrast to only 50% for HPV-negative HNSCCs, and is accompanied by severe side effects for both entities. Therefore, innovative treatment modalities are required, resulting in a better outcome for HPV-negative HNSCCs, and lowering the adverse effects for both entities. The effect of the dual PI3K/mTOR inhibitor NVP-BEZ235 on a combined treatment with cisplatin and radiation was studied in six HPV-negative and six HPV-positive HNSCC cell lines. Cisplatin alone was slightly more effective in HPV-positive cells. This could be attributed to a defect in homologous recombination, as demonstrated by depleting RAD51. Solely for HPV-positive cells, pretreatment with BEZ235 resulted in enhanced cisplatin sensitivity. For the combination of cisplatin and radiation, additive effects were observed. However, when pretreated with BEZ235, this combination changed into a synergistic interaction, with a slightly stronger enhancement for HPV-positive cells. This increase could be attributed to a diminished degree of DSB repair in G1, as visualized via the detection of γH2AX/53BP1 foci. BEZ235 can be used to enhance the effect of combined treatment with cisplatin and radiation in both HPV-negative and -positive HNSCCs.

7.
J Innate Immun ; 14(5): 461-476, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35086104

RESUMEN

Neutrophil extracellular traps (NETs) have been described as a potential trigger of severe COVID-19. NETs are known as extracellular DNA fibers released by neutrophils in response to infection. If the host is unable to balance efficient clearance of NETs by dornases (DNases), detrimental consequences occur. Elevated levels of NETs in COVID-19 patients are associated with higher risk of morbid thrombotic complications. Here, we studied the level of NET markers and DNase activity in a cohort of COVID-19 patients compared to healthy controls. Our data confirmed an increased level of NET markers in the plasma of COVID-19 patients, with a higher level in male compared to female patients. At the same time, there was an increased DNase activity detectable in COVID-19 patients compared to healthy controls. Importantly, there was a negative correlation of DNase activity with the age of male patients. The antimicrobial peptide LL-37, which is known to stabilize NETs against DNase degradation, is embedded in NETs upon severe acute respiratory syndrome coronavirus-2-infection. The LL-37 plasma level correlates with the NET-marker level in male COVID-19 patients, indicating a potential role of LL-37 in the risk of NET-associated thrombosis in male COVID-19 patients by stabilizing NETs against DNase degradation. In conclusion, our data identify two potential risk factors of elderly male patients which may lead to inefficient NET degradation and a subsequently higher risk of NET-associated thrombosis during COVID-19: reduced DNase activity and an increased LL-37 level.


Asunto(s)
COVID-19 , Trampas Extracelulares , Trombosis , Anciano , Desoxirribonucleasa I/metabolismo , Trampas Extracelulares/metabolismo , Femenino , Humanos , Masculino , Neutrófilos/metabolismo
8.
Neurooncol Adv ; 4(1): vdab180, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35274102

RESUMEN

Background: The oncogene epidermal growth factor receptor variant III (EGFRvIII) is expressed in approximately one-third of all glioblastomas (GBMs). So far it is not clear if EGFRvIII expression induces replication stress in GBM cells, which might serve as a therapeutical target. Methods: Isogenetic EGFRvIII- and EGFRvIII+ cell lines with endogenous EGFRvIII expression were used. Markers of oncogenic and replication stress such as γH2AX, RPA, 53BP1, ATR, and CHK1 were analyzed using western blot, immunofluorescence, and flow cytometry. The DNA fiber assay was performed to analyze replication, transcription was measured by incorporation of EU, and genomic instability was investigated by micronuclei and CGH-Array analysis. Immunohistochemistry staining was used to detect replication stress markers and R-loops in human GBM samples. Results: EGFRvIII+ cells exhibit an activated replication stress response, increased spontaneous DNA damage, elevated levels of single-stranded DNA, and reduced DNA replication velocity, which are all indicative characteristics of replication stress. Furthermore, we show here that EGFRvIII expression is linked to increased genomic instability. EGFRvIII-expressing cells display elevated RNA synthesis and R-loop formation, which could also be confirmed in EGFRvIII-positive GBM patient samples. Targeting replication stress by irinotecan resulted in increased sensitivity of EGFRvIII+ cells. Conclusion: This study demonstrates that EGFRvIII expression is associated with increased replication stress, R-loop accumulation, and genomic instability. This might contribute to intratumoral heterogeneity but may also be exploited for individualized therapy approaches.

9.
Emerg Microbes Infect ; 10(1): 1807-1818, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34402750

RESUMEN

Male sex was repeatedly identified as a risk factor for death and intensive care admission. However, it is yet unclear whether sex hormones are associated with disease severity in COVID-19 patients. In this study, we analysed sex hormone levels (estradiol and testosterone) of male and female COVID-19 patients (n = 50) admitted to an intensive care unit (ICU) in comparison to control non-COVID-19 patients at the ICU (n = 42), non-COVID-19 patients with the most prevalent comorbidity (coronary heart diseases) present within the COVID-19 cohort (n = 39) and healthy individuals (n = 50). We detected significantly elevated estradiol levels in critically ill male COVID-19 patients compared to all control cohorts. Testosterone levels were significantly reduced in critically ill male COVID-19 patients compared to control cohorts. No statistically significant differences in sex hormone levels were detected in critically ill female COVID-19 patients, albeit similar trends towards elevated estradiol levels were observed. Linear regression analysis revealed that among a broad range of cytokines and chemokines analysed, IFN-γ levels are positively associated with estradiol levels in male and female COVID-19 patients. Furthermore, male COVID-19 patients with elevated estradiol levels were more likely to receive ECMO treatment. Thus, we herein identified that disturbance of sex hormone metabolism might present a hallmark in critically ill male COVID-19 patients.


Asunto(s)
COVID-19/mortalidad , COVID-19/patología , Estradiol/sangre , Testosterona/sangre , Anciano , Anciano de 80 o más Años , COVID-19/sangre , Cuidados Críticos , Enfermedad Crítica , Oxigenación por Membrana Extracorpórea , Femenino , Humanos , Hipogonadismo/patología , Unidades de Cuidados Intensivos , Interferón gamma/sangre , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Distribución por Sexo
10.
Cancers (Basel) ; 12(10)2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003585

RESUMEN

Chromosomal instability (CIN) is an emerging hallmark of cancer and its role in therapeutic responses has been increasingly attracting the attention of the research community. To target the vulnerability of tumors with high CIN, it is important to identify the genes and mechanisms involved in the maintenance of CIN. In our work, we recognize the tumor suppressor gene Phosphatase and Tensin homolog (PTEN) as a potential gene causing CIN in triple-negative breast cancer (TNBC) and show that TNBC with low expression levels of PTEN can be sensitized for the treatment with poly-(ADP-ribose)-polymerase 1 (PARP1) inhibitors, independent of Breast Cancer (BRCA) mutations or a BRCA-like phenotype. In silico analysis of mRNA expression data from 200 TNBC patients revealed low expression of PTEN in tumors with a high CIN70 score. Western blot analysis of TNBC cell lines confirm lower protein expression of PTEN compared to non TNBC cell lines. Further, PTEN-deficient cell lines showed cellular sensitivity towards PARP1 inhibition treatment. DNA fiber assays and examination of chromatin bound protein fractions indicate a protective role of PTEN at stalled replication forks. In this study, we recognize PTEN as a potential CIN-causing gene in TNBC and identify its important role in the replication processes.

11.
Cells ; 9(1)2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31963582

RESUMEN

Chromosomal instability not only has a negative effect on survival in triple-negative breast cancer, but also on the well treatable subgroup of luminal A tumors. This suggests a general mechanism independent of subtypes. Increased chromosomal instability (CIN) in triple-negative breast cancer (TNBC) is attributed to a defect in the DNA repair pathway homologous recombination. Homologous recombination (HR) prevents genomic instability by repair and protection of replication. It is unclear whether genetic alterations actually lead to a repair defect or whether superior signaling pathways are of greater importance. Previous studies focused exclusively on the repair function of HR. Here, we show that the regulation of HR by the intra-S-phase damage response at the replication is of overriding importance. A damage response activated by Ataxia telangiectasia and Rad3 related-checkpoint kinase 1 (ATR-CHK1) can prevent replication stress and leads to resistance formation. CHK1 thus has a preferred role over HR in preventing replication stress in TNBC. The signaling cascade ATR-CHK1 can compensate for a double-strand break repair error and lead to resistance of HR-deficient tumors. Established methods for the identification of HR-deficient tumors for Poly(ADP-Ribose)-Polymerase 1 (PARP1) inhibitor therapies should be extended to include analysis of candidates for intra-S phase damage response.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Resistencia a Antineoplásicos/genética , Inestabilidad Genómica/genética , Recombinación Homóloga/genética , Reparación del ADN por Recombinación/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Alquilantes/farmacología , Antibióticos Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Bases de Datos Genéticas , Femenino , Inestabilidad Genómica/efectos de los fármacos , Recombinación Homóloga/efectos de los fármacos , Humanos , Microscopía Electrónica de Transmisión , Mitomicina/farmacología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación/efectos de los fármacos , Transducción de Señal/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/mortalidad , Neoplasias de la Mama Triple Negativas/patología
12.
Sci Rep ; 8(1): 16238, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389969

RESUMEN

Multi drug resistance protein 2 knockout mice (Mdr2-/-) are a mouse model of chronic liver inflammation and inflammation-induced tumour development. Here we investigated the kinetics of early heme oxygenase 1 (HO-1) induction on inflammation, tumour development, and DNA damage in Mdr2-/- mice. HO-1 was induced by intraperitoneal injection of cobalt protoporphyrin IX (CoPP) twice weekly for 9 consecutive weeks. Immediately after HO-1 induction, liver function improved and infiltration of CD4+ and CD8+ T cells was reduced. Furthermore, we observed increased p38 activation with concomitant reduction of Cyclin D1 expression in aged Mdr2-/- mice. Long-term effects of HO-1 induction included increased CD8+ T cell infiltration as well as delayed and reduced tumour growth in one-year-old animals. Unexpectedly, DNA double-strand breaks were detected predominantly in macrophages of 65-week-old Mdr2-/- mice, while DNA damage was reduced in response to early HO-1 induction in vivo and in vitro. Overall, early induction of HO-1 in Mdr2-/- mice had a beneficial short-term effect on liver function and reduced hepatic T cell accumulation. Long-term effects of early HO-1 induction were increased CD8+ T cell numbers, decreased proliferation as wells as reduced DNA damage in liver macrophages of aged animals, accompanied by delayed and reduced tumour growth.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Activadores de Enzimas/administración & dosificación , Hemo-Oxigenasa 1/metabolismo , Hepatitis/tratamiento farmacológico , Neoplasias Hepáticas/prevención & control , Proteínas de la Membrana/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Daño del ADN , Modelos Animales de Enfermedad , Femenino , Hepatitis/genética , Hepatitis/inmunología , Hepatitis/patología , Humanos , Inyecciones Intraperitoneales , Hígado/citología , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Protoporfirinas/administración & dosificación , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
13.
Methods Mol Biol ; 1510: 103-113, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27761816

RESUMEN

DNA fiber spreading assay is an invaluable technique to visualize and follow the spatial and temporal progress of individual DNA replication forks. It provides information on the DNA replication progress and its regulation under normal conditions as well as on replication stress induced by environmental genotoxic agents or cancer drugs. The method relies on the detection of incorporated thymidine analogues during DNA synthesis in the S phase of the cell cycle by indirect immunofluorescence. Here, we describe the procedure established in our laboratories for sequential pulse labeling of human cells with 5-chloro-2'-deoxyuridine (CldU) and 5-iodo-2'-deoxyuridine (IdU), cell lysis, and DNA fiber spreading on slides and sequential immunodetection of the incorporated thymidine analogues by primary antibodies recognizing specifically CldU or IdU alone. We describe also the laser scanning imaging, classification, and measurement of the detected DNA fiber tracks. The obtained quantitative data can be evaluated statistically to reveal the immediate or long-term effects of DNA-damaging agents, DNA repair inhibitors, and epigenetic modulators like HDAC inhibitors on DNA replication in normal and tumor cells.


Asunto(s)
Bioensayo , ADN/química , Desoxiuridina/análogos & derivados , Inhibidores de Histona Desacetilasas/farmacología , Idoxuridina/metabolismo , Coloración y Etiquetado/métodos , Anticuerpos/química , Benzamidas/farmacología , ADN/metabolismo , Replicación del ADN , Desoxiuridina/química , Desoxiuridina/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Células HCT116 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Idoxuridina/química , Microscopía Confocal , Piridinas/farmacología , Fase S
14.
Cell Cycle ; 15(7): 974-85, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26919204

RESUMEN

Cdc45 is an essential protein that together with Mcm2-7 and GINS forms the eukaryotic replicative helicase CMG. Cdc45 seems to be rate limiting for the initial unwinding or firing of replication origins. In line with this view, Cdc45-overexpressing cells fired at least twice as many origins as control cells. However, these cells displayed an about 2-fold diminished fork elongation rate, a pronounced asymmetry of replication fork extension, and an early S phase arrest. This was accompanied by H2AX-phosphorylation and subsequent apoptosis. Unexpectedly, we did not observe increased ATR/Chk1 signaling but rather a mild ATM/Chk2 response. In addition, we detected accumulation of long stretches of single-stranded DNA, a hallmark of replication catastrophe. We conclude that increased origin firing by upregulated Cdc45 caused exhaustion of the single-strand binding protein RPA, which in consequence diminished the ATR/Chk1 response; the subsequently occurring fork breaks led to an ATM/Chk2 mediated phosphorylation of H2AX and eventually to apoptosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Apoptosis , ADN de Cadena Simple/análisis , Células HeLa , Histonas/metabolismo , Humanos , Origen de Réplica , Puntos de Control de la Fase S del Ciclo Celular , Transducción de Señal
15.
Oncotarget ; 7(9): 9732-41, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26799421

RESUMEN

There is a need to develop new, more efficient therapies for head and neck cancer (HNSCC) patients. It is currently unclear whether defects in DNA repair genes play a role in HNSCCs' resistance to therapy. PARP1 inhibitors (PARPi) were found to be "synthetic lethal" in cancers deficient in BRCA1/2 with impaired homologous recombination. Since tumors rarely have these particular mutations, there is considerable interest in finding alternative determinants of PARPi sensitivity. Effectiveness of combined irradiation and PARPi olaparib was evaluated in ten HNSCC cell lines, subdivided into HR-proficient and HR-deficient cell lines using a GFP-based reporter assay. Both groups were equally sensitive to PARPi alone. Combined treatment revealed stronger synergistic interactions in the HR-deficient group. Because HR is mainly active in S-Phase, replication processes were analyzed. A stronger impact of treatment on replication processes (p = 0.04) and an increased number of radial chromosomes (p = 0.003) were observed in the HR-deficient group. We could show that radiosensitization by inhibition of PARP1 strongly correlates with HR competence in a replication-dependent manner. Our observations indicate that PARP1 inhibitors are promising candidates for enhancing the therapeutic ratio achieved by radiotherapy via disabling DNA replication processes in HR-deficient HNSCCs.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/terapia , Replicación del ADN/efectos de los fármacos , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/terapia , Recombinación Homóloga/genética , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Línea Celular Tumoral , Reparación del ADN/genética , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello
16.
Oncotarget ; 7(34): 55639-55648, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27489351

RESUMEN

Pro-inflammatory signaling pathways, especially interleukin 6 (IL-6), and reactive oxygen species (ROS) promote carcinogenesis in the liver. In order to elucidate the underlying oncogenic mechanism, we activated the IL-6 signal transducer glycoprotein 130 (gp130) via stable expression of a constitutively active gp130 construct (L-gp130) in untransformed telomerase-immortalized human fetal hepatocytes (FH-hTERT). As known from hepatocellular adenomas, forced gp130 activation alone was not sufficient to induce malignant transformation. However, additional challenge of FH-hTERT L-gp130 clones with oxidative stress resulted in 2- to 3-fold higher ROS levels and up to 6-fold more DNA-double strand breaks (DSB). Despite increased DNA damage, ROS-challenged FH-hTERT L-gp130 clones displayed an enhanced proliferation and rapidly developed colony growth capabilities in soft agar. As driving gp130-mediated oncogenic mechanism, we detected a decreased expression of antioxidant genes, in particular glutathione peroxidase 3 and apolipoprotein E, and an absence of P21 upregulation following ROS-conferred induction of DSB. In summary, an impaired oxidative stress response in hepatocytes with gp130 gain-of-function mutations, as detected in dysplastic intrahepatic nodules and hepatocellular adenomas, is one of the central oncogenic mechanisms in chronic liver inflammation.


Asunto(s)
Transformación Celular Neoplásica , Receptor gp130 de Citocinas/fisiología , Hepatocitos/patología , Neoplasias Hepáticas/etiología , Estrés Oxidativo , Animales , Roturas del ADN de Doble Cadena , Femenino , Células Hep G2 , Humanos , Ratones , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Telomerasa/genética
17.
Cell Cycle ; 14(19): 3190-202, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26317153

RESUMEN

In response to replication stress ATR signaling through CHK1 controls the intra-S checkpoint and is required for the maintenance of genomic integrity. Homologous recombination (HR) comprises a series of interrelated pathways that function in the repair of DNA double strand breaks and interstrand crosslinks. In addition, HR, with its key player RAD51, provides critical support for the recovery of stalled forks during replication. High levels of RAD51 are regularly found in various cancers, yet little is known about the effect of the increased RAD51 expression on intra-S checkpoint signaling. Here, we describe a role for RAD51 in driving genomic instability caused by impaired replication and intra-S mediated CHK1 signaling by studying an inducible RAD51 overexpression model as well as 10 breast cancer cell lines. We demonstrate that an excess of RAD51 decreases I-Sce-I mediated HR despite formation of more RAD51 foci. Cells with high RAD51 levels display reduced elongation rates and excessive dormant origin firing during undisturbed growth and after damage, likely caused by impaired CHK1 activation. In consequence, the inability of cells with a surplus of RAD51 to properly repair complex DNA damage and to resolve replication stress leads to higher genomic instability and thus drives tumorigenesis.


Asunto(s)
Proteínas Quinasas/metabolismo , Recombinasa Rad51/metabolismo , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Daño del ADN/fisiología , Reparación del ADN/genética , Reparación del ADN/fisiología , Replicación del ADN/genética , Replicación del ADN/fisiología , Inestabilidad Genómica/genética , Inestabilidad Genómica/fisiología , Recombinación Homóloga/genética , Recombinación Homóloga/fisiología , Humanos , Proteínas Quinasas/genética , Recombinasa Rad51/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
18.
DNA Repair (Amst) ; 24: 87-97, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25288561

RESUMEN

RAD51-associated protein 1 (RAD51AP1) is critical for homologous recombination (HR) by interacting with and stimulating the activities of the RAD51 and DMC1 recombinases. In human somatic cells, knockdown of RAD51AP1 results in increased sensitivity to DNA damaging agents and to impaired HR, but the formation of DNA damage-induced RAD51 foci is unaffected. Here, we generated a genetic model system, based on chicken DT40 cells, to assess the phenotype of fully inactivated RAD51AP1 in vertebrate cells. Targeted inactivation of both RAD51AP1 alleles has no effect on either viability or doubling-time in undamaged cells, but leads to increased levels of cytotoxicity after exposure to cisplatin or to ionizing radiation. Interestingly, ectopic expression of GgRAD51AP1, but not of HsRAD51AP1 is able to fully complement in cell survival assays. Notably, in RAD51AP1-deficient DT40 cells the resolution of DNA damage-induced RAD51 foci is greatly slowed down, while their formation is not impaired. We also identify, for the first time, an important role for RAD51AP1 in counteracting both spontaneous and DNA damage-induced replication stress. In human and in chicken cells, RAD51AP1 is required to maintain wild type speed of replication fork progression, and both RAD51AP1-depleted human cells and RAD51AP1-deficient DT40 cells respond to replication stress by a slow-down of replication fork elongation rates. However, increased firing of replication origins occurs in RAD51AP1-/- DT40 cells, likely to ensure the timely duplication of the entire genome. Taken together, our results may explain why RAD51AP1 commonly is overexpressed in tumor cells and tissues, and we speculate that the disruption of RAD51AP1 function could be a promising approach in targeted tumor therapy.


Asunto(s)
Replicación del ADN , Proteínas de Unión al ADN/genética , Secuencia de Aminoácidos , Animales , Línea Celular/efectos de los fármacos , Línea Celular/efectos de la radiación , Pollos , Cisplatino/farmacología , Daño del ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Humanos , Hidroxiurea/farmacología , Datos de Secuencia Molecular , Proteínas de Unión al ARN , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Radiación Ionizante , Vertebrados/genética
19.
Nat Commun ; 4: 2578, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24153426

RESUMEN

Besides mutations in BRCA1/BRCA2, heterozygous defects in PALB2 are important in breast cancer predisposition. PALB2 heterozygosity increases the risk of malignancy about sixfold. PALB2 interacts with BRCA1 and BRCA2 to regulate homologous recombination and mediate DNA damage response. Here we show, by analysing lymphoblastoid cell lines from heterozygous female PALB2 mutation carriers, that PALB2 haploinsufficiency causes aberrant DNA replication/damage response. Mutation carrier cells show increased origin firing and shorter distance between consecutive replication forks. Carrier cell lines also show elevated ATR protein, but not phosphorylation levels, and a majority of them display aberrant Chk1-/Chk2-mediated DNA damage response. Elevated chromosome instability is observed in primary blood lymphocytes of PALB2 mutation carriers, indicating that the described mechanisms of genome destabilization operate also at the organism level. These findings provide a new mechanism for early stages of breast cancer development that may also apply to other heterozygous homologous recombination signalling pathway gene mutations in hereditary cancer predisposition.


Asunto(s)
Neoplasias de la Mama/genética , Replicación del ADN , Regulación Neoplásica de la Expresión Génica , Heterocigoto , Mutación , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , Inestabilidad Cromosómica , Daño del ADN , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Humanos , Linfocitos/metabolismo , Linfocitos/patología , Proteínas Nucleares/metabolismo , Cultivo Primario de Células , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
20.
Radiother Oncol ; 102(3): 466-71, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22326574

RESUMEN

BACKGROUND: Replication-dependent radiosensitization of tumors ranks among the most promising tools for future improvements in tumor therapy. However, cell cycle checkpoint signaling during S phase is a key for maintaining genomic stability after ionizing irradiation allowing DNA damage repair by stabilizing replication forks, inhibiting new origin firing and recruiting DNA repair proteins. As the impact of the different types of DNA damage induced by ionizing radiation on replication fork functionality has not been investigated, this study was performed in tumor cells treated with various agents that induce specific DNA lesions. METHODS: U2OS cells were exposed to methyl methanesulfonate (MMS) to induce base damage, low or high concentrations of hydrogen peroxide for the induction of SSBs, Topotecan to induce DSBs at replication, Mitomycin C (MMC) to induce interstrand cross-links or ionizing irradiation to analyze all damages. Chk1 phosphorylation, origin firing and replication fork progression, and cell cycle distribution were analyzed. RESULTS: In our system, the extent of Chk1 phosphorylation was dependent on the type of damage induced and prolonged Chk1 phosphorylation correlated with the inhibition of replication initiation. Ionizing radiation, high concentrations of hydrogen peroxide, and Topotecan affected replication elongation much more strongly that the other agents. Almost all agents induced a slight increase in the S phase population but subsequent G2 arrest was only observed in response to those agents that strongly inhibited replication elongation and caused prolonged Chk1 phosphorylation. CONCLUSIONS: Our data suggest that to improve radiotherapy, radiosensitivity in S phase could be increased by combining irradiation with agents that induce secondary DSB or inhibit checkpoint signaling, such as inhibitors of PARP or Chk1.


Asunto(s)
Daño del ADN/efectos de la radiación , Replicación del ADN , Ciclo Celular/efectos de la radiación , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Roturas del ADN de Doble Cadena , Humanos , Neoplasias/patología , Neoplasias/radioterapia , Fosforilación , Proteínas Quinasas/metabolismo , Tolerancia a Radiación , Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA