Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(18): 9594-9609, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37702151

RESUMEN

The cohesin complex regulates higher order chromosome architecture through maintaining sister chromatid cohesion and folding chromatin by DNA loop extrusion. Impaired cohesin function underlies a heterogeneous group of genetic syndromes and is associated with cancer. Here, we mapped the genetic dependencies of human cell lines defective of cohesion regulators DDX11 and ESCO2. The obtained synthetic lethality networks are strongly enriched for genes involved in DNA replication and mitosis and support the existence of parallel sister chromatid cohesion establishment pathways. Among the hits, we identify the chromatin binding, BRCT-domain containing protein PAXIP1 as a novel cohesin regulator. Depletion of PAXIP1 severely aggravates cohesion defects in ESCO2 mutant cells, leading to mitotic cell death. PAXIP1 promotes global chromatin association of cohesin, independent of DNA replication, a function that cannot be explained by indirect effects of PAXIP1 on transcription or DNA repair. Cohesin regulation by PAXIP1 requires its binding partner PAGR1 and a conserved FDF motif in PAGR1. PAXIP1 co-localizes with cohesin on multiple genomic loci, including active gene promoters and enhancers. Possibly, this newly identified role of PAXIP1-PAGR1 in regulating cohesin occupancy on chromatin is also relevant for previously described functions of PAXIP1 in transcription, immune cell maturation and DNA repair.

2.
Nat Genet ; 39(2): 159-61, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17200672

RESUMEN

The Fanconi anemia and BRCA networks are considered interconnected, as BRCA2 gene defects have been discovered in individuals with Fanconi anemia subtype D1. Here we show that a defect in the BRCA2-interacting protein PALB2 is associated with Fanconi anemia in an individual with a new subtype. PALB2-deficient cells showed hypersensitivity to cross-linking agents and lacked chromatin-bound BRCA2; these defects were corrected upon ectopic expression of PALB2 or by spontaneous reversion.


Asunto(s)
Proteína BRCA2/fisiología , Neoplasias de la Mama/genética , Anemia de Fanconi/genética , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Predisposición Genética a la Enfermedad , Humanos , Mutación , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética
3.
Hum Mutat ; 36(5): 562-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25754594

RESUMEN

The diagnosis of VACTERL syndrome can be elusive, especially in the prenatal life, due to the presence of malformations that overlap those present in other genetic conditions, including the Fanconi anemia (FA). We report on three VACTERL cases within two families, where the two who arrived to be born died shortly after birth due to severe organs' malformations. The suspicion of VACTERL association was based on prenatal ultrasound assessment and postnatal features. Subsequent chromosome breakage analysis suggested the diagnosis of FA. Finally, by next-generation sequencing based on the analysis of the exome in one family and of a panel of Fanconi genes in the second one, we identified novel FANCL truncating mutations in both families. We used ectopic expression of wild-type FANCL to functionally correct the cellular FA phenotype for both mutations. Our study emphasizes that the diagnosis of FA should be considered when VACTERL association is suspected. Furthermore, we show that loss-of-function mutations in FANCL result in a severe clinical phenotype characterized by early postnatal death.


Asunto(s)
Canal Anal/anomalías , Esófago/anomalías , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/genética , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/genética , Riñón/anomalías , Deformidades Congénitas de las Extremidades/diagnóstico , Deformidades Congénitas de las Extremidades/genética , Mutación , Fenotipo , Columna Vertebral/anomalías , Tráquea/anomalías , Aborto Inducido , Rotura Cromosómica , Diagnóstico Diferencial , Exoma , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Recién Nacido , Nacimiento Vivo , Masculino , Embarazo , Diagnóstico Prenatal , Índice de Severidad de la Enfermedad
4.
Nat Genet ; 37(9): 934-5, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16116423

RESUMEN

The protein predicted to be defective in individuals with Fanconi anemia complementation group J (FA-J), FANCJ, is a missing component in the Fanconi anemia pathway of genome maintenance. Here we identify pathogenic mutations in eight individuals with FA-J in the gene encoding the DEAH-box DNA helicase BRIP1, also called FANCJ. This finding is compelling evidence that the Fanconi anemia pathway functions through a direct physical interaction with DNA.


Asunto(s)
Cromosomas Humanos Par 17 , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Anemia de Fanconi/genética , Mutación/genética , ARN Helicasas/deficiencia , ARN Helicasas/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Prueba de Complementación Genética , Humanos , Repeticiones de Microsatélite , Datos de Secuencia Molecular , Eliminación de Secuencia
5.
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
6.
Nat Genet ; 36(11): 1219-24, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15502827

RESUMEN

Fanconi anemia is an autosomal recessive syndrome characterized by diverse clinical symptoms, hypersensitivity to DNA crosslinking agents, chromosomal instability and susceptibility to cancer. Fanconi anemia has at least 11 complementation groups (A, B, C, D1, D2, E, F, G, I, J, L); the genes mutated in 8 of these have been identified. The gene BRCA2 was suggested to underlie complementation group B, but the evidence is inconclusive. Here we show that the protein defective in individuals with Fanconi anemia belonging to complementation group B is an essential component of the nuclear protein 'core complex' responsible for monoubiquitination of FANCD2, a key event in the DNA-damage response pathway associated with Fanconi anemia and BRCA. Unexpectedly, the gene encoding this protein, FANCB, is localized at Xp22.31 and subject to X-chromosome inactivation. X-linked inheritance has important consequences for genetic counseling of families with Fanconi anemia belonging to complementation group B. Its presence as a single active copy and essentiality for a functional Fanconi anemia-BRCA pathway make FANCB a potentially vulnerable component of the cellular machinery that maintains genomic integrity.


Asunto(s)
Cromosomas Humanos X , Anemia de Fanconi/genética , Metilación de ADN , Compensación de Dosificación (Genética) , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi , Femenino , Prueba de Complementación Genética , Ligamiento Genético , Humanos , Masculino , Mutación , Proteínas Nucleares/metabolismo , Linaje , Receptores Androgénicos/metabolismo
7.
Life Sci Alliance ; 6(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36622344

RESUMEN

The leading strand-oriented alternative PCNA clamp loader DSCC1-RFC functions in DNA replication, repair, and sister chromatid cohesion (SCC), but how it facilitates these processes is incompletely understood. Here, we confirm that loss of human DSCC1 results in reduced fork speed, increased DNA damage, and defective SCC. Genome-wide CRISPR screens in DSCC1-KO cells reveal multiple synthetically lethal interactions, enriched for DNA replication and cell cycle regulation. We show that DSCC1-KO cells require POLE3 for survival. Co-depletion of DSCC1 and POLE3, which both interact with the catalytic polymerase ε subunit, additively impair DNA replication, suggesting that these factors contribute to leading-strand DNA replication in parallel ways. An additional hit is MMS22L, which in humans forms a heterodimer with TONSL. Synthetic lethality of DSCC1 and MMS22L-TONSL likely results from detrimental SCC loss. We show that MMS22L-TONSL, like DDX11, functions in a SCC establishment pathway parallel to DSCC1-RFC. Because both DSCC1-RFC and MMS22L facilitate ESCO2 recruitment to replication forks, we suggest that distinct ESCO2 recruitment pathways promote SCC establishment following either cohesin conversion or de novo cohesin loading.


Asunto(s)
Cromátides , Replicación del ADN , Humanos , Cromátides/genética , Cromátides/metabolismo , Replicación del ADN/genética , Segregación Cromosómica/genética , Puntos de Control del Ciclo Celular , Daño del ADN/genética , ADN Polimerasa III/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , ADN Helicasas/genética , ARN Helicasas DEAD-box/metabolismo , FN-kappa B/metabolismo
8.
Sci Rep ; 12(1): 45, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997070

RESUMEN

Head-and-neck squamous cell carcinomas (HNSCCs) are relatively common in patients with Fanconi anemia (FA), a hereditary chromosomal instability disorder. Standard chemo-radiation therapy is not tolerated in FA due to an overall somatic hypersensitivity to such treatment. The question is how to find a suitable alternative treatment. We used whole-exome and whole genome mRNA sequencing to identify major genomic and transcriptomic events associated with FA-HNSCC. CRISPR-engineered FA-knockout models were used to validate a number of top hits that were likely to be druggable. We identified deletion of 18q21.2 and amplification of 11q22.2 as prevailing copy-number alterations in FA HNSCCs, the latter of which was associated with strong overexpression of the cancer-related genes YAP1, BIRC2, BIRC3 (at 11q22.1-2). We then found the drug AZD5582, a known small molecule inhibitor of BIRC2-3, to selectively kill FA tumor cells that overexpressed BIRC2-3. This occurred at drug concentrations that did not affect the viability of untransformed FA cells. Our data indicate that 11q22.2 amplifications are relatively common oncogenic events in FA-HNSCCs, as holds for non FA-HNSCC. Therefore, chemotherapeutic inhibition of overexpressed BIRC2-3 may provide the basis for an approach to develop a clinically realistic treatment of FA-HNSCCs that carry 11q22.2 amplifications.


Asunto(s)
Proteína 3 que Contiene Repeticiones IAP de Baculovirus/genética , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/metabolismo , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/genética , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Alquinos/farmacología , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/antagonistas & inhibidores , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Anemia de Fanconi/complicaciones , Anemia de Fanconi/inmunología , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/complicaciones , Neoplasias de Cabeza y Cuello/inmunología , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oligopéptidos/farmacología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Proteínas Señalizadoras YAP/genética , Proteínas Señalizadoras YAP/metabolismo
9.
Sci Adv ; 8(3): eabk0114, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35044816

RESUMEN

Budding uninhibited by benzimidazoles (BUB1) contributes to multiple mitotic processes. Here, we describe the first two patients with biallelic BUB1 germline mutations, who both display microcephaly, intellectual disability, and several patient-specific features. The identified mutations cause variable degrees of reduced total protein level and kinase activity, leading to distinct mitotic defects. Both patients' cells show prolonged mitosis duration, chromosome segregation errors, and an overall functional spindle assembly checkpoint. However, while BUB1 levels mostly affect BUBR1 kinetochore recruitment, impaired kinase activity prohibits centromeric recruitment of Aurora B, SGO1, and TOP2A, correlating with anaphase bridges, aneuploidy, and defective sister chromatid cohesion. We do not observe accelerated cohesion fatigue. We hypothesize that unresolved DNA catenanes increase cohesion strength, with concomitant increase in anaphase bridges. In conclusion, BUB1 mutations cause a neurodevelopmental disorder, with clinical and cellular phenotypes that partially resemble previously described syndromes, including autosomal recessive primary microcephaly, mosaic variegated aneuploidy, and cohesinopathies.


Asunto(s)
Segregación Cromosómica , Microcefalia , Aneuploidia , Segregación Cromosómica/genética , Humanos , Microcefalia/genética , Mutación , Proteínas Serina-Treonina Quinasas/genética
10.
Hum Mol Genet ; 18(18): 3484-95, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19561169

RESUMEN

The Fanconi anemia (FA) core complex member FANCM remodels synthetic replication forks and recombination intermediates. Thus far, only one FA patient with FANCM mutations has been described, but the relevance of these mutations for the FA phenotype is uncertain. To provide further experimental access to the FA-M complementation group we have generated Fancm-deficient mice by deleting exon 2. FANCM deficiency caused hypogonadism in mice and hypersensitivity to cross-linking agents in mouse embryonic fibroblasts (MEFs), thus phenocopying other FA mouse models. However, Fancm(Delta2/Delta2) mice also showed unique features atypical for FA mice, including underrepresentation of female Fancm(Delta2/Delta2) mice and decreased overall and tumor-free survival. This increased cancer incidence may be correlated to the role of FANCM in the suppression of spontaneous sister chromatid exchanges as observed in MEFs. In addition, FANCM appeared to have a stimulatory rather than essential role in FANCD2 monoubiquitination. The FA-M mouse model presented here suggests that FANCM functions both inside and outside the FA core complex to maintain genome stability and to prevent tumorigenesis.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/deficiencia , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Alelos , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovario/anomalías , Ovario/metabolismo , Fenotipo , Intercambio de Cromátides Hermanas , Tasa de Supervivencia , Testículo/anomalías , Testículo/metabolismo
11.
Blood ; 114(1): 174-80, 2009 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-19423727

RESUMEN

FANCM is a component of the Fanconi anemia (FA) core complex and one FA patient (EUFA867) with biallelic mutations in FANCM has been described. Strikingly, we found that EUFA867 also carries biallelic mutations in FANCA. After correcting the FANCA defect in EUFA867 lymphoblasts, a "clean" FA-M cell line was generated. These cells were hypersensitive to mitomycin C, but unlike cells defective in other core complex members, FANCM(-/-) cells were proficient in monoubiquitinating FANCD2 and were sensitive to the topoisomerase inhibitor camptothecin, a feature shared only with the FA subtype D1 and N. In addition, FANCM(-/-) cells were sensitive to UV light. FANCM and a C-terminal deletion mutant rescued the cross-linker sensitivity of FANCM(-/-) cells, whereas a FANCM ATPase mutant did not. Because both mutants restored the formation of FANCD2 foci, we conclude that FANCM functions in an FA core complex-dependent and -independent manner.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Camptotecina/farmacología , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/farmacología , ADN Helicasas/deficiencia , Resistencia a Medicamentos/genética , Resistencia a Medicamentos/fisiología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Expresión Génica , Humanos , Mutación , Tolerancia a Radiación/genética , Tolerancia a Radiación/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Ubiquitinación/genética , Rayos Ultravioleta
12.
J Exp Med ; 218(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33315086

RESUMEN

ERCC1-XPF is a multifunctional endonuclease involved in nucleotide excision repair (NER), interstrand cross-link (ICL) repair, and DNA double-strand break (DSB) repair. Only two patients with bi-allelic ERCC1 mutations have been reported, both of whom had features of Cockayne syndrome and died in infancy. Here, we describe two siblings with bi-allelic ERCC1 mutations in their teenage years. Genomic sequencing identified a deletion and a missense variant (R156W) within ERCC1 that disrupts a salt bridge below the XPA-binding pocket. Patient-derived fibroblasts and knock-in epithelial cells carrying the R156W substitution show dramatically reduced protein levels of ERCC1 and XPF. Moreover, mutant ERCC1 weakly interacts with NER and ICL repair proteins, resulting in diminished recruitment to DNA damage. Consequently, patient cells show strongly reduced NER activity and increased chromosome breakage induced by DNA cross-linkers, while DSB repair was relatively normal. We report a new case of ERCC1 deficiency that severely affects NER and considerably impacts ICL repair, which together result in a unique phenotype combining short stature, photosensitivity, and progressive liver and kidney dysfunction.


Asunto(s)
Daño del ADN/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Riñón/patología , Riñón/fisiopatología , Mutación/genética , Alelos , Sustitución de Aminoácidos , Secuencia de Bases , Línea Celular , Citoplasma/metabolismo , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Endonucleasas/deficiencia , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Luz , Hígado/patología , Hígado/fisiopatología , Proteínas Mutantes/metabolismo , Mutación Missense/genética , Estabilidad Proteica , Hermanos
13.
Elife ; 102021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33459596

RESUMEN

Germline mutations in the Folliculin (FLCN) tumor suppressor gene cause Birt-Hogg-Dubé (BHD) syndrome, a rare autosomal dominant disorder predisposing carriers to kidney tumors. FLCN is a conserved, essential gene linked to diverse cellular processes but the mechanism by which FLCN prevents kidney cancer remains unknown. Here, we show that disrupting FLCN in human renal tubular epithelial cells (RPTEC/TERT1) activates TFE3, upregulating expression of its E-box targets, including RRAGD and GPNMB, without modifying mTORC1 activity. Surprisingly, the absence of FLCN or its binding partners FNIP1/FNIP2 induces interferon response genes independently of interferon. Mechanistically, FLCN loss promotes STAT2 recruitment to chromatin and slows cellular proliferation. Our integrated analysis identifies STAT1/2 signaling as a novel target of FLCN in renal cells and BHD tumors. STAT1/2 activation appears to counterbalance TFE3-directed hyper-proliferation and may influence immune responses. These findings shed light on unique roles of FLCN in human renal tumorigenesis and pinpoint candidate prognostic biomarkers.


Asunto(s)
Proteínas Portadoras/genética , Células Epiteliales/metabolismo , Riñón/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Supresoras de Tumor/genética , Proteínas Portadoras/metabolismo , Mutación de Línea Germinal , Humanos , Interferones/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
14.
PLoS One ; 15(1): e0220348, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31935221

RESUMEN

In a process linked to DNA replication, duplicated chromosomes are entrapped in large, circular cohesin complexes and functional sister chromatid cohesion (SCC) is established by acetylation of the SMC3 cohesin subunit. Roberts Syndrome (RBS) and Warsaw Breakage Syndrome (WABS) are rare human developmental syndromes that are characterized by defective SCC. RBS is caused by mutations in the SMC3 acetyltransferase ESCO2, whereas mutations in the DNA helicase DDX11 lead to WABS. We found that WABS-derived cells predominantly rely on ESCO2, not ESCO1, for residual SCC, growth and survival. Reciprocally, RBS-derived cells depend on DDX11 to maintain low levels of SCC. Synthetic lethality between DDX11 and ESCO2 correlated with a prolonged delay in mitosis, and was rescued by knockdown of the cohesin remover WAPL. Rescue experiments using human or mouse cDNAs revealed that DDX11, ESCO1 and ESCO2 act on different but related aspects of SCC establishment. Furthermore, a DNA binding DDX11 mutant failed to correct SCC in WABS cells and DDX11 deficiency reduced replication fork speed. We propose that DDX11, ESCO1 and ESCO2 control different fractions of cohesin that are spatially and mechanistically separated.


Asunto(s)
Acetiltransferasas/genética , Proteínas de Ciclo Celular/genética , Cromátides/metabolismo , Proteínas Cromosómicas no Histona/genética , ARN Helicasas DEAD-box/genética , ADN Helicasas/genética , Células Epiteliales/enzimología , Fibroblastos/enzimología , Acetiltransferasas/metabolismo , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Línea Celular Transformada , Proliferación Celular , Cromátides/ultraestructura , Proteínas Cromosómicas no Histona/metabolismo , Rotura Cromosómica , Segregación Cromosómica , Anomalías Craneofaciales/enzimología , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , ARN Helicasas DEAD-box/metabolismo , ADN Helicasas/metabolismo , Ectromelia/enzimología , Ectromelia/genética , Ectromelia/patología , Células Epiteliales/patología , Fibroblastos/patología , Expresión Génica , Humanos , Hipertelorismo/enzimología , Hipertelorismo/genética , Hipertelorismo/patología , Ratones , Mitosis , Modelos Biológicos , Mutación , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Cohesinas
15.
Nat Commun ; 11(1): 4287, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32855419

RESUMEN

Warsaw Breakage Syndrome (WABS) is a rare disorder related to cohesinopathies and Fanconi anemia, caused by bi-allelic mutations in DDX11. Here, we report multiple compound heterozygous WABS cases, each displaying destabilized DDX11 protein and residual DDX11 function at the cellular level. Patient-derived cell lines exhibit sensitivity to topoisomerase and PARP inhibitors, defective sister chromatid cohesion and reduced DNA replication fork speed. Deleting DDX11 in RPE1-TERT cells inhibits proliferation and survival in a TP53-dependent manner and causes chromosome breaks and cohesion defects, independent of the expressed pseudogene DDX12p. Importantly, G-quadruplex (G4) stabilizing compounds induce chromosome breaks and cohesion defects which are strongly aggravated by inactivation of DDX11 but not FANCJ. The DNA helicase domain of DDX11 is essential for sister chromatid cohesion and resistance to G4 stabilizers. We propose that DDX11 is a DNA helicase protecting against G4 induced double-stranded breaks and concomitant loss of cohesion, possibly at DNA replication forks.


Asunto(s)
Anomalías Múltiples/etiología , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , G-Cuádruplex , Intercambio de Cromátides Hermanas , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Proliferación Celular , ARN Helicasas DEAD-box/química , ADN Helicasas/química , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Estabilidad Proteica , Seudogenes , ARN Helicasas/genética , ARN Helicasas/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Síndrome , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
16.
Oncogenesis ; 8(7): 38, 2019 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-31209198

RESUMEN

Head and neck squamous cell carcinomas (HNSCCs) coincide with poor survival rates. The lack of driver oncogenes complicates the development of targeted treatments for HNSCC. Here, we follow-up on two previous genome-wide RNA and microRNA interference screens in HNSCC to cross-examine tumor-specific lethality by targeting ATM, ATR, CHEK1, or CHEK2. Our results uncover CHEK1 as the most promising target for HNSCC. CHEK1 expression is essential across a panel of HNSCC cell lines but redundant for growth and survival of untransformed oral keratinocytes and fibroblasts. LY2603618 (Rabusertib), which specifically targets Chk1 kinase, kills HNSCC cells effectively and specifically. Our findings show that HNSCC cells depend on Chk1-mediated signaling to progress through S-phase successfully. Chk1 inhibition coincides with stalled DNA replication, replication fork collapses, and accumulation of DNA damage. We further show that Chk1 inhibition leads to bimodal HNSCC cell killing. In the most sensitive cell lines, apoptosis is induced in S-phase, whereas more resistant cell lines manage to bypass replication-associated apoptosis, but accumulate chromosomal breaks that become lethal in subsequent mitosis. Interestingly, CDK1 expression correlates with treatment outcome. Moreover, sensitivity to Chk1 inhibition requires functional CDK1 and CDK4/6 to drive cell cycle progression, arguing against combining Chk1 inhibitors with CDK inhibitors. In contrast, Wee1 inhibitor Adavosertib progresses the cell cycle and thereby increases lethality to Chk1 inhibition in HNSCC cell lines. We conclude that Chk1 has become a key molecule in HNSCC cell cycle regulation and a very promising therapeutic target. Chk1 inhibition leads to S-phase apoptosis or death in mitosis. We provide a potential efficacy biomarker and combination therapy to follow-up in clinical setting.

17.
DNA Repair (Amst) ; 5(5): 556-65, 2006 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-16513431

RESUMEN

The Fanconi anemia (FA) protein FANCE is an essential component of the nuclear FA core complex, which is required for monoubiquitination of the downstream target FANCD2, an important step in the FA pathway of DNA cross-link repair. FANCE is predominantly localized in the nucleus and acts as a molecular bridge between the FA core complex and FANCD2, through direct binding of both FANCC and FANCD2. At present, it is poorly understood how the nuclear accumulation of FANCE is regulated and therefore we investigated the nuclear localization of this FA protein. We found that FANCE has a strong tendency to localize in the nucleus, since the addition of a nuclear export signal does not interfere with the nuclear localization of FANCE. We also demonstrate that the nuclear accumulation of FANCE does not rely solely on its nuclear localization signal motifs, but also on FANCC. The other FA proteins are not involved in the nuclear accumulation of FANCE, indicating a tight relationship between FANCC and FANCE, as suggested from their direct interaction. Finally, we show that the region of FANCE interacting with FANCC appears to be different from the region involved in binding FANCD2. This strengthens the idea that FANCE recruits FANCD2 to the core complex, without interfering with the binding of FANCC.


Asunto(s)
Proteína del Grupo de Complementación C de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación E de la Anemia de Fanconi/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación C de la Anemia de Fanconi/química , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación E de la Anemia de Fanconi/química , Proteína del Grupo de Complementación E de la Anemia de Fanconi/genética , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Señales de Exportación Nuclear/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Técnicas del Sistema de Dos Híbridos
18.
Cell Oncol ; 29(3): 211-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17452773

RESUMEN

To identify the gene underlying Fanconi anemia (FA) complementation group I we studied informative FA-I families by a genome-wide linkage analysis, which resulted in 4 candidate regions together encompassing 351 genes. Candidates were selected via bioinformatics and data mining on the basis of their resemblance to other FA genes/proteins acting in the FA pathway, such as: degree of evolutionary conservation, presence of nuclear localization signals and pattern of tissue-dependent expression. We found a candidate, KIAA1794 on chromosome 15q25-26, to be mutated in 8 affected individuals previously assigned to complementation group I. Western blots of endogenous FANCI indicated that functionally active KIAA1794 protein is lacking in FA-I individuals. Knock-down of KIAA1794 expression by siRNA in HeLa cells caused excessive chromosomal breakage induced by mitomycin C, a hallmark of FA cells. Furthermore, phenotypic reversion of a patient-derived cell line was associated with a secondary genetic alteration at the KIAA1794 locus. These data add up to two conclusions. First, KIAA1794 is a FA gene. Second, this gene is identical to FANCI, since the patient cell lines found mutated in this study included the reference cell line for group I, EUFA592.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Adolescente , Adulto , Secuencia de Bases , Línea Celular , Niño , Inestabilidad Cromosómica/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Femenino , Genoma Humano/genética , Células HeLa , Humanos , Masculino , Datos de Secuencia Molecular , Mutación/genética , Linaje , Fenotipo , Ubiquitina/metabolismo
19.
Cancer Res ; 65(4): 1271-6, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15735012

RESUMEN

Patients with Fanconi anemia (FA) are prone to develop malignancies at an early age. Besides hematologic malignancies, squamous cell carcinomas in the anogenital region and head and neck are also frequently found in these patients. The aim of this study was to generate a panel of head and neck squamous cell carcinoma (HNSCC) cell lines and xenografts of FA HNSCC, and to characterize these cell lines in comparison with a panel of seven cell lines from patients with sporadic HNSCC. Analyses have been done on sensitivity to DNA cross-linking agents, loss of heterozygosity profile, TP53 mutations, TP53 polymorphisms and the presence of human papillomavirus. Four FA HNSCC cell lines were established. Sensitivity to DNA cross-linking agents (cisplatin) in the FA HNSCC cell lines was on average 10 times higher as compared with the sporadic HNSCC cell lines. Human papillomavirus was not detected in any of the FA or sporadic cell lines. No differences were found in loss of heterozygosity pattern, TP53 mutation frequency and TP53 polymorphism between FA and sporadic HNSCC cell lines. This is the first report on the generation of squamous cell lines of FA patients. The FA HNSCC cell lines we have generated may be utilized for future studies and might aid in the development of new preventive therapies for FA patients. The genetic characteristics of these cell lines suggest that FA HNSCC are not very different from sporadic HNSCC, except for the sensitivity to cisplatin which is consistent with the known cellular FA phenotype.


Asunto(s)
Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Anemia de Fanconi/complicaciones , Anemia de Fanconi/genética , Neoplasias de Cabeza y Cuello/etiología , Neoplasias de Cabeza y Cuello/genética , Adulto , Anciano , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Cisplatino/farmacología , Codón , ADN Viral/genética , Anemia de Fanconi/patología , Femenino , Genes p53/genética , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Metotrexato/farmacología , Repeticiones de Microsatélite , Persona de Mediana Edad , Papillomaviridae/genética , Polimorfismo Genético
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA