Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 52(3): 434-46, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24207054

RESUMEN

The replicative machinery encounters many impediments, some of which can be overcome by lesion bypass or replication restart pathways, leaving repair for a later time. However, interstrand crosslinks (ICLs), which preclude DNA unwinding, are considered absolute blocks to replication. Current models suggest that fork collisions, either from one or both sides of an ICL, initiate repair processes required for resumption of replication. To test these proposals, we developed a single-molecule technique for visualizing encounters of replication forks with ICLs as they occur in living cells. Surprisingly, the most frequent patterns were consistent with replication traverse of an ICL, without lesion repair. The traverse frequency was strongly reduced by inactivation of the translocase and DNA binding activities of the FANCM/MHF complex. The results indicate that translocase-based mechanisms enable DNA synthesis to continue past ICLs and that these lesions are not always absolute blocks to replication.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , ADN Helicasas/metabolismo , Replicación del ADN/genética , ADN/biosíntesis , Anemia de Fanconi/genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Cricetinae , ADN Helicasas/genética , Proteínas de Unión al ADN , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Humanos , Ratones , Complejos Multiproteicos/metabolismo , Proteínas Supresoras de Tumor/genética
2.
Mol Cell ; 47(1): 61-75, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22705371

RESUMEN

The Fanconi anemia (FA) protein network is necessary for repair of DNA interstrand crosslinks (ICLs), but its control mechanism remains unclear. Here we show that the network is regulated by a ubiquitin signaling cascade initiated by RNF8 and its partner, UBC13, and mediated by FAAP20, a component of the FA core complex. FAAP20 preferentially binds the ubiquitin product of RNF8-UBC13, and this ubiquitin-binding activity and RNF8-UBC13 are both required for recruitment of FAAP20 to ICLs. Both RNF8 and FAAP20 are required for recruitment of FA core complex and FANCD2 to ICLs, whereas RNF168 can modulate efficiency of the recruitment. RNF8 and FAAP20 are needed for efficient FANCD2 monoubiquitination, a key step of the FA network; RNF8 and the FA core complex work in the same pathway to promote cellular resistance to ICLs. Thus, the RNF8-FAAP20 ubiquitin cascade is critical for recruiting FA core complex to ICLs and for normal function of the FA network.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Ubiquitinación , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/química , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Lisina/química , Lisina/genética , Lisina/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , Homología de Secuencia de Aminoácido , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Mol Cell ; 37(6): 865-78, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20347428

RESUMEN

FANCM remodels branched DNA structures and plays essential roles in the cellular response to DNA replication stress. Here, we show that FANCM forms a conserved DNA-remodeling complex with a histone-fold heterodimer, MHF. We find that MHF stimulates DNA binding and replication fork remodeling by FANCM. In the cell, FANCM and MHF are rapidly recruited to forks stalled by DNA interstrand crosslinks, and both are required for cellular resistance to such lesions. In vertebrates, FANCM-MHF associates with the Fanconi anemia (FA) core complex, promotes FANCD2 monoubiquitination in response to DNA damage, and suppresses sister-chromatid exchanges. Yeast orthologs of these proteins function together to resist MMS-induced DNA damage and promote gene conversion at blocked replication forks. Thus, FANCM-MHF is an essential DNA-remodeling complex that protects replication forks from yeast to human.


Asunto(s)
ADN Helicasas/metabolismo , ADN/metabolismo , Inestabilidad Genómica , Histonas/metabolismo , Pliegue de Proteína , Multimerización de Proteína , Secuencia de Aminoácidos , Animales , Línea Celular , Pollos , ADN/genética , Daño del ADN , ADN Helicasas/química , ADN Helicasas/genética , Replicación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Evolución Molecular , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Humanos , Datos de Secuencia Molecular , Unión Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Alineación de Secuencia , Intercambio de Cromátides Hermanas
4.
Mol Cell ; 35(5): 716-23, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19748364

RESUMEN

Fanconi anemia (FA) is characterized by cellular hypersensitivity to DNA crosslinking agents, but how the Fanconi pathway protects cells from DNA crosslinks and whether FA proteins act directly on crosslinks remain unclear. We developed a chromatin-IP-based strategy termed eChIP and detected association of multiple FA proteins with DNA crosslinks in vivo. Interdependence analyses revealed that crosslink-specific enrichment of various FA proteins is controlled by distinct mechanisms. BRCA-related FA proteins (BRCA2, FANCJ/BACH1, and FANCN/PALB2), but not FA core and I/D2 complexes, require replication for their crosslink association. FANCD2, but not FANCJ and FANCN, requires the FA core complex for its recruitment. FA core complex requires nucleotide excision repair proteins XPA and XPC for its association. Consistent with the distinct recruitment mechanism, recombination-independent crosslink repair was inversely affected in cells deficient of FANC-core versus BRCA-related FA proteins. Thus, FA proteins participate in distinct DNA damage response mechanisms governed by DNA replication status.


Asunto(s)
Proteína BRCA2/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Neoplasias de la Mama/genética , Daño del ADN , Replicación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Reguladoras de la Apoptosis , Proteína BRCA2/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina/métodos , Reactivos de Enlaces Cruzados/farmacología , Proteínas de Unión al ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Femenino , Ficusina/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Proteínas Nucleares/genética , Plásmidos/metabolismo , Recombinación Genética , Factores de Tiempo , Proteínas Supresoras de Tumor/genética , Proteína de la Xerodermia Pigmentosa del Grupo A/metabolismo
5.
Am J Hum Genet ; 92(5): 800-6, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23623386

RESUMEN

Fanconi anemia (FA) is a rare genomic instability disorder characterized by progressive bone marrow failure and predisposition to cancer. FA-associated gene products are involved in the repair of DNA interstrand crosslinks (ICLs). Fifteen FA-associated genes have been identified, but the genetic basis in some individuals still remains unresolved. Here, we used whole-exome and Sanger sequencing on DNA of unclassified FA individuals and discovered biallelic germline mutations in ERCC4 (XPF), a structure-specific nuclease-encoding gene previously connected to xeroderma pigmentosum and segmental XFE progeroid syndrome. Genetic reversion and wild-type ERCC4 cDNA complemented the phenotype of the FA cell lines, providing genetic evidence that mutations in ERCC4 cause this FA subtype. Further biochemical and functional analysis demonstrated that the identified FA-causing ERCC4 mutations strongly disrupt the function of XPF in DNA ICL repair without severely compromising nucleotide excision repair. Our data show that depending on the type of ERCC4 mutation and the resulting balance between both DNA repair activities, individuals present with one of the three clinically distinct disorders, highlighting the multifunctional nature of the XPF endonuclease in genome stability and human disease.


Asunto(s)
Proteínas de Unión al ADN/genética , Desoxirribonucleasas/genética , Anemia de Fanconi/genética , Predisposición Genética a la Enfermedad/genética , Fenotipo , Apoptosis/genética , Apoptosis/efectos de la radiación , Secuencia de Bases , Exoma/genética , Anemia de Fanconi/patología , Mutación de Línea Germinal/genética , Humanos , Immunoblotting , Inmunoprecipitación , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Rayos Ultravioleta
6.
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
7.
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
8.
Hum Mutat ; 35(4): 442-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24395671

RESUMEN

Fanconi anemia (FA) is a rare recessive disorder with chromosomal instability, congenital abnormalities, and a high cancer risk. The breast cancer susceptibility gene BRCA2 (FANCD1) is one of the 16 genes involved in this recessive disease. We have identified a novel mutation of the splice donor site of intron 1 in the noncoding region of BRCA2 in a Japanese FA family. This mutation may account for the FA phenotype in a patient originally reported to have biallelic mutations in BRCA2. Subsequent functional studies revealed that one of the mutations, K2729N, was a neutral change. As reported here, a more careful analysis resulted in the identification of a novel splice site mutation. Functional analysis using a mouse embryonic stem cell-based assay revealed that it causes aberrant splicing, reduced transcript levels and hypersensitivity to DNA damaging agents, suggesting that it is likely to be pathogenic. Although similar pathogenic variants in the noncoding region of BRCA1 and 2 were not identified in a cohort of 752 familial breast cancer cases, we still think this finding is relevant for mutation analysis in Hereditary Breast and Ovarian Cancer Syndrome families in a diagnostic setting.


Asunto(s)
Proteína BRCA2/genética , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/genética , Síndrome de Cáncer de Mama y Ovario Hereditario/diagnóstico , Síndrome de Cáncer de Mama y Ovario Hereditario/genética , Animales , Proteína BRCA1/genética , Secuencia de Bases , Células Cultivadas , Análisis Mutacional de ADN , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Intrones , Ratones , Datos de Secuencia Molecular , Mutación Missense , Linaje , Sitios de Empalme de ARN
9.
Nat Genet ; 37(9): 958-63, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16116422

RESUMEN

Fanconi anemia is a genetic disease characterized by genomic instability and cancer predisposition. Nine genes involved in Fanconi anemia have been identified; their products participate in a DNA damage-response network involving BRCA1 and BRCA2 (refs. 2,3). We previously purified a Fanconi anemia core complex containing the FANCL ubiquitin ligase and six other Fanconi anemia-associated proteins. Each protein in this complex is essential for monoubiquitination of FANCD2, a key reaction in the Fanconi anemia DNA damage-response pathway. Here we show that another component of this complex, FAAP250, is mutant in individuals with Fanconi anemia of a new complementation group (FA-M). FAAP250 or FANCM has sequence similarity to known DNA-repair proteins, including archaeal Hef, yeast MPH1 and human ERCC4 or XPF. FANCM can dissociate DNA triplex, possibly owing to its ability to translocate on duplex DNA. FANCM is essential for monoubiquitination of FANCD2 and becomes hyperphosphorylated in response to DNA damage. Our data suggest an evolutionary link between Fanconi anemia-associated proteins and DNA repair; FANCM may act as an engine that translocates the Fanconi anemia core complex along DNA.


Asunto(s)
Archaea/química , ADN Helicasas/genética , Reparación del ADN , Anemia de Fanconi/genética , Hemaglutininas Virales/genética , Ligasas/genética , Proteínas Virales de Fusión/genética , Proteína BRCA1/genética , Proteína BRCA2/genética , Evolución Biológica , ADN/metabolismo , ADN Helicasas/deficiencia , ADN Helicasas/metabolismo , Anemia de Fanconi/enzimología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi , Proteína del Grupo de Complementación L de la Anemia de Fanconi , Humanos , Inmunoprecipitación , Ligasas/deficiencia , Ligasas/metabolismo , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/metabolismo , Fosforilación , Transporte de Proteínas , Ubiquitina/metabolismo , Proteínas Virales de Fusión/deficiencia
10.
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
11.
Hum Mutat ; 34(1): 93-6, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23033263

RESUMEN

Fanconi anemia (FA) is a rare genetic disorder characterized by congenital malformations, progressive bone marrow failure (BMF), and susceptibility to malignancies. FA is caused by biallelic or hemizygous mutations in one of 15 known FA genes, whose products are involved in the FA/BRCA DNA damage response pathway. Here, we report on a patient with previously unknown mutations of the most recently identified FA gene, SLX4/FANCP. Whole exome sequencing (WES) revealed a nonsense mutation and an unusual splice site mutation resulting in the partial replacement of exonic with intronic bases, thereby removing a nuclear localization signal. Immunoblotting detected no residual SLX4 protein, which was consistent with abrogated interactions with XPF/ERCC1 and MUS81/EME1. This cellular finding did not result in a more severe clinical phenotype than that of previously reported FA-P patients. Our study additionally exemplifies the versatility of WES for the detection of mutations in heterogenic disorders such as FA.


Asunto(s)
Exoma/genética , Anemia de Fanconi/genética , Mutación , Recombinasas/genética , Secuencia de Bases , Codón sin Sentido , Análisis Mutacional de ADN , Anemia de Fanconi/metabolismo , Femenino , Humanos , Immunoblotting , Datos de Secuencia Molecular , Sitios de Empalme de ARN/genética , Recombinasas/metabolismo , Adulto Joven
12.
Hum Mutat ; 34(1): 70-3, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22911665

RESUMEN

SLX4/FANCP is a recently discovered novel disease gene for Fanconi anemia (FA), a rare recessive disorder characterized by chromosomal instability and increased cancer susceptibility. Three of the 15 FA genes are breast cancer susceptibility genes in heterozygous mutation carriers--BRCA2, PALB2, and BRIP1. To investigate if defects in SLX4 also predispose to breast cancer, the gene was sequenced in a cohort of 729 BRCA1/BRCA2-negative familial breast cancer cases. We identified a single splice site mutation (c.2013+2T>A), which causes a frameshift by skipping of exon 8. We also identified 39 missense variants, four of which were selected for functional testing in a Mitomycin C-induced growth inhibition assay, and appeared indistinguishable from wild type. Although this is the first study that describes a truncating SLX4 mutation in breast cancer patients, our data indicate that germline mutations in SLX4 are very rare and are unlikely to make a significant contribution to familial breast cancer.


Asunto(s)
Neoplasias de la Mama Masculina/genética , Neoplasias de la Mama/genética , Mutación , Recombinasas/genética , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Análisis Mutacional de ADN , Salud de la Familia , Anemia de Fanconi/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Sitios de Empalme de ARN/genética
13.
J Biol Chem ; 287(2): 1007-21, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22102414

RESUMEN

Mutations in the human ChlR1 gene are associated with a unique genetic disorder known as Warsaw breakage syndrome characterized by cellular defects in sister chromatid cohesion and hypersensitivity to agents that induce replication stress. A role of ChlR1 helicase in sister chromatid cohesion was first evidenced by studies of the yeast homolog Chl1p; however, its cellular functions in DNA metabolism are not well understood. We carefully examined the DNA substrate specificity of purified recombinant human ChlR1 protein and the biochemical effect of a patient-derived mutation, a deletion of a single lysine (K897del) in the extreme C terminus of ChlR1. The K897del clinical mutation abrogated ChlR1 helicase activity on forked duplex or D-loop DNA substrates by perturbing its DNA binding and DNA-dependent ATPase activity. Wild-type ChlR1 required a minimal 5' single-stranded DNA tail of 15 nucleotides to efficiently unwind a simple duplex DNA substrate. The additional presence of a 3' single-stranded DNA tail as short as five nucleotides dramatically increased ChlR1 helicase activity, demonstrating the preference of the enzyme for forked duplex structures. ChlR1 unwound G-quadruplex (G4) DNA with a strong preference for a two-stranded antiparallel G4 (G2') substrate and was only marginally active on a four-stranded parallel G4 structure. The marked difference in ChlR1 helicase activity on the G4 substrates, reflected by increased binding to the G2' substrate, distinguishes ChlR1 from the sequence-related FANCJ helicase mutated in Fanconi anemia. The biochemical results are discussed in light of the known cellular defects associated with ChlR1 deficiency.


Asunto(s)
ARN Helicasas DEAD-box/química , ADN Helicasas/química , Trastornos por Deficiencias en la Reparación del ADN/enzimología , ADN/química , Secuencia de Aminoácidos , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/química , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN/genética , ADN/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Trastornos por Deficiencias en la Reparación del ADN/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/química , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Humanos , Eliminación de Secuencia , Especificidad por Sustrato , Síndrome
14.
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
15.
J Pathol ; 226(1): 28-39, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21915857

RESUMEN

Fanconi anaemia (FA) is a rare recessive disorder marked by developmental abnormalities, bone marrow failure, and a high risk for the development of leukaemia and solid tumours. The inactivation of FA genes, in particular FANCF, has also been documented in sporadic tumours in non-FA patients. To study whether there is a causal relationship between FA pathway defects and tumour development, we have generated a mouse model with a targeted disruption of the FA core complex gene Fancf. Fancf-deficient mouse embryonic fibroblasts displayed a phenotype typical for FA cells: they showed an aberrant response to DNA cross-linking agents as manifested by G(2) arrest, chromosomal aberrations, reduced survival, and an inability to monoubiquitinate FANCD2. Fancf homozygous mice were viable, born following a normal Mendelian distribution, and showed no growth retardation or developmental abnormalities. The gonads of Fancf mutant mice functioned abnormally, showing compromised follicle development and spermatogenesis as has been observed in other FA mouse models and in FA patients. In a cohort of Fancf-deficient mice, we observed decreased overall survival and increased tumour incidence. Notably, in seven female mice, six ovarian tumours developed: five granulosa cell tumours and one luteoma. One mouse had developed tumours in both ovaries. High-resolution array comparative genomic hybridization (aCGH) on these tumours suggests that the increased incidence of ovarian tumours correlates with the infertility in Fancf-deficient mice and the genomic instability characteristic of FA pathway deficiency.


Asunto(s)
Proteína del Grupo de Complementación F de la Anemia de Fanconi/genética , Tumor de Células de la Granulosa/genética , Luteoma/genética , Neoplasias Ováricas/genética , Animales , Hibridación Genómica Comparativa , Modelos Animales de Enfermedad , Proteína del Grupo de Complementación F de la Anemia de Fanconi/deficiencia , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Nat Genet ; 35(2): 165-70, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12973351

RESUMEN

Fanconi anemia is a recessively inherited disease characterized by congenital defects, bone marrow failure and cancer susceptibility. Cells from individuals with Fanconi anemia are highly sensitive to DNA-crosslinking drugs, such as mitomycin C (MMC). Fanconi anemia proteins function in a DNA damage response pathway involving breast cancer susceptibility gene products, BRCA1 and BRCA2 (refs. 1,2). A key step in this pathway is monoubiquitination of FANCD2, resulting in the redistribution of FANCD2 to nuclear foci containing BRCA1 (ref. 3). The underlying mechanism is unclear because the five Fanconi anemia proteins known to be required for this ubiquitination have no recognizable ubiquitin ligase motifs. Here we report a new component of a Fanconi anemia protein complex, called PHF9, which possesses E3 ubiquitin ligase activity in vitro and is essential for FANCD2 monoubiquitination in vivo. Because PHF9 is defective in a cell line derived from an individual with Fanconi anemia, we conclude that PHF9 (also called FANCL) represents a novel Fanconi anemia complementation group (FA-L). Our data suggest that PHF9 has a crucial role in the Fanconi anemia pathway as the likely catalytic subunit required for monoubiquitination of FANCD2.


Asunto(s)
Anemia de Fanconi/genética , Ligasas/genética , Proteínas Nucleares/genética , Eliminación de Secuencia , Secuencia de Aminoácidos , Proteína BRCA1/genética , Proteína BRCA2/genética , Secuencia de Bases , Aberraciones Cromosómicas , Anemia de Fanconi/enzimología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi , Proteína del Grupo de Complementación L de la Anemia de Fanconi , Humanos , Ligasas/deficiencia , Datos de Secuencia Molecular , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Ubiquitina/metabolismo
17.
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
18.
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
19.
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
20.
Mutat Res ; 668(1-2): 11-9, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19061902

RESUMEN

The capacity to maintain genomic integrity is shared by all living organisms. Multiple pathways are distinguished that safeguard genomic stability, most of which have originated in primitive life forms. In human individuals, defects in these pathways are typically associated with cancer proneness. The Fanconi anemia pathway, one of these pathways, has evolved relatively late during evolution and exists - in its fully developed form - only in vertebrates. This pathway, in which thus far 13 distinct proteins have been shown to participate, appears essential for error-free DNA replication. Inactivating mutations in the corresponding genes underlie the recessive disease Fanconi anemia (FA). In the last decade the genetic basis of this disorder has been uncovered by a variety of approaches, including complementation cloning, genetic linkage analysis and protein association studies. Here we review these approaches, introduce the encoded proteins, and discuss their possible role in ensuring genomic integrity.


Asunto(s)
Reparación del ADN , Anemia de Fanconi/genética , Transducción de Señal , Reactivos de Enlaces Cruzados/farmacología , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Prueba de Complementación Genética , Estudio de Asociación del Genoma Completo , Humanos , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA