Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
J Clin Invest ; 125(4): 1523-32, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25751062

RESUMEN

The Fanconi anemia/BRCA (FA/BRCA) pathway is a DNA repair pathway that is required for excision of DNA interstrand cross-links. The 17 known FA proteins, along with several FA-associated proteins (FAAPs), cooperate in this pathway to detect, unhook, and excise DNA cross-links and to subsequently repair the double-strand breaks generated in the process. In the current study, we identified a patient with FA with a point mutation in FANCA, which encodes a mutant FANCA protein (FANCAI939S). FANCAI939S failed to bind to the FAAP20 subunit of the FA core complex, leading to decreased stability. Loss of FAAP20 binding exposed a SUMOylation site on FANCA at amino acid residue K921, resulting in E2 SUMO-conjugating enzyme UBC9-mediated SUMOylation, RING finger protein 4-mediated (RNF4-mediated) polyubiquitination, and proteasome-mediated degradation of FANCA. Mutation of the SUMOylation site of FANCA rescued the expression of the mutant protein. Wild-type FANCA was also subject to SUMOylation, RNF4-mediated polyubiquitination, and degradation, suggesting that regulated release of FAAP20 from FANCA is a critical step in the normal FA pathway. Consistent with this model, cells lacking RNF4 exhibited interstrand cross-linker hypersensitivity, and the gene encoding RNF4 was epistatic with the other genes encoding members of the FA/BRCA pathway. Together, the results from our study underscore the importance of analyzing unique patient-derived mutations for dissecting complex DNA repair processes.


Asunto(s)
Proteína BRCA1/fisiología , Reparación del ADN/fisiología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Anemia de Fanconi/genética , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Adulto , Línea Celular Tumoral , Reparación del ADN/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/antagonistas & inhibidores , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Femenino , Genes BRCA1 , Humanos , Mutación Missense , Linaje , Fenotipo , Mutación Puntual , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional/fisiología , Proteolisis , ARN Interferente Pequeño/farmacología , Transducción de Señal/fisiología , Sumoilación , Neoplasias de la Mama Triple Negativas/genética , Ubiquitinación/fisiología
3.
Yi Chuan ; 37(2): 204-213, 2015 Feb.
Artículo en Chino | MEDLINE | ID: mdl-25665647

RESUMEN

The FANCJ-like protein family is a class of ATP-dependent helicases that can catalytically unwind duplex DNA along the 5'-3' direction. It is involved in the processes of DNA damage repair, homologous recombination and G-quadruplex DNA unwinding, and plays a critical role in maintaining genome integrity. In this study, we systemically analyzed FNACJ-like proteins from 47 eukaryotic species and discussed their sequences diversity, origin and evolution, motif organization patterns and spatial structure differences. Four members of FNACJ-like proteins, including XPD, CHL1, RTEL1 and FANCJ, were found in eukaryotes, but some of them were seriously deficient in most fungi and some insects. For example, the Zygomycota fungi lost RTEL1, Basidiomycota and Ascomycota fungi lost RTEL1 and FANCJ, and Diptera insect lost FANCJ. FANCJ-like proteins contain canonical motor domains HD1 and HD2, and the HD1 domain further integrates with three unique domains Fe-S, Arch and Extra-D. Fe-S and Arch domains are relatively conservative in all members of the family, but the Extra-D domain is lost in XPD and differs from one another in rest members. There are 7, 10 and 2 specific motifs found from the three unique domains respectively, while 5 and 12 specific motifs are found from HD1 and HD2 domains except the conserved motifs reported previously. By analyzing the arrangement pattern of these specific motifs, we found that RTEL1 and FANCJ are more closer and share two specific motifs Vb2 and Vc in HD2 domain, which are likely related with their G-quadruplex DNA unwinding activity. The evidence of evolution showed that FACNJ-like proteins were originated from a helicase, which has a HD1 domain inserted by extra Fe-S domain and Arch domain. By three continuous gene duplication events and followed specialization, eukaryotes finally possessed the current four members of FANCJ-like proteins.


Asunto(s)
Eucariontes/química , Evolución Molecular , Proteína del Grupo de Complementación A de la Anemia de Fanconi/química , Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Animales , Secuencia Conservada , Proteínas Fúngicas/química , Proteínas Fúngicas/fisiología , Humanos , Estructura Terciaria de Proteína
4.
Blood ; 117(14): 3759-69, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21273304

RESUMEN

Fanconi anemia is characterized by congenital abnormalities, bone marrow failure, and cancer predisposition. To investigate the origin, functional role, and clinical impact of FANCA mutations, we determined a FANCA mutational spectrum with 130 pathogenic alleles. Some of these mutations were further characterized for their distribution in populations, mode of emergence, or functional consequences at cellular and clinical level. The world most frequent FANCA mutation is not the result of a mutational "hot-spot" but results from worldwide dissemination of an ancestral Indo-European mutation. We provide molecular evidence that total absence of FANCA in humans does not reduce embryonic viability, as the observed frequency of mutation carriers in the Gypsy population equals the expected by Hardy-Weinberg equilibrium. We also prove that long distance Alu-Alu recombination can cause Fanconi anemia by originating large interstitial deletions involving FANCA and 2 adjacent genes. Finally, we show that all missense mutations studied lead to an altered FANCA protein that is unable to relocate to the nucleus and activate the FA/BRCA pathway. This may explain the observed lack of correlation between type of FANCA mutation and cellular phenotype or clinical severity in terms of age of onset of hematologic disease or number of malformations.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Mutación , Adolescente , Edad de Inicio , Secuencia de Bases , Técnicas de Cultivo de Célula , Células Cultivadas , Niño , Preescolar , Aberraciones Cromosómicas , Hibridación Genómica Comparativa , Análisis Mutacional de ADN , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/epidemiología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Frecuencia de los Genes , Humanos , Lactante , Modelos Biológicos , Datos de Secuencia Molecular , Mutación/fisiología , Fenotipo , España/epidemiología
5.
Biochem Biophys Res Commun ; 404(1): 206-10, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21111709

RESUMEN

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


Asunto(s)
Daño del ADN , Reparación del ADN/genética , ADN Recombinante , Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Animales , Proteína BRCA2/fisiología , Células CHO , Cricetinae , Cricetulus , Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Proteína del Grupo de Complementación C de la Anemia de Fanconi/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Formaldehído/toxicidad , Histonas/metabolismo , Ratones
6.
Nucleic Acids Res ; 37(6): 1740-54, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19129235

RESUMEN

Both Fanconi anemia (FA) and telomere dysfunction are associated with chromosome instability and an increased risk of cancer. Because of these similarities, we have investigated whether there is a relationship between the FA protein, FANCD2 and telomeres. We find that FANCD2 nuclear foci colocalize with telomeres and PML bodies in immortalized telomerase-negative cells. These cells maintain telomeres by alternative lengthening of telomeres (ALT). In contrast, FANCD2 does not colocalize with telomeres or PML bodies in cells which express telomerase. Using a siRNA approach we find that FANCA and FANCL, which are components of the FA nuclear core complex, regulate FANCD2 monoubiquitination and the telomeric localization of FANCD2 in ALT cells. Transient depletion of FANCD2, or FANCA, results in a dramatic loss of detectable telomeres in ALT cells but not in telomerase-expressing cells. Furthermore, telomere loss following depletion of these proteins in ALT cells is associated with decreased homologous recombination between telomeres (T-SCE). Thus, the FA pathway has a novel function in ALT telomere maintenance related to DNA repair. ALT telomere maintenance is therefore one mechanism by which monoubiquitinated FANCD2 may promote genetic stability.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Telómero/metabolismo , Ubiquitinación , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/fisiología , Línea Celular , Línea Celular Transformada , Proteína del Grupo de Complementación A de la Anemia de Fanconi/antagonistas & inhibidores , Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/análisis , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/antagonistas & inhibidores , Proteína del Grupo de Complementación L de la Anemia de Fanconi/fisiología , Células HeLa , Humanos , Proteínas Serina-Treonina Quinasas/fisiología , Intercambio de Cromátides Hermanas , Telómero/química , Proteína 1 de Unión a Repeticiones Teloméricas/análisis
7.
Oncogene ; 28(5): 674-85, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19015634

RESUMEN

Fanconi anemia (FA) is a recessive genome instability syndrome characterized by heightened cellular sensitivity to DNA damage, aplastic anemia and cancer susceptibility. Leukemias and squamous cell carcinomas (SCCs) are the most predominant FA-associated cancers, with the latter exhibiting markedly early disease onset and aggressiveness. Although studies of hematopoietic cells derived from FA patients have provided much insight into bone marrow deficiencies and leukemogenesis, molecular transforming events in FA-deficient keratinocytes, which are the cell type of origin for SCC, are poorly understood. We describe here the growth and molecular properties of FANCA-deficient versus FANCA-corrected HPV E6/E7 immortalized keratinocytes in monolayer and organotypic epithelial raft culture. In response to DNA damage, FANCA-deficient patient-derived keratinocyte cultures displayed a G2/M phase arrest, senescence and apoptosis. Organotypic raft cultures exhibited DNA repair-associated defects with more 53BP1 foci and TdT-mediated dNTP nick end labeling-positive cells over their corrected counterparts. Interestingly, together with reduced rates of DNA damage, FA correction resulted in a marked decrease in epithelial thickness and the presence of fewer cell layers. The observed FANCA-mediated suppression of hyperplasia correlated with the detection of fewer cells transiting through the cell cycle in the absence of gross differentiation abnormalities or apoptotic differences. Importantly, the knockdown of either FANCA or FANCD2 in HPV-positive keratinocytes was sufficient for increasing epithelial hyperplasia. Our findings support a new role for FA pathways in the maintenance of differentiation-dependent cell cycle exit, with the implication that FA deficiencies may contribute to the high risk of FA patients for developing HPV-associated SCC.


Asunto(s)
Transformación Celular Viral/genética , Células Epiteliales/patología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Papillomavirus Humano 18/fisiología , Antibióticos Antineoplásicos/farmacología , Carcinoma de Células Escamosas/genética , Línea Celular Transformada , Proliferación Celular , Daño del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos/genética , Células Epiteliales/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Prueba de Complementación Genética , Predisposición Genética a la Enfermedad , Papillomavirus Humano 18/genética , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Mitomicina/farmacología , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Técnicas de Cultivo de Órganos/métodos , Neoplasias Cutáneas/genética
8.
Exp Cell Res ; 313(11): 2283-92, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17490643

RESUMEN

Fanconi anemia (FA) cells are abnormally sensitive to DNA cross-linking agents with increased levels of apoptosis and chromosomal instability. Defects in eight FA complementation groups inhibit monoubiquitination of FANCD2, and subsequent recruitment of FANCD2 to DNA damage and S-phase-associated nuclear foci. The specific functional defect in repair or response to DNA damage in FA cells remains unknown. Damage-resistant DNA synthesis is present 2.5-5 h after cross-linker treatment of FANCC, FANCA and FANCD2-deficient cells. Analysis of the size distribution of labeled DNA replication strands revealed that diepoxybutane treatment suppressed labeling of early but not late-firing replicons in FANCC-deficient cells. In contrast, normal responses to ionizing radiation were observed in FANCC-deficient cells. Absence of this late S-phase response in FANCC-deficient cells leads to activation of secondary checkpoint responses.


Asunto(s)
Reactivos de Enlaces Cruzados/toxicidad , Replicación del ADN/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi/fisiología , Anemia de Fanconi/genética , Origen de Réplica/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular , ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN/genética , Compuestos Epoxi/toxicidad , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Humanos , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/fisiología , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Fase S/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
9.
Endocrinology ; 147(12): 5676-89, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16946016

RESUMEN

GnRH binds its cognate G protein-coupled GnRH receptor (GnRHR) located on pituitary gonadotropes and drives expression of gonadotropin hormones. There are two gonadotropin hormones, comprised of a common alpha- and hormone-specific beta-subunit, which are required for gonadal function. Recently we identified that Fanconi anemia a (Fanca), a DNA damage repair gene, is differentially expressed within the LbetaT2 gonadotrope cell line in response to stimulation with GnRH. FANCA is mutated in more than 60% of cases of Fanconi anemia (FA), a rare genetically heterogeneous autosomal recessive disorder characterized by bone marrow failure, endocrine tissue cancer susceptibility, and infertility. Here we show that induction of FANCA protein is mediated by the GnRHR and that the protein constitutively adopts a nucleocytoplasmic intracellular distribution pattern. Using inhibitors to block nuclear import and export and a GnRHR antagonist, we demonstrated that GnRH induces nuclear accumulation of FANCA and green fluorescent protein (GFP)-FANCA before exporting back to the cytoplasm using the nuclear export receptor CRM1. Using FANCA point mutations that locate GFP-FANCA to the cytoplasm (H1110P) or functionally uncouple GFP-FANCA (Q1128E) from the wild-type nucleocytoplasmic distribution pattern, we demonstrated that wild-type FANCA was required for GnRH-induced activation of gonadotrope cell markers. Cotransfection of H1110P and Q1128E blocked GnRH activation of the alphaGsu and GnRHR but not the beta-subunit gene promoters. We conclude that nucleocytoplasmic shuttling of FANCA is required for GnRH transduction of the alphaGSU and GnRHR gene promoters and propose that FANCA functions as a GnRH-induced signal transducer.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Proteínas de Transporte Nucleocitoplasmático/fisiología , Receptores LHRH/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Células Cultivadas , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Hormona Folículo Estimulante de Subunidad beta/genética , Hormonas Glicoproteicas de Subunidad alfa/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Carioferinas/fisiología , Hormona Luteinizante de Subunidad beta/genética , Proteínas Mutantes Quiméricas/metabolismo , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores LHRH/genética , Distribución Tisular , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA