Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 29(7): 1083-1095, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31628488

RESUMEN

Fanconi anemia (FA) is a chromosome instability syndrome characterized by increased cancer predisposition. Specifically, the FA pathway functions to protect genome stability during DNA replication. The central FA pathway protein, FANCD2, locates to stalled replication forks and recruits homologous recombination (HR) factors such as CtBP interacting protein (CtIP) to promote replication fork restart while suppressing new origin firing. Here, we identify alpha-thalassemia retardation syndrome X-linked (ATRX) as a novel physical and functional interaction partner of FANCD2. ATRX is a chromatin remodeler that forms a complex with Death domain-associated protein 6 (DAXX) to deposit the histone variant H3.3 into specific genomic regions. Intriguingly, ATRX was recently implicated in replication fork recovery; however, the underlying mechanism(s) remained incompletely understood. Our findings demonstrate that ATRX forms a constitutive protein complex with FANCD2 and protects FANCD2 from proteasomal degradation. ATRX and FANCD2 localize to stalled replication forks where they cooperate to recruit CtIP and promote MRE11 exonuclease-dependent fork restart while suppressing the firing of new replication origins. Remarkably, replication restart requires the concerted histone H3 chaperone activities of ATRX/DAXX and FANCD2, demonstrating that coordinated histone H3 variant deposition is a crucial event during the reinitiation of replicative DNA synthesis. Lastly, ATRX also cooperates with FANCD2 to promote the HR-dependent repair of directly induced DNA double-stranded breaks. We propose that ATRX is a novel functional partner of FANCD2 to promote histone deposition-dependent HR mechanisms in S-phase.


Asunto(s)
Proteínas Co-Represoras/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Chaperonas Moleculares/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Línea Celular , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Replicación del ADN/genética , Anemia de Fanconi/patología , Técnicas de Inactivación de Genes/métodos , Histonas/genética , Humanos , Proteína Homóloga de MRE11/genética , Recombinasa Rad51/genética , Reparación del ADN por Recombinación/genética , Transducción de Señal/genética
2.
Nucleic Acids Res ; 48(D1): D704-D715, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31701156

RESUMEN

In biology and biomedicine, relating phenotypic outcomes with genetic variation and environmental factors remains a challenge: patient phenotypes may not match known diseases, candidate variants may be in genes that haven't been characterized, research organisms may not recapitulate human or veterinary diseases, environmental factors affecting disease outcomes are unknown or undocumented, and many resources must be queried to find potentially significant phenotypic associations. The Monarch Initiative (https://monarchinitiative.org) integrates information on genes, variants, genotypes, phenotypes and diseases in a variety of species, and allows powerful ontology-based search. We develop many widely adopted ontologies that together enable sophisticated computational analysis, mechanistic discovery and diagnostics of Mendelian diseases. Our algorithms and tools are widely used to identify animal models of human disease through phenotypic similarity, for differential diagnostics and to facilitate translational research. Launched in 2015, Monarch has grown with regards to data (new organisms, more sources, better modeling); new API and standards; ontologies (new Mondo unified disease ontology, improvements to ontologies such as HPO and uPheno); user interface (a redesigned website); and community development. Monarch data, algorithms and tools are being used and extended by resources such as GA4GH and NCATS Translator, among others, to aid mechanistic discovery and diagnostics.


Asunto(s)
Biología Computacional/métodos , Genotipo , Fenotipo , Algoritmos , Animales , Ontologías Biológicas , Bases de Datos Genéticas , Exoma , Estudios de Asociación Genética , Variación Genética , Genómica , Humanos , Internet , Programas Informáticos , Investigación Biomédica Traslacional , Interfaz Usuario-Computador
3.
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
4.
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
5.
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
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.
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
8.
J Biol Chem ; 284(38): 25560-8, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19633289

RESUMEN

Genomic stability requires a functional Fanconi anemia (FA) pathway composed of an upstream "core complex" (FA proteins A/B/C/E/F/G/L/M) that mediates monoubiquitination of the downstream targets FANCD2 and FANCI. Unique among FA core complex members, FANCM has processing activities toward replication-associated DNA structures, suggesting a vital role for FANCM during replication. Using Xenopus egg extracts, we analyzed the functions of FANCM in replication and the DNA damage response. xFANCM binds chromatin in a replication-dependent manner and is phosphorylated in response to DNA damage structures. Chromatin binding and DNA damage-induced phosphorylation of xFANCM are mediated in part by the downstream FA pathway protein FANCD2. Moreover, phosphorylation and chromatin recruitment of FANCM is regulated by two mayor players in the DNA damage response: the cell cycle checkpoint kinases ATR and ATM. Our results indicate that functions of FANCM are controlled by FA- and non-FA pathways in the DNA damage response.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN/fisiología , ADN Helicasas/metabolismo , Replicación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Oocitos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Cromatina , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Oocitos/citología , Fosforilación/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética , Proteínas de Xenopus/genética , Xenopus laevis
9.
Stem Cells ; 27(12): 2884-95, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19711456

RESUMEN

Members of the Mix/Bix family of paired class homeobox genes play important roles in the development of vertebrate mesoderm and endoderm. The single Mix/Bix family member identified in the mouse, Mix-like 1 (Mixl1), is required for mesendoderm patterning during gastrulation and promotes mesoderm formation and hematopoiesis in embryonic stem cell (ESC)-derived embryoid bodies. Despite its crucial functions the transcriptional activity and targets of Mixl1 have not been well described. To investigate the molecular mechanisms of Mixl1-mediated transcriptional regulation, we have characterized the DNA-binding specificity and transcriptional properties of this homeodomain protein in differentiating ESCs. Mixl1 binds preferentially as a dimer to an 11-base pair (bp) Mixl1 binding sequence (MBS) that contains two inverted repeats separated by a 3-bp spacer. The MBS mediates transcriptional activation by Mixl1 in both NIH 3T3 cells and in a new application of an inducible ESC differentiation system. Consistent with our previous observation that early induction of Mixl1 expression in ESCs results in premature activation of Goosecoid (Gsc), we have found that Mixl1 occupies two variant MBSs within and activates transcription from the Gsc promoter in vitro and in vivo. These results strongly suggest that Gsc is a direct target gene of Mixl1 during embryogenesis. STEM CELLS 2009;27:2884-2895.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Activación Transcripcional , Animales , Secuencia de Bases , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Proteína Goosecoide/genética , Proteínas de Homeodominio/genética , Mesodermo/metabolismo , Ratones , Células 3T3 NIH , Unión Proteica , Alineación de Secuencia
10.
Database (Oxford) ; 20202020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32283553

RESUMEN

Hypothesis generation is a critical step in research and a cornerstone in the rare disease field. Research is most efficient when those hypotheses are based on the entirety of knowledge known to date. Systematic review articles are commonly used in biomedicine to summarize existing knowledge and contextualize experimental data. But the information contained within review articles is typically only expressed as free-text, which is difficult to use computationally. Researchers struggle to navigate, collect and remix prior knowledge as it is scattered in several silos without seamless integration and access. This lack of a structured information framework hinders research by both experimental and computational scientists. To better organize knowledge and data, we built a structured review article that is specifically focused on NGLY1 Deficiency, an ultra-rare genetic disease first reported in 2012. We represented this structured review as a knowledge graph and then stored this knowledge graph in a Neo4j database to simplify dissemination, querying and visualization of the network. Relative to free-text, this structured review better promotes the principles of findability, accessibility, interoperability and reusability (FAIR). In collaboration with domain experts in NGLY1 Deficiency, we demonstrate how this resource can improve the efficiency and comprehensiveness of hypothesis generation. We also developed a read-write interface that allows domain experts to contribute FAIR structured knowledge to this community resource. In contrast to traditional free-text review articles, this structured review exists as a living knowledge graph that is curated by humans and accessible to computational analyses. Finally, we have generalized this workflow into modular and repurposable components that can be applied to other domain areas. This NGLY1 Deficiency-focused network is publicly available at http://ngly1graph.org/. AVAILABILITY AND IMPLEMENTATION: Database URL: http://ngly1graph.org/. Network data files are at: https://github.com/SuLab/ngly1-graph and source code at: https://github.com/SuLab/bioknowledge-reviewer. CONTACT: asu@scripps.edu.


Asunto(s)
Investigación Biomédica/métodos , Biología Computacional/métodos , Bases de Datos Factuales , Bases del Conocimiento , Animales , Investigación Biomédica/estadística & datos numéricos , Biología Computacional/estadística & datos numéricos , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Curaduría de Datos/métodos , Minería de Datos/métodos , Humanos , Internet , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Revisiones Sistemáticas como Asunto
11.
DNA Repair (Amst) ; 7(12): 1973-81, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18786657

RESUMEN

Fanconi anemia (FA) is a recessive genetic disorder characterized by hypersensitivity to crosslinking agents that has been attributed to defects in DNA repair and/or replication. FANCD2 and the FA core complex bind to chromatin during DNA replication; however, the role of FA proteins during replication is unknown. Using Xenopus cell-free extracts, we show that FANCL depletion results in defective DNA replication restart following treatment with camptothecin, a drug that results in DSBs during DNA replication. This defect is more pronounced following treatment with mitomycin C, presumably because of an additional role of the FA pathway in DNA crosslink repair. Moreover, we show that chromatin binding of FA core complex proteins during DNA replication follows origin assembly and origin firing and is dependent on the binding of RPA to ssDNA while FANCD2 additionally requires ATR, consistent with FA proteins acting at replication forks. Together, our data suggest that FA proteins play a role in replication restart at collapsed replication forks.


Asunto(s)
Cromatina/metabolismo , Replicación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Proteína del Grupo de Complementación L de la Anemia de Fanconi/fisiología , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Afidicolina/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Camptotecina/farmacología , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Sistema Libre de Células , Daño del ADN , Reparación del ADN , Inhibidores Enzimáticos/farmacología , Anemia de Fanconi , Mitomicina/farmacología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis
12.
Mol Cancer ; 8: 133, 2009 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-20043851

RESUMEN

BACKGROUND: The Fanconi anemia (FA) pathway is a multigene DNA damage response network implicated in the repair of DNA lesions that arise during replication or after exogenous DNA damage. The FA pathway displays synthetic lethal relationship with certain DNA repair genes such as ATM (Ataxia Telangectasia Mutated) that are frequently mutated in tumors. Thus, inhibition of FANCD2 monoubiquitylation (FANCD2-Ub), a key step in the FA pathway, might target tumor cells defective in ATM through synthetic lethal interaction. Curcumin was previously identified as a weak inhibitor of FANCD2-Ub. The aim of this study is to identify derivatives of curcumin with better activity and specificity. RESULTS: Using a replication-free assay in Xenopus extracts, we screened monoketone analogs of curcumin for inhibition of FANCD2-Ub and identified analog EF24 as a strong inhibitor. Mechanistic studies suggest that EF24 targets the FA pathway through inhibition of the NF-kB pathway kinase IKK. In HeLa cells, nanomolar concentrations of EF24 inhibited hydroxyurea (HU)-induced FANCD2-Ub and foci in a cell-cycle independent manner. Survival assays revealed that EF24 specifically sensitizes FA-competent cells to the DNA crosslinking agent mitomycin C (MMC). In addition, in contrast with curcumin, ATM-deficient cells are twofold more sensitive to EF24 than matched wild-type cells, consistent with a synthetic lethal effect between FA pathway inhibition and ATM deficiency. An independent screen identified 4H-TTD, a compound structurally related to EF24 that displays similar activity in egg extracts and in cells. CONCLUSIONS: These results suggest that monoketone analogs of curcumin are potent inhibitors of the FA pathway and constitute a promising new class of targeted anticancer compounds.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Curcumina/análogos & derivados , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/antagonistas & inhibidores , Anemia de Fanconi/metabolismo , Cetonas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Curcumina/farmacología , Proteínas de Unión al ADN/metabolismo , Sinergismo Farmacológico , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Mitomicina/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Xenopus
13.
Int J Cancer ; 124(4): 783-92, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19048618

RESUMEN

The Fanconi Anemia (FA) DNA damage response pathway is involved in the processing of DNA interstrand crosslinks (ICLs). As such, inhibition of the FA pathway could chemosensitize FA-competent tumor cells to commonly used ICL agents like cisplatin. Moreover, suppression of the FA pathway is synthetic lethal with deficiencies in several other DNA repair pathways, suggesting that FA pathway inhibitors could be used in targeted therapies against specific tumors. To identify such inhibitors, we designed a novel in vitro screening assay utilizing Xenopus egg extracts. Using the DNA-stimulated monoubiquitylation of Xenopus FANCD2 (xFANCD2-L) as readout, a chemical library screen identified DDN (2,3-dichloro-5,8-dihydroxy-1,4-naphthoquinone) as a novel and potent FA pathway inhibitor. DDN inhibited xFANCD2-L formation in a dose-dependent manner in both extracts and human cells without disruption of the upstream FA core complex. DDN also inhibited the characteristic subnuclear FANCD2 foci formation following DNA damage. Moreover, DDN displayed a greater synergistic effect with cisplatin in a FA-proficient cancer cell line compared to its FA-deficient isogenic counterpart, suggesting that DDN might be a good lead candidate as cisplatin chemosensitizer in both FA-deficient and FA-competent tumors. This system constitutes the first cell-free screening assay for identifying compounds that inhibit the FA pathway and provides a new biochemical platform for mapping the functions of its various components with specific chemical inhibitors.


Asunto(s)
Evaluación Preclínica de Medicamentos/instrumentación , Evaluación Preclínica de Medicamentos/métodos , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/genética , Animales , Supervivencia Celular , Sistema Libre de Células , Cisplatino/farmacología , Reactivos de Enlaces Cruzados/farmacología , ADN/química , Daño del ADN , Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Xenopus laevis
14.
Mol Cell Biol ; 26(2): 425-37, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16382135

RESUMEN

Fanconi anemia (FA) is a multigene cancer susceptibility disorder characterized by cellular hypersensitivity to DNA interstrand cross-linking agents such as mitomycin C (MMC). FA proteins are suspected to function at the interface between cell cycle checkpoints, DNA repair, and DNA replication. Using replicating extracts from Xenopus eggs, we developed cell-free assays for FA proteins (xFA). Recruitment of the xFA core complex and xFANCD2 to chromatin is strictly dependent on replication initiation, even in the presence of MMC indicating specific recruitment to DNA lesions encountered by the replication machinery. The increase in xFA chromatin binding following treatment with MMC is part of a caffeine-sensitive S-phase checkpoint that is controlled by xATR. Recruitment of xFANCD2, but not xFANCA, is dependent on the xATR-xATR-interacting protein (xATRIP) complex. Immunodepletion of either xFANCA or xFANCD2 from egg extracts results in accumulation of chromosomal DNA breaks during replicative synthesis. Our results suggest coordinated chromatin recruitment of xFA proteins in response to replication-associated DNA lesions and indicate that xFA proteins function to prevent the accumulation of DNA breaks that arise during unperturbed replication.


Asunto(s)
Proteínas Portadoras/metabolismo , Daño del ADN/fisiología , Replicación del ADN , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Proteínas de Xenopus/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Cafeína/farmacología , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Reactivos de Enlaces Cruzados/farmacología , Reparación del ADN/fisiología , Femenino , Técnicas In Vitro , Mitomicina/farmacología , Datos de Secuencia Molecular , Oocitos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S/efectos de los fármacos , Fase S/fisiología , Homología de Secuencia de Aminoácido , Xenopus laevis
15.
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
16.
Mol Cell Biol ; 23(10): 3417-26, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12724401

RESUMEN

Bloom syndrome (BS) is a genetic disorder associated with dwarfism, immunodeficiency, reduced fertility, and an elevated risk of cancer. To investigate the mechanism of this disease, we isolated from human HeLa extracts three complexes containing the helicase defective in BS, BLM. Interestingly, one of the complexes, termed BRAFT, also contains five of the Fanconi anemia (FA) complementation group proteins (FA proteins). FA resembles BS in genomic instability and cancer predisposition, but most of its gene products have no known biochemical activity, and the molecular pathogenesis of the disease is poorly understood. BRAFT displays a DNA-unwinding activity, which requires the presence of BLM because complexes isolated from BLM-deficient cells lack such an activity. The complex also contains topoisomerase IIIalpha and replication protein A, proteins that are known to interact with BLM and could facilitate unwinding of DNA. We show that BLM complexes isolated from an FA cell line have a lower molecular mass. Our study provides the first biochemical characterization of a multiprotein FA complex and suggests a connection between the BLM and FA pathways of genomic maintenance. The findings that FA proteins are part of a DNA-unwinding complex imply that FA proteins may participate in DNA repair.


Asunto(s)
Síndrome de Bloom/genética , Anemia de Fanconi/genética , Adenosina Trifosfatasas/metabolismo , Anticuerpos Monoclonales/metabolismo , Síndrome de Bloom/metabolismo , Western Blotting , Núcleo Celular/metabolismo , ADN Helicasas/metabolismo , Reparación del ADN , ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas de Unión al ADN/metabolismo , Electroforesis en Gel de Poliacrilamida , Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Immunoblotting , Pruebas de Precipitina , RecQ Helicasas , Proteína de Replicación A , Ubiquitina/metabolismo
17.
Cancer Res ; 63(10): 2688-94, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12750298

RESUMEN

A 2-year old boy was diagnosed with Fanconi anemia (FA) and acute myeloid leukemia (AML). A cell line (termed FA-AML1) was established from blast cells obtained after a second relapse after a successful bone marrow transplant. Histochemical and surface marker analysis confirmed that the cells were derived from the myeloid lineage. Cytogenetic analysis revealed multiple chromosomal aberrations, including a ring 7. Stable proliferation of the cultured cells was absolutely dependent on the presence of granulocyte macrophage colony-stimulating factor or interleukin 3. This is the first AML cell line successfully established from a FA patient. Remarkably, FA-AML1 cells appeared to lack the characteristic cellular FA phenotype, i.e., a hypersensitivity to growth inhibition and chromosomal breakage by the cross-linking agent mitomycin C. Genomic DNA from the patient showed biallelic mutations [8415G>T (K2729N)and 8732C>A (S2835STOP)] in the breast cancer susceptibility gene FANCD1/BRCA2 [N. Howlett et al., Science (Wash. DC), 297: 606-609, 2002]. In the AML cells, however, the 8732C>A nonsense mutation was changed into a missense mutation by a secondary alteration, 8731T>G, resulting in 2835E, which restored the open-reading frame of the gene and could explain the reverted phenotype of these cells. Loss of the FA phenotype by genetic correction of a FA gene mutation during AML progression may be a common late event in the pathogenesis of AML in FA patients, which may be treatment related. This finding suggests a novel mechanistic principle of tumor progression based on the genetic correction of an early caretaker gene defect.


Asunto(s)
Anemia de Fanconi/genética , Genes BRCA2 , Leucemia Mieloide Aguda/genética , Mutación , Células Tumorales Cultivadas , Alelos , Antígenos CD/biosíntesis , División Celular/efectos de los fármacos , Preescolar , Anemia de Fanconi/complicaciones , Anemia de Fanconi/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-3/farmacología , Cariotipificación , Leucemia Mieloide Aguda/complicaciones , Leucemia Mieloide Aguda/patología , Masculino
18.
Anemia ; 2012: 481583, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22693661

RESUMEN

The Fanconi Anemia (FA) pathway consists of proteins involved in repairing DNA damage, including interstrand cross-links (ICLs). The pathway contains an upstream multiprotein core complex that mediates the monoubiquitylation of the FANCD2 and FANCI heterodimer, and a downstream pathway that converges with a larger network of proteins with roles in homologous recombination and other DNA repair pathways. Selective killing of cancer cells with an intact FA pathway but deficient in certain other DNA repair pathways is an emerging approach to tailored cancer therapy. Inhibiting the FA pathway becomes selectively lethal when certain repair genes are defective, such as the checkpoint kinase ATM. Inhibiting the FA pathway in ATM deficient cells can be achieved with small molecule inhibitors, suggesting that new cancer therapeutics could be developed by identifying FA pathway inhibitors to treat cancers that contain defects that are synthetic lethal with FA.

19.
PLoS One ; 7(1): e30042, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253870

RESUMEN

BACKGROUND: The consequences of defective homologous recombination (HR) are not understood in sporadic ovarian cancer, nor have the potential role of HR proteins other than BRCA1 and BRCA2 been clearly defined. However, it is clear that defects in HR and other DNA repair pathways are important to the effectiveness of current therapies. We hypothesize that a subset of sporadic ovarian carcinomas may harbor anomalies in HR pathways, and that a BRCAness profile (defects in HR or other DNA repair pathways) could influence response rate and survival after treatment with platinum drugs. Clinical availability of a BRCAness profile in patients and/or tumors should improve treatment outcomes. OBJECTIVE: To define the BRCAness profile of sporadic ovarian carcinoma and determine whether BRCA1, PARP, FANCD2, PTEN, H2AX, ATM, and P53 protein expression correlates with response to treatment, disease recurrence, and recurrence-free survival. MATERIALS AND METHODS: Protein microarray analysis of ovarian cancer tissue was used to determine protein expression levels for defined DNA repair proteins. Correlation with clinical and pathologic parameters in 186 patients with advanced stage III-IV and grade 3 ovarian cancer was analyzed using Chi square, Kaplan-Meier method, Cox proportional hazard model, and cumulative incidence function. RESULTS: High PARP, FANCD2 and BRCA1 expressions were significantly correlated with each other; however, elevated p53 expression was associated only with high PARP and FANCD2. Of all patients, 9% recurred within the first year. Among early recurring patients, 41% had high levels of PARP, FANCD2 and P53, compared to 19.5% of patients without early recurrence (p = 0.04). Women with high levels of PARP, FANCD2 and/or P53 had first year cumulative cancer incidence of 17% compared with 7% for the other groups (P = 0.03). CONCLUSIONS: Patients with concomitantly high levels of PARP, FANCD2 and P53 protein expression are at increased risk of early ovarian cancer recurrence and platinum resistance.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Recurrencia Local de Neoplasia/patología , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Epitelial de Ovario , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Pronóstico , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
DNA Repair (Amst) ; 10(12): 1252-61, 2011 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-22036606

RESUMEN

Fanconi anemia (FA) is a heritable disease characterized by bone marrow failure, congenital abnormalities, and cancer predisposition. The 15 identified FA genes operate in a molecular pathway to preserve genomic integrity. Within this pathway the FA core complex operates as an ubiquitin ligase that activates the complex of FANCD2 and FANCI to coordinate DNA repair. The FA core complex is formed by at least 12 proteins. However, only the FANCL subunit displays ubiquitin ligase activity. FANCA and FANCG are members of the FA core complex for which no other functions have been described than to participate in protein interactions. In this study we generated mice with combined null alleles for Fanca and Fancg to identify extended functions for these genes by characterizing the double mutant mice and cells. Double mutant a(-/-)/g(-/-) mice were born at near Mendelian frequencies without apparent developmental abnormalities. Histological analysis of a(-/-)/g(-/-) mice revealed a Leydig cell hyperplasia and frequent vacuolization of Sertoli cells in testes, while ovaries were depleted from developing follicles and displayed an interstitial cell hyperplasia. These gonadal aberrations were associated with a compromised fertility of a(-/-)/g(-/-) males and females. During the first year of life a(-/-)/g(-/-) did not develop malignancies or bone marrow failure. At the cellular level a(-/-)/g(-/-), Fanca(-/-), and Fancg(-/-) cells proved equally compromised in DNA crosslink and homology-directed repair. Overall the phenotype of a(-/-)/g(-/-) double knockout mice and cells appeared highly similar to the phenotype of Fanca or Fancg single knockouts. The lack of an augmented phenotype suggest that null mutations in Fanca or Fancg are fully epistatic, making additional important functions outside of the FA core complex highly unlikely.


Asunto(s)
Epistasis Genética/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Mutación/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Rotura Cromosómica/efectos de los fármacos , Embrión de Mamíferos , Femenino , Fertilidad/genética , Fibroblastos/citología , Fluorobencenos/farmacología , Pruebas Hematológicas , Humanos , Masculino , Ratones , Ovario/metabolismo , Ftalazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA