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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 71(1): 25-41.e6, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29937342

RESUMEN

Components of the Fanconi anemia and homologous recombination pathways play a vital role in protecting newly replicated DNA from uncontrolled nucleolytic degradation, safeguarding genome stability. Here we report that histone methylation by the lysine methyltransferase SETD1A is crucial for protecting stalled replication forks from deleterious resection. Depletion of SETD1A sensitizes cells to replication stress and leads to uncontrolled DNA2-dependent resection of damaged replication forks. The ability of SETD1A to prevent degradation of these structures is mediated by its ability to catalyze methylation on Lys4 of histone H3 (H3K4) at replication forks, which enhances FANCD2-dependent histone chaperone activity. Suppressing H3K4 methylation or expression of a chaperone-defective FANCD2 mutant leads to loss of RAD51 nucleofilament stability and severe nucleolytic degradation of replication forks. Our work identifies epigenetic modification and histone mobility as critical regulatory mechanisms in maintaining genome stability by restraining nucleases from irreparably damaging stalled replication forks.


Asunto(s)
ADN/biosíntesis , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Chaperonas Moleculares/metabolismo , Nucleosomas/metabolismo , Células A549 , ADN/genética , Replicación del ADN/fisiología , Epigénesis Genética/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Células HeLa , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Humanos , Metilación , Chaperonas Moleculares/genética , Nucleosomas/genética , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo
2.
Mol Cell ; 64(2): 388-404, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27768874

RESUMEN

Common fragile sites (CFSs) are genomic regions that are unstable under conditions of replicative stress. Although the characteristics of CFSs that render them vulnerable to stress are associated mainly with replication, the cellular pathways that protect CFSs during replication remain unclear. Here, we identify and describe a role for FANCD2 as a trans-acting facilitator of CFS replication, in the absence of exogenous replicative stress. In the absence of FANCD2, replication forks stall within the AT-rich fragility core of CFS, leading to dormant origin activation. Furthermore, FANCD2 deficiency is associated with DNA:RNA hybrid formation at CFS-FRA16D, and inhibition of DNA:RNA hybrid formation suppresses replication perturbation. In addition, we also found that FANCD2 reduces the number of potential sites of replication initiation. Our data demonstrate that FANCD2 protein is required to ensure efficient CFS replication and provide mechanistic insight into how FANCD2 regulates CFS stability.


Asunto(s)
Sitios Frágiles del Cromosoma , Replicación del ADN , ADN/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , ARN/genética , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Línea Celular Transformada , ADN/metabolismo , Anemia de Fanconi , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , 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/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Expresión Génica , Inestabilidad Genómica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Linfocitos/citología , Linfocitos/metabolismo , ARN/metabolismo
3.
Nucleic Acids Res ; 48(10): 5467-5484, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32329774

RESUMEN

Transcription-replication (T-R) conflicts are profound threats to genome integrity. However, whilst much is known about the existence of T-R conflicts, our understanding of the genetic and temporal nature of how cells respond to them is poorly established. Here, we address this by characterizing the early cellular response to transient T-R conflicts (TRe). This response specifically requires the DNA recombination repair proteins BLM and BRCA2 as well as a non-canonical monoubiquitylation-independent function of FANCD2. A hallmark of the TRe response is the rapid co-localization of these three DNA repair factors at sites of T-R collisions. We find that the TRe response relies on basal activity of the ATR kinase, yet it does not lead to hyperactivation of this key checkpoint protein. Furthermore, specific abrogation of the TRe response leads to DNA damage in mitosis, and promotes chromosome instability and cell death. Collectively our findings identify a new role for these well-established tumor suppressor proteins at an early stage of the cellular response to conflicts between DNA transcription and replication.


Asunto(s)
Replicación del ADN , Reparación del ADN por Recombinación , Transcripción Genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA2/fisiología , Línea Celular , Supervivencia Celular , Quinasa 9 Dependiente de la Ciclina/metabolismo , ADN/metabolismo , Daño del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Humanos , Mitosis/genética , Regiones Promotoras Genéticas , ARN/metabolismo , ARN Polimerasa II/metabolismo , Empalme del ARN , RecQ Helicasas/fisiología , Ubiquitinación
4.
Bioconjug Chem ; 28(11): 2729-2736, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29035511

RESUMEN

We present an approach to tuning the multifunctionality of iron oxide nanoparticles (IONs) using mixed self-assembled monolayers of cationic lipid and anionic polyethylene glycol (PEG) lipid. By forming stable, monodispersed lipid-coated IONs (L-IONs) through a solvent-exchange technique, we were able to demonstrate the relationship between surface charge, the magnetic transverse relaxivity (r2 from T2-weighted images), and the binding capacity of small interfering ribonucleic acids (siRNAs) as a function of the cationic-to-anionic (PEG) lipid ratio. These properties were controlled by the cationic charge and the PEG conformation; relaxivity and siRNA binding could be varied in the mushroom and brush regimes but not at high brush densities. In vitro results combining cell viability, uptake, and transfection efficiency using HeLa cells suggest that the functional physicochemical and biological properties of L-IONs may be best achieved using catanionic lipid coatings near equimolar ratios of cationic to anionic PEG-lipids.


Asunto(s)
Compuestos Férricos/química , Lípidos/química , Nanopartículas/química , Polietilenglicoles/química , Células HeLa , Humanos , Nanopartículas de Magnetita/química , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química , Transfección
5.
Blood ; 120(10): 2109-17, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22855611

RESUMEN

The Fanconi anemia (FA)-BRCA pathway is critical for the repair of DNA interstrand crosslinks (ICLs) and the maintenance of chromosome stability. A key step in FA-BRCA pathway activation is the covalent attachment of monoubiquitin to FANCD2 and FANCI. Monoubiquitinated FANCD2 and FANCI localize in chromatin-associated nuclear foci where they interact with several well-characterized DNA repair proteins. Importantly, very little is known about the structure, function, and regulation of FANCD2. Herein, we describe the identification and characterization of a CUE (coupling of ubiquitin conjugation to endoplasmic reticulum degradation) ubiquitin-binding domain (UBD) in FANCD2, and demonstrate that the CUE domain mediates noncovalent binding to ubiquitin in vitro. We show that although mutation of the CUE domain destabilizes FANCD2, the protein remains competent for DNA damage-inducible monoubiquitination and phosphorylation. Importantly, we demonstrate that the CUE domain is required for interaction with FANCI, retention of monoubiquitinated FANCD2, and FANCI in chromatin, and for efficient ICL repair. Our results suggest a model by which heterodimerization of monoubiquitinated FANCD2 and FANCI in chromatin is mediated in part through a noncovalent interaction between the FANCD2 CUE domain and monoubiquitin covalently attached to FANCI, and that this interaction shields monoubiquitinated FANCD2 from polyubiquitination and proteasomal degradation.


Asunto(s)
Reparación del ADN/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Transducción de Señal/genética , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Cromatina/genética , Inestabilidad Cromosómica , Daño 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 , Células HeLa , Humanos , Datos de Secuencia Molecular , Mutación , Fosforilación , Plásmidos , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Transfección , Ubiquitinación
6.
Nucleic Acids Res ; 40(17): 8348-60, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22735704

RESUMEN

p21 is a well-established regulator of cell cycle progression. The role of p21 in DNA repair, however, remains poorly characterized. Here, we describe a critical role of p21 in a replication-coupled DNA double-strand break (DSB) repair that is mechanistically distinct from its cell cycle checkpoint function. We demonstrate that p21-deficient cells exhibit elevated chromatid-type aberrations, including gaps and breaks, dicentrics and radial formations, following exposure to several DSB-inducing agents. p21-/- cells also exhibit an increased DNA damage-inducible DNA-PKCS S2056 phosphorylation, indicative of elevated non-homologous DNA end joining. Concomitantly, p21-/- cells are defective in replication-coupled homologous recombination (HR), exhibiting decreased sister chromatid exchanges and HR-dependent repair as determined using a crosslinked GFP reporter assay. Importantly, we establish that the DSB hypersensitivity of p21-/- cells is associated with increased cyclin-dependent kinase (CDK)-dependent BRCA2 S3291 phosphorylation and MRE11 nuclear foci formation and can be rescued by inhibition of CDK or MRE11 nuclease activity. Collectively, our results uncover a novel mechanism by which p21 regulates the fidelity of replication-coupled DSB repair and the maintenance of chromosome stability distinct from its role in the G1-S phase checkpoint.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN , Replicación del ADN , Animales , Proteína BRCA2/metabolismo , Camptotecina/toxicidad , Inestabilidad Cromosómica , Reactivos de Enlaces Cruzados/toxicidad , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Reparación del ADN por Unión de Extremidades , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Etopósido/toxicidad , Puntos de Control de la Fase G1 del Ciclo Celular , Células HCT116 , Células HeLa , Humanos , Proteína Homóloga de MRE11 , Ratones , Mitomicina/toxicidad , Fosforilación , Reparación del ADN por Recombinación , Inhibidores de Topoisomerasa/toxicidad
7.
Molecules ; 19(9): 13319-31, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25170952

RESUMEN

Previously, we have reported the synthesis of a homochiral l-cyclic peptide [WR]5 and its use for delivery of anti-HIV drugs and biomolecules. A physical mixture of HAuCl4 and the peptide generated peptide-capped gold nanoparticles. Here, [WR]5 and [WR]5-AuNPs were tested for their efficiency to deliver a small interfering RNA molecule (siRNA) in human cervix adenocarcinoma (HeLa) cells. Flow cytometry investigation revealed that the intracellular uptake of a fluorescence-labeled non-targeting siRNA (200 nM) was enhanced in the presence of [WR]5 and [WR]5-AuNPs by 2- and 3.8-fold when compared with that of siRNA alone after 24 h incubation. Comparative toxicity results showed that [WR]5 and [WR]5-AuNPs were less toxic in cells compared to other available carrier systems, such as Lipofectamine.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , Péptidos Cíclicos/química , ARN Interferente Pequeño/metabolismo , Transfección , Secuencia de Aminoácidos , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/metabolismo , Técnicas de Silenciamiento del Gen/métodos , Células HeLa , Humanos , Péptidos Cíclicos/metabolismo , ARN Interferente Pequeño/genética
8.
Mutat Res ; 755(2): 135-40, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23796964

RESUMEN

Hepatocellular carcinoma (HCC) is non-responsive to many chemotherapeutic agents including etoposide. The aim of this study was to examine the survival strategy of the HCC cell line HepG2 after etoposide treatment. Here we analyzed and compared spontaneous and etoposide-induced DNA damage in HepG2 (α-fetoprotein (AFP)-positive) and Chang Liver (AFP-negative) cell lines. Compared to Chang Liver cells, HepG2 cells exhibited a significantly higher degree of micronucleation and a higher nuclear division index, as determined by the cytokinesis-block micronucleus assay, following exposure to etoposide. HepG2 cells were also more resistant to etoposide-induced cytotoxicity compared to Chang Liver cells. We also establish that increased etoposide-induced multinucleation in HepG2 cells is dependent on the catalytic activity of Akt, as phosphatidylinositol-3-kinase inhibitors as well as the overexpression of kinase-defective Akt reversed this phenotype. Moreover, ectopic expression of wild type PTEN reduced the frequency of etoposide-induced multinucleated HepG2 cells, and restored HepG2 etoposide sensitivity. Taken together, these results implicate the Akt/PTEN cellular axis as a major determinant of the etoposide resistance of HCC cells.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Resistencia a Antineoplásicos/fisiología , Etopósido/farmacología , Células Hep G2/efectos de los fármacos , Proteínas de Neoplasias/fisiología , Fosfohidrolasa PTEN/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Inhibidores de Topoisomerasa II/farmacología , Fusión Celular , Supervivencia Celular , Daño del ADN , Reparación del ADN/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , Células Gigantes/efectos de los fármacos , Células Gigantes/fisiología , Células HeLa/efectos de los fármacos , Humanos , Pruebas de Micronúcleos , Proteínas de Neoplasias/genética , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
9.
bioRxiv ; 2023 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-37066159

RESUMEN

Fanconi anemia (FA) is a rare genetic disease characterized by heterogeneous congenital abnormalities and increased risk for bone marrow failure and cancer. FA is caused by mutation of any one of 23 genes, the protein products of which function primarily in the maintenance of genome stability. An important role for the FA proteins in the repair of DNA interstrand crosslinks (ICLs) has been established in vitro . While the endogenous sources of ICLs relevant to the pathophysiology of FA have yet to be clearly determined, a role for the FA proteins in a two-tier system for the detoxification of reactive metabolic aldehydes has been established. To discover new metabolic pathways linked to FA, we performed RNA-seq analysis on non-transformed FA-D2 ( FANCD2 -/- ) and FANCD2-complemented patient cells. Multiple genes associated with retinoic acid metabolism and signaling were differentially expressed in FA-D2 ( FANCD2 -/- ) patient cells, including ALDH1A1 and RDH10 , which encode for retinaldehyde and retinol dehydrogenases, respectively. Increased levels of the ALDH1A1 and RDH10 proteins was confirmed by immunoblotting. FA-D2 ( FANCD2 -/- ) patient cells displayed increased aldehyde dehydrogenase activity compared to the FANCD2-complemented cells. Upon exposure to retinaldehyde, FA-D2 ( FANCD2 -/- ) cells exhibited increased DNA double-strand breaks and checkpoint activation indicative of a defect in the repair of retinaldehyde-induced DNA damage. Our findings describe a novel link between retinoic acid metabolism and FA and identify retinaldehyde as an additional reactive metabolic aldehyde relevant to the pathophysiology of FA.

10.
R I Med J (2013) ; 104(2): 25-29, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33648315

RESUMEN

The overarching goal of the Rhode Island-IDeA Network of Biomedical Research Excellence (RI-INBRE) is to improve institutional capacity for biomedical research excellence and expand student experiential training opportunities in the State of Rhode Island. RI-INBRE comprises five major core components: The Administrative Core, the Bioinformatics Core, the Centralized Research Core Facility, the Training Core, and the Developmental Research Project Program Core. Since its inception in 2001, RI-INBRE has made significant investments and marked advancements in the biomedical research infrastructure of Rhode Island. RI-INBRE funding has increased the scale and quality of faculty research and engaged undergraduate students, graduate students, and postdoctoral fellows in structured and mentored research training experiences. Over the last 19 years, RI-INBRE has supported 212 faculty researchers and over 533 projects and has provided research-training opportunities for nearly 2,000 students, resulting in 757 publications. Through its student-training program, RI-INBRE has contributed to regional workforce development by engaging students and encouraging them to pursue careers in biomedical fields. Many of these students have been admitted to graduate or medical schools and obtained biomedical industry jobs following graduation. RI-INBRE has been particularly influential in building the research infrastructure at primarily undergraduate institutions, which have seen significant improvements in research quality and output, student training, and research infrastructure.


Asunto(s)
Investigación Biomédica , Humanos , Mentores , Rhode Island , Facultades de Medicina , Estudiantes
11.
Mol Cell Biol ; 41(8): e0023421, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34096775

RESUMEN

Fanconi anemia (FA) is a rare genetic disease characterized by increased risk for bone marrow failure and cancer. The FA proteins function together to repair damaged DNA. A central step in the activation of the FA pathway is the monoubiquitination of the FANCD2 and FANCI proteins, which occurs upon exposure to DNA-damaging agents and during the S phase of the cell cycle. The regulatory mechanisms governing S-phase monoubiquitination, in particular, are poorly understood. In this study, we have identified a cyclin-dependent kinase (CDK) regulatory phosphosite (S592) proximal to the site of FANCD2 monoubiquitination. FANCD2 S592 phosphorylation was detected by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and by immunoblotting with an S592 phospho-specific antibody. Mutation of S592 leads to abrogated monoubiquitination of FANCD2 during the S phase. Furthermore, FA-D2 (FANCD2-/-) patient cells expressing S592 mutants display reduced proliferation under conditions of replication stress and increased mitotic aberrations, including micronuclei and multinucleated cells. Our findings describe a novel cell cycle-specific regulatory mechanism for the FANCD2 protein that promotes mitotic fidelity.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/genética , Fosforilación/fisiología , Ciclo Celular/fisiología , Quinasas Ciclina-Dependientes/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 del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Humanos , Espectrometría de Masas en Tándem/métodos , Ubiquitinación/fisiología
12.
J Biol Chem ; 284(42): 28935-42, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19704162

RESUMEN

Fanconi Anemia (FA) is a rare recessive disease characterized by congenital abnormalities, bone marrow failure, and cancer susceptibility. The FA proteins and the familial breast cancer susceptibility gene products, BRCA1 and FANCD1/BRCA2, function cooperatively in the FA-BRCA pathway to repair damaged DNA and to prevent cellular transformation. Activation of this pathway occurs via the mono-ubiquitination of the FANCD2 protein, targeting it to nuclear foci where it co-localizes with FANCD1/BRCA2, RAD51, and PCNA. The regulation of the mono-ubiquitination of FANCD2, as well as its function in DNA repair remain poorly understood. In this study, we have further characterized the interaction between the FANCD2 and PCNA proteins. We have identified a highly conserved, putative FANCD2 PCNA interaction motif (PIP-box), and demonstrate that mutation of this motif disrupts FANCD2-PCNA binding and precludes the mono-ubiquitination of FANCD2. Consequently, the FANCD2 PIP-box mutant protein fails to correct the mitomycin C hypersensitivity of FA-D2 patient cells. Our results suggest that PCNA may function as a molecular platform to facilitate the mono-ubiquitination of FANCD2 and activation of the FA-BRCA pathway.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteína BRCA2/metabolismo , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Daño del ADN , Reparación del ADN , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Humanos , Mitomicina/química , Mutagénesis Sitio-Dirigida , Mutación , Ubiquitina/química
13.
Cancers (Basel) ; 12(5)2020 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-32349331

RESUMEN

Triple negative breast cancer is a collection of heterogeneous breast cancers that are immunohistochemically negative for estrogen receptor, progesterone receptor, and ErbB2 (due to deletion or lack of amplification). No dominant proliferative driver has been identified for this type of cancer, and effective targeted therapy is lacking. In this study, we hypothesized that triple negative breast cancer cells are multi-driver cancer cells, and evaluated a biphasic mathematical model for identifying potent and synergistic drug combinations for multi-driver cancer cells. The responses of two triple negative breast cancer cell lines, MDA-MB-231 and MDA-MB-468, to a panel of targeted therapy drugs were determined over a broad range of concentrations. The analyses of the drug responses by the biphasic mathematical model revealed that both cell lines were indeed dependent on multiple drivers, and inhibitors of individual drivers caused a biphasic response: a target-specific partial inhibition at low nM concentrations, and an off-target toxicity at µM concentrations. We further demonstrated that combinations of drugs, targeting each driver, cause potent, synergistic, and cell-specific cell killing. Immunoblotting analysis of the effects of the individual drugs and drug combinations on the signaling pathways supports the above conclusion. These results support a multi-driver proliferation hypothesis for these triple negative breast cancer cells, and demonstrate the applicability of the biphasic mathematical model for identifying effective and synergistic targeted drug combinations for triple negative breast cancer cells.

14.
Dev Cell ; 5(6): 903-14, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14667412

RESUMEN

Mechanisms underlying the multiple developmental defects observed in Fanconi anemia (FA) patients are not well defined. We have identified the zebrafish homolog of human FANCD2, which encodes a nuclear effector protein that is monoubiquitinated in response to DNA damage, targeting it to nuclear foci where it preserves chromosomal integrity. Fancd2-deficient zebrafish embryos develop defects similar to those found in children with FA, including shortened body length, microcephaly, and microophthalmia, which are due to extensive cellular apoptosis. Developmental defects and increased apoptosis in Fancd2-deficient zebrafish were corrected by injection of human FANCD2 or zebrafish bcl2 mRNA, or by knockdown of p53, indicating that in the absence of Fancd2, developing tissues spontaneously undergo p53-dependent apoptosis. Thus, Fancd2 is essential during embryogenesis to prevent inappropriate apoptosis in neural cells and other tissues undergoing high levels of proliferative expansion, implicating this mechanism in the congenital abnormalities observed in human infants with FA.


Asunto(s)
Anomalías Múltiples/genética , Apoptosis/fisiología , Anemia de Fanconi/genética , Proteínas Nucleares/genética , Proteína p53 Supresora de Tumor/genética , Secuencia de Aminoácidos , Animales , Reactivos de Enlaces Cruzados/farmacología , Compuestos Epoxi/farmacología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi , Regulación del Desarrollo de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/farmacología , Regulación hacia Arriba/genética , Pez Cebra
15.
Mutat Res ; 668(1-2): 27-41, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19101576

RESUMEN

It has long been hypothesized that a defect in the repair of damaged DNA is central to the etiology of Fanconi anemia (FA). Indeed, an increased sensitivity of FA patient-derived cells to the lethal effects of various forms of DNA damaging agents was described over three decades ago [A.J. Fornace, Jr., J.B. Little, R.R. Weichselbaum, DNA repair in a Fanconi's anemia fibroblast cell strain, Biochim. Biophys. Acta 561 (1979) 99-109; Y. Fujiwara, M. Tatsumi, Repair of mitomycin C damage to DNA in mammalian cells and its impairment in Fanconi's anemia cells, Biochem. Biophys. Res. Commun. 66 (1975) 592-598; A.J. Rainbow, M. Howes, Defective repair of ultraviolet- and gamma-ray-damaged DNA in Fanconi's anaemia, Int. J. Radiat. Biol. Relat. Stud. Phys. Chem. Med. 31 (1977) 191-195]. Furthermore, the cytological hallmark of FA, the DNA crosslink-induced radial chromosome formation, exemplifies an innate impairment in the repair of these particularly cytotoxic DNA lesions [A.D. Auerbach, Fanconi anemia diagnosis and the diepoxybutane (DEB) test, Exp. Hematol. 21 (1993) 731-733]. Precisely defining the collective role of the FA proteins in DNA repair, however, continues to be one of the most enigmatic and challenging questions in the FA field. The first six identified FA proteins (A, C, E, F, G, and D2) harbored no recognizable enzymatic features, precluding association with a specific metabolic process. Consequently, our knowledge of the role of the FA proteins in the DNA damage response has been gleaned primarily through biochemical association studies with non-FA proteins. Here, we provide a chronological discourse of the major FA protein interaction network discoveries, with particular emphasis on the DNA damage response, that have defined our current understanding of the molecular basis of FA.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Anemia de Fanconi/genética , Inestabilidad Cromosómica , ADN/metabolismo , Reparación del ADN , Epistasis Genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Humanos , Modelos Biológicos , Recombinación Genética
16.
Mol Cell Biol ; 39(15)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31085681

RESUMEN

Fanconi anemia (FA) is an inherited disease characterized by bone marrow failure and increased cancer risk. FA is caused by mutation of any 1 of 22 genes, and the FA proteins function cooperatively to repair DNA interstrand cross-links (ICLs). A central step in the activation of the FA pathway is the monoubiquitination of the FANCD2 and FANCI proteins, which occurs within chromatin. How FANCD2 and FANCI are anchored to chromatin remains unknown. In this study, we identify and characterize a FANCD2 histone-binding domain (HBD) and embedded methyl-lysine-binding domain (MBD) and demonstrate binding specificity for H4K20me2. Disruption of the HBD/MBD compromises FANCD2 chromatin binding and nuclear focus formation and its ability to promote error-free DNA interstrand cross-link repair, leading to increased error-prone repair and genome instability. Our study functionally describes the first FA protein chromatin reader domain and establishes an important link between this human genetic disease and chromatin plasticity.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/química , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/genética , Histonas/metabolismo , Sitios de Unión , Línea Celular , Cromatina/metabolismo , Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Inestabilidad Genómica , Células HeLa , Histonas/química , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica
17.
Mol Cancer Res ; 16(9): 1335-1345, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29858375

RESUMEN

Chromatin is a highly compact structure that must be rapidly rearranged in order for DNA repair proteins to access sites of damage and facilitate timely and efficient repair. Chromatin plasticity is achieved through multiple processes, including the posttranslational modification of histone tails. In recent years, the impact of histone posttranslational modification on the DNA damage response has become increasingly well recognized, and chromatin plasticity has been firmly linked to efficient DNA repair. One particularly important histone posttranslational modification process is methylation. Here, we focus on the regulation and function of H4K20 methylation (H4K20me) in the DNA damage response and describe the writers, erasers, and readers of this important chromatin mark as well as the combinatorial histone posttranslational modifications that modulate H4K20me recognition. Finally, we discuss the central role of H4K20me in determining if DNA double-strand breaks (DSB) are repaired by the error-prone, nonhomologous DNA end joining pathway or the error-free, homologous recombination pathway. This review article discusses the regulation and function of H4K20me2 in DNA DSB repair and outlines the components and modifications that modulate this important chromatin mark and its fundamental impact on DSB repair pathway choice. Mol Cancer Res; 16(9); 1335-45. ©2018 AACR.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Reparación del ADN , Histonas/genética , Histonas/metabolismo , Humanos , Fosforilación
18.
DNA Repair (Amst) ; 5(2): 251-7, 2006 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-16309973

RESUMEN

Nijmegen breakage syndrome, caused by mutations in the NBS1 gene, is an autosomal recessive chromosomal instability disorder characterized by cancer predisposition. Cells isolated from Nijmegen breakage syndrome patients display increased levels of spontaneous chromosome aberrations and sensitivity to ionizing radiation. Here, we have investigated DNA double strand break repair pathways of homologous recombination, including single strand annealing, and non-homologous end-joining in Nijmegen breakage syndrome patient cells. We used recently developed GFP-YFP-based plasmid substrates to measure the efficiency of DNA double strand break repair. Both single strand annealing and non-homologous end-joining processes were markedly impaired in NBS1-deficient cells, and repair proficiency was restored upon re-introduction of full length NBS1 cDNA. Despite the observed defects in the repair efficiency, no apparent differences in homologous recombination or non-homologous end-joining effector proteins RAD51, KU70, KU86, or DNA-PK(CS) were observed. Furthermore, comparative analysis of junction sequences of plasmids recovered from NBS1-deficient and NBS1-complemented cells revealed increased dependence on microhomology-mediated end-joining DNA repair process in NBS1-complemented cells.


Asunto(s)
Daño del ADN , Reparación del ADN , Síndrome de Nijmegen/genética , Antígenos Nucleares/biosíntesis , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Proteínas de Ciclo Celular/fisiología , Línea Celular , Línea Celular Transformada , Células Cultivadas , Rotura Cromosómica , ADN Complementario/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta en la Radiación , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Autoantígeno Ku , Proteínas Luminiscentes/metabolismo , Modelos Estadísticos , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Plásmidos/metabolismo , Recombinasa Rad51/metabolismo , Recombinación Genética
19.
Blood Rev ; 31(3): 93-99, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27760710

RESUMEN

Fanconi anemia (FA) is a rare autosomal and X-linked genetic disease characterized by congenital abnormalities, progressive bone marrow failure (BMF), and increased cancer risk during early adulthood. The median lifespan for FA patients is approximately 33years. The proteins encoded by the FA genes function together in the FA-BRCA pathway to repair DNA damage and to maintain genome stability. Within the past two years, five new FA genes have been identified-RAD51/FANCR, BRCA1/FANCS, UBE2T/FANCT, XRCC2/FANCU, and REV7/FANCV-bringing the total number of disease-causing genes to 21. This review summarizes the discovery of these new FA genes and describes how these proteins integrate into the FA-BRCA pathway to maintain genome stability and critically prevent early-onset BMF and cancer.


Asunto(s)
Médula Ósea/metabolismo , Médula Ósea/patología , Anemia de Fanconi/etiología , Anemia de Fanconi/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Daño del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Anemia de Fanconi/patología , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Inestabilidad Genómica , Recombinación Homóloga , Humanos , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Mutación , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
20.
Oncotarget ; 8(44): 76443-76457, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-29100324

RESUMEN

Fanconi anemia (FA) is a rare disease characterized by congenital defects, bone marrow failure, and atypically early-onset cancers. The FA proteins function cooperatively to repair DNA interstrand crosslinks. A major step in the activation of the pathway is the monoubiquitination of the FANCD2 and FANCI proteins, and their recruitment to chromatin-associated nuclear foci. The regulation and function of FANCD2 and FANCI, however, is poorly understood. In addition, how chromatin state impacts pathway activation is also unknown. In this study, we have examined the influence of chromatin state on the activation of the FA pathway. We describe potent activation of FANCD2 and FANCI monoubiquitination and nuclear foci formation following treatment of cells with the histone methyltransferase inhibitor BRD4770. BRD4770-induced activation of the pathway does not occur via the direct induction of DNA damage or via the inhibition of the G9a histone methyltransferase, a mechanism previously proposed for this molecule. Instead, we show that BRD4770-inducible FANCD2 and FANCI monoubiquitination and nuclear foci formation may be a consequence of inhibition of the PRC2/EZH2 chromatin-modifying complex. In addition, we show that inhibition of the class I and II histone deacetylases leads to attenuated FANCD2 and FANCI monoubiquitination and nuclear foci formation. Our studies establish that chromatin state is a major determinant of the activation of the FA pathway and suggest an important role for the PRC2/EZH2 complex in the regulation of this critical tumor suppressor pathway.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA