Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Cell ; 60(3): 351-61, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26593718

RESUMEN

DNA replication stress can cause chromosomal instability and tumor progression. One key pathway that counteracts replication stress and promotes faithful DNA replication consists of the Fanconi anemia (FA) proteins. However, how these proteins limit replication stress remains largely elusive. Here we show that conflicts between replication and transcription activate the FA pathway. Inhibition of transcription or enzymatic degradation of transcription-associated R-loops (DNA:RNA hybrids) suppresses replication fork arrest and DNA damage occurring in the absence of a functional FA pathway. Furthermore, we show that simple aldehydes, known to cause leukemia in FA-deficient mice, induce DNA:RNA hybrids in FA-depleted cells. Finally, we demonstrate that the molecular mechanism by which the FA pathway limits R-loop accumulation requires FANCM translocase activity. Failure to activate a response to physiologically occurring DNA:RNA hybrids may critically contribute to the heightened cancer predisposition and bone marrow failure of individuals with mutated FA proteins.


Asunto(s)
Daño del ADN , ADN Helicasas/metabolismo , Replicación del ADN , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Inestabilidad Genómica , Ácidos Nucleicos Heterodúplex/metabolismo , Animales , ADN Helicasas/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Células HeLa , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Ratones , Ratones Noqueados , Mutación , Ácidos Nucleicos Heterodúplex/genética
2.
Nucleic Acids Res ; 47(21): 11268-11283, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31586398

RESUMEN

Accurate DNA replication is critical for the maintenance of genome integrity and cellular survival. Cancer-associated alterations often involve key players of DNA replication and of the DNA damage-signalling cascade. Post-translational modifications play a fundamental role in coordinating replication and repair and central among them is ubiquitylation. We show that the E3 ligase UBR5 interacts with components of the replication fork, including the translesion synthesis (TLS) polymerase polη. Depletion of UBR5 leads to replication problems, such as slower S-phase progression, resulting in the accumulation of single stranded DNA. The effect of UBR5 knockdown is related to a mis-regulation in the pathway that controls the ubiquitylation of histone H2A (UbiH2A) and blocking this modification is sufficient to rescue the cells from replication problems. We show that the presence of polη is the main cause of replication defects and cell death when UBR5 is silenced. Finally, we unveil a novel interaction between polη and H2A suggesting that UbiH2A could be involved in polη recruitment to the chromatin and the regulation of TLS.


Asunto(s)
Daño del ADN , Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células Cultivadas , Daño del ADN/genética , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Histonas/metabolismo , Humanos , Unión Proteica , Procesamiento Proteico-Postraduccional , Fase S/genética , Ubiquitinación/fisiología
3.
PLoS Genet ; 14(10): e1007643, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30335751

RESUMEN

The Fanconi Anemia (FA) pathway is important for repairing interstrand crosslinks (ICLs) between the Watson-Crick strands of the DNA double helix. An initial and essential stage in the repair process is the detection of the ICL. Here, we report the identification of UHRF2, a paralogue of UHRF1, as an ICL sensor protein. UHRF2 is recruited to ICLs in the genome within seconds of their appearance. We show that UHRF2 cooperates with UHRF1, to ensure recruitment of FANCD2 to ICLs. A direct protein-protein interaction is formed between UHRF1 and UHRF2, and between either UHRF1 and UHRF2, and FANCD2. Importantly, we demonstrate that the essential monoubiquitination of FANCD2 is stimulated by UHRF1/UHRF2. The stimulation is mediating by a retention of FANCD2 on chromatin, allowing for its monoubiquitination by the FA core complex. Taken together, we uncover a mechanism of ICL sensing by UHRF2, leading to FANCD2 recruitment and retention at ICLs, in turn facilitating activation of FANCD2 by monoubiquitination.


Asunto(s)
Reparación del ADN/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Secuencia de Aminoácidos , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Línea Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , ADN/metabolismo , Daño del ADN/fisiología , Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Células HEK293 , Células HeLa , Humanos , Dominios y Motivos de Interacción de Proteínas , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
4.
Nucleic Acids Res ; 45(16): 9441-9454, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28934491

RESUMEN

DNA translesion synthesis (TLS) is a crucial damage tolerance pathway that oversees the completion of DNA replication in the presence of DNA damage. TLS polymerases are capable of bypassing a distorted template but they are generally considered inaccurate and they need to be tightly regulated. We have previously shown that polη is phosphorylated on Serine 601 after DNA damage and we have demonstrated that this modification is important for efficient damage bypass. Here we report that polη is also phosphorylated by CDK2, in the absence of damage, in a cell cycle-dependent manner and we identify serine 687 as an important residue targeted by the kinase. We discover that phosphorylation on serine 687 regulates the stability of the polymerase during the cell cycle, allowing it to accumulate in late S and G2 when productive TLS is critical for cell survival. Furthermore, we show that alongside the phosphorylation of S601, the phosphorylation of S687 and S510, S512 and/or S514 are important for damage bypass and cell survival after UV irradiation. Taken together our results provide new insights into how cells can, at different times, modulate DNA TLS for improved cell survival.


Asunto(s)
Ciclo Celular/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , Ciclo Celular/efectos de la radiación , Línea Celular , Supervivencia Celular , Quinasa 2 Dependiente de la Ciclina/metabolismo , Daño del ADN/efectos de la radiación , Reparación del ADN , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , Humanos , Fosforilación , Estabilidad Proteica , Serina/metabolismo , Rayos Ultravioleta
5.
Cell Mol Life Sci ; 73(16): 3097-114, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27094386

RESUMEN

Interstrand crosslinks (ICLs) are a highly toxic form of DNA damage. ICLs can interfere with vital biological processes requiring separation of the two DNA strands, such as replication and transcription. If ICLs are left unrepaired, it can lead to mutations, chromosome breakage and mitotic catastrophe. The Fanconi anemia (FA) pathway can repair this type of DNA lesion, ensuring genomic stability. In this review, we will provide an overview of the cellular response to ICLs. First, we will discuss the origin of ICLs, comparing various endogenous and exogenous sources. Second, we will describe FA proteins as well as FA-related proteins involved in ICL repair, and the post-translational modifications that regulate these proteins. Finally, we will review the process of how ICLs are repaired by both replication-dependent and replication-independent mechanisms.


Asunto(s)
Reactivos de Enlaces Cruzados/efectos adversos , Daño del ADN/efectos de los fármacos , Reparación del ADN , ADN/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Transducción de Señal , Animales , ADN/química , ADN/metabolismo , Aductos de ADN/química , Aductos de ADN/genética , Aductos de ADN/metabolismo , Replicación del ADN , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Inestabilidad Genómica , Humanos , Sustancias Intercalantes/efectos adversos , Modelos Moleculares , Procesamiento Proteico-Postraduccional
6.
Mol Cell ; 32(3): 306-12, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18995829

RESUMEN

In response to DNA damage, eukaryotic cells must rapidly load DNA repair proteins onto damaged chromatin. Chromatin recruitment often entails ubiquitination of a damage-specific DNA repair protein, interaction with a ubiquitin binding factor, assembly of a multisubunit DNA repair complex, and eventually a deubiquitination event once the DNA repair reaction has been completed. This review focuses on the recent discoveries in the Fanconi Anemia (FA) and DNA double-strand break (DSB) repair pathways, which underscore the importance of regulated chromatin loading in the DNA damage response. Interestingly, these two pathways share several features, suggesting a more general mechanism for DNA-repair regulation.


Asunto(s)
Cromatina/genética , Daño del ADN , Reparación del ADN , Anemia de Fanconi/genética , Ensamble y Desensamble de Cromatina , Roturas del ADN de Doble Cadena , Anemia de Fanconi/metabolismo , Homeostasis , Humanos , Ubiquitina/metabolismo , Enzimas Activadoras de Ubiquitina/genética , Enzimas Activadoras de Ubiquitina/metabolismo , Ubiquitinación
7.
J Biol Chem ; 287(35): 29429-41, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22778262

RESUMEN

Notch signaling is critical for development and adult tissue physiology, controlling cell fate in a context-dependent manner. Upon ligand binding, the transmembrane Notch receptor undergoes two ordered proteolytic cleavages releasing Notch intracellular domain, which regulates the transcription of Notch target genes. The strength of Notch signaling is of crucial importance and depends notably on the quantity of Notch receptor at the cell surface. Using an shRNA library screen monitoring Notch trafficking and degradation in the absence of ligand, we identified mammalian USP12 and its Drosophila melanogaster homolog as novel negative regulators of Notch signaling. USP12 silencing specifically interrupts Notch trafficking to the lysosomes and, as a consequence, leads to an increased amount of receptor at the cell surface and to a higher Notch activity. At the biochemical level, USP12 with its activator UAF1 deubiquitinate the nonactivated form of Notch in cell culture and in vitro. These results characterize a new level of conserved regulation of Notch signaling by the ubiquitin system.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Proteolisis , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Proteínas Nucleares/genética , Transporte de Proteínas/fisiología , Receptores Notch/genética , Ubiquitina Tiolesterasa/genética
8.
Nat Cell Biol ; 8(4): 339-47, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16531995

RESUMEN

Monoubiquitination is a reversible post-translational protein modification that has an important regulatory function in many biological processes, including DNA repair. Deubiquitinating enzymes (DUBs) are proteases that are negative regulators of monoubiquitination, but little is known about their regulation and contribution to the control of conjugated-substrate levels. Here, we show that the DUB ubiquitin specific protease 1 (USP1) deubiquitinates the DNA replication processivity factor, PCNA, as a safeguard against error-prone translesion synthesis (TLS) of DNA. Ultraviolet (UV) irradiation inactivates USP1 through an autocleavage event, thus enabling monoubiquitinated PCNA to accumulate and to activate TLS. Significantly, the site of USP1 cleavage is immediately after a conserved internal ubiquitin-like diglycine (Gly-Gly) motif. This mechanism is reminiscent of the processing of precursors of ubiquitin and ubiquitin-like modifiers by DUBs. Our results define a regulatory mechanism for protein ubiquitination that involves the signal-induced degradation of an inhibitory DUB.


Asunto(s)
Daño del ADN/efectos de la radiación , Endopeptidasas/metabolismo , Regulación de la Expresión Génica , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Proteínas de Arabidopsis , Replicación del ADN , Endopeptidasas/química , Endopeptidasas/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/fisiología , Humanos , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Homología de Secuencia de Aminoácido , Proteasas Ubiquitina-Específicas , Rayos Ultravioleta
9.
Cell Rep ; 42(7): 112721, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37392383

RESUMEN

The Fanconi anemia (FA) pathway repairs DNA interstrand crosslinks (ICLs) in humans. Activation of the pathway relies on loading of the FANCD2/FANCI complex onto chromosomes, where it is fully activated by subsequent monoubiquitination. However, the mechanism for loading the complex onto chromosomes remains unclear. Here, we identify 10 SQ/TQ phosphorylation sites on FANCD2, which are phosphorylated by ATR in response to ICLs. Using a range of biochemical assays complemented with live-cell imaging including super-resolution single-molecule tracking, we show that these phosphorylation events are critical for loading of the complex onto chromosomes and for its subsequent monoubiquitination. We uncover how the phosphorylation events are tightly regulated in cells and that mimicking their constant phosphorylation leads to an uncontrolled active state of FANCD2, which is loaded onto chromosomes in an unrestrained fashion. Taken together, we describe a mechanism where ATR triggers FANCD2/FANCI loading onto chromosomes.


Asunto(s)
Cromatina , Anemia de Fanconi , Humanos , Fosforilación , Anemia de Fanconi/genética , 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 , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Daño del ADN , Ubiquitinación , Reparación del ADN , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo
10.
J Biol Chem ; 285(14): 10362-9, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20147293

RESUMEN

The level of monoubiquitinated proliferating cell nuclear antigen (PCNA) is closely linked with DNA damage bypass to protect cells from a high level of mutagenesis. However, it remains unclear how the level of monoubiquitinated PCNA is regulated. Here, we demonstrate that human ELG1 protein, which comprises an alternative replication factor C (RFC) complex and plays an important role in preserving genomic stability, as an interacting partner for the USP1 (ubiquitin-specific protease 1)-UAF1 (USP1-associated factor 1) complex, a deubiquitinating enzyme complex for PCNA and FANCD2. ELG1 protein interacts with PCNAs that are localized at stalled replication forks. ELG1 knockdown specifically resulted in an increase in the level of PCNA monoubiquitination without affecting the level of FANCD2 ubiquitination. It is a novel function of ELG1 distinct from its role as an alternative RFC complex because knockdowns of any other RFC subunits or other alternative RFCs did not affect PCNA monoubiquitination. Lastly, we identified a highly conserved N-terminal domain in ELG1 that was responsible for the USP1-UAF1 interaction as well as the activity to down-regulate PCNA monoubiquitination. Taken together, ELG1 specifically directs USP1-UAF1 complex for PCNA deubiquitination.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endopeptidasas/metabolismo , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Procesamiento Proteico-Postraduccional , Ubiquitina/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas , Proteínas de Arabidopsis , Células Cultivadas , Cromatina/fisiología , Daño del ADN , Replicación del ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Endopeptidasas/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Riñón/citología , Riñón/metabolismo , Mutación/genética , Proteínas Nucleares/genética , Antígeno Nuclear de Célula en Proliferación/genética , ARN Interferente Pequeño/farmacología , Recombinación Genética , Transformación Bacteriana , Proteasas Ubiquitina-Específicas , Ubiquitinación
11.
J Biol Chem ; 285(15): 11252-7, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20147737

RESUMEN

The UAF1 (Usp1-associated factor 1) protein binds and stimulates three deubiquitinating enzymes: USP1, USP12, and USP46. Although the USP1 x UAF1 complex is required for regulation of the Fanconi anemia (FA) DNA repair pathway, less is known about the USP12 x UAF1 and the USP46 x UAF1 complexes. To understand further the nature of the USP12 and USP46 complexes, we attempted to identify proteins that interact with the USP12 and USP46 deubiquitinating enzyme complexes. We identified WDR20, a WD40-repeat containing protein, as a common binding partner of UAF1, USP12, and USP46. Further analysis showed that WDR20 associates exclusively with USP12 and USP46, not with USP1. Furthermore, we demonstrate the purification of a ternary USP12 x UAF1 x WDR20 complex. Interestingly, and consistent with the binding assays, WDR20 stimulated the enzymatic activity of USP12 x UAF1, but not of USP1 x UAF1. Consistent with our previous report that USP12 and USP46 do not regulate the FA pathway, small interference RNA-mediated depletion of WDR20 protein did not affect the FA pathway or DNA damage responses. We provide a model in which WDR20 serves as a stimulatory subunit for preserving and regulating the activity of the subset of the UAF1 x USP complexes.


Asunto(s)
Proteínas Portadoras/fisiología , Regulación Enzimológica de la Expresión Génica , Complejos Multienzimáticos/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina/química , Proteínas Portadoras/metabolismo , Línea Celular , Daño del ADN , Anemia de Fanconi/metabolismo , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Unión Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo
12.
STAR Protoc ; 2(1): 100348, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33665628

RESUMEN

Cells possess multiple DNA repair pathways to tackle a variety of DNA lesions. Often, DNA repair proteins function as large protein complexes. Here, we describe a protocol to purify DNA repair protein complexes from nuclei of mammalian cells. The method permits purification of protein complexes containing stable as well as transiently associated proteins, which subsequently can be identified by mass-spectrometry analysis. This protocol can be applied to uncover the functions and mechanism of DNA repair pathways. For complete information on the use and execution of this protocol, please refer to Socha et al. (2020).


Asunto(s)
Núcleo Celular/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Células HeLa , Humanos
13.
Cell Rep ; 32(1): 107850, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32640220

RESUMEN

The Fanconi anemia (FA) pathway repairs DNA interstrand crosslinks (ICLs). Many FA proteins are recruited to ICLs in a timely fashion so that coordinated repair can occur. However, the mechanism of this process is poorly understood. Here, we report the purification of a FANCD2-containing protein complex with multiple subunits, including WRNIP1. Using live-cell imaging, we show that WRNIP1 is recruited to ICLs quickly after their appearance, promoting repair. The observed recruitment facilitates subsequent recruitment of the FANCD2/FANCI complex. Depletion of WRNIP1 sensitizes cells to ICL-forming drugs. We find that ubiquitination of WRNIP1 and the activity of its UBZ domain are required to facilitate recruitment of FANCD2/FANCI and promote repair. Altogether, we describe a mechanism by which WRNIP1 is recruited rapidly to ICLs, resulting in chromatin loading of the FANCD2/FANCI complex in an unusual process entailing ubiquitination of WRNIP1 and the activity of its integral UBZ domain.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Reactivos de Enlaces Cruzados/química , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/química , Secuencia de Aminoácidos , Supervivencia Celular , Cromatina/metabolismo , Proteínas de Unión al ADN/química , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Dominios Proteicos , Subunidades de Proteína/metabolismo , Ubiquitinación
14.
Cell Rep ; 27(10): 2990-3005.e5, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167143

RESUMEN

Interstrand crosslinks (ICLs) of the DNA helix are a deleterious form of DNA damage. ICLs can be repaired by the Fanconi anemia pathway. At the center of the pathway is the FANCD2/FANCI complex, recruitment of which to DNA is a critical step for repair. After recruitment, monoubiquitination of both FANCD2 and FANCI leads to their retention on chromatin, ensuring subsequent repair. However, regulation of recruitment is poorly understood. Here, we report a cluster of phosphosites on FANCD2 whose phosphorylation by CK2 inhibits both FANCD2 recruitment to ICLs and its monoubiquitination in vitro and in vivo. We have found that phosphorylated FANCD2 possesses reduced DNA binding activity, explaining the previous observations. Thus, we describe a regulatory mechanism operating as a molecular switch, where in the absence of DNA damage, the FANCD2/FANCI complex is prevented from loading onto DNA, effectively suppressing the FA pathway.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Secuencia de Aminoácidos , Animales , Sistemas CRISPR-Cas/genética , Quinasa de la Caseína II/metabolismo , ADN/metabolismo , Daño del ADN , Reparación del ADN , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/química , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Células HeLa , Humanos , Fosforilación , Unión Proteica , Estructura Cuaternaria de Proteína , ARN Guía de Kinetoplastida/metabolismo , Alineación de Secuencia , Ubiquitinación
15.
Nat Commun ; 7: 12124, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27405460

RESUMEN

The Fanconi anaemia (FA) pathway is important for the repair of DNA interstrand crosslinks (ICL). The FANCD2-FANCI complex is central to the pathway, and localizes to ICLs dependent on its monoubiquitination. It has remained elusive whether the complex is recruited before or after the critical monoubiquitination. Here, we report the first structural insight into the human FANCD2-FANCI complex by obtaining the cryo-EM structure. The complex contains an inner cavity, large enough to accommodate a double-stranded DNA helix, as well as a protruding Tower domain. Disease-causing mutations in the Tower domain are observed in several FA patients. Our work reveals that recruitment of the complex to a stalled replication fork serves as the trigger for the activating monoubiquitination event. Taken together, our results uncover the mechanism of how the FANCD2-FANCI complex activates the FA pathway, and explains the underlying molecular defect in FA patients with mutations in the Tower domain.


Asunto(s)
Reparación del ADN , ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/ultraestructura , Proteínas del Grupo de Complementación de la Anemia de Fanconi/ultraestructura , Anemia de Fanconi/genética , Ubiquitinación , Microscopía por Crioelectrón , Ensayo de Cambio de Movilidad Electroforética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , 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 , Humanos , Dominios Proteicos/genética
16.
Nat Struct Mol Biol ; 22(3): 242-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25643322

RESUMEN

DNA interstrand cross-links (ICLs) prevent strand separation during DNA replication and transcription and therefore are extremely cytotoxic. In metazoans, a major pathway of ICL repair is coupled to DNA replication, and it requires the Fanconi anemia pathway. In most current models, collision of a single DNA replication fork with an ICL is sufficient to initiate repair. In contrast, we show here that in Xenopus egg extracts two DNA replication forks must converge on an ICL to trigger repair. When only one fork reaches the ICL, the replicative CMG helicase fails to unload from the stalled fork, and repair is blocked. Arrival of a second fork, even when substantially delayed, rescues repair. We conclude that ICL repair requires a replication-induced X-shaped DNA structure surrounding the lesion, and we speculate on how this requirement helps maintain genomic stability in S phase.


Asunto(s)
Reparación del ADN/fisiología , ADN/química , Modelos Genéticos , Modelos Moleculares , Animales , Replicación del ADN , Inestabilidad Genómica , Fase S , Xenopus
17.
Cell Rep ; 10(12): 1947-56, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25801034

RESUMEN

The Fanconi anemia (FA) pathway is critical for the cellular response to toxic DNA interstrand crosslinks (ICLs). Using a biochemical purification strategy, we identified UHRF1 as a protein that specifically interacts with ICLs in vitro and in vivo. Reduction of cellular levels of UHRF1 by RNAi attenuates the FA pathway and sensitizes cells to mitomycin C. Knockdown cells display a drastic reduction in FANCD2 foci formation. Using live-cell imaging, we observe that UHRF1 is rapidly recruited to chromatin in response to DNA crosslinking agents and that this recruitment both precedes and is required for the recruitment of FANCD2 to ICLs. Based on these results, we describe a mechanism of ICL sensing and propose that UHRF1 is a critical factor that binds to ICLs. In turn, this binding is necessary for the subsequent recruitment of FANCD2, which allows the DNA repair process to initiate.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/metabolismo , Secuencia de Aminoácidos , Proteínas Potenciadoras de Unión a CCAAT/genética , Cromatina/genética , Daño del ADN/fisiología , Reparación del ADN/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Humanos , Ubiquitina-Proteína Ligasas
19.
J Biol Chem ; 284(8): 5343-51, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19075014

RESUMEN

A balance between ubiquitination and deubiquitination regulates numerous cellular processes and pathways, and specific deubiquitinating enzymes often play the decisive role of controlling this balance. We recently reported that the USP1 deubiquitinating enzyme, which regulates the Fanconi anemia pathway by deubiquitinating the central player of the pathway, FANCD2, is activated by the WD40-repeat containing UAF1 protein through formation of a stable USP1/UAF1 protein complex. Here we present the isolation of two novel multisubunit deubiquitinating enzyme complexes containing USP12 and USP46, respectively. Both complexes contain the UAF1 protein as a bona fide subunit. Interestingly, UAF1 regulates the enzymatic activity of both enzyme complexes, suggesting that this activator protein may regulate a subclass of human deubiquitinating enzymes. We postulate that additional WD40-containing proteins may also form complexes with other human deubiquitinating enzymes and thereby regulate their activity and substrate specificity.


Asunto(s)
Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación/fisiología , Proteínas de Arabidopsis , Endopeptidasas/genética , Endopeptidasas/aislamiento & purificación , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/aislamiento & purificación , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/aislamiento & purificación , Proteínas Nucleares/genética , Proteínas Nucleares/aislamiento & purificación , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/aislamiento & purificación , Proteasas Ubiquitina-Específicas
20.
Mol Cell ; 28(5): 786-97, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-18082604

RESUMEN

The deubiquitinating enzyme USP1 controls the cellular levels of the DNA damage response protein Ub-FANCD2, a key protein of the Fanconi anemia DNA repair pathway. Here we report the purification of a USP1 multisubunit protein complex from HeLa cells containing stoichiometric amounts of a WD40 repeat-containing protein, USP1 associated factor 1 (UAF1). In vitro reconstitution of USP1 deubiquitinating enzyme activity, using either ubiquitin-7-amido-4-methylcoumarin (Ub-AMC) or purified monoubiquitinated FANCD2 protein as substrates, demonstrates that UAF1 functions as an activator of USP1. UAF1 binding increases the catalytic turnover (kcat) but does not increase the affinity of the USP1 enzyme for the substrate (KM). Moreover, we show that DNA damage results in an immediate shutoff of transcription of the USP1 gene, leading to a rapid decline in the USP1/UAF1 protein complex. Taken together, our results describe a mechanism of regulation of the deubiquitinating enzyme, USP1, and of DNA repair.


Asunto(s)
Daño del ADN , Endopeptidasas/genética , Endopeptidasas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Arabidopsis , Cumarinas/metabolismo , Reparación del ADN , Endopeptidasas/aislamiento & purificación , Células HeLa , Humanos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Mutación , Proteínas Nucleares/aislamiento & purificación , Subunidades de Proteína , Transducción de Señal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteasas Ubiquitina-Específicas , Ubiquitinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA