Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 180(6): 1039-1041, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32200798

RESUMEN

Two papers, by Nakazawa and Vidakovic, show how ubiquitylation of a single lysine residue in RNA polymerase II serves as a master switch to regulate transcription, RNA polymerase II degradation, and transcription-coupled nucleotide excision repair in response to DNA damage.


Asunto(s)
ARN Polimerasa II , Transcripción Genética , Daño del ADN , Reparación del ADN , Ubiquitinación
2.
Mol Cell ; 82(7): 1343-1358.e8, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35271816

RESUMEN

Nucleotide excision repair (NER) counteracts the onset of cancer and aging by removing helix-distorting DNA lesions via a "cut-and-patch"-type reaction. The regulatory mechanisms that drive NER through its successive damage recognition, verification, incision, and gap restoration reaction steps remain elusive. Here, we show that the RAD5-related translocase HLTF facilitates repair through active eviction of incised damaged DNA together with associated repair proteins. Our data show a dual-incision-dependent recruitment of HLTF to the NER incision complex, which is mediated by HLTF's HIRAN domain that binds 3'-OH single-stranded DNA ends. HLTF's translocase motor subsequently promotes the dissociation of the stably damage-bound incision complex together with the incised oligonucleotide, allowing for an efficient PCNA loading and initiation of repair synthesis. Our findings uncover HLTF as an important NER factor that actively evicts DNA damage, thereby providing additional quality control by coordinating the transition between the excision and DNA synthesis steps to safeguard genome integrity.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN , ADN/genética , ADN/metabolismo , Daño del ADN , Replicación del ADN , Proteínas de Unión al ADN/genética
3.
Cell ; 146(6): 931-41, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21925316

RESUMEN

The eukaryotic replicative DNA helicase, CMG, unwinds DNA by an unknown mechanism. In some models, CMG encircles and translocates along one strand of DNA while excluding the other strand. In others, CMG encircles and translocates along duplex DNA. To distinguish between these models, replisomes were confronted with strand-specific DNA roadblocks in Xenopus egg extracts. An ssDNA translocase should stall at an obstruction on the translocation strand but not the excluded strand, whereas a dsDNA translocase should stall at obstructions on either strand. We found that replisomes bypass large roadblocks on the lagging strand template much more readily than on the leading strand template. Our results indicate that CMG is a 3' to 5' ssDNA translocase, consistent with unwinding via "steric exclusion." Given that MCM2-7 encircles dsDNA in G1, the data imply that formation of CMG in S phase involves remodeling of MCM2-7 from a dsDNA to a ssDNA binding mode.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN , ADN/metabolismo , Xenopus/metabolismo , Animales , ADN de Cadena Simple/metabolismo , Modelos Biológicos , Fase S
4.
Proc Natl Acad Sci U S A ; 120(11): e2208860120, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36893274

RESUMEN

XPA is a central scaffold protein that coordinates the assembly of repair complexes in the global genome (GG-NER) and transcription-coupled nucleotide excision repair (TC-NER) subpathways. Inactivating mutations in XPA cause xeroderma pigmentosum (XP), which is characterized by extreme UV sensitivity and a highly elevated skin cancer risk. Here, we describe two Dutch siblings in their late forties carrying a homozygous H244R substitution in the C-terminus of XPA. They present with mild cutaneous manifestations of XP without skin cancer but suffer from marked neurological features, including cerebellar ataxia. We show that the mutant XPA protein has a severely weakened interaction with the transcription factor IIH (TFIIH) complex leading to an impaired association of the mutant XPA and the downstream endonuclease ERCC1-XPF with NER complexes. Despite these defects, the patient-derived fibroblasts and reconstituted knockout cells carrying the XPA-H244R substitution show intermediate UV sensitivity and considerable levels of residual GG-NER (~50%), in line with the intrinsic properties and activities of the purified protein. By contrast, XPA-H244R cells are exquisitely sensitive to transcription-blocking DNA damage, show no detectable recovery of transcription after UV irradiation, and display a severe deficiency in TC-NER-associated unscheduled DNA synthesis. Our characterization of a new case of XPA deficiency that interferes with TFIIH binding and primarily affects the transcription-coupled subpathway of nucleotide excision repair, provides an explanation of the dominant neurological features in these patients, and reveals a specific role for the C-terminus of XPA in TC-NER.


Asunto(s)
Neoplasias Cutáneas , Xerodermia Pigmentosa , Humanos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Alelos , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Proteína de la Xerodermia Pigmentosa del Grupo A/metabolismo , Reparación del ADN/genética , Daño del ADN/genética , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/metabolismo , Neoplasias Cutáneas/genética , Factor de Transcripción TFIIH/genética , Factor de Transcripción TFIIH/metabolismo
5.
Nucleic Acids Res ; 51(2): 631-649, 2023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36594163

RESUMEN

TRAIP is a key factor involved in the DNA damage response (DDR), homologous recombination (HR) and DNA interstrand crosslink (ICL) repair. However, the exact functions of TRAIP in these processes in mammalian cells are not fully understood. Here we identify the zinc finger protein 212, ZNF212, as a novel binding partner for TRAIP and find that ZNF212 colocalizes with sites of DNA damage. The recruitment of TRAIP or ZNF212 to sites of DNA damage is mutually interdependent. We show that depletion of ZNF212 causes defects in the DDR and HR-mediated repair in a manner epistatic to TRAIP. In addition, an epistatic analysis of Zfp212, the mouse homolog of human ZNF212, in mouse embryonic stem cells (mESCs), shows that it appears to act upstream of both the Neil3 and Fanconi anemia (FA) pathways of ICLs repair. We find that human ZNF212 interacted directly with NEIL3 and promotes its recruitment to ICL lesions. Collectively, our findings identify ZNF212 as a new factor involved in the DDR, HR-mediated repair and ICL repair though direct interaction with TRAIP.


Asunto(s)
Reparación del ADN , Anemia de Fanconi , Animales , Ratones , Humanos , Reparación del ADN/genética , Daño del ADN , Replicación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Genómica , Anemia de Fanconi/genética , Mamíferos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas del Tejido Nervioso/genética
6.
Proc Natl Acad Sci U S A ; 119(34): e2207408119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969784

RESUMEN

The xeroderma pigmentosum protein A (XPA) and replication protein A (RPA) proteins fulfill essential roles in the assembly of the preincision complex in the nucleotide excision repair (NER) pathway. We have previously characterized the two interaction sites, one between the XPA N-terminal (XPA-N) disordered domain and the RPA32 C-terminal domain (RPA32C), and the other with the XPA DNA binding domain (DBD) and the RPA70AB DBDs. Here, we show that XPA mutations that inhibit the physical interaction in either site reduce NER activity in biochemical and cellular systems. Combining mutations in the two sites leads to an additive inhibition of NER, implying that they fulfill distinct roles. Our data suggest a model in which the interaction between XPA-N and RPA32C is important for the initial association of XPA with NER complexes, while the interaction between XPA DBD and RPA70AB is needed for structural organization of the complex to license the dual incision reaction. Integrative structural models of complexes of XPA and RPA bound to single-stranded/double-stranded DNA (ss/dsDNA) junction substrates that mimic the NER bubble reveal key features of the architecture of XPA and RPA in the preincision complex. Most critical among these is that the shape of the NER bubble is far from colinear as depicted in current models, but rather the two strands of unwound DNA must assume a U-shape with the two ss/dsDNA junctions localized in close proximity. Our data suggest that the interaction between XPA and RPA70 is key for the organization of the NER preincision complex.


Asunto(s)
Reparación del ADN , Proteína de Replicación A , Proteína de la Xerodermia Pigmentosa del Grupo A , ADN/metabolismo , Daño del ADN , Unión Proteica , Dominios Proteicos , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Proteína de la Xerodermia Pigmentosa del Grupo A/metabolismo
7.
Crit Rev Biochem Mol Biol ; 56(2): 125-136, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33430640

RESUMEN

DNA damaging agents have been a cornerstone of cancer therapy for nearly a century. The discovery of many of these chemicals, particularly the alkylating agents, are deeply entwined with the development of poisonous materials originally intended for use in warfare. Over the last decades, their anti-proliferative effects have focused on the specific mechanisms by which they damage DNA, and the factors involved in the repair of such damage. Due to the variety of aberrant adducts created even for the simplest alkylating agents, numerous pathways of repair are engaged as a defense against this damage. More recent work has underscored the role of RNA damage in the cellular response to these agents, although the understanding of their role in relation to established DNA repair pathways is still in its infancy. In this review, we discuss the chemistry of alkylating agents, the numerous ways in which they damage nucleic acids, as well as the specific DNA and RNA repair pathways which are engaged to counter their effects.


Asunto(s)
Daño del ADN , ADN/genética , ARN/genética , Alquilantes/toxicidad , Alquilación/efectos de los fármacos , Animales , ADN/química , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Humanos , ARN/química
8.
Chem Res Toxicol ; 36(6): 822-837, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37208809

RESUMEN

Cisplatin (CP) is a common antitumor drug that is used to treat many solid tumors. The activity of CP is attributed to the formation of DNA-DNA cross-links, which consist of 1,2-intra-, 1,3-intra-, and interstrand cross-links. To better understand how each intrastrand cross-link contributes to the activity of CP, we have developed comprehensive ultraperformance liquid chromatography-selective ion monitoring (UPLC-SIM) assays to quantify 1,2-GG-, 1,2-AG-, 1,3-GCG-, and 1,3-GTG-intrastrand cross-links. The limit of quantitation for the developed assays ranged from 5 to 50 fmol or as low as 6 cross-links per 108 nucleotides. To demonstrate the utility of the UPLC-SIM assays, we first performed in vitro cross-link formation kinetics experiments. We confirmed that the 1,2-GG-intrastrand cross-links were the most abundant intrastrand cross-link and formed at a faster rate compared to 1,2-AG- and 1,3-intrastrand cross-links. Furthermore, we investigated the repair kinetics of intrastrand cross-links in CP-treated wild-type and nucleotide excision repair (NER)-deficient U2OS cells. We observed a slow decrease of both 1,2- and 1,3-intrastrand cross-links in wild-type cells and no evidence of direct repair in the NER-deficient cells. Taken together, we have demonstrated that our assays are capable of accurately quantifying intrastrand cross-links in CP-treated samples and can be utilized to better understand the activity of CP.


Asunto(s)
Cisplatino , Aductos de ADN , Cisplatino/farmacología , ADN/química , Cromatografía Liquida , Espectrometría de Masas , Reparación del ADN , Reactivos de Enlaces Cruzados/química
9.
Cell ; 134(6): 969-80, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18805090

RESUMEN

DNA interstrand crosslinks (ICLs) are toxic DNA lesions whose repair occurs in the S phase of metazoans via an unknown mechanism. Here, we describe a cell-free system based on Xenopus egg extracts that supports ICL repair. During DNA replication of a plasmid containing a site-specific ICL, two replication forks converge on the crosslink. Subsequent lesion bypass involves advance of a nascent leading strand to within one nucleotide of the ICL, followed by incisions, translesion DNA synthesis, and extension of the nascent strand beyond the lesion. Immunodepletion experiments suggest that extension requires DNA polymerase zeta. Ultimately, a significant portion of the input DNA is fully repaired, but not if DNA replication is blocked. Our experiments establish a mechanism for ICL repair that reveals how this process is coupled to DNA replication.


Asunto(s)
Reparación del ADN , Replicación del ADN , Animales , Sistema Libre de Células , ADN , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , Xenopus
10.
Mol Cell ; 54(3): 472-84, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24726326

RESUMEN

SLX4 binds to three nucleases (XPF-ERCC1, MUS81-EME1, and SLX1), and its deficiency leads to genomic instability, sensitivity to DNA crosslinking agents, and Fanconi anemia. However, it is not understood how SLX4 and its associated nucleases act in DNA crosslink repair. Here, we uncover consequences of mouse Slx4 deficiency and reveal its function in DNA crosslink repair. Slx4-deficient mice develop epithelial cancers and have a contracted hematopoietic stem cell pool. The N-terminal domain of SLX4 (mini-SLX4) that only binds to XPF-ERCC1 is sufficient to confer resistance to DNA crosslinking agents. Recombinant mini-SLX4 enhances XPF-ERCC1 nuclease activity up to 100-fold, directing specificity toward DNA forks. Mini-SLX4-XPF-ERCC1 also vigorously stimulates dual incisions around a DNA crosslink embedded in a synthetic replication fork, an essential step in the repair of this lesion. These observations define vertebrate SLX4 as a tumor suppressor, which activates XPF-ERCC1 nuclease specificity in DNA crosslink repair.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Recombinasas/fisiología , Animales , Secuencia de Bases , Células de la Médula Ósea/patología , Aductos de ADN/química , Daño del ADN , Proteínas de Unión al ADN/química , Endonucleasas/química , Células Madre Hematopoyéticas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/enzimología , Conformación de Ácido Nucleico , Proteínas Supresoras de Tumor
11.
Cell Mol Life Sci ; 78(24): 7925-7942, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34731255

RESUMEN

Global genome nucleotide excision repair (GG-NER) eliminates a broad spectrum of DNA lesions from genomic DNA. Genomic DNA is tightly wrapped around histones creating a barrier for DNA repair proteins to access DNA lesions buried in nucleosomal DNA. The DNA-damage sensors XPC and DDB2 recognize DNA lesions in nucleosomal DNA and initiate repair. The emerging view is that a tight interplay between XPC and DDB2 is regulated by post-translational modifications on the damage sensors themselves as well as on chromatin containing DNA lesions. The choreography between XPC and DDB2, their interconnection with post-translational modifications such as ubiquitylation, SUMOylation, methylation, poly(ADP-ribos)ylation, acetylation, and the functional links with chromatin remodelling activities regulate not only the initial recognition of DNA lesions in nucleosomes, but also the downstream recruitment and necessary displacement of GG-NER factors as repair progresses. In this review, we highlight how nucleotide excision repair leaves a mark on chromatin to enable DNA damage detection in nucleosomes.


Asunto(s)
Cromatina/genética , Daño del ADN , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Nucleosomas/fisiología , Procesamiento Proteico-Postraduccional , Animales , Cromatina/química , Enzimas Reparadoras del ADN/genética , Humanos
12.
Nucleic Acids Res ; 48(15): 8461-8473, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32633759

RESUMEN

DNA polymerase ζ (Pol ζ) and Rev1 are essential for the repair of DNA interstrand crosslink (ICL) damage. We have used yeast DNA polymerases η, ζ and Rev1 to study translesion synthesis (TLS) past a nitrogen mustard-based interstrand crosslink (ICL) with an 8-atom linker between the crosslinked bases. The Rev1-Pol ζ complex was most efficient in complete bypass synthesis, by 2-3 fold, compared to Pol ζ alone or Pol η. Rev1 protein, but not its catalytic activity, was required for efficient TLS. A dCMP residue was faithfully inserted across the ICL-G by Pol η, Pol ζ, and Rev1-Pol ζ. Rev1-Pol ζ, and particularly Pol ζ alone showed a tendency to stall before the ICL, whereas Pol η stalled just after insertion across the ICL. The stalling of Pol η directly past the ICL is attributed to its autoinhibitory activity, caused by elongation of the short ICL-unhooked oligonucleotide (a six-mer in our study) by Pol η providing a barrier to further elongation of the correct primer. No stalling by Rev1-Pol ζ directly past the ICL was observed, suggesting that the proposed function of Pol ζ as an extender DNA polymerase is also required for ICL repair.


Asunto(s)
ADN Polimerasa Dirigida por ADN/genética , ADN/genética , Nucleotidiltransferasas/genética , Proteínas de Saccharomyces cerevisiae/genética , Estructuras Cromosómicas/efectos de los fármacos , Estructuras Cromosómicas/genética , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Replicación del ADN/genética , Complejos Multiproteicos/genética , Compuestos de Mostaza Nitrogenada/farmacología , Saccharomyces cerevisiae/genética
13.
Nucleic Acids Res ; 48(6): 3328-3342, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32002546

RESUMEN

Monoubiquitination of the Fanconi anemia complementation group D2 (FANCD2) protein by the FA core ubiquitin ligase complex is the central event in the FA pathway. FANCA and FANCG play major roles in the nuclear localization of the FA core complex. Mutations of these two genes are the most frequently observed genetic alterations in FA patients, and most point mutations in FANCA are clustered in the C-terminal domain (CTD). To understand the basis of the FA-associated FANCA mutations, we determined the cryo-electron microscopy (EM) structures of Xenopus laevis FANCA alone at 3.35 Å and 3.46 Å resolution and two distinct FANCA-FANCG complexes at 4.59 and 4.84 Å resolution, respectively. The FANCA CTD adopts an arc-shaped solenoid structure that forms a pseudo-symmetric dimer through its outer surface. FA- and cancer-associated point mutations are widely distributed over the CTD. The two different complex structures capture independent interactions of FANCG with either FANCA C-terminal HEAT repeats, or the N-terminal region. We show that mutations that disturb either of these two interactions prevent the nuclear localization of FANCA, thereby leading to an FA pathway defect. The structure provides insights into the function of FANCA CTD, and provides a framework for understanding FA- and cancer-associated mutations.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi/ultraestructura , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/ultraestructura , Proteína del Grupo de Complementación G de la Anemia de Fanconi/ultraestructura , Anemia de Fanconi/genética , Animales , Núcleo Celular/genética , Núcleo Celular/ultraestructura , Microscopía por Crioelectrón , Cristalografía por Rayos X , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/ultraestructura , Anemia de Fanconi/patología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/química , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Mutación , Unión Proteica/genética , Conformación Proteica , Xenopus laevis/genética
14.
Nucleic Acids Res ; 48(4): 2173-2188, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31925419

RESUMEN

The XPA protein functions together with the single-stranded DNA (ssDNA) binding protein RPA as the central scaffold to ensure proper positioning of repair factors in multi-protein nucleotide excision repair (NER) machinery. We previously determined the structure of a short motif in the disordered XPA N-terminus bound to the RPA32C domain. However, a second contact between the XPA DNA-binding domain (XPA DBD) and the RPA70AB tandem ssDNA-binding domains, which is likely to influence the orientation of XPA and RPA on the damaged DNA substrate, remains poorly characterized. NMR was used to map the binding interfaces of XPA DBD and RPA70AB. Combining NMR and X-ray scattering data with comprehensive docking and refinement revealed how XPA DBD and RPA70AB orient on model NER DNA substrates. The structural model enabled design of XPA mutations that inhibit the interaction with RPA70AB. These mutations decreased activity in cell-based NER assays, demonstrating the functional importance of XPA DBD-RPA70AB interaction. Our results inform ongoing controversy about where XPA is bound within the NER bubble, provide structural insights into the molecular basis for malfunction of disease-associated XPA missense mutations, and contribute to understanding of the structure and mechanical action of the NER machinery.


Asunto(s)
Reparación del ADN/genética , Modelos Moleculares , Proteína de Replicación A/química , Proteína de la Xerodermia Pigmentosa del Grupo A/química , ADN/química , ADN/genética , Daño del ADN/genética , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Humanos , Espectroscopía de Resonancia Magnética , Unión Proteica/genética , Proteína de Replicación A/genética , Proteína de la Xerodermia Pigmentosa del Grupo A/genética
15.
EMBO J ; 36(11): 1605-1622, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28373211

RESUMEN

Base excision repair (BER) is one of the most frequently used cellular DNA repair mechanisms and modulates many human pathophysiological conditions related to DNA damage. Through live cell and in vitro reconstitution experiments, we have discovered a major sub-pathway of conventional long-patch BER that involves formation of a 9-nucleotide gap 5' to the lesion. This new sub-pathway is mediated by RECQ1 DNA helicase and ERCC1-XPF endonuclease in cooperation with PARP1 poly(ADP-ribose) polymerase and RPA The novel gap formation step is employed during repair of a variety of DNA lesions, including oxidative and alkylation damage. Moreover, RECQ1 regulates PARP1 auto-(ADP-ribosyl)ation and the choice between long-patch and single-nucleotide BER, thereby modulating cellular sensitivity to DNA damage. Based on these results, we propose a revised model of long-patch BER and a new key regulation point for pathway choice in BER.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , RecQ Helicasas/metabolismo , Proteína de Replicación A/metabolismo , Línea Celular , ADN/metabolismo , Daño del ADN , Humanos , Modelos Biológicos
16.
Chem Res Toxicol ; 34(7): 1790-1799, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34133118

RESUMEN

Nitrogen mustards are a widely used class of antitumor agents that exert their cytotoxic effects through the formation of DNA interstrand cross-links (ICLs). Despite being among the first antitumor agents used, the biological responses to NM ICLs remain only partially understood. We have previously reported the generation of NM ICL mimics by incorporation of ICL precursors into DNA using solid-phase synthesis at defined positions, followed by a double reductive amination reaction. However, the structure of these mimics deviated from the native NM ICLs. Using further development of our approach, we report a new class of NM ICL mimics that only differ from their native counterpart by substitution of dG with 7-deaza-dG at the ICL. Importantly, this approach allows for the synthesis of diverse NM ICLs, illustrated here with a mimic of the adduct formed by chlorambucil. We used the newly generated ICLs in reactions with replicative and translesion synthesis DNA polymerase to demonstrate their stability and utility for functional studies. These new NM ICLs will allow for the further characterization of the biological responses to this important class of antitumor agents.


Asunto(s)
Antineoplásicos Alquilantes/química , ADN/química , Sustancias Intercalantes/química , Mecloretamina/análogos & derivados , Antineoplásicos Alquilantes/síntesis química , ADN/síntesis química , ADN Polimerasa Dirigida por ADN/química , Humanos , Sustancias Intercalantes/síntesis química , Mecloretamina/síntesis química
17.
Nucleic Acids Res ; 47(16): 8337-8347, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31372632

RESUMEN

DNA repair is critical for maintaining genomic integrity. Finding DNA lesions initiates the entire repair process. In human nucleotide excision repair (NER), XPC-RAD23B recognizes DNA lesions and recruits downstream factors. Although previous studies revealed the molecular features of damage identification by the yeast orthologs Rad4-Rad23, the dynamic mechanisms by which human XPC-RAD23B recognizes DNA defects have remained elusive. Here, we directly visualized the motion of XPC-RAD23B on undamaged and lesion-containing DNA using high-throughput single-molecule imaging. We observed three types of one-dimensional motion of XPC-RAD23B along DNA: diffusive, immobile and constrained. We found that consecutive AT-tracks led to increase in proteins with constrained motion. The diffusion coefficient dramatically increased according to ionic strength, suggesting that XPC-RAD23B diffuses along DNA via hopping, allowing XPC-RAD23B to bypass protein obstacles during the search for DNA damage. We also examined how XPC-RAD23B identifies cyclobutane pyrimidine dimers (CPDs) during diffusion. XPC-RAD23B makes futile attempts to bind to CPDs, consistent with low CPD recognition efficiency. Moreover, XPC-RAD23B binds CPDs in biphasic states, stable for lesion recognition and transient for lesion interrogation. Taken together, our results provide new insight into how XPC-RAD23B searches for DNA lesions in billions of base pairs in human genome.


Asunto(s)
Enzimas Reparadoras del ADN/química , Reparación del ADN , ADN Viral/química , Proteínas de Unión al ADN/química , ADN/química , Dímeros de Pirimidina/química , Bacteriófago lambda/química , Bacteriófago lambda/genética , Sitios de Unión , ADN/genética , ADN/metabolismo , Daño del ADN , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Difusión , Humanos , Cinética , Modelos Moleculares , Conformación de Ácido Nucleico , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/metabolismo , Concentración Osmolar , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Dímeros de Pirimidina/metabolismo , Imagen Individual de Molécula
18.
Mol Cell ; 47(1): 3-4, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22793691

RESUMEN

A new pathway for the repair of DNA alkylation damage is described in this issue of Molecular Cell (Latypov et al., 2012). Alkyltransferase-like enzymes mark O(6)-alkylguanine lesions and, depending on adduct size, channel them into global genome or transcription-coupled nucleotide excision repair pathways.

19.
PLoS Genet ; 13(5): e1006813, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28542210

RESUMEN

Double strand breaks (DSBs) and interstrand crosslinks (ICLs) are toxic DNA lesions that can be repaired through multiple pathways, some of which involve shared proteins. One of these proteins, DNA Polymerase θ (Pol θ), coordinates a mutagenic DSB repair pathway named microhomology-mediated end joining (MMEJ) and is also a critical component for bypass or repair of ICLs in several organisms. Pol θ contains both polymerase and helicase-like domains that are tethered by an unstructured central region. While the role of the polymerase domain in promoting MMEJ has been studied extensively both in vitro and in vivo, a function for the helicase-like domain, which possesses DNA-dependent ATPase activity, remains unclear. Here, we utilize genetic and biochemical analyses to examine the roles of the helicase-like and polymerase domains of Drosophila Pol θ. We demonstrate an absolute requirement for both polymerase and ATPase activities during ICL repair in vivo. However, similar to mammalian systems, polymerase activity, but not ATPase activity, is required for ionizing radiation-induced DSB repair. Using a site-specific break repair assay, we show that overall end-joining efficiency is not affected in ATPase-dead mutants, but there is a significant decrease in templated insertion events. In vitro, Pol θ can efficiently bypass a model unhooked nitrogen mustard crosslink and promote DNA synthesis following microhomology annealing, although ATPase activity is not required for these functions. Together, our data illustrate the functional importance of the helicase-like domain of Pol θ and suggest that its tethering to the polymerase domain is important for its multiple functions in DNA repair and damage tolerance.


Asunto(s)
Dominio Catalítico , Reparación del ADN por Unión de Extremidades , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Animales , Enzimas Reparadoras del ADN/química , Enzimas Reparadoras del ADN/genética , ADN Polimerasa Dirigida por ADN , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética
20.
J Biol Chem ; 293(17): 6482-6496, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29514982

RESUMEN

DNA interstrand cross-links (ICLs) block the progress of the replication and transcription machineries and can weaken chromosomal stability, resulting in various diseases. FANCD2-FANCI-associated nuclease (FAN1) is a conserved structure-specific nuclease that unhooks DNA ICLs independently of the Fanconi anemia pathway. Recent structural studies have proposed two different mechanistic features for ICL unhooking by human FAN1: a specific basic pocket that recognizes the terminal phosphate of a 1-nucleotide (nt) 5' flap or FAN1 dimerization. Herein, we show that despite lacking these features, Pseudomonas aeruginosa FAN1 (PaFAN1) cleaves substrates at ∼3-nt intervals and resolves ICLs. Crystal structures of PaFAN1 bound to various DNA substrates revealed that its conserved basic Arg/Lys patch comprising Arg-228 and Lys-260 recognizes phosphate groups near the 5' terminus of a DNA substrate with a 1-nt flap or a nick. Substitution of Lys-260 did not affect PaFAN1's initial endonuclease activity but significantly decreased its subsequent exonuclease activity and ICL unhooking. The Arg/Lys patch also interacted with phosphates at a 3-nt gap, and this interaction could drive movement of the scissile phosphates into the PaFAN1-active site. In human FAN1, the ICL-resolving activity was not affected by individual disruption of the Arg/Lys patch or basic pocket. However, simultaneous substitution of both FAN1 regions significantly reduced its ICL-resolving activity, suggesting that these two basic regions play a complementary role in ICL repair. On the basis of these findings, we propose a conserved role for two basic regions in FAN1 to guide ICL unhooking and to maintain genomic stability.


Asunto(s)
Proteínas Bacterianas/química , Desoxirribonucleasa I/química , Simulación de Dinámica Molecular , Pseudomonas aeruginosa/enzimología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Desoxirribonucleasa I/genética , Desoxirribonucleasa I/metabolismo , Dominios Proteicos , Pseudomonas aeruginosa/genética , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA