Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 281
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 84(7): 1257-1270.e6, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38377993

RESUMEN

Current base editors (BEs) use DNA deaminases, including cytidine deaminase in cytidine BE (CBE) or adenine deaminase in adenine BE (ABE), to facilitate transition nucleotide substitutions. Combining CBE or ABE with glycosylase enzymes can induce limited transversion mutations. Nonetheless, a critical demand remains for BEs capable of generating alternative mutation types, such as T>G corrections. In this study, we leveraged pre-trained protein language models to optimize a uracil-N-glycosylase (UNG) variant with altered specificity for thymines (eTDG). Notably, after two rounds of testing fewer than 50 top-ranking variants, more than 50% exhibited over 1.5-fold enhancement in enzymatic activities. When eTDG was fused with nCas9, it induced programmable T-to-S (G/C) substitutions and corrected db/db diabetic mutation in mice (up to 55%). Our findings not only establish orthogonal strategies for developing novel BEs but also demonstrate the capacities of protein language models for optimizing enzymes without extensive task-specific training data.


Asunto(s)
Ácidos Alcanesulfónicos , Edición Génica , Uracil-ADN Glicosidasa , Animales , Ratones , Mutación , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo
2.
Mol Cell ; 84(11): 2036-2052.e7, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38688279

RESUMEN

Alterations of bases in DNA constitute a major source of genomic instability. It is believed that base alterations trigger base excision repair (BER), generating DNA repair intermediates interfering with DNA replication. Here, we show that genomic uracil, a common type of base alteration, induces DNA replication stress (RS) without being processed by BER. In the absence of uracil DNA glycosylase (UNG), genomic uracil accumulates to high levels, DNA replication forks slow down, and PrimPol-mediated repriming is enhanced, generating single-stranded gaps in nascent DNA. ATR inhibition in UNG-deficient cells blocks the repair of uracil-induced gaps, increasing replication fork collapse and cell death. Notably, a subset of cancer cells upregulates UNG2 to suppress genomic uracil and limit RS, and these cancer cells are hypersensitive to co-treatment with ATR inhibitors and drugs increasing genomic uracil. These results reveal unprocessed genomic uracil as an unexpected source of RS and a targetable vulnerability of cancer cells.


Asunto(s)
Reparación del ADN , Replicación del ADN , Inestabilidad Genómica , Uracil-ADN Glicosidasa , Uracilo , Humanos , Uracilo/metabolismo , Uracil-ADN Glicosidasa/metabolismo , Uracil-ADN Glicosidasa/genética , Reparación del ADN/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Daño del ADN , Línea Celular Tumoral , Neoplasias/genética , Neoplasias/patología , Neoplasias/metabolismo
3.
Mol Cell ; 81(19): 4008-4025.e7, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34508659

RESUMEN

BRCA1/2 mutant tumor cells display an elevated mutation burden, the etiology of which remains unclear. Here, we report that these cells accumulate ssDNA gaps and spontaneous mutations during unperturbed DNA replication due to repriming by the DNA primase-polymerase PRIMPOL. Gap accumulation requires the DNA glycosylase SMUG1 and is exacerbated by depletion of the translesion synthesis (TLS) factor RAD18 or inhibition of the error-prone TLS polymerase complex REV1-Polζ by the small molecule JH-RE-06. JH-RE-06 treatment of BRCA1/2-deficient cells results in reduced mutation rates and PRIMPOL- and SMUG1-dependent loss of viability. Through cellular and animal studies, we demonstrate that JH-RE-06 is preferentially toxic toward HR-deficient cancer cells. Furthermore, JH-RE-06 remains effective toward PARP inhibitor (PARPi)-resistant BRCA1 mutant cells and displays additive toxicity with crosslinking agents or PARPi. Collectively, these studies identify a protective and mutagenic role for REV1-Polζ in BRCA1/2 mutant cells and provide the rationale for using REV1-Polζ inhibitors to treat BRCA1/2 mutant tumors.


Asunto(s)
Roturas del ADN de Cadena Simple , ADN Primasa/metabolismo , Replicación del ADN , ADN de Neoplasias/biosíntesis , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Enzimas Multifuncionales/metabolismo , Neoplasias/enzimología , Nucleotidiltransferasas/metabolismo , Reparación del ADN por Recombinación , Animales , Antineoplásicos/farmacología , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Línea Celular Tumoral , ADN Primasa/genética , ADN de Neoplasias/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Femenino , Células HEK293 , Humanos , Ratones Desnudos , Enzimas Multifuncionales/genética , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/genética , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Nucleic Acids Res ; 52(2): 784-800, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38000394

RESUMEN

Activation-induced cytidine deaminase (AID) interacts with replication protein A (RPA), the major ssDNA-binding protein, to promote deamination of cytosine to uracil in transcribed immunoglobulin (Ig) genes. Uracil-DNA glycosylase (UNG) acts in concert with AID during Ig diversification. In addition, UNG preserves genome integrity by base-excision repair (BER) in the overall genome. How UNG is regulated to support both mutagenic processing and error-free repair remains unknown. UNG is expressed as two isoforms, UNG1 and UNG2, which both contain an RPA-binding helix that facilitates uracil excision from RPA-coated ssDNA. However, the impact of this interaction in antibody diversification and genome maintenance has not been investigated. Here, we generated B-cell clones with targeted mutations in the UNG RPA-binding motif, and analysed class switch recombination (CSR), mutation frequency (5' Ig Sµ), and genomic uracil in clones representing seven Ung genotypes. We show that the UNG:RPA interaction plays a crucial role in both CSR and repair of AID-induced uracil at the Ig loci. By contrast, the interaction had no significant impact on total genomic uracil levels. Thus, RPA coordinates UNG during CSR and pre-replicative repair of mutagenic uracil in ssDNA but is not essential in post-replicative and canonical BER of uracil in dsDNA.


Asunto(s)
Proteína de Replicación A , Uracil-ADN Glicosidasa , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Reparación del ADN/genética , ADN de Cadena Simple/genética , Cambio de Clase de Inmunoglobulina/genética , Isotipos de Inmunoglobulinas/genética , Inmunoglobulinas/genética , Mutágenos , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , Uracilo/metabolismo , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Humanos , Animales , Ratones
5.
Mol Cell ; 68(5): 901-912.e3, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29220655

RESUMEN

DNA double-strand breaks (DSBs) occurring within fragile zones of less than 200 base pairs account for the formation of the most common human chromosomal translocations in lymphoid malignancies, yet the mechanism of how breaks occur remains unknown. Here, we have transferred human fragile zones into S. cerevisiae in the context of a genetic assay to understand the mechanism leading to DSBs at these sites. Our findings indicate that a combination of factors is required to sensitize these regions. Foremost, DNA strand separation by transcription or increased torsional stress can expose these DNA regions to damage from either the expression of human AID or increased oxidative stress. This damage causes DNA lesions that, if not repaired quickly, are prone to nuclease cleavage, resulting in DSBs. Our results provide mechanistic insight into why human neoplastic translocation fragile DNA sequences are more prone to enzymes or agents that cause longer-lived DNA lesions.


Asunto(s)
Cromosomas Humanos/genética , Citidina Desaminasa/genética , Roturas del ADN de Doble Cadena , ADN de Hongos/genética , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Saccharomyces cerevisiae/genética , Translocación Genética , Cromosomas Humanos/química , Cromosomas Humanos/metabolismo , Citidina Desaminasa/metabolismo , ADN de Hongos/química , ADN de Hongos/metabolismo , Proteínas de Unión al ADN , Endonucleasas/genética , Endonucleasas/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Fúngica de la Expresión Génica , Humanos , Conformación de Ácido Nucleico , Peroxidasas/genética , Peroxidasas/metabolismo , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Relación Estructura-Actividad , Transcripción Genética , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo
6.
Nucleic Acids Res ; 51(13): 6554-6565, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37283083

RESUMEN

UdgX excises uracil from uracil-containing DNA to concurrently form a covalent bond with the resulting AP-DNA. Structurally, UdgX is highly similar to family-4 UDGs (F4-UDGs). However, UdgX is unique in possessing a flexible R-loop (105KRRIH109). Among the class-defining motifs, while its motif A (51GEQPG55) diverged to possess Q53 in place of A53/G53 in F4-UDGs, motif B [178HPS(S/A)(L/V)(L/V)R184] has remained unchanged. Previously, we proposed an SN1 mechanism resulting in a covalent bond between H109 and AP-DNA. In this study, we investigated several single/double mutants of UdgX. The H109A, H109S, H109G, H109Q, H109C and H109K mutants gain conventional UDG activity to varying levels. The crystal structures of UdgX mutants show topological changes in their active sites, rationalizing their UDG activities. The E52Q, E52N and E52A mutants reveal that E52 forms a catalytic dyad with H109 to enhance its nucleophilicity. The Q53A mutant supports that UdgX specific evolution of Q53 occurred essentially to stabilize the R-loop conformation. The R184A mutation (motif B) supports the role of R184 in substrate-binding. Taken together, the structural, bioinformatics, and mutational studies suggest that UdgX diverged from F4-UDGs, and the emergence of the characteristic R-loop in UdgX is functionally assisted by A53/G53 to Q53 changes in motif A.


Asunto(s)
Uracil-ADN Glicosidasa , Dominio Catalítico , ADN/química , Reparación del ADN , Mutación , Uracilo , Uracil-ADN Glicosidasa/química , Uracil-ADN Glicosidasa/genética
7.
Plant J ; 113(3): 610-625, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36565011

RESUMEN

Base editing enables precise gene editing without requiring donor DNA or double-stranded breaks. To facilitate base editing tools, a uracil DNA glycosylase inhibitor (UGI) was fused to cytidine deaminase-Cas nickase to inhibit uracil DNA glycosylase (UDG). Herein, we revealed that the bacteriophage PBS2-derived UGI of the cytosine base editor (CBE) could not inhibit archaic Type IV UDG in oligoploid cyanobacteria. To overcome the limitation of the CBE, dCas12a-assisted gene repression of the udg allowed base editing at the desired targets with up to 100% mutation frequencies, and yielded correct phenotypes of desired mutants in cyanobacteria. Compared with the original CBE (BE3), base editing was analyzed within a broader C4-C16 window with a strong TC-motif preference. Using multiplexed CyanoCBE, while udg was repressed, simultaneous base editing at two different sites was achieved with lower mutation frequencies than single CBE. Our discovery of a Type IV UDG that is not inhibited by the UGI of the CBE in cyanobacteria and the development of dCas12a-mediated base editing should facilitate the application of base editing not only in cyanobacteria, but also in archaea and green algae that possess Type IV UDGs. We revealed the bacteriophage-derived UGI of the base editor did not repress Type IV UDG in cyanobacteria. To overcome the limitation, orthogonal dCas12a interference was successfully applied to repress the UDG gene expression in cyanobacteria during base editing occurred, yielding a premature translational termination at desired targets. This study will open a new opportunity to perform base editing with Type IV UDGs in archaea and green algae.


Asunto(s)
Cianobacterias , Uracil-ADN Glicosidasa , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Edición Génica , ADN , Reparación del ADN , Cianobacterias/genética , Cianobacterias/metabolismo , Citosina
8.
Plant J ; 115(6): 1633-1646, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37278489

RESUMEN

DNA glycosylases remove mispaired or modified bases from DNA initiating the base excision repair (BER) pathway. The DNA glycosylase MBD4 (methyl-CpG-binding domain protein 4) has been functionally characterized in mammals, but not yet in plants, where it is called MBD4-like (MBD4L). Mammalian MBD4 and Arabidopsis recombinant MBD4L excise U and T mispaired with G, as well as 5-fluorouracil (5-FU) and 5-bromouracil (5-BrU) in vitro. Here, we investigate the ability of Arabidopsis MBD4L to remove some of these substrates from the nuclear genome in vivo in coordination with uracil DNA glycosylase (AtUNG). We found that mbd4l mutants are hypersensitive to 5-FU and 5-BrU, as they displayed smaller size, less root growth, and higher cell death than control plants in both media. Using comet assays, we determined BER-associated DNA fragmentation in isolated nuclei and observed reduced DNA breaks in mbd4l plants under both conditions, but particularly with 5-BrU. The use of ung and ung x mbd4l mutants in these assays indicated that both MBD4L and AtUNG trigger nuclear DNA fragmentation in response to 5-FU. Consistently, we here report the nuclear localization of AtUNG based on the expression of AtUNG-GFP/RFP constructs in transgenic plants. Interestingly, MBD4L and AtUNG are transcriptionally coordinated but display not completely overlapping functions. MBD4L-deficient plants showed reduced expression of BER genes and enhanced expression of DNA damage response (DDR) gene markers. Overall, our findings indicate that Arabidopsis MBD4L is critical for maintaining nuclear genome integrity and preventing cell death under genotoxic stress conditions.


Asunto(s)
Arabidopsis , Animales , Arabidopsis/genética , Arabidopsis/metabolismo , ADN/metabolismo , Daño del ADN , Reparación del ADN/genética , Fluorouracilo/metabolismo , Mamíferos/genética , Mamíferos/metabolismo , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo
9.
Mol Microbiol ; 120(2): 298-306, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37452011

RESUMEN

DNA glycosylases protect genetic fidelity during DNA replication by removing potentially mutagenic chemically damaged DNA bases. Bacterial Lhr proteins are well-characterized DNA repair helicases that are fused to additional 600-700 amino acids of unknown function, but with structural homology to SecB chaperones and AlkZ DNA glycosylases. Here, we identify that Escherichia coli Lhr is a uracil-DNA glycosylase (UDG) that depends on an active site aspartic acid residue. We show that the Lhr DNA helicase activity is functionally independent of the UDG activity, but that the helicase domains are required for fully active UDG activity. Consistent with UDG activity, deletion of lhr from the E. coli chromosome sensitized cells to oxidative stress that triggers cytosine deamination to uracil. The ability of Lhr to translocate single-stranded DNA and remove uracil bases suggests a surveillance role to seek and remove potentially mutagenic base changes during replication stress.


Asunto(s)
Escherichia coli , Uracil-ADN Glicosidasa , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Secuencia de Aminoácidos , ADN/metabolismo , Uracilo/química , Reparación del ADN , ADN Helicasas/metabolismo , Proteínas Bacterianas/metabolismo
10.
Biol Pharm Bull ; 47(7): 1275-1281, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38987176

RESUMEN

The generation of DNA damage causes mutations and consequently cancer. Reactive oxygen species are important sources of DNA damage and some mutation signatures found in human cancers. 8-Oxo-7,8-dihydroguanine (GO, 8-hydroxyguanine) is one of the most abundant oxidized bases and induces a G→T transversion mutation at the modified site. The damaged G base also causes untargeted base substitution mutations at the G bases of 5'-GpA-3' dinucleotides (action-at-a-distance mutations) in human cells, and the cytosine deaminase apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3 (APOBEC3) is involved in the mutation process. The deaminated cytosine, i.e., uracil, bases are expected to be removed by uracil DNA glycosylase. Most of the substitution mutations at the G bases of 5'-GpA-3' might be caused by abasic sites formed by the glycosylase. In this study, we expressed the uracil DNA glycosylase inhibitor from Bacillus subtilis bacteriophage PBS2 in human U2OS cells and examined the effects on the GO-induced action-at-a-distance mutations. The inhibition of uracil DNA glycosylase increased the mutation frequency, and in particular, the frequency of G→A transitions. These results indicated that uracil DNA glycosylase, in addition to APOBEC3, is involved in the untargeted mutation process induced by GO.


Asunto(s)
Guanina , Mutación , Uracil-ADN Glicosidasa , Humanos , Guanina/análogos & derivados , Guanina/metabolismo , Uracil-ADN Glicosidasa/metabolismo , Uracil-ADN Glicosidasa/genética , Línea Celular Tumoral , Daño del ADN , Bacillus subtilis/genética , Bacteriófagos/genética
11.
Nucleic Acids Res ; 50(9): 5384-5399, 2022 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-35544322

RESUMEN

Establishing saturated mutagenesis in a specific gene through gene editing is an efficient approach for identifying the relationships between mutations and the corresponding phenotypes. CRISPR/Cas9-based sgRNA library screening often creates indel mutations with multiple nucleotides. Single base editors and dual deaminase-mediated base editors can achieve only one and two types of base substitutions, respectively. A new glycosylase base editor (CGBE) system, in which the uracil glycosylase inhibitor (UGI) is replaced with uracil-DNA glycosylase (UNG), was recently reported to efficiently induce multiple base conversions, including C-to-G, C-to-T and C-to-A. In this study, we fused a CGBE with ABE to develop a new type of dual deaminase-mediated base editing system, the AGBE system, that can simultaneously introduce 4 types of base conversions (C-to-G, C-to-T, C-to-A and A-to-G) as well as indels with a single sgRNA in mammalian cells. AGBEs can be used to establish saturated mutant populations for verification of the functions and consequences of multiple gene mutation patterns, including single-nucleotide variants (SNVs) and indels, through high-throughput screening.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Animales , Mutación INDEL , Mamíferos/genética , Mutación , Uracil-ADN Glicosidasa/genética
12.
Nucleic Acids Res ; 50(18): 10449-10468, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36156150

RESUMEN

Single-strand selective uracil-DNA glycosylase 1 (SMUG1) initiates base excision repair (BER) of uracil and oxidized pyrimidines. SMUG1 status has been associated with cancer risk and therapeutic response in breast carcinomas and other cancer types. However, SMUG1 is a multifunctional protein involved, not only, in BER but also in RNA quality control, and its function in cancer cells is unclear. Here we identify several novel SMUG1 interaction partners that functions in many biological processes relevant for cancer development and treatment response. Based on this, we hypothesized that the dominating function of SMUG1 in cancer might be ascribed to functions other than BER. We define a bad prognosis signature for SMUG1 by mapping out the SMUG1 interaction network and found that high expression of genes in the bad prognosis network correlated with lower survival probability in ER+ breast cancer. Interestingly, we identified hsa-let-7b-5p microRNA as an upstream regulator of the SMUG1 interactome. Expression of SMUG1 and hsa-let-7b-5p were negatively correlated in breast cancer and we found an inhibitory auto-regulatory loop between SMUG1 and hsa-let-7b-5p in the MCF7 breast cancer cells. We conclude that SMUG1 functions in a gene regulatory network that influence the survival and treatment response in several cancers.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Humanos , MicroARNs/genética , Pronóstico , Uracilo/metabolismo , Uracil-ADN Glicosidasa/genética
13.
Nucleic Acids Res ; 50(21): 12039-12057, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-36444883

RESUMEN

The human APOBEC family of eleven cytosine deaminases use RNA and single-stranded DNA (ssDNA) as substrates to deaminate cytosine to uracil. This deamination event has roles in lipid metabolism by altering mRNA coding, adaptive immunity by causing evolution of antibody genes, and innate immunity through inactivation of viral genomes. These benefits come at a cost where some family members, primarily from the APOBEC3 subfamily (APOBEC3A-H, excluding E), can cause off-target deaminations of cytosine to form uracil on transiently single-stranded genomic DNA, which induces mutations that are associated with cancer evolution. Since uracil is only promutagenic, the mutations observed in cancer genomes originate only when uracil is not removed by uracil DNA glycosylase (UNG) or when the UNG-induced abasic site is erroneously repaired. However, when ssDNA is present, replication protein A (RPA) binds and protects the DNA from nucleases or recruits DNA repair proteins, such as UNG. Thus, APOBEC enzymes must compete with RPA to access their substrate. Certain APOBEC enzymes can displace RPA, bind and scan ssDNA efficiently to search for cytosines, and can become highly overexpressed in tumor cells. Depending on the DNA replication conditions and DNA structure, RPA can either be in excess or deficient. Here we discuss the interplay between these factors and how despite RPA, multiple cancer genomes have a mutation bias at cytosines indicative of APOBEC activity.


Asunto(s)
ADN de Cadena Simple , Proteína de Replicación A , Humanos , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , ADN de Cadena Simple/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Replicación del ADN/genética , Citosina/metabolismo , ADN/metabolismo , Uracilo/metabolismo , Desaminasas APOBEC/genética , Desaminasas APOBEC/metabolismo , Desaminación
14.
Chembiochem ; 24(10): e202200765, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-36883884

RESUMEN

DNA repair proteins participate in extensive protein-protein interactions that promote the formation of DNA repair complexes. To understand how complex formation affects protein function during base excision repair, we used SpyCatcher/SpyTag ligation to produce a covalent complex between human uracil DNA glycosylase (UNG2) and replication protein A (RPA). Our covalent "RPA-Spy-UNG2" complex could identify and excise uracil bases in duplex areas next to ssDNA-dsDNA junctions slightly faster than the wild-type proteins, but this was highly dependent on DNA structure, as the turnover of the RPA-Spy-UNG2 complex slowed at DNA junctions where RPA tightly engaged long ssDNA sections. Conversely, the enzymes preferred uracil sites in ssDNA where RPA strongly enhanced uracil excision by UNG2 regardless of ssDNA length. Finally, RPA was found to promote UNG2 excision of two uracil sites positioned across a ssDNA-dsDNA junction, and dissociation of UNG2 from RPA enhanced this process. Our approach of ligating together RPA and UNG2 to reveal how complex formation affects enzyme function could be applied to examine other assemblies of DNA repair proteins.


Asunto(s)
Reparación del ADN , Proteína de Replicación A , Uracil-ADN Glicosidasa , Humanos , ADN/metabolismo , Replicación del ADN , ADN de Cadena Simple , Cinética , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , Uracilo/metabolismo , Uracil-ADN Glicosidasa/genética
15.
Nat Immunol ; 12(1): 70-6, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21151102

RESUMEN

Activation-induced deaminase (AID) initiates diversity of immunoglobulin genes through deamination of cytosine to uracil. Two opposing models have been proposed for the deamination of DNA or RNA by AID. Although most data support DNA deamination, there is no physical evidence of uracil residues in immunoglobulin genes. Here we demonstrate their presence by determining the sensitivity of DNA to digestion with uracil DNA glycosylase (UNG) and abasic endonuclease. Using several methods of detection, we identified uracil residues in the variable and switch regions. Uracil residues were generated within 24 h of B cell stimulation, were present on both DNA strands and were found to replace mainly cytosine bases. Our data provide direct evidence for the model that AID functions by deaminating cytosine residues in DNA.


Asunto(s)
Linfocitos B/metabolismo , Citidina Desaminasa/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Uracil-ADN Glicosidasa/metabolismo , Animales , Variación Antigénica/genética , Linfocitos B/inmunología , Linfocitos B/patología , Células Cultivadas , Citidina Desaminasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Cambio de Clase de Inmunoglobulina , Región Variable de Inmunoglobulina , Interleucina-4/inmunología , Interleucina-4/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Químicos , Bazo/patología , Uracilo/análisis , Uracil-ADN Glicosidasa/genética
16.
PLoS Genet ; 16(12): e1008960, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33362210

RESUMEN

Most B cell lymphomas originate from B cells that have germinal center (GC) experience and bear chromosome translocations and numerous point mutations. GC B cells remodel their immunoglobulin (Ig) genes by somatic hypermutation (SHM) and class switch recombination (CSR) in their Ig genes. Activation Induced Deaminase (AID) initiates CSR and SHM by generating U:G mismatches on Ig DNA that can then be processed by Uracyl-N-glycosylase (UNG). AID promotes collateral damage in the form of chromosome translocations and off-target SHM, however, the exact contribution of AID activity to lymphoma generation and progression is not completely understood. Here we show using a conditional knock-in strategy that AID supra-activity alone is not sufficient to generate B cell transformation. In contrast, in the absence of UNG, AID supra-expression increases SHM and promotes lymphoma. Whole exome sequencing revealed that AID heavily contributes to lymphoma SHM, promoting subclonal variability and a wider range of oncogenic variants. Thus, our data provide direct evidence that UNG is a brake to AID-induced intratumoral heterogeneity and evolution of B cell lymphoma.


Asunto(s)
Citidina Desaminasa/genética , Heterogeneidad Genética , Linfoma de Células B/genética , Uracil-ADN Glicosidasa/genética , Animales , Transformación Celular Neoplásica/genética , Células Cultivadas , Evolución Clonal , Citidina Desaminasa/metabolismo , Femenino , Linfoma de Células B/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Uracil-ADN Glicosidasa/metabolismo
17.
Int J Mol Sci ; 24(11)2023 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-37298065

RESUMEN

The protein encoded by the vaccinia virus D4R gene has base excision repair uracil-DNA N-glycosylase (vvUNG) activity and also acts as a processivity factor in the viral replication complex. The use of a protein unlike PolN/PCNA sliding clamps is a unique feature of orthopoxviral replication, providing an attractive target for drug design. However, the intrinsic processivity of vvUNG has never been estimated, leaving open the question whether it is sufficient to impart processivity to the viral polymerase. Here, we use the correlated cleavage assay to characterize the translocation of vvUNG along DNA between two uracil residues. The salt dependence of the correlated cleavage, together with the similar affinity of vvUNG for damaged and undamaged DNA, support the one-dimensional diffusion mechanism of lesion search. Unlike short gaps, covalent adducts partly block vvUNG translocation. Kinetic experiments show that once a lesion is found it is excised with a probability ~0.76. Varying the distance between two uracils, we use a random walk model to estimate the mean number of steps per association with DNA at ~4200, which is consistent with vvUNG playing a role as a processivity factor. Finally, we show that inhibitors carrying a tetrahydro-2,4,6-trioxopyrimidinylidene moiety can suppress the processivity of vvUNG.


Asunto(s)
Uracil-ADN Glicosidasa , Virus Vaccinia , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Virus Vaccinia/genética , ADN/metabolismo , Replicación Viral , Reparación del ADN , Uracilo/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Replicación del ADN
18.
Molecules ; 28(3)2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36770637

RESUMEN

Although genomic DNA is predominantly duplex under physiological conditions, particular sequence motifs can favor the formation of alternative secondary structures, including the G-quadruplex. These structures can exist within gene promoters, telomeric DNA, and regions of the genome frequently found altered in human cancers. DNA is also subject to hydrolytic and oxidative damage, and its local structure can influence the type of damage and its magnitude. Although the repair of endogenous DNA damage by the base excision repair (BER) pathway has been extensively studied in duplex DNA, substantially less is known about repair in non-duplex DNA structures. Therefore, we wanted to better understand the effect of DNA damage and repair on quadruplex structure. We first examined the effect of placing pyrimidine damage products uracil, 5-hydroxymethyluracil, the chemotherapy agent 5-fluorouracil, and an abasic site into the loop region of a 22-base telomeric repeat sequence known to form a G-quadruplex. Quadruplex formation was unaffected by these analogs. However, the activity of the BER enzymes were negatively impacted. Uracil DNA glycosylase (UDG) and single-strand selective monofunctional uracil DNA glycosylase (SMUG1) were inhibited, and apurinic/apyrimidinic endonuclease 1 (APE1) activity was completely blocked. Interestingly, when we performed studies placing DNA repair intermediates into the strand opposite the quadruplex, we found that they destabilized the duplex and promoted quadruplex formation. We propose that while duplex is the preferred configuration, there is kinetic conversion between duplex and quadruplex. This is supported by our studies using a quadruplex stabilizing molecule, pyridostatin, that is able to promote quadruplex formation starting from duplex DNA. Our results suggest how DNA damage and repair intermediates can alter duplex-quadruplex equilibrium.


Asunto(s)
Reparación del ADN , Uracil-ADN Glicosidasa , Humanos , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Daño del ADN , Estrés Oxidativo/genética , ADN/química
19.
Biophys J ; 121(7): 1276-1288, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35183522

RESUMEN

Polymerase chain reaction (PCR) is a powerful tool to diagnose infectious diseases. Uracil DNA glycosylase (UDG) is broadly used to remove carryover contamination in PCR. However, UDG can contribute to false negative results when not inactivated completely, leading to DNA degradation during the amplification step. In this study, we designed novel thermolabile UDG derivatives by supercomputing molecular dynamic simulations and residual network analysis. Based on enzyme activity analysis, thermolability, thermal stability, and biochemical experiments of Escherichia coli-derived UDG and 22 derivatives, we uncovered that the UDG D43A mutant eliminated the false negative problem, demonstrated high efficiency, and offered great benefit for use in PCR diagnosis. We further obtained structural and thermodynamic insights into the role of the D43A mutation, including perturbed protein structure near D43; weakened pairwise interactions of D43 with K42, N46, and R80; and decreased melting temperature and native fraction of the UDG D43A mutant compared with wild-type UDG.


Asunto(s)
Escherichia coli , Uracil-ADN Glicosidasa , Escherichia coli/metabolismo , Mutación , Uracil-ADN Glicosidasa/química , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo
20.
J Gen Virol ; 103(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35438620

RESUMEN

The covalently closed circular DNA (cccDNA) of hepatitis B virus (HBV) plays a key role in the persistence of viral infection. We have previously shown that overexpression of an antiviral factor APOBEC3G (A3G) induces hypermutation in duck HBV (DHBV) cccDNA, whereas uracil-DNA-glycosylase (UNG) reduces these mutations. In this study, using cell-culture systems, we examined whether endogenous A3s and UNG affect HBV cccDNA mutation frequency. IFNγ stimulation induced a significant increase in endogenous A3G expression and cccDNA hypermutation. UNG inhibition enhanced the IFNγ-mediated hypermutation frequency. Transfection of reconstructed cccDNA revealed that this enhanced hypermutation caused a reduction in viral replication. These results suggest that the balance of endogenous A3s and UNG activities affects HBV cccDNA mutation and replication competency.


Asunto(s)
Virus de la Hepatitis B del Pato , Hepatitis B Crónica , Hepatitis B , Desaminasas APOBEC/genética , Desaminasas APOBEC/metabolismo , ADN Circular/genética , ADN Circular/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Virus de la Hepatitis B del Pato/genética , Virus de la Hepatitis B del Pato/metabolismo , Virus de la Hepatitis B/fisiología , Humanos , Uracilo , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA