106 results on '"Radicella JP"'
Search Results
2. Adaptive Mutation in Escherichia coli : a Role for Conjugation
- Author
-
Park Pu, Radicella Jp, and Maurice S. Fox
- Subjects
DNA Replication ,Reversion ,Lactose ,medicine.disease_cause ,F Factor ,Plasmid ,Bacterial Proteins ,Adaptive mutation ,Escherichia coli ,Lac Repressors ,medicine ,Selection, Genetic ,Allele ,Frameshift Mutation ,Alleles ,Genetics ,Mutation ,Multidisciplinary ,biology ,Escherichia coli Proteins ,biology.organism_classification ,Adaptation, Physiological ,Enterobacteriaceae ,Culture Media ,Repressor Proteins ,Lac Operon ,Conjugation, Genetic ,bacteria ,Bacteria - Abstract
When subjected to selective conditions that impose starvation, a bacterial population can accumulate mutations, called adaptive, that allow colony formation. Here, the reversion of a lac allele under selective conditions, in a model system using Escherichia coli with the lac mutation on an F' plasmid, was shown to require the conjugational capacity of the plasmid. Reversion associated with transfer was shown, and when the same lac allele was chromosomal, reversion to Lac+ was 25 to 50 times less frequent. Postplating reversion was 25 times less when mating was inhibited by the presence of detergent. Mutability associated with conjugation provides new ways of thinking about the origin of adaptive mutations.
- Published
- 1995
3. Distinct spatiotemporal patterns and PARP dependence of XRCC1 recruitment to single-strand break and base excision repair
- Author
-
Campalans, A, Kortulewski, T, Amouroux, R, Menoni, H, Vermeulen, Wim, Radicella, JP, Campalans, A, Kortulewski, T, Amouroux, R, Menoni, H, Vermeulen, Wim, and Radicella, JP
- Published
- 2013
4. Cloning and characterization of hOGG1, a human homolog of the OGG1 gene of Saccharomyces cerevisiae
- Author
-
Fox Ms, Radicella Jp, Desmaze C, C Dhérin, and Serge Boiteux
- Subjects
DNA, Complementary ,Guanine ,Base pair ,Saccharomyces cerevisiae ,Mutant ,Genes, Fungal ,Molecular Sequence Data ,DNA Glycosylases ,Substrate Specificity ,Gene product ,chemistry.chemical_compound ,Escherichia coli ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Lyase activity ,Gene ,N-Glycosyl Hydrolases ,Multidisciplinary ,biology ,Sequence Homology, Amino Acid ,Escherichia coli Proteins ,Genetic Complementation Test ,Chromosome Mapping ,Biological Sciences ,biology.organism_classification ,Molecular biology ,Phenotype ,chemistry ,Biochemistry ,DNA-Formamidopyrimidine Glycosylase ,DNA glycosylase ,Chromosomes, Human, Pair 3 ,DNA - Abstract
The OGG1 gene of Saccharomyces cerevisiae encodes a DNA glycosylase activity that is a functional analog of the Fpg protein from Escherichia coli and excises 7,8-dihydro-8-oxoguanine (8-oxoG) from damaged DNA. The repair of this ubiquitous kind of oxidative damage is essential to prevent mutations both in bacteria and in yeast. A human cDNA clone carrying an ORF displaying homology to the yeast protein was identified. The predicted protein has 345 amino acids and a molecular mass of 39 kDa. This protein shares a 38% sequence identity with the yeast Ogg1 protein, adding this novel human gene product to the growing family of enzymes that the repair of oxidatively damaged bases and are related to the E. coli endonuclease III. Northern blot analysis indicates that this gene, localized to chromosome 3p25, is ubiquitously expressed in human tissues. The cloned coding sequence was expressed in an E. coli strain that carried a disrupted fpg gene, the bacterial functional analog of OGG1 . Cell-free extracts from these cultures displayed a specific lyase activity on duplex DNA that carried an 8-oxoG/C base pair. The products of the reaction are consistent with an enzymatic activity like the one displayed by the yeast Ogg1. Analysis of the substrate specificity reveals a very strong preference for DNA fragments harboring 8-oxoG/C base pairs. The pattern of specificity correlates well with the one found for the yeast enzyme. Moreover, when the human coding sequence was expressed in a yeast strain mutant in OGG1 it was able to complement the spontaneous mutator phenotype. These results make this novel gene ( hOGG1 ) a strong candidate for the human homolog of the yeast OGG1 and suggest an important role of its product in the protection of the genome from the mutagenic effects of the oxidatively damaged purines.
- Published
- 1997
5. Réparation de l'ADN et cancer : Les gènes de réparation des bases oxydées dans l'ADN sont-ils des gènes suppresseurs de tumeurs ?
- Author
-
Boiteux, S, primary and Radicella, JP, additional
- Published
- 1998
- Full Text
- View/download PDF
6. DprA recruits ComM to facilitate recombination during natural transformation in Gram-negative bacteria.
- Author
-
Dalia TN, Machouri M, Lacrouts C, Fauconnet Y, Guerois R, Andreani J, Radicella JP, and Dalia AB
- Abstract
Natural transformation (NT) represents one of the major modes of horizontal gene transfer in bacterial species. During NT, cells can take up free DNA from the environment and integrate it into their genome by homologous recombination. While NT has been studied for >90 years, the molecular details underlying this recombination remain poorly understood. Recent work has demonstrated that ComM is an NT-specific hexameric helicase that promotes recombinational branch migration in Gram-negative bacteria. How ComM is loaded onto the post-synaptic recombination intermediate during NT, however, remains unclear. Another NT-specific recombination mediator protein that is ubiquitously conserved in both Gram-positive and Gram-negative bacteria is DprA. Here, we uncover that DprA homologs in Gram-negative species contain a C-terminal winged helix domain that is predicted to interact with ComM by AlphaFold. Using Helicobacter pylori and Vibrio cholerae as model systems, we demonstrate that ComM directly interacts with the DprA winged-helix domain, and that this interaction is critical for DprA to recruit ComM to the recombination site to promote branch migration during NT. These results advance our molecular understanding of recombination during this conserved mode of horizontal gene transfer. Furthermore, they demonstrate how structural modeling can help uncover unexpected interactions between well-studied proteins to provide deep mechanistic insight into the molecular coordination required for their activity., Significance Statement: Bacteria can acquire novel traits like antibiotic resistance and virulence through horizontal gene transfer by natural transformation. During this process, cells take up free DNA from the environment and integrate it into their genome by homologous recombination. Many of the molecular details underlying this process, however, remain incompletely understood. In this study, we identify a new protein-protein interaction between ComM and DprA, two factors that promote homologous recombination during natural transformation in Gram-negative species. Through a combination of bioinformatics, structural modeling, cell biological assays, and complementary genetic approaches, we demonstrate that this interaction is required for DprA to recruit ComM to the site of homologous recombination.
- Published
- 2024
- Full Text
- View/download PDF
7. Targeted nuclear irradiation with a proton microbeam induces oxidative DNA base damage and triggers the recruitment of DNA glycosylases OGG1 and NTH1.
- Author
-
Robeska E, Lalanne K, Vianna F, Sutcu HH, Khobta A, Busso D, Radicella JP, Campalans A, and Baldeyron C
- Subjects
- Humans, Protons, DNA Repair, Oxidative Stress, DNA Damage, DNA metabolism, DNA Glycosylases metabolism
- Abstract
DNA is the major target of radiation therapy of malignant tumors. Ionizing radiation (IR) induces a variety of DNA lesions, including chemically modified bases and strand breaks. The use of proton beam therapy for cancer treatment is ramping up, as it is expected to reduce normal tissue damage. Thus, it is important to understand the molecular mechanisms of recognition, signaling, and repair of DNA damage induced by protons in the perspective of assessing not only the risk associated with human exposure to IR but also the possibility to improve the efficacy of therapy. Here, we used targeted irradiation of nuclear regions of living cells with controlled number of protons at a high spatio-temporal resolution to detect the induced base lesions and characterize the recruitment kinetics of the specific DNA glycosylases to DNA damage sites. We show that localized irradiation with 4 MeV protons induces, in addition to DNA double strand breaks (DSBs), the oxidized bases 7,8-dihydro-8-oxoguanine (8-oxoG) and thymine glycol (TG) at the site of irradiation. Consistently, the DNA glycosylases OGG1 and NTH1, capable of excising 8-oxoG and TG, respectively, and initiating the base excision repair (BER) pathway, are recruited to the site of damage. To our knowledge, this is the first direct evidence indicating that proton microbeams induce oxidative base damage, and thus implicating BER in the repair of DNA lesions induced by protons., Competing Interests: Declaration of Competing Interest The authors declare that there are no conflicts of interest. There was no financial or non-financial assistance provided by a third party for the reported work. There are no patents or copyrights relevant to this work., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Identification of key residues of the DNA glycosylase OGG1 controlling efficient DNA sampling and recruitment to oxidized bases in living cells.
- Author
-
D'Augustin O, Gaudon V, Siberchicot C, Smith R, Chapuis C, Depagne J, Veaute X, Busso D, Di Guilmi AM, Castaing B, Radicella JP, Campalans A, and Huet S
- Subjects
- Humans, Cell Nucleus genetics, Cell Nucleus metabolism, DNA chemistry, DNA Repair, DNA Glycosylases metabolism
- Abstract
The DNA-glycosylase OGG1 oversees the detection and clearance of the 7,8-dihydro-8-oxoguanine (8-oxoG), which is the most frequent form of oxidized base in the genome. This lesion is deeply buried within the double-helix and its detection requires careful inspection of the bases by OGG1 via a mechanism that remains only partially understood. By analyzing OGG1 dynamics in the nucleus of living human cells, we demonstrate that the glycosylase constantly samples the DNA by rapidly alternating between diffusion within the nucleoplasm and short transits on the DNA. This sampling process, that we find to be tightly regulated by the conserved residue G245, is crucial for the rapid recruitment of OGG1 at oxidative lesions induced by laser micro-irradiation. Furthermore, we show that residues Y203, N149 and N150, while being all involved in early stages of 8-oxoG probing by OGG1 based on previous structural data, differentially regulate the sampling of the DNA and recruitment to oxidative lesions., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2023
- Full Text
- View/download PDF
9. A noncanonical response to replication stress protects genome stability through ROS production, in an adaptive manner.
- Author
-
Ragu S, Droin N, Matos-Rodrigues G, Barascu A, Caillat S, Zarkovic G, Siberchicot C, Dardillac E, Gelot C, Guirouilh-Barbat J, Radicella JP, Ishchenko AA, Ravanat JL, Solary E, and Lopez BS
- Subjects
- Humans, Reactive Oxygen Species metabolism, Cytokines genetics, Genomic Instability, NF-kappa B metabolism, NADPH Oxidases genetics, NADPH Oxidases metabolism
- Abstract
Cells are inevitably challenged by low-level/endogenous stresses that do not arrest DNA replication. Here, in human primary cells, we discovered and characterized a noncanonical cellular response that is specific to nonblocking replication stress. Although this response generates reactive oxygen species (ROS), it induces a program that prevents the accumulation of premutagenic 8-oxoguanine in an adaptive way. Indeed, replication stress-induced ROS (RIR) activate FOXO1-controlled detoxification genes such as SEPP1, catalase, GPX1, and SOD2. Primary cells tightly control the production of RIR: They are excluded from the nucleus and are produced by the cellular NADPH oxidases DUOX1/DUOX2, whose expression is controlled by NF-κB, which is activated by PARP1 upon replication stress. In parallel, inflammatory cytokine gene expression is induced through the NF-κB-PARP1 axis upon nonblocking replication stress. Increasing replication stress intensity accumulates DNA double-strand breaks and triggers the suppression of RIR by p53 and ATM. These data underline the fine-tuning of the cellular response to stress that protects genome stability maintenance, showing that primary cells adapt their responses to replication stress severity., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
10. OGG1 competitive inhibitors show important off-target effects by directly inhibiting efflux pumps and disturbing mitotic progression.
- Author
-
Tanushi X, Pinna G, Vandamme M, Siberchicot C, D'Augustin O, Di Guilmi AM, Radicella JP, Castaing B, Smith R, Huet S, Leteurtre F, and Campalans A
- Abstract
One of the most abundant DNA lesions induced by Reactive oxygen species (ROS) is 8-oxoG, a highly mutagenic lesion that compromises genetic instability when not efficiently repaired. 8-oxoG is specifically recognized by the DNA-glycosylase OGG1 that excises the base and initiates the Base Excision Repair pathway (BER). Furthermore, OGG1 has not only a major role in DNA repair but it is also involved in transcriptional regulation. Cancer cells are particularly exposed to ROS, thus challenging their capacity to process oxidative DNA damage has been proposed as a promising therapeutic strategy for cancer treatment. Two competitive inhibitors of OGG1 (OGG1i) have been identified, TH5487 and SU0268, which bind to the OGG1 catalytic pocket preventing its fixation to the DNA. Early studies with these inhibitors show an enhanced cellular sensitivity to cytotoxic drugs and a reduction in the inflammatory response. Our study uncovers two unreported off-targets effects of these OGG1i that are independent of OGG1. In vitro and in cellulo approaches have unveiled that OGG1i TH5487 and SU0268, despite an unrelated molecular structure, are able to inhibit some members of the ABC family transporters, in particular ABC B1 (MDR1) and ABC G2 (BCRP). The inhibition of these efflux pumps by OGG1 inhibitors results in a higher intra-cellular accumulation of various fluorescent probes and drugs, and largely contributes to the enhanced cytotoxicity observed when the inhibitors are combined with cytotoxic agents. Furthermore, we found that SU0268 has an OGG1-independent anti-mitotic activity-by interfering with metaphase completion-resulting in a high cellular toxicity. These two off-target activities are observed at concentrations of OGG1i that are normally used for in vivo studies. It is thus critical to consider these previously unreported non-specific effects when interpreting studies using TH5487 and SU0268 in the context of OGG1 inhibition. Additionally, our work highlights the persistent need for new specific inhibitors of the enzymatic activity of OGG1., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Tanushi, Pinna, Vandamme, Siberchicot, D’Augustin, Di Guilmi, Radicella, Castaing, Smith, Huet, Leteurtre and Campalans.)
- Published
- 2023
- Full Text
- View/download PDF
11. Foetal exposure to the bisphenols BADGE and BPAF impairs meiosis through DNA oxidation in mouse ovaries.
- Author
-
Abdallah S, Jampy A, Moison D, Wieckowski M, Messiaen S, Martini E, Campalans A, Radicella JP, Rouiller-Fabre V, Livera G, and Guerquin MJ
- Subjects
- Female, Mice, Animals, Benzhydryl Compounds toxicity, DNA, Aneuploidy, Meiosis, Ovary
- Abstract
Many endocrine disruptors have been proven to impair the meiotic process which is required for the production of healthy gametes. Bisphenol A is emblematic of such disruptors, as it impairs meiotic prophase I and causes oocyte aneuploidy following in utero exposure. However, the mechanisms underlying these deleterious effects remain poorly understood. Furthermore, the increasing use of BPA alternatives raises concerns for public health. Here, we investigated the effects of foetal exposure to two BPA alternatives, bisphenol A Diglycidyl Ether (BADGE) and bisphenol AF (BPAF), on oogenesis in mice. These compounds delay meiosis initiation, increase the number of MLH1 foci per cell and induce oocyte aneuploidy. We further demonstrate that these defects are accompanied by changes in gene expression in foetal premeiotic germ cells and aberrant mRNA splicing of meiotic genes. We observed an increase in DNA oxidation after exposure to BPA alternatives. Specific induction of oxidative DNA damage during foetal germ cell differentiation causes similar defects during oogenesis, as observed in 8-oxoguanine DNA Glycosylase (OGG1)-deficient mice or after in utero exposure to potassium bromate (KBrO3), an inducer of oxidative DNA damage. The supplementation of BPA alternatives with N-acetylcysteine (NAC) counteracts the effects of bisphenols on meiosis. Together, our results propose oxidative DNA lesion as an event that negatively impacts female meiosis with major consequences on oocyte quality. This could be a common mechanism of action for numerous environmental pro-oxidant pollutants, and its discovery, could lead to reconsider the adverse effect of bisphenol mixtures that are simultaneously present in our environment., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. ComFC mediates transport and handling of single-stranded DNA during natural transformation.
- Author
-
Damke PP, Celma L, Kondekar SM, Di Guilmi AM, Marsin S, Dépagne J, Veaute X, Legrand P, Walbott H, Vercruyssen J, Guérois R, Quevillon-Cheruel S, and Radicella JP
- Subjects
- Bacterial Proteins genetics, Bacterial Proteins metabolism, DNA metabolism, Membrane Proteins metabolism, Transformation, Bacterial, DNA, Single-Stranded genetics, DNA, Single-Stranded metabolism, Helicobacter pylori genetics, Helicobacter pylori metabolism
- Abstract
The ComFC protein is essential for natural transformation, a process that plays a major role in the spread of antibiotic resistance genes and virulence factors across bacteria. However, its role remains largely unknown. Here, we show that Helicobacter pylori ComFC is involved in DNA transport through the cell membrane, and is required for the handling of the single-stranded DNA once it is delivered into the cytoplasm. The crystal structure of ComFC includes a zinc-finger motif and a putative phosphoribosyl transferase domain, both necessary for the protein's in vivo activity. Furthermore, we show that ComFC is a membrane-associated protein with affinity for single-stranded DNA. Our results suggest that ComFC provides the link between the transport of the transforming DNA into the cytoplasm and its handling by the recombination machinery., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
13. Tumor resistance to radiotherapy is triggered by an ATM/TAK1-dependent-increased expression of the cellular prion protein.
- Author
-
Bernardino-Sgherri J, Siberchicot C, Auvré F, Busso D, Brocas C, El Masri G, Lioutsko A, Ferri F, Radicella JP, Romeo PH, and Bravard A
- Subjects
- Cell Line, Tumor, Humans, Neoplasms genetics, PrPC Proteins metabolism, Radiation Tolerance, Ataxia Telangiectasia Mutated Proteins metabolism, MAP Kinase Kinase Kinases metabolism, Neoplasms metabolism, Neoplasms radiotherapy, PrPC Proteins biosynthesis
- Abstract
In solid cancers, high expression of the cellular prion protein (PrPC) is associated with stemness, invasiveness, and resistance to chemotherapy, but the role of PrPC in tumor response to radiotherapy is unknown. Here, we show that, in neuroblastoma, breast, and colorectal cancer cell lines, PrPC expression is increased after ionizing radiation (IR) and that PrPC deficiency increases radiation sensitivity and decreases radiation-induced radioresistance in tumor cells. In neuroblastoma cells, IR activates ATM that triggers TAK1-dependent phosphorylation of JNK and subsequent activation of the AP-1 transcription factor that ultimately increases PRNP promoter transcriptional activity through an AP-1 binding site in the PRNP promoter. Importantly, we show that this ATM-TAK1-PrPC pathway mediated radioresistance is activated in all tumor cell lines studied and that pharmacological inhibition of TAK1 activity recapitulates the effects of PrPC deficiency. Altogether, these results unveil how tumor cells activate PRNP to acquire resistance to radiotherapy and might have implications for therapeutic targeting of solid tumors radioresistance.
- Published
- 2021
- Full Text
- View/download PDF
14. A peptide of a type I toxin-antitoxin system induces Helicobacter pylori morphological transformation from spiral shape to coccoids.
- Author
-
El Mortaji L, Tejada-Arranz A, Rifflet A, Boneca IG, Pehau-Arnaudet G, Radicella JP, Marsin S, and De Reuse H
- Subjects
- Adenosine Triphosphate metabolism, Cell Membrane drug effects, Cell Membrane metabolism, Helicobacter pylori ultrastructure, Hydrogen Peroxide toxicity, Intracellular Space metabolism, Kinetics, Membrane Potentials drug effects, Oxidative Stress drug effects, Peptidoglycan metabolism, Helicobacter pylori cytology, Helicobacter pylori drug effects, Peptides pharmacology, Toxin-Antitoxin Systems
- Abstract
Toxin-antitoxin systems are found in many bacterial chromosomes and plasmids with roles ranging from plasmid stabilization to biofilm formation and persistence. In these systems, the expression/activity of the toxin is counteracted by an antitoxin, which, in type I systems, is an antisense RNA. While the regulatory mechanisms of these systems are mostly well defined, the toxins' biological activity and expression conditions are less understood. Here, these questions were investigated for a type I toxin-antitoxin system (AapA1-IsoA1) expressed from the chromosome of the human pathogen Helicobacter pylori We show that expression of the AapA1 toxin in H. pylori causes growth arrest associated with rapid morphological transformation from spiral-shaped bacteria to round coccoid cells. Coccoids are observed in patients and during in vitro growth as a response to different stress conditions. The AapA1 toxin, first molecular effector of coccoids to be identified, targets H. pylori inner membrane without disrupting it, as visualized by cryoelectron microscopy. The peptidoglycan composition of coccoids is modified with respect to spiral bacteria. No major changes in membrane potential or adenosine 5'-triphosphate (ATP) concentration result from AapA1 expression, suggesting coccoid viability. Single-cell live microscopy tracking the shape conversion suggests a possible association of this process with cell elongation/division interference. Oxidative stress induces coccoid formation and is associated with repression of the antitoxin promoter and enhanced processing of its transcript, leading to an imbalance in favor of AapA1 toxin expression. Our data support the hypothesis of viable coccoids with characteristics of dormant bacteria that might be important in H. pylori infections refractory to treatment., Competing Interests: The authors declare no competing interest.
- Published
- 2020
- Full Text
- View/download PDF
15. Lost in the Crowd: How Does Human 8-Oxoguanine DNA Glycosylase 1 (OGG1) Find 8-Oxoguanine in the Genome?
- Author
-
D'Augustin O, Huet S, Campalans A, and Radicella JP
- Subjects
- Animals, DNA metabolism, DNA Glycosylases genetics, DNA Glycosylases physiology, DNA Repair physiology, DNA-Binding Proteins metabolism, Guanine metabolism, Guanine physiology, Humans, Oxidative Stress physiology, DNA Glycosylases metabolism, Guanine analogs & derivatives
- Abstract
The most frequent DNA lesion resulting from an oxidative stress is 7,8-dihydro-8-oxoguanine (8-oxoG). 8-oxoG is a premutagenic base modification due to its capacity to pair with adenine. Thus, the repair of 8-oxoG is critical for the preservation of the genetic information. Nowadays, 8-oxoG is also considered as an oxidative stress-sensor with a putative role in transcription regulation. In mammalian cells, the modified base is excised by the 8-oxoguanine DNA glycosylase (OGG1), initiating the base excision repair (BER) pathway. OGG1 confronts the massive challenge that is finding rare occurrences of 8-oxoG among a million-fold excess of normal guanines. Here, we review the current knowledge on the search and discrimination mechanisms employed by OGG1 to find its substrate in the genome. While there is considerable data from in vitro experiments, much less is known on how OGG1 is recruited to chromatin and scans the genome within the cellular nucleus. Based on what is known of the strategies used by proteins searching for rare genomic targets, we discuss the possible scenarios allowing the efficient detection of 8-oxoG by OGG1.
- Published
- 2020
- Full Text
- View/download PDF
16. Chromatin recruitment of OGG1 requires cohesin and mediator and is essential for efficient 8-oxoG removal.
- Author
-
Lebraud E, Pinna G, Siberchicot C, Depagne J, Busso D, Fantini D, Irbah L, Robeska E, Kratassiouk G, Ravanat JL, Epe B, Radicella JP, and Campalans A
- Subjects
- Cell Cycle Proteins genetics, Chromosomal Proteins, Non-Histone genetics, Euchromatin genetics, Genomic Instability genetics, Guanine metabolism, HeLa Cells, Humans, Oxidative Stress genetics, RNA, Small Interfering genetics, Transfection, Cohesins, Chromatin genetics, DNA Glycosylases genetics, DNA Repair genetics, Guanine analogs & derivatives
- Abstract
One of the most abundant DNA lesions induced by oxidative stress is the highly mutagenic 8-oxoguanine (8-oxoG), which is specifically recognized by 8-oxoguanine DNA glycosylase 1 (OGG1) to initiate its repair. How DNA glycosylases find small non-helix-distorting DNA lesions amongst millions of bases packaged in the chromatin-based architecture of the genome remains an open question. Here, we used a high-throughput siRNA screening to identify factors involved in the recognition of 8-oxoG by OGG1. We show that cohesin and mediator subunits are required for re-localization of OGG1 and other base excision repair factors to chromatin upon oxidative stress. The association of OGG1 with euchromatin is necessary for the removal of 8-oxoG. Mediator subunits CDK8 and MED12 bind to chromatin and interact with OGG1 in response to oxidative stress, suggesting they participate in the recruitment of the DNA glycosylase. The oxidative stress-induced association between the cohesin and mediator complexes and OGG1 reveals an unsuspected function of those complexes in the maintenance of genomic stability., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2020
- Full Text
- View/download PDF
17. MasterPATH: network analysis of functional genomics screening data.
- Author
-
Rubanova N, Pinna G, Kropp J, Campalans A, Radicella JP, Polesskaya A, Harel-Bellan A, and Morozova N
- Subjects
- Humans, Loss of Function Mutation, MicroRNAs genetics, MicroRNAs metabolism, Muscle Development genetics, Phenotype, Gene Regulatory Networks, Genomics methods, Protein Interaction Maps, Software, Transcriptome
- Abstract
Background: Functional genomics employs several experimental approaches to investigate gene functions. High-throughput techniques, such as loss-of-function screening and transcriptome profiling, allow to identify lists of genes potentially involved in biological processes of interest (so called hit list). Several computational methods exist to analyze and interpret such lists, the most widespread of which aim either at investigating of significantly enriched biological processes, or at extracting significantly represented subnetworks., Results: Here we propose a novel network analysis method and corresponding computational software that employs the shortest path approach and centrality measure to discover members of molecular pathways leading to the studied phenotype, based on functional genomics screening data. The method works on integrated interactomes that consist of both directed and undirected networks - HIPPIE, SIGNOR, SignaLink, TFactS, KEGG, TransmiR, miRTarBase. The method finds nodes and short simple paths with significant high centrality in subnetworks induced by the hit genes and by so-called final implementers - the genes that are involved in molecular events responsible for final phenotypic realization of the biological processes of interest. We present the application of the method to the data from miRNA loss-of-function screen and transcriptome profiling of terminal human muscle differentiation process and to the gene loss-of-function screen exploring the genes that regulates human oxidative DNA damage recognition. The analysis highlighted the possible role of several known myogenesis regulatory miRNAs (miR-1, miR-125b, miR-216a) and their targets (AR, NR3C1, ARRB1, ITSN1, VAV3, TDGF1), as well as linked two major regulatory molecules of skeletal myogenesis, MYOD and SMAD3, to their previously known muscle-related targets (TGFB1, CDC42, CTCF) and also to a number of proteins such as C-KIT that have not been previously studied in the context of muscle differentiation. The analysis also showed the role of the interaction between H3 and SETDB1 proteins for oxidative DNA damage recognition., Conclusion: The current work provides a systematic methodology to discover members of molecular pathways in integrated networks using functional genomics screening data. It also offers a valuable instrument to explain the appearance of a set of genes, previously not associated with the process of interest, in the hit list of each particular functional genomics screening.
- Published
- 2020
- Full Text
- View/download PDF
18. Prion protein deficiency impairs hematopoietic stem cell determination and sensitizes myeloid progenitors to irradiation.
- Author
-
Siberchicot C, Gault N, Déchamps N, Barroca V, Aguzzi A, Roméo PH, Radicella JP, Bravard A, and Bernardino-Sgherri J
- Subjects
- Hematopoietic Stem Cells, Humans, Myeloid Progenitor Cells, Prion Proteins genetics, Prions genetics, Protein Deficiency
- Abstract
Highly conserved among species and expressed in various types of cells, numerous roles have been attributed to the cellular prion protein (PrPC). In hematopoiesis, PrPC regulates hematopoietic stem cell self-renewal but the mechanisms involved in this regulation are unknown. Here we show that PrPC regulates hematopoietic stem cell number during aging and their determination towards myeloid progenitors. Furthermore, PrPC protects myeloid progenitors against the cytotoxic effects of total body irradiation. This radioprotective effect was associated with increased cellular prion mRNA level and with stimulation of the DNA repair activity of the Apurinic/pyrimidinic endonuclease 1, a key enzyme of the base excision repair pathway. Altogether, these results show a previously unappreciated role of PrPC in adult hematopoiesis, and indicate that PrPC-mediated stimulation of BER activity might protect hematopoietic progenitors from the cytotoxic effects of total body irradiation., (Copyright© 2020 Ferrata Storti Foundation.)
- Published
- 2020
- Full Text
- View/download PDF
19. Identification of the periplasmic DNA receptor for natural transformation of Helicobacter pylori.
- Author
-
Damke PP, Di Guilmi AM, Varela PF, Velours C, Marsin S, Veaute X, Machouri M, Gunjal GV, Rao DN, Charbonnier JB, and Radicella JP
- Subjects
- Bacterial Proteins genetics, Biological Transport, DNA genetics, DNA metabolism, DNA, Bacterial genetics, Helicobacter pylori genetics, Periplasm genetics, Receptors, Cell Surface genetics, Bacterial Proteins metabolism, DNA, Bacterial metabolism, Gene Transfer, Horizontal, Helicobacter pylori metabolism, Periplasm metabolism, Receptors, Cell Surface metabolism, Transformation, Bacterial
- Abstract
Horizontal gene transfer through natural transformation is a major driver of antibiotic resistance spreading in many pathogenic bacterial species. In the case of Gram-negative bacteria, and in particular of Helicobacter pylori, the mechanisms underlying the handling of the incoming DNA within the periplasm are poorly understood. Here we identify the protein ComH as the periplasmic receptor for the transforming DNA during natural transformation in H. pylori. ComH is a DNA-binding protein required for the import of DNA into the periplasm. Its C-terminal domain displays strong affinity for double-stranded DNA and is sufficient for the accumulation of DNA in the periplasm, but not for DNA internalisation into the cytoplasm. The N-terminal region of the protein allows the interaction of ComH with a periplasmic domain of the inner-membrane channel ComEC, which is known to mediate the translocation of DNA into the cytoplasm. Our results indicate that ComH is involved in the import of DNA into the periplasm and its delivery to the inner membrane translocator ComEC.
- Published
- 2019
- Full Text
- View/download PDF
20. Correction to: Mutations in the nucleotide binding and hydrolysis domains of helicobacter pylori MutS2 lead to altered biochemical activities and inactivation of its in vivo function.
- Author
-
Damke PP, Dhanaraju R, Marsin S, Radicella JP, and Rao DN
- Abstract
Following publication of the original article [1], the authors notified us of an error in the presentation of Fig. 6G.
- Published
- 2019
- Full Text
- View/download PDF
21. The C-terminal domain of HpDprA is a DNA-binding winged helix domain that does not bind double-stranded DNA.
- Author
-
Lisboa J, Celma L, Sanchez D, Marquis M, Andreani J, Guérois R, Ochsenbein F, Durand D, Marsin S, Cuniasse P, Radicella JP, and Quevillon-Cheruel S
- Subjects
- Bacterial Proteins metabolism, Binding Sites, Conserved Sequence, Crystallography, X-Ray, DNA chemistry, Helicobacter pylori chemistry, Membrane Proteins metabolism, Protein Binding, Protein Conformation, alpha-Helical, Bacterial Proteins chemistry, DNA metabolism, Membrane Proteins chemistry
- Abstract
DNA-processing protein A, a ubiquitous multidomain DNA-binding protein, plays a crucial role during natural transformation in bacteria. Here, we carried out the structural analysis of DprA from the human pathogen Helicobacter pylori by combining data issued from the 1.8-Å resolution X-ray structure of the Pfam02481 domain dimer (RF), the NMR structure of the carboxy terminal domain (CTD), and the low-resolution structure of the full-length DprA dimer obtained in solution by SAXS. In particular, we sought a molecular function for the CTD, a domain that we show here is essential for transformation in H. pylori. Albeit its structural homology to winged helix DNA-binding motifs, we confirmed that the isolated CTD does not interact with ssDNA nor with dsDNA. The key R52 and K137 residues of RF are crucial for these two interactions. Search for sequences harboring homology to either HpDprA or Rhodopseudomonas palustris DprA CTDs led to the identification of conserved patches in the two CTD. Our structural study revealed the similarity of the structures adopted by these residues in RpDprA CTD and HpDprA CTD. This argues for a conserved, but yet to be defined, CTD function, distinct from DNA binding., (© 2019 Federation of European Biochemical Societies.)
- Published
- 2019
- Full Text
- View/download PDF
22. Identification of an XRCC1 DNA binding activity essential for retention at sites of DNA damage.
- Author
-
Mok MCY, Campalans A, Pillon MC, Guarné A, Radicella JP, and Junop MS
- Subjects
- Animals, CHO Cells, Cricetulus, Escherichia coli, HeLa Cells, Humans, Protein Binding, DNA metabolism, DNA Breaks, Single-Stranded, DNA Repair, Protein Domains, X-ray Repair Cross Complementing Protein 1 chemistry, X-ray Repair Cross Complementing Protein 1 metabolism
- Abstract
Repair of two major forms of DNA damage, single strand breaks and base modifications, are dependent on XRCC1. XRCC1 orchestrates these repair processes by temporally and spatially coordinating interactions between several other repair proteins. Here we show that XRCC1 contains a central DNA binding domain (CDB, residues 219-415) encompassing its first BRCT domain. In contrast to the N-terminal domain of XRCC1, which has been reported to mediate damage sensing in vitro, we demonstrate that the DNA binding module identified here lacks binding specificity towards DNA containing nicks or gaps. Alanine substitution of residues within the CDB of XRCC1 disrupt DNA binding in vitro and lead to a significant reduction in XRCC1 retention at DNA damage sites without affecting initial recruitment. Interestingly, reduced retention at sites of DNA damage is associated with an increased rate of repair. These findings suggest that DNA binding activity of XRCC1 plays a significant role in retention at sites of damage and the rate at which damage is repaired.
- Published
- 2019
- Full Text
- View/download PDF
23. The transcription-coupled DNA repair-initiating protein CSB promotes XRCC1 recruitment to oxidative DNA damage.
- Author
-
Menoni H, Wienholz F, Theil AF, Janssens RC, Lans H, Campalans A, Radicella JP, Marteijn JA, and Vermeulen W
- Subjects
- Cell Line, DNA metabolism, DNA Helicases metabolism, DNA Repair Enzymes metabolism, HEK293 Cells, Humans, Models, Genetic, Oxidation-Reduction, Oxidative Stress, Poly-ADP-Ribose Binding Proteins metabolism, X-ray Repair Cross Complementing Protein 1 metabolism, DNA genetics, DNA Damage, DNA Helicases genetics, DNA Repair, DNA Repair Enzymes genetics, Poly-ADP-Ribose Binding Proteins genetics, Transcription, Genetic, X-ray Repair Cross Complementing Protein 1 genetics
- Abstract
Transcription-coupled nucleotide excision repair factor Cockayne syndrome protein B (CSB) was suggested to function in the repair of oxidative DNA damage. However thus far, no clear role for CSB in base excision repair (BER), the dedicated pathway to remove abundant oxidative DNA damage, could be established. Using live cell imaging with a laser-assisted procedure to locally induce 8-oxo-7,8-dihydroguanine (8-oxoG) lesions, we previously showed that CSB is recruited to these lesions in a transcription-dependent but NER-independent fashion. Here we showed that recruitment of the preferred 8-oxoG-glycosylase 1 (OGG1) is independent of CSB or active transcription. In contrast, recruitment of the BER-scaffolding protein, X-ray repair cross-complementing protein 1 (XRCC1), to 8-oxoG lesions is stimulated by CSB and transcription. Remarkably, recruitment of XRCC1 to BER-unrelated single strand breaks (SSBs) does not require CSB or transcription. Together, our results suggest a specific transcription-dependent role for CSB in recruiting XRCC1 to BER-generated SSBs, whereas XRCC1 recruitment to SSBs generated independently of BER relies predominantly on PARP activation. Based on our results, we propose a model in which CSB plays a role in facilitating BER progression at transcribed genes, probably to allow XRCC1 recruitment to BER-intermediates masked by RNA polymerase II complexes stalled at these intermediates.
- Published
- 2018
- Full Text
- View/download PDF
24. Mitochondrial maintenance under oxidative stress depends on mitochondrially localised α-OGG1.
- Author
-
Lia D, Reyes A, de Melo Campos JTA, Piolot T, Baijer J, Radicella JP, and Campalans A
- Subjects
- Cell Cycle physiology, Cell Line, Tumor, DNA Glycosylases genetics, DNA, Mitochondrial metabolism, Guanine analogs & derivatives, Guanine metabolism, HEK293 Cells, Humans, Mitochondria enzymology, Mitochondrial Membranes enzymology, Mitochondrial Membranes metabolism, RNA, Small Interfering administration & dosage, RNA, Small Interfering genetics, Reactive Oxygen Species metabolism, Transfection, DNA Glycosylases metabolism, Mitochondria metabolism, Oxidative Stress physiology
- Abstract
Accumulation of 8-oxoguanine (8-oxoG) in mitochondrial DNA and mitochondrial dysfunction have been observed in cells deficient for the DNA glycosylase OGG1 when exposed to oxidative stress. In human cells, up to eight mRNAs for OGG1 can be generated by alternative splicing and it is still unclear which of them codes for the protein that ensures the repair of 8-oxoG in mitochondria. Here, we show that the α-OGG1 isoform, considered up to now to be exclusively nuclear, has a functional mitochondrial-targeting sequence and is imported into mitochondria. We analyse the sub-mitochondrial localisation of α-OGG1 with unprecedented resolution and show that this DNA glycosylase is associated with DNA in mitochondrial nucleoids. We show that the presence of α-OGG1 inside mitochondria and its enzymatic activity are required to preserve the mitochondrial network in cells exposed to oxidative stress. Altogether, these results unveil a new role of α-OGG1 in the mitochondria and indicate that the same isoform ensures the repair of 8-oxoG in both nuclear and mitochondrial genomes. The activity of α-OGG1 in mitochondria is sufficient for the recovery of organelle function after oxidative stress., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2018. Published by The Company of Biologists Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
25. Structural basis for the substrate selectivity of Helicobacter pylori NucT nuclease activity.
- Author
-
Celma L, Corbinais C, Vercruyssen J, Veaute X, de la Sierra-Gallay IL, Guérois R, Busso D, Mathieu A, Marsin S, Quevillon-Cheruel S, and Radicella JP
- Subjects
- Amino Acid Sequence, Crystallography, X-Ray, Endonucleases chemistry, Protein Conformation, Sequence Homology, Amino Acid, Substrate Specificity, Endonucleases metabolism, Helicobacter pylori enzymology
- Abstract
The Phospholipase D (PLD) superfamily of proteins includes a group of enzymes with nuclease activity on various nucleic acid substrates. Here, with the aim of better understanding the substrate specificity determinants in this subfamily, we have characterised the enzymatic activity and the crystal structure of NucT, a nuclease implicated in Helicobacter pylori purine salvage and natural transformation and compared them to those of its bacterial and mammalian homologues. NucT exhibits an endonuclease activity with a strong preference for single stranded nucleic acids substrates. We identified histidine124 as essential for the catalytic activity of the protein. Comparison of the NucT crystal structure at 1.58 Å resolution reported here with those of other members of the sub-family suggests that the specificity of NucT for single-stranded nucleic acids is provided by the width of a positively charged groove giving access to the catalytic site.
- Published
- 2017
- Full Text
- View/download PDF
26. Incorporating interaction networks into the determination of functionally related hit genes in genomic experiments with Markov random fields.
- Author
-
Robinson S, Nevalainen J, Pinna G, Campalans A, Radicella JP, and Guyon L
- Subjects
- Algorithms, Humans, Lymphoma genetics, Lymphoma metabolism, Gene Regulatory Networks, Genomics methods, Signal Transduction
- Abstract
Motivation: Incorporating gene interaction data into the identification of 'hit' genes in genomic experiments is a well-established approach leveraging the 'guilt by association' assumption to obtain a network based hit list of functionally related genes. We aim to develop a method to allow for multivariate gene scores and multiple hit labels in order to extend the analysis of genomic screening data within such an approach., Results: We propose a Markov random field-based method to achieve our aim and show that the particular advantages of our method compared with those currently used lead to new insights in previously analysed data as well as for our own motivating data. Our method additionally achieves the best performance in an independent simulation experiment. The real data applications we consider comprise of a survival analysis and differential expression experiment and a cell-based RNA interference functional screen., Availability and Implementation: We provide all of the data and code related to the results in the paper., Contact: sean.j.robinson@utu.fi or laurent.guyon@cea.fr., Supplementary Information: Supplementary data are available at Bioinformatics online., (© The Author 2017. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com)
- Published
- 2017
- Full Text
- View/download PDF
27. ComB proteins expression levels determine Helicobacter pylori competence capacity.
- Author
-
Corbinais C, Mathieu A, Damke PP, Kortulewski T, Busso D, Prado-Acosta M, Radicella JP, and Marsin S
- Subjects
- Bacterial Proteins genetics, DNA Damage, DNA Repair, DNA, Bacterial genetics, Helicobacter pylori metabolism, Mutation genetics, Operon genetics, Transformation, Genetic, Bacterial Proteins metabolism, Gene Expression Regulation, Bacterial, Helicobacter pylori genetics
- Abstract
Helicobacter pylori chronically colonises half of the world's human population and is the main cause of ulcers and gastric cancers. Its prevalence and the increase in antibiotic resistance observed recently reflect the high genetic adaptability of this pathogen. Together with high mutation rates and an efficient DNA recombination system, horizontal gene transfer through natural competence makes of H. pylori one of the most genetically diverse bacteria. We show here that transformation capacity is enhanced in strains defective for recN, extending previous work with other homologous recombination genes. However, inactivation of either mutY or polA has no effect on DNA transformation, suggesting that natural competence can be boosted in H. pylori by the persistence of DNA breaks but not by enhanced mutagenesis. The transformation efficiency of the different DNA repair impaired strains correlates with the number of transforming DNA foci formed on the cell surface and with the expression of comB8 and comB10 competence genes. Overexpression of the comB6-B10 operon is sufficient to increase the transformation capacity of a wild type strain, indicating that the ComB complex, present in the bacterial wall and essential for DNA uptake, can be a limiting factor for transformation efficiency., Competing Interests: The authors declare no competing financial interests.
- Published
- 2017
- Full Text
- View/download PDF
28. Following transforming DNA in Helicobacter pylori from uptake to expression.
- Author
-
Corbinais C, Mathieu A, Kortulewski T, Radicella JP, and Marsin S
- Subjects
- DNA, Bacterial metabolism, Gene Expression, Gene Transfer, Horizontal, Helicobacter pylori metabolism, DNA, Bacterial genetics, Helicobacter pylori genetics, Transformation, Bacterial genetics
- Abstract
Natural transformation is a potent driver for genetic diversification in bacterial populations. It involves exogenous DNA binding, uptake, transport and internalization into the cytoplasm, where DNA can be processed and integrated into the host chromosome. Direct visualisation of transforming DNA (tDNA) has been limited to its binding to the surface or, in the case of Gram-negative species, to its entrance into the periplasm. We present here for the first time the direct visualisation of tDNA entering the bacterial cytoplasm. We used as a model the Gram-negative pathogen Helicobacter pylori, characterised by a large intraspecies variability that results from high mutation rates and efficient horizontal gene transfer. Using fluorescently labelled DNA, we followed for up to 3 h the fate of tDNA foci formed in the periplasm and eventually internalised into the cytoplasm. By tracking at the single cell level the expression of a fluorescent protein coded by the tDNA, we show that up to 50% of the cells express the transforming phenotype. The overall transformation process in H. pylori, from tDNA uptake to expression of the recombinant gene, can take place in less than 1 h, without requiring a growth arrest, and prior to the replication of the chromosome., (© 2016 John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
29. A threshold of endogenous stress is required to engage cellular response to protect against mutagenesis.
- Author
-
Saintigny Y, Chevalier F, Bravard A, Dardillac E, Laurent D, Hem S, Dépagne J, Radicella JP, and Lopez BS
- Subjects
- Animals, CHO Cells, Cricetulus, Proteomics, Thymidine metabolism, DNA Damage, Genomic Instability genetics, Mutagenesis, Oxidative Stress
- Abstract
Endogenous stress represents a major source of genome instability, but is in essence difficult to apprehend. Incorporation of labeled radionuclides into DNA constitutes a tractable model to analyze cellular responses to endogenous attacks. Here we show that incorporation of [(3)H]thymidine into CHO cells generates oxidative-induced mutagenesis, but, with a peak at low doses. Proteomic analysis showed that the cellular response differs between low and high levels of endogenous stress. In particular, these results confirmed the involvement of proteins implicated in redox homeostasis and DNA damage signaling pathways. Induced-mutagenesis was abolished by the anti-oxidant N-acetyl cysteine and plateaued, at high doses, upon exposure to L-buthionine sulfoximine, which represses cellular detoxification. The [(3)H]thymidine-induced mutation spectrum revealed mostly base substitutions, exhibiting a signature specific for low doses (GC > CG and AT > CG). Consistently, the enzymatic activity of the base excision repair protein APE-1 is induced at only medium or high doses. Collectively, the data reveal that a threshold of endogenous stress must be reached to trigger cellular detoxification and DNA repair programs; below this threshold, the consequences of endogenous stress escape cellular surveillance, leading to high levels of mutagenesis. Therefore, low doses of endogenous local stress can jeopardize genome integrity more efficiently than higher doses.
- Published
- 2016
- Full Text
- View/download PDF
30. Mutations in the nucleotide binding and hydrolysis domains of Helicobacter pylori MutS2 lead to altered biochemical activities and inactivation of its in vivo function.
- Author
-
Damke PP, Dhanaraju R, Marsin S, Radicella JP, and Rao DN
- Subjects
- Bacterial Proteins genetics, Helicobacter pylori chemistry, Helicobacter pylori genetics, Hydrolysis, Kinetics, MutS DNA Mismatch-Binding Protein genetics, Mutation, Protein Binding, Protein Structure, Tertiary, Recombination, Genetic, Adenosine Triphosphate metabolism, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Helicobacter pylori enzymology, MutS DNA Mismatch-Binding Protein chemistry, MutS DNA Mismatch-Binding Protein metabolism
- Abstract
Background: Helicobacter pylori MutS2 (HpMutS2), an inhibitor of recombination during transformation is a non-specific nuclease with two catalytic sites, both of which are essential for its anti-recombinase activity. Although HpMutS2 belongs to a highly conserved family of ABC transporter ATPases, the role of its ATP binding and hydrolysis activities remains elusive., Results: To explore the putative role of ATP binding and hydrolysis activities of HpMutS2 we specifically generated point mutations in the nucleotide-binding Walker-A (HpMutS2-G338R) and hydrolysis Walker-B (HpMutS2-E413A) domains of the protein. Compared to wild-type protein, HpMutS2-G338R exhibited ~2.5-fold lower affinity for both ATP and ADP while ATP hydrolysis was reduced by ~3-fold. Nucleotide binding efficiencies of HpMutS2-E413A were not significantly altered; however the ATP hydrolysis was reduced by ~10-fold. Although mutations in the Walker-A and Walker-B motifs of HpMutS2 only partially reduced its ability to bind and hydrolyze ATP, we demonstrate that these mutants not only exhibited alterations in the conformation, DNA binding and nuclease activities of the protein but failed to complement the hyper-recombinant phenotype displayed by mutS2-disrupted strain of H. pylori. In addition, we show that the nucleotide cofactor modulates the conformation, DNA binding and nuclease activities of HpMutS2., Conclusions: These data describe a strong crosstalk between the ATPase, DNA binding, and nuclease activities of HpMutS2. Furthermore these data show that both, ATP binding and hydrolysis activities of HpMutS2 are essential for the in vivo anti-recombinase function of the protein.
- Published
- 2016
- Full Text
- View/download PDF
31. XPC deficiency is related to APE1 and OGG1 expression and function.
- Author
-
de Melo JT, de Souza Timoteo AR, Lajus TB, Brandão JA, de Souza-Pinto NC, Menck CF, Campalans A, Radicella JP, Vessoni AT, Muotri AR, and Agnez-Lima LF
- Subjects
- Cells, Cultured, DNA Glycosylases genetics, DNA Repair physiology, DNA-(Apurinic or Apyrimidinic Site) Lyase genetics, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, Fibroblasts drug effects, Fibroblasts pathology, Gene Expression Regulation, Humans, Hydrogen Peroxide pharmacology, Immunoprecipitation, Oxidants pharmacology, Oxidative Stress, RNA, Messenger metabolism, Xeroderma Pigmentosum genetics, Xeroderma Pigmentosum pathology, DNA Glycosylases metabolism, DNA-(Apurinic or Apyrimidinic Site) Lyase metabolism, DNA-Binding Proteins metabolism
- Abstract
Oxidative DNA damage is considered to be a major cause of neurodegeneration and internal tumors observed in syndromes that result from nucleotide excision repair (NER) deficiencies, such as Xeroderma Pigmentosum (XP) and Cockayne Syndrome (CS). Recent evidence has shown that NER aids in removing oxidized DNA damage and may interact with base excision repair (BER) enzymes. Here, we investigated APE1 and OGG1 expression, localization and activity after oxidative stress in XPC-deficient cells. The endogenous APE1 and OGG1 mRNA levels were lower in XPC-deficient fibroblasts. However, XPC-deficient cells did not show hypersensitivity to oxidative stress compared with NER-proficient cells. To confirm the impact of an XPC deficiency in regulating APE1 and OGG1 expression and activity, we established an XPC-complemented cell line. Although the XPC complementation was only partial and transient, the transfected cells exhibited greater OGG1 expression and activity compared with XPC-deficient cells. However, the APE1 expression and activity did not significantly change. Furthermore, we observed a physical interaction between the XPC and APE1 proteins. Together, the results indicate that the responses of XPC-deficient cells under oxidative stress may not only be associated with NER deficiency per se but may also include new XPC functions in regulating BER proteins., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
32. Φ-score: A cell-to-cell phenotypic scoring method for sensitive and selective hit discovery in cell-based assays.
- Author
-
Guyon L, Lajaunie C, Fer F, Bhajun R, Sulpice E, Pinna G, Campalans A, Radicella JP, Rouillier P, Mary M, Combe S, Obeid P, Vert JP, and Gidrol X
- Abstract
Phenotypic screening monitors phenotypic changes induced by perturbations, including those generated by drugs or RNA interference. Currently-used methods for scoring screen hits have proven to be problematic, particularly when applied to physiologically relevant conditions such as low cell numbers or inefficient transfection. Here, we describe the Φ-score, which is a novel scoring method for the identification of phenotypic modifiers or hits in cell-based screens. Φ-score performance was assessed with simulations, a validation experiment and its application to gene identification in a large-scale RNAi screen. Using robust statistics and a variance model, we demonstrated that the Φ-score showed better sensitivity, selectivity and reproducibility compared to classical approaches. The improved performance of the Φ-score paves the way for cell-based screening of primary cells, which are often difficult to obtain from patients in sufficient numbers. We also describe a dedicated merging procedure to pool scores from small interfering RNAs targeting the same gene so as to provide improved visualization and hit selection.
- Published
- 2015
- Full Text
- View/download PDF
33. The nuclease activities of both the Smr domain and an additional LDLK motif are required for an efficient anti-recombination function of Helicobacter pylori MutS2.
- Author
-
Damke PP, Dhanaraju R, Marsin S, Radicella JP, and Rao DN
- Subjects
- Amino Acid Motifs, Bacterial Proteins genetics, Bacterial Proteins metabolism, DNA Repair, Deoxyribonucleases genetics, Deoxyribonucleases metabolism, Humans, Molecular Sequence Data, MutS Homolog 2 Protein metabolism, Protein Structure, Tertiary, Sequence Analysis, Protein, Helicobacter pylori enzymology, Helicobacter pylori genetics, MutS Homolog 2 Protein genetics, Recombination, Genetic
- Abstract
Helicobacter pylori, a human pathogen, is a naturally and constitutively competent bacteria, displaying a high rate of intergenomic recombination. While recombination events are essential for evolution and adaptation of H. pylori to dynamic gastric niches and new hosts, such events should be regulated tightly to maintain genomic integrity. Here, we analyze the role of the nuclease activity of MutS2, a protein that limits recombination during transformation in H. pylori. In previously studied MutS2 proteins, the C-terminal Smr domain was mapped as the region responsible for its nuclease activity. We report here that deletion of Smr domain does not completely abolish the nuclease activity of HpMutS2. Using bioinformatics analysis and mutagenesis, we identified an additional and novel nuclease motif (LDLK) at the N-terminus of HpMutS2 unique to Helicobacter and related ε-proteobacterial species. A single point mutation (D30A) in the LDLK motif and the deletion of Smr domain resulted in ∼ 5-10-fold loss of DNA cleavage ability of HpMutS2. Interestingly, the mutant forms of HpMutS2 wherein the LDLK motif was mutated or the Smr domain was deleted were unable to complement the hyper-recombination phenotype of a mutS2(-) strain, suggesting that both nuclease sites are indispensable for an efficient anti-recombinase activity of HpMutS2., (© 2015 John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
34. Interaction with OGG1 is required for efficient recruitment of XRCC1 to base excision repair and maintenance of genetic stability after exposure to oxidative stress.
- Author
-
Campalans A, Moritz E, Kortulewski T, Biard D, Epe B, and Radicella JP
- Subjects
- Animals, CHO Cells, Cell Line, Cricetulus, DNA Glycosylases analysis, DNA-Binding Proteins analysis, Gene Deletion, Humans, Protein Interaction Maps, X-ray Repair Cross Complementing Protein 1, DNA Glycosylases metabolism, DNA Repair, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Oxidative Stress, Polymorphism, Single Nucleotide
- Abstract
XRCC1 is an essential protein required for the maintenance of genomic stability through its implication in DNA repair. The main function of XRCC1 is associated with its role in the single-strand break (SSB) and base excision repair (BER) pathways that share several enzymatic steps. We show here that the polymorphic XRCC1 variant R194W presents a defect in its interaction with the DNA glycosylase OGG1 after oxidative stress. While proficient for single-strand break repair (SSBR), this variant does not colocalize with OGG1, reflecting a defect in its involvement in BER. Consistent with a role of XRCC1 in the coordination of the BER pathway, induction of oxidative base damage in XRCC1-deficient cells complemented with the R194W variant results in increased genetic instability as revealed by the accumulation of micronuclei. These data identify a specific molecular role for the XRCC1-OGG1 interaction in BER and provide a model for the effects of the R194W variant identified in molecular cancer epidemiology studies., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Nuclear translocation of p19INK4d in response to oxidative DNA damage promotes chromatin relaxation.
- Author
-
Sonzogni SV, Ogara MF, Castillo DS, Sirkin PF, Radicella JP, and Cánepa ET
- Subjects
- Active Transport, Cell Nucleus, Animals, Blotting, Northern, Blotting, Western, Cell Line, Cell Nucleus metabolism, Chromatin genetics, Cyclin-Dependent Kinase Inhibitor p19 genetics, DNA Repair, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, HEK293 Cells, HeLa Cells, Humans, Microscopy, Confocal, Protein Binding, Chromatin metabolism, Cyclin-Dependent Kinase Inhibitor p19 metabolism, DNA Damage, Oxidative Stress
- Abstract
DNA is continuously exposed to damaging agents that can lead to changes in the genetic information with adverse consequences. Nonetheless, eukaryotic cells have mechanisms such as the DNA damage response (DDR) to prevent genomic instability. The DNA of eukaryotic cells is packaged into nucleosomes, which fold the genome into highly condensed chromatin, but relatively little is known about the role of chromatin accessibility in DNA repair. p19INK4d, a cyclin-dependent kinase inhibitor, plays an important role in cell cycle regulation and cellular DDR. Extensive data indicate that p19INK4d is a critical factor in the maintenance of genomic integrity and cell survival. p19INK4d is upregulated by various genotoxics, improving the repair efficiency for a variety of DNA lesions. The evidence of p19INK4d translocation into the nucleus and its low sequence specificity in its interaction with DNA prompted us to hypothesize that p19INK4d plays a role at an early stage of cellular DDR. In the present study, we demonstrate that upon oxidative DNA damage, p19INK4d strongly binds to and relaxes chromatin. Furthermore, in vitro accessibility assays show that DNA is more accessible to a restriction enzyme when a chromatinized plasmid is incubated in the presence of a protein extract with high levels of p19INK4d. Nuclear protein extracts from cells overexpressing p19INK4d are better able to repair a chromatinized and damaged plasmid. These observations support the notion that p19INK4d would act as a chromatin accessibility factor that allows the access of the repair machinery to the DNA damage site.
- Published
- 2015
- Full Text
- View/download PDF
36. E2F1 and E2F2 induction in response to DNA damage preserves genomic stability in neuronal cells.
- Author
-
Castillo DS, Campalans A, Belluscio LM, Carcagno AL, Radicella JP, Cánepa ET, and Pregi N
- Subjects
- Cell Line, Tumor, Cell Survival drug effects, Cell Survival radiation effects, Cycloheximide toxicity, DNA Repair drug effects, Dactinomycin toxicity, E2F1 Transcription Factor genetics, E2F2 Transcription Factor genetics, HEK293 Cells, Histones metabolism, Humans, Hydrogen Peroxide toxicity, MAP Kinase Kinase Kinases metabolism, Neurons cytology, Neurons metabolism, Protein Synthesis Inhibitors toxicity, Rad51 Recombinase metabolism, Ultraviolet Rays, Up-Regulation drug effects, p300-CBP Transcription Factors metabolism, DNA Damage drug effects, DNA Damage radiation effects, E2F1 Transcription Factor metabolism, E2F2 Transcription Factor metabolism, Genomic Instability drug effects, Genomic Instability radiation effects
- Abstract
E2F transcription factors regulate a wide range of biological processes, including the cellular response to DNA damage. In the present study, we examined whether E2F family members are transcriptionally induced following treatment with several genotoxic agents, and have a role on the cell DNA damage response. We show a novel mechanism, conserved among diverse species, in which E2F1 and E2F2, the latter specifically in neuronal cells, are transcriptionally induced after DNA damage. This upregulation leads to increased E2F1 and E2F2 protein levels as a consequence of de novo protein synthesis. Ectopic expression of these E2Fs in neuronal cells reduces the level of DNA damage following genotoxic treatment, while ablation of E2F1 and E2F2 leads to the accumulation of DNA lesions and increased apoptotic response. Cell viability and DNA repair capability in response to DNA damage induction are also reduced by the E2F1 and E2F2 deficiencies. Finally, E2F1 and E2F2 accumulate at sites of oxidative and UV-induced DNA damage, and interact with γH2AX DNA repair factor. As previously reported for E2F1, E2F2 promotes Rad51 foci formation, interacts with GCN5 acetyltransferase and induces histone acetylation following genotoxic insult. The results presented here unveil a new mechanism involving E2F1 and E2F2 in the maintenance of genomic stability in response to DNA damage in neuronal cells.
- Published
- 2015
- Full Text
- View/download PDF
37. The prion protein is critical for DNA repair and cell survival after genotoxic stress.
- Author
-
Bravard A, Auvré F, Fantini D, Bernardino-Sgherri J, Sissoëff L, Daynac M, Xu Z, Etienne O, Dehen C, Comoy E, Boussin FD, Tell G, Deslys JP, and Radicella JP
- Subjects
- Animals, Brain enzymology, Cell Line, Cell Nucleus chemistry, Cell Survival, DNA-(Apurinic or Apyrimidinic Site) Lyase metabolism, Humans, Methyl Methanesulfonate toxicity, Mice, Mice, Inbred C57BL, Mutagens toxicity, Neurons drug effects, Neurons metabolism, Prion Proteins, Prions analysis, Prions biosynthesis, Prions genetics, Transcriptional Activation, DNA Repair, Prions metabolism
- Abstract
The prion protein (PrP) is highly conserved and ubiquitously expressed, suggesting that it plays an important physiological function. However, despite decades of investigation, this role remains elusive. Here, by using animal and cellular models, we unveil a key role of PrP in the DNA damage response. Exposure of neurons to a genotoxic stress activates PRNP transcription leading to an increased amount of PrP in the nucleus where it interacts with APE1, the major mammalian endonuclease essential for base excision repair, and stimulates its activity. Preventing the induction of PRNP results in accumulation of abasic sites in DNA and impairs cell survival after genotoxic treatment. Brains from Prnp(-/-) mice display a reduced APE1 activity and a defect in the repair of induced DNA damage in vivo. Thus, PrP is required to maintain genomic stability in response to genotoxic stresses., (© The Author(s) 2014. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2015
- Full Text
- View/download PDF
38. hOGG1-Cys326 variant cells are hypersensitive to DNA repair inhibition by nitric oxide.
- Author
-
Moritz E, Pauly K, Bravard A, Hall J, Radicella JP, and Epe B
- Subjects
- Cell Line, DNA Damage drug effects, DNA Glycosylases metabolism, Genomic Instability, Humans, Hyperthermia, Induced, Nitric Oxide pharmacology, Oxidative Stress genetics, Polymorphism, Genetic, Protein Stability, Reactive Oxygen Species metabolism, DNA Glycosylases genetics, DNA Repair drug effects, Genetic Variation, Nitric Oxide metabolism
- Abstract
The repair of 8-oxo-7,8-dihydroguanine in the DNA of mammalian cells is initiated by 8-oxoguanine DNA glycosylase (OGG1). A frequent polymorphism in the human OGG1 gene, rs1052133, causes the substitution of serine by cysteine at amino acid 326 of the protein and has been associated with an altered risk for various types of cancer in some populations. The OGG1-Cys326 protein appears to have normal enzymatic activity, but greater sensitivity to oxidation than the serine variant. Here, we describe a comparison of the cellular repair by the two OGG1 variants under stress conditions characteristic of inflammation, namely in cells pretreated with nitric oxide (NO) or pre-exposed to hyperthermia. The results show that NO at concentrations causing negligible DNA damage and little cytotoxicity strongly reduces the repair rates of oxidized purines in the DNA of HeLa cells overexpressing the OGG1-Cys326 variant. The reduction in repair was much less pronounced in isogenic cells overexpressing the OGG1-Ser326 variant. Similar results were observed in EBV-transformed lymphocytes from donors homozygous for the two OGG1 variant alleles. In contrast, hyperthermia-induced stress caused a repair retardation that was independent of the OGG1 polymorphism. The repair inhibition by NO in the variant cells gave rise to increased genetic instability, measured as increased micronuclei formation after oxidant exposure. The results could explain a higher risk of malignant transformation in inflamed tissues of carriers of this variant allele., (© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2014
- Full Text
- View/download PDF
39. Replication fork collapse is a major cause of the high mutation frequency at three-base lesion clusters.
- Author
-
Sedletska Y, Radicella JP, and Sage E
- Subjects
- Cell Line, Transformed, DNA Breaks, Double-Stranded, DNA Replication, Deoxyribonuclease (Pyrimidine Dimer) metabolism, Escherichia coli Proteins metabolism, Guanine analogs & derivatives, Guanine metabolism, Humans, Uracil analogs & derivatives, Uracil metabolism, DNA Damage, DNA Repair, Mutagenesis, Mutation Rate
- Abstract
Unresolved repair of clustered DNA lesions can lead to the formation of deleterious double strand breaks (DSB) or to mutation induction. Here, we investigated the outcome of clusters composed of base lesions for which base excision repair enzymes have different kinetics of excision/incision. We designed multiply damaged sites (MDS) composed of a rapidly excised uracil (U) and two oxidized bases, 5-hydroxyuracil (hU) and 8-oxoguanine (oG), excised more slowly. Plasmids harboring these U-oG/hU MDS-carrying duplexes were introduced into Escherichia coli cells either wild type or deficient for DNA n-glycosylases. Induction of DSB was estimated from plasmid survival and mutagenesis determined by sequencing of surviving clones. We show that a large majority of MDS is converted to DSB, whereas almost all surviving clones are mutated at hU. We demonstrate that mutagenesis at hU is correlated with excision of the U placed on the opposite strand. We propose that excision of U by Ung initiates the loss of U-oG-carrying strand, resulting in enhanced mutagenesis at the lesion present on the opposite strand. Our results highlight the importance of the kinetics of excision by base excision repair DNA n-glycosylases in the processing and fate of MDS and provide evidence for the role of strand loss/replication fork collapse during the processing of MDS on their mutational consequences.
- Published
- 2013
- Full Text
- View/download PDF
40. Rapid inactivation and proteasome-mediated degradation of OGG1 contribute to the synergistic effect of hyperthermia on genotoxic treatments.
- Author
-
Fantini D, Moritz E, Auvré F, Amouroux R, Campalans A, Epe B, Bravard A, and Radicella JP
- Subjects
- Cell Nucleus metabolism, Cell Proliferation, DNA Glycosylases genetics, DNA Repair, Enzyme Stability, HeLa Cells, Humans, Photosensitizing Agents pharmacology, Proteasome Endopeptidase Complex, Protein Transport, Pyrrolidines pharmacology, Quinolizines pharmacology, Ubiquitin-Protein Ligases metabolism, Ubiquitination, DNA Damage, DNA Glycosylases metabolism, Heat-Shock Response, Proteolysis
- Abstract
Inhibition of DNA repair has been proposed as a mechanism underlying heat-induced sensitization of tumour cells to some anticancer treatments. Base excision repair (BER) constitutes the main pathway for the repair of DNA lesions induced by oxidizing or alkylating agents. Here, we report that mild hyperthermia, without toxic consequences per se, affects cellular DNA glycosylase activities, thus impairing BER. Exposure of cells to mild hyperthermia leads to a rapid and selective inactivation of OGG1 (8-oxoguanine DNA glycosylase) associated with the relocalisation of the protein into a detergent-resistant cellular fraction. Following its inactivation, OGG1 is ubiquitinated and directed to proteasome-mediated degradation, through a CHIP (C-terminus of HSC70-interacting protein) E3 ligase-mediated process. Moreover, the residual OGG1 accumulates in the perinuclear region leading to further depletion from the nucleus. As a consequence, HeLa cells subjected to hyperthermia and exposed to a genotoxic treatment have a reduced capacity to repair OGG1 cognate base lesions and an enhanced cell growth defect. The partial alleviation of this response by OGG1 overexpression indicates that heat-induced glycosylase inactivation contributes to the synergistic effect of hyperthermia on genotoxic treatments. Taken together, our results suggest that OGG1 inhibition contributes to heat-induced chemosensitisation of cells and could lay the basis for new anticancer therapeutic protocols that include hyperthermia., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
41. Distinct spatiotemporal patterns and PARP dependence of XRCC1 recruitment to single-strand break and base excision repair.
- Author
-
Campalans A, Kortulewski T, Amouroux R, Menoni H, Vermeulen W, and Radicella JP
- Subjects
- Animals, Cell Line, Cell Nucleus metabolism, DNA Glycosylases metabolism, DNA-Binding Proteins chemistry, Euchromatin genetics, Euchromatin metabolism, Heterochromatin genetics, Heterochromatin metabolism, Humans, Mice, Oxidation-Reduction, Poly (ADP-Ribose) Polymerase-1, Poly(ADP-ribose) Polymerases physiology, Protein Binding, Protein Structure, Tertiary, Protein Transport, Single-Cell Analysis, X-ray Repair Cross Complementing Protein 1, DNA Breaks, Single-Stranded, DNA Repair, DNA-Binding Proteins metabolism, Poly(ADP-ribose) Polymerases metabolism
- Abstract
Single-strand break repair (SSBR) and base excision repair (BER) of modified bases and abasic sites share several players. Among them is XRCC1, an essential scaffold protein with no enzymatic activity, required for the coordination of both pathways. XRCC1 is recruited to SSBR by PARP-1, responsible for the initial recognition of the break. The recruitment of XRCC1 to BER is still poorly understood. Here we show by using both local and global induction of oxidative DNA base damage that XRCC1 participation in BER complexes can be distinguished from that in SSBR by several criteria. We show first that XRCC1 recruitment to BER is independent of PARP. Second, unlike SSBR complexes that are assembled within minutes after global damage induction, XRCC1 is detected later in BER patches, with kinetics consistent with the repair of oxidized bases. Third, while XRCC1-containing foci associated with SSBR are formed both in eu- and heterochromatin domains, BER complexes are assembled in patches that are essentially excluded from heterochromatin and where the oxidized bases are detected.
- Published
- 2013
- Full Text
- View/download PDF
42. Biochemical and cellular characterization of Helicobacter pylori RecA, a protein with high-level constitutive expression.
- Author
-
Orillard E, Radicella JP, and Marsin S
- Subjects
- Animals, Bacterial Proteins genetics, DNA Damage, DNA Repair, Gamma Rays, Gene Dosage, Gene Expression Regulation, Enzymologic radiation effects, Helicobacter pylori genetics, Helicobacter pylori growth & development, Helicobacter pylori radiation effects, Humans, Protein Transport radiation effects, Rec A Recombinases genetics, Ultraviolet Rays, Bacterial Proteins metabolism, Gene Expression Regulation, Bacterial radiation effects, Helicobacter pylori enzymology, Rec A Recombinases metabolism
- Abstract
Helicobacter pylori is a bacterial pathogen colonizing half of the world's human population. It has been implicated in a number of gastric diseases, from asymptomatic gastritis to cancer. It is characterized by an amazing genetic variability that results from high mutation rates and efficient DNA homologous recombination and transformation systems. Here, we report the characterization of H. pylori RecA (HpRecA), a protein shown to be involved in DNA repair, transformation, and mouse colonization. The biochemical characterization of the purified recombinase reveals activities similar to those of Escherichia coli RecA (EcRecA). We show that in H. pylori, HpRecA is present in about 80,000 copies per cell during exponential growth and decreases to about 50,000 copies in stationary phase. The amount of HpRecA remains unchanged after induction of DNA lesions, suggesting that HpRecA is always expressed at a high level in order to repair DNA damage or facilitate recombination. We performed HpRecA localization analysis by adding a Flag tag to the protein, revealing two different patterns of localization. During exponential growth, RecA-Flag presents a diffuse pattern, overlapping with the DAPI (4',6-diamidino-2-phenylindole) staining of DNA, whereas during stationary phase, the protein is present in more defined areas devoid of DAPI staining. These localizations are not affected by inactivation of competence or DNA recombination genes. Neither UV irradiation nor gamma irradiation modified HpRecA localization, suggesting the existence of a constitutive DNA damage adaptation system.
- Published
- 2011
- Full Text
- View/download PDF
43. Unexpected role for Helicobacter pylori DNA polymerase I as a source of genetic variability.
- Author
-
García-Ortíz MV, Marsin S, Arana ME, Gasparutto D, Guérois R, Kunkel TA, and Radicella JP
- Subjects
- DNA Polymerase I chemistry, DNA Repair, DNA Replication, Exonucleases genetics, Genome, Bacterial, Helicobacter pylori genetics, Mutagenesis, Phenotype, Sequence Alignment, DNA Polymerase I genetics, DNA, Bacterial genetics, Exonucleases chemistry, Genetic Variation, Genomic Instability, Helicobacter pylori enzymology
- Abstract
Helicobacter pylori, a human pathogen infecting about half of the world population, is characterised by its large intraspecies variability. Its genome plasticity has been invoked as the basis for its high adaptation capacity. Consistent with its small genome, H. pylori possesses only two bona fide DNA polymerases, Pol I and the replicative Pol III, lacking homologues of translesion synthesis DNA polymerases. Bacterial DNA polymerases I are implicated both in normal DNA replication and in DNA repair. We report that H. pylori DNA Pol I 5'- 3' exonuclease domain is essential for viability, probably through its involvement in DNA replication. We show here that, despite the fact that it also plays crucial roles in DNA repair, Pol I contributes to genomic instability. Indeed, strains defective in the DNA polymerase activity of the protein, although sensitive to genotoxic agents, display reduced mutation frequencies. Conversely, overexpression of Pol I leads to a hypermutator phenotype. Although the purified protein displays an intrinsic fidelity during replication of undamaged DNA, it lacks a proofreading activity, allowing it to efficiently elongate mismatched primers and perform mutagenic translesion synthesis. In agreement with this finding, we show that the spontaneous mutator phenotype of a strain deficient in the removal of oxidised pyrimidines from the genome is in part dependent on the presence of an active DNA Pol I. This study provides evidence for an unexpected role of DNA polymerase I in generating genomic plasticity., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2011
- Full Text
- View/download PDF
44. Critical lysine residues within the overlooked N-terminal domain of human APE1 regulate its biological functions.
- Author
-
Fantini D, Vascotto C, Marasco D, D'Ambrosio C, Romanello M, Vitagliano L, Pedone C, Poletto M, Cesaratto L, Quadrifoglio F, Scaloni A, Radicella JP, and Tell G
- Subjects
- Acetylation, Amino Acid Sequence, Binding Sites, DNA-(Apurinic or Apyrimidinic Site) Lyase classification, DNA-(Apurinic or Apyrimidinic Site) Lyase metabolism, HeLa Cells, Humans, Molecular Sequence Data, Nucleophosmin, Peptides metabolism, Phylogeny, Protein Structure, Tertiary, RNA metabolism, Sequence Analysis, Protein, DNA-(Apurinic or Apyrimidinic Site) Lyase chemistry, Lysine metabolism
- Abstract
Apurinic/apyrimidinic endonuclease 1 (APE1), an essential protein in mammals, is involved in base excision DNA repair (BER) and in regulation of gene expression, acting as a redox co-activator of several transcription factors. Recent findings highlight a novel role for APE1 in RNA metabolism, which is modulated by nucleophosmin (NPM1). The results reported in this article show that five lysine residues (K24, K25, K27, K31 and K32), located in the APE1 N-terminal unstructured domain, are involved in the interaction of APE1 with both RNA and NPM1, thus supporting a competitive binding mechanism. Data from kinetic experiments demonstrate that the APE1 N-terminal domain also serves as a device for fine regulation of protein catalytic activity on abasic DNA. Interestingly, some of these critical lysine residues undergo acetylation in vivo. These results suggest that protein-protein interactions and/or post-translational modifications involving APE1 N-terminal domain may play important in vivo roles, in better coordinating and fine-tuning protein BER activity and function on RNA metabolism.
- Published
- 2010
- Full Text
- View/download PDF
45. Genetic dissection of Helicobacter pylori AddAB role in homologous recombination.
- Author
-
Marsin S, Lopes A, Mathieu A, Dizet E, Orillard E, Guérois R, and Radicella JP
- Subjects
- Bacterial Proteins genetics, Exodeoxyribonucleases genetics, Helicobacter pylori genetics, Bacterial Proteins metabolism, Exodeoxyribonucleases metabolism, Helicobacter pylori enzymology, Recombination, Genetic
- Abstract
Helicobacter pylori infects the stomach of about half of the world's human population, frequently causing chronic inflammation at the origin of several gastric pathologies. One of the most remarkable characteristics of the species is its remarkable genomic plasticity in which homologous recombination (HR) plays a critical role. Here, we analyzed the role of the H. pylori homologue of the AddAB recombination protein. Bioinformatics analysis of the proteins unveils the similarities and differences of the H. pylori AddAB complex with respect to the RecBCD and AddAB complexes from Escherichia coli and Bacillus subtilis, respectively. Helicobacter pylori mutants lacking functional addB or/and addA show the same level of sensitivity to DNA-damaging agents such as UV or irradiation and of deficiency in intrachromosomal RecA-dependent HR. Epistasis analyses of both DNA repair and HR phenotypes, using double and triple recombination mutants, demonstrate that, in H. pylori, AddAB and RecOR complexes define two separate presynaptic pathways with little functional overlap. However, neither of these complexes participates in the RecA-dependent process of transformation of these naturally competent bacteria., (© 2010 Federation of European Microbiological Societies. Published by Blackwell Publishing Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
46. Oxidative stress triggers the preferential assembly of base excision repair complexes on open chromatin regions.
- Author
-
Amouroux R, Campalans A, Epe B, and Radicella JP
- Subjects
- Bromates toxicity, Cell Line, Chromatin chemistry, Chromatin enzymology, DNA Glycosylases analysis, DNA-(Apurinic or Apyrimidinic Site) Lyase analysis, DNA-Binding Proteins analysis, Euchromatin chemistry, Guanosine analogs & derivatives, Guanosine metabolism, Humans, X-ray Repair Cross Complementing Protein 1, DNA Damage, DNA Glycosylases metabolism, DNA Repair, Euchromatin enzymology, Oxidative Stress
- Abstract
How DNA repair machineries detect and access, within the context of chromatin, lesions inducing little or no distortion of the DNA structure is a poorly understood process. Removal of oxidized bases is initiated by a DNA glycosylase that recognises and excises the damaged base, initiating the base excision repair (BER) pathway. We show that upon induction of 8-oxoguanine, a mutagenic product of guanine oxidation, the mammalian 8-oxoguanine DNA glycosylase OGG1 is recruited together with other proteins involved in BER to euchromatin regions rich in RNA and RNA polymerase II and completely excluded from heterochromatin. The underlying mechanism does not require direct interaction of the protein with the oxidized base, however, the release of the protein from the chromatin fraction requires completion of repair. Inducing chromatin compaction by sucrose results in a complete but reversible inhibition of the in vivo repair of 8-oxoguanine. We conclude that after induction of oxidative DNA damage, the DNA glycosylase is actively recruited to regions of open chromatin allowing the access of the BER machinery to the lesions, suggesting preferential repair of active chromosome regions.
- Published
- 2010
- Full Text
- View/download PDF
47. HO* radicals induce an unexpected high proportion of tandem base lesions refractory to repair by DNA glycosylases.
- Author
-
Bergeron F, Auvré F, Radicella JP, and Ravanat JL
- Subjects
- 8-Hydroxy-2'-Deoxyguanosine, Animals, Cattle, DNA drug effects, DNA metabolism, DNA radiation effects, Deoxyadenosines metabolism, Deoxyguanosine analogs & derivatives, Deoxyguanosine metabolism, Gamma Rays, Hydroxyl Radical metabolism, In Vitro Techniques, Models, Biological, Oxidation-Reduction, DNA Damage, DNA Glycosylases metabolism, DNA Repair physiology, Hydroxyl Radical pharmacology
- Abstract
Reaction of HO(*) radicals with double-stranded calf thymus DNA produces high levels of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodGuo) and, to a minor extent, 8-oxo-7,8-dihydro-2'-deoxyadenosine (8-oxodAdo). Formation of the hydroxylated purine lesions is explained by addition of HO(*) to the C8 position of the purine moiety. It has been reported that tandem lesions containing a formylamine residue neighboring 8-oxodGuo could be produced through addition of a transiently generated pyrimidine peroxyl radical onto the C8 of an adjacent purine base. Formation of such tandem lesions accounted for approximately 10% of the total 8-oxodGuo. In the present work we show that addition of HO(*) onto the C8 of purine accounts for only approximately 5% of the generated 8-oxodGuo. About 50% of the 8-hydroxylated purine lesions, including 8-oxodGuo and 8-oxodAdo, are involved in tandem damage and are produced by peroxyl addition onto the C8 of a vicinal purine base. In addition, the remaining 45% of the 8-oxodGuo are produced by an electron transfer reaction, providing an explanation for the higher yield of formation of 8-oxodGuo compared to 8-oxodAdo. Interestingly, we show that >40% of the 8-oxodGuo involved in tandem lesions is refractory to excision by DNA glycosylases. Altogether our results demonstrate that, subsequently to a single oxidation event, peroxidation reactions significantly increase the yield of formation of hydroxylated purine modifications, generating a high proportion of tandem lesions partly refractory to base excision repair.
- Published
- 2010
- Full Text
- View/download PDF
48. Inactivation by oxidation and recruitment into stress granules of hOGG1 but not APE1 in human cells exposed to sub-lethal concentrations of cadmium.
- Author
-
Bravard A, Campalans A, Vacher M, Gouget B, Levalois C, Chevillard S, and Radicella JP
- Subjects
- Cell Line, Tumor, Cell Nucleus metabolism, Cytoplasmic Granules metabolism, DNA Glycosylases metabolism, Down-Regulation, Humans, Oxidation-Reduction, Oxidative Stress, RNA Processing, Post-Transcriptional, Cadmium toxicity, DNA Glycosylases drug effects, DNA-(Apurinic or Apyrimidinic Site) Lyase drug effects
- Abstract
The induction of mutations in mammalian cells exposed to cadmium has been associated with the oxidative stress triggered by the metal. There is increasing evidence that the mutagenic potential of Cd is not restricted to the induction of DNA lesions. Cd has been shown to inactivate several DNA repair enzymes. Here we show that exposure of human cells to sub-lethal concentrations of Cd leads to a time- and concentration-dependent decrease in hOGG1 activity, the major DNA glycosylase activity responsible for the initiation of the base excision repair (BER) of 8-oxoguanine, an abundant and mutagenic form of oxidized guanine. Although there is a slight effect on the level of hOGG1 transcripts, we show that the inhibition of the 8-oxoguanine DNA glycosylase activity is mainly associated with an oxidation of the hOGG1 protein and its disappearance from the soluble fraction of total cell extracts. Confocal microscopy analyses show that in cells exposed to Cd hOGG1-GFP is recruited to discrete structures in the cytoplasm. These structures were identified as stress granules. Removal of Cd from the medium allows the recovery of the DNA glycosylase activity and the presence of hOGG1 in a soluble form. In contrast to hOGG1, we show here that exposure to Cd does not affect the activity of the second enzyme of the pathway, the major AP endonuclease APE1., (Copyright (c) 2009 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
49. Expression of 8-oxoguanine DNA glycosylase (Ogg1) in mouse retina.
- Author
-
Bigot K, Leemput J, Vacher M, Campalans A, Radicella JP, Lacassagne E, Provost A, Masson C, Menasche M, and Abitbol M
- Subjects
- Analysis of Variance, Animals, DNA Polymerase beta genetics, DNA Polymerase beta metabolism, DNA Repair genetics, DNA-(Apurinic or Apyrimidinic Site) Lyase genetics, DNA-(Apurinic or Apyrimidinic Site) Lyase metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Eye metabolism, Gene Expression Profiling methods, Immunohistochemistry, In Situ Hybridization, Mice, Polymerase Chain Reaction, X-ray Repair Cross Complementing Protein 1, DNA Glycosylases genetics, DNA Glycosylases metabolism, Gene Expression, Retina metabolism
- Abstract
Purpose: The retina is highly exposed to oxidative stress due to the high level of oxygen consumption in this tissue and its exposure to light. The main DNA base lesion generated by oxygen free radicals is 8-oxoguanine (8-oxoG). However, its presence in retinal cells and the mechanisms underlying its repair remain undetermined., Methods: 8-oxoguanine DNA glycosylase (Ogg1) gene expression and messenger localization in adult mouse ocular tissues was analyzed by RT-PCR and in situ hybridization. Using immunohistochemistry, we determined the localization of Ogg1 protein and three base excision repair (BER) enzymes: apurinic/apyrimidic endonuclease (APE1), DNA polymerase beta, and X-ray repair cross-complementation group 1 (XRCC1). Ogg1 and AP-lyase activities in the neuroretina were obtained using double-stranded oligonucleotides harboring either an 8-oxoG residue or a tetrahydrofuran., Results: We report here that 8-oxoG is abundant in the retina. Ogg1, the enzyme responsible for the recognition and excision of the oxidized base, is present in its active form and found mainly in ganglion cells and photoreceptor inner segments. We show that APE1 and DNA polymerase beta, two BER proteins involved in 8-oxoG repair, are also present in these cells. The cellular distribution of these proteins was similar to that of Ogg1. XRRC1 is present in both inner nuclear and ganglion cells layers; however, this protein is absent from photoreceptor inner segments., Conclusions: This is the first study to demonstrate the presence of a functional 8-oxoG BER pathway in retinal neurons. The study of three BER proteins involved in 8-oxoG elimination demonstrates that XRCC1 localization differs from those of Ogg1, APE1, and DNA polymerase beta. This result suggests that the elimination of 8-oxoG is coordinated through two pathways, which differ slightly according to the cellular localization of the abnormally oxidized guanine.
- Published
- 2009
50. Oxidation status of human OGG1-S326C polymorphic variant determines cellular DNA repair capacity.
- Author
-
Bravard A, Vacher M, Moritz E, Vaslin L, Hall J, Epe B, and Radicella JP
- Subjects
- Alleles, Cell Line, Humans, Lymphocytes enzymology, Lymphocytes physiology, Oxidation-Reduction, Polymorphism, Single Nucleotide, DNA Glycosylases genetics, DNA Glycosylases metabolism, DNA Repair physiology
- Abstract
The hOGG1 gene encodes the DNA glycosylase that removes the mutagenic lesion 7,8-dihyro-8-oxoguanine (8-oxoG) from DNA. A frequently found polymorphism resulting in a serine to cysteine substitution at position 326 of the OGG1 protein has been associated in several molecular epidemiologic studies with cancer development. To investigate whether the variant allele encodes a protein with altered OGG1 function, we compared the 8-oxoG repair activity, both in vivo and in cell extracts, of lymphoblastoid cell lines established from individuals carrying either Ser/Ser or Cys/Cys genotypes. We show that cells homozygous for the Cys variant display increased genetic instability and reduced in vivo 8-oxoG repair rates. Consistently, their extracts have an almost 2-fold lower basal 8-oxoG DNA glycosylase activity when compared with the Ser variant. Treatment with reducing agents of either the Cys variant cells directly or of protein extracts from these cells increases the repair capacity to the level of the Ser variant, whereas it does not affect the activity in cells or extracts from the latter. Furthermore, the DNA glycosylase activity of cells carrying the Cys/Cys alleles is more sensitive to inactivation by oxidizing agents when compared with that of the Ser/Ser cells. Analysis of the redox status of the OGG1 protein in the cells confirms that the lower activity of OGG1-Cys326 is associated with the oxidation of Cys326 to form a disulfide bond. Our findings support the idea that individuals homozygous for the OGG1-Cys variant could more readily accumulate mutations under conditions of oxidative stress.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.