Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Genes Dev ; 32(17-18): 1242-1251, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30181361

ABSTRACT

During homologous recombination, cells must coordinate repair, DNA damage checkpoint signaling, and movement of chromosomal loci to facilitate homology search. In Saccharomyces cerevisiae, increased movement of damaged loci (local mobility) and undamaged loci (global mobility) precedes homolog pairing in mitotic cells. How cells modulate chromosome mobility in response to DNA damage remains unclear. Here, we demonstrate that global chromosome mobility is regulated by the Rad51 recombinase and its mediator, Rad52. Surprisingly, rad51Δ rad52Δ cells display checkpoint-dependent constitutively increased mobility, indicating that a regulatory circuit exists between recombination and checkpoint machineries to govern chromosomal mobility. We found that the requirement for Rad51 in this circuit is distinct from its role in recombination and that interaction with Rad52 is necessary to alleviate inhibition imposed by mediator recruitment to ssDNA. Thus, interplay between recombination factors and the checkpoint restricts increased mobility until recombination proteins are assembled at damaged sites.


Subject(s)
Chromosomes, Fungal/metabolism , DNA Damage , Homologous Recombination , Rad51 Recombinase/physiology , Rad52 DNA Repair and Recombination Protein/physiology , Saccharomyces cerevisiae Proteins/physiology , Mutation , Rad51 Recombinase/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
2.
Genes Dev ; 32(7-8): 524-536, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29636371

ABSTRACT

Chromosomal deletion rearrangements mediated by repetitive elements often involve repeats separated by several kilobases and sequences that are divergent. While such rearrangements are likely induced by DNA double-strand breaks (DSBs), it has been unclear how the proximity of DSBs relative to repeat sequences affects the frequency of such events. We generated a reporter assay in mouse cells for a deletion rearrangement involving repeats separated by 0.4 Mb. We induced this repeat-mediated deletion (RMD) rearrangement with two DSBs: the 5' DSB that is just downstream from the first repeat and the 3' DSB that is varying distances upstream of the second repeat. Strikingly, we found that increasing the 3' DSB/repeat distance from 3.3 kb to 28.4 kb causes only a modest decrease in rearrangement frequency. We also found that RMDs are suppressed by KU70 and RAD51 and promoted by RAD52, CtIP, and BRCA1. In addition, we found that 1%-3% sequence divergence substantially suppresses these rearrangements in a manner dependent on the mismatch repair factor MSH2, which is dominant over the suppressive role of KU70. We suggest that a DSB far from a repeat can stimulate repeat-mediated rearrangements, but multiple pathways suppress these events.


Subject(s)
Chromosome Breakage , Chromosome Deletion , Repetitive Sequences, Nucleic Acid , Animals , DNA/chemistry , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/physiology , Ku Autoantigen/physiology , Mice , MutS Homolog 2 Protein/physiology , Rad51 Recombinase/physiology , Rad52 DNA Repair and Recombination Protein/physiology , Tumor Suppressor p53-Binding Protein 1/physiology
3.
Genome Biol ; 19(1): 37, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29548335

ABSTRACT

BACKGROUND: Genomic instability promotes evolution and heterogeneity of tumors. Unraveling its mechanistic basis is essential for the design of appropriate therapeutic strategies. In a previous study, we reported an unexpected oncogenic property of p21WAF1/Cip1, showing that its chronic expression in a p53-deficient environment causes genomic instability by deregulation of the replication licensing machinery. RESULTS: We now demonstrate that p21WAF1/Cip1 can further fuel genomic instability by suppressing the repair capacity of low- and high-fidelity pathways that deal with nucleotide abnormalities. Consequently, fewer single nucleotide substitutions (SNSs) occur, while formation of highly deleterious DNA double-strand breaks (DSBs) is enhanced, crafting a characteristic mutational signature landscape. Guided by the mutational signatures formed, we find that the DSBs are repaired by Rad52-dependent break-induced replication (BIR) and single-strand annealing (SSA) repair pathways. Conversely, the error-free synthesis-dependent strand annealing (SDSA) repair route is deficient. Surprisingly, Rad52 is activated transcriptionally in an E2F1-dependent manner, rather than post-translationally as is common for DNA repair factor activation. CONCLUSIONS: Our results signify the importance of mutational signatures as guides to disclose the repair history leading to genomic instability. We unveil how chronic p21WAF1/Cip1 expression rewires the repair process and identifies Rad52 as a source of genomic instability and a candidate therapeutic target.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Repair , Genomic Instability , Mutation , Rad52 DNA Repair and Recombination Protein/physiology , Tumor Suppressor Protein p53/physiology , Cell Line , DNA/biosynthesis , Humans
4.
Mech Ageing Dev ; 167: 56-63, 2017 10.
Article in English | MEDLINE | ID: mdl-29024686

ABSTRACT

Aging may be characterized as the progressive increase of the risk of death caused by a decrease of almost all bodily functions. While a great number of model organism studies have established the role of DNA double strand breaks (DSBs) as one of the main causes of aging, few studies have examined whether common polymorphisms in human DSB repair genes influence aging and mortality. More importantly, to the best of our knowledge, no longitudinal study has thus far examined the link between polymorphisms in DSB repair and the risk of death. This longitudinal study thus analyses whether four common polymorphisms (rs2155209, rs7963551, rs17105278, rs2735383) in four selected DSB repair genes (MRE11A, RAD52, RAD51B, NBS1) influence the hazard of age-adjusted death in a cohort of patients with typical symptoms of ischemic heart disease. The results have shown that rs7963551 G/T heterozygotes exhibit a significantly increased hazard of death when compared with the combined GG and TT homozygotes (HR=1.42, 95% CI: 1.06-1.91, p=0.018). This study indicates that the SNP affecting efficiency of DSB repair may influence aging in humans.


Subject(s)
Cardiovascular Diseases/genetics , Polymorphism, Single Nucleotide , Rad52 DNA Repair and Recombination Protein/genetics , Aged , Cardiovascular Diseases/mortality , Cohort Studies , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Female , Genetic Predisposition to Disease , Genotype , Heterozygote , Humans , Longitudinal Studies , Male , Middle Aged , Multivariate Analysis , Mutation , Proportional Hazards Models , Rad52 DNA Repair and Recombination Protein/physiology , Risk
5.
Nucleic Acids Res ; 42(7): e61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24500205

ABSTRACT

Gene targeting is a genetic technique to modify an endogenous DNA sequence in its genomic location via homologous recombination (HR) and is useful both for functional analysis and gene therapy applications. HR is inefficient in most organisms and cell types, including mammalian cells, often limiting the effectiveness of gene targeting. Therefore, increasing HR efficiency remains a major challenge to DNA editing. Here, we present a new concept for gene correction based on the development of DNA aptamers capable of binding to a site-specific DNA binding protein to facilitate the exchange of homologous genetic information between a donor molecule and the desired target locus (aptamer-guided gene targeting). We selected DNA aptamers to the I-SceI endonuclease. Bifunctional oligonucleotides containing an I-SceI aptamer sequence were designed as part of a longer single-stranded DNA molecule that contained a region with homology to repair an I-SceI generated double-strand break and correct a disrupted gene. The I-SceI aptamer-containing oligonucleotides stimulated gene targeting up to 32-fold in yeast Saccharomyces cerevisiae and up to 16-fold in human cells. This work provides a novel concept and research direction to increase gene targeting efficiency and lays the groundwork for future studies using aptamers for gene targeting.


Subject(s)
Aptamers, Nucleotide/chemistry , Gene Targeting , DNA/chemistry , Deoxyribonucleases, Type II Site-Specific/genetics , HEK293 Cells , Humans , Nucleic Acid Conformation , Rad52 DNA Repair and Recombination Protein/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Sequence Homology, Nucleic Acid
6.
Oncogene ; 32(30): 3552-8, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-22964643

ABSTRACT

Synthetic lethality is an approach to study selective cell killing based on genotype. Previous work in our laboratory has shown that loss of RAD52 is synthetically lethal with BRCA2 deficiency, while exhibiting no impact on cell growth and viability in BRCA2-proficient cells. We now show that this same synthetically lethal relationship is evident in cells with deficiencies in BRCA1 or PALB2, which implicates BRCA1, PALB2 and BRCA2 in an epistatic relationship with one another. When RAD52 was depleted in BRCA1- or PALB2-deficient cells, a severe reduction in plating efficiency was observed, with many abortive attempts at cell division apparent in the double-depleted background. In contrast, when RAD52 was depleted in a BRCA1- or PALB2-wildtype background, a negligible decrease in colony survival was observed. The frequency of ionizing radiation-induced RAD51 foci formation and double-strand break-induced homologous recombination (HR) was decreased by 3- and 10-fold, respectively, when RAD52 was knocked down in BRCA1- or PALB2-depleted cells, with minimal effect in BRCA1- or PALB2-proficient cells. RAD52 function was independent of BRCA1 status, as evidenced by the lack of any defect in RAD52 foci formation in BRCA1-depleted cells. Collectively, these findings suggest that RAD52 is an alternative repair pathway of RAD51-mediated HR, and a target for therapy in cells deficient in the BRCA1-PALB2-BRCA2 repair pathway.


Subject(s)
Genes, BRCA1 , Genes, BRCA2 , Homologous Recombination/physiology , Nuclear Proteins/genetics , Rad51 Recombinase/physiology , Rad52 DNA Repair and Recombination Protein/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , DNA Repair/drug effects , DNA Repair/genetics , Fanconi Anemia Complementation Group N Protein , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Silencing , Genes, BRCA1/physiology , Genes, BRCA2/physiology , Genes, Lethal/genetics , Genes, Lethal/physiology , Homologous Recombination/genetics , Humans , RNA, Small Interfering/pharmacology , Rad51 Recombinase/genetics , Rad52 DNA Repair and Recombination Protein/physiology
7.
PLoS One ; 7(1): e30622, 2012.
Article in English | MEDLINE | ID: mdl-22292001

ABSTRACT

The SCF (Skp1-Cul1-F-box) complex contributes to a variety of cellular events including meiotic cell cycle control, but its function during meiosis is not understood well. Here we describe a novel function of SCF/Skp1 in meiotic recombination and subsequent chromosome segregation. The skp1 temperature-sensitive mutant exhibited abnormal distribution of spindle microtubules in meiosis II, which turned out to originate from abnormal bending of the spindle in meiosis I. Bent spindles were reported in mitosis of this mutant, but it remained unknown how SCF could affect spindle morphology. We found that the meiotic bent spindle in skp1 cells was due to a hypertension generated by chromosome entanglement. The spindle bending was suppressed by inhibiting double strand break (DSB) formation, indicating that the entanglement was generated by the meiotic recombination machinery. Consistently, Rhp51/Rad51-Rad22/Rad52 foci persisted until meiosis I in skp1 cells, proving accumulation of recombination intermediates. Intriguingly bent spindles were also observed in the mutant of Fbh1, an F-box protein containing the DNA helicase domain, which is involved in meiotic recombination. Genetic evidence suggested its cooperation with SCF/Skp1. Thus, SCF/Skp1 together with Fbh1 is likely to function in the resolution of meiotic recombination intermediates, thereby ensuring proper chromosome segregation.


Subject(s)
Chromosome Segregation/genetics , Meiosis/genetics , Recombination, Genetic , SKP Cullin F-Box Protein Ligases/physiology , Chromosome Segregation/physiology , DNA Helicases/genetics , DNA Helicases/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Genetic Complementation Test , Organisms, Genetically Modified , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/physiology , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Rad52 DNA Repair and Recombination Protein/physiology , Recombination, Genetic/genetics , Recombination, Genetic/physiology , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces/metabolism , Schizosaccharomyces/physiology , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Tubulin/genetics , Tubulin/physiology
8.
Proc Natl Acad Sci U S A ; 108(26): 10448-53, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21670257

ABSTRACT

The human tumor suppressor protein BRCA2 plays a key role in recombinational DNA repair. BRCA2 recruits RAD51 to sites of DNA damage through interaction with eight conserved motifs of approximately 35 amino acids, the BRC repeats; however, the specific function of each repeat remains unclear. Here, we investigated the function of the individual BRC repeats by systematically analyzing their effects on RAD51 activities. Our results reveal the existence of two categories of BRC repeats that display unique functional characteristics. One group, comprising BRC1, -2, -3, and -4, binds to free RAD51 with high affinity. The second group, comprising BRC5, -6, -7, and -8, binds to free RAD51 with low affinity but binds to the RAD51-ssDNA filament with high affinity. Each member of the first group reduces the ATPase activity of RAD51, whereas none of the BRC repeats of the second group affects this activity. Thus, through different mechanisms, both types of BRC repeats bind to and stabilize the RAD51 nucleoprotein filament on ssDNA. In addition, members of the first group limit binding of RAD51 to duplex DNA, where members of the second group do not. Only the first group enhances DNA strand exchange by RAD51. Our results suggest that the two groups of BRC repeats have differentially evolved to ensure efficient formation of a nascent RAD51 filament on ssDNA by promoting its nucleation and growth, respectively. We propose that the BRC repeats cooperate in a partially redundant but reinforcing manner to ensure a high probability of RAD51 filament formation.


Subject(s)
BRCA2 Protein/physiology , Rad52 DNA Repair and Recombination Protein/physiology , Repetitive Sequences, Amino Acid , Adenosine Triphosphatases/metabolism , BRCA2 Protein/chemistry , BRCA2 Protein/metabolism , Electrophoretic Mobility Shift Assay , Humans , Protein Binding , Rad52 DNA Repair and Recombination Protein/metabolism
10.
Cell Cycle ; 7(15): 2418-26, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18677117

ABSTRACT

Specific ataxia telangiectasia and Rad3-related (ATR) mutations confer higher frequencies of homologous recombination. The genetic requirements for hyper-recombination in ATR mutants are unknown. MEC1, the essential yeast ATR/ATM homolog, controls S and G(2) checkpoints and the DNA damage-inducibility of genes after radiation exposure. Since the mec1-Delta (null) mutant is defective in both S and G(2) checkpoints, we measured spontaneous and DNA damage-associated sister chromatid exchange (SCE), homolog (heteroallelic) recombination, and homology-directed translocations in the mec1-21 hypomorphic mutant, which is defective in the S phase checkpoint but retains some G(2) checkpoint function. We observed a sixfold, tenfold and 30-fold higher rate of spontaneous SCE, heteroallelic recombination, and translocations, respectively, in mec1-21 mutants compared to wild type. The mec1-21 hyper-recombination was partially reduced in rad9, pds1 and chk1 mutants, and abolished in rad52 mutants, suggesting the hyper-recombination results from RAD52-dependent recombination pathway(s) that require G(2) checkpoint functions. The HU and UV sensitivities of mec1-21 rad9 and mec1-21 rad52 were synergistically increased, compared to the single mutants, indicating that mec1-21, rad52 and rad9 mutants are defective in independent pathways for HU and UV resistance. G(2)-arrested mec1-21 rad9 cells exhibit more UV resistance than non-synchronized cells, indicating that one function of RAD9 in conferring UV resistance in mec1-21 is by triggering G(2) arrest. We suggest that checkpoint genes that function in the RAD9-mediated pathway are required for either homologous recombination or DNA damage resistance in the S phase checkpoint mutant mec1-21.


Subject(s)
Cell Cycle Proteins/physiology , Genes, cdc/physiology , Nuclear Proteins/physiology , Protein Kinases/physiology , Recombination, Genetic/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Cell Cycle Proteins/genetics , Checkpoint Kinase 1 , G2 Phase/genetics , Intracellular Signaling Peptides and Proteins , Models, Biological , Mutant Proteins/genetics , Mutant Proteins/physiology , Nuclear Proteins/genetics , Organisms, Genetically Modified , Phenotype , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Rad52 DNA Repair and Recombination Protein/physiology , Securin , Sequence Homology
11.
Curr Biol ; 18(15): 1142-6, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18656357

ABSTRACT

In eukaryotes, homologous recombination proteins such as RAD51 and RAD52 play crucial roles in DNA repair and genome stability. Human RAD52 is a member of a large single-strand annealing protein (SSAP) family [1] and stimulates Rad51-dependent recombination [2, 3]. In prokaryotes and phages, it has been difficult to establish the presence of RAD52 homologs with conserved sequences. Putative SSAPs were recently found in several phages that infect strains of Lactococcus lactis[4]. One of these SSAPs was identified as Sak and was found in the virulent L. lactis phage ul36, which belongs to the Siphoviridae family [4, 5]. In this study, we show that Sak is homologous to the N terminus of human RAD52. Purified Sak binds single-stranded DNA (ssDNA) preferentially over double-stranded DNA (dsDNA) and promotes the renaturation of long complementary ssDNAs. Sak also binds RecA and stimulates homologous recombination reactions. Mutations shown to modulate RAD52 DNA binding [6] affect Sak similarly. Remarkably, electron-microscopic reconstruction of Sak reveals an undecameric (11) subunit ring, similar to the crystal structure of the N-terminal fragment of human RAD52 [7, 8]. For the first time, we propose a viral homolog of RAD52 at the amino acid, phylogenic, functional, and structural levels.


Subject(s)
DNA-Binding Proteins/chemistry , Rad52 DNA Repair and Recombination Protein/chemistry , Siphoviridae/genetics , Viral Proteins/chemistry , Amino Acid Sequence , DNA Repair , DNA-Binding Proteins/physiology , DNA-Binding Proteins/ultrastructure , Humans , Lactococcus lactis/virology , Models, Molecular , Molecular Sequence Data , Phylogeny , Rad52 DNA Repair and Recombination Protein/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Structural Homology, Protein , Viral Proteins/physiology , Viral Proteins/ultrastructure
12.
FEMS Yeast Res ; 8(3): 351-61, 2008 May.
Article in English | MEDLINE | ID: mdl-18248415

ABSTRACT

To identify new human proteins implicated in homologous recombination (HR), we set up 'a papillae assay' to screen a human cDNA library using the RS112 strain of Saccharomyces cerevisiae containing an intrachromosomal recombination substrate. We isolated 23 cDNAs, 11 coding for complete proteins and 12 for partially deleted proteins that increased HR when overexpressed in yeast. We characterized the effect induced by the overexpression of the complete human proteasome subunit beta 2, the partially deleted proteasome subunits alpha 3 and beta 8, the ribosomal protein L12, the brain abundant membrane signal protein (BASP1) and the human homologue to v-Ha-RAS (HRAS), which elevated HR by 2-6.5-fold over the control. We found that deletion of the RAD52 gene, which has a key role in most HR events, abolished the increase of HR induced by the proteasome subunits and HRAS; by contrast, the RAD52 deletion did not affect the high level of HR due to BASP1 and RPL12. This suggests that the proteins stimulated yeast HR via different mechanisms. Overexpression of the complete beta 2 human proteasome subunit or the partially deleted alpha 3 and beta 8 subunits increased methyl methanesulphonate (MMS) resistance much more in the rad52 Delta mutant than in the wild-type. Overexpression of RPL12 and BASP1 did not affect MMS resistance in both the wild-type and the rad52 Delta mutant, whereas HRAS decreased MMS resistance in the rad52 Delta mutant. The results indicate that these proteins may interfere with the pathway(s) involved in the repair of MMS-induced DNA damage. Finally, we provide further evidence that yeast is a helpful tool to identify human proteins that may have a regulatory role in HR.


Subject(s)
Recombination, Genetic , Saccharomyces cerevisiae/genetics , DNA Repair , Gene Library , Genes, ras , Humans , Membrane Proteins/genetics , Methyl Methanesulfonate/pharmacology , Nerve Tissue Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/physiology , Repressor Proteins/genetics , Ribosomal Proteins/genetics
13.
Mutagenesis ; 23(2): 119-29, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18267950

ABSTRACT

Protection from chronic exposure to cosmic radiation, which is primarily composed of protons, in future manned missions to Mars and beyond is considered to be a key unresolved issue. To model the effects of cosmic radiation on a living cell, we used Saccharomyces cerevisiae cells harboring various deletions of DNA repair genes to investigate the response of cells to DNA strand breaks caused by exposure to 250 MeV proton irradiation (linear energy transfer of 0.41 keV/microm). In our study, DNA strand breaks induced by exposure to protons were predominantly repaired via the homologous recombination and postreplication repair pathways. We simulated chronic exposure to proton irradiation by treating cells from colonies that survived proton treatment, after several rounds of subculturing, to a second proton dose, as well as additional cell stressors. In general, cells cultured from proton surviving colonies were not more sensitive to secondary cell stressors. However, cells from rad52delta colonies that survived proton treatment showed increased resistance to secondary stressors, such as gamma-rays (1.17 and 1.33 MeV; 0.267 keV/microm), ultraviolet (UV) and proton irradiation and elevated temperatures. Resistance to secondary stressors was also observed in rad52delta cells that survived exposure to gamma-rays, rather than protons, but this was not observed to occur in rad52delta cells after UV irradiation. rad52delta cells that survived exposure to protons, followed by gamma-rays (proton surviving colonies were cultured prior to gamma-ray exposure), exhibited an additive effect, whereby these cells had a further increase in stress resistance. A genetic analysis indicated that increased stress resistance is most likely due to a second-site mutation that suppresses the rad52delta phenotype. We will discuss possible origins of these second-site mutations.


Subject(s)
DNA Breaks , DNA Repair/genetics , DNA/radiation effects , Protons , Rad52 DNA Repair and Recombination Protein/physiology , Recombination, Genetic , Gamma Rays , Gene Deletion , Mutation , Rad52 DNA Repair and Recombination Protein/genetics , Radiation Tolerance/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/radiation effects , Ultraviolet Rays
14.
Mol Cell Biol ; 28(3): 897-906, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18039855

ABSTRACT

Homologous recombination (HR) is critical for DNA double-strand break (DSB) repair and genome stabilization. In yeast, HR is catalyzed by the Rad51 strand transferase and its "mediators," including the Rad52 single-strand DNA-annealing protein, two Rad51 paralogs (Rad55 and Rad57), and Rad54. A Rad51 homolog, Dmc1, is important for meiotic HR. In wild-type cells, most DSB repair results in gene conversion, a conservative HR outcome. Because Rad51 plays a central role in the homology search and strand invasion steps, DSBs either are not repaired or are repaired by nonconservative single-strand annealing or break-induced replication mechanisms in rad51Delta mutants. Although DSB repair by gene conversion in the absence of Rad51 has been reported for ectopic HR events (e.g., inverted repeats or between plasmids), Rad51 has been thought to be essential for DSB repair by conservative interchromosomal (allelic) gene conversion. Here, we demonstrate that DSBs stimulate gene conversion between homologous chromosomes (allelic conversion) by >30-fold in a rad51Delta mutant. We show that Rad51-independent allelic conversion and break-induced replication occur independently of Rad55, Rad57, and Dmc1 but require Rad52. Unlike DSB-induced events, spontaneous allelic conversion was detected in both rad51Delta and rad52Delta mutants, but not in a rad51Delta rad52Delta double mutant. The frequencies of crossovers associated with DSB-induced gene conversion were similar in the wild type and the rad51Delta mutant, but discontinuous conversion tracts were fivefold more frequent and tract lengths were more widely distributed in the rad51Delta mutant, indicating that heteroduplex DNA has an altered structure, or is processed differently, in the absence of Rad51.


Subject(s)
Cell Cycle Proteins/physiology , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/physiology , Rad51 Recombinase/physiology , Rad52 DNA Repair and Recombination Protein/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Adenosine Triphosphatases , DNA Repair Enzymes , Gene Conversion , Nucleic Acid Conformation , Nucleic Acid Heteroduplexes/chemistry , Rad51 Recombinase/genetics , Rad52 DNA Repair and Recombination Protein/genetics
16.
Biochem Biophys Res Commun ; 357(3): 800-3, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17442269

ABSTRACT

Bleomycins are small glycopeptide cancer chemotherapeutics that give rise to 3'-modified DNA double-strand breaks (DSBs). In Saccharomyces cerevisiae, DSBs are predominantly repaired by RAD52-dependent homologous recombination (HR) with some support by Yku70/Yku80 (KU)-dependent pathways. The main DSB repair function of KU is believed to be as part of the non-homologous end-joining (NHEJ) pathway, but KU also functions in a "chromosome healing" pathway that seals DSBs by de novo telomere addition. We report here that rad52Deltayku70Delta double mutants are considerably more bleomycin hypersensitive than rad52Deltalig4Delta cells that lack the NHEJ-specific DNA ligase 4. Moreover, the telomere-specific KU mutation yku80-135i also dramatically increases rad52Delta bleomycin hypersensitivity, almost to the level of rad52Deltayku80Delta. The results indicate that telomere-specific functions of KU play a more prominent role in the repair of bleomycin-induced damage than its NHEJ functions, which could have important clinical implications for bleomycin-based combination chemotherapies.


Subject(s)
Antigens, Nuclear/physiology , Bleomycin/toxicity , DNA Damage , DNA Repair/physiology , DNA-Binding Proteins/physiology , Telomere/metabolism , Antibiotics, Antineoplastic/toxicity , Antigens, Nuclear/genetics , DNA Ligase ATP , DNA Ligases/genetics , DNA Ligases/physiology , DNA Repair/genetics , DNA, Fungal/drug effects , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , Ku Autoantigen , Microbial Viability/drug effects , Microbial Viability/genetics , Mutation , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/physiology , Recombination, Genetic , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Telomere/genetics
17.
J Biol Chem ; 281(52): 40001-9, 2006 Dec 29.
Article in English | MEDLINE | ID: mdl-17040915

ABSTRACT

We examined the double-stranded DNA (dsDNA) binding preference of the Saccharomyces cerevisiae Rad52 protein and its homologue, the Rad59 protein. In nuclease protection assays both proteins protected an internal sequence and the dsDNA ends equally well. Similarly, using electrophoretic mobility shift assays, we found the affinity of both Rad52 and Rad59 proteins for DNA ends to be comparable with their affinity for internal sequences. The protein-DNA complexes were also directly visualized using atomic force microscopy. Both proteins formed discrete complexes, which were primarily found (90-94%) at internal dsDNA sites. We also measured the DNA end binding behavior of human Rad52 protein and found a slight preference for dsDNA ends. Thus, these proteins have no strong preference for dsDNA ends over internal sites, which is inconsistent with their function at a step of dsDNA break repair that precedes DNA processing. Therefore, we conclude that S. cerevisiae Rad52 and Rad59 proteins and their eukaryotic counterparts function by binding to single-stranded DNA formed as intermediates of recombination rather than by binding to the unprocessed DNA double-strand break.


Subject(s)
DNA, Fungal/metabolism , DNA-Binding Proteins/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , Recombination, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Nucleic Acid , DNA Breaks, Double-Stranded , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/physiology , Humans , Protein Binding/genetics , Protein Processing, Post-Translational/genetics , Rad52 DNA Repair and Recombination Protein/physiology , Saccharomyces cerevisiae Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...