Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
2.
Mutat Res ; 663(1-2): 7-14, 2009 Apr 26.
Article in English | MEDLINE | ID: mdl-19150617

ABSTRACT

Irradiation of cells with UVC light induces two types of mutagenic DNA photoproducts, i.e. cyclobutane pyrimidine dimers (CPD) and pyrimidine (6-4) pyrimidone photoproducts (6-4 PP). To investigate the relationship between the frequency of UV-induced photolesions at specific sites and their ability to induce mutations, we quantified CPD formation at the nucleotide level along exons 3 and 8 of the hprt gene using ligation-mediated PCR, and determined the mutational spectrum of 132 UV-induced hprt mutants in the AA8 hamster cell line and of 165 mutants in its nucleotide excision repair-defective derivative UV5. In AA8 cells, transversions predominated with a strong strand bias towards thymine-containing photolesions in the non-transcribed strand. As hamster AA8 cells are proficient in global genome repair of 6-4 PP but selectively repair CPD from the transcribed strand of active genes, most mutations probably resulted from erroneous bypass of CPD in the non-transcribed strand. However, the relative incidence of CPD and the positions where mutations most frequently arose do not correlate. In fact some major damage sites hardly gave rise to the formation of mutations. In the repair-defective UV5 cells, mutations were almost exclusively C>T transitions caused by photoproducts at PyC sites in the transcribed strand. Even though CPD were formed at high frequencies at some TT sites in UV5, these photoproducts did not contribute to mutation induction at all. We conclude that, even in the absence of repair, large variations in the level of induction of CPD at different sites throughout the two exons do not correspond to frequencies of mutation induction.


Subject(s)
DNA Repair/radiation effects , Mutation/genetics , Pyrimidine Dimers/metabolism , Ultraviolet Rays , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , Exons/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Molecular Sequence Data
3.
Mutat Res ; 640(1-2): 131-8, 2008 Apr 02.
Article in English | MEDLINE | ID: mdl-18249417

ABSTRACT

Estimates of genotoxic effects of mutagens at low and protracted doses are often based on linear extrapolation of data obtained at relatively high doses. To test the validity of such an approach, a comparison was made between the mutagenicity of N-methyl-N-nitrosourea (MNU) in T-lymphocytes of the rat following two treatment protocols, i.e. sub-chronic exposure to a low dose (15-45 repeated exposures to 1mg/kg of MNU) or acute exposure to a single high dose (15, 30 or 45 mg/kg of MNU). Mutation induction appeared dramatically lower following sub-chronic treatment compared to treatment with a single high exposure. Furthermore, DNA sequence analysis of the coding region of the hprt gene in MNU-induced mutants showed that acute high dose treatment causes mainly GC-->AT base pair changes, whereas sub-chronic treatment results in a significant contribution of AT base pair changes to mutation induction. We hypothesize that O(6)-methylguanine-DNA methyltransferase is saturated after acute treatments, while after sub-chronic treatment most O(6)-methylguanine is efficiently repaired. These data suggest (i) that risk estimations at low and protracted doses of MNU on the basis of linear extrapolation of effects measured at high dose are too high and (ii) that the protective effects of DNA repair processes are relatively strong at low sub-chronic exposure.


Subject(s)
Methylnitrosourea/toxicity , Mutagenesis , Spleen/drug effects , Alkylating Agents , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Male , Methylation , Methylnitrosourea/administration & dosage , Rats , Rats, Wistar , Time Factors
4.
Mol Cell Biol ; 23(16): 5755-67, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12897146

ABSTRACT

Nucleotide excision repair (NER) is the main DNA repair pathway in mammals for removal of UV-induced lesions. NER involves the concerted action of more than 25 polypeptides in a coordinated fashion. The xeroderma pigmentosum group A protein (XPA) has been suggested to function as a central organizer and damage verifier in NER. How XPA reaches DNA lesions and how the protein is distributed in time and space in living cells are unknown. Here we studied XPA in vivo by using a cell line stably expressing physiological levels of functional XPA fused to green fluorescent protein and by applying quantitative fluorescence microscopy. The majority of XPA moves rapidly through the nucleoplasm with a diffusion rate different from those of other NER factors tested, arguing against a preassembled XPA-containing NER complex. DNA damage induced a transient ( approximately 5-min) immobilization of maximally 30% of XPA. Immobilization depends on XPC, indicating that XPA is not the initial lesion recognition protein in vivo. Moreover, loading of replication protein A on NER lesions was not dependent on XPA. Thus, XPA participates in NER by incorporation of free diffusing molecules in XPC-dependent NER-DNA complexes. This study supports a model for a rapid consecutive assembly of free NER factors, and a relatively slow simultaneous disassembly, after repair.


Subject(s)
DNA Repair , DNA-Binding Proteins/physiology , Cell Line , Cell Nucleus/metabolism , DNA Damage , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Radiation , Fibroblasts/metabolism , Green Fluorescent Proteins , Humans , Immunoblotting , Light , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Peptides/chemistry , Protein Structure, Tertiary , Time Factors , Transfection , Ultraviolet Rays , Xeroderma Pigmentosum Group A Protein
5.
Mutat Res ; 615(1-2): 143-52, 2007 Feb 03.
Article in English | MEDLINE | ID: mdl-17208257

ABSTRACT

The Saccharomyces cerevisiae Rad52 protein has a crucial role in the repair of DNA double-strand breaks by homologous recombination. In vitro, Rad52 displays DNA binding and strand annealing activities and promotes Rad51-mediated strand exchange. Schizosaccharomyces pombe has two Rad52 homologues, Rad22A and Rad22B. Whereas rad22A deficient strains exhibit severe defects in repair and recombination, rad22B mutants have a much less severe phenotype. To better understand the role of Rad22A and Rad22B in double-strand break repair, both proteins were purified to near homogeneity. Using gel retardation and filter binding assays, binding of Rad22A and Rad22B to short single-stranded DNAs was demonstrated. Binding of Rad22A to double-stranded oligonucleotides or linearized plasmid molecules containing blunt ends or short single-stranded overhangs could not be detected. Rad22B also does not bind efficiently to short duplex oligonucleotides but binds readily to DNA fragments containing 3'-overhangs. Rad22A as well as Rad22B efficiently promote annealing of complementary single-stranded DNAs. In the presence of Rad22A annealing of complementary DNAs is almost 90%. Whereas in reactions containing Rad22B the maximum level of annealing is 60%, most likely due to inhibition of the reaction by duplex DNA. Gel-filtration experiments and electron microscopic analyses indicate self-association of Rad22A and Rad22B and the formation of multimeric structures as has been observed for Rad52 in yeast and man.


Subject(s)
DNA-Binding Proteins/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/metabolism , Base Sequence , DNA Breaks, Double-Stranded , DNA Repair , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Microscopy, Electron , Multiprotein Complexes , Mutation , Phenotype , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
6.
DNA Repair (Amst) ; 4(5): 571-82, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15811629

ABSTRACT

Previous studies point to the XPC-hHR23B complex as the principal initiator of global genome nucleotide excision repair (NER) pathway, responsible for the repair of UV-induced cyclobutane pyrimidine dimers (CPD) and 6-4 photoproducts (6-4PP) in human cells. However, the UV-damaged DNA binding protein (UV-DDB) has also been proposed as a damage recognition factor involved in repair of UV-photoproducts, especially CPD. Here, we show in human XP-E cells (UV-DDB deficient) that the incision complex formation at UV-induced lesions was severely diminished in locally damaged nuclear spots. Repair kinetics of CPD and 6-4PP in locally and globally UV-irradiated normal human and XP-E cells demonstrate that UV-DDB can mediate efficient targeting of XPC-hHR23B and other NER factors to 6-4PP. The data is consistent with a mechanism in which UV-DDB forms a stable complex when bound to a 6-4PP, allowing subsequent repair proteins--starting with XPC-hHR23B--to accumulate, and verify the lesion, resulting in efficient 6-4PP repair. These findings suggest that (i) UV-DDB accelerates repair of 6-4PP, and at later time points also CPD, (ii) the fraction of 6-4PP that can be bound by UV-DDB is limited due to its low cellular quantity and fast UV dependent degradation, and (iii) in the absence of UV-DDB a slow XPC-hHR23B dependent pathway is capable to repair 6-4PP, and to some extent also CPD.


Subject(s)
DNA Damage/radiation effects , DNA Repair , DNA/metabolism , DNA/radiation effects , Pyrimidine Dimers , Xeroderma Pigmentosum , Cell Nucleus/radiation effects , DNA/genetics , DNA Damage/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Fibroblasts/drug effects , Fibroblasts/radiation effects , Humans , Photochemistry , Ultraviolet Rays , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group A Protein
7.
DNA Repair (Amst) ; 4(10): 1121-8, 2005 Sep 28.
Article in English | MEDLINE | ID: mdl-16009599

ABSTRACT

RAD52 and RAD54 genes from Saccharomyces cerevisiae are required for double-strand break repair through homologous recombination and show epistatic interactions i.e., single and double mutant strains are equally sensitive to DNA damaging agents. In here we combined mutations in RAD52 and RAD54 homologs in Schizosaccharomyces pombe and mice. The analysis of mutant strains in S. pombe demonstrated nearly identical sensitivities of rhp54, rad22A and rad22B double and triple mutants to X-rays, cis-diamminedichloroplatinum and hydroxyurea. In this respect, the fission yeast homologs of RAD54 and RAD52 closely resemble their counterparts in S. cerevisiae. To verify if inactivation of RAD52 affects the DNA damage sensitivities of RAD54 deficient mice, several endpoints were studied in double mutant mice and in bone marrow cells derived from these animals. Haemopoietic depression in bone marrow and the formation of micronuclei after in vivo exposure to mitomycine C (MMC) was not increased in either single or double mutant mice in comparison to wildtype animals. The induction of sister chromatid exchanges in splenocytes was slightly reduced in the RAD54 mutant. A similar reduction was detected in the double mutant. However, a deficiency of RAD52 exacerbates the MMC survival of RAD54 mutant mice and also has a distinct effect on the survival of bone marrow cells after exposure to ionizing radiation. These findings may be explained by additive defects in HR in the double mutant but may also indicate a more prominent role for single-strand annealing in the absence of Rad54.


Subject(s)
Nuclear Proteins/genetics , Schizosaccharomyces/genetics , Alkylating Agents/pharmacology , Animals , Bone Marrow/drug effects , DNA Damage/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Epistasis, Genetic , Erythrocytes/drug effects , Female , Hematopoiesis/genetics , Hematopoiesis/radiation effects , Male , Mice , Mice, Knockout , Micronucleus Tests , Mitomycin/toxicity , Mutation , Radiation Tolerance/genetics , Schizosaccharomyces pombe Proteins/genetics , Sister Chromatid Exchange/genetics
8.
Cancer Res ; 64(3): 889-94, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871817

ABSTRACT

Defects in nucleotide excision repair have been shown to be associated with the photosensitive form of the disorder trichothiodystrophy (TTD). Most repair-deficient TTD patients are mutated in the XPD gene, a subunit of the transcription factor TFIIH. Knowledge of the kinetics and efficiency of repair of the two major UV-induced photolesions in TTD is critical to understand the role of unrepaired lesions in the process of carcinogenesis and explain the absence of enhanced skin cancer incidence in TTD patients contrarily to the xeroderma pigmentosum D patients. In this study, we used different approaches to quantify repair of UV-induced cyclobutane pyrimidine dimers (CPD) and pyrimidine (6-4) pyrimidone photoproducts (6-4PP) at the gene and the genome overall level. In cells of two TTD patients, repair of CPD and 6-4PP was reduced compared with normal human cells, but the reduction was more severe in confluent cells than in exponentially growing cells. Moreover, the impairment of repair was more drastic for CPD than 6-4PP. Most notably, exponentially growing TTD cells displayed complete repair 6-4PP over a broad dose range, albeit at a reduced rate compared with normal cells. Strand-specific analysis of CPD repair in a transcriptional active gene revealed that TTD cells were capable to perform transcription-coupled repair. Taken together, the data suggest that efficient repair of 6-4PP in dividing TTD cells in concert with transcription-coupled repair might account for the absence of increased skin carcinogenesis in TTD patients.


Subject(s)
DNA Repair/physiology , Hair Diseases/genetics , Pyrimidine Dimers/genetics , Cells, Cultured , Fibroblasts/physiology , Fibroblasts/radiation effects , Hair Diseases/pathology , Humans , Immunoblotting , Pyrimidine Dimers/metabolism , Ultraviolet Rays
9.
BMC Genomics ; 6: 6, 2005 Jan 18.
Article in English | MEDLINE | ID: mdl-15656902

ABSTRACT

BACKGROUND: During excessive pressure or volume overload, cardiac cells are subjected to increased mechanical stress (MS). We set out to investigate how the stress response of cardiac cells to MS can be compared to genotoxic stresses induced by DNA damaging agents. We chose for this purpose to use ionising radiation (IR), which during mediastinal radiotherapy can result in cardiac tissue remodelling and diminished heart function, and ultraviolet radiation (UV) that in contrast to IR induces high concentrations of DNA replication- and transcription-blocking lesions. RESULTS: Cultures enriched for neonatal rat cardiac myocytes (CM) or fibroblasts were subjected to any one of the three stressors. Affymetrix microarrays, analysed with Linear Modelling on Probe Level, were used to determine gene expression patterns at 24 hours after (the start of) treatment. The numbers of differentially expressed genes after UV were considerably higher than after IR or MS. Remarkably, after all three stressors the predominant gene expression response in CM-enriched fractions was up-regulation, while in fibroblasts genes were more frequently down-regulated. To investigate the activation or repression of specific cellular pathways, genes present on the array were assigned to 25 groups, based on their biological function. As an example, in the group of cholesterol biosynthesis a significant proportion of genes was up-regulated in CM-enriched fractions after MS, but down-regulated after IR or UV. CONCLUSION: Gene expression responses after the types of cellular stress investigated (MS, IR or UV) have a high stressor and cell type specificity.


Subject(s)
Fibroblasts/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Myocytes, Cardiac/metabolism , Oligonucleotide Array Sequence Analysis/methods , Animals , Cells, Cultured , DNA/metabolism , DNA Damage , DNA Repair , DNA, Complementary/metabolism , Down-Regulation , Fibroblasts/radiation effects , Gene Expression , Genetic Techniques , Myocytes, Cardiac/radiation effects , Nucleic Acid Hybridization , Polymerase Chain Reaction , Pressure , Radiation, Ionizing , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stress, Mechanical , Tenascin/metabolism , Transcription, Genetic , Ultraviolet Rays , Up-Regulation , X-Rays
10.
DNA Repair (Amst) ; 2(12): 1297-308, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14642560

ABSTRACT

Nucleotide excision repair (NER) is the principal pathway for the removal of a wide range of DNA helix-distorting lesions. Two NER subpathways have been identified, i.e. global genome repair (GGR) and transcription-coupled repair (TCR). Little is known about the expression of NER pathways in differentiated cells. We assessed the repair of UV-induced cyclobutane pyrimidine dimers (CPD) and 6-4-photoproducts (6-4 PP) in terminally differentiated myocytes and proliferating fibroblasts isolated from the hearts of neonatal rats. Myocytes and fibroblasts were found to carry out efficient removal of 6-4 PP but display poor repair of CPD by GGR. Furthermore, both cell types were found to carry out TCR of CPD, thus mimicking the repair phenotype of established rodent cell lines. The inefficient repair of CPD at the genome overall level occurs in the absence of massive apoptosis, but goes along with an undetectable level of transcription of the p48 gene, known to be mutated in xeroderma pigmentosum group E (XP-E) patients and recently proposed to be essential for repair of CPD in nonexpressed DNA. Taken together, the results suggest that primary non-dividing cardiac myocytes and proliferating fibroblasts from rat heart selectively remove CPD from the transcribed strand of transcriptionally active genes. GGR of CPD is poor due to the absence of p48 expression.


Subject(s)
DNA Repair/genetics , DNA/radiation effects , Genome , Heart/radiation effects , Pyrimidine Dimers/genetics , Animals , Apoptosis/radiation effects , Caspase 3 , Caspases/metabolism , Cell Differentiation , Cell Division , Cells, Cultured , DNA Damage , Fibroblasts/radiation effects , Male , Muscle Cells/radiation effects , RNA/genetics , RNA/metabolism , RNA/radiation effects , Rats , Rats, Wistar , Ultraviolet Rays
11.
Radiat Res ; 164(4 Pt 1): 383-90, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16187740

ABSTRACT

The adaptive response to ionizing radiation may be mediated by the induction of antioxidant defense mechanisms, accelerated repair or altered cell cycle progression after the conditioning dose. To gain new insight into the mechanism of the adaptive response, nondividing lymphocytes and fibroblasts were used to eliminate possible contributions of cell cycle effects. The effect of conditioning doses of 0.05 or 0.1 Gy followed by challenging doses up to 8 Gy (with a 4-h interval between exposures) on induction and repair of DNA damage was determined by single-cell gel electrophoresis (comet assay), premature chromosome condensation, and immunofluorescence labeling for gamma-H2AX. The conditioning dose reduced the induction of DNA strand breaks, but the kinetics of strand break rejoining was not influenced by the conditioning dose in nondividing cells of either cell type. We conclude that adaptation in nondividing cells is not mediated by enhanced strand break rejoining and that protection against the induction of DNA damage is rather small. Therefore, the adaptive response is most likely a reflection of perturbation of cell cycle progression.


Subject(s)
DNA Damage , DNA Repair , X-Rays , Cells, Cultured , Chromosome Aberrations , Histones/metabolism , Humans , Phosphorylation
12.
Mutat Res ; 577(1-2): 170-8, 2005 Sep 04.
Article in English | MEDLINE | ID: mdl-15949822

ABSTRACT

UV-induced cyclobutane pyrimidine dimers (CPDs) are removed with accelerated speed from the transcribed strand of expressed genes in cultured mammalian cells by a process called transcription-coupled repair (TCR). It has been previously shown that this phenomenon has consequences for the molecular nature of the mutations induced by UV-light. Here, we review these data and show that TCR has not only a clear impact on UV-induced mutations in cultured mammalian cells but also on genes involved in tumor formation in the skin of UV-exposed mice. Mutations observed in the p53 gene in UV-induced squamous cell carcinoma are predominantly found at sites of dipyrimidines in the non-transcribed strand. In contrast, in UVC-irradiated Csb(-/-) Chinese hamster cells and in UVB-induced tumors in the Csb(-/-) mouse, almost all mutations are at positions of dipyrimidine sites in the transcribed strand of the mutated gene. Csb(-/-) mice appear to be susceptible to UVB-induced skin cancer in contrast to the human CSB patients. We speculate that the UVB-induced cancer susceptibility of Csb(-/-) mice is related to the absence of TCR as well as to a lack of a compensating global genome repair system for CPDs in mice.


Subject(s)
DNA Repair , Mutagenesis , Skin Neoplasms/genetics , Transcription, Genetic , Ultraviolet Rays , Animals , Cells, Cultured , Mice
13.
Toxicology ; 193(1-2): 79-90, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14599769

ABSTRACT

Nucleotide excision repair (NER) is a multistep process capable to remove a variety of DNA distorting lesions from prokaryotic and eukaryotic genomes. In eukaryotic cells, the process requires more than 30 proteins to perform the different steps, i.e. recognition of DNA damage, single strand incisions and excision of the lesion-containing DNA fragment and DNA repair synthesis/ligation. NER can operate via two subpathways: global genome repair (GGR) and a specialized pathway coupled to active transcription (transcription-coupled repair, TCR) and directed to DNA lesions in the transcribed strand of active genes. Both in vivo as well as in cultured cells the fast removal of transcription blocking lesions by TCR is crucial to escape from lethal effects of inhibited transcription inhibition The most delicate step in NER is the recognition of the DNA lesions in their different chromatin context and the mechanism of damage recognition in GGR and TCR is principally different and requires specific proteins. In GGR, the XPC-HR23B is essential for the formation of the incision complex. In TCR the Cockayne syndrome (CS) gene products are key players in the recognition of a stalled RNA polymerase the presumed signaling structure for repair of transcribed strands. In this study, we show that the extent of recovery of UV-inhibited transcription and TCR strictly depends on the amount of CSB protein as well as the amount of DNA damage present in the cell. This indicates that the ratio between DNA damage frequency and CSB protein concentration in the cell is rather critical for acute cellular response, i.e. recovery of inhibited transcription upon DNA damage infliction, and hence cellular survival.


Subject(s)
DNA Repair/genetics , Transcription, Genetic/genetics , Animals , Cockayne Syndrome/genetics , DNA Damage/genetics , Humans , Organelles/genetics
14.
Photochem Photobiol ; 88(1): 147-53, 2012.
Article in English | MEDLINE | ID: mdl-22017241

ABSTRACT

Cellular protection against deleterious effects of DNA damaging agents requires an intricate network of defense mechanisms known as the DNA damage response (DDR). Ionizing radiation (IR) mediated activation of the DDR induces a transcriptional upregulation of genes that are also involved in nucleotide excision repair (NER). This suggests that pre-exposure to X-rays might stimulate NER in human cells. Here, we demonstrate in normal human fibroblasts that UV-induced NER is augmented by pre-exposure to IR and that this increased repair is accompanied by elevated mRNA and protein levels of the NER factors XPC and DDB2. Furthermore, when IR exposure precedes local UV irradiation, the presence of XPC and DDB2 at the sites of local UV damages is increased. This increase might be p53 dependent, but the mechanism of X-ray specific stabilization of p53 is unclear as both X-rays and UV stabilize p53.


Subject(s)
DNA Repair , Fibroblasts/radiation effects , Radiation, Ionizing , Base Sequence , DNA Primers , Humans , Reverse Transcriptase Polymerase Chain Reaction
15.
J Cell Biol ; 192(3): 401-15, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21282463

ABSTRACT

Single-stranded DNA gaps that might arise by futile repair processes can lead to mutagenic events and challenge genome integrity. Nucleotide excision repair (NER) is an evolutionarily conserved repair mechanism, essential for removal of helix-distorting DNA lesions. In the currently prevailing model, NER operates through coordinated assembly of repair factors into pre- and post-incision complexes; however, its regulation in vivo is poorly understood. Notably, the transition from dual incision to repair synthesis should be rigidly synchronized as it might lead to accumulation of unprocessed repair intermediates. We monitored NER regulatory events in vivo using sequential UV irradiations. Under conditions that allow incision yet prevent completion of repair synthesis or ligation, preincision factors can reassociate with new damage sites. In contrast, replication protein A remains at the incomplete NER sites and regulates a feedback loop from completion of DNA repair synthesis to subsequent damage recognition, independently of ATR signaling. Our data reveal an important function for replication protein A in averting further generation of DNA strand breaks that could lead to mutagenic and recombinogenic events.


Subject(s)
DNA Repair , Replication Protein A/physiology , Cells, Cultured , DNA Replication , DNA, Single-Stranded/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , Genome, Human , Humans , Replication Protein A/genetics , Replication Protein A/metabolism , Ultraviolet Rays/adverse effects
16.
Radiat Res ; 175(4): 432-43, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21299404

ABSTRACT

Cockayne syndrome (CS) cells are defective in transcription-coupled repair (TCR) and sensitive to oxidizing agents, including ionizing radiation. We examined the hypothesis that TCR plays a role in ionizing radiation-induced oxidative DNA damage repair or alternatively that CS plays a role in transcription elongation after irradiation. Irradiation with doses up to 100 Gy did not inhibit RNA polymerase II-dependent transcription in normal and CS-B fibroblasts. In contrast, RNA polymerase I-dependent transcription was severely inhibited at 5 Gy in normal cells, indicating different mechanisms of transcription response to X rays. The frequency of radiation-induced base damage was 2 × 10(-7) lesions/base/Gy, implying that 150 Gy is required to induce one lesion/30-kb transcription unit; no TCR of X-ray-induced base damage in the p53 gene was observed. Therefore, it is highly unlikely that defective TCR underlies the sensitivity of CS to ionizing radiation. Overall genome repair levels of radiation-induced DNA damage measured by repair replication were significantly reduced in CS-A and CS-B cells. Taken together, the results do not provide evidence for a key role of TCR in repair of radiation-induced oxidative damages in human cells; rather, impaired repair of oxidative lesions throughout the genome may contribute to the CS phenotype.


Subject(s)
Cell Survival/radiation effects , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , DNA Damage/genetics , DNA Repair/genetics , DNA Repair/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Humans , Radiation Dosage
18.
Mol Cell ; 23(4): 471-82, 2006 08.
Article in English | MEDLINE | ID: mdl-16916636

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the editors. Molecular Cell has retracted this article following the results of an investigation carried out by Leiden University Medical Center's Committee of Scientific Integrity, which concluded that unacceptable data manipulation by the first author Maria Fousteri led to breaches of scientific integrity, making these results unreliable. These manipulations include duplications (Figures 1C, 2A, 3D [CSB panel], and 5C [p300 panel]), image tilt correction (Figure 4D [CSB panel]), and aesthetic corrections. Additional details can be found in the redacted version of the investigation report (https://www.lumc.nl/cen/att/80813053317221/1263833/report-lumc-committee-scientific-integrity).


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA Repair , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Cells, Cultured , Cross-Linking Reagents , DNA/radiation effects , DNA Damage , DNA-Binding Proteins/metabolism , Fibroblasts/radiation effects , Humans , Models, Genetic , Poly-ADP-Ribose Binding Proteins , RNA Splicing Factors , Transcription, Genetic/radiation effects , Transcriptional Elongation Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ultraviolet Rays
19.
Genes Dev ; 19(11): 1376-89, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15937223

ABSTRACT

In meiotic prophase, synaptonemal complexes (SCs) closely appose homologous chromosomes (homologs) along their length. SCs are assembled from two axial elements (AEs), one along each homolog, which are connected by numerous transverse filaments (TFs). We disrupted the mouse gene encoding TF protein Sycp1 to analyze the role of TFs in meiotic chromosome behavior and recombination. Sycp1(-/-) mice are infertile, but otherwise healthy. Sycp1(-/-) spermatocytes form normal AEs, which align homologously, but do not synapse. Most Sycp1(-/-) spermatocytes arrest in pachynema, whereas a small proportion reaches diplonema, or, exceptionally, metaphase I. In leptotene Sycp1(-/-) spermatocytes, gammaH2AX (indicative of DNA damage, including double-strand breaks) appears normal. In pachynema, Sycp1(-/-) spermatocytes display a number of discrete gammaH2AX domains along each chromosome, whereas gammaH2AX disappears from autosomes in wild-type spermatocytes. RAD51/DMC1, RPA, and MSH4 foci (which mark early and intermediate steps in pairing/recombination) appear in similar numbers as in wild type, but do not all disappear, and MLH1 and MLH3 foci (which mark late steps in crossing over) are not formed. Crossovers were rare in metaphase I of Sycp1(-/-) mice. We propose that SYCP1 has a coordinating role, and ensures formation of crossovers. Unexpectedly, Sycp1(-/-) spermatocytes did not form XY bodies.


Subject(s)
Meiosis/physiology , Nuclear Proteins/physiology , Recombination, Genetic/physiology , Synaptonemal Complex/ultrastructure , Animals , Apoptosis , Base Sequence , Crossing Over, Genetic , DNA Primers , DNA-Binding Proteins , Female , In Situ Nick-End Labeling , Infertility, Female/genetics , Infertility, Male/genetics , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Spermatocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL