Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Nucleic Acids Res ; 39(16): 6986-97, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21596788

ABSTRACT

Ataxia telangiectasia (ATM) mutated and Artemis, the proteins defective in ataxia telangiectasia and a class of Radiosensitive-Severe Combined Immunodeficiency (RS-SCID), respectively, function in the repair of DNA double strand breaks (DSBs), which arise in heterochromatic DNA (HC-DSBs) following exposure to ionizing radiation (IR). Here, we examine whether they have protective roles against oxidative damage induced and/or endogenously induced DSBs. We show that DSBs generated following acute exposure of G0/G1 cells to the oxidative damaging agent, tert-butyl hydroperoxide (TBH), are repaired with fast and slow components of similar magnitude to IR-induced DSBs and have a similar requirement for ATM and Artemis. Strikingly, DSBs accumulate in ATM(-/-) mouse embryo fibroblasts (MEFs) and in ATM or Artemis-defective human primary fibroblasts maintained for prolonged periods under confluence arrest. The accumulated DSBs localize to HC-DNA regions. Collectively, the results provide strong evidence that oxidatively induced DSBs arise in HC as well as euchromatic DNA and that Artemis and ATM function in their repair. Additionally, we show that Artemis functions downstream of ATM and is dispensable for HC-relaxation and for pKAP-1 foci formation. These findings are important for evaluating the impact of endogenously arising DNA DSBs in ATM and Artemis-deficient patients.


Subject(s)
Cell Cycle Proteins/physiology , DNA Breaks, Double-Stranded , DNA-Binding Proteins/physiology , Heterochromatin/metabolism , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Proliferation , Cellular Senescence , DNA Ligase ATP , DNA Ligases/physiology , DNA Repair , DNA-Binding Proteins/genetics , Endonucleases , Fibroblasts/enzymology , Fibroblasts/metabolism , Gene Knockdown Techniques , Histones/metabolism , Humans , Mice , Nuclear Proteins/genetics , Oxidative Stress , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Repressor Proteins/metabolism , Tripartite Motif-Containing Protein 28 , Tumor Suppressor Proteins/genetics
2.
Science ; 260(5105): 207-10, 1993 Apr 09.
Article in English | MEDLINE | ID: mdl-8469973

ABSTRACT

Cells maintain the integrity of their genome through an intricate network of repair systems that recognize and remove lesions from DNA. The only known site-directed recombination process in vertebrates is the V(D)J recombination of lymphocyte antigen receptor genes. A large panel of cell lines deficient in DNA repair were tested for the ability to perform V(D)J recombination after introduction of the RAG-1 and RAG-2 genes. Two mutants failed to generate normal V(D)J recombination, and further analysis provided evidence for two distinct nonlymphoid-specific genes that encode factors involved in both DNA repair and V(D)J recombination.


Subject(s)
DNA Repair , Gene Rearrangement, T-Lymphocyte , Genes, RAG-1 , Receptors, Antigen, T-Cell/genetics , Animals , Base Sequence , CHO Cells , Cell Line , Cricetinae , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Immunoglobulin Heavy Chains , Molecular Sequence Data , Mutation , Recombination, Genetic , VDJ Recombinases
3.
Science ; 265(5177): 1442-5, 1994 Sep 02.
Article in English | MEDLINE | ID: mdl-8073286

ABSTRACT

The radiosensitive mutant xrs-6, derived from Chinese hamster ovary cells, is defective in DNA double-strand break repair and in ability to undergo V(D)J recombination. The human XRCC5 DNA repair gene, which complements this mutant, is shown here through genetic and biochemical evidence to be the 80-kilodalton subunit of the Ku protein. Ku binds to free double-stranded DNA ends and is the DNA-binding component of the DNA-dependent protein kinase. Thus, the Ku protein is involved in DNA repair and in V(D)J recombination, and these results may also indicate a role for the Ku-DNA-dependent protein kinase complex in those same processes.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA Repair/genetics , DNA-Binding Proteins/genetics , Genes, Immunoglobulin , Nuclear Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Recombination, Genetic , Animals , Base Sequence , CHO Cells , Cell Survival/radiation effects , Cloning, Molecular , Cricetinae , DNA Damage , DNA-Binding Proteins/metabolism , Genetic Complementation Test , Humans , Hybrid Cells , Ku Autoantigen , Molecular Sequence Data , Nuclear Proteins/metabolism , Transfection
4.
Trends Biochem Sci ; 20(10): 412-5, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8533154

ABSTRACT

Two processes involving DNA double-strand breaks (DSBs) are the repair of DNA damage induced by ionizing radiation, and V(D)J recombination, the genomic rearrangement that creates antigen-receptor diversity in vertebrates. Recent evidence indicates that DNA-dependent protein kinase (DNA-PK), which is activated by DNA ends, is a central component of both the DNA DSB repair and V(D)J recombination machineries.


Subject(s)
DNA Repair , DNA-Binding Proteins , Gene Rearrangement , Protein Serine-Threonine Kinases/genetics , Recombination, Genetic , Animals , DNA , DNA Damage/radiation effects , DNA-Activated Protein Kinase , Genetic Complementation Test , Mammals/genetics , Models, Genetic , Mutation
5.
Bone Marrow Transplant ; 41(11): 983-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18278071

ABSTRACT

Several human disorders mutated in core components of the major DNA double-strand break (DSB) repair pathway, non-homologous end joining (NHEJ), have been described. Cell lines from these patients are characterized by sensitivity to DSB-inducing agents. DNA ligase IV syndrome (LIG4) patients specifically, for unknown reasons, respond particularly badly following treatment for malignancy or BMT. We report the first systematic evaluation of the response of LIG4 syndrome to compounds routinely employed for BMT conditioning. We found human pre-B lymphocytes, a key target population for BMT conditioning, when deficient for DNA ligase IV, unexpectedly exhibit significant sensitivity to CsA the principal prophylaxis for GVHD. Furthermore, we found that CsA treatment alone or in combination with BU and fludarabine resulted in increased levels of DSBs specifically in LIG4 syndrome cells compared to wild-type or Artemis-deficient cells. Our study shows that CsA can induce DSBs and that LIG4 syndrome patient's fail to adequately repair this damage. These DSBs likely arise as a consequence of DNA replication in the presence of CsA. This work has implications for BMT and GVHD management in general and specifically for LIG4 syndrome.


Subject(s)
Bone Marrow Transplantation , Cyclosporine/adverse effects , DNA Breaks, Double-Stranded/drug effects , DNA Ligases/drug effects , DNA Repair-Deficiency Disorders/chemically induced , Immunosuppressive Agents/adverse effects , Precursor Cells, B-Lymphoid/drug effects , Transplantation Conditioning/adverse effects , Cell Line , DNA Ligase ATP , DNA Ligases/deficiency , DNA Repair-Deficiency Disorders/physiopathology , Humans , Syndrome
6.
Curr Biol ; 8(2): R49-51, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9427640

ABSTRACT

Cell lines and mice defective in poly(ADP-ribose) polymerase (PARP) have elevated spontaneous genetic rearrangements and abnormal responses to stresses. These results may be explained by an altered response to damage induced by free radicals, and suggest that PARP limits genomic instability from such damage.


Subject(s)
DNA Repair , Poly(ADP-ribose) Polymerases/metabolism , Animals , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Mice , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics
7.
Curr Biol ; 9(13): 699-702, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10395545

ABSTRACT

The major mechanism for the repair of DNA double-strand breaks (DSBs) in mammalian cells is non-homologous end-joining (NHEJ), a process that involves the DNA-dependent protein kinase [1] [2], XRCC4 and DNA ligase IV [3] [4] [5] [6]. Rodent cells and mice defective in these components are radiation-sensitive and defective in V(D)J-recombination, showing that NHEJ also functions to rejoin DSBs introduced during lymphocyte development [7] [8]. 180BR is a radiosensitive cell line defective in DSB repair, which was derived from a leukaemia patient who was highly sensitive to radiotherapy [9] [10] [11]. We have identified a mutation within a highly conserved motif encompassing the active site in DNA ligase IV from 180BR cells. The mutated protein is severely compromised in its ability to form a stable enzyme-adenylate complex, although residual activity can be detected at high ATP concentrations. Our results characterize the first patient with a defect in an NHEJ component and suggest that a significant defect in NHEJ that leads to pronounced radiosensitivity is compatible with normal human viability and does not cause any major immune dysfunction. The defect, however, may confer a predisposition to leukaemia.


Subject(s)
DNA Ligases/genetics , DNA Repair , DNA-Binding Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Radiation Tolerance/genetics , Animals , Blotting, Western , Cell Line, Transformed , DNA Ligase ATP , DNA Ligases/metabolism , DNA Repair/genetics , DNA-Activated Protein Kinase , DNA-Binding Proteins/genetics , Fibroblasts/radiation effects , Humans , Mutation , Nuclear Proteins , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rabbits , Radiation, Ionizing , Sequence Analysis, DNA
8.
Trends Genet ; 14(8): 312-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9724963

ABSTRACT

Ataxia-telangiectasia (A-T) is an autosomal recessive human disorder that, because of its multisystem nature, is of interest to scientists and clinicians from many disciplines. A-T patients have defects in the neurological and immune systems, telangiectasia in the eyes and face, and are, in addition, cancer-prone and radiation-sensitive. A-T cell lines have a range of diverse phenotypes including sensitivity to ionizing radiation and defects in cell-cycle checkpoint control. The ATM protein is a member of the PI 3-kinase-like superfamily, and it has been widely accepted that A-T cells represent mammalian cell-cycle checkpoint mutants and that the radiation sensitivity is a consequence of this defect. However, several lines of evidence suggest that A-T cells have distinct repair and checkpoint defects. A-T cells therefore appear to harbour dual checkpoint/repair defects. Here, we review the evidence supporting this contention and consider its implications for an analysis of the A-T phenotype.


Subject(s)
Ataxia Telangiectasia/genetics , Protein Serine-Threonine Kinases , Proteins/genetics , Animals , Ataxia Telangiectasia/complications , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , DNA Repair/genetics , DNA-Binding Proteins , Humans , Mice , Mice, Knockout , Mice, SCID , Mutation , Neoplasms/etiology , Neoplasms/genetics , Neoplasms/metabolism , Phenotype , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Tumor Suppressor Proteins
9.
Mol Cell Biol ; 6(8): 2944-9, 1986 Aug.
Article in English | MEDLINE | ID: mdl-2431295

ABSTRACT

Six X-ray-sensitive (xrs) strains of the CHO-K1 cell line were shown to revert at a very high frequency after treatment with 5-azacytidine. This suggested that there was a methylated xrs+ gene in these strains which was structurally intact, but not expressed. The xrs strains did not complement one another, and the locus was autosomally located. In view of the frequency of their isolation and their somewhat different phenotypes, we propose that the xrs strains are mutants derived from an active wild-type gene. However, there is in addition a methylated silent gene present in the genome. Azacytidine treatment reactivated this gene. We present a model for the functional hemizygosity of mammalian cell lines, which is based on the inactivation of genes by de novo hypermethylation. In contrast to results with xrs strains, other repair-defective lines were found not to be reverted by azacytidine.


Subject(s)
Azacitidine/pharmacology , DNA Repair/drug effects , Animals , Cell Line , Cricetinae , Cricetulus , Ethyl Methanesulfonate/pharmacology , Female , Methylation , Mutation , Ovary/cytology , X-Rays
10.
Mol Cell Biol ; 19(5): 3267-77, 1999 May.
Article in English | MEDLINE | ID: mdl-10207052

ABSTRACT

Ku is a heterodimeric protein with double-stranded DNA end-binding activity that operates in the process of nonhomologous end joining. Ku is thought to target the DNA-dependent protein kinase (DNA-PK) complex to the DNA and, when DNA bound, can interact and activate the DNA-PK catalytic subunit (DNA-PKcs). We have carried out a 3' deletion analysis of Ku80, the larger subunit of Ku, and shown that the C-terminal 178 amino acid residues are dispensable for DNA end-binding activity but are required for efficient interaction of Ku with DNA-PKcs. Cells expressing Ku80 proteins that lack the terminal 178 residues have low DNA-PK activity, are radiation sensitive, and can recombine the signal junctions but not the coding junctions during V(D)J recombination. These cells have therefore acquired the phenotype of mouse SCID cells despite expressing DNA-PKcs protein, suggesting that an interaction between DNA-PKcs and Ku, involving the C-terminal region of Ku80, is required for DNA double-strand break rejoining and coding but not signal joint formation. To gain further insight into important domains in Ku80, we report a point mutational change in Ku80 in the defective xrs-2 cell line. This residue is conserved among species and lies outside of the previously reported Ku70-Ku80 interaction domain. The mutational change nonetheless abrogates the Ku70-Ku80 interaction and DNA end-binding activity.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic/genetics , Animals , CHO Cells , Cell Survival/genetics , Clone Cells/metabolism , Clone Cells/radiation effects , Cricetinae , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Enzyme Activation/genetics , Gene Expression/genetics , Humans , Ku Autoantigen , Nuclear Proteins/metabolism , Point Mutation/genetics , Sequence Deletion/genetics , Transfection
11.
Mol Cell Biol ; 17(3): 1264-73, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032253

ABSTRACT

The gene product defective in radiosensitive CHO mutants belonging to ionizing radiation complementation group 5, which includes the extensively studied xrs mutants, has recently been identified as Ku80, a subunit of the Ku protein and a component of DNA-dependent protein kinase (DNA-PK). Several group 5 mutants, including xrs-5 and -6, lack double-stranded DNA end-binding and DNA-PK activities. In this study, we examined additional xrs mutants at the molecular and biochemical levels. All mutants examined have low or undetectable levels of Ku70 and Ku80 protein, end-binding, and DNA-PK activities. Only one mutant, xrs-6, has Ku80 transcript levels detectable by Northern hybridization, but Ku80 mRNA was detectable by reverse transcription-PCR in most other mutants. Two mutants, xrs-4 and -6, have altered Ku80 transcripts resulting from mutational changes in the genomic Ku80 sequence affecting RNA splicing, indicating that the defects in these mutants lie in the Ku80 gene rather than a gene controlling its expression. Neither of these two mutants has detectable wild-type Ku80 transcript. Since the mutation in both xrs-4 and xrs-6 cells results in severely truncated Ku80 protein, both are likely candidates to be null mutants. Azacytidine-induced revertants of xrs-4 and -6 carried both wild-type and mutant transcripts. The results with these revertants strongly support our model proposed earlier, that CHO-K1 cells carry a copy of the Ku80 gene (XRCC5) silenced by hypermethylation. Site-directed mutagenesis studies indicate that previously proposed ATP-binding and phosphorylation sites are not required for Ku80 activity, whereas N-terminal deletions of more than the first seven amino acids result in severe loss of activities.


Subject(s)
Antigens, Nuclear , CHO Cells , DNA Helicases , DNA-Binding Proteins/genetics , Mutation , Nuclear Proteins/genetics , Radiation Tolerance/genetics , Animals , Azacitidine/pharmacology , CHO Cells/radiation effects , Cricetinae , DNA/metabolism , DNA, Complementary/genetics , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Gamma Rays , Gene Dosage , Genetic Complementation Test , Ku Autoantigen , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , RNA, Messenger/analysis , Sequence Analysis, DNA , Sequence Deletion
12.
Nucleic Acids Res ; 28(7): 1506-13, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10710416

ABSTRACT

DNA-PKcs, the catalytic subunit of DNA-dependent protein kinase (DNA-PK), has a phosphoinositol 3-kinase (PI 3-K) domain close to its C-terminus. Cell lines derived from the SCID mouse have been utilised as a model DNA-PKcs-defective system. The SCID mutation results in truncation of DNA-Pkcs at the extreme C-terminus leaving the PI 3-K domain intact. The mutated protein is expressed at low levels in most SCID cell lines, leaving open the question of whether the mutation abolishes kinase activity. Here, we show that a SCID cell line that expresses the mutant protein normally has dramatically impaired kinase activity. We estimate that the residual kinase activity typically present in SCID fibroblast cell lines is at least two orders of magnitude less than that found in control cells. Our results substantiate evidence that DNA-PKcs kinase activity is required for DSB rejoining and V(D)J recombination and show that the extreme C-terminal region of DNA-PKcs, present in PI 3-K-related protein kinases but absent in bona fide PI 3 lipid kinases, is required for DNA-PKcs to function as a protein kinase. We also show that expression of mutant DNA-PKcs protein confers a growth disadvantage, providing an explanation for the lack of DNA-PKcs expression in most SCID cell lines.


Subject(s)
DNA-Binding Proteins , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Animals , B-Lymphocytes/enzymology , Base Sequence , CHO Cells , Cell Line , Chromosomes, Artificial, Yeast/genetics , Conserved Sequence , Cricetinae , DNA Primers/genetics , DNA Repair/genetics , DNA Repair/physiology , DNA-Activated Protein Kinase , Hematopoietic Stem Cells/enzymology , Mice , Mice, SCID , Mutagenesis, Site-Directed , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary
13.
Cancer Res ; 61(10): 3886-93, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11358801

ABSTRACT

Two major pathways for repairing DNA double-strand breaks (DSBs) have been identified in mammalian cells, nonhomologous end-joining (NHEJ) and homologous recombination (HR). Inactivation of NHEJ is known to lead to an elevated level of spontaneous and radiation-induced chromosomal rearrangements associated with an increased risk of tumorigenesis. This has raised the idea of a caretaker role for NHEJ. It is, however, not known whether NHEJ itself can also cause rearrangements. To investigate, on the DNA level, the influence of a defect in NHEJ on the formation of genomic rearrangements, we applied an assay based on Southern hybridization that allows the identification and quantification of incorrectly rejoined DSB ends produced by ionizing radiation. After 80 Gy of X-irradiation at a high dose rate (23 Gy/min), wild-type cells repaired 50% of the induced DSBs within 24 h by incorrect rejoining. This frequency of DSB misrejoining is considerably reduced in NHEJ-deficient cells. Low-dose-rate experiments, in which the cells were exposed to 80 Gy over a period of 14 days under repair conditions, led to no detectable misrejoining in wild-type cells but revealed a misrejoining frequency of 10% in NHEJ-deficient cells. This shows that in situations of separated breaks, NHEJ deficiency leads to genomic rearrangements, in agreement with chromosomal studies. However, if multiple DSBs coincide, even wild-type cells form genomic rearrangements frequently. These repair events are absent in Ku80-, DNA-PKcs-, and DNA ligase IV-deficient cells but are present in RAD54(-/-) cells. This strongly suggests that NHEJ has, in addition to its caretaker role, also the potential to effect genomic rearrangements. We propose that it serves as an efficient pathway for rejoining correct break ends in situations of separated breaks but generates genomic rearrangements if DSBs are close in time and space.


Subject(s)
DNA Damage , DNA Repair/radiation effects , DNA-Binding Proteins , DNA/radiation effects , Saccharomyces cerevisiae Proteins , Animals , CHO Cells , Cell Line , Cricetinae , DNA/genetics , DNA/metabolism , DNA Helicases , DNA Ligase ATP , DNA Ligases/metabolism , DNA Repair/genetics , DNA Repair Enzymes , DNA-Activated Protein Kinase , Fibroblasts , Fungal Proteins/genetics , Fungal Proteins/physiology , G1 Phase/physiology , Gene Rearrangement/radiation effects , Humans , Mice , Nuclear Proteins , Protein Serine-Threonine Kinases/metabolism
14.
Cancer Res ; 49(24 Pt 1): 7057-63, 1989 Dec 15.
Article in English | MEDLINE | ID: mdl-2479475

ABSTRACT

xrs-1 is an ionizing radiation sensitive Chinese hamster ovary (CHO) strain and has a defect in double strand break rejoining. It is also highly sensitive to topoisomerase II inhibiting anticancer drugs. Sensitivity is specific for those drugs that inhibit the breakage/reunion mechanism of topoisomerase II. xrs-1 and its parent strain CHO-K1 have equal levels of topoisomerase II activity, assayed by their ability to unknot complex knotted P4 head DNA. Drug stimulated protein-DNA complex formation was similar in xrs-1 and CHO-K1, showing that they accrued equal levels of drug induced lesions. Thus sensitivity most likely results from subsequent differences in the processing of these lesions rather than the rate of formation. Drug sensitivity is directly related to the xrs phenotype since drug and gamma-ray resistance are coordinately reactivated by azacytidine treatment. All six members of the xrs complementation group are hypersensitive to etoposide. Sensitivity is not a feature common to all X-ray sensitive mutants but is shown by another complementation group, which also has a defect in double strand break rejoining. These results thus demonstrate a correlation between an inability to rejoin double strand breaks and sensitivity to topoisomerase II targeting antitumor drugs.


Subject(s)
DNA Damage , DNA Repair , DNA/genetics , Mutation , Topoisomerase II Inhibitors , Animals , Azacitidine/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Cricetinae , Cricetulus , DNA/drug effects , DNA-Binding Proteins/metabolism , Electrophoresis, Agar Gel , Female , Novobiocin/pharmacology
15.
Cancer Res ; 60(17): 4881-8, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10987302

ABSTRACT

Cells derived from Nijmegen Breakage Syndrome (NBS) patients display radiosensitivity and cell cycle checkpoint defects. Here, we examine whether the radiosensitivity of NBS cells is the result of a repair defect or whether it can be attributed to impaired checkpoint arrest. We report a small increased fraction of unrejoined double strand breaks and, more significantly, increased chromosome breaks in noncycling NBS cells at 24 h after irradiation. One of the NBS lines examined (347BR) was atypical in showing a nearly normal checkpoint response. In contrast to the mild checkpoint defect, 347BR displays marked y-ray sensitivity similar to that shown by other NBS lines. Thus, the gamma-ray sensitivity correlates with the repair defect rather than impaired checkpoint control. Taken together, the results provide direct evidence for a repair defect in NBS cells and are inconsistent with the suggestion that the radiosensitivity is attributable only to impaired checkpoint arrest. 347BR also displays elevated spontaneous damage that cannot be attributed to impaired G2-M arrest, suggesting a function of Nbsl in decreasing or limiting the impact of spontaneously arising double strand breaks.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , DNA Repair , Protein Serine-Threonine Kinases , Radiation Tolerance/physiology , Abnormalities, Multiple/metabolism , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Cell Cycle/physiology , Cell Cycle/radiation effects , Cell Line , Cell Survival/radiation effects , Checkpoint Kinase 2 , Chromosome Breakage , Chromosomes, Human/radiation effects , DNA/radiation effects , DNA Damage , Fibroblasts/pathology , Fibroblasts/radiation effects , Humans , Interphase/genetics , Mitosis/genetics , Phosphorylation , Protein Kinases/metabolism , Radiation Tolerance/genetics , Syndrome , Tumor Suppressor Protein p53/biosynthesis
16.
Cancer Res ; 55(6): 1235-8, 1995 Mar 15.
Article in English | MEDLINE | ID: mdl-7882315

ABSTRACT

The radiosensitive mutant xrs-6, derived from Chinese hamster ovary cell line CHO-K1, has been demonstrated to be defective in DNA double-strand break repair and also in its proficiency to undergo V(D)J recombination. Recent work has provided both genetic and biochemical evidence that the M(r) 80,000 subunit of the Ku protein is able to complement the radiosensitivity and the V(D)J recombination defect in the xrs-6 mutant. We demonstrate here that complementation of the radiosensitive phenotype in xrs-6 cells by the introduction of Ku80 cDNA is accompanied by the concomitant restoration of DNA double-strand break rejoining proficiency to almost that of the parental CHO-K1 cells, as measured both by neutral single-cell microgel electrophoresis (Comet) technique and by pulsed-field gel electrophoresis. These results provide further biochemical evidence for the involvement of the Ku protein in the repair of DNA double-strand breaks.


Subject(s)
Antigens, Nuclear , DNA Damage , DNA Helicases , DNA Repair , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Animals , CHO Cells , Cricetinae , DNA-Binding Proteins/physiology , Ku Autoantigen , Nuclear Proteins/physiology , Radiation Tolerance , Transfection
17.
Cancer Res ; 59(14): 3454-60, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10416610

ABSTRACT

Severe immunodeficiency characterized by lymphopenia was found in two siblings, one of whom was examined in detail. The calcium flux, pattern of tyrosine phosphorylation of proteins, and interleukin 2 (IL-2) production and proliferation in response to mitogens suggested that the peripheral blood T cells activated normally. The peripheral blood T cells were shown to have an activated phenotype with increased expression of CD45RO+ and CD95/Fas. Increased spontaneous apoptosis occurred in unstimulated lymphocyte cultures. The elevated apoptosis was not due to alterations in expression or to mutations in Bcl-2, Bcl-X(L), or Flip, nor could the spontaneous apoptosis be prevented by blocking Fas, suggesting that it was independent of Fas signaling. This is the first inherited combined immunodeficiency associated with impaired lymphocyte survival. Fibroblasts derived from the patient showed appreciable radiosensitivity in clonal assays, but apoptosis was not elevated. Our results show that the fibroblasts represent a new radiosensitive phenotype not associated with cell cycle checkpoint defects, V(D)J recombination defects, or elevated chromosome breakage. We suggest that the affected gene plays a role in an undetermined damage response mechanism that results in elevated spontaneous apoptosis in lymphoid cells and radiosensitivity in fibroblasts.


Subject(s)
Apoptosis , Fibroblasts/radiation effects , Immunologic Deficiency Syndromes/pathology , Lymphocytes/radiation effects , Severe Combined Immunodeficiency/pathology , Apoptosis/radiation effects , Child , Child, Preschool , Chromosome Inversion , Chromosomes, Human, Pair 7/ultrastructure , DNA Damage , DNA Repair , DNA, Complementary/genetics , Female , Fibroblasts/pathology , Gamma Rays , Humans , Lymphocytes/pathology , Male , Radiation Tolerance , Severe Combined Immunodeficiency/genetics , Signal Transduction/physiology , Translocation, Genetic
18.
DNA Repair (Amst) ; 3(8-9): 1227-35, 2004.
Article in English | MEDLINE | ID: mdl-15279811

ABSTRACT

Around 15-20 hereditary disorders associated with impaired DNA damage response mechanisms have been previously described. The range of clinical features associated with these disorders attests to the significant role that these pathways play during development. Recently, three new such disorders have been reported extending the importance of the damage response pathways to human health. LIG4 syndrome is conferred by hypomorphic mutations in DNA ligase IV, an essential component of DNA non-homologous end-joining (NHEJ), and is associated with pancytopaenia, developmental and growth delay and dysmorphic facial features. Radiosensitive severe combined immunodeficiency (RS-SCID) is caused by mutations in Artemis, a protein that plays a subsidiary role in non-homologous end-joining although it is not an essential component. RS-SCID is characterised by severe combined immunodeficiency but patients have no overt developmental abnormalities. ATR-Seckel syndrome is caused by mutations in ataxia telangiectasia and Rad3 related protein (ATR), a component of a DNA damage signalling pathway. ATR-Seckel syndrome patients have dramatic microcephaly and marked growth and developmental delay. The clinical features of these patients are considered in the light of the function of the defective protein.


Subject(s)
Cell Cycle Proteins/genetics , DNA Ligases/genetics , DNA Sequence, Unstable , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Protein Serine-Threonine Kinases/genetics , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , Ataxia Telangiectasia Mutated Proteins , DNA Damage , DNA Ligase ATP , DNA Repair , DNA-Binding Proteins , Endonucleases , Homozygote , Humans , Models, Biological , Models, Genetic , Mutation , Nuclear Proteins/genetics , Recombination, Genetic , Signal Transduction , Syndrome
19.
J Mol Biol ; 279(2): 375-85, 1998 Jun 05.
Article in English | MEDLINE | ID: mdl-9642044

ABSTRACT

Mutations in genes controlling the correct functioning of the replicative, repair and recombination machineries may lead to genomic instability. A high level of spontaneous chromosomal aberrations amplified by treatment with DNA cross-linking agents is the hallmark of Fanconi anemia (FA), an inherited chromosomal instability syndrome associated with cancer proneness. Two of the eight FA genes have been cloned (FAA and FAC), but their function has not yet been defined. The lack of homology with known genes suggests the involvement of FA genes in a novel pathway specific to vertebrates. Using a DNA end-joining assay in cultured cells, we studied the processing of both blunt and cohesive-ended double strand breaks (DSB) in normal and FA cells. The results show that: (i) the overall ligation efficiency is normal in FA lymphoblasts; (ii) in FA-C, error-free processing of blunt-ended DSB is markedly decreased, resulting in a higher deletion frequency and larger deletion size; (iii) the fidelity of processing of blunt-DSB is completely restored in FACC cells (complemented with wild-type FAC gene) and the deletion size shifted to values similar to that observed in normal cells; (iv) the fidelity of cohesive end-joining is not affected in FA cells; (v) activities and/or expression of known factors involved in DSB processing, such as the components of the DNA-PK complex and XRCC4, are normal in FA cells. Our results provide strong evidence that the lack of a functional FAC gene results in loss of fidelity of end-joining, which likely accounts for the FA-C phenotype of chromosome instability. We conclude that FAC, and perhaps all FA gene products, are likely to play a role in the fidelity of end-joining of specific DSB.


Subject(s)
Cell Cycle Proteins , DNA Damage/genetics , DNA-Binding Proteins , DNA/genetics , DNA/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Nuclear Proteins , Proteins/genetics , Proteins/metabolism , Amino Acid Sequence , Cell Line , Chromosome Aberrations , DNA Repair/genetics , DNA Replication , DNA-Activated Protein Kinase , Fanconi Anemia Complementation Group Proteins , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Phenotype , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Sequence Deletion , Substrate Specificity
20.
Radiat Res ; 164(1): 53-62, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15966765

ABSTRACT

We examined telomere maintenance in cells of 11 primary fibroblast cell lines with differing genetic defects that confer sensitivity to ionizing radiation. These included cell lines derived from patients with ataxia telangiectasia, Nijmegen breakage syndrome, Fanconi anemia, defective Artemis, DNA ligase I and DNA ligase IV, an immunodeficient patient with a defect in DNA double-strand break repair, and a patient diagnosed with xeroderma pigmentosum who, in addition, showed severe clinical sensitivity to ionizing radiation. Our results, based on Southern blot, flow-FISH and Q-FISH (quantitative FISH) measurements, revealed an accelerated rate of telomere shortening in most cell lines derived from the above patients compared to cell lines from normal individuals or a cell line isolated from a heterozygotic parent of one radiosensitive patient. This accelerated telomere shortening was accompanied by the formation of chromatin bridges in anaphase cells, indicative of the early loss of telomere capping function and in some cases low levels of chromosome abnormalities in metaphase cells. We also analyzed telomere maintenance in mouse embryonic stem cells deficient in Brca1, another defect that confers radiosensitivity. Similarly, these cells showed accelerated telomere shortening and mild telomere dysfunction in comparison to control cells. Our results suggest that mechanisms that confer sensitivity to ionizing radiation may be linked with mechanisms that cause telomere dysfunction.


Subject(s)
Cell Survival/genetics , Cell Survival/radiation effects , Chromosome Aberrations , Radiation Tolerance/genetics , Telomere/genetics , Animals , Cell Line , Dose-Response Relationship, Radiation , Fibroblasts/physiology , Fibroblasts/ultrastructure , Humans , Mice , Radiation Dosage , Stem Cells/physiology , Stem Cells/ultrastructure , Telomere/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL