Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters










Publication year range
1.
Clin Oncol (R Coll Radiol) ; 26(5): 243-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24630811

ABSTRACT

Substantial insight into the mechanisms responding to DNA double-strand breaks has been gained from molecular, biochemical and structural approaches. Attention is now focusing on understanding the interplay between the pathways, how they interface through the cell cycle and the communication with other DNA transactions, such as replication and transcription. Understanding these aspects will facilitate an assessment of how cancer cells have modified these processes to achieve unlimited proliferative capacity and adaptability, and pave the way to identify targets suitable for therapy. Here, we briefly overview the processes responding to double-strand breaks and discuss our current understanding of their interplay in a cellular context.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/physiology , Ataxia Telangiectasia/genetics , Cell Cycle/physiology , DNA End-Joining Repair/physiology , Homologous Recombination , Humans , Mutation , Signal Transduction/physiology
2.
Br J Radiol ; 87(1035): 20130685, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24363387

ABSTRACT

DNA double strand breaks (DSBs) are potential lethal lesions but can also lead to chromosome rearrangements, a step promoting carcinogenesis. DNA non-homologous end-joining (NHEJ) is the major DSB rejoining process and occurs in all cell cycle stages. Homologous recombination (HR) can additionally function to repair irradiation-induced two-ended DSBs in G2 phase. In mammalian cells, HR predominantly uses a sister chromatid as a template for DSB repair; thus HR functions only in late S/G2 phase. Here, we review current insight into the interplay between HR and NHEJ in G2 phase. We argue that NHEJ represents the first choice pathway, repairing approximately 80% of X-ray-induced DSBs with rapid kinetics. However, a subset of DSBs undergoes end resection and repair by HR. 53BP1 restricts resection, thereby promoting NHEJ. During the switch from NHEJ to HR, 53BP1 is repositioned to the periphery of enlarged irradiation-induced foci (IRIF) via a BRCA1-dependent process. K63-linked ubiquitin chains, which also form at IRIF, are also repositioned as well as receptor-associated protein 80 (RAP80), a ubiquitin binding protein. RAP80 repositioning requires POH1, a proteasome component. Thus, the interfacing barriers to HR, 53BP1 and RAP80 are relieved by POH1 and BRCA1, respectively. Removal of RAP80 from the IRIF core is required for loss of the ubiquitin chains and 53BP1, and for efficient replication protein A foci formation. We propose that NHEJ is used preferentially to HR because it is a compact process that does not necessitate extensive chromatin changes in the DSB vicinity.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Recombinational DNA Repair , Animals , DNA/genetics , G2 Phase/genetics , Genes, BRCA1/physiology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Proteasome Endopeptidase Complex/physiology , Trans-Activators/physiology , Tumor Suppressor p53-Binding Protein 1
3.
Cell Death Differ ; 20(8): 1089-100, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23722522

ABSTRACT

DNA double strand breaks (DSBs) are the most common form of DNA damage and are repaired by non-homologous-end-joining (NHEJ) or homologous recombination (HR). Several protein components function in NHEJ, and of these, DNA Ligase IV is essential for performing the final 'end-joining' step. Mutations in DNA Ligase IV result in LIG4 syndrome, which is characterised by growth defects, microcephaly, reduced number of blood cells, increased predisposition to leukaemia and variable degrees of immunodeficiency. In this manuscript, we report the creation of a human induced pluripotent stem cell (iPSC) model of LIG4 deficiency, which accurately replicates the DSB repair phenotype of LIG4 patients. Our findings demonstrate that impairment of NHEJ-mediated-DSB repair in human iPSC results in accumulation of DSBs and enhanced apoptosis, thus providing new insights into likely mechanisms used by pluripotent stem cells to maintain their genomic integrity. Defects in NHEJ-mediated-DSB repair also led to a significant decrease in reprogramming efficiency of human cells and accumulation of chromosomal abnormalities, suggesting a key role for NHEJ in somatic cell reprogramming and providing insights for future cell based therapies for applications of LIG4-iPSCs. Although haematopoietic specification of LIG4-iPSC is not affected per se, the emerging haematopoietic progenitors show a high accumulation of DSBs and enhanced apoptosis, resulting in reduced numbers of mature haematopoietic cells. Together our findings provide new insights into the role of NHEJ-mediated-DSB repair in the survival and differentiation of progenitor cells, which likely underlies the developmental abnormalities observed in many DNA damage disorders. In addition, our findings are important for understanding how genomic instability arises in pluripotent stem cells and for defining appropriate culture conditions that restrict DNA damage and result in ex vivo expansion of stem cells with intact genomes.


Subject(s)
DNA End-Joining Repair/physiology , DNA Ligases/deficiency , Genomic Instability/physiology , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Apoptosis/physiology , Cell Line , Cell Survival/physiology , Cells, Cultured , DNA Ligase ATP , DNA Ligases/physiology , Hematopoietic Stem Cells/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Phenotype , Tumor Suppressor Protein p53/physiology , Up-Regulation/physiology
4.
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
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
7.
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
8.
Clin Immunol ; 113(2): 214-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15451479

ABSTRACT

Fanconi anemia (FA), an autosomal recessive chromosomal instability syndrome, is characterized clinically by developmental abnormalities, growth retardation, progressive bone marrow failure, pancytopenia, and pronounced cancer predisposition. Nijmegen Breakage Syndrome (NBS) is a related disorder that shares overlapping clinical features, principally, developmental delay, microcephaly, and cancer predisposition. The diagnosis has relied on chromosomal instability following exposure to DNA cross-linking agents in FA and to ionizing radiation (IR) in NBS. We describe two patients who clinically had FA, but showed sensitivity to both DNA cross-linking agents and ionizing radiation, and who were found to have a rare mutation in the NBS gene. The importance of genetic diagnosis with respect to treatment and prognosis is discussed.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/immunology , Cell Cycle Proteins/genetics , Chromosome Breakage/genetics , Fanconi Anemia/genetics , Nuclear Proteins/genetics , Abnormalities, Multiple/pathology , Blotting, Western , Chromosome Breakage/immunology , Diagnosis, Differential , Fanconi Anemia/immunology , Fanconi Anemia/physiopathology , Female , Humans , Immunoglobulins/blood , Infant, Newborn , Lymphocytes/immunology , Male , Mutation , Phenotype , Receptors, Antigen, T-Cell/genetics
9.
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
10.
Proc Natl Acad Sci U S A ; 100(5): 2462-7, 2003 Mar 04.
Article in English | MEDLINE | ID: mdl-12604777

ABSTRACT

DNA nonhomologous end-joining (NHEJ) is the major pathway for repairing DNA double-strand breaks in mammalian cells. It also functions to carry out rearrangements at the specialized breaks introduced during V(D)J recombination. Here, we describe a patient with T(-)B(-) severe combined immunodeficiency, whose cells have defects closely resembling those of NHEJ-defective rodent cells. Cells derived from this patient show dramatic radiosensitivity, decreased double-strand break rejoining, and reduced fidelity in signal and coding joint formation during V(D)J recombination. Detailed examination indicates that the patient is defective neither in the known factors involved in NHEJ in mammals (Ku70, Ku80, DNA-dependent protein kinase catalytic subunit, Xrcc4, DNA ligase IV, or Artemis) nor in the Mre11/Rad50/Nbs1 complex, whose homologue in Saccharomyces cerevisiae functions in NHEJ. These results provide strong evidence that additional activities are crucial for NHEJ and V(D)J recombination in mammals.


Subject(s)
DNA Nucleotidyltransferases/chemistry , DNA Repair , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Animals , Catalytic Domain , Cells, Cultured , DNA Damage , DNA Ligase ATP , DNA Ligases/metabolism , DNA Nucleotidyltransferases/metabolism , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Radiation , Fibroblasts/metabolism , Humans , Immunoblotting , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured , VDJ Recombinases
11.
J Med Genet ; 39(12): 900-5, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12471202

ABSTRACT

The DNA double stranded break (DSB) repair mechanism, non-homologous end joining (NHEJ) represents an essential step in antigen receptor gene rearrangement mechanisms, processes believed to be intimately involved in the aetiology of lymphoproliferative disease. We investigated the potential impact that previously undescribed polymorphisms identified within NHEJ DNA ligase IV (LIG4) have upon predisposition to several lymphoproliferative disorders, including leukaemia, lymphoma, and multiple myeloma. Two LIG4 polymorphisms were examined, both C>T transitions, which result in the amino acid substitutions A3V and T9I. Inheritance of the LIG4 A3V CT genotype was found to be significantly associated with a two-fold reduction in risk of developing multiple myeloma (OR 0.49, 95% CI 0.27 to 0.89). Similarly, inheritance of the LIG4 T9I CT and the T9I TT genotypes were found to associate with a 1.5-fold reduction (OR 0.77, 95% CI 0.51 to 1.17) and a four-fold reduction (OR 0.22, 95% CI 0.07 to 0.70) in risk of developing multiple myeloma respectively, suggesting a gene dosage effect for this polymorphism. The LIG4 A3V and T9I variant alleles are in linkage disequilibrium (D'=0.95, p<0.0001), and the protective effect associated with these polymorphisms was found to be the result of inheritance of the A3V-T9I CT and A3V-T9I TT haplotypes. These data suggest that genetic variants of NHEJ LIG4 may modulate predisposition to multiple myeloma, a tumour characterised by aberrant immunoglobulin (Ig) class switch recombination.


Subject(s)
DNA Ligases/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Immunoglobulin Class Switching/genetics , Multiple Myeloma/genetics , Polymorphism, Genetic/genetics , Base Sequence , Case-Control Studies , DNA Ligase ATP , Gene Frequency , Haplotypes/genetics , Humans , Linkage Disequilibrium/genetics , Molecular Sequence Data , Odds Ratio
12.
Radiat Prot Dosimetry ; 99(1-4): 117-22, 2002.
Article in English | MEDLINE | ID: mdl-12194262

ABSTRACT

Ionising radiation (IR) induces a range of DNA damage similar to that which arises endogenously from reactive oxygen species generated as by-products of metabolism. However, due to non-homogeneous energy deposition, the damage from IR frequently occurs in clusters producing unique 'complex' lesions. Cells have evolved a range of mechanisms to respond to DNA damage, which include pathways of DNA repair and processes that prevent the proliferation of damaged cells. However, the repair mechanisms are not fool proof and clustered radiation-induced lesions pose a particular problem. Whether DNA damage created by IR can be repaired accurately, mis-repaired or not repaired at all is of utmost importance in considering the impact of radiation exposure. Here, the current knowledge is discussed of the repair of double strand breaks, a biologically important lesion induced by IR, in the context of the fidelity of the repair mechanisms and the consequences of mis-repair or lack of repair.


Subject(s)
DNA Damage/radiation effects , DNA Repair/radiation effects , Cell Cycle/radiation effects , Models, Molecular , Nucleic Acid Conformation , Radiation, Ionizing , Reactive Oxygen Species/radiation effects , Reproducibility of Results
13.
Radiat Res ; 156(2): 167-76, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11448237

ABSTRACT

The DNA-dependent protein kinase (DNA-PK), comprised of the Ku70/Ku80 (now known as G22p1/Xrcc5) heterodimer and the catalytic subunit DNA-PKcs (now known as Prkdc), is required for the nonhomologous end joining (NHEJ) pathway of DNA double-strand break repair. The mechanism of action of DNA-PK remains unclear. We have investigated whether DNA-PK regulates gene transcription in vivo after DNA damage using the subtractive hybridization technique of cDNA representational difference analysis (cDNA RDA). Differential transcription, both radiation-dependent and independent, was detected and confirmed in primary mouse embryo fibroblasts from DNA-PKcs(-/-) and DNA-PKcs(+/+) mice. We present evidence that transcription of the extracellular matrix gene laminin alpha 4 (Lama4) is regulated by DNA-PK in a radiation-independent manner. However, screening of both primary and immortalized DNA-PKcs-deficient cell lines demonstrates that the majority of differences were not consistently dependent on DNA-PK status. Similar results were obtained in experiments using KU mutant hamster cell lines, indicating heterogeneity of transcription between closely related cell lines. Our results suggest that while DNA-PK may be involved in limited gene-specific transcription, it does not play a major role in the transcriptional response to DNA damage.


Subject(s)
Antigens, Nuclear , DNA Helicases , Protein Serine-Threonine Kinases/deficiency , Transcription, Genetic , 3T3 Cells , Animals , CHO Cells , Cells, Cultured , Cricetinae , DNA Damage , DNA Repair , DNA, Complementary/genetics , DNA, Complementary/radiation effects , DNA-Activated Protein Kinase , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Ku Autoantigen , Laminin/genetics , Mice , Mice, Knockout , Mutation , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
14.
Radiother Oncol ; 60(1): 75-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11410307

ABSTRACT

BACKGROUND: Fanconi anaemia is a rare disease associated with cellular sensitivity to chemicals (e.g. mitomycin C and diepoxybutane); variable but mild cellular radiosensitivity has also been reported. MATERIALS AND METHODS: A 32-year-old patient with Fanconi anaemia and tonsillar carcinoma, treated by radiotherapy, was found to exhibit profound clinical radiosensitivity. Confluent, ulcerating oropharyngeal mucositis developed after a conventionally fractionated dose of 34Gy and healing was incomplete by 2 months after cessation of therapy. RESULTS: Cellular radiosensitivity assays and RPLD studies from this patient did not suggest any major detectable radiosensitivity. CONCLUSION: There is a discrepancy between the observed clinical radiosensitivity and the usual "predictive" radiosensitivity assays in this patient with Fanconi anaemia.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Fanconi Anemia/complications , Radiation Tolerance , Tonsillar Neoplasms/radiotherapy , Adult , Carcinoma, Squamous Cell/complications , Cells, Cultured , Fanconi Anemia/genetics , Female , Fibroblasts/radiation effects , Humans , Radiation Tolerance/genetics , Tonsillar Neoplasms/complications
15.
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
16.
J Biol Chem ; 276(33): 31124-32, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11349135

ABSTRACT

DNA ligase IV functions in DNA non-homologous end-joining, in V(D)J recombination, and during brain development. We previously reported a homozygous mutation (R278H) in DNA ligase IV in a developmentally normal leukemia patient who overresponded to radiotherapy. The impact of this hypomorphic mutation has been evaluated using cellular, biochemical, and structural approaches. Structural modeling using T7 DNA ligase predicts that the activity and conformational stability of the protein is likely to be impaired. We show that wild type DNA ligase IV-Xrcc4 is an efficient double-stranded ligase with distinct optimal requirements for adenylate complex formation versus rejoining. The mutation impairs the formation of an adenylate complex as well as reducing the rejoining activity. Additionally, it imparts temperature-sensitive activity to the protein consistent with the predictions of the structural modeling. At the cellular level, the mutation confers a unique V(D)J recombination phenotype affecting the fidelity of signal joint formation with little effect on the frequency of the reaction. These findings suggest that hypomorphic mutations in ligase IV may allow normal development but confer marked radiosensitivity.


Subject(s)
DNA Ligases/chemistry , Mutation , Radiation Tolerance , Adenosine Monophosphate/metabolism , Cell Line , DNA/metabolism , DNA Ligase ATP , DNA Ligases/genetics , DNA Repair , Humans , Models, Structural , Recombination, Genetic , Structure-Activity Relationship , Temperature
17.
EMBO J ; 20(1-2): 210-21, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11226171

ABSTRACT

DNA double strand break (DSB) repair by non-homologous end joining (NHEJ) in mammalian cells requires the Ku70-Ku80 heterodimer, the DNA-PK catalytic subunit DNA-PKcs, as well as DNA ligase IV and Xrcc4. NHEJ of plasmid DSBs in Saccharomyces cerevisiae requires Ku, Xrcc4 and DNA ligase IV, as well as Mre11, Rad50, Xrs2 and DNA damage checkpoint proteins. Saccharomyces cerevisiae Ku is also required for telomere length maintenance and transcriptional silencing. We have characterized NHEJ in Schizosaccharomyces pombe using an extrachromosomal assay and find that, as anticipated, it is Ku70 and DNA ligase IV dependent. Unexpectedly, we find that Rad32, Rad50 (the S.pombe homologues of Mre11 and Rad50, respectively) and checkpoint proteins are not required for NHEJ. Furthermore, although S.pombe Ku70 is required for maintenance of telomere length, it is dispensable for transcriptional silencing at telomeres and is located throughout the nucleus rather than concentrated at the telomeres. Together, these results provide insight into the mechanism of NHEJ and contrast significantly with recent studies in S.cerevisiae.


Subject(s)
Antigens, Nuclear , DNA Damage , DNA Helicases , DNA Ligases/metabolism , DNA Repair , DNA, Fungal/genetics , DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Schizosaccharomyces/genetics , Animals , Base Sequence , Bleomycin/pharmacology , Cell Nucleus/genetics , Cell Nucleus/physiology , DNA Ligase ATP , DNA, Fungal/metabolism , Gamma Rays , Gene Silencing , Ku Autoantigen , Mammals , Molecular Sequence Data , Restriction Mapping , Schizosaccharomyces/drug effects , Schizosaccharomyces/radiation effects , Telomere/genetics , Telomere/physiology , Temperature , Transcription Factors/metabolism
18.
J Photochem Photobiol B ; 65(2-3): 88-96, 2001 Dec 31.
Article in English | MEDLINE | ID: mdl-11809363

ABSTRACT

DNA damage response mechanisms serve to protect cells from exogenous and endogenous DNA damaging agents with the aim of maintaining genomic stability. In contrast, the generation of an efficient immune response requires the creation of a repertoire of distinct immunoglobulin and T cell receptor genes able to recognise the huge array of antigens that may be encountered in a lifetime. Surprisingly, cells have exploited the same mechanisms used to maintain genomic integrity to create genetic diversity during immune development. Here, we review the damage response mechanisms operating on DNA double strand breaks and their function during development of the immune response. We discuss disorders that are associated with immunodeficiency and defective responses to the presence of DNA double strand breaks.


Subject(s)
DNA Damage/immunology , DNA Repair/immunology , Animals , Genetic Variation , Genome , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology
19.
Mol Cell ; 8(6): 1175-85, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11779494

ABSTRACT

DNA ligase IV functions in DNA nonhomologous end-joining and V(D)J recombination. Four patients with features including immunodeficiency and developmental and growth delay were found to have mutations in the gene encoding DNA ligase IV (LIG4). Their clinical phenotype closely resembles the DNA damage response disorder, Nijmegen breakage syndrome (NBS). Some of the mutations identified in the patients directly disrupt the ligase domain while others impair the interaction between DNA ligase IV and Xrcc-4. Cell lines from the patients show pronounced radiosensitivity. Unlike NBS cell lines, they show normal cell cycle checkpoint responses but impaired DNA double-strand break rejoining. An unexpected V(D)J recombination phenotype is observed involving a small decrease in rejoining frequency coupled with elevated imprecision at signal junctions.


Subject(s)
DNA Ligases/genetics , Developmental Disabilities/genetics , Immunologic Deficiency Syndromes/genetics , Mutation/genetics , Nuclear Proteins , Cell Cycle , Cell Cycle Proteins/physiology , Cells, Cultured , Child , Chromosome Breakage/genetics , DNA Damage/genetics , DNA Ligase ATP , DNA Ligases/metabolism , DNA Mutational Analysis , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Developmental Disabilities/enzymology , Fibroblasts , Gene Rearrangement/genetics , Genetic Complementation Test , Humans , Immunologic Deficiency Syndromes/enzymology , Middle Aged , Phenotype , Protein Binding , Radiation Tolerance/genetics , Recombinant Proteins/metabolism , Recombination, Genetic/genetics , Syndrome , Transfection
20.
Nucleic Acids Res ; 28(23): 4778-82, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11095690

ABSTRACT

DNA non-homologous end joining, the major mechanism for the repair of DNA double-strands breaks (DSB) in mammalian cells requires the DNA-dependent protein kinase (DNA-PK), a complex composed of a large catalytic subunit of 460 kDa (DNA-PKcs) and the heterodimer Ku70-Ku80 that binds to double-stranded DNA ends. Mutations in any of the three subunits of DNA-PK lead to extreme radiosensitivity and DSB repair deficiency. Here we show that the 283 C-terminal amino acids of Ku80 introduced into the Chinese hamster ovary cell line CHO-K1 have a dominant negative effect. Expression of Ku(449-732) in CHO cells was verified by northern blot analysis and resulted in decreased Ku-dependent DNA end-binding activity, a diminished capacity to repair DSBs as determined by pulsed field gel electrophoresis and decreased radioresistance determined by clonogenic survival. The stable modifications observed at the molecular and cellular level suggest that this fragment of Ku80 confers a dominant negative effect providing an important mechanism to sensitise radioresistant cells.


Subject(s)
Antigens, Nuclear , Cell Division/radiation effects , DNA Helicases , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Animals , CHO Cells , Cell Division/genetics , Cell Survival/genetics , Cell Survival/radiation effects , Cricetinae , DNA Repair , DNA, Recombinant , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dose-Response Relationship, Radiation , Gene Expression , Ku Autoantigen , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oligonucleotides/metabolism , Phenotype , Plasmids/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...