Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Res ; 61(6): 2649-55, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289143

ABSTRACT

DNA double-strand breaks (DSBs) can be induced by a number of endogenous and exogenous agents and are lethal events if left unrepaired. DNA DSBs can be repaired by homologous recombination (HR) and nonhomologous end joining (NHEJ). In mammals and higher eukaryotes, NHEJ is thought to be the primary pathway for repair, but the role for each pathway in DNA DSB repair has not been fully elucidated. To define the relative contributions of HR and NHEJ in mammalian DNA DSB repair, cells defective in both pathways were produced. Double-mutant cells were created by expressing a dominant mutant hRAD54 protein in a DNA-dependent protein kinase (DNA-PK)-deficient severe combined immunodeficient cell line. Double-mutant cells demonstrate an increase in ionizing radiation sensitivity and a decrease in DNA DSB repair as compared with either single mutant, whereas single-mutant hRAD54 cells exhibit a wild-type phenotype. Unexpectedly, DNA-PK-null cells were more resistant to mitomycin-C damage than were wild-type cells. Chromosome aberration analysis reveals numerous incomplete chromatid exchange aberrations in the majority of the double-mutant cells after ionizing radiation exposure. Our findings confirm a role for HR in DSB repair in higher eukaryotes, yet indicate that its role is not evident unless the primary repair pathway, NHEJ, is nonfunctional. Mitomycin-C resistance in DNA-PK-null cells compared with wild-type cells suggests that the HR pathway may be more efficient in cross-link repair in the absence of NHEJ. Lastly, the incorrectly repaired chromatid damage observed in double-mutant cells may result from failed recombination or another error-prone repair process that is apparent in the absence of the two primary repair pathways.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins , Nuclear Proteins/genetics , Alkylating Agents/toxicity , Animals , Chromosome Aberrations/genetics , Cricetinae , DNA Damage , DNA Helicases , DNA, Complementary/drug effects , DNA, Complementary/genetics , DNA, Complementary/metabolism , DNA-Activated Protein Kinase , Humans , Mice , Mice, SCID , Mitomycin/toxicity , Nuclear Proteins/biosynthesis , Point Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic/genetics , Sister Chromatid Exchange/genetics , Transfection
2.
Curr Biol ; 10(15): 886-95, 2000.
Article in English | MEDLINE | ID: mdl-10959836

ABSTRACT

BACKGROUND: The response of eukaryotic cells to double-strand breaks in genomic DNA includes the sequestration of many factors into nuclear foci. Recently it has been reported that a member of the histone H2A family, H2AX, becomes extensively phosphorylated within 1-3 minutes of DNA damage and forms foci at break sites. RESULTS: In this work, we examine the role of H2AX phosphorylation in focus formation by several repair-related complexes, and investigate what factors may be involved in initiating this response. Using two different methods to create DNA double-strand breaks in human cells, we found that the repair factors Rad50 and Rad51 each colocalized with phosphorylated H2AX (gamma-H2AX) foci after DNA damage. The product of the tumor suppressor gene BRCA1 also colocalized with gamma-H2AX and was recruited to these sites before Rad50 or Rad51. Exposure of cells to the fungal inhibitor wortmannin eliminated focus formation by all repair factors examined, suggesting a role for the phosphoinositide (PI)-3 family of protein kinases in mediating this response. Wortmannin treatment was effective only when it was added early enough to prevent gamma-H2AX formation, indicating that gamma-H2AX is necessary for the recruitment of other factors to the sites of DNA damage. DNA repair-deficient cells exhibit a substantially reduced ability to increase the phosphorylation of H2AX in response to ionizing radiation, consistent with a role for gamma-H2AX in DNA repair. CONCLUSIONS: The pattern of gamma-H2AX foci that is established within a few minutes of DNA damage accounts for the patterns of Rad50, Rad51, and Brca1 foci seen much later during recovery from damage. The evidence presented strongly supports a role for the gamma-H2AX and the PI-3 protein kinase family in focus formation at sites of double-strand breaks and suggests the possibility of a change in chromatin structure accompanying double-strand break repair.


Subject(s)
DNA Damage , DNA Repair Enzymes , DNA Repair , Histones/metabolism , Acid Anhydride Hydrolases , Androstadienes/pharmacology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Bromodeoxyuridine/pharmacology , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Gamma Rays , Humans , Lasers , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Rad51 Recombinase , Tumor Cells, Cultured , Ultraviolet Rays , Wortmannin
3.
Radiat Res ; 153(2): 125-30, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10629611

ABSTRACT

M059J is a radiosensitive cell line established from a human glioblastoma tumor that fails to express the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs, now known as PRKDC). Another cell line, M059K, established from the same tumor is radioresistant. Neither M059J nor M059K cells have been fully characterized, beyond the lack of expression of PRKDC and low expression of ATM in M059J cells. To determine whether its radiosensitive phenotype is due to a defect in the gene that encodes PRKDC, we show here that M059J cells can be complemented with the PRKDC gene by introducing a fragment of human chromosome 8 containing a copy of the human PRKDC gene. Two hybrid cell lines that retain an extra copy of PRKDC display active kinase activity and are radioresistant, demonstrating that the primary defect in M059J cells is in PRKDC. In addition, these cell lines derived from M059J cells provide us with a closer genetic match to M059J than M059K cells in studies to elucidate the function of DNA-PK.


Subject(s)
DNA-Binding Proteins , Genetic Complementation Test , Protein Serine-Threonine Kinases/genetics , Radiation Tolerance/genetics , Animals , Cell Line , Chromosomes, Human, Pair 8 , DNA-Activated Protein Kinase , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Mice , Mice, SCID , Nuclear Proteins , Phenotype
4.
Cancer Res ; 58(11): 2316-22, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9622065

ABSTRACT

Nijmegen breakage syndrome (NBS), which in the past also has been classified as a variant of ataxia telangiectasia (AT), is characterized by cancer proneness and extreme sensitivity to ionizing radiation. We investigated the DNA damage responses of four independent primary NBS fibroblast cell lines. Following a low dose of ionizing radiation, p53 is mostly induced with slower kinetics and shows more transient induction in NBS fibroblasts. Nonetheless, this damage-induced protein appears biologically functional: unsynchronized and synchronized NBS cells show a G1 arrest after ionizing radiation as determined by bivariate flow cytometry. Neither an AT cell line nor a NBS cell line transformed with human papillomavirus genes E6 and E7 shows a G1 arrest. Furthermore, NBS cells show a normal G2 block, unlike that shown for AT cells. These data provide a cellular distinction between NBS and AT, thereby clearly separating the NBS from the AT syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Cell Cycle/radiation effects , Chromosome Breakage , Ataxia Telangiectasia/genetics , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/radiation effects , Flow Cytometry , G1 Phase/radiation effects , G2 Phase/radiation effects , Humans , Microcephaly/genetics , Radiation Tolerance/genetics , Syndrome , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects
5.
Radiat Res ; 149(5): 455-62, 1998 May.
Article in English | MEDLINE | ID: mdl-9588356

ABSTRACT

Repair of DNA double-strand breaks (DSBs) is linked to the V(D)J recombination pathway through investigations of radiation-sensitive mutants. Here we report a possible association between the distribution of DSBs within higher-order chromatin structures and this pathway. Both murine severe combined immunodeficient (SCID) and Chinese hamster XR-1 cells exhibit defective DNA DSB repair and defective V(D)J recombination. The DSB repair defect is not complete, with only a subset of slowly repairing lesions affected by the mutations in these cell lines. We used a modified neutral filter elution procedure which retained elements of higher-order chromatin structures, namely nuclear matrix-DNA interactions. X-ray-induced DSBs that occurred as multiples within looped DNA structures were nonrepairable in SCID and XR-1 cells. In contrast, these lesions were repaired in radioresistant wild-type cells. Cell lines complemented with human DNA containing the respective complementing genes (XRCC7 and XRCC4) showed an increased rate of DSB repair. These results agree with previous findings with xrs5 cells (a member of the XRCC5 group). Xrs5 cells are defective for the Ku p80 subunit of the V(D)J recombination complex and show repair and V(D)J recombination defects similar to those of SCID and XR-1 cells.


Subject(s)
Chromatin/chemistry , DNA Damage , DNA Repair , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/genetics , Recombination, Genetic , Animals , Cricetinae , Cricetulus , Humans , Kinetics , Mice , Mice, SCID , Radiation Tolerance
6.
Proc Natl Acad Sci U S A ; 93(24): 13825-30, 1996 Nov 26.
Article in English | MEDLINE | ID: mdl-8943020

ABSTRACT

Severe combined immunodeficient (SCID) mice display an increased sensitivity to ionizing radiation compared with the parental, C.B-17, strain due to a deficiency in DNA double-strand break repair. The catalytic subunit of DNA-dependent protein kinase (DNA-PKCS) has previously been identified as a strong candidate for the SCID gene. DNA-PK phosphorylates many proteins in vitro, including p53 and replication protein A (RPA), two proteins involved in the response of cells of DNA damage. To determine whether p53 and RPA are also substrates of DNA-PK in vivo following DNA damage, we compared the response of SCID and MO59J (human DNA-PKcs-deficient glioblastoma) cells with their respective wild-type parents following ionizing radiation. Our findings indicate that (i) p53 levels are increased in SCID cells following ionizing radiation, and (ii) RPA p34 is hyperphosphorylated in both SCID cells and MO59J cells following ionizing radiation. The hyperphosphorylation of RPA p34 in vivo is concordant with a decrease in the binding of RPA to single-stranded DNA in crude extracts derived from both C.B-17 and SCID cells. These results suggest that DNA-PK is not the only kinase capable of phosphorylating RPA. We conclude that the DNA damage response involving p53 and RPA is not associated with the defect in DNA repair in SCID cells and that the physiological substrate(s) for DNA-PK essential for DNA repair has not yet been identified.


Subject(s)
DNA Damage , DNA Replication , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/deficiency , Tumor Suppressor Protein p53/biosynthesis , Animals , Cell Line , DNA-Activated Protein Kinase , Fibroblasts/physiology , Fibroblasts/radiation effects , Glioblastoma , Humans , Mice , Mice, SCID , Nuclear Proteins , Oligodeoxyribonucleotides , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Replication Protein A , Substrate Specificity , Tumor Cells, Cultured
7.
Radiat Res ; 145(1): 39-46, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8532835

ABSTRACT

Severe combined immunodeficient (scid) murine cells, which are defective in both repair of DNA double-strand breaks and V(D)J recombination, are deficient in DNA-dependent protein kinase (DNA-PK), a protein which forms an activated complex with the DNA end-binding Ku proteins (p80 and p70) upon association with damaged DNA. Xrs 5 cells are deficient in the Ku p80 protein and also fail to form an active DNA-PK repair complex. Since both scid and xrs cells are defective in the same protein complex, we compared the kinetics of chromosome repair in scid cells to results published previously for xrs 5 cells. C.B-17 cells, scid cells and scid cells complemented with a single human chromosome 8 were irradiated with 6 Gy and allowed to repair from 0-24 h before fusion to HeLa cells for chromosome condensation. Breaks and dicentrics were visualized by fluorescence in situ hybridization. All cells had the same initial amount of chromosome damage, but scid cells had a slower rate of rejoining, more unrejoined breaks and more dicentrics than C.B-17 and scid cells with human chromosome 8. The scid cells appear to respond differently than xrs 5 cells, despite both cells lacking an essential component of the same DNA repair complex.


Subject(s)
Antigens, Nuclear , Chromosome Aberrations , Chromosomes, Human, Pair 8 , Chromosomes/radiation effects , DNA Damage , DNA Helicases , DNA Repair , Severe Combined Immunodeficiency/genetics , Algorithms , Animals , Cell Fusion , Cell Line , Chromosomes/physiology , DNA Nucleotidyltransferases/deficiency , DNA Nucleotidyltransferases/genetics , DNA-Activated Protein Kinase , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Kinetics , Ku Autoantigen , Mice , Mice, SCID , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Radiation Tolerance , Transcription Factors/metabolism , VDJ Recombinases
8.
J Med Genet ; 32(12): 925-9, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8825916

ABSTRACT

X chromosome inactivation has been hypothesised to play a role in the aetiology and clinical expression of the fragile X syndrome. The identification of the FMR1 gene involved in fragile X syndrome allows testing of the assumption that the fragile X locus is normally subject to X inactivation. We studied the expression of the FMR1 gene from inactive X chromosomes by reverse transcription of RNA followed by PCR (RT-PCR), both in somatic cell hybrids which retain an active or inactive human X chromosome and in a female patient with a large deletion surrounding the FMR1 gene. In both analyses, the data indicate that FMR1 is not normally expressed from the inactive X chromosome and is, therefore, subject to X chromosome inactivation. This finding is consistent with the results of previous studies of DNA methylation of FMR1 on active and inactive X chromosomes, verifies previous assumptions about the fragile X locus, and supports the involvement of X inactivation in the variable phenotype of females with full mutations of the FMR1 gene.


Subject(s)
Dosage Compensation, Genetic , Fragile X Syndrome/genetics , Nerve Tissue Proteins/genetics , RNA-Binding Proteins , Animals , Base Sequence , DNA Primers , Fragile X Mental Retardation Protein , Humans , Hybrid Cells , Mice , Molecular Sequence Data , Polymerase Chain Reaction , X Chromosome/genetics
9.
Science ; 267(5201): 1178-83, 1995 Feb 24.
Article in English | MEDLINE | ID: mdl-7855601

ABSTRACT

Severe combined immunodeficient (SCID) mice are deficient in a recombination process utilized in both DNA double-strand break repair and in V(D)J recombination. The phenotype of these mice involves both cellular hypersensitivity to ionizing radiation and a lack of B and T cell immunity. The catalytic subunit of DNA-dependent protein kinase, p350, was identified as a strong candidate for the murine gene SCID. Both p350 and a gene complementing the SCID defect colocalize to human chromosome 8q11. Chromosomal fragments expressing p350 complement the SCID phenotype, and p350 protein levels are greatly reduced in cells derived from SCID mice compared to cells from wild-type mice.


Subject(s)
DNA-Binding Proteins , Protein Serine-Threonine Kinases/genetics , Severe Combined Immunodeficiency/genetics , Animals , Base Sequence , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 8 , Cloning, Molecular , DNA Repair/genetics , DNA-Activated Protein Kinase , Gamma Rays , Genetic Complementation Test , Genetic Markers , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, SCID , Molecular Sequence Data , Nuclear Proteins , Phenotype , Protein Serine-Threonine Kinases/metabolism , Radiation Tolerance , Recombination, Genetic , Severe Combined Immunodeficiency/enzymology
10.
Cancer Res ; 53(24): 6011-6, 1993 Dec 15.
Article in English | MEDLINE | ID: mdl-8261415

ABSTRACT

Severe combined immunodeficient (scid) C.B-17 mice are deficient in variable (diversity) joining region recombination, the process of assembling the immunoglobulin and T-cell receptor genes from gene segments, thereby creating much of the enormous diversity of antigen-binding capacity, scid mice are also sensitive to ionizing radiation, as a result of their deficiency in double-strand break repair. Here we report the complementation of the radiation-sensitive scid phenotype by transferring human chromosome 8 into scid cells. Somatic cell hybrids were generated by fusing scid cells with human HT-1080 cells, resulting in radioresistant hybrids with several human chromosomes. One of the identified human chromosomes in the radioresistant scid cell line 4.61, which retains only two human chromosomes, is a rearranged 8/21 translocation. Proof that chromosome 8 confers the complementation was achieved by transferring only human chromosome 8 into scid cells by microcell-mediated chromosome transfer (scid/hu8 cell line). The presence of chromosome 8 in our scid/hu8 cell line was monitored by fluorescence in situ hybridization and polymerase chain reaction. We demonstrated the radioresistance of this hybrid not only to high dose rate but also to low dose rate radiation. We also showed that transference of human chromosome 8 to scid cells fully complements the DNA double-strand break repair deficiency and the high sensitivity of scid cells to radiation-induced chromosome aberrations. Mapping the scid gene to human chromosome 8 is an important first step in cloning the scid gene, which will enhance our understanding of double-strand break repair pathways in humans.


Subject(s)
Chromosomes, Human, Pair 8 , Mice, SCID/genetics , Radiation Tolerance , Animals , Base Sequence , Cell Line , Chromosome Aberrations , DNA Repair , Genetic Complementation Test , Humans , Hybrid Cells , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phenotype
11.
Am J Hum Genet ; 49(1): 184-91, 1991 Jul.
Article in English | MEDLINE | ID: mdl-1905878

ABSTRACT

A compound (AC)n repeat located 1,000 bp downstream from the human synapsin I gene and within the last intron of the A-raf-1 gene has been identified. DNA data-base comparisons of the sequences surrounding the repeat indicate that the synapsin I gene and the A-raf-1 gene lie immediately adjacent to each other, in opposite orientation. PCR amplification of this synapsin I/A-raf-1 associated repeat by using total genomic DNA from members of the 40 reference pedigree families of the Centre d'Etude du Polymorphisme Humaine showed it to be highly polymorphic, with a PIC value of .84 and a minimum of eight alleles. Because the synapsin I gene has been mapped previously to the short arm of the human X chromosome at Xp11.2, linkage analysis was performed with markers on the proximal short arm of the X chromosome. The most likely gene order is DXS7SYN/ARAF1TIMPDXS255DXS146, with a relative probability of 5 x 10(8) as compared with the next most likely order. This highly informative repeat should serve as a valuable marker for disease loci mapped to the Xp11 region.


Subject(s)
Genetic Linkage , Nerve Tissue Proteins/genetics , Repetitive Sequences, Nucleic Acid , X Chromosome/chemistry , Amino Acid Sequence , Chromosome Banding , Chromosome Mapping , Gene Amplification , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Synapsins
12.
Brain Dev ; 9(5): 469-74, 1987.
Article in English | MEDLINE | ID: mdl-3124657

ABSTRACT

The characteristics of Rett syndrome suggest that it is an X-linked neurodegenerative disorder. Laboratory investigations to date have not revealed any metabolic abnormalities in affected individuals. Synapsin I is a neuron-specific protein thought to play a fundamental role in neuronal function. In this report we summarize the circumstantial evidence suggesting that a defect in synapsin I gene structure or expression might be involved in Rett syndrome. This evidence includes analysis of structural and functional aspects of synapsin I primary structure, characterization of synapsin I messenger RNAs, location of the synapsin I gene on the human X chromosome and preliminary analysis of synapsin I gene structure in Rett individuals.


Subject(s)
Brain Diseases/genetics , DNA/genetics , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Female , Humans , Molecular Sequence Data , Rats , Synapsins , Syndrome , X Chromosome/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...