Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters










Publication year range
1.
Cytogenet Genome Res ; 125(3): 165-75, 2009.
Article in English | MEDLINE | ID: mdl-19738377

ABSTRACT

The Bloom protein (BLM) and Topoisomerase IIIalpha are found in association with proteins of the Fanconi anemia (FA) pathway, a disorder manifesting increased cellular sensitivity to DNA crosslinking agents. In order to determine if the association reflects a functional interaction for the maintenance of genome stability, we have analyzed the effects of siRNA-mediated depletion of the proteins in human cells. Depletion of Topoisomerase IIIalpha or BLM leads to increased radial formation, as is seen in FA. BLM and Topoisomerase IIIalpha are epistatic to the FA pathway for suppression of radial formation in response to DNA interstrand crosslinks since depletion of either of them in FA cells does not increase radial formation. Depletion of Topoisomerase IIIalpha or BLM also causes an increase in sister chromatid exchanges, as is seen in Bloom syndrome cells. Human Fanconi anemia cells, however, do not demonstrate increased sister chromatid exchanges, separating this response from radial formation. Primary cell lines from mice defective in both Blm and Fancd2 have the same interstrand crosslink-induced genome instability as cells from mice deficient in the Fancd2 protein alone. These observations demonstrate that the association of BLM and Topoisomerase IIIalpha with Fanconi proteins is a functional one, delineating a BLM-Topoisomerase IIIalpha-Fanconi pathway that is critical for suppression of chromosome radial formation.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA/metabolism , Fanconi Anemia/metabolism , RecQ Helicases/metabolism , Animals , Cell Line , Cross-Linking Reagents/pharmacology , DNA Topoisomerases, Type I/genetics , Fanconi Anemia/genetics , Genomic Instability/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitomycin/pharmacology , RNA, Small Interfering/genetics , RecQ Helicases/genetics , Sister Chromatid Exchange
2.
Cytogenet Genome Res ; 121(3-4): 174-80, 2008.
Article in English | MEDLINE | ID: mdl-18758156

ABSTRACT

High levels of interstrand cross-link damage in mammalian cells cause chromatid breaks and radial formations recognizable by cytogenetic examination. The mechanism of radial formation observed following DNA damage has yet to be determined. Due to recent findings linking homologous recombination and non-homologous end-joining to the action of the Fanconi anemia pathway, we speculated that radials might be the result of defects in either of the pathways of DNA repair. To test this hypothesis, we have investigated the role of homologous recombination proteins RAD51 and RAD52, non-homologous end-joining proteins Ku70 and LIG4, and protein MRE11 in radial formation and cell survival following interstrand crosslink damage with mitomycin C. For the studies we used small inhibitory RNA to deplete the proteins from cells, allowing for evaluation of radial formation and cell survival. In transformed normal human fibroblasts, depletion of these proteins increased interstrand crosslink sensitivity as manifested by decreased cell survival and increased radial formation. These results demonstrate that inactivation of proteins from either of the two separate DNA repair pathways increases cellular sensitivity to interstrand crosslinks, indicating each pathway plays a role in the normal response to interstrand crosslink damage. We can also conclude that homologous recombination or non-homologous end-joining are not required for radial formation, since radials occur with depletion of these pathways.


Subject(s)
DNA Damage , Recombination, Genetic , Cell Line, Transformed , DNA/drug effects , Humans , Mitomycin/toxicity , RNA, Small Interfering
3.
Mol Genet Metab ; 94(1): 38-45, 2008 May.
Article in English | MEDLINE | ID: mdl-18180189

ABSTRACT

The protein encoded by SNM1 in Saccharomyces cerevisiae has been shown to act specifically in DNA interstrand crosslinks (ICL) repair. There are five mammalian homologs of SNM1, including Artemis, which is involved in V(D)J recombination. Cells from mice constructed with a disruption in the Snm1 gene are sensitive to the DNA interstrand crosslinker, mitomycin (MMC), as indicated by increased radial formation following exposure. The mice reproduce normally and have normal life spans. However, a partial perinatal lethality, not seen in either homozygous mutant alone, can be noted when the Snm1 disruption is combined with a Fancd2 disruption. To explore the role of hSNM1 and its homologs in ICL repair in human cells, we used siRNA depletion in human fibroblasts, with cell survival and chromosome radials as the end points for sensitivity following treatment with MMC. Depletion of hSNM1 increases sensitivity to ICLs as detected by both end points, while depletion of Artemis does not. Thus hSNM1 is active in maintenance of genome stability following ICL formation. To evaluate the epistatic relationship between hSNM1 and other ICL repair pathways, we depleted hSNM1 in Fanconi anemia (FA) cells, which are inherently sensitive to ICLs. Depletion of hSNM1 in an FA cell line produces additive sensitivity for MMC. Further, mono-ubiquitination of FANCD2, an endpoint of the FA pathway, is not disturbed by depletion of hSNM1 in normal cells. Thus, hSNM1 appears to represent a second pathway for genome stability, distinct from the FA pathway.


Subject(s)
DNA Repair Enzymes/genetics , Genomic Instability , Nuclear Proteins/genetics , Animals , Cell Cycle Proteins , DNA Repair , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fibroblasts/metabolism , Humans , Mice , Mice, Transgenic , RNA, Small Interfering/metabolism , Transfection
4.
Mutat Res ; 487(3-4): 73-83, 2001 Dec 19.
Article in English | MEDLINE | ID: mdl-11738934

ABSTRACT

Yeast mutants, snm1 (pso2-1), rev3 (pso1-1), and rad51, which display significant sensitivity to interstrand crosslinks (ICLs) have low relative sensitivity to other DNA damaging agents. SNM1, REV3, and RAD51 were disrupted in the same haploid strain, singly and in combination. The double mutants, snm1 Delta rev3 Delta, snm1 Delta rad51 Delta and rev3 Delta rad51 Delta were all more sensitive to ICLs than any of the single mutants, indicating that they are in separate epistasis groups for survival. A triple mutant displayed greater sensitivity to ICLs than any of the double mutants, with one ICL per genome being lethal. Therefore, Saccharomyces cerevisiae appears to have three separate ICL repair pathways, but no more. S-phase delay was not observed after ICL damage introduced by cisplatin (CDDP) or 8-methoxypsoralen (8-MOP) during the G1-phase, in any of the above mutants, or in an isogenic rad14 Delta mutant deficient in nucleotide excision repair. However, the psoralen analog angelicin (monoadduct damage) induced a significant S-phase delay in the rad14 Delta mutant. Thus, normal S-phase in the presence of ICLs does not seem to be due to rapid excision repair. The results also indicate that monoadduct formation by CDDP or 8-MOP at the doses used is not sufficient to delay S-phase in the rad14 Delta mutant. While the sensitivity of a rev3 Delta mutant indicates Pol zeta is needed for optimal ICL repair, isogenic cells deficient in Pol eta (rad30 Delta cells) were not significantly more sensitive to ICL agents than wild-type cells, and have no S-phase delay.


Subject(s)
DNA Repair/physiology , DNA, Fungal/drug effects , DNA-Binding Proteins/physiology , Fungal Proteins/physiology , Nuclear Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , DNA Damage , DNA Repair/genetics , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/physiology , Endodeoxyribonucleases , Epistasis, Genetic , Fungal Proteins/genetics , Furocoumarins/pharmacology , Methoxsalen/pharmacology , Nuclear Proteins/genetics , Rad51 Recombinase , S Phase/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/genetics
5.
Article in English | MEDLINE | ID: mdl-11701643

ABSTRACT

Inherited defects in DNA repair or the processing of DNA damage can lead to disease. Both autosomal recessive and autosomal dominant modes of inheritance are represented. The diseases as a group are characterized by genomic instability, with eventual appearance of cancer. The inherited defects frequently have a specific DNA damage sensitivity, with cells from affected individuals showing normal resistance to other genotoxic agents. The known defects are subtle alterations in transcription, replication, or recombination, with alternate pathways of processing permitting cellular viability. Distinct diseases may arise from different mutations in one gene; thus, clinical phenotypes may reflect the loss of different partial functions of a gene. The findings indicate that partial defects in transcription or recombination lead to genomic instability, cancer, and characteristic disease phenotypes.


Subject(s)
DNA Damage , Cell Cycle , DNA Repair , Genes, Dominant , Genes, Recessive , Genome, Human , Humans , Recombination, Genetic
6.
Mutat Res ; 461(1): 1-13, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10980408

ABSTRACT

The role of Snm1, Rev3 and Rad51 in S-phase after cisplatin (CDDP) DNA treatment has been examined. When isogenic deletion mutants snm1 delta, rev3 delta and rad51 delta were arrested in G1 and treated with doses of CDDP causing significant lethality (<20% survival in the mutant strains), they progressed through S-phase with normal kinetics. The mutants arrested in G2 like wild-type cells, however they did not exit the arrest and reenter the cell cycle. This finding demonstrates that these genes are not required to allow DNA replication in the presence of damage. Therefore, Snm1, Rev3 and Rad51 may act after S to allow repair. At high levels of damage (<40% survival in wild-type cells) S-phase was slowed in a MEC1-dependent fashion. The cross-link incision kinetics of snm1 delta and rev3 delta mutants were also examined; both showed no deficiencies in incision of cross-linked DNA.


Subject(s)
Cisplatin/pharmacology , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase , Fungal Proteins/genetics , Interphase/genetics , Nuclear Proteins/genetics , Saccharomyces cerevisiae Proteins , Antineoplastic Agents/pharmacology , Cross-Linking Reagents/pharmacology , DNA Damage , DNA Repair , DNA Replication , Endodeoxyribonucleases , Furocoumarins/pharmacology , G2 Phase/genetics , Gene Deletion , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Rad51 Recombinase , S Phase/genetics , Saccharomyces cerevisiae
7.
Am J Hum Genet ; 66(5): 1540-51, 2000 May.
Article in English | MEDLINE | ID: mdl-10762542

ABSTRACT

Fanconi anemia (FA) is a rare autosomal recessive disease manifested by bone-marrow failure and an elevated incidence of cancer. Cells taken from patients exhibit spontaneous chromosomal breaks and rearrangements. These breaks and rearrangements are greatly elevated by treatment of FA cells with the use of DNA cross-linking agents. The FA complementation group D gene (FANCD) has previously been localized to chromosome 3p22-26, by use of microcell-mediated chromosome transfer. Here we describe the use of noncomplemented microcell hybrids to identify small overlapping deletions that narrow the FANCD critical region. A 1.2-Mb bacterial-artificial-chromosome (BAC)/P1 contig was constructed, bounded by the marker D3S3691 distally and by the gene ATP2B2 proximally. The contig contains at least 36 genes, including the oxytocin receptor (OXTR), hOGG1, the von Hippel-Lindau tumor-suppressor gene (VHL), and IRAK-2. Both hOGG1 and IRAK-2 were excluded as candidates for FANCD. BACs were then used as probes for FISH analyses, to map the extent of the deletions in four of the noncomplemented microcell hybrid cell lines. A narrow region of common overlapping deletions limits the FANCD critical region to approximately 200 kb. The three candidate genes in this region are TIGR-A004X28, SGC34603, and AA609512.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 3/genetics , Fanconi Anemia/genetics , Blotting, Southern , Cell Line , Chromosome Breakage/genetics , Contig Mapping , DNA, Complementary/genetics , DNA-Formamidopyrimidine Glycosylase , Expressed Sequence Tags , Fanconi Anemia/pathology , Genetic Complementation Test , Genetic Linkage/genetics , Genetic Markers/genetics , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Interleukin-1 Receptor-Associated Kinases , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/physiology , Protein Kinases/genetics , Protein Kinases/physiology , Sequence Tagged Sites
8.
Mutat Res ; 434(1): 29-39, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10377946

ABSTRACT

Cisplatin (CDDP) has been used as a DNA cross-linking agent to evaluate whether there is a specific cell cycle checkpoint response to such damage in Saccharomyces cerevisiae (S. cerevisiae). Fluorescent-activated cell sorting (FACS) analysis showed only a G2/M checkpoint, normal exit from G1 and progression through S-phase following alpha-factor arrest and CDDP treatment. Of the checkpoint mutants tested, rad9, rad17 and rad24, did not show increased sensitivity to CDDP compared to isogenic wild-type cells. However, other checkpoint mutants tested (mec1, mec3 and rad53) showed increased sensitivity to CDDP, as did controls with a defect in excision repair (rad1 and rad14) or a defect in recombination (rad51 and rad52). Thus, by survival and cell cycle kinetics, it appears that DNA cross-links do not inhibit entry into S-phase or slow DNA replication and that replication continues after cisplatin treatment in yeast.


Subject(s)
Cell Cycle Proteins , Cell Cycle/drug effects , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , DNA, Fungal/drug effects , DNA-Binding Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/drug effects , Cell Cycle/genetics , DNA Damage , DNA Repair/genetics , DNA Repair Enzymes , DNA, Fungal/genetics , DNA, Fungal/metabolism , Endonucleases/genetics , Flow Cytometry , Fungal Proteins/genetics , G2 Phase/drug effects , Gene Expression Regulation, Fungal/drug effects , Genes, Fungal/genetics , Microbial Sensitivity Tests , Mitosis/drug effects , Mutation , Protein Kinases/genetics , Protein Serine-Threonine Kinases , S Phase/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
9.
J Biol Chem ; 273(29): 18605-9, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9660833

ABSTRACT

Antibodies raised against the 51C/SHIP2 inositol polyphosphate 5'-phosphatase were used to examine the effects of growth factors and insulin on the metabolism of this protein. Immunoblot analysis revealed that the 51C/SHIP2 protein was widely expressed in fibroblast and nonhematopoietic tumor cell lines, unlike the SHIP protein, which was found only in cell lines of hematopoietic origin. The 51C/SHIP2 antiserum precipitated a protein of approximately 145 kDa along with an activity which hydrolyzed phosphatidylinositol 3,4, 5-trisphosphate to phosphatidylinositol 3,4-bisphosphate. Tyrosine phosphorylation of the 51C/SHIP2 protein occurred in response to treatment of cells with epidermal growth (EGF), platelet-derived growth factor (PDGF), nerve growth factor (NGF), insulin-like growth factor-1 (IGF-1), or insulin. EGF and PDGF induced transient tyrosine phosphorylation of 51C/SHIP2, with maximal tyrosine phosphorylation occurring at 5-10 min following treatment and returning to near basal levels within 20 min. In contrast, treatment of cells with NGF, IGF-1, or insulin resulted in prolonged tyrosine phosphorylation of 51C/SHIP2 protein, with 40-80% maximal phosphorylation sustained for up to 2 h following agonist treatment. The kinetics of activation of the Akt/PKB protein kinase by the various factors correlated well with the kinetics of tyrosine phosphorylation of 51C/SHIP2. EGF, NGF, and PDGF stimulated the association of 51C/SHIP2 protein with the Shc adapter protein; however, no Shc could be detected in 51C/SHIP2-immune precipitates from cells treated with IGF-1 or insulin. The data suggest that 51C/SHIP2 may play a significant role in regulation of phosphatidylinositol 3'-kinase signaling by growth factors and insulin.


Subject(s)
Growth Substances/pharmacology , Insulin/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Tyrosine/pharmacology , src Homology Domains , 3T3 Cells , Animals , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphorylation/drug effects , Rabbits , Signal Transduction , Tumor Cells, Cultured
10.
Nucleic Acids Res ; 26(4): 1124-5, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9461477

ABSTRACT

Bacterial Artificial Chromosomes (BACs) have been used to complement a metabolic defect and to transfer a drug resistance marker into mammalian cells by electroporation. The selectable markers are stable and the recipient cells have BAC DNA integrated into the chromosomes as shown by fluorescent in situ hybridization, PCR and Southern hybridization.


Subject(s)
Chromosomes, Bacterial/genetics , Electroporation , Genetic Complementation Test , Base Sequence , Cell Line , DNA Primers/genetics , Gene Transfer Techniques , Genetic Markers , Genetic Techniques , Humans , In Situ Hybridization, Fluorescence , Polymerase Chain Reaction
12.
Somat Cell Mol Genet ; 23(1): 1-7, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9217996

ABSTRACT

Fanconi anemia is a rare autosomal recessive disease characterized by developmental defects of the thumb and radius, childhood onset of pancytopenic anemia and increased risk of leukemia. At least five complementation groups (A-E) have been defined but only the FAC gene has been cloned. Cells can be assigned to complementation group C by direct mutation analysis. To facilitate the search for additional FA genes and to measure the frequency of complementation groups, we have established new genetically marked immortalized FA-A and FA-D fibroblast cell lines and show their usefulness as universal fusion donors. These reference FA cell lines facilitated somatic cell fusion analysis and enabled us to assign the complementation group in 16 unrelated FA patients from North America. The majority of patients, belong to FA complementation group A (69%), followed by FA-C (18%), FA-D (4%) and FA-B or FA-E (9%).


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Fanconi Anemia/genetics , Nuclear Proteins , Cell Fusion , Cell Survival , DNA Mutational Analysis , Epoxy Compounds/toxicity , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Genes , Genetic Complementation Test , Genetic Linkage , Humans , Mitomycin/toxicity , North America , Proteins/genetics , Transfection
14.
Somat Cell Mol Genet ; 23(6): 371-7, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9661700

ABSTRACT

Cells from patients with Fanconi anemia (FA) show decreased viability and decreased chromosome stability after treatment with DNA cross-linking agents, compared to normal cells. FA cells also show a relative accumulation at the G2/M transition after such treatment. This has suggested a possible checkpoint abnormality. In the studies presented here, treatment with hydroxyurea, caffeine or inhibitors of cell cycle kinases did not reveal abnormalities in survival or chromosome stability in FA-A or FA-D cells. Chromosomal breaks introduced by hydrogen peroxide or methyl methanesulfonate accumulated to the same extent in FA-A or FA-D cells as in normal cells. We conclude that FA-A and FA-D cells respond normally to agents known to alter the cell cycle or introduce DNA strand breaks. FA cells process strand breaks and a variety of DNA monoadducts normally. Our results are compatible with repair of DNA crosslinks being slower in FA than in normal cells and FA cells having normal cell cycle checkpoints.


Subject(s)
Caffeine/pharmacology , Cell Cycle/drug effects , Central Nervous System Stimulants/pharmacology , Cross-Linking Reagents/pharmacology , Fanconi Anemia/pathology , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Cells, Cultured/drug effects , Chromosome Breakage , DNA Adducts , Enzyme Inhibitors/pharmacology , Fanconi Anemia/genetics , Humans , Hydrogen Peroxide/pharmacology , Hydroxyurea/pharmacology , Methyl Methanesulfonate/pharmacology , Mitomycin/pharmacology , Oxidants/pharmacology
15.
Somat Cell Mol Genet ; 22(2): 151-7, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8782494

ABSTRACT

Fanconi anemia (FA) is an autosomal recessive disease characterized by birth defects, progressive bone marrow failure and increased risk for leukemia. FA cells display chromosome breakage and increased cell killing in response to DNA crosslinking agents. At least 5 genes have been defined by cell complementation studies, but only one of these, FAC has been cloned to date. Efforts to map and isolate new FA genes by functional complementation have been hampered by the lack of immortalized FA fibroblast cell lines. Here we report the use of a novel immortalization strategy to create 4 new immortalized FA fibroblast lines, including one from the rare complementation group D.


Subject(s)
Cell Line, Transformed , Fanconi Anemia , Fibroblasts/cytology , Cell Culture Techniques , Cell Fusion , Cell Survival/drug effects , Chromosome Aberrations , Epoxy Compounds/pharmacology , Ethyl Methanesulfonate/pharmacology , Fibroblasts/drug effects , Genetic Complementation Test , Humans , Karyotyping , Mitomycin/pharmacology , Mutagens/pharmacology , Phenotype , Skin/cytology
16.
Biochim Biophys Acta ; 1264(2): 243-8, 1995 Nov 07.
Article in English | MEDLINE | ID: mdl-7495870

ABSTRACT

The gene for Thermus aquaticus (Taq) DNA polymerase enzyme (Taq Pol I) was mutagenized and sixty-two candidate clones were screened for enzyme activity. Two of the clones expressed enzymes (*Taq-3 and *Taq-5) that showed very reduced 5'-3' exonuclease activity and normal DNA polymerase activity. These two enzymes showed heat resitance and storage stability similar to Taq Pol I and had similar effectiveness in PCR. Processivity of the polymerases was compared by measuring the extension of an end-labeled primer annealed to a single stranded DNA, as well as by a PCR method. The processivity of *Taq-3 and *Taq-5 was similar to Taq Pol I (50-80 nucleotides) and more processive than a Taq Pol I deficient in the 5'-3' exonuclease due to absence of the first 290 amino acids (Stoffel fragment). The results indicate two amino acids which are required for normal 5'-3' exonuclease activity in Taq Pol I (Arg-25 and Arg-74).


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Exodeoxyribonucleases/metabolism , Thermus/enzymology , Cloning, Molecular , DNA Primers , DNA-Directed DNA Polymerase/biosynthesis , Enzyme Stability , Exodeoxyribonuclease V , Exodeoxyribonucleases/antagonists & inhibitors , Exodeoxyribonucleases/biosynthesis , Genes, Bacterial , Mutagenesis , Nucleic Acid Synthesis Inhibitors , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Restriction Mapping , Taq Polymerase , Thermus/genetics
17.
Genomics ; 29(1): 285-7, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-8530088

ABSTRACT

We have cloned a novel human cDNA, INPPL1 (GenBank Accession No. L36818), which maps to 11q23. The corresponding mRNA is 4657 nt in length and is widely expressed in both fetal and adult tissues. An open reading frame of 3441 nt encodes a putative polypeptide that shares several domains with inositol triphosphate phosphatases. Several polymorphisms have been mapped to the 3'-untranslated region, yet the putative coding region showed no polymorphisms in nine independent cDNA samples.


Subject(s)
Chromosomes, Human, Pair 11 , Hominidae/genetics , Phosphoric Monoester Hydrolases/genetics , Adult , Amino Acid Sequence , Animals , Chromosome Mapping , Cloning, Molecular , Conserved Sequence , DNA, Complementary , Fetus , Humans , Molecular Sequence Data , Open Reading Frames , Phosphoric Monoester Hydrolases/biosynthesis , Polymorphism, Genetic , Sequence Homology, Amino Acid
18.
Exp Cell Res ; 217(2): 272-9, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7698226

ABSTRACT

Human diploid fibroblasts (HDF) IMR90, starting at various population doubling levels (PDL), were serially cultured under four different oxygen conditions; the conventional atmospheric 20% O2 condition and three lower oxygen conditions (1, 6, and 12% O2). All cultures from different PDLs showed that a longer replicative life span was achieved under the lower oxygen conditions. When the starting culture PDL was between 33 and 62, the increased life span rate was constant (average 22%) at 1% O2, but the rate decreased when the starting PDL was higher. The growth advantage under the 1% O2 condition was also observed in the very late passages of cultures to some extent, but terminal cultures with senescent cells were not stimulated by the low oxygen condition. When cultures at an extended PDL under the 1% O2 condition were shifted back to the 20% O2 condition, the cells rapidly senesced. HDF from a subject with Werner syndrome, a premature aging disease, which are known to have reduced replicative potential in vitro, also showed 43% increase in life span under the lowest oxygen conditions. When SV40 large T-transformed IMR90 cells at preimmortal stages were tested, no significant growth differences were observed under different oxygen conditions, and all the cultures died out at a similar PDL. These results suggest: (1) The atmospheric 20% oxygen tension hastens HDF senescence. (2) Young cells are more resistant to oxygen tension than old cells. (3) SV40 large T transformation abolishes the oxygen effect on cellular aging.


Subject(s)
Cellular Senescence/physiology , Fibroblasts/cytology , Oxygen/physiology , Cell Division/physiology , Cell Line , Cell Line, Transformed , Diploidy , Humans , Werner Syndrome/pathology
20.
Hum Genet ; 93(5): 583-6, 1994 May.
Article in English | MEDLINE | ID: mdl-8168839

ABSTRACT

Cells from Fanconi anemia (FA) patients have defective DNA repair and are hypersensitive to DNA crosslinking agents such as mitomycin C (MMC). We examined the possibility that topoisomerase I is involved in the DNA crosslink repair system and is deficient in FA group A cells. FA cells and control cells were exposed to MMC with or without camptothecin (CPT), a topoisomerase I inhibitor. The cells did not show any increased sensitivity to killing by MMC with CPT, suggesting that the topoisomerase I is not involved in MMC-damaged DNA repair. However, FA cells showed increased sensitivity to CPT in comparison to control cells, raising the possibility of altered topoisomerase I in FA cells. Therefore, a mutation analysis was performed on topoisomerase I cDNA from FA cells by using chemical cleavage mismatch scanning and nucleotide sequencing. No mutation was detected from GM1309, a group A FA cell line. A base transition (C to T) at position 241, causing an amino acid change (His to Tyr), was found in GM2061, a FA cell line of unknown complementation group. However, allele-specific oligonucleotide hybridization analysis showed that this is a gene polymorphism. We conclude that FA cells have normal gene structure for topoisomerase I.


Subject(s)
DNA Topoisomerases, Type I/genetics , Fanconi Anemia/enzymology , Base Sequence , Camptothecin/pharmacology , Cell Line/drug effects , Cell Survival/drug effects , Cell Transformation, Viral , Cells, Cultured , DNA/biosynthesis , DNA Mutational Analysis , DNA Primers , Fanconi Anemia/pathology , Humans , Mitomycin/pharmacology , Molecular Sequence Data , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...