Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Environ Mol Mutagen ; 54(5): 327-37, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23677513

ABSTRACT

The highly conserved DNA glycosylase MutY is implicated in repair of oxidative DNA damage, in particular in removing adenines misincorporated opposite 7,8-dihydro-8-oxoguanine (8-oxo-G). The MutY homologues (MutYH) physically associate with proteins implicated in replication, DNA repair, and checkpoint signaling, specifically with the DNA damage sensor complex 9-1-1 proteins. Here, we ask whether MutYH could have a broader function in sensing and repairing different types of DNA damage induced by conventional chemotherapeutics. Thus, we examined if deletion of the Schizosaccharomyces pombe MutY homologue, Myh1, alone or in combination with deletion of either component of the 9-1-1 sensor complex, influences survival after exposure to different classes of DNA damaging chemotherapeutics that do not act primarily by causing 8-oxoG lesions. We show that Myh1 contributes to survival on genotoxic stresses induced by the oxidizing, DNA double strand break-inducing, bleomycins, or the DNA crosslinking platinum compounds, particularly in a rad1 mutant background. Exposure of cells to cisplatin leads to a moderate overall accumulation of Myh1 protein. Interestingly, we found that DNA damage induced by phleomycin results in increased chromatin association of Myh1. Further, we demonstrate that Myh1 relocalizes to the nucleus after exposure to hydrogen peroxide or chemotherapeutics, most prominently seen after phleomycin treatment. These observations indicate a wider role of Myh1 in DNA repair and DNA damage-induced checkpoint activation than previously thought.


Subject(s)
Antineoplastic Agents/toxicity , Cross-Linking Reagents/toxicity , DNA Glycosylases/metabolism , DNA Repair/physiology , DNA, Fungal/drug effects , Schizosaccharomyces pombe Proteins/metabolism , Active Transport, Cell Nucleus/drug effects , Blotting, Western , Cisplatin/toxicity , DNA Glycosylases/genetics , Fluorescent Antibody Technique , Gene Deletion , Microbial Viability/drug effects , Microscopy, Fluorescence , Organoplatinum Compounds/toxicity , Oxaliplatin , Phleomycins/toxicity , Schizosaccharomyces pombe Proteins/genetics
2.
Nucleic Acids Res ; 35(2): 353-62, 2007.
Article in English | MEDLINE | ID: mdl-17170004

ABSTRACT

The inaccurate repair of DNA double-strand breaks (DSBs) can result in genomic instability, and additionally cell death or the development of cancer. Elg1, which forms an alternative RFC-like complex with RFC2-5, is required for the maintenance of genome stability in Saccharomyces cerevisiae, and its function has been linked to DNA replication or damage checkpoint response. Here, we show that Elg1 is involved in homologous recombination (HR)-mediated DSB repair. Mutants of elg1 were partially defective in HR induced by methylmethanesufonate (MMS) and phleomycin. Deletion of ELG1 resulted in less efficient repair of phleomycin-induced DSBs in G2/M phase-arrested cells. During HR between MAT and HML loci, Elg1 associated with both the MAT locus near the HO endonuclease-induced DSB site, and the HML homologous donor locus. The association of Elg1 with the MAT locus was not dependent on Rad52. However, Elg1 association with the HML locus depended on Rad52. Importantly, we found that two of the later steps in HR-mediated repair of an HO endonuclease-induced DSB, primer extension after strand invasion and ligation, were less efficient in elg1 mutants. Our results suggest that Elg1 is involved in DSB repair by HR.


Subject(s)
Carrier Proteins/physiology , DNA Breaks, Double-Stranded , DNA Repair , Carrier Proteins/genetics , Cell Cycle , Genes, Mating Type, Fungal , Mutation , Phleomycins/toxicity , Proliferating Cell Nuclear Antigen/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , Recombination, Genetic , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins
3.
Environ Mol Mutagen ; 47(9): 707-17, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17078097

ABSTRACT

The Saccharomyces cerevisiae SPT10 protein possesses a DNA-binding domain that is fused to a putative histone acetyltransferase domain. It binds specifically to upstream-activating sequence elements in the core histone promoters and plays a direct role in histone gene regulation. SPT10 is also required for cell-cycle-specific K56 acetylation at histone genes, allowing the recruitment of the nucleosome remodeling factor Snf5 and subsequent regulation of gene transcription. We reisolated the SPT10 gene in a functional genome-wide screen designed to identify haploid yeast mutants that are hypersensitive to the antitumor drug bleomycin, which acts by damaging DNA. In addition to bleomycin, we show that spt10Delta mutants are also hypersensitive to a limited set of genotoxic agents that create DNA strand breaks, but not to 254-nm ultraviolet light or 4-nitroquinoline-1-oxide, which generate helix distortion. The hypersensitivities of the spt10Delta mutant to the genotoxic agents are rescued by a single copy plasmid carrying the SPT10 gene. We further showed that spt10Delta mutants displayed a modest twofold increase spontaneous mutant frequency, as compared to the parent. Following exposure to bleomycin, these mutants accumulate unrepaired lesions, e.g., DNA strand breaks with blocked 3'-ends in the chromosomal DNA. This defect is not due to the altered expression level or the enzymatic activities of a key DNA repair enzyme, APN1, which is known to repair DNA strand breaks with blocked ends. We propose that SPT10 mediates repair of a subset of DNA lesions by acetylating histones to promote recruitment of DNA repair enzymes.


Subject(s)
Mutagens/toxicity , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/drug effects , Transcription Factors/genetics , 4-Nitroquinoline-1-oxide/toxicity , Bleomycin/toxicity , Chromatin/genetics , DNA Damage , DNA Repair , Gamma Rays/adverse effects , Histone Acetyltransferases , Hydrogen Peroxide/toxicity , Methyl Methanesulfonate/toxicity , Phleomycins/toxicity , Quinolones/toxicity , Rad51 Recombinase/genetics , Saccharomyces cerevisiae/genetics , Ultraviolet Rays/adverse effects
4.
Proc Natl Acad Sci U S A ; 93(3): 1238-42, 1996 Feb 06.
Article in English | MEDLINE | ID: mdl-8577747

ABSTRACT

P-glycoprotein (Pgp), a transmembrane efflux pump encoded by the MDR1 gene, transports various lipophilic drugs that enter the cell by passive diffusion through the lipid bilayer. Pgp-expressing multidrug-resistant cell lines are not usually cross-resistant to a hydrophilic antifolate methotrexate (MTX). MTX enters cells primarily through a folate carrier, but passive diffusion becomes the primary mode of MTX uptake in carrier-deficient cells. To test if a deficiency in MTX carrier would allow Pgp to confer resistance to MTX, a MTX carrier-deficient cell line (3T6-C26) was infected with a recombinant retrovirus expressing the human MDR1 gene. The infected 3T6-C26 cells showed increased survival in MTX relative to uninfected cells. Multistep selection of the infected cells with vinblastine led to increased Pgp expression and a concomitant increase in resistance to MTX. MTX resistance of Pgp-expressing 3T6-C26 cells was reduced by Pgp inhibitors, including a Pgp-specific monoclonal antibody UTC2. In contrast, the expression and the inhibition of Pgp had no effect on MTX resistance in 3T6 cells with normal carrier-mediated MTX uptake. Thus, a deficiency in the MTX carrier enables Pgp to confer resistance to MTX, suggesting that hydrophilic compounds may become Pgp substrates when such compounds enter cells by passive diffusion.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters , Carrier Proteins/metabolism , Drug Resistance, Multiple , Folic Acid Antagonists/toxicity , Methotrexate/metabolism , Methotrexate/toxicity , Neoplasm Proteins , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Antibodies, Monoclonal , Biological Transport , Carrier Proteins/antagonists & inhibitors , Cell Line , Clone Cells , Colony-Forming Units Assay , Drug Resistance, Multiple/genetics , Fibroblasts , Flow Cytometry , Folic Acid Antagonists/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Kinetics , Mice , Phleomycins/toxicity , Pyrimidines/toxicity , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Retroviridae , Transfection , Vinblastine/toxicity
5.
Mutat Res ; 336(1): 19-27, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7528892

ABSTRACT

The recombinogenicity of damaged chromosomes in diploid Saccharomyces cerevisiae cells treated with bleomycin and structurally related phleomycin was measured, along with aneuploidy and mutation events. Phleomycin was substantially (up to 26-fold) more effective than bleomycin in producing genetic changes at all concentrations, even when colony-forming abilities of cells growing in the presence of bleomycin or phleomycin were similar. These results suggest that the DNA lesions produced by the two structurally related analogs could differ in their nature or frequency, or could be processed differently by the cells. Bioassays were developed and used to compare the cytotoxicities of freshly dissolved bleomycin and phleomycin with the cytotoxicities of lysates prepared from bleomycin- and phleomycin-treated cells. Unexpectedly, lysates prepared from bleomycin-treated cells were 1.5-3.5 times more cytotoxic than freshly dissolved bleomycin after 45-min treatments (3-33 x 10(-6) M). In contrast, lysates prepared from phleomycin-treated cells were 3-38 times less cytotoxic than freshly dissolved phleomycin (0.5-6.4 x 10(-6) M). Cytotoxicities of all lysates were higher after 36-h treatments than after 45-min treatments. At 3.3 x 10(-6) M, this increase was eightfold for bleomycin and 15-fold for phleomycin. Nevertheless, lysates from phleomycin-treated cells were considerably more cytotoxic than lysates from bleomycin-treated cells or freshly prepared bleomycin, consistent with the higher effectiveness of phleomycin than bleomycin in producing chromosomal breaks, genetic changes, and cell killing.


Subject(s)
Bleomycin/toxicity , Chromosomes, Fungal/drug effects , DNA Damage , Phleomycins/toxicity , Aneuploidy , Cell-Free System , DNA, Fungal/drug effects , Mitosis/drug effects , Mutation , Saccharomyces cerevisiae/genetics , Time Factors
6.
Mutat Res ; 193(2): 157-65, 1988 Mar.
Article in English | MEDLINE | ID: mdl-2450277

ABSTRACT

We have previously reported the isolation of 3 mutants of Chinese hamster ovary cells which exhibit hypersensitivity to bleomycin. 2 mutants were isolated on the basis of bleomycin-sensitivity [designated BLM-1 and BLM-2, Robson et al., Cancer Res., 45 (1985) 5304-5309] and 1 as adriamycin-sensitive [ADR-1, Robson et al., Cancer Res., 47 (1987) 1560-1565]. Because bleomycin generates DNA-strand breaks via a free-radical mechanism, we have studied the survival response of these mutants to a range of drugs which also generate free radicals and consequently DNA-strand breaks. The mutants are all hypersensitive to phleomycin, which differs from bleomycin in being unable to intercalate due to a modified bithiazole moiety. However, BLM-2 cells alone are hypersensitive to pepleomycin, a semi-synthetic bleomycin analogue. In contrast, BLM-1 cells are more sensitive than BLM-2 to streptonigrin (which operates via a hydroquinone intermediate). ADR-1 cells show wild-type resistance to streptonigrin. The results obtained with neocarzinostatin, an antibiotic requiring thiol activation, are unusual in that both BLM-1 and BLM-2 are approximately 3-fold more resistant than parental cells. However, the steady-state intracellular level of the major non-protein thiol, glutathione, is not altered in BLM-1 or BLM-2 cells. ADR-1 cells show essentially wild-type resistance to neocarzinostatin. Analysis of cell hybrids shows that BLM-1 and BLM-2 cells are phenotypically recessive in combination with parental CHO-K1 cells and represent different genetic complementation groups not only from one another, but also from the bleomycin-sensitive mutant xrs-6, isolated on the basis of X-ray sensitivity by Jeggo and Kemp [Mutation Res., 112 (1983) 313-319]. These results indicate that at least 3 gene products are involved in cellular protection against bleomycin toxicity in mammalian cells.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Bleomycin/toxicity , Mutation , Zinostatin/pharmacology , Animals , Cell Line , Cricetinae , Cricetulus , Drug Resistance , Genetic Complementation Test , Glutathione/metabolism , Intracellular Fluid/drug effects , Peplomycin , Phleomycins/toxicity , X-Rays
8.
J Antibiot (Tokyo) ; 37(4): 376-83, 1984 Apr.
Article in English | MEDLINE | ID: mdl-6202668

ABSTRACT

Significant and often substantial enhancement of the antitumor properties of several individual phleomycins , by co- administration via intraperitoneal injection of a number of purine analogues, is demonstrated in rats and mice having three diverse tumors. It is evident that the dose levels of both the phleomycin and the amplifier are very significant and that optimal levels vary widely with the actual agents used. Constant serum levels of amplifier can be maintained for several days by administration via silastic-pellet implantation rather than injection, and this route of administration is an effective alternative for amplifiers of low solubility.


Subject(s)
Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bleomycin/toxicity , Carcinoma 256, Walker/drug therapy , Carcinoma, Ehrlich Tumor/drug therapy , Phleomycins/toxicity , Purines/toxicity , Animals , Male , Mice , Purines/blood , Rats , Rats, Inbred Strains , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...