Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 58(17): 3965-73, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731510

ABSTRACT

DNA repair status is recognized as an important determinant of the clinical efficacy of cancer chemotherapy. To assess the role that a mammalian DNA glycosylase plays in modulating the toxicity and clastogenicity of the chemotherapeutic DNA cross-linking alkylating agents, we compared the sensitivity of wild-type murine cells to that of isogenic cells bearing homozygous null mutations in the 3-methyladenine DNA glycosylase gene (Aag). We show that Aag protects against the toxic and clastogenic effects of 1,3-bis(2-chloroethyl)-1-nitrosourea and mitomycin C (MMC), as measured by cell killing, sister chromatid exchange, and chromosome aberrations. This protection is accompanied by suppression of apoptosis and a slightly reduced p53 response. Our results identify 3-methyladenine DNA glycosylase-initiated base excision repair as a potentially important determinant of the clinical efficacy and, possibly, the carcinogenicity of these widely used chemotherapeutic agents. However, Aag does not contribute significantly to protection against the toxic and clastogenic effects of several chemotherapeutic nitrogen mustards (namely, mechlorethamine, melphalan, and chlorambucil), at least in the mouse embryonic stem cells used here. We also compare the Aag null phenotype with the Fanconi anemia phenotype, a human disorder characterized by cellular hypersensitivity to DNA cross-linking agents, including MMC. Although Aag null cells are sensitive to MMC-induced growth delay and cell cycle arrest, their sensitivity is modest compared to that of Fanconi anemia cells.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Carmustine/toxicity , DNA Repair , Guanine/analogs & derivatives , Mitomycin/toxicity , Mutagens/toxicity , N-Glycosyl Hydrolases/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , DNA Glycosylases , G2 Phase/drug effects , Guanine/metabolism , Humans , Mice , Mitosis/drug effects
2.
J Biol Chem ; 273(9): 5412-8, 1998 Feb 27.
Article in English | MEDLINE | ID: mdl-9479003

ABSTRACT

DNA-damaging agents produce a plethora of cellular responses that include p53 induction, cell cycle arrest, and apoptosis. It is generally assumed that it is the DNA damage produced by these agents that triggers such responses, but there is limited direct evidence to support this assumption. Here, we used DNA alkylation repair proficient and deficient isogenic mouse cell lines to demonstrate that the signal to trigger p53 induction, cell cycle arrest, and apoptosis in response to alkylating agents does emanate from DNA damage. Moreover, we established that 3-methyladenine, a relatively minor DNA lesion produced by most methylating agents (which form mainly 7-methylguanine), can specifically induce sister chromatid exchange, chromatid and chromosome gaps and breaks, S phase arrest, the accumulation of p53, and apoptosis. This study was made possible by the generation of 3-methyladenine DNA glycosylase null mutant cells by targeted homologous recombination and by the chemical synthesis of a methylating agent that almost exclusively produces 3-methyladenine DNA lesions. The combined use of these two experimental tools has defined the biological consequences of 3-methyladenine, a DNA lesion produced by endogenous cellular metabolites, environmental carcinogens, and chemotherapeutic alkylating agents.


Subject(s)
Adenine/analogs & derivatives , DNA Damage , DNA Glycosylases , DNA Repair , N-Glycosyl Hydrolases/deficiency , Adenine/metabolism , Alkylating Agents , Animals , Apoptosis , Cell Line , Chromosome Aberrations , DNA Adducts/metabolism , DNA Methylation , Mice , Netropsin/analogs & derivatives , Netropsin/pharmacology , Sister Chromatid Exchange , Tumor Suppressor Protein p53/biosynthesis
3.
EMBO J ; 15(4): 945-52, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8631315

ABSTRACT

In Escherichia coli, the repair of 3-methyladenine (3MeA) DNA lesions prevents alkylation-induced cell death because unrepaired 3MeA blocks DNA replication. Whether this lesion is cytotoxic to mammalian cells has been difficult to establish in the absence of 3MeA repair-deficient cell lines. We previously isolated and characterized a mouse 3MeA DNA glycosylase cDNA (Aag) that provides resistance to killing by alkylating agents in E. coli. To determine the in vivo role of Aag, we cloned a large fragment of the Aag gene and used it to create Aag-deficient mouse cells by targeted homologous recombination. Aag null cells have no detectable Aag transcripts or 3MeA DNA glycosylase activity. The loss of Aag renders cells significantly more sensitive to methyl methanesulfonate-induced chromosome damage, and to cell killing induced by two methylating agents, one of which produces almost exclusively 3MeAs. Aag null embryonic stem cells become sensitive to two cancer chemotherapeutic alkylating agents, namely 1,3-bis(2-chloroethyl)-1-nitrosourea and mitomycin C, indicating that Aag status is an important determinant of cellular resistance to these agents. We conclude that this mammalian 3MeA DNA glycosylase plays a pivotal role in preventing alkylation-induced chromosome damage and cytotoxicity.


Subject(s)
DNA Damage , DNA Glycosylases , N-Glycosyl Hydrolases/metabolism , Alkylating Agents/toxicity , Animals , Cell Division , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Repair , Mice , Mice, Knockout , Mutagenesis , Sister Chromatid Exchange , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...