Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(9): e44817, 2012.
Article in English | MEDLINE | ID: mdl-23028632

ABSTRACT

O6-Methylguanine, one of alkylated DNA bases, is especially mutagenic. Cells containing this lesion are eliminated by induction of apoptosis, associated with the function of mismatch repair (MMR) proteins. A retrovirus-mediated gene-trap mutagenesis was used to isolate new genes related to the induction of apoptosis, triggered by the treatment with an alkylating agent, N-methyl-N-nitrosourea (MNU). This report describes the identification of a novel gene, MAPO2 (O6-methylguanine-induced apoptosis 2), which is originally annotated as C1orf201. The MAPO2 gene is conserved among a wide variety of multicellular organisms and encodes a protein containing characteristic PxPxxY repeats. To elucidate the function of the gene product in the apoptosis pathway, a human cell line derived from HeLa MR cells, in which the MAPO2 gene was stably knocked down by expressing specific miRNA, was constructed. The knockdown cells grew at the same rate as HeLa MR, thus indicating that MAPO2 played no role in the cellular growth. After exposure to MNU, HeLa MR cells and the knockdown cells underwent cell cycle arrest at G2/M phase, however, the production of the sub-G1 population in the knockdown cells was significantly suppressed in comparison to that in HeLa MR cells. Moreover, the activation of BAK and caspase-3, and depolarization of mitochondrial membrane, hallmarks for the induction of apoptosis, were also suppressed in the knockdown cells. These results suggest that the MAPO2 gene product might positively contribute to the induction of apoptosis triggered by O6-methylguanine.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/drug effects , Apoptosis/genetics , Guanine/analogs & derivatives , Amino Acid Motifs , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Gene Knockdown Techniques , Guanine/pharmacology , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/genetics , Methylnitrosourea/pharmacology , Mice , Molecular Sequence Data , Rats
2.
Biochem Pharmacol ; 76(3): 303-11, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18573489

ABSTRACT

O6-Methylguanine and O6-chloroethylguanine are primary DNA lesions produced by two types of antineoplastic drugs, 8-carbamoyl-3-methylimidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one (temozolomide, TMZ) and 1-(4-amino-2-methyl-5-pyrimidinyl) methyl-3-(2-chloroethyl)-3-nitrosourea (ACNU), respectively. They can be repaired by O6-methylguanine-DNA methyltransferase, coded by the Mgmt gene. Otherwise, these two types of lesions induce apoptosis in different ways. O6-Chloroethylguanine blocks DNA replication thereby inducing apoptosis. On the other hand, O6-methylguanine does not block DNA replication and the resulting O6-methylguanine-thymine mispair is recognized by mismatch repair-related proteins, including MLH1, thereby inducing apoptosis. Reflecting this, mouse cells lacking both MGMT and MLH1 are resistant to TMZ, but not to ACNU. The translocation of phosphatidylserine in cell membrane as well as a change of mitochondrial transmembrane potentials occurred in an MLH1-dependent manner after treatment with TMZ, but no such MLH1 dependency was observed in the case of ACNU treatment. By using cell lines defective in both APAF-1 and MGMT, it was revealed that the APAF-1 function is required for execution of apoptosis induced by either TMZ or ACNU. There is almost 12h delay in occurrence of apoptosis-related mitochondrial depolarization in TMZ-treated cells in comparison to those of ACNU-treated cells, reflecting the fact that at least one cycle of DNA replication is required to trigger apoptosis in the former case, but not in the latter.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA Damage , DNA Repair , Dacarbazine/analogs & derivatives , Fibroblasts/drug effects , Nimustine/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Antineoplastic Agents/chemistry , Apoptosis/genetics , Cell Line , Dacarbazine/chemistry , Dacarbazine/pharmacology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Knockout , MutL Protein Homolog 1 , Nimustine/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/physiology , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/physiology , Phosphatidylserines/metabolism , Structure-Activity Relationship , Temozolomide
3.
Carcinogenesis ; 28(12): 2657-63, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17881774

ABSTRACT

O(6)-Methylguanine and O(6)-chloroethylguanine, which are the primary cytotoxic DNA lesions produced by 5-(3,3-dimethyl-1-triazeno)imidazole-4-carboxamide (dacarbazine) and 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea (ACNU), respectively, can be repaired by O(6)-methylguanine-DNA methyltransferase (MGMT), coded by the MGMT gene. However, the two types of drugs exhibit different effects on cells defective in both MGMT and MLH1 functions, the latter being related to the cellular activity to recognize mismatched bases of DNA for inducing apoptosis. Human cells deficient in both MGMT and MLH1 are resistant to the killing effect of dacarbazine and exhibit an increased mutant frequency after treatment with dacarbazine. On the other hand, these doubly deficient cells are sensitive to the killing action of ACNU and there is no significant increase in ACNU-induced mutant frequency. A mismatch recognition complex, composed of MSH2, MSH6, MLH1, PMS2 and PCNA, is formed after exposing MGMT-deficient cells to dacarbazine, but not in cells treated with ACNU. In contrast, the phosphorylation of Chk1 efficiently occurs in cells treated with dacarbazine as well as with ACNU, the former being in MLH1-dependent manner, whereas the latter in MLH1-independent manner. Therefore, the signals delivered from different sources would merge at the step of Chk1 activation or at an earlier step, and the subsequent process leading to apoptosis appears to be common.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Dacarbazine/pharmacology , Nimustine/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/metabolism , Checkpoint Kinase 1 , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Humans , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , MutS Homolog 2 Protein/metabolism , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
4.
DNA Repair (Amst) ; 3(4): 413-20, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15010317

ABSTRACT

Among various types of drugs designed for use in cancer chemotherapy, some have the potential for alkylation. After metabolic activation, these chemicals attack DNA and alkylate their bases, thereby preventing multiplication of rapidly growing tumor cells. Some of alkylated bases cause mutations, leading to untoward induction of tumors. To search for the rationale to separate lethal and mutagenic effects of alkylation drugs, we investigated actions of dacarbazine, a monofunctional triazene, on mouse and human cell lines defective in the Mgmt and/or the Mlh1 gene, the former encoding a DNA repair methyltransferase and the latter a protein involved in mismatch repair and induction of apoptosis. Mgmt-deficient cells are hypersensitive to the killing action of dacarbazine. On the other hand, cells defective in both Mgmt and Mlh1 genes are as resistant to the drug as are wild-type cells, in terms of survival, but do have many mutations after dacarbazine treatment. Thus, the killing and mutagenic actions of dacabazine can be dissociated by manipulating actions of these gene products.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Dacarbazine/toxicity , Mutation , Neoplasm Proteins/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins , Humans , Mice , MutL Protein Homolog 1 , Nuclear Proteins
5.
DNA Repair (Amst) ; 2(10): 1135-46, 2003 Oct 07.
Article in English | MEDLINE | ID: mdl-13679151

ABSTRACT

To examine involvement of mismatch repair system in alkylation-induced apoptosis and mutagenesis, cell lines defective in the Mgmt gene encoding a DNA repair enzyme, O(6)-methylguanine-DNA methyltransferase, and/or the Mlh1 gene encoding a protein involved in mismatch repair were established from gene-targeted mice. Mgmt(-/-) cells are hypersensitive to the killing effect of N-methyl-N-nitrosourea (MNU) and this effect of MNU was overcome by introducing an additional mutation in the Mlh1 gene. Mgmt(-/-)Mlh1(-/-) cells are more resistant to MNU than are wild-type cells. When the human Mgmt cDNA sequence with a strong promoter was introduced, the wild-type cells acquired the same high level of resistance to MNU as that of Mgmt(-/-)Mlh1(-/-) cells. Although no apparent increase in MNU-induced mutant frequency was observed in such methyltransferase-overproducing wild-type cells, mutant frequency of Mgmt(-/-)Mlh1(-/-) cells became 10-fold higher after being treated with MNU. Mgmt(-/-)Mlh1(+/-) cells carrying approximately half the normal level of MLH1 protein showed a normal level of spontaneous mutant frequency, yet were still highly responsive to the mutagenic effect of the alkylating carcinogen. This haploinsufficient character of Mlh1 mutation was also observed in cell survival assays; Mgmt(-/-)Mlh1(+/-) cells were as resistant to MNU as were Mgmt(-/-)Mlh1(-/-) cells. While caspase-3 was induced in Mgmt(-/-)Mlh1(+/+) cells after treatment with MNU, no induction occurred in Mgmt(-/-)Mlh1(+/-) cells or in Mgmt(-/-)Mlh1(-/-) cells. The cellular content of MLH1 protein seems to be critical for determining if damaged cells enter into either a death or mutation-inducing pathway. The haploinsufficient phenotype of Mlh1-heterozygous cells may be explained by competition in heterodimer formation between MLH1 homologues.


Subject(s)
Apoptosis/drug effects , Mutagenesis/drug effects , Neoplasm Proteins/physiology , O(6)-Methylguanine-DNA Methyltransferase/physiology , Adaptor Proteins, Signal Transducing , Alkylating Agents/pharmacology , Alkylation , Animals , Base Pair Mismatch , Carrier Proteins , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , Methylnitrosourea/pharmacology , Mice , Mice, Knockout , MutL Protein Homolog 1 , Mutation , Neoplasm Proteins/genetics , Nuclear Proteins , O(6)-Methylguanine-DNA Methyltransferase/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...