Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
4.
Mutat Res ; 480-481: 37-50, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11506797

RESUMO

The induction of double-strand breaks (DSBs) in DNA by exposure to DNA damaging agents or as intermediates in normal cellular processes, creates a severe threat for the integrity of the genome. Unrepaired or incorrectly repaired DSBs lead to broken chromosomes and/or gross chromosomal rearrangements which are frequently associated with tumor formation in mammals. To maintain the integrity of the genome and to prevent the formation of chromosomal aberrations, several pathways exist in eukaryotes: homologous recombination (HR), non-homologous end joining (NHEJ) and single-strand annealing (SSA). These mechanisms are conserved in evolution, but the relative contribution depends on the organism, cell type and stage of the cell cycle. In yeast, DSBs are primarily repaired via HR while in higher eukaryotes, both HR and NHEJ are important. In mammals, defects in both HR or NHEJ lead to a predisposition to cancer and at the cellular level, the frequency of chromosomal aberrations is increased. This review summarizes our current knowledge about DSB-repair with emphasis on recent progress in understanding the precise biochemical activities of individual proteins involved.


Assuntos
Quebra Cromossômica/fisiologia , Reparo do DNA/fisiologia , DNA/genética , DNA/metabolismo , Genoma , Animais , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila melanogaster/genética , Células Eucarióticas/metabolismo , Humanos , Recombinação Genética/fisiologia , Saccharomyces cerevisiae/genética , Schizosaccharomyces/genética
5.
Mutat Res ; 485(3): 177-85, 2001 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-11267829

RESUMO

Nijmegen breakage syndrome (NBS) and ataxia telangiectasia (AT) are rare autosomal recessive hereditary disorders characterized by radiosensitivity, chromosomal instability, immunodeficiency and proneness to cancer. Although the clinical features of both syndromes are quite distinct, the cellular characteristics are very similar. Cells from both NBS and AT patients are hypersensitive to ionizing radiation (IR), show elevated levels of chromosomal aberrations and display radioresistant DNA synthesis (RDS). The proteins defective in NBS and AT, NBS1 and ATM, respectively, are involved in the same pathway, but their exact relationship is not yet fully understood. Stumm et al. (Am. J. Hum. Genet. 60 (1997) 1246) have reported that hybrids of AT and NBS lymphoblasts were not complemented for chromosomal aberrations. In contrast, we found that X-ray-induced cell killing as well as chromosomal aberrations were complemented in proliferating NBS-1LBI/AT5BIVA hybrids, comparable to that in NBS-1LBI cells after transfer of a single human chromosome 8 providing the NBS1 gene. RDS observed in AT5BIVA cells was reduced in these hybrids to the level of that seen in immortal NBS-1LBI cells. However, the level of DNA synthesis, following ionizing radiation, in SV40 transformed wild-type cell lines was the same as in NBS-1LBI cells. Only primary wild-type cells showed stronger inhibition of DNA synthesis. In summary, these results clearly indicate that RDS cannot be used as an endpoint in functional complementation studies with immortal NBS-1LBI cells, whereas the cytogenetic assay is suitable for complementation studies with immortal AT and NBS cells.


Assuntos
Anormalidades Múltiplas/genética , Ataxia Telangiectasia/genética , Aberrações Cromossômicas , Tolerância a Radiação/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular , Morte Celular , Replicação do DNA/efeitos da radiação , Proteínas de Ligação a DNA , Relação Dose-Resposta à Radiação , Teste de Complementação Genética , Predisposição Genética para Doença , Humanos , Células Híbridas , Camundongos , Proteínas Nucleares , Proteínas Serina-Treonina Quinases , Síndrome , Proteínas Supressoras de Tumor , Raios X
6.
Genes Chromosomes Cancer ; 30(4): 323-35, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11241785

RESUMO

A combination of flow cytometry and microsatellite analysis was used to investigate loss of expression of HLA-A and/or HLA-B alleles and concurrent LOH at polymorphic chromosome 6 loci both in freshly isolated lymphocytes (in vivo mutations) and in lymphocytes cultured ex vivo. The fraction of in vivo mutants that showed LOH at 6p appeared to vary from 0%-49% for various donors. During culturing ex vivo, HLA-A(-) cells arose at a high rate and showed simultaneous loss of expression at the linked HLA-B locus. Up to 90% of the ex vivo arisen HLA-A2(-) cell population showed LOH of multiple 6p markers, and 50% had lost heterozygosity at 6q. This ex vivo spectrum resembles that found in HLA-A2 mutants obtained from lymphoblastoid cells. The HLA-A2 mutants present in vivo may reflect only a small fraction of the mutants that can be detected ex vivo. In normal lymphocytes, in vivo only mitotic recombination appears to be sustained, indicating the importance of this mechanism for tumor initiation in normal cells. Although mutations resulting in LOH at both chromosome 6 arms were shown to result in nonviable cells in normal lymphocytes, they have been shown to result in viable mutants in lymphoblastoid cells. We hypothesize that these types of mutations also occur in vivo but only survive in cells that already harbor a mutated genetic background. In light of the high rate at which these types of mutations occur, they may contribute to cancer progression.


Assuntos
Perda de Heterozigosidade/genética , Linfócitos T/metabolismo , Células Cultivadas , Análise Mutacional de DNA , Citometria de Fluxo , Antígeno HLA-A2/biossíntese , Antígeno HLA-A2/genética , Antígeno HLA-A3/biossíntese , Antígeno HLA-A3/genética , Teste de Histocompatibilidade , Humanos , Contagem de Linfócitos , Repetições de Microssatélites/genética , Deleção de Sequência/genética , Linfócitos T/química
7.
Mutat Res ; 461(1): 59-69, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10980412

RESUMO

The DNA-dependent protein kinase (DNA-PK) complex plays a key role in DNA double-strand break (DSB) repair and V(D)J recombination. Using a genetic approach we have isolated cell mutants sensitive to ionizing radiation (IR) in the hope of elucidating the mechanism and components required for these pathways. We describe here, an X-ray-sensitive and DSB repair defective Chinese hamster ovary (CHO) cell line, XR-C2, which was assigned to the X-Ray Cross Complementation (XRCC) group 7. This group of mutants is defective in the XRCC7/SCID/Prkdc gene, which encodes the catalytic subunit of DNA-PK (DNA-PKcs). Despite the fact that XR-C2 cells expressed normal levels of DNA-PKcs protein, no DNA-PK catalytic activity could be observed in XR-C2, confirming the genetic analyses that these cells harbor a dysfunctional gene for DNA-PKcs. In contrast to other IR group 7 mutants, which contain undetectable or low levels of DNA-PKcs protein and which show a severe defect in V(D)J recombination, XR-C2 cells manifested only a mild defect in both coding and signal junction formation. The unique phenotype of the XR-C2 mutant suggests that a normal level of kinase activity is critical for radiation resistance but not for V(D)J recombination, whereas the overall structure of the DNA-PKcs protein appears to be of great importance for this process.


Assuntos
Reparo do DNA/genética , Proteínas de Ligação a DNA , Mutação , Proteínas Serina-Treonina Quinases/genética , Tolerância a Radiação/genética , Recombinação Genética/genética , Animais , Células CHO , Cricetinae , Proteína Quinase Ativada por DNA , Relação Dose-Resposta à Radiação , Teste de Complementação Genética , Mutagênicos/farmacologia , Raios X
8.
Proc Natl Acad Sci U S A ; 97(15): 8403-8, 2000 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-10900004

RESUMO

Somatic mutation accumulation has been implicated as a major cause of cancer and aging. By using a transgenic mouse model with a chromosomally integrated lacZ reporter gene, mutational spectra were characterized at young and old age in two organs greatly differing in proliferative activity, i.e., the heart and small intestine. At young age the spectra were nearly identical, mainly consisting of G. C to A.T transitions and 1-bp deletions. At old age, however, distinct patterns of mutations had developed. In small intestine, only point mutations were found to accumulate, including G.C to T.A, G.C to C.G, and A.T to C.G transversions and G.C to A.T transitions. In contrast, in heart about half of the accumulated mutations appeared to be large genome rearrangements, involving up to 34 centimorgans of chromosomal DNA. Virtually all other mutations accumulating in the heart appeared to be G.C to A.T transitions at CpG sites. These results suggest that distinct mechanisms lead to organ-specific genome deterioration and dysfunction at old age.


Assuntos
Envelhecimento/genética , Coração , Intestino Delgado , Mutação , Animais , Linhagem Celular Transformada , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação Puntual
9.
Mutat Res ; 428(1-2): 237-54, 1999 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-10517997

RESUMO

Numerous reactive mutagenic electrophiles are present in the environment or are formed in the human body through metabolizing processes. Those electrophiles can directly react with DNA and are considered to be ultimate carcinogens. In the past decades more than 200 in vitro and in vivo genotoxic tests have been described to identify, monitor and characterize the exposure of humans to such agents. When the responses of such genotoxic tests are quantified by a weight-of-evidence analysis, it is found that the intrinsic potency of electrophiles being mutagens does not differ much for the majority of the agents studied. Considering the fact that under normal environmental circumstances human are exposed to low concentration of about a million electrophiles, the relation between exposure to such agents and adverse health effects (e.g., cancer) will become a 'Pandora's box'. For quantitative risk assessment it will be necessary not only to detect whether the agent is genotoxic, but also understand the mechanism of interaction of the agent with the DNA in target cells needs to be taken into account. Examples are given for a limited group of important environmental and carcinogenic agents for which such an approach is feasible. The groups identified are agents that form cross-links with DNA or are mono-alkylating agents that react with base-moieties in the DNA strands. Quantitative hazard ranking of the mutagenic potency of these groups of chemical can be performed and there is ample evidence that such a ranking corresponds with the individual carcinogenic potency of those agents in rodents. Still, in practice, with the exception of certain occupational or accidental exposure situations, these approaches have not be successful in preventing cancer death in the human population. However, this is not only due to the described 'Pandora's box' situation. At least three other factors are described. Firstly, in the industrial world the medical treatment of cancer in patients occurs with high levels of extremely mutagenic agents. Actually, both in number of persons and in exposure levels such medical treatment is the single largest exposure of humans to known carcinogens. Although such treatments are very effective in curing the tumor as present in the patient, the recurrence of cancer in those patients later in life is very high. In other words: "curing cancer is not the same as preventing cancer death in the human population". Secondly, the rate of cancer death in the human population is also determined by the efficacy in which other major causes of death are prevented. For instance, cardiovascular diseases are the major cause of death in humans in the industrialized world. There is evidence that the treatment of cardiovascular diseases is more successful than that of cancer. On a population level this will result in increase of cancer being the ultimate death cause. Finally, the improvement of medical treatment of diseases together with an improved quality of life will lead to increase average age of the population. Because the onset of most cancer is long after the exposure to carcinogens-in human often more than 30 years-cancer is predominantly a disease of the old age. This means that if the average age of human increases, there will be a selective preference of cancer becoming an even more important cause of death. This especially will be pronounced in those countries were the age distribution in a population is abnormal.


Assuntos
Carcinógenos Ambientais/toxicidade , Mutagênicos/toxicidade , Medição de Risco/métodos , Envelhecimento , Carcinógenos Ambientais/metabolismo , DNA/efeitos dos fármacos , DNA/metabolismo , Dano ao DNA , Exposição Ambiental , Feminino , Humanos , Masculino , Testes de Mutagenicidade/métodos , Testes de Mutagenicidade/estatística & dados numéricos , Mutagênicos/metabolismo , Neoplasias/etiologia , Neoplasias/mortalidade , Neoplasias/prevenção & controle , Países Baixos/epidemiologia , Dinâmica Populacional
10.
Nucleic Acids Res ; 27(16): 3276-82, 1999 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10454634

RESUMO

In this study the role of nucleotide excision repair (NER) in protecting mouse embryonic stem (ES) cells against the genotoxic effects of UV-photolesions was analysed. Repair of cyclobutane pyrimidine dimers (CPD) in transcribed genes could not be detected whereas the removal of (6-4) photoproducts (6-4PP) was incomplete, already reaching its maximum (30%) 4 h after irradiation. Measurements of repair replication revealed a saturation of NER activity at UV doses >5 J/m2 while at a lower dose (2.5 J/m2) the repair kinetics were similar to those in murine embryonic fibroblasts (MEFs). Cytotoxic and mutagenic effects of photolesions were determined in ES cells differing in NER activity. ERCC1-deficient ES cells were hypermutable (10-fold) compared to wild-type cells, indicating that at physiologically relevant doses ES cells efficiently remove photolesions. The effect of the NER deficiency on cytoxicity was only 2-fold. Exposure to high UV doses (10 J/m2) resulted in a rapid and massive induction of apoptosis. Possibly, to avoid the accumulation of mutated cells, ES cells rely on the induction of a strong apoptotic response with a simultaneous shutting down of NER activity.


Assuntos
Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA , Embrião de Mamíferos/efeitos da radiação , Endonucleases , Células-Tronco/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Linhagem Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Genes p53 , Hipoxantina Fosforribosiltransferase/genética , Camundongos , Mutação , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-mos/genética , Dímeros de Pirimidina/genética , Células-Tronco/metabolismo , Raios Ultravioleta
11.
Mutat Res ; 434(1): 17-27, 1999 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-10377945

RESUMO

Nijmegen Breakage Syndrome (NBS) is a very rare autosomal recessive chromosomal instability disorder characterized by microcephaly, growth retardation, immunodeficiency and a high incidence of malignancies. Cells from NBS patients are hypersensitive to ionizing radiation (IR) and display radioresistant DNA synthesis (RDS). NBS is caused by mutations in the NBS1 gene on chromosome 8q21 encoding a protein called nibrin. This protein is a component of the hMre11/hRad50 protein complex, suggesting a defect in DNA double-strand break (DSB) repair and/or cell cycle checkpoint function in NBS cells. We established SV40 transformed, immortal NBS fibroblasts, from primary cells derived from a Polish patient, carrying the common founder mutation 657del5. Immortalized NBS cells, like primary cells, are X-ray sensitive (2-fold) and display RDS following IR. They show an increased sensitivity to bleomycin (3.5-fold), etoposide (2.5-fold), camptothecin (3-fold) and mitomycin C (1.5-fold), but normal sensitivity towards UV-C. Despite the clear hypersensitivity towards DSB-inducing agents, the overall rates of DSB-rejoining in NBS cells as measured by pulsed field gel electrophoresis were found to be very similar to those of wild type cells. This indicates that the X-ray sensitivity of NBS cells is not directly caused by an overt defect in DSB repair.


Assuntos
Anormalidades Múltiplas/genética , Transformação Celular Viral , Quebra Cromossômica , Fibroblastos/virologia , Anormalidades Múltiplas/patologia , Antineoplásicos/farmacologia , Bleomicina/farmacologia , Camptotecina/farmacologia , Linhagem Celular , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Pré-Escolar , DNA/efeitos dos fármacos , DNA/genética , DNA/efeitos da radiação , Dano ao DNA , Reparo do DNA , Etoposídeo/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Células HeLa , Humanos , Mitomicina/farmacologia , Mutação , Síndrome , Raios X
12.
Nucleic Acids Res ; 27(8): 1795-801, 1999 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10101186

RESUMO

The protein O 6-alkylguanine-DNA alkyltransferase(alkyltransferase) is involved in the repair of O 6-alkylguanine and O 4-alkylthymine in DNA and plays an important role in most organisms in attenuating the cytotoxic and mutagenic effects of certain classes of alkylating agents. A genomic clone encompassing the Drosophila melanogaster alkyltransferase gene ( DmAGT ) was identified on the basis of sequence homology with corresponding genes in Saccharomyces cerevisiae and man. The DmAGT gene is located at position 84A on the third chromosome. The nucleotide sequence of DmAGT cDNA revealed an open reading frame encoding 194 amino acids. The MNNG-hypersensitive phenotype of alkyltransferase-deficient bacteria was rescued by expression of the DmAGT cDNA. Furthermore, alkyltransferase activity was identified in crude extracts of Escherichia coli harbouring DmAGT cDNA and this activity was inhibited by preincubation of the extract with an oligonucleotide containing a single O6-methylguanine lesion. Similar to E.coli Ogt and yeast alkyltransferase but in contrast to the human alkyltransferase, the Drosophila alkyltransferase is resistant to inactivation by O 6-benzylguanine. In an E.coli lac Z reversion assay, expression of DmAGT efficiently suppressed MNNG-induced G:C-->A:T as well as A:T-->G:C transition mutations in vivo. These results demonstrate the presence of an alkyltransferase specific for the repair of O 6-methylguanine and O 4-methylthymine in Drosophila.


Assuntos
Drosophila melanogaster/enzimologia , Proteínas de Escherichia coli , Metiltransferases , O(6)-Metilguanina-DNA Metiltransferase/genética , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Sequência de Bases , Clonagem Molecular , Reparo do DNA , DNA Complementar , Drosophila melanogaster/genética , Inibidores Enzimáticos/farmacologia , Escherichia coli , Deleção de Genes , Guanina/análogos & derivados , Guanina/metabolismo , Guanina/farmacologia , Humanos , Metilnitronitrosoguanidina , Dados de Sequência Molecular , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Fenótipo , Timina/análogos & derivados , Timina/metabolismo , Fatores de Transcrição
13.
Mutat Res ; 423(1-2): 1-10, 1999 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-10029664

RESUMO

Loss of heterozygosity (LOH) contributes significantly to the inactivation of tumor suppressor genes and may involve a variety of mechanisms. Studying loss of HLA-A2 alleles in human lymphoblastoid cell lines, we previously showed that mitotic recombination and chromosome loss with concomitant duplication of the non-selected chromosome were the most frequent mechanisms of LOH. In the present study we used the HLA system to determine the rate and spectrum of LOH mutations in the EBV transformed lymphoblastoid cell line R83-4915. Spontaneous loss of HLA-A2 in R83-4915 occurred with a rate of 7.9x10-7 which was 5 to 10-times lower compared to the previously observed rate of loss of HLA-A2 in other lymphoblastoid cell lines. Among the HLA-A2 mutants, 27% did not show LOH of additional chromosome 6 markers. Molecular analysis showed that neither large deletion nor gene conversion was the cause for their mutant phenotype. The remaining mutants showed LOH, which was caused by mitotic recombination (40%) and chromosome loss (33%). However, the chromosome loss observed in mutants of R83-4915 was not accompanied by the duplication of the remaining chromosome. Instead 3 out of 5 mutants became polyploid suggesting that different mechanisms exist to compensate for chromosome loss. In conclusion, the rate and types of LOH that can be observed in cell lines obtained from various donors may depend on the genetic make-up or the transformation status of these cells


Assuntos
Perda de Heterozigosidade/genética , Linfócitos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Transformada , Células Cultivadas , Deleção Cromossômica , Células Clonais , DNA/análise , Antígeno HLA-A2/genética , Humanos , Hibridização in Situ Fluorescente , Linfócitos/química , Linfócitos/imunologia
15.
Proc Natl Acad Sci U S A ; 95(23): 13759-64, 1998 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-9811874

RESUMO

Genetic events leading to the loss of heterozygosity (LOH) have been shown to play a crucial role in the development of cancer. However, LOH events do not occur only in genetically unstable cancer cells but also have been detected in normal somatic cells of mouse and man. Mice, in which one of the alleles for adenine phosphoribosyltransferase (Aprt) has been disrupted by gene targeting, were used to investigate the potency of carcinogens to induce LOH in vivo. After 7,12-dimethyl-1,2-benz[a]anthracene (DMBA) exposure, a 3-fold stronger mutagenic response was detected at the autosomal Aprt gene than at the X chromosomal hypoxantine-guanine phosphoribosyltransferase (Hprt) gene in splenic T-lymphocytes. Allele-specific PCR analysis showed that the normal, nontargeted Aprt allele was lost in 70% of the DMBA-induced Aprt mutants. Fluorescence in situ hybridization analysis demonstrated that the targeted allele had become duplicated in almost all DMBA-induced mutants that displayed LOH at Aprt. These results indicate that the main mechanisms by which DMBA caused LOH were mitotic recombination or chromosome loss and duplication but not deletion. However, after treatment with the alkylating agent N-ethyl-N-nitrosourea, Aprt had a similar mutagenic response to Hprt while the majority (90%) of N-ethyl-N-nitrosourea-induced Aprt mutants had retained both alleles. Unexpectedly, irradiation with x-rays, which induce primarily large deletions, resulted in a significant increase of the mutant frequency at Hprt but not at Aprt. This in vivo study clearly indicates that, in normal somatic cells, carcinogen exposure can result in the induction of LOH events that are compatible with cell survival and may represent an initiating event in tumorigenesis.


Assuntos
9,10-Dimetil-1,2-benzantraceno/toxicidade , Adenina Fosforribosiltransferase/genética , Carcinógenos/toxicidade , Perda de Heterozigosidade/genética , Alelos , Animais , Feminino , Humanos , Perda de Heterozigosidade/efeitos dos fármacos , Masculino , Camundongos , Neoplasias/genética
16.
Mol Cell Biol ; 18(11): 6423-9, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9774658

RESUMO

The RAD52 epistasis group is required for recombinational repair of double-strand breaks (DSBs) and shows strong evolutionary conservation. In Saccharomyces cerevisiae, RAD52 is one of the key members in this pathway. Strains with mutations in this gene show strong hypersensitivity to DNA-damaging agents and defects in recombination. Inactivation of the mouse homologue of RAD52 in embryonic stem (ES) cells resulted in a reduced frequency of homologous recombination. Unlike the yeast Scrad52 mutant, MmRAD52(-/-) ES cells were not hypersensitive to agents that induce DSBs. MmRAD52 null mutant mice showed no abnormalities in viability, fertility, and the immune system. These results show that, as in S. cerevisiae, MmRAD52 is involved in recombination, although the repair of DNA damage is not affected upon inactivation, indicating that MmRAD52 may be involved in certain types of DSB repair processes and not in others. The effect of inactivating MmRAD52 suggests the presence of genes functionally related to MmRAD52, which can partly compensate for the absence of MmRad52 protein.


Assuntos
Dano ao DNA/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/fisiologia , Recombinação Genética/genética , Animais , Linfócitos B/metabolismo , Sobrevivência Celular/efeitos da radiação , Citometria de Fluxo , Região de Troca de Imunoglobulinas/genética , Camundongos , Camundongos Knockout , Fenótipo , Proteína Rad52 de Recombinação e Reparo de DNA , Radiação Ionizante , Saccharomyces cerevisiae/fisiologia , Células-Tronco/metabolismo , Linfócitos T/metabolismo , Raios X
17.
Mutat Res ; 400(1-2): 509-40, 1998 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-9685708

RESUMO

In the past years, several methodologies were developed for potency ranking of genotoxic carcinogens and germ cell mutagens. In this paper, we analyzed six sub-classes of covalent deoxyribonucleic acid (DNA) binding antineoplastic drugs comprising a total of 37 chemicals and, in addition, four alkyl-epoxides, using four approaches for the ranking of genotoxic agents on a potency scale: the EPA/IARC genetic activity profile (GAP) database, the ICPEMC agent score system, and the analysis of qualitative and quantitative structure-activity and activity-activity relationships (SARs, AARs) between types of DNA modifications and genotoxic endpoints. Considerations of SARs and AARs focused entirely on in vivo data for mutagenicity in male germ cells (mouse, Drosophila), carcinogenicity (TD50s) and acute toxicity (LD50s) in rodents, whereas the former two approaches combined the entire database on in vivo and in vitro mutagenicity tests. The analysis shows that the understanding and prediction of rank positions of individual genotoxic agents requires information on their mechanism of action. Based on SARs and AARs, the covalent DNA binding antineoplastic drugs can be divided into three categories. Category 1 comprises mono-functional alkylating agents that primarily react with N7 and N3 moieties of purines in DNA. Efficient DNA repair is the major protective mechanism for their low and often not measurable genotoxic effects in repair-competent germ cells, and the need of high exposure doses for tumor induction in rodents. Due to cell type related differences in the efficiency of DNA repair, a strong target cell specificity in various species regarding the potency of these agents for adverse effects is found. Three of the four evaluation systems rank category 1 agents lower than those of the other two categories. Category 2 type mutagens produce O-alkyl adducts in DNA in addition to N-alkyl adducts. In general, certain O-alkyl DNA adducts appear to be slowly repaired, or even not at all, which make this kind of agents potent carcinogens and germ cell mutagens. Especially the inefficient repair of O-alkyl-pyrimidines causes the high mutational response of cells to these agents. Agents of this category give high potency scores in all four expert systems. The major determinant for the high rank positions on any scale of genotoxic of category 3 agents is their ability to induce primarily structural chromosomal changes. These agents are able to cross-link DNA. Their high intrinsic genotoxic potency appears to be related to the number of DNA cross-links per target dose unit they can induce. A confounding factor among category 3 agents is that often the genotoxic endpoints occur close to or at toxic levels, and that the width of the mutagenic dose range, i.e., the dose area between the lowest observed effect level and the LD50, is smaller (usually no more than 1 logarithmic unit) than for chemicals of the other two categories. For all three categories of genotoxic agents, strong correlations are observed between their carcinogenic potency, acute toxicity and germ cell specificity.


Assuntos
Antineoplásicos/metabolismo , Testes de Carcinogenicidade , DNA/metabolismo , Testes de Mutagenicidade , Animais , Testes de Carcinogenicidade/métodos , Testes de Carcinogenicidade/estatística & dados numéricos , Aberrações Cromossômicas , Deleção Cromossômica , Reagentes de Ligações Cruzadas , Bases de Dados Factuais , Relação Dose-Resposta a Droga , Genes/efeitos dos fármacos , Células Germinativas/efeitos dos fármacos , Humanos , Dose Letal Mediana , Meiose/efeitos dos fármacos , Testes de Mutagenicidade/métodos , Testes de Mutagenicidade/estatística & dados numéricos , Mutação/efeitos dos fármacos , Mutação/genética , Segunda Neoplasia Primária/induzido quimicamente , Valor Preditivo dos Testes , Medição de Risco , Especificidade da Espécie
18.
Environ Mol Mutagen ; 31(1): 18-31, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9464312

RESUMO

To investigate tissue-specific relations between DNA adducts and mutagenesis in vivo, lambda lacZ transgenic mice were treated i.p. with N-ethyl-N-nitrosourea (ENU), diethylnitrosamine (DEN), and ethyl methanesulphonate (EMS). In liver, bone marrow, and brain DNA from mice sacrificed at several time points after treatment O6-ethylguanine (O6-EtG) and N7-ethylguanine (N7-EtG) levels were determined as well as the mutant frequency (MF) in lacZ. In liver DNA of ENU- and DEN-treated mice, the bulk of O6-EtG was removed at 3 days after treatment, while the MF continued to increase thereafter. This suggests that O6-EtG is not the major premutagenic lesion in the liver. Indeed, sequence analysis of mutants showed only 24% GC-->AT transitions, consistent with the O6-EtG lesion, and 28% TA-->AT transversions, expected from O2-ethylthymine. In bone marrow after ENU treatment, a maximum mutation induction occurred at 3 days post-treatment, of which 43% were GC-->AT mutations and 22% were TA-->AT mutations. This suggests that in bone marrow O6-EtG may be a major premutagenic lesion at the 3-day time point. In liver and bone marrow, EMS treatment gave rise to a high level of N7-EtG and a low level of O6-EtG but no increase in MF. No adducts or mutation induction were observed in bone marrow of DEN-treated mice. No MF increase was observed in the brain of either ENU- or EMS-treated mice, although O6- and N7-adducts were present.


Assuntos
Adutos de DNA , Dietilnitrosamina/toxicidade , Metanossulfonato de Etila/toxicidade , Etilnitrosoureia/toxicidade , Óperon Lac , Mutagênicos/toxicidade , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Guanina/análogos & derivados , Guanina/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Transgênicos
19.
Genes Chromosomes Cancer ; 21(1): 30-8, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9443039

RESUMO

Loss of heterozygosity (LOH) plays an important role in the expression of recessive mutations in mammalian cells. To gain insight into the rate and mechanisms of LOH the autosomal HLA-A gene was used as a model system. Spontaneous HLA-A2 mutants originated with a rate of respectively 4.1 x 10(-6) and 6.9 x 10(-6) per cell per generation in TK6 and WI-L2-NS, two isogenic lymphoblastoid cell lines which differ in TP53 status. The rate of loss of HLA-A2 is 10-50 times higher compared to the mutation rate of the X-linked HPRT gene. The homozygous TP53 mutation in WI-L2-NS had no effect on the rate of HLA-A2 loss or the spectrum of these mutations. Microsatellite analysis of most of the HLA-A2 mutants (84%) showed LOH for multiple markers on chromosome arm 6p telomeric of a recombination breakpoint, LOH for all 6p markers, or LOH for markers on both the 6p- and 6q-arms. Cytogenetic analysis showed that these mechanisms gave mutant cells which harbored two intact chromosomes 6 and which were indistinguishable from non-mutant cells. Therefore, loss of HLA-A2 is mainly caused by somatic recombination (33-50%) or chromosome loss with duplication of the remaining chromosome (34-40%). These findings correspond to the mechanisms behind loss of the wild-type RBI allele in retinoblastoma and suggest that both somatic recombination and chromosome loss followed by duplication contribute to tumorigenesis.


Assuntos
Deleção Cromossômica , Antígenos HLA-A/genética , Perda de Heterozigosidade , Recombinação Genética , Linhagem Celular Transformada , Cromossomos Humanos Par 6/genética , Humanos , Hibridização in Situ Fluorescente
20.
Mol Cell Biol ; 17(10): 6097-104, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9315669

RESUMO

The RAD54 gene of Saccharomyces cerevisiae plays a crucial role in recombinational repair of double-strand breaks in DNA. Here the isolation and functional characterization of the RAD54 homolog of the fruit fly Drosophila melanogaster, DmRAD54, are described. The putative Dmrad54 protein displays 46 to 57% identity to its homologs from yeast and mammals. DmRAD54 RNA was detected at all stages of fly development, but an increased level was observed in early embryos and ovarian tissue. To determine the function of DmRAD54, a null mutant was isolated by random mutagenesis. DmRADS4-deficient flies develop normally, but the females are sterile. Early development appears normal, but the eggs do not hatch, indicating an essential role for DmRAD54 in development. The larvae of mutant flies are highly sensitive to X rays and methyl methanesulfonate. Moreover, this mutant is defective in X-ray-induced mitotic recombination as measured by a somatic mutation and recombination test. These phenotypes are consistent with a defect in the repair of double-strand breaks and imply that the RAD54 gene is crucial in repair and recombination in a multicellular organism. The results also indicate that the recombinational repair pathway is functionally conserved in evolution.


Assuntos
Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila , Drosophila melanogaster/genética , Proteínas do Ovo , Recombinação Genética/fisiologia , Sequência de Aminoácidos , Animais , Dano ao DNA , DNA Helicases , Proteínas de Ligação a DNA/fisiologia , Drosophila melanogaster/embriologia , Drosophila melanogaster/crescimento & desenvolvimento , Olho/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos/genética , Genes de Insetos/fisiologia , Larva/efeitos dos fármacos , Larva/efeitos da radiação , Masculino , Metanossulfonato de Metila/farmacologia , Mitose/genética , Dados de Sequência Molecular , Mutagênese , Mutagênicos/farmacologia , RNA Mensageiro/análise , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA