Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Bull Cancer ; 98(3): 291-303, 2011 Mar.
Artigo em Francês | MEDLINE | ID: mdl-21459716

RESUMO

MGMT status is now regarded as a strong predictive factor of response to standard treatment of newly diagnosed glioblastomas involving temozolomide (TMZ) and radiotherapy. MGMT promoter methylation is also a prognostic factor - independent of treatment - in anaplastic gliomas. The predictive function can be explained by the role of the DNA repair enzyme MGMT, which antagonizes the effects of alkylating agents such as TMZ. MGMT promoter methylation could also reflect a particular molecular phenotype with its own specific prognostic significance. Since MGMT status determination is becoming a crucial biological marker in new clinical glioma trials, and is beginning to be used in day-to-day clinical practice, there is currently a strong need to determine the best technique for MGMT analysis. A French multicenter study has been set up for this purpose.


Assuntos
Glioma/enzimologia , Proteínas de Neoplasias/análise , O(6)-Metilguanina-DNA Metiltransferase/análise , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/enzimologia , Reparo do DNA , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Glioma/tratamento farmacológico , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Prognóstico , Temozolomida
2.
Carcinogenesis ; 31(12): 2111-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20732909

RESUMO

Methylating agents are widely distributed environmental carcinogens. Moreover, they are being used in cancer chemotherapy. The primary target of methylating agents is DNA, and therefore, DNA repair is the first-line barrier in defense against their toxic and carcinogenic effects. Methylating agents induce in the DNA O(6)-methylguanine (O(6)MeG) and methylations of the ring nitrogens of purines. The lesions are repaired by O(6)-methylguanine-DNA methyltransferase (Mgmt) and by enzymes of the base excision repair (BER) pathway, respectively. Whereas O(6)MeG is well established as a pre-carcinogenic lesion, little is known about the carcinogenic potency of base N-alkylation products such as N3-methyladenine and N3-methylguanine. To determine their role in cancer formation and the role of BER in cancer protection, we checked the response of mice with a targeted gene disruption of Mgmt or N-alkylpurine-DNA glycosylase (Aag) or both Mgmt and Aag, to azoxymethane (AOM)-induced colon carcinogenesis, using non-invasive mini-colonoscopy. We demonstrate that both Mgmt- and Aag-null mice show a higher colon cancer frequency than the wild-type. With a single low dose of AOM (3 mg/kg) Aag-null mice showed an even stronger tumor response than Mgmt-null mice. The data provide evidence that both BER initiated by Aag and O(6)MeG reversal by Mgmt are required for protection against alkylation-induced colon carcinogenesis. Further, the data indicate that non-repaired N-methylpurines are not only pre-toxic but also pre-carcinogenic DNA lesions.


Assuntos
Neoplasias do Colo/prevenção & controle , Reparo do DNA , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Animais , Neoplasias do Colo/genética , DNA Glicosilases/fisiologia , Feminino , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL
3.
Biochem Pharmacol ; 80(9): 1317-25, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20654586

RESUMO

O(6)-Alkylguanine-DNA alkyltransferase (AGT) mediates tumor resistance to alkylating agents that generate guanine O(6)-chloroethyl (Onrigin™ and carmustine) and O(6)-methyl (temozolomide) lesions; however, the relative efficiency of AGT protection against these lesions and the degree of resistance to these agents that a given number of AGT molecules produces are unclear. Measured from differential cytotoxicity in AGT-ablated and AGT-intact HL-60 cells containing 17,000 AGT molecules/cell, AGT produced 12- and 24-fold resistance to chloroethylating (90CE) and methylating (KS90) analogs of Onrigin™, respectively. For 50% growth inhibition, KS90 and 90CE generated 5,600 O(6)-methylguanines/cell and ∼300 O(6)-chloroethylguanines/cell, respectively. AGT repaired O(6)-methylguanines until the AGT pool was exhausted, while its repair of O(6)-chloroethylguanines was incomplete due to progression of the lesions to AGT-irreparable interstrand DNA cross-links. Thus, the smaller number of O(6)-chloroethylguanine lesions needed for cytotoxicity accounted for the marked degree of resistance (12-fold) to 90CE produced by AGT. Transfection of human or murine AGT into AGT deficient transplantable tumor cells (i.e., EMT6, M109 and U251) generated transfectants expressing AGT ranging from 4,000 to 700,000 molecules/cell. In vitro growth inhibition assays using these transfectants treated with 90CE revealed that AGT caused a concentration dependent resistance up to a level of ∼10,000 AGT molecules/cell. This finding was corroborated by in vivo studies where expression of 4,000 and 10,000 murine AGT molecules/cell rendered EMT6 tumors partially and completely resistant to Onrigin™, respectively. These studies imply that the antitumor activity of Onrigin™ stems from guanine O(6)-chloroethylation and define the threshold concentration of AGT that negates its antineoplastic activity.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA , Guanina/análogos & derivados , Hidrazinas/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Sulfonamidas/farmacologia , Animais , Carmustina/farmacologia , Linhagem Celular Tumoral , Metilação de DNA , Reparo do DNA , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Guanina/análise , Humanos , Camundongos , O(6)-Metilguanina-DNA Metiltransferase/análise , Temozolomida
4.
Biochem Pharmacol ; 79(11): 1553-61, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20005211

RESUMO

1,2-Bis(methylsulfonyl)-1-(2-chloroethyl)-2-[[1-(4-nitrophenyl)ethoxy]carbonyl]hydrazine (KS119) is a prodrug of the 1,2-bis(sulfonyl)hydrazine class of antineoplastic agents designed to exploit the oxygen-deficient regions of cancerous tissue. Thus, under reductive conditions in hypoxic cells this agent decomposes to produce the reactive intermediate 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE), which in turn generates products that alkylate the O(6)-position of guanine in DNA. Comparison of the cytotoxicity of KS119 in cultured cells lacking O(6)-alkylguanine-DNA alkyltransferase (AGT) to an agent such as Onrigin, which through base catalyzed activation produces the same critical DNA G-C cross-link lesions by the generation of 90CE, indicates that KS119 is substantially more potent than Onrigin under conditions of oxygen deficiency, despite being incompletely activated. In cell lines expressing relatively large amounts of AGT, the design of the prodrug KS119, which requires intracellular activation by reductase enzymes to produce a cytotoxic effect, results in an ability to overcome resistance derived from the expression of AGT. This appears to derive from the ability of a small portion of the chloroethylating species produced by the activation of KS119 to slip through the cellular protection afforded by AGT to generate the few DNA G-C cross-links that are required for tumor cell lethality. The findings also demonstrate that activation of KS119 under oxygen-deficient conditions is ubiquitous, occurring in all of the cell lines tested thus far, suggesting that the enzymes required for reductive activation of this agent are widely distributed in many different tumor types.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Hidrazinas/farmacologia , Hipóxia , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Pró-Fármacos/farmacologia , Animais , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Humanos , Hidrazinas/farmacocinética , O(6)-Metilguanina-DNA Metiltransferase/deficiência , Oxirredução , Oxirredutases/metabolismo , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacocinética
5.
Anticancer Res ; 29(10): 3759-68, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19846906

RESUMO

O(6)-Methylguanine-DNA-methyltransferase (MGMT) is a unique protein, which both repairs O(6)-alkylguanine lesions stoichiometrically without a multi-enzymatic pathway and self-inactivates. It has recently been linked to the therapeutic success of alkylating agent chemotherapy, specifically temozolomide treatment. This drug affects the MGMT pathway to induce cell death in tumor tissue. Low levels of functional MGMT have been correlated with success of treatment, while high levels bring about failure of therapy. Expression of MGMT protein varies in normal and tumoral tissue. Furthermore, its epigenetic silencing due to promoter methylation has been linked to its lack of expression in many types of tumor, including gliomas. Great enthusiasm surrounds the utility of this protein in cancer treatment. Not only has there been success in manipulating MGMT levels to enhance alkylating agent therapy, but studies also suggest a possible role of MGMT in protecting hematopoietic cells from the myelosuppressive effects of high-dose chemotherapy. Innovative research into this protein will no doubt be rewarding. This review presents a summary of what is known about this unique protein, including its structure, function in its pathway, polymorphisms, expression in normal and tumoral tissue, relation to alkylating agent therapy, and possible future applications.


Assuntos
Neoplasias/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Humanos , Neoplasias/diagnóstico , Neoplasias/patologia , Neoplasias/terapia , O(6)-Metilguanina-DNA Metiltransferase/química , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Prognóstico
6.
Brain Nerve ; 61(7): 849-54, 2009 Jul.
Artigo em Japonês | MEDLINE | ID: mdl-19618863

RESUMO

Older patients are frequently excluded from randomized studies; further, it is unclear whether the morbidity associated with chemoradiotherapy with temozolomide (TMZ) outweighs the possible survival benefit in this population. TMZ administered at a dose of 150-200 mg/m2 for 5 days every 4 weeks is the standard of care in operated glioblastoma (GBM) after concurrent chemoradiotherapy. Alternative dosing regimens, such as 1-week on/1-week off, or 3-week on/1-week off, that deliver more prolonged exposure have been observed to result in higher cumulative doses than the standard 5-day regimen and may deplete tumor-derived O6-methylguanine-DNA methyltransferase (MGMT) in tumor cells, thus sensitizing tumor cells to the effects of TMZ. Currently, chemotherapy with TMZ is an interesting alternative to radiotherapy in patients with very large tumors or in the elderly who are exposed to a higher risk of delayed neurotoxicity. The DNA damage induced by nitrosoureas and TMZ is partially repaired by MGMT. Thus, administration of the combination of nitrosoureas and TMZ might overcome MGMT-mediated resistance via MGMT depletion, yielding superior treatment results compared to the administration of treatment alone. However, the results of 2 studies that administered BCNU and CCNU with TMZ reported contradictory results. The introduction of TMZ has enabled the extension of chemotherapy treatment by 1-3 years due to the improved toxicity profile and lack of cumulative toxicity. Treatment-induced myelodysplastic syndrome with or without acute myeloblastic leukemia is a well-recognized late treatment-related complication associated with TMZ administration.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Glioblastoma/terapia , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Alquilantes/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/enzimologia , Carmustina/administração & dosagem , Dano ao DNA , Dacarbazina/administração & dosagem , Dacarbazina/efeitos adversos , Glioblastoma/enzimologia , Humanos , Leucemia Mieloide Aguda/induzido quimicamente , Lomustina/administração & dosagem , Síndromes Mielodisplásicas/induzido quimicamente , Segunda Neoplasia Primária/induzido quimicamente , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Temozolomida
7.
Proc Natl Acad Sci U S A ; 106(7): 2142-7, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19164530

RESUMO

O(6)-methylguanine (O(6)meG) and related modifications of guanine in double-stranded DNA are functionally severe lesions that can be produced by many alkylating agents, including N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), a potent carcinogen. O(6)meG is repaired through its demethylation by the O(6)-alkylguanine-DNA alkyltransferase (AGT). This protein is called Mgmt (or MGMT) in mammals and Mgt1 in the yeast Saccharomyces cerevisiae. AGT proteins remove methyl and other alkyl groups from an alkylated O(6) in guanine by transferring the adduct to an active-site cysteine residue. The resulting S-alkyl-Cys of AGT is not restored back to Cys, so repair proteins of this kind can act only once. We report here that S. cerevisiae Mgt1 is cotargeted for degradation, through a degron near its N terminus, by 2 ubiquitin-mediated proteolytic systems, the Ubr1/Rad6-dependent N-end rule pathway and the Ufd4/Ubc4-dependent ubiquitin fusion degradation (UFD) pathway. The cotargeting of Mgt1 by these pathways is synergistic, in that it increases not only the yield of polyubiquitylated Mgt1, but also the processivity of polyubiquitylation. The N-end rule and UFD pathways comediate both the constitutive and MNNG-accelerated degradation of Mgt1. Yeast cells lacking the Ubr1 and Ufd4 ubiquitin ligases were hyperresistant to MNNG but hypersensitive to the toxicity of overexpressed Mgt1. We consider ramifications of this discovery for the control of DNA repair and mechanisms of substrate targeting by the ubiquitin system.


Assuntos
O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Saccharomyces cerevisiae/genética , Animais , Domínio Catalítico , Códon , Reparo do DNA , Proteínas Fúngicas , Modelos Biológicos , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo , Temperatura , Ubiquitina/química
8.
Biochem Pharmacol ; 76(3): 303-11, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18573489

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Dano ao DNA , Reparo do DNA , Dacarbazina/análogos & derivados , Fibroblastos/efeitos dos fármacos , Nimustina/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Antineoplásicos/química , Apoptose/genética , Linhagem Celular , Dacarbazina/química , Dacarbazina/farmacologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteína 1 Homóloga a MutL , Nimustina/química , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Fosfatidilserinas/metabolismo , Relação Estrutura-Atividade , Temozolomida
9.
Mutagenesis ; 23(5): 341-6, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18477655

RESUMO

O(6)-methylguanine DNA methyltransferase (MGMT) deficiency is associated with an increased susceptibility to alkylating agent toxicity. To understand the contribution of MGMT in protecting against cyclophosphamide (CP)-induced toxicity, mutagenesis and tumorigenesis, we compared the biological effects of this agent in transgenic Mgmt knockout and wild-type mice. In addition, neurofibromin (Nf1)+/- background was used to increase the likelihood of CP-induced tumorigenesis. Cohorts of Mgmt-proficient or -deficient mice (either Nf1+/+ or Nf1+/-) were given 6 weekly injections of a maximally tolerated dose of CP (250 mg/kg) or vehicle and followed for 15 months. CP-treated mice had more deaths than control mice but there was no difference in the long-term survival between Mgmt+/+ and Mgmt-/- mice (12 of 83 Mgmt+/+ mice died compared to 12 of 80 Mgmt-/- mice, disregarding Nf1 status). Lymphomas and adrenal tumours were the most frequent malignancies. Interestingly, CP-treated, Mgmt-deficient mice developed fewer tumours than controls. Ten of 71 (14%) Mgmt-proficient mice developed tumours after CP treatment compared to only 2 of 68 (3%) Mgmt-deficient mice (P = 0.02). Mgmt-/-, Nf1+/- mice developed fewer tumours (1 of 35, 3%) following CP compared to Mgmt+/+, Nf1+/- mice (7 of 37, 19%) (P = 0.03). Hypoxanthine-guanine phosphoribosyltransferase mutation assays showed no significant increases in mutant frequencies in Mgmt-/- (18.1 x 10(6)) compared to Mgmt+/+ mice (12.9 x 10(6)). These data indicate that MGMT deficiency does not protect against long-term toxicity or mutagenicity from CP and appears to attenuate the occurrence of CP-induced tumours in an Nf1+/- background.


Assuntos
Alquilantes/toxicidade , Transformação Celular Neoplásica/induzido quimicamente , Ciclofosfamida/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Animais , Transformação Celular Neoplásica/genética , Hematopoese/efeitos dos fármacos , Hipoxantina Fosforribosiltransferase/genética , Camundongos , Camundongos Knockout , Mutagênese , Mutação , Neurofibromina 1/genética , O(6)-Metilguanina-DNA Metiltransferase/genética
10.
Br Med Bull ; 85: 17-33, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18245773

RESUMO

INTRODUCTION: Alkylating agents are frequently used in the chemotherapy of many types of cancer. This group of drugs mediates cell death by damaging DNA and therefore, understandably, cellular DNA repair mechanisms can influence both their antitumour efficacy and their dose-limiting toxicities. SOURCES OF DATA: This review focuses on the mechanism of action of the DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT) and its exploitation in cancer therapy and reviews the current literature. AREAS OF AGREEMENT: MGMT can provide resistance to alkylating agents by DNA damage reversal. Inhibition of tumour MGMT by pseudosubstrates to overcome tumour resistance is under clinical evaluation. In addition, MGMT overexpression in haematopoietic stem cells has been shown in animal models to protect normal cells against the myelosuppressive effects of chemotherapy: this strategy has also entered clinical trials. AREAS OF CONTROVERSY: MGMT inhibitors enhance the myelotoxic effect of O(6)-alkylating drugs and therefore reduce the maximum-tolerated dose of these agents. Retroviral vectors used for chemoprotective gene therapy are associated with insertional mutagenesis and leukaemia development. GROWING POINTS: The results of ongoing preclinical and clinical research involving various aspects of MGMT modulation should provide new prospects for the treatment of glioma, melanoma and other cancer types. AREAS TIMELY FOR DEVELOPING RESEARCH: Tissue- and tumour-specific approaches to the modulation of MGMT together with other DNA repair functions and in combination with immuno- or radiotherapy are promising strategies to improve alkylating agent therapy.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Dano ao DNA , Enzimas Reparadoras do DNA/metabolismo , Terapia Genética/métodos , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Enzimas Reparadoras do DNA/genética , Resistencia a Medicamentos Antineoplásicos , Humanos , O(6)-Metilguanina-DNA Metiltransferase/fisiologia
11.
Eur J Cancer ; 43(12): 1791-801, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17588740

RESUMO

An increased DNA-repair activity in tumour cells has been associated with resistance to treatment to DNA-directed drugs, while defects in DNA repair pathways result in hypersensitivity to these agents. In the past years the unravelling of the molecular basis of these DNA pathways, with a better understanding of the DNA damage caused by different anticancer agents, has provided the rationale for the use of some DNA repair inhibitors to optimise the therapeutic use of DNA-damaging agents currently used in the treatment of tumours. In addition, the possibility to specifically target the differences in DNA repair capacity between normal and tumour cells has recently emerged as an exciting possibility. The present review will mainly cover those approaches that are currently under clinical investigation.


Assuntos
Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Neoplasias/terapia , Inibidores Enzimáticos/uso terapêutico , Humanos , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Inibidores de Poli(ADP-Ribose) Polimerases
12.
Chin Med J (Engl) ; 120(8): 714-7, 2007 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-17517190

RESUMO

BACKGROUND: O(6)-methylguanine-DNA-methyltransferase (MGMT) is a specific DNA revising enzyme transferring alkylated groups from DNA to its cysteine residue to avoid the abnormal twisting of DNA. Therefore, it is one of the drug resistant genes targeted in the treatment of cancer. This study explored the protective effect of MGMT gene transferred into mammalian cells. METHODS: Mammalian expression vector containing the MGMT gene cloned from human hepatocytes by RT-PCR was constructed and transferred into K562 cells and human peripheral blood mononuclear cells (PBMCs) via liposome, then assayed for gene expression at RNA and protein levels. MTT assay was used to check the drug resistance of cells transfected with MGMT gene. RESULTS: MGMT gene was successfully cloned. Real-time PCR showed that the mRNA expression in gene transfected groups in K562 cell line and PBMC were 13.4 and 4.0 times that of the empty vector transfected groups respectively. RESULTS: of Western blotting showed distinct higher expression of MGMT in gene transfected group than in other two groups. The IC(50) values increased to 7 and 2 times that of the original values respectively in stable transfected K562 cells and transient transfected PBMC. CONCLUSION: The alkylating resistance of eukaryotic cells is enhanced after being transfected with MGMT gene which protein product performs the protective function, and may provide the reference for the protective model of peripheral blood cells in cancer chemotherapy.


Assuntos
O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Células K562 , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Microscopia de Fluorescência , Compostos de Mostarda Nitrogenada/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
13.
Int J Oncol ; 29(4): 785-97, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16964376

RESUMO

Clinically achievable concentrations of temozolomide (TMZ) produce cytotoxic effects only in mismatch repair (MMR)-proficient cells endowed with low O6-methylguanine-DNA methyltransferase (MGMT) activity. Aim of the present study was to investigate the molecular mechanisms underlying acquired resistance of melanoma cells to TMZ and the effect of O6-benzylguanine (BG), a specific MGMT inhibitor, on the development of a TMZ-resistant phenotype. Three MMR-proficient melanoma cell clones with low or no MGMT activity were treated daily for 5 days with 50 micromol/l TMZ, alone or in combination with 5 micromol/l BG. Parental clones and sublines established after one or four cycles of treatment were analyzed for sensitivity to TMZ or TMZ+BG and for other parameters. The sublines established after one cycle of TMZ or TMZ+BG exhibited a marked increase in MGMT activity and resistance to TMZ alone. BG only partially reversed acquired resistance to the drug. In some cases, alterations in the MMR system accounted for MGMT-independent resistance to TMZ. Up-regulation of MGMT activity was associated with either demethylation of the MGMT promoter or hypermethylation of the body of the gene, and partially reversed by 5-aza-2'-deoxycytidine. The sublines established after four cycles of TMZ or TMZ+BG did not show a further increase in resistance to TMZ alone. However, two out of three sublines established after TMZ+BG treatment exhibited increased resistance to TMZ+BG. In conclusion, our data demonstrate that a single cycle of TMZ is sufficient to induce high levels of drug resistance in melanoma clones, principally, but not exclusively, via up-regulation of MGMT expression. Exposure to TMZ+BG favors the development of MGMT-independent mechanisms of TMZ resistance.


Assuntos
Antineoplásicos/farmacologia , Pareamento Incorreto de Bases/fisiologia , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Guanina/análogos & derivados , Melanoma/enzimologia , Melanoma/genética , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Metilação de DNA , Reparo do DNA , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Guanina/farmacologia , Humanos , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , O(6)-Metilguanina-DNA Metiltransferase/genética , Regiões Promotoras Genéticas , Temozolomida , Células Tumorais Cultivadas , Regulação para Cima
14.
Mol Ther ; 13(2): 391-400, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16226060

RESUMO

Severe adverse events related to insertional mutagenesis have reinforced interest in self-inactivating (SIN) retroviral vectors lacking enhancer-promoter sequences in the long terminal repeats (LTRs). Here, we have compared the potency of gammaretroviral and lentiviral vectors expressing the P140K mutant of O(6)-methylguanine-DNA methyltransferase (MGMT). MGMT-P140K is a clinically relevant selection marker that mediates a strong survival advantage in hematopoietic cells exposed to alkylating agents. We designed gammaretroviral and lentiviral vectors that contained identical enhancer-promoter sequences located either in the LTR or downstream of the packaging region, for internal initiation of transcription from SIN backbones. Gammaretroviral vectors with intact LTRs containing enhancer-promoter sequences showed both higher titers and higher expression levels than the lentiviral counterparts, likely a result of suboptimal RNA processing of the lentiviral leader region. In the SIN context, gammaretroviral and lentiviral vectors with comparable internal cassettes had similar expression properties. Interestingly, gammaretroviral SIN vectors pseudotyped with RD114/TR had a higher transduction efficiency on proliferating human CD34(+) cells than lentiviral counterparts. These results encourage further investigations into the formation of retroviral hybrid vectors that combine the desired properties of high efficiency and increased biosafety.


Assuntos
Regulação Viral da Expressão Gênica/fisiologia , Vetores Genéticos , Células-Tronco Hematopoéticas/enzimologia , Lentivirus/genética , Vírus da Leucemia Murina/genética , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , O(6)-Metilguanina-DNA Metiltransferase/genética , Transdução Genética , Animais , Células Cultivadas , Células-Tronco Hematopoéticas/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Insercional , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Processamento Pós-Transcricional do RNA
15.
Chembiochem ; 7(1): 194-202, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16342318

RESUMO

Protein microarrays are an attractive approach for the high-throughput analysis of protein function, but their impact on proteomics has been limited by the technical difficulties associated with their generation. Here we demonstrate that fusion proteins of O6-alkylguanine-DNA alkyltransferase (AGT) can be used for the simple and reliable generation of protein microarrays for the analysis of protein function. Important features of the approach are the selectivity of the covalent immobilization; this allows for direct immobilization of proteins out of cell extracts, and the option both to label and to immobilize AGT fusion proteins, which allows for direct screening for protein-protein interactions between different AGT fusion proteins. In addition to the identification of protein-protein interactions, AGT-based protein microarrays can be used for the characterization of small molecule-protein interactions or post-translational modifications. The potential of the approach was demonstrated by investigating the post-translational modification of acyl carrier protein (ACP) from E. coli by different phosphopantetheine transferases (PPTases), yielding insights into the role of selected ACP amino acids in the ACP-PPTase interaction.


Assuntos
Proteínas de Transporte/fisiologia , Corantes Fluorescentes/química , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Análise Serial de Proteínas/métodos , Proteínas Recombinantes de Fusão/química , Proteínas de Transporte/química , Humanos , Imobilização , Modelos Moleculares , Estrutura Molecular , O(6)-Metilguanina-DNA Metiltransferase/química , Conformação Proteica , Estrutura Terciária de Proteína , Proteômica/métodos , Proteínas Recombinantes de Fusão/fisiologia , Relação Estrutura-Atividade , Transferases/química , Transferases/fisiologia
16.
J Pharmacol Exp Ther ; 316(2): 946-54, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16258022

RESUMO

Two camptothecin-resistant cell lines, CPT30 and KB100, were established and characterized previously in our laboratory. Because enhanced sensitivity to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and decreased expression of O(6)-methylguanine-DNA methyltransferase (MGMT) protein were observed in these lines, we hypothesized that MGMT may be a determinant of cytotoxicity associated with camptothecin-derived DNA topoisomerase I inhibitors (CPTs). We used the Tet-On system to induce expression of MGMT in Chinese hamster ovary (CHO) cells and RNA interference to knock down MGMT expression in human nasopharyngeal carcinoma HONE-1 cells in order to identify any correlations between MGMT expression and CPTs cytotoxicity. CHO-derived Tet-On-inducible cells (S12+) showed MGMT overexpression and statistically significant more resistance to BCNU, camptothecin, 7-ethyl-10-hydrocamptothecin (SN38), and topotecan than parental CHO cells (p < 0.05), but there was less resistance to CPTs than to BCNU. Knockdown of MGMT expression with small interfering RNA in HONE-1 cells conferred increased sensitivity to BCNU and CPTs compared with mock control. Furthermore, alteration of MGMT expression coincides with CPT-induced cell death and poly(ADP-ribose) polymerase cleavage. There were no differences in protein levels and catalytic activity of topoisomerase I between MGMT-proficient and MGMT-deficient cells from the Tet-On-inducible and small interfering RNA (siRNA) systems. Resistance to CPTs coincided with decreased amounts of protein-linked DNA breaks generated by CPTs in MGMT-proficient cells and vice versa in MGMT-deficient cells. Our data indicate that MGMT can modulate cytotoxicity of CPT-derived topoisomerase I inhibitors.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Enzimas Reparadoras do DNA/fisiologia , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Inibidores da Topoisomerase I , Animais , Células CHO , Camptotecina/análogos & derivados , Catálise , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Clonagem Molecular , Cricetinae , Cricetulus , Enzimas Reparadoras do DNA/biossíntese , Enzimas Reparadoras do DNA/genética , Resistencia a Medicamentos Antineoplásicos , Inativação Gênica , Humanos , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , O(6)-Metilguanina-DNA Metiltransferase/genética , RNA Interferente Pequeno/genética , Transfecção
17.
Mol Cancer Ther ; 4(11): 1755-63, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16275997

RESUMO

Cloretazine (VNP40101M; 101M; 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)-2-[(methylamino)carbonyl]hydrazine) is a sulfonylhydrazine prodrug that generates both chloroethylating and carbamoylating species on activation. To explore the molecular mechanisms underlying the broad anticancer activity observed in preclinical studies, cloretazine and chloroethylating-only [i.e., 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine] and carbamoylating-only (i.e., 1,2-bis(methylsulfonyl)-1-[(methylamino)carbonyl]hydrazine) analogues were evaluated in five murine hematopoietic cell lines. These cell lines were separable into two groups by virtue of their sensitivity to 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine; the sensitive group included L1210, P388, and F-MEL leukemias (IC50s, 6-8 micromol/L) and the resistant group consisted of Ba/F3 bone marrow and WEHI-3B leukemia cells (IC50s, 50-70 micromol/L). Resistant cells expressed O6-alkylguanine-DNA alkyltransferase (AGT), whereas sensitive cells did not. A correlation existed between AGT expression and the functional status of p53; AGT- cells possessed defective p53, whereas AGT+ cells contained wild-type p53. Based on recent findings on regulation of AGT gene expression by others, we suspect that silencing of the AGT gene by promoter hypermethylation frequently occurs during tumor progression involving p53 inactivation. O6-Chloroethylguanine is the initial DNA lesion that progresses to lethal interstrand DNA cross-links. Cloretazine exhibited a much higher preference toward the O6-chloroethylation of guanine, as measured by the difference in IC50s to wild-type and AGT-transfected L1210 cells, than 1,3-bis(2-chloroethyl)-1-nitrosourea, which targets the same site in DNA. Preferential toxicity of cloretazine against AGT- tumor cells coupled with decreased toxicity to AGT+ cells in host tissues constitute the therapeutic basis for cloretazine.


Assuntos
Hidrazinas/farmacologia , Neoplasias/tratamento farmacológico , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Sulfonamidas/farmacologia , Animais , Antineoplásicos/farmacologia , Northern Blotting , Southern Blotting , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Metilação de DNA , DNA Complementar/metabolismo , Progressão da Doença , Relação Dose-Resposta a Droga , Inativação Gênica , Glicerol/análogos & derivados , Glicerol/química , Guanina/química , Células-Tronco Hematopoéticas/efeitos dos fármacos , Concentração Inibidora 50 , Camundongos , Modelos Químicos , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Pró-Fármacos/química , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Transfecção , Proteína Supressora de Tumor p53/metabolismo
18.
Curr Genet ; 46(6): 317-30, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15614491

RESUMO

DNA-damaging agents constantly challenge cellular DNA; and efficient DNA repair is therefore essential to maintain genome stability and cell viability. Several DNA repair mechanisms have evolved and these have been shown to be highly conserved from bacteria to man. DNA repair studies were originally initiated in very simple organisms such as Escherichia coli and Saccharomyces cerevisiae, bacteria being the best understood organism to date. As a consequence, bacterial DNA repair genes encoding proteins with well characterized functions have been transferred into higher organisms in order to increase repair capacity, or to complement repair defects, in heterologous cells. While indicating the contribution of these repair functions to protection against the genotoxic effects of DNA-damaging agents, heterologous expression studies also highlighted the role of the DNA lesions that are substrates for such processes. In addition, bacterial DNA repair-like functions could be identified in higher organisms using this approach. We heterologously expressed three well characterized E. coli repair genes in S. cerevisiae cells of different genetic backgrounds: (1) the ada gene encoding O(6)-methylguanine DNA-methyltransferase, a protein involved in the repair of alkylation damage to DNA, (2) the recA gene encoding the main recombinase in E. coli and (3) the nth gene, the product of which (endonuclease III) is responsible for the repair of oxidative base damage. Here, we summarize our results and indicate the possible implications they have for a better understanding of particular DNA repair processes in S. cerevisiae.


Assuntos
Reparo do DNA , Proteínas de Escherichia coli/fisiologia , Escherichia coli/genética , Variação Genética , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/genética , Sobrevivência Celular , Escherichia coli/enzimologia , Regulação da Expressão Gênica , Mutação , Saccharomyces cerevisiae/enzimologia
19.
Oncogene ; 23(35): 5931-40, 2004 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15208683

RESUMO

O6-methylguanine methyltransferase, Mgmt, constitutes the first line of defense against O6-alkylguanine, which can result in G : C to A : T transitions upon DNA replication. Mgmt has been found in organisms as diverse as archaebacteria and mammals. This evolutionary conservation suggests that all organisms may be exposed to either endogenous or environmental alkylating agents. We thus hypothesized that tissues of Mgmt-/- mice would exhibit elevated mutant frequencies. Employing the Big Blue trade mark transgenic system, we evaluated lacI mutants rescued from liver and small intestinal DNA of young Mgmt-/- mice. Interestingly, while there was a small difference between Mgmt-/- mice and controls with respect to lacI mutant frequency, no differences attributable to Mgmt deficiency were apparent in the mutational spectra. Although mutations stemming from O6-guanine alkylations would be predicted to be cumulative, we found no evidence of an Mgmt-dependent alteration in mutation spectrum in DNA samples from 12 month-old mice. To optimize our ability to detect mutations resulting from O6-alkylguanine-induced G : T mismatches, mice with combined deficiencies of Mgmt and the DNA mismatch repair molecule, Msh6, were analysed. In spite of this strategy, we observed no significant differences between Mgmt-/- Msh6-/- and Msh6-/- mouse lacI mutations, except for a trend towards a greater percentage (of total transitions) of G : C to A : T changes in Mgmt-/-Msh6-/- livers. Therefore, despite the striking evolutionary conservation of Mgmt, deficiency of this gene did not significantly impact the spontaneous lacI mutational spectrum in vivo.


Assuntos
Pareamento Incorreto de Bases , Proteínas de Ligação a DNA/fisiologia , Guanina/análogos & derivados , Guanina/toxicidade , Mutação , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Fatores Etários , Animais , Proteínas de Ligação a DNA/genética , Feminino , Óperon Lac , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , O(6)-Metilguanina-DNA Metiltransferase/genética , Transgenes
20.
Chem Res Toxicol ; 17(6): 742-52, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15206895

RESUMO

O(6)-Alkylguanine-DNA alkyltransferase (AGT) is a DNA repair protein that removes O(6)-alkylguanine adducts. The interaction of dibromomethane (CH(2)Br(2)) and bromomethyl acetate (BrCH(2)OAc) with AGT was studied in vitro, and the effect of AGT on their toxicity and mutagenicity was investigated using Escherichia coli strain TRG8 (lacking endogenous AGT) that expressed human AGT or its inactive C145A mutant. Both CH(2)Br(2) and BrCH(2)OAc reacted with AGT at its cysteine acceptor site, abolishing its DNA repair activity with the latter agent being much more potent. The formation of AGT-Cys(145)S-CH(2)OAc by BrCH(2)OAc was confirmed by mass spectral analysis, but the presumed AGT-Cys(145)S-CH(2)Br adduct from CH(2)Br(2) was too unstable for such characterization. In the presence of CH(2)Br(2), AGT was covalently cross-linked to an oligodeoxyribonucleotide, 5'-d(AG)(8)-3', but no cross-link was formed by BrCH(2)OAc. Survival of cells exposed to CH(2)Br(2) was reduced, and the number of mutants was greatly increased when wild-type AGT was present. The cytotoxicity of CH(2)Br(2) was similar to that of BrCH(2)CH(2)Br(2), but the mutagenicity was about four times less. Virtually all of the AGT-mediated mutants induced by CH(2)Br(2) in the rpoB gene were at G:C sites with equal numbers of transitions to A:T and transversions to T:A. In contrast, BrCH(2)OAc was more than 10-fold less genotoxic than CH(2)Br(2) and the survival of cells exposed to BrCH(2)OAc was not affected by AGT. The number of mutations (almost all G:C to A:T transitions) induced by BrCH(2)OAc was slightly reduced by the presence of wild-type AGT and substantially increased by the inactive C145A mutant. These results with CH(2)Br(2) are consistent with a mechanism in which reaction at the active site Cys145 residue followed by attack of AGT-Cys(145)S-CH(2)Br at guanine in DNA forms a covalent adduct, which leads to cytotoxicity and to mutagenicity. The results with BrCH(2)OAc suggest that it reacts directly with DNA to form O(6)-(CH(2)OAc)guanine, which, if unrepaired, causes G:C to A:T transitions. Our experiments reveal two novel pathways (direct inactivation of AGT and formation of AGT-Cys(145)S-CH(2)-DNA adducts) by which CH(2)Br(2) may cause damage to the genome in addition to the well-recognized pathway involving activation by GSTs.


Assuntos
Dano ao DNA/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Hidrocarbonetos Bromados/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , RNA Polimerases Dirigidas por DNA/metabolismo , Eletroforese em Gel de Poliacrilamida , Escherichia coli/enzimologia , Espectrometria de Massas , Testes de Mutagenicidade , Plasmídeos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA