Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 177: 113988, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32330495

RESUMEN

Chloroethylnitrosoureas (CENUs) are bifunctional antitumor alkylating agents, which exert their antitumor activity through inducing the formation of dG-dC interstrand crosslinks (ICLs) within DNA double strand. However, the complex process of tumor biology enables tumor cells to escape the killing triggered by CENUs, as for instance with the detoxifying activity of O6-methylguanine DNA methyltransferase (MGMT) to accomplish DNA damage repair. Considering the fact that most tumor cells highly depend on aerobic glycolysis to provide energy for survival even in the presence of oxygen (Warburg effect), inhibition of aerobic glycolysis may be an attractive strategy to overcome the resistance and improve the chemotherapeutic effects of CENUs. Especially, 3-bromopyruvate (3-BrPA), a small molecule alkylating agent, has been emerged as an effective glycolytic inhibitor (energy blocker) in cancer treatment. In view of its tumor specificity and inhibition on cellular multiple targets, it is likely to reduce the chemoresistance when chemotherapeutic drugs are combined with 3-BrPA. In this study, we investigated the effects of 3-BrPA on the chemosensitivity of two human hepatocellular carcinoma (HCC) cell lines to the cytotoxic effects of l,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and the underlying molecular mechanism. The sensitivity of SMMC-7721 and HepG2 cells to BCNU was significantly increased by 2 h pretreatment with micromolar dosage of 3-BrPA. Moreover, 3-BrPA decreased the cellular ATP and GSH levels, and extracellular lactate excreted by tumor cells, and the effects were more effective when 3-BrPA was combined with BCNU. Cellular hexokinase-II (HK-II) activity was also reduced after exposure to the treatment of 3-BrPA plus BCNU. Based on the above results, the effects of 3-BrPA on the formation of dG-dC ICLs induced by BCNU was investigated by stable isotope dilution high-performance liquid chromatography electrospray ionization tandem mass spectrometry (HPLC-ESI-MS/MS). The results indicated that BCNU produced higher levels of dG-dC ICLs in SMMC-7721 and HepG2 cells pretreated with 3-BrPA compared to that without 3-BrPA pretreatment. Notably, in MGMT-deficient HepG2 cells, the levels of dG-dC ICLs were significantly higher than MGMT-proficient SMMC-7721 cells. In general, these findings revealed that 3-BrPA, as an effective glycolytic inhibitor, may be considered as a potential clinical chemosensitizer to optimize the therapeutic index of CENUs.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Carmustina/farmacología , Reactivos de Enlaces Cruzados/farmacología , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica , Glucólisis/efectos de los fármacos , Piruvatos/farmacología , Adenosina Trifosfato/biosíntesis , Línea Celular Tumoral , ADN/química , ADN/genética , ADN/metabolismo , Daño del ADN , Reparación del ADN/efectos de los fármacos , ADN de Neoplasias/química , ADN de Neoplasias/metabolismo , Combinación de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Glutatión/metabolismo , Glucólisis/genética , Células Hep G2 , Hexoquinasa/antagonistas & inhibidores , Hexoquinasa/genética , Hexoquinasa/metabolismo , Humanos , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/genética
2.
Mol Cancer Ther ; 11(11): 2440-50, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22986464

RESUMEN

The dismal prognosis of glioblastoma multiforme (GBM) is mostly due to the high propensity of GBM tumor cells to invade. We reported an inverse relationship between GBM angiogenicity and expression of the DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT), which has been extensively characterized for its role in resistance to alkylating agents used in GBM treatment. In the present study, given the major role of angiogenesis and invasion in GBM aggressiveness, we aimed to investigate the relationship between MGMT expression and GBM invasion. Stable overexpression of MGMT in the U87MG cell line significantly decreased invasion, altered expression of invasion-related genes, decreased expression of α(5)ß(1) integrin and focal adhesion kinase, and reduced their spindle-shaped morphology and migration compared with the empty vector control. Conversely, short hairpin RNA-mediated stable knockdown of MGMT or its pharmacologic depletion in the MGMT-positive T98G cell line were required for increased invasion. The inverse relationship between MGMT and invasion was further validated in primary GBM patient-derived cell lines. Using paraffin-embedded tumors from patients with newly diagnosed GBM (n = 59), tumor MGMT promoter hypermethylation (MGMT gene silencing) was significantly associated with increased immunohistochemical expression of the proinvasive matricellular protein secreted protein acidic and rich in cysteine (SPARC; P = 0.039, χ(2) test). Taken together, our findings highlight for the first time the role of MGMT as a negative effector of GBM invasion. Future studies are warranted to elucidate the role of SPARC in the molecular mechanisms underlying the inverse relationship between MGMT and GBM invasion and the potential use of MGMT and SPARC as biomarkers of GBM invasion.


Asunto(s)
Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Glioblastoma/enzimología , Glioblastoma/patología , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Movimiento Celular , Forma de la Célula , Activación Enzimática , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioblastoma/genética , Humanos , Inmunohistoquímica , Invasividad Neoplásica , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Osteonectina/metabolismo , Fenotipo , ARN Interferente Pequeño/metabolismo
3.
DNA Cell Biol ; 31(5): 856-66, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22196440

RESUMEN

The abnormal function of O(6)-methylguanine-DNA methyltransferase (MGMT) is reported to be associated with the occurrence of various tumors and malignant tumor progression. However, little evidence is available to describe its role in esophageal carcinogenesis. To address this issue, we constructed a stable MGMT-silenced esophageal cancer cell line by RNA interference, and exposed the cells to N-methyl-N-nitro-N-nitrosoguanidine (MNNG) to investigate the role that MGMT plays in toxicity. During this time, we also observed the malignant behavior of cells in vitro and in vivo. In addition, two-dimensional electrophoresis and mass spectrometry were used to detect and confirm the proteins that were differentially expressed in the MGMT-deficient and MGMT-proficient cells, which might be responsible for the malignant alteration of cells. Results showed that the IC(50) of MGMT-deficient and MGMT-proficient cells exposed to MNNG was 30 µM and 65 µM, respectively, and MGMT-deficient cells had more aggressive motility and invasive abilities compared with MGMT-proficient cells. Nineteen differentially expressed proteins were detected between the MGMT-deficient and MGMT-proficient cells, 14 of which were identified, including the membrane-cytoskeleton linker protein, Ezrin, which was confirmed by both mass spectrometry and western blot analysis. The correlation between MGMT, Ezrin expression, and the malignant behavior of one normal epithelial esophageal cell line and seven esophageal cancer lines is discussed. In conclusion, loss of MGMT expression leads EC109 esophageal cancer cells to have increased malignant behavior, which may correlate with its high Ezrin protein expression.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Animales , Western Blotting , Células CHO , Adhesión Celular , Movimiento Celular , Proliferación Celular , Cricetinae , Electroforesis en Gel Bidimensional , Esófago/metabolismo , Esófago/patología , Técnica del Anticuerpo Fluorescente , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación/genética , O(6)-Metilguanina-ADN Metiltransferasa/antagonistas & inhibidores , O(6)-Metilguanina-ADN Metiltransferasa/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transfección
4.
Biochem Pharmacol ; 79(11): 1553-61, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20005211

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Hidrazinas/farmacología , Hipoxia , O(6)-Metilguanina-ADN Metiltransferasa/fisiología , Profármacos/farmacología , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Humanos , Hidrazinas/farmacocinética , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Oxidación-Reducción , Oxidorreductasas/metabolismo , Profármacos/metabolismo , Profármacos/farmacocinética
5.
Eur J Endocrinol ; 161(4): 553-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19589911

RESUMEN

CONTEXT: Crooke's cell adenoma (CCA), characterized by massive Crooke's hyaline change in corticotroph adenoma, causes a rare subtype of Cushing's disease. In contrast to ordinary corticotroph adenomas, CCAs are generally aggressive and present as invasive macroadenomas, which are refractory to both surgery and radiotherapy and have a high-recurrence rate. Moreover, some patients with CCA present with distant or craniospinal metastases. Currently, there are no effective standard therapies for CCA. OBJECTIVE: We report a patient with Crooke's cell carcinoma who presented with local invasion and liver metastases, which was refractory to conventional therapeutic modalities including transsphenoidal surgery, radiosurgery, medications, and hepatic transcatheter arterial embolization. After all these treatments failed, the patient had monthly temozolomide administrations, resulting in gradual clinical improvement and biochemical data that were consistent with tumor shrinkage. In glioblastoma, low O(6)-methylguanine DNA methyltransferase (MGMT) expression is associated with epigenetic gene silencing and predicts a better response to temozolomide. METHODS: We thus investigated MGMT expression, immunohistochemically, in seven CCAs (five invasive macroadenomas and two invasive microadenomas) and 17 ordinary-type adenomas (OTAs; three noninvasive macroadenomas, 12 noninvasive microadenomas, and two invasive microadenomas) from patients with Cushing's disease. RESULTS: In seven CCAs, all five invasive macroadenomas exhibited low MGMT expression, defined as <5% nuclear MGMT staining. In 17 OTAs, only one adenoma showed low MGMT expression. CONCLUSION: In Cushing's disease, invasive macroadenomas including CCA usually have low-MGMT expression. Temozolomide thus may be a new therapeutic option for invasive macroadenomas such as CCA particularly when conventional treatments are ineffective.


Asunto(s)
O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/genética , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/enzimología , Neoplasias Hipofisarias/enzimología , Adolescente , Hormona Adrenocorticotrópica/sangre , Adulto , Anciano , Antineoplásicos Alquilantes/uso terapéutico , Dacarbazina/análogos & derivados , Dacarbazina/uso terapéutico , Femenino , Humanos , Hidrocortisona/sangre , Inmunohistoquímica , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , O(6)-Metilguanina-ADN Metiltransferasa/biosíntesis , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/patología , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/cirugía , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/cirugía , Temozolomida , Tomografía Computarizada por Rayos X , Adulto Joven
6.
Clin Cancer Res ; 15(1): 338-45, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19118063

RESUMEN

PURPOSE: Recent studies suggest that temozolomide has activity in neuroendocrine tumors. Low levels of the DNA repair enzyme, O(6)-methylguanine DNA methyltransferase (MGMT), are associated with sensitivity to temozolomide in other tumor types. We evaluated the prevalence of MGMT deficiency in neuroendocrine tumors and correlated MGMT deficiency with treatment response to temozolomide-based regimens. EXPERIMENTAL DESIGN: The prevalence of MGMT deficiency, measured by immunohistochemistry, was assessed in 97 archival neuroendocrine tumor specimens. Rates of treatment response and survival were next evaluated in a cohort of 101 consecutive neuroendocrine tumor patients who had received treatment with a temozolomide-based regimen at one of three institutions. MGMT expression was directly correlated with treatment response in 21 patients who had available tumor tissue and response data. RESULTS: In archival specimens, MGMT deficiency was observed in 19 of 37 (51%) pancreatic neuroendocrine tumors and 0 of 60 (0%) carcinoid tumors (P < 0.0001). In the clinical cohort, 18 of 53 (34%) patients with pancreatic neuroendocrine tumors but only 1 of 44 (2%) patients with carcinoid tumors (P < 0.001) experienced a partial or complete response to temozolomide-based therapy. Among 21 patients with evaluable tumor tissue who had also received treatment with temozolomide, 4 of 5 patients with MGMT-deficient tumors (all pancreatic neuroendocrine tumors) and 0 of 16 patients with tumors showing intact MGMT expression responded to treatment (P = 0.001). CONCLUSIONS: MGMT deficiency, measured by immunohistochemistry, is more common in pancreatic neuroendocrine tumors than in carcinoid tumors as is treatment response to temozolomide-based therapy. Absence of MGMT may explain the sensitivity of some pancreatic neuroendocrine tumors to treatment.


Asunto(s)
Dacarbazina/análogos & derivados , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/enzimología , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Antineoplásicos Alquilantes/uso terapéutico , Tumor Carcinoide/tratamiento farmacológico , Tumor Carcinoide/enzimología , Dacarbazina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/enzimología , Temozolomida , Resultado del Tratamiento
7.
DNA Repair (Amst) ; 6(8): 1127-33, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17500046

RESUMEN

The DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT) is a cardinal defense against the mutagenic and carcinogenic effects of alkylating agents. We have reported evidence that absence of detectable MGMT activity (MGMT(-) phenotype) in human brain is a predisposing factor for primary brain tumors that affects ca. 12% of individuals [J.R. Silber, A. Blank, M.S. Bobola, B.A. Mueller, D.D. Kolstoe, G.A. Ojemann, M.S. Berger, Lack of the DNA repair protein O(6)-methylguanine-DNA methyltransferase in histologically normal brain adjacent to primary brain tumors, Proc. Natl. Acad. Sci. U.S.A. 93 (1996) 6941-6946]. We report here that MGMT(-) phenotype in the brain of children and adults, and the apparent increase in risk of neurocarcinogenesis, may arise during gestation. We found that MGMT activity in 71 brain specimens at 6-19 weeks post-conception was positively correlated with gestational age (P

Asunto(s)
Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/etiología , Encéfalo/embriología , Encéfalo/enzimología , Metilasas de Modificación del ADN/deficiencia , Enzimas Reparadoras del ADN/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Proteínas Supresoras de Tumor/deficiencia , Adulto , Encéfalo/crecimiento & desarrollo , Neoplasias Encefálicas/genética , Niño , Reparación del ADN , Feto/enzimología , Edad Gestacional , Humanos , Mutación , Fenotipo
8.
DNA Repair (Amst) ; 6(8): 1145-54, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17485251

RESUMEN

O(6)-Methylguanine-DNA methyltransferase (MGMT) is a DNA repair protein that protects cells from the biological consequences of alkylating agents by removing alkyl groups from the O(6)-position of guanine. Cyclophosphamide and ifosfamide are oxazaphosphorines used clinically to treat a wide variety of cancers; however, the role of MGMT in recognizing DNA damage induced by these agents is unclear. In vitro evidence suggests that MGMT may protect against the urotoxic oxazaphosphorine metabolite, acrolein. Here, we demonstrate that Chinese hamster ovary cells transfected with MGMT are protected against cytotoxicity following treatment with chloroacetaldehyde (CAA), a neuro- and nephrotoxic metabolite of cyclophosphamide and ifosfamide. The mechanism by which MGMT recognizes damage induced by acrolein and CAA is unknown. CHO cells expressing a mutant form of MGMT (MGMT(R128A)), known to have >1000-fold less repair activity towards alkylated DNA while maintaining full active site transferase activity towards low molecular weight substrates, exhibited equivalent CAA- and acrolein-induced cytotoxicity to that of CHO cells transfected with plasmid control. These results imply that direct reaction of acrolein or CAA with the active site cysteine residue of MGMT, i.e. scavenging, is unlikely a mechanism to explain MGMT protection from CAA and acrolein-induced toxicity. In vivo, no difference was detected between Mgmt-/- and Mgmt+/+ mice in the lethal effects of cyclophosphamide. While MGMT may be important at the cellular level, mice deficient in MGMT are not significantly more susceptible to cyclophosphamide, acrolein or CAA. Thus, our data does not support targeting MGMT to improve oxazaphosphorine therapy.


Asunto(s)
Ciclofosfamida/toxicidad , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Acetaldehído/análogos & derivados , Acetaldehído/toxicidad , Acroleína/toxicidad , Alquilantes/metabolismo , Alquilantes/toxicidad , Animales , Células CHO , Cricetinae , Cricetulus , Ciclofosfamida/metabolismo , Reparación del ADN , Resistencia a Antineoplásicos , Ifosfamida/metabolismo , Ifosfamida/toxicidad , Ratones , Ratones Noqueados , Mutación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/enzimología , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Trasplante Heterólogo
9.
Mol Carcinog ; 44(3): 193-201, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16086375

RESUMEN

The prepubertal breast is more susceptible than the mature breast to the carcinogenic effects of ionizing radiation, and potentially to cigarette smoke and alkylating chemotherapeutics. Mammary epithelial cells (MECs) from sexually immature (3-week (wk)-old) Fischer 344 rats were more sensitive than mature (8-wk-old) rats to the carcinogenic, lethal, and mutagenic effects of N-nitroso-N-methylurea (NMU). The work reported here was undertaken to better define this age-specific susceptibility of the mammary gland to NMU. Using the alkaline comet assay, it was found that MECs from immature but not mature rats displayed an increase in single-strand DNA breaks or alkali-labile lesions 2 h following NMU treatment. Hoechst staining indicated apoptosis was not responsible for the increase. Inhibition of methylguanine methyltransferase (MGMT) did not affect immature MECs but caused mature MECs to recapitulate the immature response to NMU. Direct measurement of MGMT activity revealed that immature MECs are significantly deficient in MGMT activity relative to mature MECs. MECs had the lowest MGMT activity of all organs tested. Immature kidneys, which preferentially developed nephroblastomas after NMU treatment, also displayed significantly lower MGMT activity than mature kidneys. These results suggest that age-related differences in MGMT activity may play a significant role in age-differential susceptibility to rat mammary gland and kidney carcinogenesis, and argue the importance of extending these studies to humans. They also provide a mechanistic basis for studying, as potentially initiating events in breast cancer, exposures of prepubertal girls to alkylating agents, to which humans are exposed in cigarette smoke, the diet, and as chemotherapy.


Asunto(s)
Diferenciación Celular , Susceptibilidad a Enfermedades , Células Epiteliales/patología , Neoplasias Mamarias Animales/inducido químicamente , Metilnitrosourea/farmacología , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinógenos/farmacología , Células Cultivadas , Ensayo Cometa , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Femenino , Neoplasias Mamarias Animales/enzimología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , O(6)-Metilguanina-ADN Metiltransferasa/genética , Ratas , Ratas Endogámicas F344
10.
Cancer Res ; 65(8): 3319-27, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15833865

RESUMEN

DNA repair capacity of eukaryotic cells has been studied extensively in recent years. Mammalian cells have been engineered to overexpress recombinant nuclear DNA repair proteins from ectopic genes to assess the impact of increased DNA repair capacity on genome stability. This approach has been used in this study to specifically target O(6)-methylguanine DNA methyltransferase (MGMT) to the mitochondria and examine its impact on cell survival after exposure to DNA alkylating agents. Survival of human hematopoietic cell lines and primary hematopoietic CD34(+) committed progenitor cells was monitored because the baseline repair capacity for alkylation-induced DNA damage is typically low due to insufficient expression of MGMT. Increased DNA repair capacity was observed when K562 cells were transfected with nuclear-targeted MGMT (nucl-MGMT) or mitochondrial-targeted MGMT (mito-MGMT). Furthermore, overexpression of mito-MGMT provided greater resistance to cell killing by 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) than overexpression of nucl-MGMT. Simultaneous overexpression of mito-MGMT and nucl-MGMT did not enhance the resistance provided by mito-MGMT alone. Overexpression of either mito-MGMT or nucl-MGMT also conferred a similar level of resistance to methyl methanesulfonate (MMS) and temozolomide (TMZ) but simultaneous overexpression in both cellular compartments was neither additive nor synergistic. When human CD34(+) cells were infected with oncoretroviral vectors that targeted O(6)-benzylguanine (6BG)-resistant MGMT (MGMT(P140K)) to the nucleus or the mitochondria, committed progenitors derived from infected cells were resistant to 6BG/BCNU or 6BG/TMZ. These studies indicate that mitochondrial or nuclear targeting of MGMT protects hematopoietic cells against cell killing by BCNU, TMZ, and MMS, which is consistent with the possibility that mitochondrial DNA damage and nuclear DNA damage contribute equally to alkylating agent-induced cell killing during chemotherapy.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Dacarbazina/análogos & derivados , Guanina/análogos & derivados , Mitocondrias/enzimología , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Antígenos CD34/biosíntesis , Carmustina/farmacología , Muerte Celular/efectos de los fármacos , Núcleo Celular/enzimología , Daño del ADN , Reparación del ADN , Dacarbazina/farmacología , Resistencia a Antineoplásicos , Guanina/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/enzimología , Humanos , Células K562 , Metilmetanosulfonato/farmacología , O(6)-Metilguanina-ADN Metiltransferasa/biosíntesis , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/genética , Temozolomida , Transfección
11.
Mol Cancer Ther ; 3(2): 123-7, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14985452

RESUMEN

Temozolomide is an alkylating agent that mediates its cytotoxic effects via O(6)-methylguanine (O(6)-meG) adducts in DNA and their recognition and processing by the postreplication mismatch repair system (MMR). O(6)-meG adducts can be repaired by the DNA repair protein O(6)-alkylguanine-DNA-alkyltransferase (MGMT), which therefore constitutes a major resistance mechanism to the drug. Resistance to Temozolomide can also be mediated by loss of MMR, which is frequently mediated by methylation of the hMLH1 gene promoter. Methylation of hMLH1 can be reversed by treatment of cells with 5-aza-2'-deoxycytidine, while the MGMT pseudosubstrate O(6)-(4-bromothenyl)guanine (PaTrin-2) can deplete MGMT activity. Using a drug-resistant cell line which expresses MGMT and has methylated hMLH1, we show that while either of these treatments can individually sensitize cells to Temozolomide, the combined treatment leads to substantially greater sensitization. The increased sensitization is not observed in matched MMR proficient cells.


Asunto(s)
Reparación del ADN , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Resistencia a Medicamentos , Guanina/análogos & derivados , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Disparidad de Par Base/efectos de los fármacos , Proteínas Portadoras , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Guanina/farmacología , Humanos , Homólogo 1 de la Proteína MutL , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Temozolomida
12.
Cancer Res ; 63(18): 5793-8, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-14522901

RESUMEN

The preponderance of G:C to A:T transitions in inherited and somatic human mutations has led to the hypothesis that some of these mutations arise as a result of formation of O(6)-methylguanine in DNA. To test this hypothesis, the fine structure map of N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)-induced mutations was determined in human lymphoblastoid cells in the human hypoxanthine-guanine-phosphoribosyltransferase (HPRT) gene and compared with HPRT mutations observed in somatic T lymphocytes from normal individuals. Human TK6 cells, which are methylguanine methyltransferase deficient (MGMT(-)), were treated with the methylating agent MNNG to create a level of O(6)-methylguanine in cellular DNA equal to that found in normal human tissues. A total of 676 bp of the HPRT gene was scanned using constant denaturing capillary electrophoresis and high-fidelity PCR. MNNG induced 14 predominant hot spots, all which were G:C to A:T transitions. Thirteen of these 14 MNNG-induced hot spots were found among the in vivo set, and 10 of the MNNG-induced hot spots were among 75 putative in vivo hot spots (mutations observed two or more times in vivo). Using a hypergeometric test for concordance, the MNNG-induced hot spots were found to be a significant subset of the putative in vivo hot spots (P < 4 x 10(-7)). The set of shared hot spots comprise some 18% of the HPRT in vivo hot spot spectrum and strongly suggest that MNNG-induced hot spots in vitro share a common mutational pathway with a significant subset of somatic mutations in vivo.


Asunto(s)
Guanina/análogos & derivados , Hipoxantina Fosforribosiltransferasa/genética , Metilnitronitrosoguanidina/farmacología , Mutación , Linfocitos T/enzimología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Análisis Mutacional de ADN , Exones , Guanina/metabolismo , Humanos , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Linfocitos T/efectos de los fármacos , Linfocitos T/fisiología
13.
Clin Cancer Res ; 8(9): 3008-18, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12231548

RESUMEN

Alkylating agents are standard components of adjuvant chemotherapy for gliomas. We provide evidence here that Ape1/Ref-1, the major mammalian apurinic/apyrimidinic endonuclease (Ap endo), contributes to alkylating agent resistance in human glioma cells by incising DNA at abasic sites. We show that antisense oligonucleotides directed against Ape1/Ref-1 in SNB19, a human glioma cell line lacking O(6)-methylguanine-DNA-methyltransferase, mediate both reduction in Ape1/Ref-1 protein and Ap endo activity and concurrent reduction in resistance to methyl methanesulfonate and the clinical alkylators temozolomide and 1,3-(2-chloroethyl)-1-nitrosourea. An accompanying increase in the level of abasic sites indicates that the DNA repair activity of Ape1/Ref-1 contributes to resistance. Conversely, we also show that exposure of SNB19 cells to HOCl, a generator of reactive oxygen species (ROS), results in elevated Ape1/Ref-1 protein and Ap endo activity, enhanced alkylator resistance, and reduced levels of abasic sites. Given current evidence that heightened oxidative stress prevails within brain tumors, the finding that ROS increase resistance to clinical alkylators in glioma cells may have significance for the response of gliomas to alkylating agent-based chemotherapy. Our results may also be relevant to the design of therapeutic regimens using concurrent ionizing radiation (a generator of ROS) and alkylating agent-based chemotherapy.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/enzimología , Liasas de Carbono-Oxígeno/fisiología , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos/fisiología , Glioblastoma/enzimología , Proteínas de Neoplasias/fisiología , Ácido Apurínico/análisis , Neoplasias Encefálicas/patología , Liasas de Carbono-Oxígeno/biosíntesis , Carmustina/farmacología , Aductos de ADN , Daño del ADN , ADN de Neoplasias/análisis , ADN de Neoplasias/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa , Dacarbazina/farmacología , Inducción Enzimática , Glioblastoma/patología , Humanos , Ácido Hipocloroso/farmacología , Metilmetanosulfonato/farmacología , Proteínas de Neoplasias/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Estrés Oxidativo , Especies Reactivas de Oxígeno/farmacología , Temozolomida , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/enzimología
14.
Anticancer Res ; 22(2A): 697-701, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12014639

RESUMEN

BCNU was reported to have about a 6- to 8- fold lower cytotoxic potency than ACNU in cell lines naturally deficient in O6-AGT. In seven tumor cell lines with an O6-AGT activity ranging from 40 to 360 fmol/mg the cytotoxic potency of BCNU, ACNU and HeCNU, without and after O6-AGT depletion by O6-BG, was determined. Without O6-AGT depletion, BCNU was superior to both other drugs in tumor cells with high O6-AGT activity. After O6-AGT depletion, the cytotoxic potency (comparison of IC50 values) of ACNU was higher than that of BCNU (p=0.016) or that of HeCNU (p=0.016) in all tumor cell lines. We conclude that (without O6-AGT depletion) BCNU is the drug of choice especially in tumor cells with high transferase activity. The higher cytotoxic potency of ACNU after O6-AGT depletion as compared to BCNU after O6-AGT depletion is countered by the higher toxicity of ACNU in patients necessitating a clinical dose reduction as compared to BCNU. Thus, we would not expect superiority of ACNU + O6-BG over BCNU+ O6-BG after systemic administration.


Asunto(s)
Antineoplásicos Alquilantes/toxicidad , Guanina/análogos & derivados , Compuestos de Nitrosourea/toxicidad , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Carmustina/toxicidad , Sinergismo Farmacológico , Guanina/metabolismo , Guanina/farmacología , Humanos , Nimustina/toxicidad , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Células Tumorales Cultivadas
15.
Cancer Res ; 61(5): 1957-63, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11280752

RESUMEN

Temozolomide (TMZ) is a DNA-methylating agent that has recently been introduced into Phase II and III trials for the treatment of gliomas. TMZ produces O6-methylguanine in DNA, which mispairs with thymine during the next cycle of DNA replication. Subsequent futile cycles of DNA mismatch repair can lead to a p53-associated apoptotic cell death, although this mechanism has been described mostly in hematopoietic neoplasms. We studied the action of TMZ in gliomas and the role p53 might play by using U87 glioma cells that were either p53-wild-type or p53-deficient (by virtue of expression of the viral oncoprotein E6). LN-Z308 cells, in which p53 gene is deleted, were also used. p53-proficient U87 MG cells underwent a prolonged, p53- and p21(Waf1/Cip1)-associated G2-M arrest beginning 2 days after TMZ treatment. Although very few of these cells underwent apoptosis, most underwent senescence over a 10-day period. p53-deficient (E6-transfected U87 and LN-Z308) cells similarly underwent G2-M arrest in response to TMZ, but this arrest was accompanied by only minor changes in p53 or p21(Waf1/Cip1) and was reversed within 7 days of TMZ treatment in association with the appearance of cells with either 8n or subG1 DNA content. These results suggest that glioma cells respond to TMZ by undergoing G2-M arrest. p53 is not necessary for this G2-M arrest to occur but is important in the duration of G2-M arrest and in the ultimate fate of TMZ-treated cells. Therefore, the integrity of the G2-M cell cycle checkpoint may be important in the cytotoxicity of TMZ in glioma cells.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Fase G2/efectos de los fármacos , Glioblastoma/patología , Mitosis/efectos de los fármacos , Proteína p53 Supresora de Tumor/fisiología , Disparidad de Par Base , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Fase G2/fisiología , Glioblastoma/tratamiento farmacológico , Humanos , Mitosis/fisiología , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Temozolomida , Células Tumorales Cultivadas/efectos de los fármacos , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo
16.
Cancer Res ; 61(1): 53-8, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11196197

RESUMEN

Alkylating agents such as N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) are known to covalently link alkyl groups at the position 6 of guanines (O6MG) in DNA. O6-alkylguanine-DNA alkyltransferase (AGT) specifically removes the methyl group of the O6MG. Using purified human topoisomerase I (Top1), we found an 8-10-fold enhancement of Top1 cleavage complexes when O6MG is incorporated in oligonucleotides at the +1 position relative to a unique Top1 cleavage site. Top1 poisoning by O6MG is attributable to a decrease of the Top1-mediated DNA religation as well as an increase in the enzyme cleavage step. Increased cleavage is probably linked to a change in the hydrogen bonding pattern, such as in the case of the 8-oxoguanine, whereas inhibition of religation could be attributed to altered base pairing, such as abasic sites or base mismatches, because incorporation of a 6-thioguanine did not affect Top1 activity. Top1-DNA covalent complexes are also induced in MNNG-treated CHO cells constitutively lacking the AGT enzyme. Conversely, no increase could be detected in CHO cells transfected with the wild-type human AGT. Moreover, we show that yeasts overexpressing the human Top1 are more sensitive to MNNG, whereas knock-out Top1 strain cells display some resistance to the drug. Altogether, these results suggest a role for Top1 poisoning by alkylated bases in the antiproliferative activity of alkylating agents as well as in the DNA lesions resulting from endogenous and carcinogenic DNA modifications.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , Guanina/análogos & derivados , Guanina/metabolismo , Metilnitronitrosoguanidina/toxicidad , Alquilantes/toxicidad , Animales , Células CHO/efectos de los fármacos , Células CHO/enzimología , Células CHO/metabolismo , Cricetinae , ADN/metabolismo , Humanos , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/genética , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/enzimología , Transfección
17.
Carcinogenesis ; 21(10): 1879-83, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11023546

RESUMEN

O(6)-methylguanine-DNA methyltransferase plays vital roles in preventing induction of mutations and cancer as well as cell death related to alkylating agents. Mice defective in the MGMT: gene, encoding the methyltransferase, were used to evaluate cell death-inducing and tumorigenic activities of therapeutic agents which have alkylation potential. MGMT(-/-) mice were considerably more sensitive to dacarbazine, a monofunctional triazene, than were wild-type mice, in terms of survival. When dacarbazine was administered i.p. to 6-week-old mice and survival at 30 days was enumerated, LD(50) values of MGMT(-/-) and MGMT(+/+) mice were 20 and 450 mg/kg body wt, respectively. Increased sensitivity of MGMT(-/-) mice to 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosou rea (ACNU), a bifunctional nitrosourea, was also noted. On the other hand, there was no difference in survival of MGMT(+/+) and MGMT(-/-) mice exposed to cyclophosphamide, a bifunctional nitrogen mustard. It appears that dacarbazine and ACNU produce O(6)-alkylguanine as a major toxic lesion, while cyclophosphamide yields other types of modifications in DNA which are not subjected to the action of the methyltransferase. MGMT(-/-) mice seem to be less refractory to the tumor-inducing effect of dacarbazine than are MGMT(+/+) mice. Thus, the level of O(6)-methylguanine-DNA methyltransferase activity is an important factor when determining susceptibility to drugs with the potential for alkylation.


Asunto(s)
Antineoplásicos Alquilantes/toxicidad , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Adenoma/inducido químicamente , Animales , Muerte Celular/efectos de los fármacos , Ciclofosfamida/toxicidad , Dacarbazina/toxicidad , Femenino , Dosificación Letal Mediana , Neoplasias Pulmonares/inducido químicamente , Linfoma/inducido químicamente , Masculino , Ratones , Ratones Noqueados , Nimustina/toxicidad , O(6)-Metilguanina-ADN Metiltransferasa/genética , Testículo/efectos de los fármacos , Neoplasias del Timo/inducido químicamente
18.
Cancer Res ; 60(15): 4105-11, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10945617

RESUMEN

To determine whether N-methyl-N-nitrosourea (MNU) can induce malignant transformation of human fibroblasts and whether O6-methylguanine (O6-MeG) is involved, two populations of infinite life span cell strain MISU-1.1, differing only in level of O6-alkylguanine-DNA alkyltransferase, were treated with MNU and assayed for focus formation. MNU caused a dose-dependent increase in the frequency of foci in both groups, but the dose required was significantly lower in the cells lacking O6-alkylguanine-DNA alkyltransferase, indicating that O6-MeG was causally involved. Of 35 independent focus-derived strains assayed for p53 transactivating abilily, one was heterozygous, and 15 had lost all activity, 1 of 7 from untreated cells and 14 of 27 from MNU-treated cells. These results indicate that loss of p53 is not required for focus formation but may permit cells to form foci. Of 35 strains assayed for tumorigenicity, 10 formed malignant tumors with a short latency, all 10 lacked wild-type p53. The p53 heterozygous strain also formed tumors after a long latency, and the cells from those tumors lacked p53 transactivating ability. None of the 19 strains with wild-type p53 formed tumors. These results indicate that although loss of p53 is not sufficient for malignant transformation of MSU-1.1 cells, it may be necessary. Analysis of the p53 cDNA from several focus-derived strains lacking p53 activity revealed that each contained the same mutation, an A to G transition at codon 215, resulting in a change from serine to glycine. Because p53 can be inactivated by mutations at any one of a large number of sites, finding the same mutation in each strain assayed strongly suggests that the target population included a subpopulation of cells with this codon 215 mutation in one allele. Further analysis showed that all 15 focus-derived cells strains that lacked p53 transactivating activity contained two alleles, each with the same codon 215 mutation, and that the mutant allele in the heterozygous strain also had that mutatation. Analysis of the p arm of chromosome 17 of the focus-derived cell strains containing the codon 215 mutation revealed seven patterns of loss of heterozygosity, evidence of mitotic homologous recombination. Similar analysis of a separate series of cell strains, derived from foci induced by cobalt-60, revealed four patterns of loss of heterozygosity, only two of which had been found with those induced by MNU. These data suggest that homologous mitotic recombination, induced by O6-MeG in a subpopulation of cells heterozygous for p53 mutation, rendered the cells homozygous for loss of p53 activity, that this allowed the cells to form foci, and that although loss of p53 is not sufficient for malignant transformation, it predisposes cells to acquire the additional changes needed for such transformation.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Genes p53/genética , Metilnitrosourea/toxicidad , Recombinación Genética/genética , Alelos , Línea Celular , Codón/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Eliminación de Gen , Guanina/análogos & derivados , Guanina/farmacología , Homocigoto , Humanos , Pérdida de Heterocigocidad , O(6)-Metilguanina-ADN Metiltransferasa/antagonistas & inhibidores , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Recombinación Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
19.
Leukemia ; 14(7): 1301-9, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10914556

RESUMEN

Primary effusion lymphoma (PEL) represents a peculiar type of B cell lymphoma which associates with HHV-8 infection and preferentially grows in liquid phase in the serous body cavities. In this report, we provide the detailed characterization of a newly established PEL cell line, termed CRO-AP/6. The cell line was obtained from the pleural effusion of a HIV-positive patient with PEL. Its derivation from the tumor clone was established by immunogenotypic analysis. Detailed phenotypic investigations defined that CRO-AP/6 reflects pre-terminally differentiated B cells expressing the CD138/syndecan-1 antigen. Karyotypic studies of CRO-AP/6 identified several chromosomal abnormalities, whereas genotypic studies ruled out the involvement of molecular lesions associated with other types of B cell lymphoma. Both CRO-AP/6 and the parental tumor sample harbored infection by HHV-8. Conversely, EBV infection was present in the parental tumor sample although not in CROAP/6, indicating that CRO-AP/6 originated from the selection of an EBV-negative tumor subclone. The pattern of viral (HHV-8 v-cyclin) and cellular (p27Kip1) regulators of cell cycle expressed by CRO-AP/6, together with the results of growth fraction analysis, point to abrogation of the physiological inverse relationship between proliferation and p27Kip1 expression. Also, both CRO-AP/6 and the parental tumor sample display biallelic inactivation of the DNA repair enzyme gene O6-methylguanine-DNA methyltransferase (MGMT) by promoter methylation. Overall, the CRO-AP/6 cell line may help understand cell cycle control of PEL cells, may clarify the relative contribution of HHV-8 and EBV to the disease growth and development and may facilitate the identification of recurrent cytogenetic abnormalities highlighting putative novel cancer related loci relevant to PEL.


Asunto(s)
Infecciones por Herpesviridae/patología , Herpesvirus Humano 8/patogenicidad , Linfoma Relacionado con SIDA/patología , Linfoma de Células B/virología , Proteínas de Neoplasias/fisiología , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Derrame Pleural Maligno/patología , Células Tumorales Cultivadas/virología , Infecciones Tumorales por Virus/patología , Adulto , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Ciclo Celular , Aberraciones Cromosómicas , Células Clonales/patología , Células Clonales/virología , Ciclinas/biosíntesis , Ciclinas/genética , Metilación de ADN , Activación Enzimática , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Regulación Neoplásica de la Expresión Génica , Regulación Viral de la Expresión Génica , Genes Supresores de Tumor , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/aislamiento & purificación , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 8/inmunología , Herpesvirus Humano 8/aislamiento & purificación , Humanos , Inmunofenotipificación , Linfoma Relacionado con SIDA/etiología , Linfoma Relacionado con SIDA/genética , Linfoma Relacionado con SIDA/virología , Linfoma de Células B/genética , Linfoma de Células B/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , O(6)-Metilguanina-ADN Metiltransferasa/genética , O(6)-Metilguanina-ADN Metiltransferasa/fisiología , Derrame Pleural Maligno/genética , Derrame Pleural Maligno/virología , Regiones Promotoras Genéticas , Proto-Oncogenes , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/virología , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Latencia del Virus
20.
Carcinogenesis ; 21(2): 301-5, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10657972

RESUMEN

Mice with mutations in both alleles of the Mgmt and the Mlh1 gene, the former encoding a DNA repair methyltransferase and the latter a protein functioning at an early step of mismatch repair, are as resistant to the killing action of alkylating agents as are wild-type mice. These mice yielded a large number of tumors when exposed to alkylating carcinogens, but this characteristic was subdued since they also showed a relatively high level of spontaneous tumorigenicity, as a consequence of the defect in mismatch repair. This complexity is now resolved by introducing the Mlh1(+/-) mutation, instead of Mlh1(-/-), in these methyltransferase-deficient mice. Mgmt(-/-) Mlh1(+/-) mice, with about half the amount of MLH1 protein as Mgmt(-/-) Mlh1(+/+) mice, were resistant to the killing action of N-methyl-N-nitrosourea (MNU), up to the level of 30 mg/kg body wt. Eight weeks after exposure to this dose of MNU, 40% of MNU-treated Mgmt(-/-) Mlh1(+/-) mice had thymic lymphomas and there were no tumors in those mice not given the treatment. It seems that the cellular content of MLH1 protein is a critical factor for determining if damaged cells enter into either one of the two pathways leading to mutation induction or to apototic cell death. Loss of Mlh1 expression was frequently observed in tumors of Mgmt(-/-) Mlh1(+/-) mice and this might be related to progression of the tumors.


Asunto(s)
Alquilantes/toxicidad , Alelos , Carcinógenos/toxicidad , Eliminación de Gen , Neoplasias Intestinales/genética , Linfoma/genética , Metilnitrosourea/toxicidad , Proteínas de Neoplasias/genética , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , Proteínas Adaptadoras Transductoras de Señales , Alquilantes/farmacología , Animales , Carcinógenos/farmacología , Proteínas Portadoras , Codón/genética , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Progresión de la Enfermedad , Genes ras/efectos de los fármacos , Predisposición Genética a la Enfermedad , Genotipo , Neoplasias Intestinales/inducido químicamente , Dosificación Letal Mediana , Linfoma/inducido químicamente , Metilnitrosourea/farmacología , Ratones , Ratones Noqueados , Homólogo 1 de la Proteína MutL , Proteínas de Neoplasias/fisiología , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Proteínas Nucleares , O(6)-Metilguanina-ADN Metiltransferasa/genética , O(6)-Metilguanina-ADN Metiltransferasa/fisiología , Neoplasias del Timo/inducido químicamente , Neoplasias del Timo/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...