Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Nutr Metab Cardiovasc Dis ; 29(2): 191-200, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30573307

RESUMEN

BACKGROUND: There are no data on the prevalence of non-alcoholic fatty liver disease (NAFLD) in general population samples in Guatemala or in other Central American countries. The prevalence and distribution of NAFLD and its associated risk factors were evaluated in a population-based sample of adults in Guatemala. METHODS: Cross-sectional study of 411 men and women 40 years of age or older residing in urban and rural areas of Guatemala. Metabolic outcomes included obesity, central obesity, hypercholesterolemia, diabetes, and metabolic syndrome (MetS). Liver disease outcomes included elevated liver enzymes, elevated Fatty Liver Index (FLI), and elevated FIB-4 score. RESULTS: The overall prevalence of obesity, central obesity, diabetes, and MetS were 30.9, 74.3, 21.6, and 64.2%, respectively. The fully-adjusted prevalence ratios (95% CI) for obesity, central obesity, diabetes, and MetS comparing women to men were 2.83 (1.86-4.30), 1.72 (1.46-2.02), 1.18 (1.03-1.34), and 1.87 (1.53-2.29), respectively. The overall prevalence of elevated liver enzymes (ALT or AST), elevated FLI, and elevated FIB-4 scores were 38.4, 60.1, and 4.1%, respectively. The fully-adjusted prevalence ratios (95% CI) for elevated liver enzymes (either ALT or AST) and elevated FLI score comparing women to men were 2.99 (1.84-4.86) and 1.47 (1.18-1.84), respectively. CONCLUSIONS: The prevalence of metabolic abnormalities and liver outcomes in this general population study was very high. The prevalence of metabolic and liver abnormalities was particularly high among women, an observation that could explain the atypical 1:1 male to female ratio of liver cancer in Guatemala.


Asunto(s)
Síndrome Metabólico/epidemiología , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Adulto , Pruebas Enzimáticas Clínicas , Estudios Transversales , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/epidemiología , Femenino , Guatemala/epidemiología , Humanos , Hipercolesterolemia/diagnóstico , Hipercolesterolemia/epidemiología , Pruebas de Función Hepática , Masculino , Síndrome Metabólico/diagnóstico , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/diagnóstico , Obesidad Abdominal/diagnóstico , Obesidad Abdominal/epidemiología , Prevalencia , Factores de Riesgo , Salud Rural , Salud Urbana
2.
Nat Genet ; 28(1): 29-35, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11326271

RESUMEN

Hepatocellular carcinoma (HCC) is a major cause of cancer death, but the molecular mechanism for its development beyond its initiation has not been well characterized. Suppressor of cytokine signaling (SOCS-1; also known as JAB and SSI-1) switches cytokine signaling 'off' by means of its direct interaction with Janus kinase (JAK). We identified aberrant methylation in the CpG island of SOCS-1 that correlated with its transcription silencing in HCC cell lines. The incidence of aberrant methylation was 65% in the 26 human primary HCC tumor samples analyzed. Moreover, the restoration of SOCS-1 suppressed both growth rate and anchorage-independent growth of cells in which SOCS-1 was methylation-silenced and JAK2 was constitutively activated. This growth suppression was caused by apoptosis and was reproduced by AG490, a specific, chemical JAK2 inhibitor that reversed constitutive phosphorylation of STAT3 in SOCS-1 inactivated cells. The high prevalence of the aberrant SOCS-1 methylation and its growth suppression activity demonstrated the importance of the constitutive activation of the JAK/STAT pathway in the development of HCC. Our results also indicate therapeutic strategies for the treatment of HCC including use of SOCS-1 in gene therapy and inhibition of JAK2 by small molecules, such as AG490.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas Portadoras/genética , Metilación de ADN , Silenciador del Gen , Genes Supresores de Tumor , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas , Proteínas Represoras , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/terapia , Proteínas Portadoras/metabolismo , Islas de CpG , Proteínas de Unión al ADN/metabolismo , Terapia Genética , Humanos , Janus Quinasa 2 , Neoplasias Hepáticas/terapia , Datos de Secuencia Molecular , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Factor de Transcripción STAT3 , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Transactivadores/metabolismo , Tirfostinos/uso terapéutico
3.
Nat Biotechnol ; 16(13): 1352-6, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9853618

RESUMEN

A method has been developed to produce small DNA fragments from PCR products for analysis of defined DNA variations by mass spectrometry. The genomic region to be analyzed is PCR-amplified with primers containing a sequence for the type IIS restriction endonuclease Bpml. Bpml digestion of the resultant PCR products yields fragments as small as seven bases, which are then analyzed by electrospray ionization mass spectrometry. The approach was validated using seven different variants within the APC tumor suppressor gene, in which a perfect correlation was obtained with DNA sequencing. Both the sense and antisense strands were analyzed independently, and several variants can be analyzed simultaneously. These results provide the basis for a generally applicable and highly accurate method that directly queries the mass of variant DNA sequences.


Asunto(s)
ADN/genética , Genotipo , Espectrometría de Masas/métodos , Secuencia de Bases , Codón , ADN/química , Genes APC , Humanos , Reacción en Cadena de la Polimerasa , Mapeo Restrictivo
4.
Nucleic Acids Res ; 28(7): E24, 2000 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10710441

RESUMEN

Solar ultraviolet (UV) radiation induces DNA photoproducts in skin cells and is the predominant cause of human skin cancers. To understand human susceptibility to skin cancer and to facilitate the development of prevention measures, highly specific reagents to detect and quantitate UV-induced DNA adducts in human skin will be needed. One approach towards this end is the use of monoclonal antibody-based molecular dosimetry methods. To facilitate the development of photoproduct-specific antibody reagents we have: (i) cloned and sequenced a single chain variable fragment (ScFv) gene coding for one such high affinity monoclonal antibody, [alpha]UVssDNA-1 (mAb C3B6), recognizing the thymidine(6-4)thymidine photoproduct; (ii) expressed and displayed the cloned ScFv gene on the surface of phage; (iii) selected functional recombinant phage by panning; (iv) purified the ScFv peptide; (v) shown that the purified ScFv peptide binds to UV-irradiated polythymidylic acid but not unirradiated polythymidylic acid. This is the first demonstration of the use of phage display to select a ScFv recognizing DNA damage. In addition, this is the initial step towards immortalizing the antibody gene for genetic manipulation, structure-function studies and application to human investigations.


Asunto(s)
Aductos de ADN/análisis , Daño del ADN , Región Variable de Inmunoglobulina/genética , Biblioteca de Péptidos , Timidina/análisis , Secuencia de Aminoácidos , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Bacteriófagos/genética , Clonación Molecular , Aductos de ADN/química , Aductos de ADN/inmunología , Ensayo de Inmunoadsorción Enzimática , Escherichia coli , Región Variable de Inmunoglobulina/inmunología , Datos de Secuencia Molecular , Fotoquímica , Poli T/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Análisis de Secuencia de ADN , Timidina/análogos & derivados , Timidina/inmunología , Rayos Ultravioleta
5.
J Natl Cancer Inst ; 91(4): 347-54, 1999 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-10050868

RESUMEN

BACKGROUND: Residents of Qidong, People's Republic of China, are at high risk for development of hepatocellular carcinoma, in part due to consumption of foods contaminated with aflatoxins, which require metabolic activation to become carcinogenic. In a randomized, placebo-controlled, double-blind phase IIa chemoprevention trial, we tested oltipraz, an antischistosomal drug that has been shown to be a potent and effective inhibitor of aflatoxin-induced hepatocarcinogenesis in animal models. METHODS: In 1995, 234 adults from Qidong were enrolled. Healthy eligible individuals were randomly assigned to receive by mouth 125 mg oltipraz daily, 500 mg oltipraz weekly, or a placebo. Sequential immunoaffinity chromatography and liquid chromatography coupled to mass spectrometry or to fluorescence detection were used to identify and quantify phase 1 and phase 2 metabolites of aflatoxin B1 in the urine of study participants. Reported P values are two-sided. RESULTS: One month of weekly administration of 500 mg oltipraz led to a 51% decrease in median levels of the phase 1 metabolite aflatoxin M1 excreted in urine compared with administration of a placebo (P = .030), but it had no effect on levels of a phase 2 metabolite, aflatoxin-mercapturic acid (P = .871). By contrast, daily intervention with 125 mg oltipraz led to a 2.6-fold increase in median aflatoxin-mercapturic acid excretion (P = .017) but had no effect on excreted aflatoxin M1 levels (P = .682). CONCLUSIONS: Intermittent, high-dose oltipraz inhibited phase 1 activation of aflatoxins, and sustained low-dose oltipraz increased phase 2 conjugation of aflatoxin, yielding higher levels of aflatoxin-mercapturic acid. While both mechanisms can contribute to protection, this study highlights the feasibility of inducing phase 2 enzymes as a chemopreventive strategy in humans.


Asunto(s)
Aflatoxina B1/antagonistas & inhibidores , Anticarcinógenos/uso terapéutico , Carcinógenos/antagonistas & inhibidores , Neoplasias Hepáticas/prevención & control , Neoplasias Hepáticas/orina , Pirazinas/uso terapéutico , Acetilcisteína/orina , Aflatoxina B1/orina , Anticarcinógenos/administración & dosificación , Carcinógenos/metabolismo , China , Citocromo P-450 CYP1A2/metabolismo , Método Doble Ciego , Esquema de Medicación , Estudios de Factibilidad , Cromatografía de Gases y Espectrometría de Masas , Glutatión Transferasa/metabolismo , Humanos , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/enzimología , Pirazinas/administración & dosificación , Reproducibilidad de los Resultados , Tionas , Tiofenos , Resultado del Tratamiento
6.
Cancer Res ; 54(7 Suppl): 1907s-1911s, 1994 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-8137310

RESUMEN

The rapidly expanding understanding of the progressive processes of carcinogenesis provides opportunities for the identification of molecular biological markers reflecting events from exposure through clinical disease. These molecular biological markers can be classified into categories of markers of exposure reflecting the dose of toxic agents, markers of effect indicating a biological response to exposure, and markers of susceptibility providing information about the inherent sensitivity of individuals to the toxic agents. By definition some of these markers are chemical agent specific, such as a carcinogen-DNA or -protein adduct, while others are biological process specific, such as the altered expression of a gene. This article reviews the development and validation of molecular biomarkers of aflatoxins using experimental and human population studies. The development of molecular biomarkers for aflatoxins is based upon the extensive research database available about their metabolism, macromolecular adduct formation, and general mechanisms of action. The long-term goal of the research described in this paper is the application of aflatoxin biomarkers to the development of preventive interventions in human populations at high risk for liver cancer.


Asunto(s)
Aflatoxinas/toxicidad , Biomarcadores/análisis , Neoplasias/epidemiología , Neoplasias/prevención & control , Animales , Carcinógenos/metabolismo , ADN/metabolismo , Expresión Génica , Humanos , Neoplasias Hepáticas/epidemiología , Neoplasias Hepáticas/prevención & control , Proteínas/metabolismo , Factores de Riesgo
7.
Cancer Res ; 46(8): 3924-31, 1986 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2873884

RESUMEN

The effects of dietary administration of ethoxyquin (EQ) on aflatoxin B1 (AFB1) metabolism, DNA adduct formation and removal, and hepatic tumorigenesis were examined in male Fischer rats. Rats were fed a semipurified diet containing 0.4% EQ for 1 wk, gavaged with 250 micrograms of AFB1 per kg 5 times a wk during the next 2 wk, and, finally, restored to the control diet 1 wk after cessation of dosing. At 4 mo, focal areas of hepatocellular alteration were identified and quantitated by staining sections of liver for gamma-glutamyl transpeptidase. Treatment with EQ reduced by greater than 95% both area and volume of liver occupied by gamma-glutamyl transpeptidase-positive foci. Utilizing the same multiple dosing protocol, patterns of covalent modifications of DNA by AFB1 were determined. EQ produced a dramatic reduction in the binding of AFB1 to hepatic DNA: 18-fold initially and 3-fold at the end of the dosing period. Although binding was detectable at 3 and 4 mo postdosing, no effect of EQ was observed, suggesting that these persistent adducts are not of primary relevance to AFB1 carcinogenesis. Analysis of nucleic acid bases by high-performance liquid chromatography revealed no qualitative differences in adduct species between treatment groups. The inhibitory effect of EQ on AFB1 binding to DNA and tumorigenesis appears related to induction of detoxication enzymes. Rats fed 0.4% EQ for 7 days showed a 5-fold increase in hepatic cytosolic glutathione S-transferase (GST)-specific activities. Multiple molecular forms of GST were induced, and concomitant elevations in messenger RNA levels coding for the synthesis of GST subunits were observed. Correspondingly, biliary elimination of AFB1-glutathione conjugate was increased 4.5-fold in animals on the EQ diet during the first 2 h following p.o. administration of 250 micrograms of AFB1 per kg. Thus, induction by EQ of enzymes important to AFB1 detoxication, such as GST, can lead to enhanced carcinogen elimination, as well as reductions of AFB1-DNA adduct formation and subsequent expression of preneoplastic lesions, and, ultimately, neoplasia.


Asunto(s)
Aflatoxinas/metabolismo , ADN/metabolismo , Etoxiquina/farmacología , Glutatión Transferasa/biosíntesis , Guanina/análogos & derivados , Neoplasias Hepáticas Experimentales/inducido químicamente , Quinolinas/farmacología , Aflatoxina B1 , Aflatoxinas/toxicidad , Animales , Bilis/metabolismo , Hidroxianisol Butilado/farmacología , Hidroxitolueno Butilado/farmacología , Inducción Enzimática , Guanina/metabolismo , Hígado/enzimología , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/prevención & control , Masculino , Ratas , Ratas Endogámicas F344 , gamma-Glutamiltransferasa/análisis
8.
Cancer Res ; 52(2): 267-74, 1992 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-1728400

RESUMEN

The development of molecular dosimetry methods will simplify the identification of people at high risk for cancer. A combined monoclonal antibody immunoaffinity chromatography/high performance liquid chromatography method has been devised to isolate and quantify aflatoxin-DNA adducts and other metabolites in rat urine samples. We report the production of 11 different monoclonal antibodies recognizing aflatoxin B1, aflatoxin Q1, aflatoxin G1, aflatoxicol, and aflatoxin M1 and the application of these antibodies to a multiple monoclonal antibody affinity chromatography technique. Using the multiple monoclonal antibody affinity column with rat urines obtained from dosed animals, between 90 and 95% of total aflatoxin metabolites can be bound to the column and isolated. Analytical immunoaffinity chromatography/high performance liquid chromatography analysis of these isolated aflatoxins reveals that more than 55% of the aflatoxins in rat urine are aflatoxin-dihydrodiol, aflatoxin-N7-guanine, aflatoxin Q1, aflatoxin M1, aflatoxin P1, and aflatoxin B1, accounting for 1.5, 9.6, 1.8, 34.5, 8.0, and 1.0% of the total aflatoxins, respectively. Further, a perchloric acid digestion of the aflatoxin-N7-guanine peak was used to confirm its identity by its conversion to guanine. The measurement of aflatoxin-N7-guanine excretion in rat urine was examined to assess its utility as a marker of DNA adduct formation in the liver, and a dose-dependent excretion in urine was found with a correlation coefficient of 0.99. A comparison of the dose-dependent residual levels of aflatoxin binding to liver DNA with the amount of aflatoxin-N7-guanine excreted in urine showed a correlation coefficient of 0.98. Besides the nucleic acid adduct excretion data, aflatoxin M1 and aflatoxin P1 were evaluated as molecular dosimeters in the urine. Aflatoxin M1 was found to be an excellent marker, whereas no linear relationship between dose and aflatoxin P1 excretion in urine was found.


Asunto(s)
Aflatoxina B1/análogos & derivados , Aflatoxinas/inmunología , Guanina/análogos & derivados , Aflatoxina B1/inmunología , Aflatoxina B1/orina , Aflatoxinas/análisis , Aflatoxinas/metabolismo , Animales , Anticuerpos Monoclonales , Afinidad de Anticuerpos , Cromatografía de Afinidad , Cromatografía Líquida de Alta Presión , Guanina/inmunología , Guanina/orina , Ratas , Análisis de Regresión
9.
Cancer Res ; 47(16): 4271-7, 1987 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-2886217

RESUMEN

1,2-Dithiol-3-thiones, reported constituents of cruciferous vegetables, are five-membered cyclic sulfur-containing compounds with antioxidant, chemotherapeutic, and chemoprotective activities. The effects of dietary administration of a substituted 1,2-dithiol-3-thione, oltipraz [5-(2-pyrazinyl)-4-methyl-1,2-dithiol-3-thione], a potent antischistosomal agent, on aflatoxin B1 (AFB1) metabolism, DNA adduct formation, and hepatic tumorigenesis were examined in male F344 rats. Rats were fed graded doses of oltipraz (0.01-0.1%) for 4 wk. During the second and third wk of oltipraz feeding rats were gavaged with 250 micrograms of AFB1/kg five times a wk. Rats were finally restored to control diet 1 wk after cessation of AFB1 dosing. At 4 months focal areas of hepatocellular alteration were identified and quantitated by staining sections of liver for gamma-glutamyl transpeptidase activity. Treatment with oltipraz at all doses reduced by greater than 90% the volume of liver occupied by gamma-glutamyl transpeptidase-positive foci. Levels of AFB1 bound to hepatic DNA were reduced between 40 and 80% in animals fed increasing doses of dietary oltipraz (0.01-0.1%) for 1 wk prior to a single exposure to AFB1. Feeding of the higher levels of oltipraz led to marked increases in the specific activity of glutathione S-transferases, presumably serving to facilitate the detoxication of the ultimate electrophilic form of AFB1, the 8,9-oxide. At low dietary concentrations of oltipraz (0.01%), the only inductive effects seen were on the activities of selected cytochrome P-450 monooxygenases. Therefore, the protection afforded by oltipraz may be due to both the enhancement of electrophile detoxication pathways as well as modified oxidative metabolism of AFB1. In in vitro metabolism studies with hepatic post-mitochondrial supernatant, low-dose oltipraz pretreatment facilitated the oxidative production of aflatoxins P1 and Q1, but not M1, from AFB1. High-dose (0.1%) oltipraz pretreatment enhanced the primary metabolism of AFB1 to aflatoxins P1, M1, and Q1 as well as the formation of chloroform-insoluble metabolites. Feeding studies with a series of 1,2-dithiol-3-thione and 1,2-dithiol-3-one derivatives of oltipraz demonstrated that the inductive activity for cytochrome P-450-dependent monooxygenases and electrophile detoxication enzymes, such as glutathione S-transferases, could be readily separated by minor modifications of the 1,2-dithiol-3-thione structure. The unsubstituted 1,2-dithiol-3-thione nucleus strongly induced electrophile detoxication enzymes, but not the monooxygenases, and was the most effective inhibitor of the binding of AFB1 to hepatic DNA in vivo.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Aflatoxinas/toxicidad , Antineoplásicos/farmacología , Neoplasias Hepáticas Experimentales/prevención & control , Pirazinas/farmacología , Tionas/farmacología , Tiofenos/farmacología , Aflatoxina B1 , Aflatoxinas/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/biosíntesis , ADN/metabolismo , Inducción Enzimática/efectos de los fármacos , Glutatión Transferasa/biosíntesis , Compuestos Heterocíclicos/farmacología , Neoplasias Hepáticas Experimentales/inducido químicamente , Masculino , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/prevención & control , Ratas , Ratas Endogámicas F344 , Relación Estructura-Actividad , gamma-Glutamiltransferasa/análisis
10.
Cancer Res ; 51(20): 5501-6, 1991 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-1680553

RESUMEN

Previous studies have demonstrated that dietary administration of the schistosomicidal drug 5-(2-pyrazinyl)-4-methyl-1,2-dithiole-3-thione (oltipraz) ameliorates the hepatotoxicity of aflatoxin B1 (AFB1). Notably, mortality, altered hepatic function, hepatic AFB1-DNA adduct levels, and expression of hepatic enzyme-altered foci were markedly reduced in the rat by concurrent feeding of oltipraz during exposures to AFB1. Collectively, these studies prompted us to evaluate the chemoprotective properties of oltipraz against AFB1-induced liver cancer. In addition, preliminary molecular dosimetry studies were undertaken to determine the utility of measurements of urinary aflatoxin-N7-guanine excretion as a marker of relative risk for hepatocarcinogenesis in AFB1-exposed rats. For the carcinogenesis studies, 5-wk-old male F344 rats were randomly divided into two groups. One group (55 rats) received the AIN-76A diet, and the other group (56 rats) received the AIN-76A diet supplemented with 0.075% oltipraz. The oltipraz-supplemented diet was fed for 4 wk. Beginning 1 wk after starting the experimental diets, all rats in both groups received 25 micrograms of AFB1/rat/day by gavage for 5 days per wk over the next 2 wk. One wk following cessation of dosing with AFB1, oltipraz was removed from the diet, and all rats were fed the AIN-76A diet for the remainder of the experiment. At 3 mo after dosing, livers of ten sentinel rats from each group were analyzed for the burden of gamma-glutamyltranspeptidase-positive foci. In accord with previous findings, rats fed the oltipraz-supplemented diet exhibited substantial reductions in the focal burden (97% reduction; P less than 0.05) of these AFB1-induced lesions. The remaining rats were maintained for the cancer study until they became moribund or the termination of the experiment at 23 mo. Gross liver lesions were identified at autopsy and confirmed by microscopic evaluation. An 11% incidence of hepatocellular carcinoma was observed in the AFB1-treated, control diet-fed rats. An additional 9% of this group had hepatocellular adenomas. Oltipraz afforded complete protection against both AFB1-induced hepatocellular neoplasms. Using Kaplan-Meier survival analyses, rats in the oltipraz group had a significantly (P less than 0.02) longer life span and an increased survival free of liver tumors (P less than 0.0002). Molecular dosimetry studies used rats fed either the oltipraz-supplemented or control diet for 1 wk and then challenged with a single dose of AFB1 to examine the initial rates of 8,9-dihydro-8-(N7-guanyl)-9-hydroxyaflatoxin B1 excreted in the urine.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Adenoma/prevención & control , Aflatoxina B1/antagonistas & inhibidores , Neoplasias Hepáticas Experimentales/prevención & control , Pirazinas/farmacología , Adenoma/inducido químicamente , Adenoma/mortalidad , Aflatoxina B1/metabolismo , Aflatoxina B1/toxicidad , Animales , ADN/metabolismo , Crecimiento/efectos de los fármacos , Hígado/enzimología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/mortalidad , Masculino , Ratas , Ratas Endogámicas F344 , Análisis de Supervivencia , Tionas , Tiofenos , gamma-Glutamiltransferasa/análisis
11.
Cancer Res ; 52(20): 5668-73, 1992 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-1394191

RESUMEN

The mutagenic spectrum induced by aflatoxin-DNA lesions in DNA repair deficient and repair proficient human cells was investigated. The reactive metabolite aflatoxin B1-8,9-epoxide was synthesized and reacted in vitro with the shuttle vector plasmid pS189. Plasmids were transfected into human fibroblasts and allowed to replicate, and the recovered plasmids were screened in indicator bacteria for plasmid survival and mutations in the supF marker gene. Sequence data were obtained from 71 independently arising mutants recovered from DNA repair deficient xeroderma pigmentosum (XP) cells [XP12BE(SV40)] and 60 mutants recovered from a DNA repair proficient cell line (GM0637). Plasmid survival was lower and mutation frequency higher with the XP cells, and the mutation hotspots differed substantially for the 2 cell lines. Most mutations (> 90%) were base substitutions at G:C pairs, only about one-half of which were G:C-->T:A transversions, the expected predominant mutation. One-third of the mutations at GG sites and none of those at isolated Gs were G:C-->A:T transitions. Tandem base substitutions also occurred only at GG sites and were found only with XP cells. The location of mutation hotspots with either cell line did not correlate with the level of modification within the sequence as assessed by a DNA polymerase stop assay. These results suggest that the DNA repair deficiency associated with XP can influence not only the overall frequency of mutations but also the distribution of mutations within a gene. The finding of transition mutations exclusively at GG sites may be of predictive value in attempts to link dietary aflatoxin exposure to cancers associated with specific mutations in the c-ras oncogene and the p53 tumor suppressor gene.


Asunto(s)
Aflatoxina B1/toxicidad , Reparación del ADN , Replicación del ADN/efectos de los fármacos , Mutación/efectos de los fármacos , Plásmidos/efectos de los fármacos , Xerodermia Pigmentosa/genética , Aflatoxina B1/genética , Secuencia de Bases/efectos de los fármacos , Línea Celular Transformada , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/toxicidad , Vectores Genéticos/efectos de los fármacos , Humanos , Datos de Secuencia Molecular , Pruebas de Mutagenicidad , Xerodermia Pigmentosa/patología
12.
Cancer Res ; 57(4): 641-5, 1997 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9044840

RESUMEN

The in vivo activation of aflatoxin B1 (AFB1) was assessed by using two transgenic mouse lines, M2 and M10, in which the human fetus-specific CYP3A7 was expressed in the kidney (M2) and the liver (M10), respectively. Male mice of 8 weeks old from these two lines were treated with a single i.p. injection of AFB1 (4 mg/kg body weight). AFB1-N7-guanine adduct was quantified by high-performance liquid chromatography. DNA damage was measured using the alkaline elution technique 2 and 6 h after AFB1 treatment. Administration of AFB1 resulted in a significantly higher level of AFB1-N7-guanine in the livers of M10 transgenic mice compared with their nontransgenic littermates (16.5 +/- 4.2 versus 10.4 +/- 1.2 ng/mg DNA, P < 0.01). The level of this biomarker was also significantly higher in the kidney of the M2 mice compared with control mice (73.0 +/- 6.3 versus 50.2 +/- 9.5 ng/mg DNA, P < 0.01). Similar results were also observed with DNA damage expressed as normalized area above curve (NAAC) in the two transgenic lineages, e.g., NAAC values were significantly higher in the livers of M10 and the kidneys of M2 mice. A dose-response relationship of NAAC values was observed in the livers of M10 mice when treated with AFB1 at different doses ranging from 1 to 16 mg/kg body weight, whereas in nontransgenic mice, only slight but not statistically significant increases of NAAC values were observed. Both the mouse CYP3A11 and GST-Yc subunit were expressed at identical levels in these transgenic lines. The results of this study present further evidence that human fetuses are also under the carcinogenic attack of AFB1 as adults if exposed to this potent carcinogen.


Asunto(s)
Aflatoxina B1/farmacocinética , Hidrocarburo de Aril Hidroxilasas , Sistema Enzimático del Citocromo P-450/genética , Daño del ADN , Animales , Biotransformación , Carcinógenos/toxicidad , Citocromo P-450 CYP3A , ADN/efectos de los fármacos , Dimetilsulfóxido/toxicidad , Feto , Guanina/metabolismo , Humanos , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL/genética , Ratones Transgénicos
13.
Cancer Res ; 42(8): 3120-4, 1982 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6807538

RESUMEN

Five monoclonal antibodies were obtained after fusion of mouse P3 x 63 myeloma cells with spleen cells isolated from BALB/c mice that had been immunized with aflatoxin B1-adducted DNA complexes with methylated bovine serum albumin. Selected hybridomas were found to produce monoclonal antibodies specific for modified DNA containing both the 2,3-dihydro-2-(N7-guanyl)-3-hydroxyaflatoxin B1 and the putative 2,3-dihydro-2-(N5-formyl-2',5',6'-triamino-4'-oxo-N5-pyrimidyl)-3-hydroxyaflatoxin B1, suggesting that these DNA adducts share a common antigenic determinant. Enzyme immunoassay conditions using these monoclonal antibodies were optimized, and DNA isolated from the livers of rats given dosages of aflatoxin B1 ranging from 0.01 to 1.0 mg aflatoxin B1 per kg body weight was tested. A level of modification in DNA of 1 aflatoxin B1 residue per 1,355,000 nucleotides can be quantitatively measured. Monoclonal antibodies will be useful probes for studying the molecular interactions of aflatoxin B1 with DNA and the occurrence of aflatoxin B1:DNA adducts in tissues and cells of humans exposed to this environmental carcinogen.


Asunto(s)
Aflatoxinas/farmacología , Anticuerpos Monoclonales , Carcinógenos/farmacología , Núcleo Celular/metabolismo , ADN/metabolismo , Hígado/metabolismo , Aflatoxina B1 , Aflatoxinas/metabolismo , Animales , Núcleo Celular/efectos de los fármacos , ADN/inmunología , Femenino , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Endogámicas
14.
Cancer Res ; 48(22): 6336-42, 1988 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-3180051

RESUMEN

To assess target-tissue exposure to the human urinary bladder carcinogen 4-aminobiphenyl (ABP), we have developed a sensitive immunochemical method for measuring the major arylamine-DNA adduct formed, N-(guan-8-yl)-ABP (Gua-C8-ABP). High-affinity polyclonal antisera from rabbits immunized with N-(guanosin-8-yl)-ABP coupled to keyhole limpet hemocyanin were characterized and shown to have high specificity for antigenic determinants on the purine and biphenyl rings of Gua-C8-ABP and minimal cross-reactivity with ABP, deoxyguanosine, or hydrolyzed DNA. Assay standards containing ABP-modified DNA were prepared by reacting [3H]N-hydroxy-ABP with calf thymus DNA. DNA samples were hydrolyzed with trifluoroacetic acid and dried under vacuum, and the residues were dissolved in dimethyl sulfoxide under argon. Using a streptavidin-biotin amplified enzyme-linked immunosorbent assay, DNA hydrolysates competed at 25 micrograms DNA/microtiter well for a limiting amount of anti-keyhole limpet hemocyanin-(Gua-C8-ABP) in the presence of excess solid-phase bovine serum albumin-(Gua-C8-ABP) coating antigen. The limit of sensitivity for this assay using 25 micrograms DNA was 2 adducts/10(8) nucleotides. Gua-C8-ABP adducts in liver and bladder epithelial DNAs were readily quantified after p.o. administration of 5 mg/kg ABP to dogs. This methodology is capable of detecting adducts at levels of biological significance and should be applicable to human target-tissue dosimetry.


Asunto(s)
Compuestos de Aminobifenilo/metabolismo , Carcinógenos/metabolismo , ADN/metabolismo , Compuestos de Aminobifenilo/inmunología , Animales , Reacciones Cruzadas , ADN/inmunología , Ensayo de Inmunoadsorción Enzimática , Sueros Inmunes/inmunología , Conejos , Neoplasias de la Vejiga Urinaria/inducido químicamente
15.
Cancer Res ; 45(5): 2116-22, 1985 May.
Artículo en Inglés | MEDLINE | ID: mdl-3921236

RESUMEN

A significant population of humans (5 to 10%) are phenotypic poor metabolizers of debrisoquine. We have isolated the cytochrome P-450 isozyme from rat liver responsible for this activity and have shown that antibodies raised against the protein are able to inhibit this catalytic activity in human liver microsomes (Distlerath, L. M., and Guengerich, F. P., Proc. Natl. Acad. Sci. USA, 81: 7348-7352, 1984). These antibodies were utilized to determine which metabolic transformations are linked to debrisoquine 4-hydroxylation in human liver microsomes using techniques of immunochemical inhibition. The antibodies almost completely inhibited debrisoquine 4-hydroxylation and bufuralol 1'-hydroxylation in microsomes prepared from several different human livers. The oxidation of the pyrrolizidine alkaloids lasiocarpine and monocrotaline were inhibited by roughly one-third. The antibodies did not inhibit N,N-dimethylnitrosamine N-demethylation, oxidation of vinylidene chloride to 2,2-chloroacetaldehyde, oxidation of trichloroethylene to chloral, N-oxidation of azoprocarbazine, morphine N-demethylation, diazepam N-demethylation, oxidation of benzo(a)pyrene to alkali-soluble metabolites, oxidation of benzo(a)pyrene 7,8-dihydrodiol to products covalently bound to DNA, the N- and ring-oxidation of 1- and 2-naphthylamine and 2-aminofluorene, or the conversion of aflatoxin B1 to DNA adducts or aflatoxin Q1. Studies with space-filling models of the drugs the metabolism of which is associated with debrisoquine 4-hydroxylase in the literature indicated that all can be fitted to a general structure in which a basic nitrogen is about 5 A away from the site of carbon hydroxylation and a hydrophobic domain is near the site of hydroxylation. These results may be useful in predicting which chemicals may or may not be metabolized in an atypical manner by a segment of the human population.


Asunto(s)
Sistema Enzimático del Citocromo P-450/análisis , Hígado/enzimología , Oxigenasas de Función Mixta/análisis , Aflatoxina B1 , Aflatoxinas/metabolismo , Citocromo P-450 CYP2D6 , Sistema Enzimático del Citocromo P-450/inmunología , Debrisoquina/metabolismo , Humanos , Hidroxilación , Inmunoquímica , Microsomas Hepáticos/enzimología , Especificidad por Sustrato
16.
Cancer Res ; 53(15): 3499-504, 1993 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8339253

RESUMEN

Oltipraz [5-(2-pyrazinyl)-4-methyl-1,2-dithiole-3-thione] protects against aflatoxin B1-induced hepatocarcinogenesis in rats when fed before and during carcinogen exposure; however, such an exposure-chemoprotection intervention paradigm is not directly relevant to most human populations. To model and assess the possible efficacy of short term interventions targeted at individuals at risk for sustained exposure to aflatoxins, 175-g male F344 rats were treated daily with 25 micrograms of aflatoxin B1, p.o., for 28 days. One week after the start of aflatoxin B1 exposure, half of the animals were fed a diet supplemented with 0.075% oltipraz for 10 days; these rats were then restored to the unsupplemented AIN-76A diet for the remainder of the experimental period. Livers were analyzed 2 or 3 months after the last aflatoxin B1 dose for burden of glutathione S-transferase P (GST-P)-positive foci, as an index of presumptive preneoplastic tumors. The transient intervention with oltipraz reduced the volume percent of hepatic GST-P-positive foci by 54% (P = 0.047) and 72% (P = 0.004) at 2 and 3 months, respectively. A strong positive correlation was also observed between the extent of fibrosis in the livers of these animals and the hepatic burden of GST-P-positive foci, implying that cytotoxicity is associated with the tumorigenic process. This protection may reflect alterations in the metabolism and disposition of aflatoxin B1 induced by oltipraz. Glutathione S-transferase catalyze the detoxication of aflatoxin-8,9-oxide and were found to be rapidly induced in the livers of animals after the beginning of the oltipraz intervention. Glutathione S-transferase activity remained significantly (P < 0.05) higher until 9 days after the end of the oltipraz intervention. In contrast, levels of hepatic aflatoxin-DNA adducts were not significantly reduced until 4 days after the beginning of the intervention but remained significantly (P < 0.05) lower up to 11 days after the end of the intervention. The cumulative reduction in levels of hepatic aflatoxin-DNA adducts (approximately 25%) by the oltipraz intervention underestimated the reduction in the hepatic burden of GST-P-positive foci. The significant protection against presumptive preneoplastic tumors, despite the delay of intervention, suggests that oltipraz may exert substantial activity against the cytotoxic and autopromoting action of repeated exposures to aflatoxin B1 and supports the utility of intervention trials with oltipraz in individuals chronically consuming aflatoxin B1-contaminated foods, particularly in regions with high incidences of liver cancer.


Asunto(s)
Aflatoxina B1/toxicidad , Anticarcinógenos/farmacología , Aductos de ADN , Neoplasias Hepáticas Experimentales/prevención & control , Pirazinas/farmacología , Aflatoxina B1/metabolismo , Animales , ADN/metabolismo , Glutatión Transferasa/análisis , Glutatión Transferasa/antagonistas & inhibidores , Neoplasias Hepáticas Experimentales/inducido químicamente , Masculino , Ratas , Ratas Endogámicas F344 , Tionas , Tiofenos
17.
Cancer Res ; 52(1): 45-52, 1992 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-1727385

RESUMEN

Hepatocellular carcinoma is one of the five leading human cancers causing at least 250,000 deaths each year. One of the major risk factors for this disease is exposure to dietary aflatoxins, and the development of appropriate molecular dosimetry biomarkers would facilitate the identification of individuals at risk. This study was undertaken to explore the relationship between dietary intake of aflatoxins and the excretion of the major aflatoxin-DNA adduct and other metabolites into the urine of chronically exposed people. The following protocol was developed for this investigation in Guangxi Autonomous Region, People's Republic of China, where the diets of 30 males and 12 females (ages, 25-64 years) were monitored for 1 week and aflatoxin intake levels determined each day. Starting on the fourth day, total urine volumes were obtained in consecutive 12-h fractions for 3 or 4 days. High performance liquid chromatography and competitive radioimmunoassay analyses were done on each of the urine samples, and the relationships between excretion of total aflatoxin metabolites, aflatoxin-N7-guanine, aflatoxin M1, aflatoxin P1, and aflatoxin B1, and aflatoxin B1 intake values were determined. The average intake of aflatoxin B1 by men was 48.4 micrograms/day, giving a total mean exposure during the study period of 276.8 micrograms. The average daily intake by women was 77.4 micrograms/day, resulting in a total average exposure during the 7-day period of 542.6 micrograms aflatoxin B1. Initial efforts to characterize aflatoxin metabolites in urine samples were with an analysis by competitive radioimmunoassay. The analysis by linear regression of the association between aflatoxin B1 intake/day and total aflatoxin metabolite excretion/day showed a correlation coefficient of only 0.26. These findings stimulated the immunoaffinity/analytical high performance liquid chromatography analysis for individual metabolites. When the data were analyzed by linear regression analysis, the aflatoxin N7-guanine excretion and aflatoxin B1 intake from the previous day showed a correlation coefficient of 0.65 and P less than 0.000001. Similar analysis for aflatoxin M1 resulted in a correlation coefficient of 0.55 and P less than 0.00001, whereas there was no positive statistical association between exposure in the diet and aflatoxin P1 excretion, despite aflatoxin P1 being quantitatively a major metabolite. Analysis of the total aflatoxin-N7-guanine excretion in the urine during the complete collection period plotted against the total aflatoxin B1 exposure in the diet for each of the individuals, smoothing the day to day variations, revealed a correlation coefficient of 0.80 and P less than 0.0000001.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Aflatoxina B1/orina , ADN/orina , Adulto , Aflatoxina B1/administración & dosificación , Aflatoxina B1/metabolismo , Aflatoxina M1/orina , Aflatoxinas/orina , China , Cromatografía Líquida de Alta Presión , ADN/metabolismo , Exposición a Riesgos Ambientales , Femenino , Contaminación de Alimentos , Humanos , Masculino , Persona de Mediana Edad , Radioinmunoensayo , Zea mays
18.
Cancer Res ; 61(1): 103-9, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11196146

RESUMEN

The prostate has been identified as a target for 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP)-induced carcinogenesis. Humans are exposed to PhIP through ingestion of well-done cooked meats, and there is evidence from epidemiological studies that implicates red meat consumption in prostate carcinogenesis. The alpha and pi class isoforms of glutathione S-transferases (GSTs) have been shown to inhibit adduction of activated PhIP metabolites to DNA in cell-free systems. In humans, silencing of GST pi(GSTP1) through CpG island hypermethylation is found in nearly all prostate carcinomas and is believed to be an early event in prostate carcinogenesis. We hypothesized that suppressed GSTP1 expression in prostate cells would increase their vulnerability to cytotoxicity and DNA adduct formation mediated by activated PhIP metabolites. To test this hypothesis, the human prostate adenocarcinoma cell line, LNCaP, which contains a silenced GSTP1 gene, was genetically modified to constitutively express high levels of GSTP1. Both LNCaP and LNCaP-GSTP1 cells exposed to N-OH-PhIP, but not parent PhIP, for 24 h showed a dose-dependent decrease in cell viability. GSTP1-overexpressing cells had LC50s 30-40% higher than cells transfected with the vector alone. PhIP-DNA adducts isolated from LNCaP-derived cells and primary human prostate tissue cultures exposed to N-OH-PhIP were analyzed by liquid chromatography/electrospray ionization mass spectrometry. Primary cultures of human prostate tissue and LNCaP-GSTP1 cells had approximately 50% lower adduct levels than parental LNCaP and vector control cells. Bioactivation assays using LNCaP cytosols showed that enzymatic activation of N-OH-PhIP to a DNA binding species was dependent on ATP and could be inhibited by recombinant human GSTP1 in the presence of glutathione. This evidence confirms that N-OH-PhIP can be bioactivated to a DNA binding species in human prostate and human prostate-derived cells. These observations provide the basis for using LNCaP and LNCaP-GSTP1 cells as a model system for studying the role of this enzyme in protection against N-OH-PhIP induced DNA damage in prostate carcinogenesis. Loss of GSTP1 expression in human prostate may, therefore, enhance its susceptibility to carcinogenic insult by compounds such as N-OH-PhIP. Conversely, induction of GSTs in early-stage prostate carcinogenesis may be a useful protective strategy.


Asunto(s)
Carcinógenos/toxicidad , Aductos de ADN/biosíntesis , Glutatión Transferasa/metabolismo , Imidazoles/toxicidad , Isoenzimas/metabolismo , Próstata/efectos de los fármacos , Piridinas/toxicidad , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Adenocarcinoma/prevención & control , Anciano , Biotransformación , Carcinógenos/antagonistas & inhibidores , Carcinógenos/farmacocinética , Citosol/metabolismo , ADN/efectos de los fármacos , ADN/metabolismo , Gutatión-S-Transferasa pi , Glutatión Transferasa/biosíntesis , Glutatión Transferasa/genética , Humanos , Imidazoles/antagonistas & inhibidores , Imidazoles/metabolismo , Imidazoles/farmacocinética , Isoenzimas/biosíntesis , Isoenzimas/genética , Masculino , Persona de Mediana Edad , Próstata/enzimología , Próstata/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/prevención & control , Piridinas/antagonistas & inhibidores , Piridinas/farmacocinética , Transfección , Células Tumorales Cultivadas
19.
Cancer Res ; 61(1): 33-5, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11196182

RESUMEN

Hepatocellular carcinoma (HCC), a common cause of cancer deaths worldwide, has several major etiological risk factors, including infection with the hepatitis viruses and exposure to aflatoxin B1. A specific missense mutation resulting from a guanine to thymine transversion at the third position of codon 249 in the p53 tumor suppressor gene has been reported in 10-70% of HCCs from areas of high dietary exposure to aflatoxin B1. Short oligonucleotide mass analysis was compared with DNA sequencing in 25 HCC samples for specific p53 mutations. Mutations were detected in 10 samples by short oligonucleotide mass analysis in agreement with DNA sequencing. Analysis of another 20 plasma and tumor pairs showed 11 tumors containing the specific mutation, and this change was detected in six of the paired plasma samples. Four of the plasma samples had detectable levels of the mutation; however, the tumors were negative, suggesting possible multiple independent HCCs. Ten plasma samples from healthy individuals were all negative. This molecular diagnostic technique has implications for prevention trials and for the early diagnosis of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Genes p53/genética , Neoplasias Hepáticas/genética , Espectrometría de Masa por Ionización de Electrospray/métodos , Carcinoma Hepatocelular/sangre , Estudios de Cohortes , Análisis Mutacional de ADN/métodos , ADN de Neoplasias/análisis , ADN de Neoplasias/sangre , ADN de Neoplasias/genética , Femenino , Humanos , Neoplasias Hepáticas/sangre , Masculino , Mutación , Oligonucleótidos/análisis , Oligonucleótidos/genética , Estudios Prospectivos
20.
Pharmacol Ther ; 34(2): 321-34, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3120202

RESUMEN

Primary hepatocellular carcinoma is one of the most lethal and common cancers in the world. It is particularly prevalent on the continents of Africa and Asia. A number of epidemiological studies have associated the exposure status of people to aflatoxin B1 as being important in the etiology of liver cancer. However, to date these studies have relied upon the criteria of presumptive intake data, rather than relying upon quantitative analyses of aflatoxin-DNA adduct and metabolite content obtained by monitoring biological fluids from exposed people. Information obtained by monitoring exposed individuals for specific DNA adducts and metabolites will define the pharmacokinetics of aflatoxin B1 in people, thereby facilitating risk assessments. We have developed monoclonal antibodies specific for aflatoxin metabolites, especially the DNA adducts. These monoclonal antibodies are used in solid phase immunoassays for the preparative purification of these aflatoxin derivatives from biological fluids. These methods in conjunction with other analytical procedures have resulted in the development of rapid protocols used to quantitatively measure aflatoxins in urine obtained from people dietarily exposed to this carcinogen. The people examined live in the Gambia and high liver cancer regions in the People's Republic of China. We have recently completed a pilot study in China where we have now begun to identify the pharmacokinetic parameters associated with chronic exposure of aflatoxin B1 in the diet. Despite the development of these new technologies, we must continue to define animal model systems which can be used to interpret the human data. Our work using animal models based on the differential effects of ethoxyquin on the kinetics of aflatoxin-DNA adducts and gamma glutamyl transpeptidase-positive foci formation indicate that the direct linear extrapolation of total adduct content in target tissues to dose may be inappropriate to assign risk to people (or rats). However, our findings do support the concept that measurement of the major, rapidly excised AFB1-N7-Gua adduct in tissues and fluids is an appropriate dosimeter for estimating exposure status and risk in individuals consuming this mycotoxin. Further studies employing different classes of modifiers of aflatoxin carcinogenesis in rodent models should better define the relationships between hepatic and urinary levels of AFB1-N7-Gua and susceptibility to neoplasia.


Asunto(s)
Aflatoxinas/toxicidad , Anticuerpos Monoclonales , Daño del ADN , Reparación del ADN , Aflatoxina B1 , Animales , Cromatografía de Afinidad , Humanos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda