Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Food Chem Toxicol ; 41(2): 193-200, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12480297

RESUMEN

Dose-response studies have shown a sharp threshold for the renal papillary toxic effect in male rats between 0.25% and 0.5% ethoxyquin (Eto) in the diet over 6 months. Although similar elevated urinary protein (albumin) levels resulted from dietary Eto (0.5%) in both males and females, papillary necrosis was male specific. Following [14C]Eto administration, radiolabel was associated with urinary albumin but not alpha 2 globulin (alpha(2mu)-g). Autoradiographic studies indicated that the sex differences in nephrotoxicity do not involve differences in distribution or retention of Eto. Faecal and urinary metabolic profiles were also similar in the two sexes. The sharp threshold of toxicity in the male rat could indicate a fine balance between toxifying/detoxifying metabolism of Eto.


Asunto(s)
Antioxidantes/toxicidad , Etoxiquina/toxicidad , Enfermedades Renales/inducido químicamente , Médula Renal/efectos de los fármacos , Administración Oral , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacocinética , Dieta , Relación Dosis-Respuesta a Droga , Etoxiquina/administración & dosificación , Etoxiquina/farmacocinética , Femenino , Enfermedades Renales/patología , Médula Renal/patología , Masculino , Necrosis , Ratas , Ratas Endogámicas F344 , Caracteres Sexuales
2.
Food Addit Contam ; 18(4): 329-41, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11339268

RESUMEN

One of the most important concerns in the decontamination of aflatoxin-containing feed commodities is the safety of the products for food-producing animals and for human consumption of products derived from these animals. A new method, based on the use of florisil and C18 solid phase extraction columns, was developed for the preparation of extracts from decontaminated peanut meal, which allowed testing with in vitro genotoxicity assays without interference of the residual aflatoxin B1. Recovery of degradation products in the extracts was evaluated by the use of radiolabelled [14C]-aflatoxin B1 (AFB1) added to naturally-contaminated peanut meal (3.5 mg AFB1/kg). The meal was treated by a small-scale version of an industrial decontamination process based on ammoniation. Following decontamination, more than 90% of the label could not be extracted from the meal. AFB1 accounted for about 10% of the radiolabel present in the extractable fraction, indicating a total AFB1 reduction of more than 99%. Decontamination of the meal by a number of other small- and industrial-scale ammonia-based processes resulted in similar efficiencies. Application of the extraction procedure resulted in AFB1-rich and AFB1-poor fractions, the latter containing half of the extractable decontamination products but less than 1% of the residual AFB1. Testing in the Salmonella/microsome mutagenicity assay (TA 100, with S9-mix) of the original crude extracts and AFB1-rich fractions prepared from non-treated and decontaminated meal, showed the positive results expected from the AFB1 contents as determined by HPLC analysis. Analysis and testing of the AFB1-poor fractions showed that the various decontamination processes not only resulted in a successful degradation of AFB1 but also did not produce other potent mutagenic compounds. Slight positive results obtained with these extracts were similar for the untreated and treated meals and may be due to unknown compounds originally present in the meal. Results obtained with an unscheduled DNA synthesis (UDS) and Comet assay with rat hepatocytes supported this conclusion. Positive results obtained with the micronucleus assay, using immortalized mouse hepatocytes (GKB), did not clearly reflect the mycotoxin levels and require further examination. It is concluded that the newly developed extraction procedure yields highly reproducible fractions and hence is very suitable for examining the possible formation of less potent degradation products of aflatoxins in short-term genotoxicity tests.


Asunto(s)
Aflatoxinas/análisis , Alimentación Animal/análisis , Arachis/química , Descontaminación , Animales , Cromatografía Líquida de Alta Presión , Electroforesis/métodos , Masculino , Pruebas de Mutagenicidad , Ratas , Ratas Endogámicas F344 , Sensibilidad y Especificidad
3.
Food Addit Contam ; 18(2): 137-49, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11288911

RESUMEN

A sample of peanut meal, highly contaminated with aflatoxins, has been subjected to decontamination by two commercial ammonia-based processes. The original contaminated and the two decontaminated meals were fed to rats for 90 days. No lesions associated with aflatoxin-induced hepatocarcinogenesis were detected histologically following feeding with the two detoxified meals. There were, however, clear differences between the two meals in respect of growth rates of the rats. In addition, feeding one of the detoxified meals resulted in hepatic abnormalities detected using novel immunohistochemical reagents. Differences between the two detoxified meals were also indicated by the results of studies using meals 'spiked' with [14C]-aflatoxin B1 prior to being subjected to the detoxification processes. The meals differed in the bioavailability of the label. It was concluded that peanut meal where an initial, unacceptable level of contamination with aflatoxins had been reduced by two ammonia-based processes to comparable, acceptable levels, may still have different effects in vivo when incorporated into animal diets.


Asunto(s)
Aflatoxinas/toxicidad , Amoníaco , Arachis , Contaminación de Alimentos , Aflatoxinas/farmacocinética , Animales , Disponibilidad Biológica , Descontaminación/métodos , Ingestión de Alimentos/efectos de los fármacos , Técnicas para Inmunoenzimas , Riñón/patología , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Masculino , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Aumento de Peso/efectos de los fármacos
4.
Food Chem Toxicol ; 39(3): 287-91, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11278061

RESUMEN

Six plant species (Cassia sophera, Chamaecrista nigricans, Mitragyna inermis, Ocimum americanum, Securidaca longepedunculata and Synedrella nodiflora) traditionally used in Ghana to control insect pests of stored grain and legumes were screened in the laboratory at three concentrations (0.5, 1 and 5%, w/w) against four common storage pests (Rhyzopertha dominica, Callosobruchus maculatus, Sitophilus zeamais and Prostephanus truncatus). All the plants showed some ability to control all or some of the test insect species. Levels of efficacy varied according to test concentration with the highest concentration tested providing the best control. The S. longepedunculata plant induced the highest percent mortality and was the best at reducing emergence of the F(1) generation. The six plants were also incorporated into standard rat diet at two concentrations (1 and 5%, w/w) and fed to rats over a 6-week period to assess potential deleterious effects against vertebrates. None of the plants demonstrated any neurotoxicological or neurobehavioural effects to the rats over the course of the trial. However, S. longepedunculata and C. nigricans caused a significant reduction in rat growth rate when incorporated at 5% in the diet, induced cell hyperplasia in the liver, and reduced the mean weight of the liver and kidneys, compared to the control group of rats. Kidney pathology was affected only by the 5% concentration of S. longepedunculata which caused a reduced accumulation of alpha2mu-globulin. The implications of these results are discussed in the context of farmer usage of insecticidal plants for stored product protection.


Asunto(s)
Insectos/fisiología , Insecticidas/toxicidad , Plantas , Vertebrados/fisiología , Animales , Escarabajos , Dieta , Ghana , Ratas , Tritio , Aumento de Peso
5.
Cancer Res ; 60(4): 957-69, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10706111

RESUMEN

Structurally diverse compounds can confer resistance to aflatoxin B1 (AFB1) hepatocarcinogenesis in the rat. Treatment with either phytochemicals [benzyl isothiocyanate, coumarin (CMRN), or indole-3-carbinol] or synthetic antioxidants and other drugs (butylated hydroxyanisole, diethyl maleate, ethoxyquin, beta-naphthoflavone, oltipraz, phenobarbital, or trans-stilbene oxide) has been found to increase hepatic aldo-keto reductase activity toward AFB1-dialdehyde and glutathione S-transferase (GST) activity toward AFB1-8,9-epoxide in both male and female rats. Under the conditions used, the natural benzopyrone CMRN was a major inducer of the AFB1 aldehyde reductase (AFAR) and the aflatoxin-conjugating class-alpha GST A5 subunit in rat liver, causing elevations of between 25- and 35-fold in hepatic levels of these proteins. Induction was not limited to AFAR and GSTA5: treatment with CMRN caused similar increases in the amount of the class-pi GST P1 subunit and NAD(P)H: quinone oxidoreductase in rat liver. Immunohistochemistry demonstrated that the overexpression of AFAR, GSTA5, GSTP1, and NAD(P)H:quinone oxidoreductase affected by CMRN is restricted to the centrilobular (periacinar) zone of the lobule, sometimes extending almost as far as the portal tract. This pattern of induction was also observed with ethoxyquin, oltipraz, and trans-stilbene oxide. By contrast, induction of these proteins by beta-naphthoflavone and diethyl maleate was predominantly periportal. Northern blotting showed that induction of these phase II drug-metabolizing enzymes by CMRN was accompanied by similar increases in the levels of their mRNAs. To assess the biological significance of enzyme induction by dietary CMRN, two intervention studies were performed in which the ability of the benzopyrone to inhibit either AFB1-initiated preneoplastic nodules (at 13 weeks) or AFB1-initiated liver tumors (at 50 weeks) was investigated. Animals pretreated with CMRN for 2 weeks prior to administration of AFB1, and with continued treatment during exposure to the carcinogen for a further 11 weeks, were protected completely from development of hepatic preneoplastic lesions by 13 weeks. In the longer-term dietary intervention, treatment with CMRN before and during exposure to AFB1 for a total of 24 weeks was found to significantly inhibit the number and size of tumors that subsequently developed by 50 weeks. These data suggest that consumption of a CMRN-containing diet provides substantial protection against the initiation of AFB1 hepatocarcinogenesis in the rat.


Asunto(s)
Aflatoxina B1/toxicidad , Aldehído Reductasa/biosíntesis , Cumarinas/farmacología , Glutatión Transferasa/biosíntesis , Neoplasias Hepáticas Experimentales/prevención & control , Quinona Reductasas/biosíntesis , Animales , Inducción Enzimática/efectos de los fármacos , Femenino , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/inducido químicamente , Masculino , Lesiones Precancerosas/prevención & control , Conejos , Ratas , Ratas Endogámicas F344 , Factores Sexuales
6.
Cancer Lett ; 137(2): 173-81, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10374839

RESUMEN

Ingestion of aflatoxin B1 is implicated in the high incidence of human liver cancers in several developing countries. An association has been detected between human exposure to aflatoxins, and mutations in the third base of codon 249 of the p53 gene in hepatomas. In vitro experiments using human cell line cells and aflatoxin B1 have demonstrated the induction of p53 mutations in codon 249 and adjacent codons. It was therefore of interest to see if this correlation between the in vivo and in vitro situations held for other species. The present study examined a rat liver-derived cell line, transformed in vitro with aflatoxin B1, for the presence of mutations associated with in vivo aflatoxin-induced hepatocarcinogenesis. In an in vivo rodent model systems using the aflatoxin B1-sensitive male F344 rat, previous studies have shown that hepatocarcinogenesis is accompanied by significant incidences of codon 12 mutations in K-ras and codon 13 mutations in N-ras genes, but in contrast to the human, apparently not by mutations in codon 243 of the p53 gene (which corresponds to codon 249 in the human gene). In contrast to the situation in humans, mutation in the third base of codon 243 in the rat would not result in any changes in amino acid sequence, but mutations in codon 250, as seen in in vitro human systems, would be expressed in the rat p53 protein. In the present study, an immortalised, non-transformed liver epithelial cell line derived from a male F344 rat was transformed in vitro by aflatoxin B1 as demonstrated by tumour formation in nude mice. The transformation was dependent on metabolic activation of the aflatoxin B1. Transfection of DNA, extracted from these tumours, into NIH 3T3 fibroblasts conferred a stable, malignant transforming capacity. However, no mutations in codon 12 of the K-ras or codon 13 of the N-ras genes were detected in any of these tumours. These results indicate that in vitro transformation does not necessarily involve the same mutations, as those observed in vivo. Also, no mutations in codon 243 or adjacent codons of the p53 gene, paralleling those observed in the human cell line treated with aflatoxin B1, were detected. The results serve to emphasise the in vivo and in vitro variation in the oncogene activation in the same target organ or cell lines derived from that organ, even when using a single carcinogen activated by a known metabolic pathway.


Asunto(s)
Aflatoxina B1/toxicidad , Transformación Celular Neoplásica/genética , Hígado/efectos de los fármacos , Mutágenos/toxicidad , Mutación/efectos de los fármacos , Células 3T3 , Animales , Southern Blotting , Pruebas de Carcinogenicidad , Línea Celular , Transformación Celular Neoplásica/inducido químicamente , Análisis Mutacional de ADN , Células Epiteliales/efectos de los fármacos , Genes p53/efectos de los fármacos , Genes p53/genética , Genes ras/efectos de los fármacos , Genes ras/genética , Hígado/química , Masculino , Ratones , Ratones Desnudos , Ratas , Ratas Endogámicas F344 , Análisis de Secuencia de ADN , Transfección
7.
Biochem Soc Symp ; 64: 141-68, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10207627

RESUMEN

Cancer chemopreventive agents can act by inhibiting either the acquisition of mutations or the neoplastic processes that occur subsequent to mutagenesis. Compounds that reduce the rate at which mutations arise, referred to as blocking agents, exert their effects largely through their ability to induce the expression of antioxidant and detoxification proteins. This is achieved by the transcriptional activation of a small number of genes that are co-regulated through the presence of an antioxidant responsive element (ARE) in their promoters. Blocking agents can cause gene induction by producing oxidative and/or chemical stress within the cell and, as the inducible proteins act to ameliorate the metabolic insult, the process represents a form of adaptive response. The transcription factors which mediate this response through the ARE are members of the basic leucine zipper superfamily. The mechanism whereby cells sense and respond to the chemical signal(s) generated by chemopreventive blocking agents is discussed.


Asunto(s)
Anticarcinógenos/farmacología , Antioxidantes/metabolismo , Estrés Oxidativo , Adaptación Fisiológica , Secuencia de Aminoácidos , Animales , Inducción Enzimática , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Inactivación Metabólica/genética , Datos de Secuencia Molecular , Transducción de Señal , Activación Transcripcional
8.
Biochem J ; 339 ( Pt 1): 95-101, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10085232

RESUMEN

We have used homology modelling, based on the crystal structure of the human glutathione S-transferase (GST) A1-1, to obtain the three-dimensional structures of rat GSTA3 and rat GSTA5 subunits bound to S-aflatoxinyl-glutathione. The resulting models highlight two residues, at positions 208 and 108, that could be important for determining, either directly or indirectly, substrate specificity for aflatoxin-exo-8,9-epoxide among the Alpha-class GSTs. Residues at these positions were mutated in human GSTA1-1 (Met-208, Leu-108), rat GSTA3-3 (Glu-208, His-108) and rat GSTA5-5 (Asp-208, Tyr-108): in the active rat GSTA5-5 to those in the inactive GSTA1-1; and in the inactive human GSTA1-1 and rat GSTA3-3 to those in the active rat GSTA5-5. These studies show clearly that, in all three GSTs, an aspartate residue at position 208 is a prerequisite for high activity in aflatoxin-exo-8,9-epoxide conjugation, although this alone is not sufficient; other residues in the vicinity, particularly residues 103-112, are important, perhaps for the optimal orientation of the aflatoxin-exo-8,9-epoxide in the active site for catalysis to occur.


Asunto(s)
Aflatoxina B1/análogos & derivados , Glutatión Transferasa/metabolismo , Aflatoxina B1/metabolismo , Secuencia de Aminoácidos , Animales , Catálisis , Glutatión Transferasa/química , Glutatión Transferasa/genética , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Ratas , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
9.
Carcinogenesis ; 19(10): 1829-36, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9806166

RESUMEN

Indole-3-carbinol (I3C) was examined for its ability to inhibit aflatoxin B1 (AFB1)-induced hepatocarcinogenesis in male Fischer rats when administered either before or after the carcinogen. After 13 weeks, animals pretreated with I3C (0.5% in the diet) for 2 weeks prior to administration of AFB1 and with continuing treatment during exposure to the carcinogen were protected from development of preneoplastic lesions, as determined by the classical markers gamma-glutamyltranspeptidase (GGT) and glutathione S-transferase (GST) P. In animals receiving AFB1 for 6 weeks before treatment with I3C, there was no obvious protective effect at 13 weeks compared with animals receiving only AFB1. Using cytokeratin 18 expression as a marker, animals fed AFB1 alone had a small number of positive foci at 13 weeks. However, no cytokeratin-positive foci were visible in the majority of livers from either group receiving I3C in combination with AFB1 and after 43 weeks all animals in these groups were protected from liver tumour formation. These results suggest that expression of cytokeratin 18, a later phenotypic change in foci than induction of GST-P and GGT, correlates more closely with tumour outcome in this model. I3C appeared to retard progression of AFB1-induced carcinogenesis at both the initiation and promotion stages. Continuous treatment with I3C for 13 weeks caused significant induction of CYP1A1, 1A2, 3A and 2B1/2, GST Yc2, aflatoxin B1 aldehyde reductase and quinone reductase. Such alteration of the drug metabolizing capacity of the liver by I3C contributes to blocking of initiation, while the observed inhibition of ornithine decarboxylase, a rate limiting enzyme in polyamine biosynthesis, and of tyrosine kinase activity may contribute to the suppressive effect of I3C.


Asunto(s)
Aflatoxina B1/toxicidad , Anticarcinógenos/farmacología , Carcinógenos/toxicidad , Indoles/farmacología , Neoplasias Hepáticas Experimentales/prevención & control , Aflatoxina B1/antagonistas & inhibidores , Aldehído Reductasa/metabolismo , Animales , Biomarcadores de Tumor , Sistema Enzimático del Citocromo P-450/metabolismo , Glutatión Transferasa/metabolismo , Queratinas/metabolismo , Neoplasias Hepáticas Experimentales/inducido químicamente , Masculino , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/prevención & control , Ratas , Ratas Endogámicas F344 , gamma-Glutamiltransferasa/metabolismo
10.
Biochem J ; 335 ( Pt 3): 619-30, 1998 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9794803

RESUMEN

A characteristic feature of the class Theta glutathione S-transferase (GST) T1-1 is its ability to activate dichloromethane and dibromoethane by catalysing the formation of mutagenic conjugates. The level of the GSTT1 subunit within tissues is an important determinant of susceptibility to the carcinogenic effects of these dihaloalkanes. In the present study it is demonstrated that hepatic GST activity towards these compounds can be elevated significantly in female and male Fischer-344 rats by feeding these animals on diets supplemented with cancer chemopreventive agents. Immunoblotting experiments showed that increased activity towards the dihaloalkanes is associated with elevated levels of the GSTT1 subunit in rat liver. Sex-specific effects were observed in the induction of GSTT1 protein. Amongst the chemopreventive agents tested, indole-3-carbinol proved to be the most potent inducer of hepatic GSTT1 in male rats (6.2-fold), whereas coumarin was the most potent inducer of this subunit in the livers of female rats (3. 5-fold). Phenobarbital showed significant induction of GSTT1 only in male rat liver and had little effect in female rat liver. Western blotting showed that class Alpha, Mu and Pi GST subunits are not co-ordinately induced with GSTT1, indicating that the expression of GSTT1 is determined, at least in part, by mechanisms distinct from those that regulate levels of other transferases. The increase in amount of hepatic GSTT1 protein was also reflected by an increase in the steady-state level of mRNA in response to treatment with chemopreventive agents and model inducers. Immunohistochemical detection of GSTT1 in rat liver supported the Western blotting data, but showed, in addition to cytoplasmic staining, significant nuclear localization of the enzyme in hepatocytes from some treated animals, including those fed on an oltipraz-containing diet. Significantly, the hepatic level of cytochrome P-450 2E1, an enzyme which offers a detoxification pathway for dihaloalkanes, was unchanged by the various inducing agents studied. It is concluded that the induction of GSTT1 by dietary components and its localization within cells are important factors that should be considered when assessing the risk dihaloalkanes pose to human health.


Asunto(s)
Anticarcinógenos/farmacología , Glutatión Transferasa/biosíntesis , Hidrocarburos Bromados/farmacocinética , Isoenzimas/biosíntesis , Hígado/enzimología , Cloruro de Metileno/farmacocinética , Xenobióticos/farmacocinética , Animales , Anticarcinógenos/administración & dosificación , Biotransformación , Carcinógenos/farmacocinética , Carcinógenos/toxicidad , Suplementos Dietéticos , Inducción Enzimática , Femenino , Humanos , Hidrocarburos Bromados/toxicidad , Hígado/efectos de los fármacos , Masculino , Cloruro de Metileno/toxicidad , Fenobarbital/farmacología , Ratas , Ratas Endogámicas F344 , Caracteres Sexuales , Xenobióticos/toxicidad
11.
Toxicol Appl Pharmacol ; 151(1): 152-8, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9705898

RESUMEN

Aflatoxin M1 (AFM1) is the principal hydroxylated aflatoxin metabolite present in the milk of dairy cows fed a diet contaminated with aflatoxin B1, (AFB1) and the metabolite is also present in the milk of human nursing mothers consuming foodstuffs containing the toxin. AFM1 is usually considered to be a detoxification product of AFB1 and this appears warranted if the biological endpoints involved are carcinogenicity and mutagenicity. However, it may not be a valid conclusion in the case of cytotoxicity. The metabolism of AFM1 and AFB1 have been studied in vitro using human liver microsomes. Formation of primary metabolites associated with metabolic activation to the respective epoxides reflected the differences between the carcinogenic potentials of the two toxins and, similar to AFB1, the conjugation of AFM1 epoxide with reduced GSH was catalyzed by mouse, but not human liver cytosol. Although the majority of the binding of [3H]AFB1 to microsomal protein was dependent on metabolic activation, a high level of retention of [3H]AFM1 by microsomes, nonextractable in methanol and unrelated to metabolic activation, was observed. It appears possible that this property is related to the high cytotoxicity of AFM1. Experiments using human cell line cells either expressing or not expressing human cytochrome P450 enzymes in assays of acute toxicity (MTT assays) have demonstrated a directly toxic potential of AFM1 in the absence of metabolic activation, in contrast to AFB1. Caution therefore needs to be exercised in designating the formation of AFM1 as essentially detoxification when considering a biological response in which cytotoxicity may play a significant role, e.g., immunotoxicity.


Asunto(s)
Aflatoxina B1/toxicidad , Aflatoxina M1/toxicidad , Carcinógenos/toxicidad , Microsomas Hepáticos/efectos de los fármacos , Mutágenos/toxicidad , Aflatoxina B1/metabolismo , Aflatoxina M1/metabolismo , Biotransformación/efectos de los fármacos , Línea Celular , Sistema Enzimático del Citocromo P-450/biosíntesis , Citosol/efectos de los fármacos , Citosol/metabolismo , Citotoxicidad Inmunológica , Compuestos Epoxi/metabolismo , Glutatión/metabolismo , Humanos , Microsomas Hepáticos/metabolismo
12.
Chem Biol Interact ; 111-112: 51-67, 1998 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-9679543

RESUMEN

The rat can be protected against aflatoxin B1 (AFB1) hepatocarcinogenesis by being fed on a diet containing the synthetic antioxidant ethoxyquin. Evidence suggests that chemoprotection against AFB1 is due to increased detoxification of the mycotoxin by one or more inducible drug-metabolising enzymes. The glutathione S-transferase (GST) isoenzymes in rat liver that contribute to ethoxyquin-induced chemoprotection against AFB1 have been identified by protein purification. This approach resulted in the isolation of several heterodimeric class alpha GST, all of which contained the A5 subunit and possessed at least 50-fold greater activity towards AFB1-8,9-epoxide than previously studied transferases. Molecular cloning and heterologous expression of rat GSTA5-5 has led to the demonstration that it exhibits substantially greater activity for AFB1-8,9-epoxide than other rat transferases. The A5 homodimer can also catalyse the conjugation of glutathione with other epoxides, such as trans-stilbene oxide and 1,2-epoxy-3-(4'-nitrophenoxy)propane, and possesses high catalytic activity for the reactive aldehyde 4-hydroxynonenal. Western blotting has shown that the A5 subunit is not only induced by ethoxyquin but that it is also induced by other cancer chemopreventive agents, such as butylated hydroxyanisole, oltipraz, benzyl isothiocyanate, indole-3-carbinol and coumarin. In addition to GSTA5, we have identified a novel aflatoxin-aldehyde reductase (AFAR) that is similarly induced by ethoxyquin. However, immunoblotting has shown that GSTA5 and AFAR are not always co-ordinately regulated by chemoprotectors. In order to gain a better understanding of the mechanisms responsible for the induction of GSTA5 protein, the GSTA5 gene has been cloned. It was isolated on two overlapping bacteriophage lambda clones and found to be approximately 12 kb in length. The transcriptional start site of GSTA5 has been identified 228 bp upstream from the ATG translational initiation codon. Computer-assisted analysis of the upstream sequence has indicated the presence of a putative antioxidant responsive element (located between -421 and -429 bp) which may be responsible for the induction of GSTA5 by chemopreventive agents.


Asunto(s)
Aflatoxina B1/toxicidad , Etoxiquina/farmacología , Glutatión Transferasa/metabolismo , Aflatoxina B1/farmacocinética , Aldehído Reductasa/biosíntesis , Secuencia de Aminoácidos , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Secuencia de Bases , Biotransformación , Clonación Molecular , ADN/genética , Resistencia a Medicamentos , Inducción Enzimática/efectos de los fármacos , Glutatión Transferasa/química , Glutatión Transferasa/genética , Inactivación Metabólica , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/prevención & control , Datos de Secuencia Molecular , Estrés Oxidativo , Conformación Proteica , Ratas
14.
Biochem J ; 332 ( Pt 1): 21-34, 1998 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9576847

RESUMEN

The masking of charged amino or carboxy groups by N-phthalidylation and O-phthalidylation has been used to improve the absorption of many drugs, including ampicillin and 5-fluorouracil. Following absorption of such prodrugs, the phthalidyl group is hydrolysed to release 2-carboxybenzaldehyde (2-CBA) and the pharmaceutically active compound; in humans, 2-CBA is further metabolized to 2-hydroxymethylbenzoic acid by reduction of the aldehyde group. In the present work, the enzyme responsible for the reduction of 2-CBA in humans is identified as a homologue of rat aflatoxin B1-aldehyde reductase (rAFAR). This novel human aldo-keto reductase (AKR) has been cloned from a liver cDNA library, and together with the rat protein, establishes the AKR7 family of the AKR superfamily. Unlike its rat homologue, human AFAR (hAFAR) appears to be constitutively expressed in human liver, and is widely expressed in extrahepatic tissues. The deduced human and rat protein sequences share 78% identity and 87% similarity. Although the two AKR7 proteins are predicted to possess distinct secondary structural features which distinguish them from the prototypic AKR1 family of AKRs, the catalytic- and NADPH-binding residues appear to be conserved in both families. Certain of the predicted structural features of the AKR7 family members are shared with the AKR6 beta-subunits of voltage-gated K+-channels. In addition to reducing the dialdehydic form of aflatoxin B1-8,9-dihydrodiol, hAFAR shows high affinity for the gamma-aminobutyric acid metabolite succinic semialdehyde (SSA) which is structurally related to 2-CBA, suggesting that hAFAR could function as both a SSA reductase and a 2-CBA reductase in vivo. This hypothesis is supported in part by the finding that the major peak of 2-CBA reductase activity in human liver co-purifies with hAFAR protein.


Asunto(s)
Oxidorreductasas de Alcohol/química , Aldehído Reductasa/química , Hígado/enzimología , Aflatoxina B1/análogos & derivados , Aflatoxina B1/metabolismo , Aldo-Ceto Reductasas , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Humanos , Concentración de Iones de Hidrógeno , Inmunohistoquímica , Isoenzimas/aislamiento & purificación , Datos de Secuencia Molecular , Canales de Potasio/química , Estructura Secundaria de Proteína , ARN Mensajero/metabolismo , Ratas , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Ácido gamma-Aminobutírico/análogos & derivados , Ácido gamma-Aminobutírico/metabolismo
15.
Carcinogenesis ; 18(9): 1729-38, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9328168

RESUMEN

A range of potential chemoprotective agents, most of them natural dietary constituents, has been examined for ability to modulate both phase I (cytochrome P450 1A1, 1A2, 2B1/2, 2C11, 2E1, 3A, 4A) and phase II drug metabolizing enzymes (glutathione S-transferases, in particular subunits Yc2 and P, aflatoxin B1-aldehyde reductase and quinone reductase) in rat liver. In addition to assays of total enzyme activity and Western blots for individual isozymes, the ability of microsomes to metabolize aflatoxin B1, and of cytosols to conjugate aflatoxin B1 (AFB1)-epoxide to GSH and to produce AFB1-dialcohol, were measured. Induction of gamma-glutamyl transpeptidase activity was examined by histochemistry. Differing patterns of induction were observed, reflecting differences in the control of expression of the individual enzymes studied. Of the compounds examined, butylated hydroxytoluene, ethoxyquin, indole-3-carbinol and phenethyl isothiocyanate were the most potent bifunctional agents (inducing both phase I and II activities). Oltipraz, while only weakly inducing CYP1A2 and 2B1/2, was a potent inducer of phase II enzymes. Caffeic acid, garlic oil, sinigrin and propyl gallate all showed some ability to induce phase II enzymes. 4-Methyl catechol, alpha-tocopherol and red wine decreased certain phase I enzyme activities, while inducing total GST activity. Butylated hydroxytoluene, ethoxyquin, garlic oil and indole-3-carbinol induced gamma glutamyltranspeptidase in periportal hepatocytes. Particularly because of their ability to induce the detoxifying activities of glutathione S-transferase Yc2 and aldehyde reductase, butylated hydroxytoluene, ethoxyquin, indole-3-carbinol, oltipraz, phenethyl isothiocyanate and sinigrin will be effective blocking agents in rodents, if administered prior to AFB1. While these studies indicate the relative contributions of phase I and II metabolism in the overall protective effect in rat, care should be taken that a similar balance is achieved in man, and that relevant enzymes or iso forms are induced.


Asunto(s)
Aflatoxina B1/metabolismo , Anticarcinógenos/farmacología , Dieta , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Animales , Anticarcinógenos/administración & dosificación , Peso Corporal , Sistema Enzimático del Citocromo P-450/metabolismo , Relación Dosis-Respuesta a Droga , Inducción Enzimática , Inmunohistoquímica , Masculino , Tamaño de los Órganos , Sustancias Protectoras/farmacología , Ratas , Ratas Endogámicas F344
16.
Cancer Res ; 57(19): 4257-66, 1997 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9331086

RESUMEN

Fischer 344 rats fed on a diet that is deficient in selenium are more resistant to the hepatocarcinogen aflatoxin B1 (AFB1) than those fed on a selenium-sufficient diet. Hepatic cytosol from either selenium-deficient Fischer 344 rats or Hooded Lister rats possesses a marked increase in both reductase activity toward AFB1-dialdehyde and glutathione S-transferase (GST) activity toward AFB(1)-8,9-epoxide than hepatic cytosol from selenium-sufficient rats. The elevation in hepatic AFB1-aldehyde reductase (AFAR) activity in selenium-deficient animals is accompanied by an increase of 11- and 15-fold in the levels of AFAR protein in liver cytosol from Fischer 344 and Hooded Lister rats, respectively. The amount of AFAR protein in selenium-sufficient and -deficient Fischer rats was modulated by treatment with N-acetylcysteine; this antioxidant reduced basal expression of AFAR but did not modulate the relative overexpression of AFAR during selenium deficiency. The enhanced capacity to conjugate glutathione with AFB(1)-8,9-epoxide in selenium-deficient livers from Fischer 344 and Hooded Lister rats is associated with a 5- and 7-fold increase, respectively, in the hepatic levels of the AFB1-metabolizing alpha-class GSTA5 subunit. The elevated levels of AFAR and GSTA5 protein in the selenium-deficient animals coincided with increases in the steady-state levels of their mRNAs. In selenium-deficient Fischer 344 rats, AFAR and GSTA5 were both found to be expressed throughout the centrilobular and midzonal areas of the liver lobule but were essentially absent from periportal hepatocytes. The effect of selenium insufficiency is pleiotropic, and it was also noted that the theta-class GSTT1 is overexpressed 3- and 10-fold in livers of selenium-deficient Hooded Lister and Fischer 344 rats. Inasmuch as GSTT1 is responsible for the metabolic activation of dihaloalkanes, selenium deficiency may increase the susceptibility of rats to mutagens such as dichloromethane.


Asunto(s)
Aflatoxina B1/toxicidad , Aldehído Reductasa/metabolismo , Glutatión Transferasa/metabolismo , Neoplasias Hepáticas Experimentales/prevención & control , Hígado/enzimología , Selenio/deficiencia , Acetilcisteína/farmacología , Aldehído Reductasa/genética , Animales , Antioxidantes/farmacología , Citosol/enzimología , Aductos de ADN , Daño del ADN , Glutatión Transferasa/genética , Inactivación Metabólica , Hígado/efectos de los fármacos , Masculino , Estrés Oxidativo , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas F344 , Selenio/fisiología
17.
Carcinogenesis ; 18(5): 905-10, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9163674

RESUMEN

Activation of ras proto-oncogenes occurs frequently in vivo in chemically induced rodent tumours, including rat hepatomas induced by aflatoxin B1. This study examines the in vitro activation of a human ras gene by this mycotoxin. A plasmid containing the human Ha-ras proto-oncogene, together with a neomycin resistance gene (pECneo), was incubated in vitro with a microsomal system generating aflatoxin B1 8,9-epoxide. Subsequent transfection of the plasmid into mouse NIH 3T3 fibroblasts, followed by G418 selection and s.c. injection of surviving cells into immunodeficient mice demonstrated that the proto-oncogene had acquired transforming capacity. Although a single tumour resulted from similar treatment of incubated unconjugated plasmid, no tumours were produced by a secondary round of transfections using DNA from this tumour. Selective PCR amplification of the human Ha-ras gene in extracted tumour DNA followed by sequencing demonstrated the presence of G-->T transversions either at the first or middle base of codon 12 in tumours resulting from transfection with the aflatoxin-B1-modified pECneo plasmid, but this was not detected in the single tumour resulting from transfection with the unmodified plasmid. Thus, although a mutation in the Ha-ras gene has not been reported for human primary hepatomas occurring in aflatoxin-exposed populations, metabolically activated aflatoxin B1 is capable of mutating this proto-oncogene to its oncogenic form in vitro. No mutations were observed in codon 61. It appears that, in contrast to the frequently reported G-->T transversions in codon 249 of the p53 gene in primary hepatomas in aflatoxin-exposed humans, the failure to detect Ha-ras mutations in these tumours is not due to an inability of aflatoxin B1 to activate this proto-oncogene. The G-->T transversions observed in this study contrast with the most frequent aflatoxin B1 in vivo induced mutations, G-->A transitions in the rat Ki-ras gene. Possible mechanisms for these differences are discussed.


Asunto(s)
Aflatoxina B1/farmacología , Genes ras , Mutagénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/genética , Células 3T3 , Animales , Humanos , Ratones , Ratones Desnudos , Mutación Puntual , Proto-Oncogenes Mas , Proto-Oncogenes , Transfección
18.
Cancer Res ; 56(12): 2758-66, 1996 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8665510

RESUMEN

Feeding rats on diets containing the synthetic antioxidants ethoxyquin, butylated hydroxyanisole, and oltipraz results in 15-, 9-, and 6-fold increases, respectively, in the hepatic levels of aflatoxin B1-dialdehyde reductase (AFAR) protein. By contrast, treatment of rats with either of the inducing agents phenobarbital or 3-methylcholanthrene results in an approximate increase of only 1.4-fold in the amount of AFAR in rat liver. Northern blotting has shown that these increases in levels of hepatic AFAR protein are accompanied by corresponding increases in AFAR mRNA. Immunodepletion of AFAR from rat liver extracts has revealed that AFAR makes a considerable contribution to carbonyl metabolism in livers from animals treated with synthetic antioxidants and that it is the major reductase that can utilize aflatoxin B1-dialdehyde as a substrate. The immunodepletion experiments also revealed the presence of at least one other inducible carbonyl-reducing enzyme that, like AFAR, can metabolize 9,10-phenanthraquinone. Carbonyl-reducing activity from rat liver has been resolved into six enzyme-containing peaks by anion-exchange chromatography on Q-Sepharose. This method has been used to show that, in addition to AFAR, two other rat liver carbonyl-reducing enzymes are induced by ethoxyquin, and that these are distinct from NAD(P)H: quinone oxidoreductase. Collectively, these data show that synthetic antioxidants can influence substantially the capacity of rat liver to metabolize reactive carbonyl-containing compounds.


Asunto(s)
Aldehído Reductasa/metabolismo , Antioxidantes/farmacología , Hígado/enzimología , Aldehído Reductasa/fisiología , Animales , Barbitúricos/farmacología , Hidroxianisol Butilado/farmacología , Carcinógenos/farmacología , Inducción Enzimática/efectos de los fármacos , Etoxiquina/farmacología , Metilcolantreno/farmacología , Oxidación-Reducción , Fenobarbital/farmacología , Pirazinas/farmacología , Ratas , Tionas , Tiofenos
19.
IARC Sci Publ ; (139): 165-73, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8923029

RESUMEN

The cytochrome P450-dependent mono-oxygenase system has evolved as one of our primary defences against toxic chemicals present in our environment. This multi-enzyme system functions as an adaptive response to environmental challenge in that exposure to specific toxic agents induces the expression of cytochrome P450 isozymes active in their metabolism. In most cases, such metabolism leads to an increased rate of chemical detoxification, but in certain cases it can also lead to an increased rate of chemical activation to toxic products. The induction of cytochrome P450s leading to cytoprotection is a major mechanism of chemoprotection, and it is well documented that this pathway prevents a large number of toxic reactions. Incredibly, despite the importance of this metabolic pathway, the effects of a wide range of chemoprotective agents with different mechanisms of action on the expression of specific cytochrome P450 isozymes remains poorly understood. In this chapter, we discess the ability of different chemical compounds to modulate cytochrome P450 both at the transcriptional and post-transcriptional levels, in a way which affects its ability to metabolize or detoxify chemical carcinogens.


Asunto(s)
Anticarcinógenos/farmacología , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Isoenzimas/efectos de los fármacos , Animales , Carcinógenos/farmacocinética , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Inducción Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Inactivación Metabólica , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Neoplasias/enzimología , Neoplasias/prevención & control
20.
IARC Sci Publ ; (139): 175-87, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8923030

RESUMEN

A number of xenobiotics, including the synthetic antioxidant ethoxyquin, inhibit aflatoxin B1 (AFB1)-induced hepatocarcinogenesis in the rat. Two detoxification enzymes that mediate ethoxyquin-induced chemoprotection against AFB1 have been identified by protein purification: a glutathione S-transferase (GST) Yc2 subunit with at least 100-fold greater activity towards AFB1-8,9-epoxide than previously studied transferases, and a unique aldehyde reductase with activity towards the dialdehydic form of AFB1-8,9-dihydrodiol. Molecular cloning has revealed that the Yc2 subunit is a class alpha GST and that the aflatoxin-metabolizing aldehyde reductase (AFAR) is a distant member of the aldo-keto reductase superfamily. Enzyme assay and western blotting have shown that many chemoprotectors, such as ethoxyquin, butylated hydroxyanisole, butylated hydroxytoluene, oltipraz and indole-3-carbinol, that inhibit AFB1-mediated hepatocarcinogenesis induce both GST Yc2 and AFAR. However, western blotting suggests that these enzymes are not always coordinately regulated, as treatment with phenobarbital and beta-naphthoflavone results in differences in the relative increase in hepatic GST Yc2 and AFAR. These findings indicate that GST Yc2 and AFAR represent important resistance mechanisms against AFB1 in the rat. This conclusion is supported by the observation that GST Yc2 and AFAR are overexpressed in rat liver preneoplastic nodules, which display pleiotropic drug resistance.


Asunto(s)
Aflatoxina B1/toxicidad , Aldehído Reductasa/efectos de los fármacos , Anticarcinógenos/farmacología , Carcinógenos/toxicidad , Glutatión Transferasa/efectos de los fármacos , Aflatoxina B1/farmacocinética , Aldehído Reductasa/metabolismo , Animales , Biotransformación , Carcinógenos/farmacocinética , Resistencia a Medicamentos , Etoxiquina/farmacología , Glutatión Transferasa/metabolismo , Humanos , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/prevención & control , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...