Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicology ; 299(2-3): 69-79, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22640941

ABSTRACT

Diet and its various components are consistently identified as among the most important 'risk factors' for cancer worldwide, yet great uncertainty remains regarding the relative contribution of nutritive (e.g., vitamins, calories) vs. non-nutritive (e.g., phytochemicals, fiber, contaminants) factors in both cancer induction and cancer prevention. Among the most potent known human dietary carcinogens is the mycotoxin, aflatoxin B(1) (AFB). AFB and related aflatoxins are produced as secondary metabolites by the molds Aspergillus flavus and Aspergillus parasiticus that commonly infect poorly stored foods including peanuts, pistachios, corn, and rice. AFB is a potent hepatocarcinogenic agent in numerous animal species, and has been implicated in the etiology of human hepatocellular carcinoma. Recent research has shown that many diet-derived factors have great potential to influence AFB biotransformation, and some efficiently protect from AFB-induced genotoxicity. One key mode of action for reducing AFB-induced carcinogenesis in experimental animals was shown to be the induction of detoxification enzymes such as certain glutathione-S-transferases that are regulated through the Keap1-Nrf2-ARE signaling pathway. Although initial studies utilized the dithiolthione drug, oltipraz, as a prototypical inducer of antioxidant response, dietary components such as suforaphane (SFN) are also effective inducers of this pathway in rodent models. However, human GSTs in general do not appear to be extensively induced by SFN, and GSTM1 - the only human GST with measurable catalytic activity toward aflatoxin B(1)-8,9-epoxide (AFBO; the genotoxic metabolite of AFB), does not appear to be induced by SFN, at least in human hepatocytes, even though its expression in human liver cells does appear to offer considerable protection against AFB-DNA damage. Although induction of detoxification pathways has served as the primary mechanistic focus of chemoprevention studies, protective effects of chemoprotective dietary components may also arise through a decrease in the rate of activation of AFB to AFBO. Dietary consumption of apiaceous vegetables inhibits CYP1A2 activity in humans, and it has been demonstrated that some compounds in those vegetables act as potent inhibitors of human CYP1A2 and cause reduced hCYP1A2-mediated mutagenicity of AFB. Other dietary compounds of different origin (e.g., constituents of brassica vegetables and hops) have been shown to modify expression of human hepatic enzymes involved in the oxidation of AFB. SFN has been shown to protect animals from AFB-induced tumors, to reduce AFB biomarkers in humans in vivo and to reduce efficiently AFB adduct formation in human hepatocytes, although it appears that this protective effect is the result of repression of human hepatic CYP3A4 expression, rather than induction of protective GSTs, at least in human hepatocytes. If this mechanism were to occur in vivo in humans, it would raise safety concerns for the use of SFN as a chemoprotective agent as it may have important implications for drug-drug interactions in humans. A dietary chemoprevention pathway that is independent of AFB biotransformation is represented by the potential for dietary components, such as chlorophyllin, to tightly bind to and reduce the bioavailability of aflatoxins. Chlorophyllin has been shown to significantly reduce genotoxic AFB biomarkers in humans, and it therefore holds promise as a practical means of reducing the incidence of AFB-induced liver cancer. Recent reports have demonstrated that DNA repair mechanisms are inducible in mammalian systems and some diet-derived compounds elevated significantly the gene expression of enzymes potentially involved in nucleotide excision repair of AFB-DNA adducts. However, these are initial observations and more research is needed to determine if dietary modulation of DNA repair is a safe and effective approach to chemoprevention of AFB-induced liver cancer.


Subject(s)
Aflatoxin B1/pharmacokinetics , Aflatoxin B1/poisoning , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Aflatoxin B1/toxicity , Animals , Brassica , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Humans , Inactivation, Metabolic , Liver Neoplasms/enzymology , Mutagens/metabolism , Mutagens/toxicity
2.
Toxicol Sci ; 116(2): 422-32, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20442190

ABSTRACT

Primary cultures of human hepatocytes were used to investigate whether the dietary isothiocyanates, sulforaphane (SFN), and phenethyl isothiocyanate (PEITC) can reduce DNA adduct formation of the hepatocarcinogen aflatoxin B(1) (AFB). Following 48 h of pretreatment, 10 and 50 microM SFN greatly decreased AFB-DNA adduct levels, whereas 25muM PEITC decreased AFB-DNA adducts in some but not all hepatocyte preparations. Microarray and quantitative reverse transcriptase (RT)-PCR analyses of gene expression in SFN and PEITC-treated hepatocytes demonstrated that SFN greatly decreased cytochrome P450 (CYP) 3A4 mRNA but did not induce the expression of either glutathione S-transferase (GST) M1 or GSTT1. The protective effects of SFN required pretreatment; cotreatment of hepatocytes with SFN and AFB in the absence of pretreatment had no effect on AFB-DNA adduct formation. When AFB-DNA adduct formation was evaluated by GST genotype, the presence of one or two functional alleles of GSTM1 was associated with a 75% reduction in AFB-DNA adducts, compared with GSTM1 null. In conclusion, these results demonstrate that the inhibition of AFB-DNA adduct formation by SFN is dependent on changes in gene expression rather than direct inhibition of catalytic activity. Transcriptional repression of genes involved in AFB bioactivation (CYP3A4 and CYP1A2), but not transcriptional activation of GSTs, may be responsible for the protective effects of SFN. Although GSTM1 expression was not induced by SFN, the presence of a functional GSTM1 allele can afford substantial protection against AFB-DNA damage in human liver. The downregulation of CYP3A4 by SFN may have important implications for drug interactions.


Subject(s)
Aflatoxin B1/toxicity , Cytochrome P-450 CYP3A/genetics , Glutathione Transferase/genetics , Hepatocytes/drug effects , Isothiocyanates/pharmacology , Thiocyanates/pharmacology , Aflatoxin B1/pharmacokinetics , Cells, Cultured , DNA Adducts/analysis , DNA Repair , Genotype , Hepatocytes/metabolism , Humans , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sulfoxides
3.
Toxicol Sci ; 112(2): 303-10, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19770484

ABSTRACT

This study employed cultured human primary hepatocytes to investigate the ability of the putative chemopreventive phytochemicals curcumin (CUR), 3,3'-diindolylmethane (DIM), isoxanthohumol (IXN), or 8-prenylnaringenin (8PN) to reduce DNA adduct formation of the hepatocarcinogen aflatoxin B1 (AFB). Following 48 h of pretreatment, DIM and 8PN significantly increased AFB-DNA adduct levels, whereas CUR and IXN had no effect. DIM greatly enhanced the transcriptional expression of cytochrome P450 (CYP) 1A1 and CYP1A2 mRNA. Glutathione S-transferase mRNAs were not increased by any of the treatments. In vitro enzyme activity assays demonstrated that 8PN and DIM, but not CUR or IXN, inhibited human CYP1A1, CYP1A2, and CYP3A4 activities. To distinguish between treatment effects on transcription versus direct effects on enzyme activity for DIM, we evaluated the effects of pretreatment alone (transcriptional activation) versus cotreatment alone (enzyme inhibition). The results demonstrated that effects on gene expression, but not catalytic activity, are responsible for the observed effects of DIM on AFB-DNA adduct formation. The increase in AFB-DNA damage following DIM treatment may be explained through its substantial induction of CYP1A2 and/or its downregulation of GSTM1, both of which were significant. The increase in DNA damage by DIM raises potential safety risks for dietary supplements of DIM and its precursor indole-3-carbinol.


Subject(s)
Aflatoxin B1/toxicity , Curcumin/pharmacology , Flavonoids/pharmacology , Indoles/pharmacology , Mutagens/toxicity , Propiophenones/pharmacology , Aflatoxin B1/metabolism , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , DNA Adducts/metabolism , Gene Expression Regulation, Enzymologic , Humans , Polymerase Chain Reaction , RNA, Messenger/genetics
4.
Food Chem Toxicol ; 44(9): 1474-84, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16762476

ABSTRACT

In humans, apiaceous vegetables (carrots, parsnips, celery, parsley, etc.) inhibit cytochrome P-450 1A2, a biotransformation enzyme known to activate several procarcinogens, including aflatoxin B1 (AFB). We evaluated eight phytochemicals from apiaceous vegetables for effects on human cytochrome P-450 1A2 (hCYP1A2) activity using a methoxyresorufin O-demethylase (MROD) assay and a trp-recombination assay. Saccharomyces cerevisiae was used for heterologous CYP1A2 expression and this yeast strain is also diploid and auxotrophic for tryptophan due to mutations in the trp5 alleles. When these two alleles undergo AFB-induced mitotic recombination, gene conversion occurs, allowing yeast to grow in the absence of tryptophan. The apiaceous constituents psoralen, 5-methoxypsoralen (5-MOP), 8-methoxypsoralen (8-MOP), and apigenin were potent inhibitors of hCYP1A2-mediated MROD activity in yeast microsomes, whereas quercetin was a modest hCYP1A2 inhibitor. Naringenin, caffeic acid, and chlorogenic acid did not inhibit hCYP1A2-mediated MROD activity. The 2-h pretreatment of intact yeast cells with psoralen, 5-MOP, and 8-MOP significantly improved cell survival after subsequent 4-h AFB treatment and reduced hCYP1A2-mediated mutagenicity of AFB. Apigenin also significantly decreased mutagenicity. These results suggest that in vivo CYP1A2 inhibition by apiaceous vegetables may be due to the phytochemicals present and imply that apiaceous vegetable intake may be chemopreventive by inhibiting CYP1A2-mediated carcinogen activation.


Subject(s)
Aflatoxin B1/pharmacokinetics , Apiaceae , Cytochrome P-450 CYP1A2 Inhibitors , Enzyme Inhibitors/pharmacology , Mutagens/pharmacokinetics , Plant Extracts/pharmacology , Poisons/pharmacokinetics , Aflatoxin B1/toxicity , Apiaceae/chemistry , Biotransformation , Cell Survival/drug effects , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemistry , Flavonoids/pharmacology , Furocoumarins/pharmacology , Humans , Inhibitory Concentration 50 , Microsomes/drug effects , Microsomes/enzymology , Mutagens/toxicity , Oxidoreductases/metabolism , Plant Extracts/chemistry , Poisons/toxicity , Recombination, Genetic , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics
5.
J Agric Food Chem ; 50(4): 938-45, 2002 Feb 13.
Article in English | MEDLINE | ID: mdl-11829671

ABSTRACT

It is still unclear whether the carcinogenic mycotoxin ochratoxin A (OTA) is bioactivated to DNA-binding metabolites in rodents and humans. Therefore, we have incubated cultured rat and human primary hepatocytes with noncytotoxic concentrations of (3)H-OTA ranging from 10(-7) to 10(-5) M for 8 h and determined its metabolism and covalent DNA binding. In rat hepatocytes, OTA was metabolized to small amounts of three products, which were further studied by electrospray ionization (ESI)-MS/MS techniques. In addition to 4-hydroxy-OTA, which is a known product of OTA biotransformation, two novel metabolites were detected and tentatively identified as hexose and pentose conjugates of OTA. The in vitro induction with 3-methylcholanthrene (3MC) increased the formation of 4-hydroxy-OTA but did not alter the formation of the conjugated metabolites. No covalent binding of (3)H-OTA or its metabolites to DNA was observed in rat hepatocytes with or without 3MC induction with a limit of detection of 2 adducts per 10(9) nucleotides. However, the cellular ratio of reduced glutathione to oxidized glutathione was significantly decreased by treatment with OTA. In cultured human hepatocytes, (3)H-OTA was only very poorly metabolized, and no covalent DNA binding was observed. In conclusion, the results of this in vitro study do not support the notion that OTA has the potential to undergo metabolic activation and form covalent DNA adducts in rodents and humans.


Subject(s)
DNA/metabolism , Hepatocytes/metabolism , Ochratoxins/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Glutathione/analysis , Glutathione/metabolism , Humans , Male , Methylcholanthrene/pharmacology , Ochratoxins/administration & dosage , Ochratoxins/analysis , Ochratoxins/pharmacology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...