Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Chem Res Toxicol ; 29(4): 659-68, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-26952143

ABSTRACT

Estragole is a known hepatocarcinogen in rodents at high doses following metabolic conversion to the DNA-reactive metabolite 1'-sulfooxyestragole. The aim of the present study was to model possible levels of DNA adduct formation in (individual) humans upon exposure to estragole. This was done by extending a previously defined PBK model for estragole in humans to include (i) new data on interindividual variation in the kinetics for the major PBK model parameters influencing the formation of 1'-sulfooxyestragole, (ii) an equation describing the relationship between 1'-sulfooxyestragole and DNA adduct formation, (iii) Monte Carlo modeling to simulate interindividual human variation in DNA adduct formation in the population, and (iv) a comparison of the predictions made to human data on DNA adduct formation for the related alkenylbenzene methyleugenol. Adequate model predictions could be made, with the predicted DNA adduct levels at the estimated daily intake of estragole of 0.01 mg/kg bw ranging between 1.6 and 8.8 adducts in 10(8) nucleotides (nts) (50th and 99th percentiles, respectively). This is somewhat lower than values reported in the literature for the related alkenylbenzene methyleugenol in surgical human liver samples. The predicted levels seem to be below DNA adduct levels that are linked with tumor formation by alkenylbenzenes in rodents, which were estimated to amount to 188-500 adducts per 10(8) nts at the BMD10 values of estragole and methyleugenol. Although this does not seem to point to a significant health concern for human dietary exposure, drawing firm conclusions may have to await further validation of the model's predictions.


Subject(s)
Anisoles/metabolism , Carcinogens/metabolism , DNA Adducts/metabolism , Liver/metabolism , Sulfones/metabolism , Adolescent , Adult , Aged , Allylbenzene Derivatives , Child, Preschool , Computer Simulation , Female , Humans , Infant , Kinetics , Liver/drug effects , Male , Middle Aged , Models, Biological , Monte Carlo Method , NAD/metabolism , Oxidation-Reduction , Young Adult
2.
J Agric Food Chem ; 64(6): 1256-63, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26808477

ABSTRACT

To study the effect of metabolic conjugation of flavonoids on the potential to inhibit protein kinase activity, the inhibitory effects of the dietary flavonol kaempferol and its major plasma conjugate kaempferol-3-O-glucuronide on protein kinases were studied. To this end, the inhibition of the phosphorylation activity of recombinant protein kinase A (PKA) and of cell lysate from the hepatocellular carcinoma cell line HepG2 on 141 putative serine/threonine phosphorylation sites derived from human proteins was assessed. Glucuronidation reduced the inhibitory potency of kaempferol on the phosphorylation activity of PKA and HepG2 lysate on average about 16 and 3.5 times, respectively, but did not appear to affect the target selectivity for kinases present in the lysate. The data demonstrate that, upon glucuronidation, kaempferol retains part of its intrinsic kinase inhibition potential, which implies that K3G does not necessarily need to be deconjugated to the aglycone for a potential inhibitory effect on protein kinases.


Subject(s)
Enzyme Inhibitors/chemistry , Kaempferols/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Cell Line , Glucuronic Acid/chemistry , Humans , Kinetics , Molecular Structure , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
3.
J Steroid Biochem Mol Biol ; 154: 245-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26361015

ABSTRACT

Non-prenylated isoflavone aglycones are known to have phyto-estrogenic properties and act as agonistic ligands on ERα and ERß due to their structural resemblance to 17ß-estradiol (E2). Genistein and daidzein are the two main dietary isoflavones; upon uptake they are extensively metabolized and exist nearly exclusively as their conjugated forms in biological fluids. Little is known about the effect of conjugation on the intrinsic estrogenic activities of these isoflavones. To characterize and compare the intrinsic estrogenic activities of genistein and daidzein, and their respective 7-O-glucuronide metabolites a cell-free assay system was employed that determines the ligand-induced changes in ERα- and ERß-ligand binding domain (LBD) interactions with 154 different binding motifs derived from 66 different nuclear receptor coregulators. The glucuronides were 8 to 4400 times less potent than their respective aglycones to modulate ERα-LBD and ERß-LBD-coregulator interactions. Glucuronidation changed the preferential activation of genistein from ERß-LBD to ERα-LBD and further increased the slightly preferential activation of daidzein for ERα-LBD. The tested isoflavone compounds were less potent than E2 (around 5 to 1580 times for the aglycones) but modulated the LBD-coregulator interactions in a manner similar to E2. Our results show that genistein and daidzein remain agonistic ligands of ERα-LBD and ERß-LBD in their conjugated form with a higher relative preference for ERα-LBD than the corresponding aglycones. This shift in receptor preference is of special interest as the preferential activation of ERß is considered one of the possible modes of action underlying the supposed beneficial instead of adverse health effects of isoflavones.


Subject(s)
Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Genistein/pharmacology , Glucuronides/metabolism , Isoflavones/pharmacology , Genistein/chemistry , Humans , Isoflavones/chemistry , Protein Binding
4.
Food Funct ; 6(4): 1098-107, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25765892

ABSTRACT

The consumption of dietary flavonoids has been associated with a variety of health benefits, including effects mediated by the activation of peroxisome proliferator-activated receptor-gamma (PPAR-γ). Flavonoids are extensively metabolized during and after uptake and there is little known on the biological effects of these conjugated metabolites of flavonoids that are found in plasma. To investigate the effect of glucuronidation on the ability of flavonoids to activate PPAR-γ we studied and compared the activity of quercetin, kaempferol and their relevant plasma conjugates quercetin-3-O-glucuronide (Q3G) and kaempferol-3-O-glucuronide (K3G) on different PPAR-γ related endpoints. The flavonoid aglycones increased PPAR-γ mediated gene expression in a stably transfected reporter gene cell line and glucuronidation diminished their effect. To study the intrinsic activity of the test compounds to activate PPAR-γ we used a novel microarray technique to study ligand induced ligand binding domain (LBD) - nuclear receptor coregulator interactions. In this cell-free system we demonstrate that, unlike the known PPAR-γ agonist rosiglitazone, neither the flavonoid aglycones nor the conjugates are agonistic ligands of the receptor. The increases in reporter gene expression in the reporter cells were accompanied by increased PPAR-γ receptor-mRNA expression and quercetin synergistically increased the effect of rosiglitazone in the reporter gene assay. It is concluded that flavonoids affect PPAR-γ mediated gene transcription by a mode of action different from agonist binding. Increases in PPAR-γ receptor mRNA expression and synergistic effects with endogenous PPAR-γ agonists may play a role in this alternative mode of action. Glucuronidation reduced the activity of the flavonoid aglycones.


Subject(s)
Glucuronides/pharmacology , Kaempferols/pharmacology , PPAR gamma/metabolism , Quercetin/analogs & derivatives , Cell Line , Drug Synergism , Endpoint Determination , Gene Expression Profiling , Gene Expression Regulation , Genes, Reporter , Humans , Ligands , PPAR gamma/genetics , Quercetin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rosiglitazone , Thiazolidinediones/pharmacology
5.
Toxicol Appl Pharmacol ; 283(2): 117-26, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25549870

ABSTRACT

The present study aims at predicting the level of formation of the ultimate carcinogenic metabolite of methyleugenol, 1'-sulfooxymethyleugenol, in the human population by taking variability in key bioactivation and detoxification reactions into account using Monte Carlo simulations. Depending on the metabolic route, variation was simulated based on kinetic constants obtained from incubations with a range of individual human liver fractions or by combining kinetic constants obtained for specific isoenzymes with literature reported human variation in the activity of these enzymes. The results of the study indicate that formation of 1'-sulfooxymethyleugenol is predominantly affected by variation in i) P450 1A2-catalyzed bioactivation of methyleugenol to 1'-hydroxymethyleugenol, ii) P450 2B6-catalyzed epoxidation of methyleugenol, iii) the apparent kinetic constants for oxidation of 1'-hydroxymethyleugenol, and iv) the apparent kinetic constants for sulfation of 1'-hydroxymethyleugenol. Based on the Monte Carlo simulations a so-called chemical-specific adjustment factor (CSAF) for intraspecies variation could be derived by dividing different percentiles by the 50th percentile of the predicted population distribution for 1'-sulfooxymethyleugenol formation. The obtained CSAF value at the 90th percentile was 3.2, indicating that the default uncertainty factor of 3.16 for human variability in kinetics may adequately cover the variation within 90% of the population. Covering 99% of the population requires a larger uncertainty factor of 6.4. In conclusion, the results showed that adequate predictions on interindividual human variation can be made with Monte Carlo-based PBK modeling. For methyleugenol this variation was observed to be in line with the default variation generally assumed in risk assessment.


Subject(s)
Carcinogens/pharmacokinetics , Eugenol/analogs & derivatives , Models, Biological , Monte Carlo Method , Carcinogens/toxicity , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Drug Evaluation/methods , Eugenol/pharmacokinetics , Eugenol/toxicity , Humans , Kinetics , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology
6.
Food Chem Toxicol ; 66: 373-84, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24508526

ABSTRACT

Safrole, present in mace and its essential oils, causes liver tumors in rodents at high dose levels due to formation of a DNA reactive 1'-sulfooxysafrole. The present study identifies malabaricone C as a mace constituent able to inhibit safrole DNA adduct formation at the level of sulfotransferase mediated bioactivation. This inhibition was incorporated into physiologically based biokinetic rat and human models. Dosing safrole at 50mg/kg body weight and malabaricone C-containing mace extract at a ratio reflecting the relative presence in mace, and assuming 100% or 1% uptake of malabaricone C-containing mace extract, the model predicted inhibition of 1'-sulfooxysafrole formation for rats and humans by 90% and 100% or 61% and 91%, respectively. To validate the model, mace extract and safrole were co-administered orally to Sprague-Dawley rats. LC-ECI-MS/MS based quantification of DNA adduct levels revealed a significant (p<0.01) 55% reduction of safrole DNA adduct formation by malabaricone C-containing mace extract in the liver of rats exposed to safrole. The data obtained were used to perform a refined risk assessment of safrole. Overall, the results suggest a lower tumor incidence when safrole would be tested within a relevant food matrix containing sulfotransferase inhibitors compared to dosing pure safrole.


Subject(s)
DNA Adducts/biosynthesis , Resorcinols/pharmacology , Safrole/pharmacokinetics , Animals , Chromatography, High Pressure Liquid , Humans , In Vitro Techniques , Proton Magnetic Resonance Spectroscopy , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
7.
Food Chem Toxicol ; 59: 564-71, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23831728

ABSTRACT

Methyleugenol (ME) occurs naturally in a variety of spices, herbs, including basil, and their essential oils. ME induces hepatomas in rodent bioassays following its conversion to a DNA reactive metabolite. In the present study, the basil constituent nevadensin was shown to be able to inhibit SULT-mediated DNA adduct formation in HepG2 cells exposed to the proximate carcinogen 1'-hydroxymethyleugenol in the presence of nevadensin. To investigate possible in vivo implications of SULT inhibition by nevadensin on ME bioactivation, the rat physiologically based kinetic (PBK) model developed in our previous work to describe the dose-dependent bioactivation and detoxification of ME in male rat was combined with the recently developed PBK model describing the dose-dependent kinetics of nevadensin in male rat. The resulting binary ME-nevadensin PBK model was used to predict the possible nevadensin mediated reduction in ME DNA adduct formation and resulting carcinogenicity at the doses of ME used by the NTP carcinogenicity study. Using these data an updated risk assessment using the Margin of Exposure (MOE) approach was performed. The results obtained point at a potential reduction of the cancer risk when rodents are orally exposed to ME within a relevant food matrix containing SULT inhibitors compared to exposure to pure ME.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinogens/pharmacokinetics , Eugenol/analogs & derivatives , Flavones/pharmacology , Hepatocytes/metabolism , Models, Biological , Animals , Anticarcinogenic Agents/blood , Anticarcinogenic Agents/metabolism , Anticarcinogenic Agents/pharmacokinetics , Biotransformation/drug effects , Carcinogens/administration & dosage , Carcinogens/metabolism , Carcinogens/toxicity , DNA Adducts/antagonists & inhibitors , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/blood , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Eugenol/administration & dosage , Eugenol/metabolism , Eugenol/pharmacokinetics , Eugenol/toxicity , Female , Flavones/blood , Flavones/metabolism , Flavones/pharmacokinetics , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Male , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Risk Assessment , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/metabolism , Tissue Distribution/drug effects
8.
Mol Nutr Food Res ; 57(11): 1969-78, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23894034

ABSTRACT

SCOPE: The present work investigates whether the previous observation that the basil flavonoid nevadensin is able to inhibit sulfotransferase (SULT)-mediated estragole DNA adduct formation in primary rat hepatocytes could be validated in vivo. METHODS AND RESULTS: Estragole and nevadensin were co-administered orally to Sprague-Dawley rats, at a ratio reflecting their presence in basil. Moreover, previously developed physiologically based biokinetic (PBBK) models to study this inhibition in rat and in human liver were refined by including a submodel describing nevadensin kinetics. Nevadensin resulted in a significant 36% reduction in the levels of estragole DNA adducts formed in the liver of rats. The refined PBBK model predicts the formation of estragole DNA adducts in the liver of rat with less than twofold difference compared to in vivo data and suggests more potent inhibition in the liver of human compared to rat due to less efficient metabolism of nevadensin in human liver and intestine. CONCLUSION: Given the role of the SULT-mediated DNA adduct formation in the hepatocarcinogenicity of estragole, the results of the present study suggest that the likelihood of bioactivation and subsequent adverse effects in rodent bioassays may be lower when estragole is dosed with nevadensin compared to dosing of pure estragole.


Subject(s)
Anisoles/adverse effects , DNA Adducts/drug effects , Flavones/pharmacology , Liver/drug effects , Ocimum basilicum/chemistry , Sulfotransferases/metabolism , Allylbenzene Derivatives , Animals , DNA Adducts/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Liver/cytology , Liver/metabolism , Male , Models, Molecular , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity
9.
Int J Sport Nutr Exerc Metab ; 23(6): 584-92, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23689036

ABSTRACT

Recent studies have demonstrated a direct link between increased exogenous CHO oxidation (CHOexog) and enhanced performance. The limiting factor for CHOexog appears to be at the level of intestinal transporters, with sodium/glucose cotransporter 1 (SGLT1) and glucose transporter Type 5 (GLUT5) responsible for glucose and fructose transport, respectively. Studies in animal models have shown that SGLT1 and intestinal glucose uptake are up-regulated by high carbohydrate diets or noncaloric sweeteners. The aim of this study was to determine the effect of preexercise ingestion of noncaloric sweeteners on CHOexog during exercise in athletes. In a randomized, crossover, double-blind fashion twenty-three healthy male cyclists (age = 29 ± 7 yrs, mass = 73.6 ± 7.4 kg, VO2peak = 68.3 ± 9.3 ml/kg/min) consumed 8 × 50 ml doses of either placebo (CON) or 1mM sucralose (SUCRA) every 15 min starting 120 min before the onset of exercise. This was followed by 2h of cycling at 48.5 ± 8.6% of VO2peak with continual ingestion of a maltodextrin drink (1.2 g/min; 828 ml/ hr). Average CHOexog during the first hour of exercise did not differ between SUCRA and CON conditions (0.226 ± 0.081 g/min vs. 0.212 ± 0.076 g/min, Δ =0.015 g/min, 95% CI -0.008 g/min, 0.038 g/min, p = .178). Blood glucose, plasma insulin and lactate, CHO and fat substrate utilization, heart rate, ratings of perceived exertion, and gastrointestinal symptoms did not differ between conditions. Our data suggest that consumption of noncaloric sweeteners in the immediate period before exercise does not lead to a significant increase in CHOexog during exercise.


Subject(s)
Bicycling/physiology , Carbohydrate Metabolism/drug effects , Exercise/physiology , Sports Nutritional Physiological Phenomena , Sucrose/analogs & derivatives , Adult , Blood Glucose/metabolism , Cross-Over Studies , Double-Blind Method , Energy Metabolism , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Heart Rate , Humans , Insulin/blood , Lactic Acid/blood , Male , Oxidation-Reduction/drug effects , Oxygen Consumption , Physical Endurance , Polysaccharides/administration & dosage , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Sucrose/administration & dosage , Young Adult
10.
Mutagenesis ; 27(6): 653-63, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22844077

ABSTRACT

Estragole is a naturally occurring food-borne genotoxic compound found in a variety of food sources, including spices and herbs. This results in human exposure to estragole via the regular diet. The objective of this study was to quantify the dose-dependent estragole-DNA adduct formation in rat liver and the urinary excretion of 1'-hydroxyestragole glucuronide in order to validate our recently developed physiologically based biodynamic (PBBD) model. Groups of male outbred Sprague Dawley rats (n = 10, per group) were administered estragole once by oral gavage at dose levels of 0 (vehicle control), 5, 30, 75, 150, and 300mg estragole/kg bw and sacrificed after 48h. Liver, kidney and lungs were analysed for DNA adducts by LC-MS/MS. Results obtained revealed a dose-dependent increase in DNA adduct formation in the liver. In lungs and kidneys DNA adducts were detected at lower levels than in the liver confirming the occurrence of DNA adducts preferably in the target organ, the liver. The results obtained showed that the PBBD model predictions for both urinary excretion of 1'-hydroxyestragole glucuronide and the guanosine adduct formation in the liver were comparable within less than an order of magnitude to the values actually observed in vivo. The PBBD model was refined using liver zonation to investigate whether its predictive potential could be further improved. The results obtained provide the first data set available on estragole-DNA adduct formation in rats and confirm their occurrence in metabolically active tissues, i.e. liver, lung and kidney, while the significantly higher levels found in liver are in accordance with the liver as the target organ for carcinogenicity. This opens the way towards future modelling of dose-dependent estragole liver DNA adduct formation in human.


Subject(s)
Anisoles/toxicity , DNA Adducts/drug effects , Models, Biological , Administration, Oral , Allylbenzene Derivatives , Animals , Anisoles/urine , Chromatography, Liquid , Dose-Response Relationship, Drug , Glucuronides/urine , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
11.
Food Funct ; 3(10): 1008-18, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22751838

ABSTRACT

Diets rich in flavonoids are associated with various positive health effects. Most in vitro research conducted to elucidate the modes of action of flavonoids uses flavonoid aglycones, but not their circulating conjugated metabolites. Conjugation alters the physico-chemical properties of flavonoids and it is widely assumed that this can affect their biological activity. This article gives a state-of-the-art overview of scientific literature reporting on the effect of metabolic conjugation on the biological activity of flavonoids. The biological activity of flavonoid aglycones is compared to that of their conjugates for a broad range of endpoints. Even though there is only limited literature available, it is shown that contrary to common belief, conjugation does not always decrease the biological activity of flavonoids. There are also endpoints which are unaffected by conjugation, and endpoints on which the conjugates have a higher or inverse activity when compared to the aglycone. The effects of conjugation can differ depending on the type and position of conjugation, the flavonoid concentration, the endpoint studied and the assay system used so that no general rules can be deducted. It is concluded that further studies on the effects of conjugation have to be done on a case-by-case basis, and characterization of the stability and metabolic fate of the flavonoids in the assay system under consideration is needed to avoid false positive or false negative outcomes.


Subject(s)
Flavonoids/metabolism , Angiogenesis Inducing Agents/metabolism , Cell Adhesion/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Diet , Endpoint Determination , Humans , Lipoproteins/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Vasodilation/drug effects
12.
Toxicol Sci ; 129(1): 174-87, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22649189

ABSTRACT

The alkenylbenzene estragole is a constituent of several herbs and spices. It induces hepatomas in rodents at high doses following bioactivation by cytochrome P450s and sulfotransferases (SULTs) giving rise to the ultimate carcinogenic metabolite 1'-sulfooxyestragole which forms DNA adducts. Methanolic extracts from different alkenylbenzene-containing herbs and spices were able to inhibit SULT activity. Flavonoids including quercetin, kaempferol, myricetin, apigenin, and nevadensin were the major constituents responsible for this inhibition with Ki values in the nano to micromolar range. In human HepG2 cells exposed to the proximate carcinogen 1'-hydroxyestragole, the various flavonoids were able to inhibit estragole DNA adduct formation and shift metabolism in favor of glucuronidation which is a detoxification pathway for 1'-hydroxyestragole. In a next step, the kinetics for SULT inhibition were incorporated in physiologically based biokinetic (PBBK) models for estragole in rat and human to predict the effect of co-exposure to estragole and (mixtures of) the different flavonoids on the bioactivation in vivo. The PBBK-model-based predictions indicate that the reduction of estragole bioactivation in rat and human by co-administration of the flavonoids is dependent on whether the intracellular liver concentrations of the flavonoids can reach their Ki values. It is expected that this is most easily achieved for nevadensin which has a Ki value in the nanomolar range and is, due to its methyl ation, more metabolically stable than the other flavonoids.


Subject(s)
Anisoles/pharmacokinetics , Benzene Derivatives/pharmacokinetics , Spices/analysis , Allylbenzene Derivatives , Anisoles/pharmacology , Benzene Derivatives/pharmacology , Cell Line , Chromatography, High Pressure Liquid , Humans , Microsomes, Liver/metabolism , Oxidation-Reduction , Spectrophotometry, Ultraviolet
13.
Toxicol Sci ; 128(2): 301-16, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22588462

ABSTRACT

A physiologically based biokinetic (PBBK) model for the alkenylbenzene safrole in humans was developed based on in vitro- and in silico-derived kinetic parameters. With the model obtained, the time- and dose-dependent formation of the proximate and ultimate carcinogenic metabolites, 1-hydroxysafrole and 1-sulfooxysafrole in human liver were estimated and compared with previously predicted levels of these metabolites in rat liver. In addition, Monte Carlo simulations were performed to predict interindividual variation in the formation of these metabolites in the overall population. For the evaluation of the model performance, a comparison was made between the predicted total amount of urinary metabolites of safrole and the reported total levels of metabolites in the urine of humans exposed to safrole, which adequately matched. The model results revealed no dose-dependent shifts in safrole metabolism and no relative increase in bioactivation at dose levels up to 100mg/kg body weight/day. Species differences were mainly observed in the detoxification pathways of 1-hydroxysafrole, with the formation of 1-oxosafrole being a main detoxification pathway of 1-hydroxysafrole in humans but a minor pathway in rats, and glucuronidation of 1-hydroxysafrole being less important in humans than in rats. The formation of 1-sulfooxysafrole was predicted to vary 4- to 17-fold in the population (fold difference between the 95th and median, and 95th and 5th percentile, respectively), with the median being three to five times higher in human than in rat liver. Comparison of the PBBK results for safrole with those previously obtained for the related alkenylbenzenes estragole and methyleugenol revealed that differences in 1-sulfooxy metabolite formation are limited, being only twofold to fivefold.


Subject(s)
Models, Molecular , Safrole/pharmacokinetics , Animals , Biotransformation , Chromatography, High Pressure Liquid , Humans , Male , Rats
14.
Toxicol Appl Pharmacol ; 260(3): 271-84, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22445790

ABSTRACT

This study defines a physiologically based kinetic (PBK) model for methyleugenol (ME) in human based on in vitro and in silico derived parameters. With the model obtained, bioactivation and detoxification of methyleugenol (ME) at different doses levels could be investigated. The outcomes of the current model were compared with those of a previously developed PBK model for methyleugenol (ME) in male rat. The results obtained reveal that formation of 1'-hydroxymethyleugenol glucuronide (1'HMEG), a major metabolic pathway in male rat liver, appears to represent a minor metabolic pathway in human liver whereas in human liver a significantly higher formation of 1'-oxomethyleugenol (1'OME) compared with male rat liver is observed. Furthermore, formation of 1'-sulfooxymethyleugenol (1'HMES), which readily undergoes desulfonation to a reactive carbonium ion (CA) that can form DNA or protein adducts (DA), is predicted to be the same in the liver of both human and male rat at oral doses of 0.0034 and 300 mg/kg bw. Altogether despite a significant difference in especially the metabolic pathways of the proximate carcinogenic metabolite 1'-hydroxymethyleugenol (1'HME) between human and male rat, the influence of species differences on the ultimate overall bioactivation of methyleugenol (ME) to 1'-sulfooxymethyleugenol (1'HMES) appears to be negligible. Moreover, the PBK model predicted the formation of 1'-sulfooxymethyleugenol (1'HMES) in the liver of human and rat to be linear from doses as high as the benchmark dose (BMD10) down to as low as the virtual safe dose (VSD). This study shows that kinetic data do not provide a reason to argue against linear extrapolation from the rat tumor data to the human situation.


Subject(s)
Computer Simulation , Eugenol/analogs & derivatives , Microsomes, Liver/metabolism , Models, Biological , Administration, Oral , Animals , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Eugenol/administration & dosage , Eugenol/pharmacokinetics , Eugenol/toxicity , Female , Humans , Male , Rats , Species Specificity
15.
Biopharm Drug Dispos ; 32(9): 530-5, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22083890

ABSTRACT

The citrus flavonoid hesperetin (4'-methoxy-3',5,7-trihydroxyflavanone) is the aglycone of hesperidin, the major flavonoid present in sweet oranges. Hesperetin 7-O-glucuronide (H7G) and hesperetin 3'-O-glucuronide (H3'G) are the two most abundant metabolites of hesperetin in vivo. In this study, their interaction with specific ABC transporters, believed to play a role in the disposition and bioavailability of hesperetin, was studied using Sf9 membranes from cells overexpressing human BCRP (ABCG2), MRP2 (ABCC2) and MRP3 (ABCC3). Both H7G and H3'G were tested for their potential to activate and inhibit ATPase activity, and to inhibit vesicular transport by these transporters. Both H7G and H3'G demonstrated interaction with all tested ABC transporters, especially with BCRP and MRP3. An interesting difference between H7G and H3'G was seen with respect to the interaction with BCRP: H7G stimulated the ATPase activity of BCRP up to 76% of the maximal effect generated by the reference activator sulfasalazine, with an EC(50) of 0.45 µM, suggesting that H7G is a high affinity substrate of BCRP, whereas H3'G did not stimulate BCRP ATPase activity. Only moderate inhibition of BCRP ATPase activity at high H3'G concentrations was observed. This study provides information on the potential of hesperetin glucuronide conjugates to act as specific ABC transporter substrates or inhibitors and indicates that regio-specific glucuronidation could affect the disposition of hesperetin.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Glucuronides/pharmacology , Hesperidin/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/metabolism , Animals , Baculoviridae/genetics , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/genetics , Spodoptera/genetics , Transport Vesicles/drug effects , Transport Vesicles/metabolism
16.
Mutagenesis ; 26(5): 605-18, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21642616

ABSTRACT

This study aimed at quantitatively comparing the occurrence/formation of DNA adducts with the carcinogenicity induced by a selection of DNA-reactive genotoxic carcinogens. Contrary to previous efforts, we used a very uniform set of data, limited to in vivo rat liver studies in order to investigate whether a correlation can be obtained, using a benchmark dose (BMD) approach. Dose-response data on both carcinogenicity and in vivo DNA adduct formation were available for six compounds, i.e. 2-acetylaminofluorene, aflatoxin B1, methyleugenol, safrole, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline and tamoxifen. BMD(10) values for liver carcinogenicity were calculated using the US Environmental Protection Agency BMD software. DNA adduct levels at this dose were extrapolated assuming linearity of the DNA adduct dose response. In addition, the levels of DNA adducts at the BMD(10) were compared to available data on endogenous background DNA damage in the target organ. Although for an individual carcinogen the tumour response increases when adduct levels increase, our results demonstrate that when comparing different carcinogens, no quantitative correlation exists between the level of DNA adduct formation and carcinogenicity. These data confirm that the quantity of DNA adducts formed by a DNA-reactive compound is not a carcinogenicity predictor but that other factors such as type of adduct and mutagenic potential may be equally relevant. Moreover, comparison to background DNA damage supports the notion that the mere occurrence of DNA adducts above or below the level of endogenous DNA damage is neither correlated to development of cancer. These data strongly emphasise the need to apply the mode of action framework to understand the contribution of other biological effect markers playing a role in carcinogenicity.


Subject(s)
Carcinogens/toxicity , DNA Adducts/metabolism , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/metabolism , Animals , Carcinogenicity Tests , Carcinogens/administration & dosage , Carcinogens/pharmacology , DNA Damage/drug effects , Dose-Response Relationship, Drug , Female , Incidence , Liver/drug effects , Liver/metabolism , Male , Mice , Neoplasms, Experimental/epidemiology , Rabbits
17.
Toxicol In Vitro ; 25(1): 267-85, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20828604

ABSTRACT

The present study defines a physiologically based biokinetic (PBBK) model for the alkenylbenzene methyleugenol in rat based on in vitro metabolic parameters determined using relevant tissue fractions, in silico derived partition coefficients, and physiological parameters derived from the literature. The model was based on the model previously developed for the related alkenylbenzene estragole and consists of eight compartments including liver, lung, and kidney as metabolizing compartments, and separate compartments for fat, arterial blood, venous blood, richly perfused and slowly perfused tissues. Evaluation of the model was performed by comparing the PBBK predicted concentration of methyleugenol in the venous compartment to methyleugenol plasma levels reported in the literature, by comparing the PBBK predicted dose-dependent percentage of formation of 2-hydroxy-4,5-dimethoxyallylbenzene, 3-hydroxy-4-methoxyallylbenzene, and 1'-hydroxymethyleugenol glucuronide to the corresponding percentage of metabolites excreted in urine reported in the literature, which were demonstrated to be in the same order of magnitude. With the model obtained the relative extent of bioactivation and detoxification of methyleugenol at different oral doses was examined. At low doses, formation of 3-(3,4-dimethoxyphenyl)-2-propen-1-ol and methyleugenol-2',3'-oxide leading to detoxification appear to be the major metabolic pathways, occurring in the liver. At high doses, the model reveals a relative increase in the formation of the proximate carcinogenic metabolite 1'-hydroxymethyleugenol, occurring in the liver. This relative increase in formation of 1'-hydroxymethyleugenol leads to a relative increase in formation of 1'-hydroxymethyleugenol glucuronide, 1'-oxomethyleugenol, and 1'-sulfooxymethyleugenol the latter being the ultimate carcinogenic metabolite of methyleugenol. These results indicate that the relative importance of different metabolic pathways of methyleugenol may vary in a dose-dependent way, leading to a relative increase in bioactiviation of methyleugenol at higher doses.


Subject(s)
Alkenes/chemistry , Carcinogens/metabolism , Carcinogens/pharmacokinetics , Eugenol/analogs & derivatives , Microsomes, Liver/metabolism , Models, Biological , Mutagens/metabolism , Mutagens/pharmacokinetics , Animals , Biocatalysis , Biotransformation , Carcinogens/administration & dosage , Computational Biology , Dose-Response Relationship, Drug , Eugenol/administration & dosage , Eugenol/metabolism , Eugenol/pharmacokinetics , Expert Systems , Female , Kinetics , Male , Mutagens/administration & dosage , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Sex Characteristics , Species Specificity
18.
Front Genet ; 2: 21, 2011.
Article in English | MEDLINE | ID: mdl-22303317

ABSTRACT

Comprehensive investigation of nutritional health effects at the molecular level requires the understanding of the interplay between three genomes, the food, the gut microbial, and the human host genome. Food genomes are researched for discovery and exploitation of macro- and micronutrients as well as specific bioactives, with those genes coding for bioactive proteins and peptides being of central interest. The human gut microbiota encompasses a complex ecosystem in the intestine with profound impact on host metabolism. It is being studied at genomic and, more recently, also at proteomic and metabonomic level. Humans are being characterized at the level of genetic pre-disposition and inter-individual variability in terms of (i) response to nutritional interventions and direction of health trajectories; (ii) epigenetic, metabolic programming at certain life stages with health consequences later in life and even for subsequent generations; and (iii) acute genomic expression as a holistic response to diet, monitored at gene transcript, protein and metabolite level. Modern nutrition science explores health-related aspects of bioactive food components, thereby promoting health, preventing, or delaying the onset of disease, optimizing performance and assessing benefits and risks in individuals and subpopulations. Personalized nutrition means adapting food to individual needs, depending on the human host's life stage, -style, and -situation. Traditionally, nutrigenomics and nutri(epi)genetics are seen as the key sciences to understand human variability in preferences and requirements for diet as well as responses to nutrition. This article puts the three nutrition and health-relevant genomes into perspective, namely the food, the gut microbial and the human host's genome, and calls for an "extended nutrigenomics" approach in order to build the future tools for personalized nutrition, health maintenance, and disease prevention. We discuss examples of these genomes, proteomes, transcriptomes, and metabolomes under the definition of genomics as the overarching term covering essentially all Omics rather than the sole study of DNA and RNA.

19.
J Agric Food Chem ; 58(10): 6119-25, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20441166

ABSTRACT

The flavanone hesperetin ((+/-)-4'-methoxy-3',5,7-trihydroxyflavanone) is the aglycone of hesperidin, which is the major flavonoid present in sweet oranges. Hesperetin contains a chiral C-atom and so can exist as an S- and R-enantiomer, however, in nature 2S-hesperidin and its S-hesperetin aglycone are predominant. The present study reports a chiral HPLC method to separate S- and R-hesperetin on an analytical and semipreparative scale. This allowed characterization of the stereoselective differences in metabolism and transport in the intestine and activity in a selected bioassay of the separated hesperetin enantiomers in in vitro model systems: (1) with human small intestinal fractions containing UDP-glucuronosyl transferases (UGTs) or sulfotransferases (SULTs); (2) with Caco-2 cell monolayers as a model for the intestinal transport barrier; (3) with mouse Hepa-1c1c7 cells transfected with human EpRE-controlled luciferase to test induction of EpRE-mediated gene expression. The results obtained indicate some significant differences in the metabolism and transport characteristics and bioactivity between S- and R-hesperetin, however, these differences are relatively small. This indicates that for these end points, including intestinal metabolism and transport and EpRE-mediated gene induction, experiments performed with racemic hesperetin may adequately reflect what can be expected for the naturally occurring S-enantiomer. This is an important finding since at present hesperetin is only commercially available as a racemic mixture, while it exists in nature mainly as an S-enantiomer.


Subject(s)
Hesperidin/chemistry , Hesperidin/metabolism , Animals , Biological Transport , Caco-2 Cells , Cell Line, Tumor , Citrus sinensis/chemistry , Cytosol/metabolism , Fruit/chemistry , Gene Expression/drug effects , Glucuronides/metabolism , Hesperidin/pharmacology , Humans , Intestine, Small/metabolism , Intestine, Small/ultrastructure , Liver Neoplasms, Experimental , Mice , Microsomes, Liver/metabolism , Response Elements/genetics , Stereoisomerism , Structure-Activity Relationship , Sulfonic Acids/metabolism , Transfection
20.
Toxicol Appl Pharmacol ; 245(1): 57-66, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20144636

ABSTRACT

Estragole has been shown to be hepatocarcinogenic in rodent species at high-dose levels. Translation of these results into the likelihood of formation of DNA adducts, mutation, and ultimately cancer upon more realistic low-dose exposures remains a challenge. Recently we have developed physiologically based biokinetic (PBBK) models for rat and human predicting bioactivation of estragole. These PBBK models, however, predict only kinetic characteristics. The present study describes the extension of the PBBK model to a so-called physiologically based biodynamic (PBBD) model predicting in vivo DNA adduct formation of estragole in rat liver. This PBBD model was developed using in vitro data on DNA adduct formation in rat primary hepatocytes exposed to 1'-hydroxyestragole. The model was extended by linking the area under the curve for 1'-hydroxyestragole formation predicted by the PBBK model to the area under the curve for 1'-hydroxyestragole in the in vitro experiments. The outcome of the PBBD model revealed a linear increase in DNA adduct formation with increasing estragole doses up to 100 mg/kg bw. Although DNA adduct formation of genotoxic carcinogens is generally seen as a biomarker of exposure rather than a biomarker of response, the PBBD model now developed is one step closer to the ultimate toxic effect of estragole than the PBBK model described previously. Comparison of the PBBD model outcome to available data showed that the model adequately predicts the dose-dependent level of DNA adduct formation. The PBBD model predicts DNA adduct formation at low levels of exposure up to a dose level showing to cause cancer in rodent bioassays, providing a proof of principle for modeling a toxicodynamic in vivo endpoint on the basis of solely in vitro experimental data.


Subject(s)
Anisoles/toxicity , Carcinogens/toxicity , DNA Adducts/metabolism , Hepatocytes/drug effects , Models, Biological , Allylbenzene Derivatives , Animals , Anisoles/chemistry , Anisoles/metabolism , Hepatocytes/metabolism , Humans , Male , Mutagenicity Tests , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...