Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Arch Toxicol ; 91(1): 393-406, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26872951

ABSTRACT

Human primary hepatocytes represent a gold standard in in vitro liver research. Due to their low availability and high costs alternative liver cell models with comparable morphological and biochemical characteristics have come into focus. The human hepatocarcinoma cell line HepG2 is often used as a liver model for toxicity studies. However, under two-dimensional (2D) cultivation conditions the expression of xenobiotic-metabolizing enzymes and typical liver markers such as albumin is very low. Cultivation for 21 days in a three-dimensional (3D) Matrigel culture system has been reported to strongly increase the metabolic competence of HepG2 cells. In our present study we further compared HepG2 cell cultivation in three different 3D systems: collagen, Matrigel and Alvetex culture. Cell morphology, albumin secretion, cytochrome P450 monooxygenase enzyme activities, as well as gene expression of xenobiotic-metabolizing and liver-specific enzymes were analyzed after 3, 7, 14, and 21 days of cultivation. Our results show that the previously reported increase of metabolic competence of HepG2 cells is not primarily the result of 3D culture but a consequence of the duration of cultivation. HepG2 cells grown for 21 days in 2D monolayer exhibit comparable biochemical characteristics, CYP activities and gene expression patterns as all 3D culture systems used in our study. However, CYP activities did not reach the level of HepaRG cells. In conclusion, the increase of metabolic competence of the hepatocarcinoma cell line HepG2 is not due to 3D cultivation but rather a result of prolonged cultivation time.


Subject(s)
Cell Culture Techniques , Gene Expression Regulation, Enzymologic , Hepatocytes/metabolism , Tissue Scaffolds/chemistry , Toxicity Tests , Animals , Biomarkers/metabolism , Cell Shape , Cell Survival/drug effects , Cells, Cultured , Collagen/chemistry , Drug Combinations , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/drug effects , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Imaging, Three-Dimensional , Laminin/chemistry , Microscopy, Confocal , Proteoglycans/chemistry , RNA, Messenger/metabolism , Time Factors , Xenobiotics/toxicity
2.
Arch Toxicol ; 90(8): 2025-36, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26374342

ABSTRACT

The ingestion of seafood contaminated with the marine biotoxin okadaic acid (OA) can lead to diarrhetic shellfish poisoning with symptoms like nausea, vomiting and abdominal cramps. Both rat and the human hepatic cytochrome P450 monooxygenases (CYP) metabolize OA. However, liver cell toxicity of metabolized OA is mainly unclear. The aim of our study was to detect the cellular effects in HepG2 cells exposed to OA in the presence of recombinant CYP enzymes of both rat and human for the investigation of species differences. The results should be set in correlation with a CYP-specific metabolite pattern. Comparative metabolite profiles of OA after incubation in rat and human recombinant CYP enzymes were established by using LC-MS/MS technique. Results demonstrated that metabolism of OA to oxygenated metabolites correlates with detoxification which was mainly catalyzed by human CYP3A4 and CYP3A5. Detoxification by rat Cyp3a1 was lower compared to human CYP3A enzymes and activation of OA by Cyp3a2 was observed, coincident with minor overall conversion capacity of OA. By contrast human and rat CYP1A2 seem to activate OA into cytotoxic intermediates. In conclusion, different mechanisms of OA metabolism may occur in the liver. At low OA doses, the human liver is likely well protected against cytotoxic OA, but for high shellfish consumers a potential risk cannot be excluded.


Subject(s)
Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Liver/drug effects , Marine Toxins/toxicity , Okadaic Acid/toxicity , Animals , Cell Culture Techniques , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP3A/genetics , Hep G2 Cells , Humans , Liver/enzymology , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity , Tandem Mass Spectrometry
3.
Anal Biochem ; 487: 45-8, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26212314

ABSTRACT

Reporter gene assays are widely used for the assessment of transcription factor activation following xenobiotic exposure of cells. A critical issue with such assays is the possibility of interference of test compounds with the test system, for example, by direct inhibition of the reporter enzyme. Here we show that the pyrrolizidine alkaloid heliotrine interferes with reporter signals derived from GAL4-based nuclear receptor transactivation assays by a mechanism independent of luciferase enzyme inhibition. These data highlight the necessity to conduct proper control experiments in order to avoid perturbation of reporter assays by test chemicals.


Subject(s)
Genes, Reporter/drug effects , Luciferases, Firefly/antagonists & inhibitors , Luciferases, Renilla/antagonists & inhibitors , Pyrrolizidine Alkaloids/pharmacology , Animals , Fireflies , Genes, Reporter/genetics , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Luciferases, Renilla/genetics , Luciferases, Renilla/metabolism , Pyrrolizidine Alkaloids/chemistry , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renilla
4.
Toxicol In Vitro ; 29(7): 1669-82, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26100227

ABSTRACT

1,2-unsaturated pyrrolizidine alkaloids (PA) are plant metabolites predominantly occurring in the plant families Asteraceae and Boraginaceae. Acute and chronic PA poisoning causes severe hepatotoxicity. So far, the molecular mechanisms of PA toxicity are not well understood. To analyze its mode of action, primary human hepatocytes were exposed to a non-cytotoxic dose of 100 µM of four structurally different PA: echimidine, heliotrine, senecionine, senkirkine. Changes in mRNA expression were analyzed by a whole genome microarray. Employing cut-off values with a |fold change| of 2 and a q-value of 0.01, data analysis revealed numerous changes in gene expression. In total, 4556, 1806, 3406 and 8623 genes were regulated by echimidine, heliotrine, senecione and senkirkine, respectively. 1304 genes were identified as commonly regulated. PA affected pathways related to cell cycle regulation, cell death and cancer development. The transcription factors TP53, MYC, NFκB and NUPR1 were predicted to be activated upon PA treatment. Furthermore, gene expression data showed a considerable interference with lipid metabolism and bile acid flow. The associated transcription factors FXR, LXR, SREBF1/2, and PPARα/γ/δ were predicted to be inhibited. In conclusion, though structurally different, all four PA significantly regulated a great number of genes in common. This proposes similar molecular mechanisms, although the extent seems to differ between the analyzed PA as reflected by the potential hepatotoxicity and individual PA structure.


Subject(s)
Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Pyrrolizidine Alkaloids/toxicity , Cell Survival/drug effects , Gene Expression Profiling , Genome, Human , Hep G2 Cells , Hepatocytes/metabolism , Humans , RNA, Messenger/metabolism
5.
Mol Nutr Food Res ; 59(6): 1117-29, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25788151

ABSTRACT

SCOPE: Quercetin is widespread in plant kingdom and consumed regularly with human diet (16 mg/day). Due to reported positive effects on health, quercetin supplements with recommended doses up to 2 g/day are offered. However, molecular effects of such high doses on human liver have not been assessed yet. Therefore, molecular effects on human hepatocytes were analyzed to help assessing the risk of quercetin supplementation. METHODS AND RESULTS: Molecular effects of three different quercetin concentrations on gene expression in human hepatocytes were investigated by microarray analysis. Possible new signaling pathways were investigated using reporter gene assays. Quercetin concentrations representing the normal intake showed weak effects on mRNA expression in liver cells. In contrast, supplemental doses affect immune response and p53 signaling and might be associated with cancer. Additionally, quercetin showed inhibition of transcriptional activation and mRNA-expression of HNF4α and its target genes. Inhibitory effects were also found for FXR, LXRα, and PXR. CONCLUSION: Normal intake of quercetin seems to play a minor regulatory role, while supplement doses may have great effects on gene expression in hepatocytes. However, since it is not clarified whether such high doses of quercetin exert positive or negative effects, a careful handling of quercetin supplements is advised.


Subject(s)
Hepatocytes/drug effects , Quercetin/pharmacology , Signal Transduction , Transcriptome , Antioxidants/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Genes, Reporter , HEK293 Cells , Hep G2 Cells , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Chem Biol Interact ; 224: 36-41, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25451572

ABSTRACT

The ATP-binding cassette transporters Breast Cancer Resistance Protein (Abcg2) and Multidrug Resistance-associated Protein 2 (Abcc2) play an important role for the hepatobiliary elimination of drugs and toxins as well as their metabolites. Previous in vitro transport studies showed that both transporters are involved in the active efflux of phase II metabolites of carcinogenic benzo[a]pyrene (BP), however the role of these carriers in hepatobiliary elimination in vivo is still unknown. In the present study, Abcg2(-/-) and Abcc2(-/-) knockout mice were used to elucidate the role of Abcg2 and Abcc2 for the hepatobiliary excretion of BP and its metabolites. After intravenous application of [(3)H]BP the hepatobiliary excretion was significantly reduced in these mice: whereas wild type mice excreted on average 25.4% of the applied dose into the bile over 90min, Abcg2(-/-) knockout mice only excreted 10.7% and Abcc2(-/-) knockout mice 8.6%. As a consequence, [(3)H]BP concentrations were in general higher in the plasma and in most of the organs of the Abcg2 and Abcc2 knockout mice. Both transporters may have a protective function for BP-induced carcinogenesis in humans, due to its crucial importance for the hepatobiliary elimination of BP via bile. Subjects with reduced ABCG2 or ABCC2 expression might have higher oral bioavailability for BP due to a reduced excretion and so might be more susceptible to BP-induced carcinogenesis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Benzo(a)pyrene/metabolism , Bile/metabolism , Liver/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Animals , Benzo(a)pyrene/administration & dosage , Biological Transport , Brain/metabolism , Gastric Mucosa/metabolism , Lung/metabolism , Male , Metabolic Clearance Rate , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Myocardium/metabolism , RNA, Messenger/metabolism
7.
Drug Metab Rev ; 46(3): 291-324, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24666398

ABSTRACT

In general, xenobiotic metabolizing enzymes (XMEs) are expressed in lower levels in the extrahepatic tissues than in the liver, making the former less relevant for the clearance of xenobiotics. Local metabolism, however, may lead to tissue-specific adverse responses, e.g. organ toxicities, allergies or cancer. This review summarizes the knowledge on the expression of phase I and phase II XMEs and transporters in extrahepatic tissues at the body's internal-external interfaces. In the lung, CYPs of families 1, 2, 3 and 4 and epoxide hydrolases are important phase I enzymes, while conjugation is less relevant. In skin, phase I-related enzymatic reactions are considered less relevant. Predominant skin XMEs are phase II enzymes, whereby glucuronosyltransferases (UGT) 1, glutathione-S-transferase (GST) and N-acetyltransferase (NAT) 1 are important for detoxification. The intestinal epithelium expresses many transporters and phase I XME with high levels of CYP3A4 and CYP3A5 and phase II metabolism is mainly related to UGT, NAT and Sulfotransferases (SULT). In the kidney, conjugation reactions and transporters play a major role for excretion processes. In the bladder, CYPs are relevant and among the phase II enzymes, NAT1 is involved in the activation of bladder carcinogens. Expression of XMEs is regulated by several mechanisms (nuclear receptors, epigenetic mechanisms, microRNAs). However, the understanding why XMEs are differently expressed in the various tissues is fragmentary. In contrast to the liver - where for most XMEs lower expression is demonstrated in early life - the XME ontogeny in the extrahepatic tissues remains to be investigated.


Subject(s)
Biological Transport/physiology , Xenobiotics/metabolism , Animals , Cytochrome P-450 Enzyme System/metabolism , Humans , Membrane Transport Proteins/metabolism
8.
Toxicol Lett ; 225(2): 311-7, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24374049

ABSTRACT

The consumption of okadaic acid (OA) contaminated shellfish can induce acute toxic symptoms in humans such as diarrhea, nausea, vomiting and abdominal pain; carcinogenic and embryotoxic effects have also been described. Toxicokinetic studies with mice have shown that high cytotoxic doses of OA can pass the gastrointestinal barrier presumably by paracellular passage. However, in vitro studies using human intestinal Caco-2 cell monolayers to represent the intestinal barrier have shown that at low-dose exposure OA is transported against a concentration gradient suggesting an active efflux mechanism. Since P-glycoprotein (P-gp) transports a wide variety of substrates, we investigated its possible influence on the observed elimination of OA. We used two different cellular transwell models: (i) Caco-2 cell monolayer endogenously expressing human P-gp and simulating the intestinal barrier and (ii) MDCK-II cell monolayer stably over-expressing P-gp. Our study demonstrates clearly that OA at non-cytotoxic concentrations passes the monolayer barrier only to a low degree, and that it is actively eliminated by P-gp over the apical membrane. Therefore, our in vitro data indicate that humans appear to have efficient defense mechanisms to protect themselves against low-dose contaminated shellfish by exhibiting a low bioavailability as a result of active elimination of OA by P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Carcinogens/pharmacokinetics , Intestines/drug effects , Okadaic Acid/pharmacokinetics , Animals , Biological Availability , Biological Transport , Caco-2 Cells , Carcinogens/toxicity , Chromatography, High Pressure Liquid , Dogs , Dose-Response Relationship, Drug , Humans , Inactivation, Metabolic , Intestinal Mucosa/metabolism , Madin Darby Canine Kidney Cells , Marine Toxins/pharmacokinetics , Marine Toxins/toxicity , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Okadaic Acid/toxicity , Solid Phase Extraction , Tandem Mass Spectrometry
9.
Mol Nutr Food Res ; 58(5): 995-1004, 2014 May.
Article in English | MEDLINE | ID: mdl-24375927

ABSTRACT

SCOPE: 1,2-Unsaturated pyrrolizidine alkaloids (PA) are found in plants such as Asteraceae and Boraginaceae families. Acute PA poisoning via contaminated food or feed causes severe damage to liver depending on species-specific oral bioavailability. For assessing PA bioavailability, their passage across the intestinal barrier was investigated using Caco-2 cells. METHODS: Differentiated Caco-2 cells were exposed in transport chambers to the PA heliotrine (Hn), echimidine (Em), senecionine (Sc), and senkirkine (Sk). Cell supernatants were analyzed by LC-MS/MS. RESULTS: PA pass Caco-2 monolayer from the apical into basolateral compartment depending on their chemical structure. Compared to the cyclic diesters Sc and Sk with a passage rate of 47% ± 4 and 40% ± 3, respectively, the transferred amount of the monoester Hn (32% ± 3) and open-chained diester Em (13% ± 2) was substantially lower. This suggested an active transport of Hn and Em. Using Madin-Darby canine kidney II/P-glycoprotein (ABCB1)-overexpressing cells, the active excretion of Hn and Em by ABCB1 from the gastrointestinal epithelium into the gut lumen was shown. CONCLUSION: PA cross the intestinal barrier structure-dependently. The passage of the noncyclic PA Hn and Em is reduced by an ABCB1-driven efflux into the gastrointestinal lumen resulting in a decreased oral bioavailability.


Subject(s)
Gastrointestinal Tract/drug effects , Plant Extracts/chemistry , Pyrrolizidine Alkaloids/chemistry , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Asteraceae/chemistry , Biological Availability , Biological Transport/drug effects , Biological Transport, Active/drug effects , Caco-2 Cells , Chromatography, Liquid , Dogs , Gastrointestinal Tract/metabolism , Humans , Madin Darby Canine Kidney Cells , Plant Extracts/pharmacology , Pyrrolizidine Alkaloids/pharmacology , Structure-Activity Relationship , Tandem Mass Spectrometry
10.
Food Chem Toxicol ; 62: 797-804, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24140465

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are well-known food contaminants comprising compounds with carcinogenic properties. Pyrene (PYR) is an important non-carcinogenic PAH because its metabolites are frequently used as biomarkers to assess human PAH exposure. This study investigated for the first time the formation and transport of PYR metabolites in the human small intestinal Caco-2 cell model using HPLC technique. The intermediate phase I metabolite 1-hydroxypyrene formed by cytochrome P450 monooxygenases is subsequently conjugated by phase II enzymes to the water-soluble PYR 1-glucuronide as minor and PYR 1-sulfate as major metabolites. The formation of the latter is mediated by human sulfotransferases 1A1*Arg, 1A2*1, 1A3, and 1B1. Caco-2 monolayer experiments revealed a predominantly apical transport of both conjugates mediated by the breast cancer resistance protein (BCRP/ABCG2). Additional treatment with several aryl hydrocarbon receptor (AhR) agonists indicate an AhR-driven induction of PYR-metabolizing enzymes and/or ABCG2. Overall, this study provides advanced mechanistic insights into the bioavailability of PYR and underlines a key role of the human small intestinal epithelium for the first pass metabolism of contaminants in food.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Neoplasm Proteins/metabolism , Polycyclic Aromatic Hydrocarbons/pharmacokinetics , Pyrenes/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Arylsulfotransferase/metabolism , Biological Availability , Biological Transport , Biomarkers/metabolism , Caco-2 Cells/drug effects , Cytochrome P-450 Enzyme System/metabolism , Food Contamination , Humans , Pyrenes/pharmacokinetics , Sulfotransferases/metabolism
11.
Toxicol Lett ; 222(2): 180-8, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-23845848

ABSTRACT

Metabolic activation of polycyclic aromatic hydrocarbons (PAH) is mediated mainly by cytochrome P450 monooxygenases (CYP) CYP1A1, 1A2 and 1B1. Several PAH are known to induce these CYP via aryl hydrocarbon receptor (AhR) signaling. Recently, it was shown that the PAH benzo[a]pyrene (BaP) can induce CYP3A4 as well. The induction was suggested to be mediated by the pregnane X receptor (PXR) rather than AhR. Metabolism by CYP3A4 is only known for dihydrodiol metabolites of PAH but not for their parent compounds. In the present study, a CYP3A4 reporter gene assay, requiring the overexpression of PXR, was used to investigate whether the PAH parent compounds BaP, benzo[c]phenanthrene (BcP) and dibenzo[a,l]pyrene (DBalP) as well as their corresponding phase I metabolites, the respective dihydrodiols and diol epoxides, can induce CYP3A4 promoter activity. BaP, BcP and their dihydrodiols were found to significantly activate the CYP3A4 promoter. Moreover, activation of PXR by all four compounds was detected by using a PXR transactivation assay, supporting that PXR mediates CYP3A4 induction by PAH. Taken together, these results show that both PAH parent compounds as well as their phase I metabolites induce CYP3A4 promoter via the transcription factor PXR.


Subject(s)
Carcinogens, Environmental/toxicity , Cytochrome P-450 CYP3A/biosynthesis , Polycyclic Aromatic Hydrocarbons/toxicity , Promoter Regions, Genetic/drug effects , Receptors, Steroid/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Benzo(a)pyrene/metabolism , Benzo(a)pyrene/toxicity , Benzopyrenes/metabolism , Benzopyrenes/toxicity , Carcinogens, Environmental/metabolism , Cell Survival/drug effects , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Dihydroxydihydrobenzopyrenes/metabolism , Dihydroxydihydrobenzopyrenes/toxicity , Enzyme Induction/drug effects , Epoxy Compounds/metabolism , Epoxy Compounds/toxicity , Genes, Reporter/drug effects , HEK293 Cells , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Ligands , Polycyclic Aromatic Hydrocarbons/metabolism , Pregnane X Receptor , Receptors, Steroid/biosynthesis , Receptors, Steroid/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism
12.
Arch Toxicol ; 87(2): 269-80, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22899102

ABSTRACT

A wide variety of contaminants are ingested through food, among them the pro-carcinogenic polycyclic aromatic hydrocarbon benzo[a]pyrene (BP) that is resorbed and partially metabolized in the enterocytes of the small intestine. Previous in vitro studies have revealed that BP phenols are excreted as Phase II metabolites including glucuronides and sulfates. This export is mediated by the breast cancer resistance protein (ABCG2). The ultimate carcinogenic Phase I BP metabolite anti-BP-7,8-dihydrodiol-9,10-epoxide (BPDE) can be detoxified by glutathione conjugate formation catalyzed by glutathione S-transferases. In the present study, differentiated human intestinal Caco-2 cells were used as a model for the human small intestine to investigate the detoxification of BPDE and excretion of stereoisomeric glutathione conjugates in the presence of an inhibitor of the glutathione-cleaving enzyme γ-glutamyl transpeptidase at the cell surface. The results indicate that the glutathione conjugates of BPDE are formed and excreted mainly to the apical and to a minor extent to the basolateral side of polarized Caco-2 monolayers. Inhibition studies revealed that the multidrug resistance-associated proteins (ABCCs) are involved in the transport of BPDE glutathione conjugates. Stable ABCC1, ABCC2 and ABCC3 knockdown cell lines were generated, thus making it possible to demonstrate that ABCC1 mediates the basolateral and ABCC2 the apical excretion of BPDE glutathione conjugates. In conclusion, the ultimate carcinogen BPDE is detoxified via glutathione conjugation and subsequently excreted by Caco-2 cells in both apical and basolateral directions. This finding is equivalent to a transport into feces as well as blood system in the in vivo situation.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Caco-2 Cells/drug effects , Carcinogens/toxicity , Drug Resistance, Multiple/physiology , Glutathione/metabolism , Multidrug Resistance-Associated Proteins/metabolism , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/metabolism , Biological Transport/drug effects , Caco-2 Cells/metabolism , Carcinogens/metabolism , Gene Expression , Gene Knockdown Techniques , Glutathione Transferase/metabolism , Humans , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Time Factors
13.
Toxicology ; 279(1-3): 196-202, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21093526

ABSTRACT

The marine biotoxin okadaic acid (OA), produced by dinoflagellates, can accumulate in various bivalve molluscs. In humans, oral consumption of shellfish contaminated with OA induces acute toxic effects like diarrhea, nausea, vomiting and abdominal pain. However, tumorigenic and embryotoxic effects of OA have been also described. Current toxicokinetic studies with mice were performed with high cytotoxic oral doses leading presumably to a paracellular passage of OA through the gastrointestinal barrier. There are no studies available analyzing the absorption at low concentrations, which represent a realistic dietary exposure, making a reliable risk assessment difficult. Therefore, we performed a low-dose study using the human intestinal Caco-2 cell model to simulate the intestinal barrier. Low level exposure of 20-200 nM OA to the cell monolayer allows an only limited passage from the "luminal" to the "blood side". Furthermore, we could detect a significant efflux of OA, which led to the suggestion that active transport mechanisms are involved in the elimination process of OA. In conclusion, our results indicate that besides the well known defense mechanisms of humans against this marine biotoxin--vomiting and diarrhea--further detoxification mechanisms are available to limit the absorption of toxic OA.


Subject(s)
Intestinal Absorption , Marine Toxins/pharmacokinetics , Okadaic Acid/pharmacokinetics , Biological Availability , Biological Transport, Active , Caco-2 Cells , Dose-Response Relationship, Drug , Humans , Marine Toxins/administration & dosage , Okadaic Acid/administration & dosage , Tissue Distribution
14.
Toxicol Lett ; 197(2): 151-5, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20562004

ABSTRACT

All-trans retinoic acid (atRA) is the most active metabolite of vitamin A. It is a ligand of retinoic acid receptors (RAR) as well as of retinoid X receptors (RXR) and effectively stimulates the RAR/RXR signalling pathway. In this study effects of atRA on the detoxification of the food contaminant benzo[a]pyrene (B[a]P) was elucidated by using the Caco-2 cell line as model system for the human small intestine. Caco-2 cells express a number of phase I and II xenobiotic-metabolising enzymes as well as several transport proteins of the ATP-binding cassette (ABC) superfamily. Pre-treatment of the cells with atRA resulted in enhanced apical excretion of B[a]P-3-sulfate, a phase II metabolite of B[a]P. Gene expression analysis revealed that the Breast Cancer Resistance Protein (BCRP), an ABC-transporter known to be involved in B[a]P-3-sulfate excretion, was strongly stimulated already at low concentrations of atRA. Furthermore co-incubation of the intestinal cell with RAR agonist and RXR agonist resulted in a strong additive induction of mRNA expression of BCRP. Thus, atRA was shown to induce BCRP gene expression probably via the RAR/RXR signalling pathway, resulting in effective removal of B[a]P metabolites from intestinal cells.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Benzo(a)pyrene/metabolism , Gene Expression Regulation/drug effects , Neoplasm Proteins/metabolism , Tretinoin/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Benzoates/pharmacology , Caco-2 Cells , Humans , Molecular Sequence Data , Neoplasm Proteins/genetics , Receptors, Retinoic Acid/agonists , Retinoic Acid Receptor alpha , Retinoid X Receptors/agonists , Retinoids/pharmacology , Signal Transduction/drug effects , Tetrahydronaphthalenes/pharmacology , Tretinoin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...