Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Arch Toxicol ; 93(2): 369-383, 2019 02.
Article in English | MEDLINE | ID: mdl-30467583

ABSTRACT

The antihistaminic drug methapyrilene was withdrawn from the market in 1979 because of hepatocarcinogenicity in rats. Since then, the drug has been used as a model hepatotoxin especially for transcriptomic analyses using material from in vivo studies. Much less transcriptomics data are available from in vitro studies, and no studies have investigated proteomic effects of methapyrilene in vitro. Thus, the present study was aimed to characterize the proteomic response of primary rat hepatocytes to methapyrilene, to broaden our knowledge on the molecular mechanisms of methapyrilene toxicity, and to compare the results of collagen sandwich-cultured hepatocytes to in vivo data. In vitro methapyrilene concentrations (0.39 µM, 6.25 µM, and 100 µM) were chosen to cover an in vivo-relevant range. Based on published pharmacokinetic data they correspond to concentrations in portal vein blood for previously in vivo-tested doses of methapyrilene, up to a concentration showing slight cytotoxicity. Analysis of proteomic alterations by two-dimensional gel electrophoresis and mass-spectrometric protein identification demonstrated consistent and concentration-dependent effects of methapyrilene, in particular on mitochondrial proteins. Data suggest substantial deregulation of amino acid and ammonia metabolism and effects on mitochondrial energy supply pathways. The effects identified in vitro concur well with into previous in vivo observations. Several effects, for example, the influence of methapyrilene on S-adenosylmethionine metabolism, have not been described previously. The data suggest that already non-toxic concentrations of methapyrilene alter components of the intermediary metabolism, such as branched-chain amino acid metabolism, as well as urea and tricarboxylic cycle enzymes. In summary, data substantially add to our knowledge on molecular mechanisms of methapyrilene hepatotoxicity at the protein level.


Subject(s)
Hepatocytes/drug effects , Hepatocytes/metabolism , Methapyrilene/toxicity , Proteins/metabolism , Animals , Cell Culture Techniques/methods , Chemical and Drug Induced Liver Injury/metabolism , Computational Biology , Gene Expression Profiling , Histamine H1 Antagonists/toxicity , Male , Mass Spectrometry , Proteins/genetics , Proteomics/methods , Rats, Wistar , Toxicity Tests/methods
2.
Food Chem Toxicol ; 121: 214-223, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30157460

ABSTRACT

The increasing number of man-made chemicals in the environment that may pose a carcinogenic risk highlights the need for developing reliable time- and cost-effective approaches for carcinogen detection and identification. To address this issue, we investigated the utility of high-throughput microarray gene expression and next-generation genome-wide DNA methylation sequencing for the in vitro identification of genotoxic and non-genotoxic carcinogens. Terminally differentiated and metabolically competent human liver HepaRG cells were treated at minimally cytotoxic concentrations of (i) the genotoxic human liver carcinogen aflatoxin B1 (AFB1) and its structural non-carcinogenic analog aflatoxin B2 (AFB2); (ii) the genotoxic human lung carcinogen benzo[a]pyrene (B[a]P) and its non-carcinogenic isomer benzo[e]pyrene (B[e]P); and (iii) the non-genotoxic liver carcinogen methapyrilene for 72 h and transcriptomic and DNA methylation profiles were examined. Treatment of HepaRG cells with the liver carcinogens AFB1 and methapyrilene generated distinct gene-expression profiles, whereas B[a]P had only a slight effect on gene expression. In contrast to transcriptomic alterations, treatment of HepaRG cells with the carcinogenic and non-carcinogenic chemicals resulted in profound changes in the DNA methylation footprint; however, the correlation between gene-specific DNA methylation and gene expression changes was minimal. Among the carcinogen-altered genes, transferrin (TF) emerged as sensitive marker for an initial screening of chemicals for their potential liver carcinogenicity. Potential liver carcinogens (i.e., chemicals causing altered TF gene expression) could then be subjected to gene-expression analyses to differentiate genotoxic from non-genotoxic liver carcinogens. This approach may substantially enhance the identification and assessment of potential liver carcinogens.


Subject(s)
Aflatoxin B1/toxicity , Benzo(a)pyrene/toxicity , Methapyrilene/toxicity , Cell Line , DNA Adducts , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Hepatocytes , Humans , Nucleic Acid Amplification Techniques , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells , Transcription, Genetic/drug effects , Transcriptome
3.
Toxicol Lett ; 281: 65-73, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-28935588

ABSTRACT

The liver, a central detoxification organ and main regulator of systemic iron homeostasis, is prone to damage by xenobiotics. In the present study, we investigated the effect of the hepatotoxicant and hepatocarcinogen methapyrilene hydrochloride on iron metabolism in rat liver in a repeat-dose in vivo toxicity study and in human HepaRG cells in vitro. Treatment of male Fischer 344 (F344) rats with methapyrilene at doses 40 and 80mg/kg body weight (bw)/day by gavage for 6 weeks resulted in changes in the expression of classic hepatotoxicity-related marker genes and iron homeostasis-related genes, especially a prominent, dose-dependent down-regulation of the transferrin (Tf) gene and an up-regulation of the ferritin, light chain (Ftl) gene. A decrease in the level of TF and an increase in the level of FTL also occurred in methapyrilene-treated differentiated HepaRG cells, indicating the existence of interspecies and in vitro-in vivo similarities in the disturbance of cellular iron homeostasis upon liver injury. In contrast, there was minimal overlap in the expression of liver toxicity-marker genes in the livers of rats and in HepaRG cells treated with methapyrilene. Importantly, the decrease of transferrin at mRNA and protein levels occurred after the treatment with a low dose of methapyrilene that exhibited minimal cytotoxicity. These results demonstrate the significance of the dysregulation of hepatic iron metabolism in the pathogenesis and mechanism of chemical-induced liver toxicity and suggest that these changes may be sensitive and useful indicators of potentially hepatotoxic chemicals.


Subject(s)
Iron/metabolism , Liver/drug effects , Methapyrilene/toxicity , Animals , Cell Line , Dose-Response Relationship, Drug , Down-Regulation , Ferritins/genetics , Ferritins/metabolism , Genetic Markers , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Transferrin/genetics , Transferrin/metabolism , Up-Regulation
4.
Toxicology ; 386: 1-10, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28529062

ABSTRACT

Recently, bile acids (BAs) were reported as promising markers for drug-induced liver injury (DILI). BAs have been suggested to correlate with hepatocellular and hepatobiliary damage; however a clear connection of BA patterns with different types of DILI remains to be established. To investigate if BAs can improve the assessment of liver injury, 20 specific BAs were quantitatively profiled via LC-MS/MS in plasma and liver tissue in a model of methapyrilene-induced liver injury in rats. Methapyrilene, a known hepatotoxin was dosed daily over 14-days at doses of 30 and 80mg/kg, followed by a recovery phase of 10days. Conventional preclinical safety endpoints were related to BA perturbations and to hepatic gene expression profiling for a mechanistic interpretation of effects. Histopathological signs of hepatocellular and hepatobiliary damage with significant changes of clinical chemistry markers were accompanied by significantly increased levels of indivdual BAs in plasma and liver tissue. BA perturbations were already evident at the earliest time point after 30mg/kg treatment, and thereby indicating better sensitivity than clinical chemistry parameters. Furthermore, the latter markers suggested recovery of liver injury, whereas BA levels in plasma and liver remained significantly elevated during the recovery phase, in line with persistent histopathological findings of bile duct hyperplasia (BDH) and bile pigment deposition. Gene expression profiling revealed downregulation of genes involved in BA synthesis (AMACR, BAAT, ACOX2) and hepatocellular uptake (NTCP, OATs), and upregulation for efflux transporters (MRP2, MRP4), suggesting an adaptive hepatocellular protection mechanism against cytotoxic bile acid accumulation. In summary, our data suggests that specific BAs with high reliability such as cholic acid (CA) and chenodeoxycholic acid (CDCA) followed by glycocholic acid (GCA), taurocholic acid (TCA) and deoxycholic acid (DCA) can serve as additional biomarkers for hepatocellular/hepatobiliary damage in the liver in rat toxicity studies.


Subject(s)
Bile Acids and Salts/metabolism , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Methapyrilene/toxicity , Animals , Chemical and Drug Induced Liver Injury/pathology , Chromatography, Liquid , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Liver/pathology , Male , Methapyrilene/administration & dosage , Rats , Rats, Wistar , Reproducibility of Results , Tandem Mass Spectrometry , Up-Regulation/drug effects
5.
Toxicology ; 386: 120-132, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28552552

ABSTRACT

Lipid dysregulation is a common hepatic adverse outcome after exposure to toxic drugs and chemicals. A donor-free rat hepatocyte-like (B-13/H) cell was therefore examined as an in vitro model for investigating mechanisms. The B-13/H cell irreversibly accumulated triglycerides (steatosis) in a time- and dose-dependent manner when exposed to fatty acids, an effect that was potentiated by the combined addition of hyperglycaemic levels of glucose and insulin. B-13/H cells also expressed the LXR nuclear receptors and exposure to their activators - T0901317 or GW3965 - induced luciferase expression from a transfected LXR-regulated reporter gene construct and steatosis in a dose-dependent manner with T0901317. Exposing B-13/H cells to a variety of cationic amphiphilic drugs - but not other hepatotoxins - also resulted in a time- and dose-dependent accumulation of phospholipids (phospholipidosis), an effect that was reduced by over-expression of lysosomal phospholipase A2. Through application of this model, hepatotoxin methapyrilene exposure was shown to induce phospholipidosis in both B-13 and B-13/H cells in a time- and dose-dependent manner. However, methapyrilene was only toxic to B-13/H cells and inhibitors of hepatotoxicity enhanced phospholipidosis, suggesting phospholipidosis is not a pathway in toxicity for this withdrawn drug. In contrast, pre-existing steatosis had minimal effect on methapyrilene hepatotoxicity in B-13/H cells. These data demonstrate that the donor free B-13 cell system for generating hepatocyte-like cells may be employed in studies of fatty acid- and LXR activator-induced steatosis and phospholipidosis and in the dissection of pathways leading to adverse outcomes such as hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/physiopathology , Fatty Acids/metabolism , Hepatocytes/drug effects , Lipid Metabolism/drug effects , Triglycerides/metabolism , Animals , Benzoates/administration & dosage , Benzoates/toxicity , Benzylamines/administration & dosage , Benzylamines/toxicity , Cell Line , Chemical and Drug Induced Liver Injury/etiology , Dose-Response Relationship, Drug , Fatty Liver/metabolism , Hepatocytes/metabolism , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/toxicity , Liver X Receptors/metabolism , Methapyrilene/administration & dosage , Methapyrilene/toxicity , Phospholipids/metabolism , Rats , Sulfonamides/administration & dosage , Sulfonamides/toxicity , Time Factors
6.
Toxicol Sci ; 156(1): 190-198, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28013212

ABSTRACT

Continuous lifetime exposure to certain natural and man-made chemicals is a major cause of cancers in humans; therefore, evaluating the carcinogenic risks of chemicals remains important. Currently, substantial progress has been made in identification of genotoxic carcinogens; in contrast, predicting the carcinogenic potential of nongenotoxic compounds is a challenge due to many different modes of action that may lead to tumorigenesis. In the present study, we investigated the effects of the nongenotoxic liver carcinogen methapyrilene and the nongenotoxic noncarcinogen usnic acid, at doses that do not exhibit organ cytotoxicity, on epigenomic alterations in the livers and kidneys of Fischer 344 (F344) rats. We demonstrate that a repeat-dose oral treatment of male F344 rats with methapyrilene for 6 weeks caused target organ-specific epigenetic alterations in the livers. In contrast, only very slight epigenetic changes were found in the livers of F344 rats treated with hepatotoxicant, but noncarcinogen, usnic acid. The methapyrilene-induced epigenetic changes consisted of changes in histone lysine acetylation and methylation, with the greatest increase occurring in global and gene-specific histone H3 lysine 9 (H3K9) deacetylation. Importantly, the results of the present study show an association between gene-specific histone H3K9 deacetylation and a reduced expression of critical cancer-related genes, including prospero homeobox 1 (Prox1), HNF1 homebox A (Hnf1a), and peroxisome proliferator activated receptor alpha (Ppara), which provides a mechanistic link between methapyrilene-induced epigenetic aberrations and liver carcinogenesis.


Subject(s)
Carcinogens/toxicity , Chromatin Assembly and Disassembly/drug effects , Epigenesis, Genetic/drug effects , Histones/metabolism , Liver/drug effects , Methapyrilene/toxicity , Protein Processing, Post-Translational/drug effects , Acetylation/drug effects , Administration, Oral , Animals , Benzofurans/administration & dosage , Benzofurans/toxicity , Carcinogens/administration & dosage , Dose-Response Relationship, Drug , Hepatocyte Nuclear Factor 1-alpha/antagonists & inhibitors , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kidney/drug effects , Kidney/metabolism , Liver/metabolism , Lysine/metabolism , Male , Methapyrilene/administration & dosage , Methylation/drug effects , Organ Specificity , PPAR alpha/antagonists & inhibitors , PPAR alpha/genetics , PPAR alpha/metabolism , Random Allocation , Rats, Inbred F344 , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
J Appl Toxicol ; 36(9): 1194-206, 2016 09.
Article in English | MEDLINE | ID: mdl-26946349

ABSTRACT

Zebrafish phenotypic assays have shown promise to assess human hepatotoxicity, though scoring of liver morphology remains subjective and difficult to standardize. Liver toxicity in zebrafish larvae at 5 days was assessed using gene expression as the biomarker approach, complementary to phenotypic analysis and analytical data on compound uptake. This approach aimed to contribute to improved hepatotoxicity prediction, with the goal of identifying biomarker(s) as a step towards the development of transgenic models for prioritization. Morphological effects of hepatotoxic compounds (acetaminophen, amiodarone, coumarin, methapyrilene and myclobutanil) and saccharin as the negative control were assessed after exposure in zebrafish larvae. The hepatotoxic compounds induced the expected zebrafish liver degeneration or changes in size, whereas saccharin did not have any phenotypic adverse effect. Analytical methods based on liquid chromatography-mass spectrometry were optimized to measure stability of selected compounds in exposure medium and internal concentration in larvae. All compounds were stable, except amiodarone for which precipitation was observed. There was a wide variation between the levels of compound in the zebrafish larvae with a higher uptake of amiodarone, methapyrilene and myclobutanil. Detection of hepatocyte markers (CP, CYP3A65, GC and TF) was accomplished by in situ hybridization of larvae to coumarin and myclobutanil and confirmed by real-time reverse transcription-quantitative polymerase chain reaction. Experiments showed decreased expression of all markers. Next, other liver-specific biomarkers (i.e. FABP10a and NR1H4) and apoptosis (i.e. CASP-3 A and TP53) or cytochrome P450-related (CYP2K19) and oxidoreductase activity-related (ZGC163022) genes, were screened. Links between basic mechanisms of liver injury and results of biomarker responses are described. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Genetic Markers , Liver/drug effects , Zebrafish/genetics , Acetaminophen/toxicity , Amiodarone/toxicity , Animals , Apoptosis/drug effects , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Coumarins/toxicity , Female , Gene Expression Regulation , Genome-Wide Association Study , Humans , In Situ Hybridization , Larva/drug effects , Larva/genetics , Liver/metabolism , Male , Methapyrilene/toxicity , Nitriles/toxicity , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Phenotype , Toxicity Tests , Transferrin/genetics , Transferrin/metabolism , Triazoles/toxicity , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
8.
J Toxicol Sci ; 40(6): 855-71, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26558467

ABSTRACT

We aimed to clarify the hepatocarcinogen-specific disruption of cell cycle checkpoint functions and its time course after repeated administration of hepatocarcinogens. Thus, rats were repeatedly administered with hepatocarcinogens (methapyrilene, carbadox and thioacetamide), a marginal hepatocarcinogen (leucomalachite green), hepatocarcinogenic promoters (oxfendazole and ß-naphthoflavone) or non-carcinogenic hepatotoxicants (promethazine and acetaminophen) for 7, 28 or 90 days, and the temporal changes in cell proliferation, expression of G1/S and spindle checkpoint-related molecules, and apoptosis were examined using immunohistochemistry and/or real-time RT-PCR analysis. Hepatocarcinogens facilitating cell proliferation at day 28 of administration also facilitated cell proliferation and apoptosis at day 90. Hepatocarcinogen- or hepatocarcinogenic promoter-specific cellular responses were not detected by immunohistochemical single molecule analysis even after 90 days. Expression of Cdkn1a, Mad2l1, Chek1 and Rbl2 mRNA also lacked specificity to hepatocarcinogens or hepatocarcinogenic promoters. In contrast, all hepatocarcinogens and the marginally hepatocarcinogenic leucomalachite green induced Mdm2 upregulation or increase in the number of phosphorylated MDM2(+) cells from day 28, irrespective of the lack of cell proliferation facilitation by some compounds. However, different Tp53 expression levels suggest different mechanisms of induction or activation of MDM2 among hepatocarcinogens. On the other hand, hepatocarcinogenic methapyrilene and carbadox downregulated the number of both ubiquitin D(+) cells and proliferating cells remaining in M phase at day 28 and/or day 90, irrespective of the lack of cell proliferation facilitation in the latter. These results suggest that hepatocarcinogens disrupt spindle checkpoint function after 28 or 90 days of administration, which may be induced ahead of cell proliferation facilitation.


Subject(s)
Carbadox/toxicity , Carcinogens/administration & dosage , Carcinogens/toxicity , Cell Proliferation/drug effects , Liver/cytology , M Phase Cell Cycle Checkpoints/drug effects , Methapyrilene/toxicity , Thioacetamide/toxicity , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzimidazoles/administration & dosage , Benzimidazoles/toxicity , Carbadox/administration & dosage , Cell Proliferation/genetics , Checkpoint Kinase 1 , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression/drug effects , Male , Methapyrilene/administration & dosage , Protein Kinases/genetics , Protein Kinases/metabolism , Rats, Inbred F344 , Rosaniline Dyes/toxicity , Time Factors , Ubiquitins/genetics , Ubiquitins/metabolism
9.
J Toxicol Sci ; 39(5): 785-94, 2014.
Article in English | MEDLINE | ID: mdl-25242409

ABSTRACT

We previously reported a toxicogenomics-based prediction model for hepatocarcinogens in which the expression patterns of signature genes following repeated doses of either genotoxic or non genotoxic compounds were similar. Based on the results of our prediction model, we hypothesized that repeated doses of non-genotoxic carcinogens might have initiating potential. Here, we conducted a two stage hepatocarcinogenesis study in rats exposed to the initiating agent nitrosodiethylamine (DEN), and hepatotoxic compounds thioacetamide (TAA), methapyrilene (MP) and acetaminophen (APAP) for 1-2 weeks followed by the liver tumor promoter phenobarbital (PB). The duration of initial treatment was determined based on positive results from our prediction model. Combined treatment of 3 or 30 mg/kg of genotoxic DEN and PB induced marked increases in altered hepatocellular foci and a DEN dose-dependent increase in the number and area of glutathione S-transferase-placental form (GST-P)-positive foci. A low number of altered hepatocellular foci were also observed in rats treated with TAA at a dose of 45 mg/kg.MP at a dose of 100 mg/kg induced a very low number of foci, but APAP did not. Hierarchical clustering analysis using gene expression data revealed that 2-week treatment with TAA at a dose of 30 mg/kg and MP at 45 mg/kg induced specific expression of DNA damage-related genes, similar to 1-week treatment with DEN at a dose of 30 mg/kg. These results suggest that TAA and MP induce DNA damage, which partially supports our hypothesis. Although this study does not indicate whether tumor growth in response to these compounds can be assessed in this model, our results suggest that cumulative treatment with non genotoxic TAA might have initiating potential in the liver.


Subject(s)
Carcinoma, Hepatocellular/chemically induced , Liver Neoplasms/chemically induced , Methapyrilene/toxicity , Mutagenicity Tests/methods , Thioacetamide/toxicity , Acetaminophen/toxicity , Animals , Carcinoma, Hepatocellular/genetics , DNA Damage/drug effects , DNA Damage/genetics , Diethylnitrosamine/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , Liver Neoplasms/genetics , Male , Oxidative Stress/genetics , Phenobarbital/toxicity , Rats, Sprague-Dawley
10.
Arch Toxicol ; 87(2): 337-45, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23274394

ABSTRACT

Cultivated hepatocytes represent a well-established in vitro system. However, the applicability of hepatocytes in toxicogenomics is still controversially discussed. Recently, an in vivo/in vitro discrepancy has been described, whereby the non-genotoxic rat liver carcinogen methapyrilene alters the expression of the metabolizing genes SULT1A1 and ABAT, as well as the DNA damage response gene GADD34 in vitro, but not in vivo. If the collagen sandwich cultures of hepatocytes really produce false-positive data, this would compromise its application in toxicogenomics. To revisit the putative in vivo/in vitro discrepancy, we first analyzed and modeled methapyrilene concentrations in the portal vein of rats. The relatively short half-life of 2.8 h implies a rapid decrease in orally administered methapyrilene in vivo below concentrations that can cause gene expression alterations. This corresponded to the time-dependent alteration levels of GADD34, ABAT and SULT1A1 RNA in the liver: RNA levels are altered 1, 6 and 12 h after methapyrilene administration, but return to control levels after 24 and 72 h. In contrast, methapyrilene concentrations in the culture medium supernatant of primary rat hepatocyte cultures decreased slowly. This explains why GADD34, ABAT and SULT1A1 were still deregulated after 24 h exposure in vitro, but not in vivo. It should also be considered that the earliest analyzed time point in the previous in vivo studies was 24 h after methapyrilene administration. In conclusion, previously observed in vitro/in vivo discrepancy can be explained by different pharmacokinetics present in vitro and in vivo. When the in vivo half-life is short, levels of some initially altered genes may have returned to control levels already 24 h after administration.


Subject(s)
Carcinogens/pharmacokinetics , Gene Expression Regulation, Neoplastic/drug effects , Hepatocytes/drug effects , Liver/drug effects , Methapyrilene/pharmacokinetics , 4-Aminobutyrate Transaminase/genetics , Animals , Antigens, Differentiation/genetics , Arylsulfotransferase/genetics , Carcinogens/toxicity , Cells, Cultured , Half-Life , Hepatocytes/metabolism , Liver/metabolism , Male , Methapyrilene/toxicity , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
11.
Toxicology ; 290(2-3): 249-57, 2011 Dec 18.
Article in English | MEDLINE | ID: mdl-22015589

ABSTRACT

The genotoxicity of methapyrilne (MP) has been evaluated in a number of assays since it was found to be a rat hepatocarcinogen with subsequent withdrawal as an over-the-counter antihistamine. Whilst it has not been classified as a genotoxin, there are reports of positive findings from mammalian cell gene mutation and transformation assays. To investigate further the genotoxic potential of MP, the alkaline Comet assay was used to evaluate DNA damage both in primary hepatocytes in culture and in vivo in the rat. To confirm bioactivation was required to induce the hepatotoxic mechanism, aminobenzotriazole, a broad spectrum cytochrome P450 enzyme inhibitor was used as a pre-treatment. The levels of glutathione and glutathione disulfide were determined in both hepatocytes in culture and in the liver following in vivo exposure. MP showed significant increases in DNA damage in freshly isolated male rat hepatocyte suspensions that could be significantly reduced by pre-incubation of aminobenzotriazole (ABT). DNA damage showed a marked sex difference, with male hepatocytes being more susceptible, and showing a concurrent depletion of glutathione (GSH) compared with female hepatocytes. Modulation of the GSH levels by diethylmaleate and γ-glutamylcysteinylethyl ester, elevated and reduced the levels of DNA damage, respectively. In the in vivo Comet assay, there was no evidence of DNA damage following MP (150mg/kg p.o) treatment for three consecutive days, although histological and liver enzyme changes were seen. Total protein GSH content was elevated in MP-treated animals and superoxide dismutase levels were increased specifically in periportal regions. Taken together, these data support the potential for MP to induce oxidative stress. The differences in DNA damage detected by the Comet assay in vitro, and in rat liver in vivo, could be attributed to differences in metabolism and response to oxidant insult or the inability of the assay to discriminate damage in a small number of individual cells in the whole liver.


Subject(s)
DNA Damage/drug effects , Hepatocytes/drug effects , Histamine H1 Antagonists/toxicity , Methapyrilene/toxicity , Oxidative Stress/drug effects , Animals , Comet Assay , Female , Glutathione/drug effects , Glutathione/metabolism , Hepatocytes/pathology , Liver/drug effects , Liver/pathology , Male , Mutagenicity Tests , Rats , Rats, Wistar , Sex Factors , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Triazoles/pharmacology
12.
J Pharm Biomed Anal ; 54(5): 1128-35, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21176868

ABSTRACT

Ophthalmic acid (OA), an endogenous tripeptide analogue of glutathione, has been suggested as a potential biomarker for paracetamol/acetaminophen hepatotoxicity. Here HPLC-MS/MS methods have been developed for the precise, sensitive and specific detection and quantification of OA in in vitro cell culture medium and plasma. For the cell culture medium the LLOQ was found to be 1 ng/ml, with less than 1% between sample carry over at all concentrations and precision below 15% for within day and below 9% for between day analyses. For rat plasma the presence of endogenous OA resulted in the LLOQ being 25 ng/ml (defined as the lowest concentration on the calibration curve where the base peak was less than 20% of the LLOQ). For the plasma assay the percentage carry over was less than 1% for all concentrations and within and between batch precision was below 21%. The methods were linear for both sample types from the LLOQ up to 5 µg/ml. The method was successfully applied to the determination of OA in samples obtained following the chronic administration of the rat hepatotoxin methapyrilene, where plasma OA concentrations were observed to show a weak negative correlation with those of established liver injury biomarkers such as aspartate aminotransferase (AST).


Subject(s)
Chromatography, High Pressure Liquid/methods , Culture Media/analysis , Epithelial Cells/metabolism , Liver/metabolism , Oligopeptides/blood , Tandem Mass Spectrometry/methods , Animals , Biomarkers/blood , Cell Line , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Epithelial Cells/cytology , Humans , Limit of Detection , Liver/cytology , Male , Methapyrilene/toxicity , Oligopeptides/analysis , Rats , Reproducibility of Results
13.
Regul Toxicol Pharmacol ; 58(3): 369-81, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20801182

ABSTRACT

In order to determine a threshold for nongenotoxic carcinogens, the traditional risk assessment approach has been to identify a mode of action (MOA) with a nonlinear dose-response. The dose-response for one or more key event(s) linked to the MOA for carcinogenicity allows a point of departure (POD) to be selected from the most sensitive effect dose or no-effect dose. However, this can be challenging because multiple MOAs and key events may exist for carcinogenicity and oftentimes extensive research is required to elucidate the MOA. In the present study, a microarray analysis was conducted to determine if a POD could be identified following short-term oral rat exposure with two nongenotoxic rodent carcinogens, fenofibrate and methapyrilene, using a benchmark dose analysis of genes aggregated in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) biological processes, which likely encompass key event(s) for carcinogenicity. The gene expression response for fenofibrate given to rats for 2days was consistent with its MOA and known key events linked to PPARα activation. The temporal response from daily dosing with methapyrilene demonstrated biological complexity with waves of pathways/biological processes occurring over 1, 3, and 7days; nonetheless, the benchmark dose values were consistent over time. When comparing the dose-response of toxicogenomic data to tumorigenesis or precursor events, the toxicogenomics POD was slightly below any effect level. Our results suggest that toxicogenomic analysis using short-term studies can be used to identify a threshold for nongenotoxic carcinogens based on evaluation of potential key event(s) which then can be used within a risk assessment framework.


Subject(s)
Carcinogens/toxicity , Fenofibrate/toxicity , Methapyrilene/analysis , Methapyrilene/toxicity , Neoplasms/chemically induced , Toxicogenetics/methods , Animals , Carcinogens/administration & dosage , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Dose-Response Relationship, Drug , Female , Fenofibrate/administration & dosage , Gene Expression , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Methapyrilene/administration & dosage , Neoplasms/genetics , No-Observed-Adverse-Effect Level , Oligonucleotide Array Sequence Analysis , Rats , Risk Assessment
14.
Toxicol Appl Pharmacol ; 239(3): 297-305, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19523481

ABSTRACT

UNLABELLED: Methapyrilene, [N,N-dimethyl-N'-pyridyl-N'(2-thienylmethyl)-1,2-ethanediamine] (MP) was withdrawn from, clinical use due to reported periportal hepatic necrosis and hepatocarcinogenicity in the rat, via S-oxidation of the thiophene group. In this study MP is used as a model hepatotoxin to further characterise the functional consequences of S-oxidation of the thiophene group in vivo, in rat models and in vitro, in freshly isolated rat hepatocyte suspensions. In vivo histological studies revealed the early depletion of glutathione (GSH), which was confined to the damaged periportal area, in contrast to an increase in GSH levels in the centrilobular region. Additionally, the induction of cell defence was demonstrated by an increase in the protein levels of heme-oxygenase 1 (HO-1) and glutamate cysteine ligase, catalytic subunit (GCLC) in vivo. Histological examination demonstrated that cytotoxicity progresses initially via apoptosis before an increase in necrosis over the 3-day administration. An apoptotic-like mechanism was observed in vitro via the measurement of cytochrome c release and caspase activation. CONCLUSION: This study provides evidence for a complex pathway of MP-induced hepatotoxicity which progresses through early adaptation, apoptosis, necrosis and inflammation, all underpinned by the zonal induction and depletion of GSH within the liver.


Subject(s)
Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Methapyrilene/toxicity , Thiophenes/metabolism , Animals , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Glutamate-Cysteine Ligase/biosynthesis , Glutathione/metabolism , Heme Oxygenase-1/biosynthesis , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/metabolism , Liver/pathology , Male , Methapyrilene/pharmacokinetics , Necrosis , Oxidation-Reduction , Rats , Rats, Wistar
15.
Arch Toxicol ; 82(12): 923-31, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18987846

ABSTRACT

Recent studies have presented evidence that in vivo obtained gene expression data can be used for carcinogen classification, for instance to differentiate between genotoxic and non-genotoxic carcinogens. However, although primary rat hepatocytes represent a well-established in vitro system for drug metabolism and enzyme induction, they have not yet been systematically optimized for toxicogenomic studies. The latter may be confounded by the fact that cultured hepatocytes show strong spontaneous alterations in gene expression patterns. Therefore, we addressed the following questions: (1) which culture system is optimal, comparing sandwich, Matrigel and 2D cultures, (2) how critical is the impact of culture period on substance-induced alterations in gene expression and (3) do these substance-induced alterations in cultured hepatocytes occur already at in vivo relevant concentrations? For this purpose we analyzed the expression of four genes, namely Abat, Gsk3beta, Myd116 and Sult1a1 that recently have been reported to be influenced by the antihistamine and non-genotoxic carcinogen methapyrilene (MPy). The most reproducible effects of MPy were observed in sandwich cultures. Induction factors of Gsk3beta and Myd116 at 100 microM MPy were 2 and 4 (medians), respectively, whereas expression of Abat and Sult1a1 were inhibited by factors of 7 and 5, respectively. Similar results were observed in hepatocytes maintained for 24 h or 3 weeks in sandwich culture with respect to the influence of MPy on the expression of Abat, Gsk3beta, Myd116 and Sult1a1. To determine whether MPy influences gene expression at in vivo relevant concentrations, 3.5 mg/kg MPy were administered to male Wistar rats intraperitoneally, resulting in plasma concentrations ranging between 1.72 and 0.32 microM 5 and 80 min after injection. Inhibition of Abat and Sult1a1 expression in vitro already occurred at in vivo relevant concentrations of 0.39 microM MPy. Induction of Myd116 was observed at 6.25 microM which is higher but in the same order of magnitude as in vivo relevant concentrations. In conclusion, the presented data strongly suggest that sandwich cultures are most adequate for detection of MPy-induced gene expression alterations and the effect of MPy was detected at in vivo relevant concentrations.


Subject(s)
Cell Culture Techniques/methods , Collagen/drug effects , Gene Expression/drug effects , Hepatocytes/drug effects , Laminin/drug effects , Methapyrilene/toxicity , Proteoglycans/drug effects , Animals , Antigens, Differentiation/metabolism , Arylsulfotransferase/metabolism , Carcinogens/toxicity , Cells, Cultured , Culture Media, Serum-Free , Dose-Response Relationship, Drug , Drug Combinations , Extracellular Matrix/drug effects , Extracellular Matrix/enzymology , Hepatocytes/enzymology , Hepatocytes/metabolism , Male , Methapyrilene/blood , Proto-Oncogene Proteins/metabolism , Rats , Rats, Wistar , Repressor Proteins/metabolism , Time Factors , Toxicogenetics
16.
Toxicology ; 250(1): 15-26, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18619722

ABSTRACT

For assessing carcinogenicity in animals, it is difficult and costly, an alternative strategy has been desired. We explored the possibility of applying a toxicogenomics approach by using comprehensive gene expression data in rat liver treated with various compounds. As prototypic non-genotoxic hepatocarcinogens, thioacetamide (TAA) and methapyrilene (MP) were selected and 349 commonly changed genes were extracted by statistical analysis. Taking both compounds as positive with six compounds, acetaminophen, aspirin, phenylbutazone, rifampicin, alpha-naphthylisothiocyanate, and amiodarone as negative, prediction analysis of microarray (PAM) was performed. By training and 10-fold cross validation, a classifier containing 112 probe sets that gave an overall success rate of 95% was obtained. The validity of the present discriminator was checked for 30 chemicals. The PAM score showed characteristic time-dependent increases by treatment with several non-genotoxic hepatocarcinogens, including TAA, MP, coumarin, ethionine and WY-14643, while almost all of the non-carcinogenic samples were correctly predicted. Measurement of hepatic glutathione content suggested that MP and TAA cause glutathione depletion followed by a protective increase, but the protective response is exhausted during repeated administration. Therefore, the presently obtained PAM classifier could predict potential non-genotoxic hepatocarcinogenesis within 24 h after single dose and the inevitable pseudo-positives could be eliminated by checking data of repeated administrations up to 28 days. Tests for carcinogenicity using rats takes at least 2 years, while the present work suggests the possibility of lowering the time to 28 days with high precision, at least for a category of non-genotoxic hepatocarcinogens causing oxidative stress.


Subject(s)
Carcinogenicity Tests/methods , Carcinogens/toxicity , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Toxicogenetics/methods , Animals , Gene Expression/drug effects , Glutathione/drug effects , Glutathione/metabolism , Liver/metabolism , Liver Neoplasms, Experimental/genetics , Male , Methapyrilene/toxicity , Microarray Analysis , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Thioacetamide/toxicity , Time Factors
17.
J Pharmacol Exp Ther ; 326(2): 657-71, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18451316

ABSTRACT

Methapyrilene (MP), a 2-thiophene H(1)-receptor antagonist, is a model toxicant in the genomic and proteomic analyses of hepatotoxicity. In rats, it causes an unusual periportal necrosis that is hypothetically attributed to chemically reactive and cytotoxic metabolites. We have characterized the bioactivation of MP by hepatic microsomes and primary rat hepatocytes, and we established a possible causal linkage with cytotoxicity. Methapyrilene tritiated at C-2 of the diaminoethane moiety ([3H]MP) was metabolized via an NADPH-dependent pathway to intermediates that combined irreversibly with microsomes (rat > mouse approximately human). This binding was attenuated by the cytochrome P450 (P450) inhibitor 1-aminobenzotriazole and thiols but not by trapping agents for iminium ions and aldehydes. Reactive intermediates were trapped as thioether adducts of monooxygenated MP. Mass spectrometric and hydrogen/deuterium exchange analysis of the glutathione adduct produced by rat liver microsomes indicated that the metabolite was most probably a thioether of MP S-oxide substituted in the thiophene ring. The glutathione adduct was formed by rat hepatocytes and eliminated in bile by rats administered [3H]MP intravenously. MP produced concentration- and time-dependent cytotoxicity, depleted glutathione, and underwent irreversible binding to the hepatocytes before a significant increase in cell damage was observed. P450 inhibitors reduced turnover of the drug, production of the glutathione adduct, irreversible binding, and cytotoxicity but inhibited glutathione depletion selectively. MP underwent lesser turnover and bioactivation in mouse hepatocytes and was not cytotoxic. Analogs with phenyl and p-methoxyphenyl rings were much less hepatocytotoxic than MP. Hepatotoxicity in rats was diminished by predosing with 1-aminobenzotriazole. For the first time, a thiophene ring substituent is identified as a bioactivation-dependent toxicophore in hepatocytes.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , Histamine H1 Antagonists , Methapyrilene , Microsomes, Liver/drug effects , Thiophenes/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chromatography, High Pressure Liquid , Glutathione/metabolism , Hepatocytes/metabolism , Histamine H1 Antagonists/chemistry , Histamine H1 Antagonists/pharmacokinetics , Histamine H1 Antagonists/toxicity , Humans , Male , Methapyrilene/chemistry , Methapyrilene/pharmacokinetics , Methapyrilene/toxicity , Mice , Mice, Inbred Strains , Microsomes, Liver/metabolism , Molecular Structure , Rats , Rats, Wistar , Thiophenes/chemistry
18.
J Toxicol Sci ; 33(1): 37-50, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18303183

ABSTRACT

The present study was conducted as a model case of the toxicogenomics approach for analyzing toxicological mechanisms and toxicity assessments in the early stage of drug development by comparing with classical toxicology data. Methapyrilene (MP) 100 mg/kg produced obvious histopathological changes in liver of rats by single or repeated dose up to 28 days with significant elevation of ALT and AST. In the middle dose groups (30 mg/kg MP), no apparent changes were noted in blood biochemical data by single dosing or repeated dosing up to one week, and no obvious histopathological changes were observed except a slight hypertrophy in the hepatocytes. Comprehensive gene expression changes were analyzed using Affymetrix GeneChip and differentially expressed probe sets were statistically extracted. These contained many genes related to "glutathione metabolism", "apoptosis", "MAPK signaling pathway" and "regulation of cell cycle", which were all thought to be involved in the development of presently observed phenotypes. In the high dose groups, TGP1 scores (developed in our system in order to overview the responsiveness of drugs to multiple marker gene lists) for these categories were markedly increased from the early time point after single dose and kept their high expression throughout the repeated dose period. In the middle dose groups, the increment of the scores were noted not only at the time points when apparent pathological changes emerged, but also at the earlier stage of repeated dosing and even after single dosing. We conclude that toxicogenomics would enable a more sensitive assessment at the earlier time point than classical toxicology evaluation.


Subject(s)
Gene Expression Profiling , Liver Diseases/genetics , Methapyrilene/toxicity , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Cell Cycle/drug effects , Chemical and Drug Induced Liver Injury , Gene Expression Regulation/drug effects , Glutathione/metabolism , Liver Diseases/blood , Liver Diseases/pathology , Male , Mitogen-Activated Protein Kinases/metabolism , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Toxicogenetics
19.
Environ Health Perspect ; 115(4): 572-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17450226

ABSTRACT

BACKGROUND: Toxicogenomics experiments often reveal thousands of transcript alterations that are related to multiple processes, making it difficult to identify key gene changes that are related to the toxicity of interest. OBJECTIVES: The objective of this study was to compare gene expression changes in a nontarget tissue to the target tissue for toxicity to help identify toxicity-related genes. METHODS: Male rats were given the hepatotoxicant methapyrilene at two dose levels, with livers and kidneys removed 24 hr after one, three, and seven doses for gene expression analysis. To identify gene changes likely to be related to toxicity, we analyzed genes on the basis of their temporal pattern of change using a program developed at the National Institute of Environmental Health Sciences, termed "EPIG" (extracting gene expression patterns and identifying co-expressed genes). RESULTS: High-dose methapyrilene elicited hepatic damage that increased in severity with the number of doses, whereas no treatment-related lesions were observed in the kidney. High-dose methapyrilene elicited thousands of gene changes in the liver at each time point, whereas many fewer gene changes were observed in the kidney. EPIG analysis identified patterns of gene expression correlated to the observed toxicity, including genes associated with endoplasmic reticulum stress and the unfolded protein response. CONCLUSIONS: By factoring in dose level, number of doses, and tissue into the analysis of gene expression elicited by methapyrilene, we were able to identify genes likely to not be implicated in toxicity, thereby allowing us to focus on a subset of genes to identify toxicity-related processes.


Subject(s)
Gene Expression Profiling , Histamine H1 Antagonists/toxicity , Liver/drug effects , Methapyrilene/toxicity , Up-Regulation/drug effects , Animals , Dose-Response Relationship, Drug , Kidney/drug effects , Kidney/pathology , Liver/pathology , Male , Rats , Rats, Sprague-Dawley
20.
J Proteome Res ; 5(7): 1586-601, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16823966

ABSTRACT

Administration of high doses of the histamine antagonist methapyrilene to rats causes periportal liver necrosis. The mechanism of toxicity is ill-defined and here we have utilized an integrated systems approach to understanding the toxic mechanisms by combining proteomics, metabonomics by 1H NMR spectroscopy and genomics by microarray gene expression profiling. Male rats were dosed with methapyrilene for 3 days at 150 mg/kg/day, which was sufficient to induce liver necrosis, or a subtoxic dose of 50 mg/kg/day. Urine was collected over 24 h each day, while blood and liver tissues were obtained at 2 h after the final dose. The resulting data further define the changes that occur in signal transduction and metabolic pathways during methapyrilene hepatotoxicity, revealing modification of expression levels of genes and proteins associated with oxidative stress and a change in energy usage that is reflected in both gene/protein expression patterns and metabolites. The difficulties of combining and interpreting multiomic data are considered.


Subject(s)
Chemical and Drug Induced Liver Injury , Genomics , Histamine H1 Antagonists/toxicity , Liver/drug effects , Methapyrilene/toxicity , Proteomics , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Histamine H1 Antagonists/administration & dosage , Histamine H1 Antagonists/urine , Lipid Metabolism , Liver/chemistry , Liver/metabolism , Liver/pathology , Liver/physiology , Liver Diseases/genetics , Liver Diseases/metabolism , Male , Methapyrilene/administration & dosage , Methapyrilene/urine , Necrosis/chemically induced , Oxidative Stress/drug effects , Proteins/analysis , Rats , Rats, Wistar , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL