Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Food Chem Toxicol ; 136: 111069, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31883992

ABSTRACT

Ionic liquids are a diverse range of charged chemicals with low volatility and often liquids at ambient temperatures. This characteristic has in part lead to them being considered environmentally-friendly replacements for existing volatile solvents. However, methylimidazolium ionic liquids are slow to break down in the environment and a recent study at Newcastle detected 1 octyl 3 methylimidazolium (M8OI) - an 8 carbon variant methylimidazolium ionic liquid - in soils in close proximity to a landfill site. The current M8OI toxicity database in cultured mammalian cells, in experimental animal studies and in model indicators of environmental impact are reviewed. Selected analytical data from the Newcastle study suggest the soils in close proximity to the landfill site, an urban soil lacking overt contamination, had variable levels of M8OI. The potential for M8OI - or a structurally related ionic liquid - to trigger primary biliary cholangitis (PBC), an autoimmune liver disease thought to be triggered by an unknown agent(s) in the environment, is reviewed.


Subject(s)
Imidazoles/toxicity , Ionic Liquids/toxicity , Liver/drug effects , Animals , Cell Line , Humans
2.
J Hepatol ; 69(5): 1123-1135, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30006067

ABSTRACT

BACKGROUND & AIMS: Primary biliary cholangitis (PBC) is an autoimmune-associated chronic liver disease triggered by environmental factors, such as exposure to xenobiotics, which leads to a loss of tolerance to the lipoic acid-conjugated regions of the mitochondrial pyruvate dehydrogenase complex, typically to the E2 component. We aimed to identify xenobiotics that might be involved in the environmental triggering of PBC. METHODS: Urban landfill and control soil samples from a region with high PBC incidence were screened for xenobiotic activities using analytical, cell-based xenobiotic receptor activation assays and toxicity screens. RESULTS: A variety of potential xenobiotic classes were ubiquitously present, as identified by their interaction with xenobiotic receptors - aryl hydrocarbon receptor, androgen receptor and peroxisome proliferator activated receptor alpha - in cell-based screens. In contrast, xenoestrogens were present at higher levels in soil extracts from around an urban landfill. Furthermore, two landfill sampling sites contained a chemical(s) that inhibited mitochondrial oxidative phosphorylation and induced the apoptosis of a hepatic progenitor cell. The mitochondrial effect was also demonstrated in human liver cholangiocytes from three separate donors. The chemical was identified as the ionic liquid [3-methyl-1-octyl-1H-imidazol-3-ium]+ (M8OI) and the toxic effects were recapitulated using authentic pure chemical. A carboxylate-containing human hepatocyte metabolite of M8OI, bearing structural similarity to lipoic acid, was also enzymatically incorporated into the E2 component of the pyruvate dehydrogenase complex via the exogenous lipoylation pathway in vitro. CONCLUSIONS: These results identify, for the first time, a xenobiotic in the environment that may be related to and/or be a component of an environmental trigger for PBC. Therefore, further study in experimental animal models is warranted, to determine the risk of exposure to these ionic liquids. LAY SUMMARY: Primary biliary cholangitis is a liver disease in which most patients have antibodies to mitochondrial proteins containing lipoic acid binding site(s). This paper identified a man-made chemical present in soils around a waste site. It was then shown that this chemical was metabolized into a product with structural similarity to lipoic acid, which was capable of replacing lipoic acid in mitochondrial proteins.


Subject(s)
Cholangitis/chemically induced , Imidazoles/toxicity , Soil Pollutants/toxicity , Xenobiotics/toxicity , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Estrogen Receptor alpha/drug effects , Hep G2 Cells , Humans , Imidazoles/isolation & purification , Liver/drug effects , Mice , Mitochondria, Liver/drug effects , Pesticides/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Rats , Soil Pollutants/analysis , Xenobiotics/isolation & purification
3.
Differentiation ; 102: 10-18, 2018.
Article in English | MEDLINE | ID: mdl-29857331

ABSTRACT

The rodent pancreatic AR42J-B13 (B-13) cell line differentiates into non-replicative hepatocyte-like cells in response to glucocorticoid mediated via the glucocorticoid receptor (GR). The aims of this study were to identify a human cell line that responds similarly and investigate the mechanisms underpinning any alteration in differentiation. Exposing the human pancreatic adenocarcinoma (HPAC) cell line to 1-10 µM concentrations of dexamethasone (DEX) resulted an inhibition of proliferation, suppressed carcinoembryonic antigen expression, limited expression of pancreatic acinar and hepatic gene expression and significant induction of the constitutively-expressed hepatic CYP3A5 mRNA transcript. These changes were associated with a pulse of genomic DNA methylation and suppressed notch signalling activity. HPAC cells expressed high levels of GR transcript in contrast to other nuclear receptors - such as the glucocorticoid-activated pregnane X receptor (PXR) - and GR transcriptional function was activated by DEX in HPAC cells. Expression of selected hepatocyte transcripts in response to DEX was blocked by co-treatment with the GR antagonist RU486. These data indicate that the HPAC response to glucocorticoid exposure includes an inhibition in proliferation, alterations in notch signalling and a limited change in the expression of genes associated with an acinar and hepatic phenotype. This is the first demonstration of a human cell responding to similarly to the rodent B-13 cell regarding formation of hepatocyte-like cells in response to glucocorticoid. Identifying and modulating the ablating factor(s) may enhance the hepatocyte-like forming capacity of HPAC cells after exposure to glucocorticoid and generate an unlimited in vitro supply of human hepatocytes for toxicology studies and a variety of clinical applications.


Subject(s)
Cell Differentiation/drug effects , Glucocorticoids/pharmacology , Hepatocytes/drug effects , Pancreas/cytology , Cell Line , Dexamethasone/pharmacology , Hepatocytes/cytology , Humans , Liver/metabolism , Pancreatic Neoplasms/drug therapy , Receptors, Glucocorticoid/drug effects , Pancreatic Neoplasms
4.
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
5.
Toxicol Lett ; 273: 55-68, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28356238

ABSTRACT

Tartrazine is a food colour that activates the transcriptional function of the human oestrogen receptor alpha in an in vitro cell model. Since oestrogens are cholestatic, we hypothesised tartrazine will cause periportal injury to the liver in vivo. To test this hypothesis, tartrazine was initially administered systemically to mice resulting in a periportal recruitment of inflammatory cells, increased serum alkaline phosphatase activity and mild periportal fibrosis. To determine whether an oestrogenic effect may be a key event in this response, tartrazine, sulphonated metabolites and a food additive contaminant were screened for their ability to interact with murine oestrogen receptors. In all cases, there were no interactions as agonists or antagonists and further, no oestrogenicity was observed with tartrazine in an in vivo uterine growth assay. To examine the relevance of the hepatic effects of tartrazine to its use as a food additive, tartrazine was orally administered to transgenic NF-κB-Luc mice. Pre- and concurrent oral treatment with alcohol was incorporated given its potential to promote gut permeability and hepatic inflammation. Tartrazine alone induced NF- κB activities in the colon and liver but there was no periportal recruitment of inflammatory cells or fibrosis. Tartrazine, its sulphonated metabolites and the contaminant inhibited sulphotransferase activities in murine hepatic S9 extracts. Given the role of sulfotransferases in bile acid excretion, the initiating event giving rise to periportal inflammation and subsequent hepatic pathology through systemic tartrazine exposure is therefore potentially associated an inhibition of bile acid sulphation and excretion and not on oestrogen receptor-mediated transcriptional function. However, these effects were restricted to systemic exposures to tartrazine and did not occur to any significant effect after oral exposure.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Food Coloring Agents/toxicity , Liver/drug effects , Tartrazine/toxicity , Administration, Oral , Animals , Cell Line , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Ethanol/toxicity , Female , Injections, Intraperitoneal , Liver/metabolism , Liver Function Tests , Luciferases, Firefly/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics
6.
Toxicol Sci ; 156(1): 54-71, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28013213

ABSTRACT

High systemic levels of oestrogens are cholestatic and primary biliary cholangitis (PBC)-which is characterized by hepatic ductular inflammation-is thought to be triggered by exposure to xenobiotics such as those around landfill sites. Xenoestrogens may be a component of this chemical trigger. We therefore hypothesized that xenoestrogens are present at higher levels in the proximity of landfill sites. To test this hypothesis, soil samples were collected, extracts prepared and biological oestrogenic activity examined using cell-based reporter gene assays. Extracts from several sample sites around a landfill site contained a chemical(s) which activated the human ERα in a dose-dependent manner. Extracts from 3 separate control sampling sites were absent of any detectable activity. The mouse ERα and 2 variant mouse ERß cDNAs were cloned and extracts from sample sites around a landfill site also activated these receptors. One variant murine ERß was constitutively active when expressed in cholangiocytes, was readily inactivated by ICI182780 and activated in a dose-responsive, ICI182780-inhibitable manner by oestrogen. However, when this receptor was activated by extracts from landfill site soils, ICI182780 failed to antagonize activation. ERß was readily detectable in murine cholangiocytes and exposing mice acutely to a pooled ER activating soil extracts also gave rise to a mild cholestatic injury. These data indicate that the environment around landfill sites may contain higher levels of xenoestrogens; that these chemicals have "super-activating" characteristics with a variant ERß and therefore these chemicals could be a component of a xenobiotic insult that triggers PBC.


Subject(s)
Alternative Splicing , Bile Ducts/drug effects , Cholestasis/chemically induced , Estrogen Receptor beta/agonists , Estrogens/toxicity , Soil Pollutants/toxicity , Animals , Bile Ducts/cytology , Bile Ducts/metabolism , Bile Ducts/pathology , Cell Line , Cell Line, Tumor , Cells, Cultured , Cholestasis/metabolism , Cholestasis/pathology , Cholestasis/prevention & control , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/chemistry , Estrogens/isolation & purification , Female , Genes, Reporter/drug effects , Humans , Kinetics , Male , Mice , Mice, Nude , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Soil Pollutants/antagonists & inhibitors , Soil Pollutants/isolation & purification , United Kingdom , Waste Disposal Facilities
7.
PLoS One ; 11(3): e0150959, 2016.
Article in English | MEDLINE | ID: mdl-26954030

ABSTRACT

The proliferative B-13 pancreatic cell line is unique in its ability to generate functional hepatocyte-like (B-13/H) cells in response to exposure to glucocorticoid. In these studies, quantitatively comparable hepatic levels of liver-specific and liver-enriched transcription factor and hepatocyte defining mRNA transcripts were expressed after 10-14 days continuous treatment with glucocorticoid. This conversion in phenotype was associated with increased Gr-α mRNA expression and translation of a functional N-terminally truncated variant protein that localized to the nucleus in B-13/H cells. A short (6 hours) pulse exposure to glucocorticoid was also sufficient to transiently activate the Gr and irreversibly drive near identical conversion to B-13/H cells. Examination of epigenetic-related mechanisms demonstrated that B-13 DNA was rapidly methylated and de-methylated over the initial 2 days in response to both continuous or pulse exposure with glucocorticoid. DNA methylation and glucocorticoid-dependent conversion to an hepatic B-13/H phenotype was blocked by the methylation inhibitor, 5-azacytidine. Conversion to an hepatic B-13/H phenotype was also blocked by histone deacetylase inhibitors. Previous experiments have identified N-terminal Sgk1 variant proteins as pivotal to the mechanism(s) associated with pancreatic-hepatic differentiation. Both continuous and pulse exposure to DEX was sufficient to result in a near-similar robust transcriptional increase in Sgk1c mRNA expression from undetectable levels in B-13 cells. Notably, expression of Sgk1c mRNA remained constitutive 14 days later; including after pulse exposure to glucocorticoid and this induction was inhibited by 5-azacytidine or by histone deacetylase inhibitors. These data therefore suggest that exposing B-13 cells to glucocorticoid results in a Gr-dependent pulse in DNA methylation and likely other epigenetic changes such as histone modifications that leads to constitutive expression of Sgk1c and irreversible reprogramming of B-13 cells into B-13/H cells. Understanding and application of these mechanism(s) may enhance the functionality of stem cell-derived hepatocytes generated in vitro.


Subject(s)
Cell Transdifferentiation/genetics , Epigenesis, Genetic , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Pancreas/cytology , Animals , Cell Line , Cell Transdifferentiation/drug effects , DNA Methylation , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Liver/metabolism , Male , Organ Specificity/genetics , Pancreas/metabolism , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Transcription, Genetic
8.
Toxicol Lett ; 243: 98-110, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26739637

ABSTRACT

Rat B-13 progenitor cells are readily converted into functional hepatocyte-like B-13/H cells capable of phase I cytochrome P450-dependent activation of pro-carcinogens and induction of DNA damage. The aim of the present study was to investigate whether the cells are also capable of Phase II sulphotransferase (SULT)-dependent activation of a pro-carcinogen to an ultimate carcinogen. To this end we therefore examined the bioactivation of the model hepatic (hepato- and cholangio-) carcinogen estragole and its proximate SULT1A1-activated genotoxic metabolite 1'-hydroxyestragole. Exposing B-13 or B-13/H cells to estragole (at concentrations up to 1mM) resulted in the production of low levels of 1'-hydroxyestragole, but did not result in detectable DNA damage. Exposing B-13/H cells - but not B-13 cells - to 1'-hydroxyestragole resulted in a dose-dependent increase in DNA damage in comet assays, confirmed by detection of N(2)-(trans-isoestragol-3'-yl)-2'-deoxyguanosine adducts. Genotoxicity was inhibited by general SULT inhibitors, supporting a role for SULTS in the activation of 1-hydroxyestragole in B-13/H cells. However, B-13 and B-13/H cells did not express biologically significant levels of SULT1A1 as determined by qRT-PCR, Western blotting and its associated 7-hydroxycoumarin sulphation activity. B-13 and B-13/H cells expressed - relative to intact rat liver - high levels of SULT2B1 (primarily the b isoform) and SULT4A1 mRNAs and proteins. B-13 and B-13/H cells also expressed the 3'-phosphoadenosine 5'-phosphosulphate synthase 1 required for the generation of activated sulphate cofactor 3'-phosphoadenosine 5'-phosphosulphate. However, only B-13/H cells expressed functional SULT activities towards SULT2B1 substrates DHEA, pregnenolone and 4 methylumbelliferone. Since liver progenitor cells are bi-potential and also form cholangiocytes, we therefore hypothesised that B-13 cells express a cholangiocyte-like SULT profile. To test this hypothesis, the expression of SULTs was examined in liver by RT-PCR and immunohistochemistry. SULT2B1 - but not SULT1A1 - was determined to be expressed in both rat and human cholangiocytes. Since 1'-hydroxyestragole exposure readily produced DNA injury in B-13/H cells, these data suggest that cholangiocarcinomas generated in rats fed estragole may be dependent, in part, on SULT2B1 activation of the 1'-hydroxyestragole metabolite.


Subject(s)
Anisoles/toxicity , DNA Damage/drug effects , Hepatocytes/drug effects , Sulfotransferases/genetics , Allylbenzene Derivatives , Animals , Anisoles/administration & dosage , Arylsulfotransferase/antagonists & inhibitors , Arylsulfotransferase/genetics , Arylsulfotransferase/metabolism , Carcinogens/administration & dosage , Carcinogens/toxicity , Cell Line , Comet Assay , Dehydroepiandrosterone/pharmacology , Gene Expression Regulation , Humans , Hymecromone/pharmacology , Liver/cytology , Liver/drug effects , Liver/metabolism , Pregnenolone/pharmacology , Rats , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/metabolism
9.
PLoS One ; 10(8): e0136173, 2015.
Article in English | MEDLINE | ID: mdl-26302150

ABSTRACT

Liver grafts donated after cardiac death are increasingly used to expand the donor pool but are prone to ischaemic-type biliary lesions. The anti-inflammatory effects of the activated pregnane X receptor have previously been shown to be beneficial in a number of inflammatory liver conditions. However, its role in reducing peri-portal inflammation and fibrosis following ischaemia-reperfusion injury has not been investigated. Hepatic injury and its response to pregnane X receptor activation was examined after partial hepatic ischaemia-reperfusion injury induced by surgically clamping the left and middle lobar blood vessels in rats. Molecular and pathological changes in the liver were examined over the following 28 days. Ischaemia-reperfusion injury resulted in transient cholestasis associated with microvillar changes in biliary epithelial cell membranes and hepatocellular injury which resolved within days after reperfusion. However, in contrast to chemically-induced acute liver injuries, this was followed by sustained elevation in isoprostane E2, peri-portal inflammation and fibrosis that remained unresolved in the ischaemic reperfused lobe for at least 28 days after clamping. Administration of pregnenolone-16α-carbonitrile--a rodent-specific pregnane X receptor activator--resulted in significant reductions in cholestasis, hepatic injury, ischaemic lobe isoprostane E2 levels, peri-portal inflammation and fibrosis. Hepatic ischaemia-reperfusion injury therefore results in inflammatory and fibrotic changes that persist well beyond the initial ischaemic insult. Drug-mediated activation of the pregnane X receptor reduced these adverse changes in rats, suggesting that the pregnane X receptor is a viable drug target to reduce ischaemic-type biliary lesions in recipients of liver transplants donated after cardiac death.


Subject(s)
Cholestasis/physiopathology , Inflammation/drug therapy , Isoprostanes/biosynthesis , Liver Cirrhosis/drug therapy , Receptors, Steroid/biosynthesis , Animals , Bile Ducts/drug effects , Bile Ducts/metabolism , Bile Ducts/pathology , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/pathology , Cholestasis/chemically induced , Constriction , Humans , Inflammation/metabolism , Inflammation/physiopathology , Isoprostanes/metabolism , Liver/drug effects , Liver/injuries , Liver/metabolism , Liver/physiopathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Transplantation , Pregnane X Receptor , Pregnenolone Carbonitrile/administration & dosage , Rats , Receptors, Steroid/metabolism , Reperfusion Injury/chemically induced , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology
10.
Toxicol Sci ; 137(2): 350-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24235770

ABSTRACT

AR42J-B-13 (B-13) cells form hepatocyte-like (B-13/H) cells in response to glucocorticoid treatment. To establish its utility in toxicity and genotoxicity screening, cytochrome P450 (CYP) induction, susceptibility to toxins, and transporter gene expression were examined. Conversion to B-13/H cells resulted in expression of male-specific CYP2C11 and sensitivity to methapyrilene. B-13/H cells constitutively expressed CYP1A, induced expression in response to an aryl hydrocarbon receptor agonist, and activated benzo[α]pyrene to a DNA-damaging species. Functional CYP1A2 was not expressed due to deletions in the Cyp1a2 gene. A B-13 cell line stably expressing the human CYP1A2 was therefore engineered (B-13(-TR/h1A2)) and the derived B-13/H cells expressed metabolically functional CYP1A2. Treatment with the cooked food mutagen 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine resulted in a dose-dependent increase in DNA damage. B-13/H cells expressed constitutive androstane receptor (CAR) and induced CYP2B1 mRNA levels in response to classical CAR activators. However, translation to functional CYP2B1 protein was low and increased minimally by CAR activator treatment. B-13/H cells expressed high levels of pregnane X-receptor (PXR) and induced CYP3A1 in response to classical PXR activators. CYP3A genes were inducible, functional, and activated aflatoxin B1 to a DNA-damaging species. All 23 major hepatic transporters were induced when B-13 cells were converted to B-13/H cells, although in many cases, levels remained below those present in adult rat liver. However, bile salt export pump, Abcb1b, multidrug resistance-associated protein, and breast cancer resistance protein transporters were functional in B-13/H cells. These data demonstrate that the B-13 cell generates hepatocyte-like cells with functional drug metabolism and transporter activities, which can alone--or in a humanized form--be used to screen for hepatotoxic and genotoxic endpoints in vitro.


Subject(s)
DNA Damage , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Liver/drug effects , Toxicity Tests , Animal Testing Alternatives , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Dexamethasone/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Enzyme Induction , Hepatocytes/drug effects , Hepatocytes/enzymology , Liver/enzymology , Liver/pathology , Male , Mutagenicity Tests/methods , Mutagens/toxicity , Rats , Sex Characteristics , Toxicity Tests/methods , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...