Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Chem Res Toxicol ; 35(8): 1383-1392, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35830964

ABSTRACT

To reduce the number of animals and studies needed to fulfill the information requirements as required by Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH) (EC no. 1907/2006), a read-across approach was used to support approximately 30 higher olefins. This study aimed to assess the absorption potential of higher olefins through the gut wall as the experimentally determined bioavailability which would strengthen the read-across hypothesis and justification, reducing the need for toxicity studies on all of the higher olefins. The absorption potential of a series of higher olefins (carbon range from 6 to 28, with five configurations of the double bond) was determined in the in vitro everted rat small intestinal sac model and subsequently ranked. In addition, in silico approaches were applied to predict the reactivity, lipophilicity, and permeability of higher olefins. In the in vitro model, everted sacs were incubated in "fed-state simulated small intestinal fluid" saturated with individual higher olefins. The sac contents were then collected, extracted, and analyzed for olefin content using gas chromatography with a flame ionization detector. The C6 to C10 molecules were readily absorbed into the intestinal sacs. Marked inter-compound differences were observed, with the amount of absorption generally decreasing with the increase in carbon number. Higher olefins with ≥C14 carbons were either not absorbed or very poorly absorbed. In the reactivity simulation study, the reactivity is well described by the position of the double bond rather than the number of carbon atoms. In the lipophilicity and permeability analysis, both parameter descriptors depend mainly on the number of carbon atoms and less on the position of the double bond. In conclusion, these new approach methodologies provide supporting information on any trends or breakpoints in intestinal uptake and a hazard matrix based on carbon number and position of the double bond. This matrix will further assist in the selection of substances for inclusion in the mammalian toxicity testing programme.


Subject(s)
Alkenes , Intestinal Absorption , Animals , Carbon/metabolism , Intestine, Small , Mammals , Permeability , Rats
2.
Toxicology ; 426: 152282, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31465819

ABSTRACT

In a 2-year study the herbicide metazachlor (BAS 479H) was shown to significantly increase the incidence of liver tumours in female Wistar rats at a dietary level of 8000 ppm. As metazachlor is not a genotoxic agent, a series of in vivo and in vitro investigative studies were undertaken to elucidate the mode of action (MOA) for metazachlor-induced female rat liver tumour formation. Male and female Wistar rats were given diets containing 0 (control), 200 and 8000 ppm metazachlor for 3, 7, 14 and 28 days. The treatment of male rats with 200 and 8000 ppm metazachlor and female rats with 8000 ppm metazachlor resulted in significant increases in relative liver weight, which was associated with a centrilobular hepatocyte hypertrophy. Hepatocyte replicative DNA synthesis (RDS) was significantly increased in male rats given 8000 ppm metazachlor for 3 and 7 days and in female rats given 200 ppm metazachlor for 7-28 days and 8000 ppm metazachlor for 3-28 days. Significant increases in relative liver weight, centrilobular hepatocyte hypertrophy and hepatocyte RDS were also observed in male and female Wistar rats given and 500 ppm sodium phenobarbital (NaPB) for 3-28 days. The treatment of female Wistar rats with either 8000 ppm metazachlor for 7 days or with 500 ppm NaPB for 3 and 7 days resulted in the nuclear translocation of the hepatic constitutive androstane receptor (CAR). Treatment of male and female Wistar rats with 8000 ppm metazachlor for 14 days resulted in significant increases in hepatic microsomal total cytochrome P450 (CYP) content, CYP2B subfamily-dependent enzyme activities and mRNA levels, together with some increases in CYP3A enzyme activity and mRNA levels. The treatment of male Wistar rat hepatocytes with metazachlor (concentration range 0.5-50 µM) and NaPB (500 µM) for 4 days resulted in increased CYP2B enzyme activities and mRNA levels; with metazachlor and NaPB also producing significant increases in hepatocyte RDS levels. Studies were also performed with hepatocytes from male Sprague-Dawley wild type (WT) rats and CAR knockout (CAR KO) rats. While both treatment with metazachlor and NaPB for 4 days increased CYP2B enzyme activities and mRNA levels in WT rat hepatocytes, only minor effects were observed in CAR KO rat hepatocytes. Treatment with both metazachlor and NaPB only increased RDS in WT but not in CAR KO rat hepatocytes. The treatment of hepatocytes from two male human donors with 0.5-25 µM metazachlor or 500 µM NaPB for 4 days resulted in increases in CYP2B6 and CYP3A4 mRNA levels but had no effect on hepatocyte RDS. EGF as concurrently used positive control demonstrated the expected RDS response in all rat and human hepatocyte cultures. In conclusion, a series of in vivo and in vitro investigative studies have demonstrated that metazachlor is a CAR activator in rat liver, with similar properties to the prototypical CAR activator phenobarbital. A robust MOA for metazachlor-induced female rat liver tumour formation has been established. Based on the lack of effect of metazachlor on RDS in human hepatocytes, it is considered that the MOA for metazachlor-induced rat liver tumour formation is qualitatively not plausible for humans.


Subject(s)
Acetamides/toxicity , Herbicides/toxicity , Liver Neoplasms, Experimental/chemically induced , Animals , Cells, Cultured , Coxsackie and Adenovirus Receptor-Like Membrane Protein/drug effects , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , DNA Replication/drug effects , Female , Gene Knockout Techniques , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Liver/pathology , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Translocation, Genetic/drug effects
3.
Xenobiotica ; 49(2): 227-238, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29424600

ABSTRACT

The hepatic and thyroid gland effects of the constitutive androstane receptor (CAR) activator sodium phenobarbital (NaPB) and the pregnane X receptor (PXR) activator pregnenolone-16α-carbonitrile (PCN) were examined in male Sprague-Dawley wild-type (WT) and knockout (KO) rats lacking both hepatic CAR and PXR receptors (CAR KO/PXR KO rats). The treatment of WT rats for 7 d with 500 ppm NaPB in the diet and 100 mg/kg/d PCN by gavage resulted in increased relative liver weight, hepatocyte hypertrophy, increased hepatocyte replicative DNA synthesis (RDS) and induction of cytochrome P450 CYP2B and CYP3A subfamily enzymes. NaPB and PCN also induced thyroid gland follicular cell RDS and hepatic microsomal UDP-glucuronosyltransferase activity towards thyroxine as substrate. These effects were not observed in the liver and thyroid gland of CAR KO/PXR KO rats. Male C57BL/6 J (WT) and CAR KO/PXR KO mice were given 1000 ppm NaPB in the diet for 7 d. In WT, but not in CAR KO/PXR KO, mice NaPB treatment resulted in liver hypertrophy and induction of hepatocyte RDS and Cyp2b enzymes. These results suggest that the CAR KO/PXR KO rat and mouse models are useful experimental models for mode of action studies with rodent CAR activators.


Subject(s)
Liver/drug effects , Phenobarbital/pharmacology , Pregnane X Receptor/genetics , Pregnenolone Carbonitrile/pharmacology , Receptors, Cytoplasmic and Nuclear/genetics , Thyroid Gland/drug effects , Animals , Constitutive Androstane Receptor , DNA Replication/drug effects , Gene Knockout Techniques , Male , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
4.
Toxicology ; 400-401: 20-27, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29548889

ABSTRACT

A number of chemicals produce liver and thyroid gland tumours in rodents by nongenotoxic modes of action (MOAs). In this study the hepatic and thyroid gland effects of the constitutive androstane receptor (CAR) activator sodium phenobarbital (NaPB) were examined in male Sprague-Dawley wild type (WT) rats and in CAR knockout (CAR KO) rats and the effects of the pregnane X receptor (PXR) activator pregnenolone-16α-carbonitrile (PCN) were examined in WT and PXR knockout (PXR KO) rats. Rats were either fed diets containing 0 (control) or 500 ppm NaPB or were dosed with 0 (control) or 100 mg/kg/day PCN orally for 7 days. The treatment of WT rats with NaPB and PCN for 7 days resulted in increased relative liver weight, increased hepatocyte replicative DNA synthesis (RDS) and the induction of cytochrome P450 CYP2B and CYP3A subfamily enzyme, mRNA and protein levels. In marked contrast, the treatment of CAR KO rats with NaPB and PXR KO rats with PCN did not result in any increases in liver weight and induction of CYP2B and CYP3A enzymes. The treatment of CAR KO rats with NaPB had no effect on hepatocyte RDS, while PCN produced only a small increase in hepatocyte RDS in PXR KO rats. Treatment with NaPB had no effect on thyroid gland weight in WT and CAR KO rats, whereas treatment with PCN resulted in an increase in relative thyroid gland weight in WT, but not in PXR KO, rats. Thyroid gland follicular cell RDS was increased by the treatment of WT rats with NaPB and PCN, with NaPB also producing a small increase in thyroid gland follicular cell RDS in CAR KO rats. Overall, the present study with CAR KO rats demonstrates that a functional CAR is required for NaPB-mediated increases in liver weight, stimulation of hepatocyte RDS and induction of hepatic CYP enzymes. The studies with PXR KO rats demonstrate that a functional PXR is required for PCN-mediated increases in liver weight and induction of hepatic CYP enzymes; with induction of hepatocyte RDS also being largely mediated through PXR. The hepatic effects of NaPB in CAR KO rats and of PCN in PXR KO rats are in agreement with those observed in other recent literature studies. These results suggest that CAR KO and PXR KO rats are useful experimental models for liver MOA studies with rodent CAR and PXR activators and may also be useful for thyroid gland MOA studies.


Subject(s)
Hepatocytes/metabolism , Phenobarbital/pharmacology , Pregnane X Receptor/deficiency , Pregnenolone Carbonitrile/pharmacology , Receptors, Cytoplasmic and Nuclear/deficiency , Thyroid Gland/metabolism , Animals , Constitutive Androstane Receptor , Hepatocytes/drug effects , Liver/drug effects , Liver/metabolism , Male , Pregnane X Receptor/genetics , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Receptors, Cytoplasmic and Nuclear/genetics , Thyroid Gland/drug effects
5.
Toxicol Sci ; 163(1): 293-306, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29462473

ABSTRACT

A phase 1 dose-escalation trial assessed the chemotherapeutic potential of ammonium perfluorooctanoate (APFO). Forty-nine primarily solid-tumor cancer patients who failed standard therapy received weekly APFO doses (50-1200 mg) for 6 weeks. Clinical chemistries and plasma PFOA (anionic APFO) were measured predose and weekly thereafter. Several clinical measures including total cholesterol, high-density lipoproteins (HDLs), thyroid stimulating hormone (TSH), and free thyroxine (fT4), relative to PFOA concentrations were examined by: Standard statistical analyses using generalized estimating equations (GEE) and a probabilistic analysis using probability distribution functions (pdf) at various PFOA concentrations; and a 2-compartment pharmacokinetic/pharmacodynamic (PK/PD) model to directly estimate mean changes. Based on the GEE, the average rates of change in total cholesterol and fT4 associated with increasing PFOA were approximately -1.2×10-3 mmol/l/µM and 2.8×10-3 pmol/l/µM, respectively. The PK/PD model predicted more closely the trends observed in the data as well as the pdfs of biomarkers. A decline in total cholesterol was observed, with a clear transition in shape and range of the pdfs, manifested by the maximum value of the Kullback-Leibler (KL) divergence, that occurred at plasma PFOA between 420 and 565 µM (175 000-230 000 ng/ml). High-density lipoprotein was unchanged. An increase in fT4 was observed at a higher PFOA transition point, albeit TSH was unchanged. Our findings are consistent with some animal models and may motivate re-examination of the epidemiologic studies to PFOA at levels several orders of magnitude lower than this study. These observational studies have reported contrary associations, but currently understood biology does not support the existence of such conflicting effects.


Subject(s)
Antineoplastic Agents , Caprylates , Fluorocarbons , Models, Biological , Neoplasms/drug therapy , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Caprylates/pharmacokinetics , Caprylates/pharmacology , Caprylates/toxicity , Cholesterol/blood , Dose-Response Relationship, Drug , Female , Fluorocarbons/pharmacokinetics , Fluorocarbons/pharmacology , Fluorocarbons/toxicity , Humans , Liver/drug effects , Liver/enzymology , Liver Function Tests , Male , Middle Aged , Neoplasms/blood , Thyroid Gland/drug effects , Thyroxine/blood , Treatment Outcome
6.
Toxicology ; 396-397: 23-32, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29425889

ABSTRACT

Phenobarbital (PB), a constitutive androstane receptor (CAR) activator, produces liver tumours in rodents by a mitogenic mode of action involving CAR activation. In this study, the hepatic effects of sodium phenobarbital (NaPB) were compared in male C57BL/6J wild type (WT) mice and in humanized mice, where both the mouse CAR and pregnane X receptor (PXR) have been replaced by their human counterparts (hCAR/hPXR mice). Investigations were also performed in cultured male C57BL/6J and CD-1 mouse, male Sprague-Dawley rat and male and female human hepatocytes. The treatment of WT and hCAR/hPXR mice with 186-984 ppm NaPB in the diet for 7 days resulted in increased relative liver weight, hypertrophy and induction of cytochrome P450 (CYP) enzyme activities. Treatment with NaPB also produced dose-dependent increases in hepatocyte replicative DNA synthesis (RDS), with the effect being more marked in WT than in hCAR/hPXR mice. While the treatment of cultured C57BL/6J and CD-1 mouse, Sprague-Dawley rat and human hepatocytes with 100 and/or 1000 µM NaPB for 4 days induced CYP enzyme activities, increased RDS was only observed in mouse and rat hepatocytes. However, as a positive control, epidermal growth factor increased RDS in hepatocytes from all three species. In summary, although human hepatocytes are refractory to the mitogenic effects of NaPB, treatment with NaPB induced RDS in vivo in hCAR/hPXR mice, which is presumably due to the human CAR and PXR receptors operating in a mouse hepatocyte regulatory environment. As the response of the hCAR/hPXR mouse to the CAR activator NaPB differs markedly from that of human hepatocytes, the hCAR/hPXR mouse is thus not a suitable animal model for studies on the hepatic effects of nongenotoxic rodent CAR activators.


Subject(s)
Hepatocytes/drug effects , Hypnotics and Sedatives/pharmacology , Phenobarbital/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Steroid/drug effects , Animals , Cells, Cultured , Constitutive Androstane Receptor , Cytochromes/metabolism , DNA Replication/drug effects , Epidermal Growth Factor/metabolism , Female , Humans , Hypnotics and Sedatives/blood , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Organ Size , Phenobarbital/blood , Pregnane X Receptor , Rats , Rats, Sprague-Dawley
7.
Toxicol Sci ; 139(2): 501-11, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24690595

ABSTRACT

The constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) are closely related nuclear receptors involved in drug metabolism and play important roles in the mechanism of phenobarbital (PB)-induced rodent nongenotoxic hepatocarcinogenesis. Here, we have used a humanized CAR/PXR mouse model to examine potential species differences in receptor-dependent mechanisms underlying liver tissue molecular responses to PB. Early and late transcriptomic responses to sustained PB exposure were investigated in liver tissue from double knock-out CAR and PXR (CAR(KO)-PXR(KO)), double humanized CAR and PXR (CAR(h)-PXR(h)), and wild-type C57BL/6 mice. Wild-type and CAR(h)-PXR(h) mouse livers exhibited temporally and quantitatively similar transcriptional responses during 91 days of PB exposure including the sustained induction of the xenobiotic response gene Cyp2b10, the Wnt signaling inhibitor Wisp1, and noncoding RNA biomarkers from the Dlk1-Dio3 locus. Transient induction of DNA replication (Hells, Mcm6, and Esco2) and mitotic genes (Ccnb2, Cdc20, and Cdk1) and the proliferation-related nuclear antigen Mki67 were observed with peak expression occurring between 1 and 7 days PB exposure. All these transcriptional responses were absent in CAR(KO)-PXR(KO) mouse livers and largely reversible in wild-type and CAR(h)-PXR(h) mouse livers following 91 days of PB exposure and a subsequent 4-week recovery period. Furthermore, PB-mediated upregulation of the noncoding RNA Meg3, which has recently been associated with cellular pluripotency, exhibited a similar dose response and perivenous hepatocyte-specific localization in both wild-type and CAR(h)-PXR(h) mice. Thus, mouse livers coexpressing human CAR and PXR support both the xenobiotic metabolizing and the proliferative transcriptional responses following exposure to PB.


Subject(s)
Cell Cycle/drug effects , Liver/drug effects , Phenobarbital/toxicity , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Transcriptome/drug effects , Animals , Cell Cycle/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Constitutive Androstane Receptor , Gene Expression Profiling , Humans , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenobarbital/pharmacokinetics , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Species Specificity , Xenobiotics/pharmacokinetics , Xenobiotics/toxicity
8.
Crit Rev Toxicol ; 44(1): 64-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24180433

ABSTRACT

The constitutive androstane receptor (CAR) and pregnane X receptor (PXR) are important nuclear receptors involved in the regulation of cellular responses from exposure to many xenobiotics and various physiological processes. Phenobarbital (PB) is a non-genotoxic indirect CAR activator, which induces cytochrome P450 (CYP) and other xenobiotic metabolizing enzymes and is known to produce liver foci/tumors in mice and rats. From literature data, a mode of action (MOA) for PB-induced rodent liver tumor formation was developed. A MOA for PXR activators was not established owing to a lack of suitable data. The key events in the PB-induced liver tumor MOA comprise activation of CAR followed by altered gene expression specific to CAR activation, increased cell proliferation, formation of altered hepatic foci and ultimately the development of liver tumors. Associative events in the MOA include altered epigenetic changes, induction of hepatic CYP2B enzymes, liver hypertrophy and decreased apoptosis; with inhibition of gap junctional intercellular communication being an associative event or modulating factor. The MOA was evaluated using the modified Bradford Hill criteria for causality and other possible MOAs were excluded. While PB produces liver tumors in rodents, important species differences were identified including a lack of cell proliferation in cultured human hepatocytes. The MOA for PB-induced rodent liver tumor formation was considered to be qualitatively not plausible for humans. This conclusion is supported by data from a number of epidemiological studies conducted in human populations chronically exposed to PB in which there is no clear evidence for increased liver tumor risk.


Subject(s)
Liver Neoplasms/pathology , Liver/drug effects , Phenobarbital/toxicity , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Aryl Hydrocarbon Hydroxylases , Cell Proliferation/drug effects , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B6 , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Neoplasms/chemically induced , Pregnane X Receptor , Receptors, Steroid/metabolism , Xenobiotics/toxicity
9.
Toxicol Sci ; 132(2): 443-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358192

ABSTRACT

Previous analysis of in utero dibutylphthalate (DBP)-exposed fetal rat testes indicated that DBP's antiandrogenic effects were mediated, in part, by indirect inhibition of steroidogenic factor 1 (SF1), suggesting that peroxisome proliferator-activated receptor alpha (PPARα) might be involved through coactivator (CREB-binding protein [CBP]) sequestration. To test this hypothesis, we have performed chromatin immunoprecipitation (ChIP) microarray analysis to assess the DNA binding of PPARα, SF1, CBP, and RNA polymerase II in DBP-induced testicular maldevelopment target genes. Pathway analysis of expression array data in fetal rat testes examined at gestational day (GD) 15, 17, or 19 indicated that lipid metabolism genes regulated by SF1 and PPARα, respectively, were overrepresented, and the time dependency of changes to PPARα-regulated lipid metabolism genes correlated with DBP-mediated repression of SF1-regulated steroidogenesis genes. ChIP microarrays were used to investigate whether DBP-mediated repression of SF1-regulated genes was associated with changes in SF1 binding to genes involved in DBP-induced testicular maldevelopment. DBP treatment caused reductions in SF1 binding in CYP11a, StAR, and CYP17a. Follicle-stimulating hormone receptor (FSHR), regulated by SF1 but unaffected by DBP-treatment, also contained SF1-binding peaks, but DBP did not change this compared with control. GD15 and GD19 fetal testes contained PPARα protein-binding peaks in CYP11a, StAR, and CYP17a regulatory regions. In contrast to its repressive effect on SF1, DBP treatment caused increases in these peaks compared with control. PPARα-binding peaks in the FSHR promoter were not detected in GD15 samples. Hence, the repressive effect of DBP on SF1-regulated steroidogenic genes correlates with inhibition of SF1-DNA binding and increased PPARα-DNA binding. The data indicate that PPARα may act as an indirect transrepressor of SF1 on steroidogenic genes in fetal rat testes in response to DBP treatment.


Subject(s)
Dibutyl Phthalate/toxicity , Testis/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Female , Immunochemistry , Male , Rats , Rats, Wistar , Testis/metabolism
10.
Toxicol Sci ; 131(2): 375-86, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23091169

ABSTRACT

The molecular events during nongenotoxic carcinogenesis and their temporal order are poorly understood but thought to include long-lasting perturbations of gene expression. Here, we have investigated the temporal sequence of molecular and pathological perturbations at early stages of phenobarbital (PB) mediated liver tumor promotion in vivo. Molecular profiling (mRNA, microRNA [miRNA], DNA methylation, and proteins) of mouse liver during 13 weeks of PB treatment revealed progressive increases in hepatic expression of long noncoding RNAs and miRNAs originating from the Dlk1-Dio3 imprinted gene cluster, a locus that has recently been associated with stem cell pluripotency in mice and various neoplasms in humans. PB induction of the Dlk1-Dio3 cluster noncoding RNA (ncRNA) Meg3 was localized to glutamine synthetase-positive hypertrophic perivenous hepatocytes, suggesting a role for ß-catenin signaling in the dysregulation of Dlk1-Dio3 ncRNAs. The carcinogenic relevance of Dlk1-Dio3 locus ncRNA induction was further supported by in vivo genetic dependence on constitutive androstane receptor and ß-catenin pathways. Our data identify Dlk1-Dio3 ncRNAs as novel candidate early biomarkers for mouse liver tumor promotion and provide new opportunities for assessing the carcinogenic potential of novel compounds.


Subject(s)
Biomarkers, Tumor/genetics , Genomic Imprinting , Intercellular Signaling Peptides and Proteins/genetics , Iodide Peroxidase/genetics , Liver Neoplasms, Experimental/genetics , Multigene Family , RNA, Untranslated/genetics , Animals , Calcium-Binding Proteins , Constitutive Androstane Receptor , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transcriptome , beta Catenin/metabolism
11.
Toxicology ; 293(1-3): 30-40, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22239858

ABSTRACT

In a prior 28-day dietary study in rats with 20 and 100 ppm K⁺ PFOS, activation of PPARα and CAR/PXR were concluded to be etiological factors in K⁺ PFOS-induced hepatomegaly and hepatic tumorigenesis. The objective of this study was to evaluate persistence/resolution of K⁺ PFOS-induced, liver-related effects in male Sprague Dawley rats following a 7-day dietary exposure to K⁺ PFOS at 20 or 100 ppm. Groups of 10 rats per treatment were observed on recovery Day(s) 1, 28, 56, and 84 following treatment. Changes consistent with hepatic PPARα and CAR/PXR activation noted on recovery Day 1 included: increased liver weight; decreased plasma cholesterol, alanine aminotransferase, and triglycerides; decreased liver DNA concentration and increased hepatocellular cytosolic CYP450 concentration; increased liver activity of acyl CoA oxidase, CYP4A, CYP2B, and CYP3A; increased liver proliferative index and decreased liver apoptotic index; decreased hepatocellular glycogen-induced vacuoles; increased centrilobular hepatocellular hypertrophy. Most effects resolved to control levels during recovery. Effects on plasma cholesterol, hepatocellular cytosolic CYP450 concentrations, liver apoptotic index, CYP3A, and centrilobular hepatocellular hypertrophy persisted through the end of the recovery period. Thyroid parameters (histology, apoptosis, and proliferation) were unaffected at all time points. Mean serum PFOS concentrations on recovery Day 1 were 39 and 140 µg/mL (20 ppm and 100 ppm K⁺ PFOS, respectively), decreasing to 4 and 26 µg/mL by recovery Day 84. Thus, hepatic effects in male rats resulting from K⁺ PFOS-induced activation of PPARα and CAR/PXR resolved slowly or were still present after 84-days following a 7-day dietary treatment, consistent with the slow elimination rate of PFOS.


Subject(s)
Alkanesulfonic Acids/pharmacokinetics , Alkanesulfonic Acids/toxicity , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Fluorocarbons/pharmacokinetics , Fluorocarbons/toxicity , Hepatomegaly/chemically induced , Liver/drug effects , Thyroid Gland/drug effects , Alkanesulfonic Acids/administration & dosage , Alkanesulfonic Acids/metabolism , Animals , Apoptosis/drug effects , Carcinogens/administration & dosage , Carcinogens/metabolism , Cell Proliferation/drug effects , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/metabolism , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/toxicity , Fluorocarbons/administration & dosage , Fluorocarbons/metabolism , Half-Life , Hepatomegaly/blood , Hepatomegaly/metabolism , Hepatomegaly/pathology , Liver/metabolism , Liver/pathology , Male , PPAR alpha/metabolism , Pregnane X Receptor , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Thyroid Gland/pathology , Toxicity Tests, Subacute
12.
Toxicology ; 293(1-3): 16-29, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22245121

ABSTRACT

The present study investigated the potential role for activation of PPARα and CAR/PXR by potassium PFOS (K⁺ PFOS) with respect to the etiology of hepatic hypertrophy and hepatocellular adenoma in rats. Male Sprague-Dawley rats were fed K⁺ PFOS (20 or 100 ppm) for either 1, 7, or 28 days. Wyeth 14,643 (Wy 14,643, 50 ppm) and phenobarbital (PB, 500 ppm) were the controls for PPARα and CAR/PXR activation, respectively. Measurements included: plasma ALT, AST, cholesterol, triglycerides, and glucose; liver protein and DNA content; liver activities of palmitoyl CoA oxidase (ACOX), Cyp4A, CYP2B, and CYP3A; induction of liver CYP4A1, CYP2E1, CYP2B1/2, and CYP3A1 proteins (SDS-PAGE and Western blots); liver and thyroid microscopic histopathology, apoptotic index, and cell proliferation index. Terminal body weight was decreased by K⁺ PFOS (100 ppm) and Wy 14,643. All test-compound treatments increased liver weight. Plasma lipids were decreased by both PFOS and Wy 14,643. After treatment for 1 day, K⁺ PFOS (100 ppm), PB, and Wy 14,643 increased mean hepatic DNA concentration and total hepatic DNA, and total DNA remained elevated after treatment for 7 days and 28 days (PB and Wy 14,643 only). Hepatic P450 concentration was elevated after 7 and 28 days by K⁺ PFOS and by PB. K⁺ PFOS and Wy 14,643 increased liver activities of ACOX and CYP4A as well as increased liver CYP4A1 protein. By 28 days of treatment, K⁺ PFOS and PB increased liver activities of CYP2B and CYP3A as well as increased liver CYP2B1/2 and CYP3A1 proteins, and Wy 14,643 increased CYP2B enzyme activity to a slight extent. All test compounds increased the liver cell proliferative index and decreased the liver apoptotic index. No histological changes of the thyroid were noted; however, PB and WY increased thyroid follicular cell proliferation index (seven-day treatment only), while K⁺ PFOS did not. The thyroid follicular cell apoptotic index did not differ between groups. The hepatomegaly and hepatocellular adenoma observed after dietary exposure of Sprague-Dawley rats to K⁺ PFOS likely are due to the increased expression of xenosensor nuclear receptors PPARα and CAR/PXR. Given the markedly lower or absent response of human hepatocytes to the proliferative stimulus from activation of PPARα and CAR/PXR, the hepatocellular proliferative response from activation of these receptors by PFOS observed in rats is not expected to be of human relevance.


Subject(s)
Adenoma, Liver Cell/chemically induced , Alkanesulfonic Acids/toxicity , Cell Proliferation/drug effects , Fluorocarbons/toxicity , Liver Neoplasms/chemically induced , PPAR alpha/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Adenoma, Liver Cell/chemistry , Adenoma, Liver Cell/metabolism , Adenoma, Liver Cell/pathology , Alkanesulfonic Acids/administration & dosage , Alkanesulfonic Acids/analysis , Alkanesulfonic Acids/pharmacokinetics , Animals , Apoptosis/drug effects , Carcinogens/administration & dosage , Carcinogens/analysis , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/analysis , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/toxicity , Fluorocarbons/administration & dosage , Fluorocarbons/analysis , Fluorocarbons/pharmacokinetics , Hepatomegaly/chemically induced , Liver/chemistry , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms/chemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Neoplasm Proteins/metabolism , Pregnane X Receptor , Random Allocation , Rats , Rats, Sprague-Dawley , Thyroid Gland/drug effects , Thyroid Gland/pathology , Toxicity Tests, Subacute
13.
Toxicol Sci ; 121(2): 207-33, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21447610

ABSTRACT

Transgenic animal models are powerful tools for developing a more detailed understanding on the roles of specific genes in biological pathways and systems. Applications of these models have been made within the field of toxicology, most notably for the screening of mutagenic and carcinogenic potential and for the characterization of toxic mechanisms of action. It has long been a goal of research toxicologists to use the data from these models to refine hazard identification and characterization to better inform human health risk assessments. This review provides an overview on the applications of transgenic animal models in the assessment of mutagenicity and carcinogenicity, their use as reporter systems, and as tools for understanding the roles of xenobiotic-metabolizing enzymes and biological receptors in the etiology of chemical toxicity. Perspectives are also shared on the future outlook for these models in toxicology and risk assessment and how transgenic technologies are likely to be an integral tool for toxicity testing in the 21st century.


Subject(s)
Animals, Genetically Modified , Toxicity Tests/methods , Toxicity Tests/trends , Animals , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Humans , Mice , Mice, Knockout , Models, Animal , Mutation , PPAR alpha/genetics , PPAR alpha/metabolism , Pregnane X Receptor , Rats , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Risk Assessment/methods , Sequence Analysis, DNA , Xenobiotics/metabolism
14.
Arch Toxicol ; 84(10): 787-98, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20614104

ABSTRACT

Ammonium perfluorooctanoate (APFO), a processing aid used in the production of fluoropolymers, produces hepatomegaly and hepatocellular hypertrophy in rodents. In mice, APFO-induced hepatomegaly is associated with increased activation of the xenosensor nuclear receptors, PPARα and CAR/PXR. Although non-genotoxic, chronic dietary treatment of Sprague-Dawley (S-D) rats with APFO produced an increase in benign tumours of the liver, acinar pancreas, and testicular Leydig cells. Most of the criteria for establishing a PPARα-mediated mode of action for the observed hepatocellular tumours have been previously established with the exception of the demonstration of increased hepatocellular proliferation. The present study evaluates the potential roles for APFO-induced activation of PPARα and CAR/PXR with respect to liver tumour production in the S-D rat and when compared to the specific PPARα agonist, 4-chloro-6-(2,3-xylidino)-2-pyrimidinylthioacetic acid (Wy 14,643). Male S-D rats were fed APFO (300 ppm in diet) or Wy 14,643 (50 ppm in diet) for either 1, 7, or 28 days. Effects of treatment with APFO included: decreased body weight; hepatomegaly, hepatocellular hypertrophy, hepatocellular hyperplasia (microscopically and by BrdU labelling index), and hepatocellular glycogen loss; increased activation of PPARα (peroxisomal ß-oxidation and microsomal CYP4A1 protein); decreased plasma triglycerides, cholesterol, and glucose; increased activation of CAR (CYP2B1/2 protein) and CAR/PXR (CYP3A1 protein). Responses to treatment with Wy 14,643 were consistent with increased activation of PPARα, specifically: increased CYP4A1 and peroxisomal ß-oxidation; increased hepatocellular hypertrophy and cell proliferation; decreased apoptosis; and hypolipidaemia. With the exception of decreased apoptosis, the effects observed with Wy 14,643 were noted with APFO, and APFO was less potent. These data clearly demonstrate an early hepatocellular proliferative response to APFO treatment and suggest that the hepatomegaly and tumours observed after chronic dietary exposure of S-D rats to APFO likely are due to a proliferative response to combined activation of PPARα and CAR/PXR. This mode of action is unlikely to pose a human hepatocarcinogenic hazard.


Subject(s)
Caprylates/toxicity , Cell Proliferation/drug effects , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Hepatocytes/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Apoptosis/drug effects , Body Weight/drug effects , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , DNA/metabolism , Hepatocytes/pathology , Hepatomegaly/chemically induced , Hypertrophy , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Organ Size/drug effects , PPAR alpha/metabolism , Peroxisomes/metabolism , Pregnane X Receptor , Rats , Rats, Sprague-Dawley , Receptors, Steroid/metabolism
15.
Toxicol Sci ; 116(2): 452-66, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20403969

ABSTRACT

Mouse nongenotoxic hepatocarcinogens phenobarbital (PB) and chlordane induce hepatomegaly characterized by hypertrophy and hyperplasia. Increased cell proliferation is implicated in the mechanism of tumor induction. The relevance of these tumors to human health is unclear. The xenoreceptors, constitutive androstane receptors (CARs), and pregnane X receptor (PXR) play key roles in these processes. Novel "humanized" and knockout models for both receptors were developed to investigate potential species differences in hepatomegaly. The effects of PB (80 mg/kg/4 days) and chlordane (10 mg/kg/4 days) were investigated in double humanized PXR and CAR (huPXR/huCAR), double knockout PXR and CAR (PXRKO/CARKO), and wild-type (WT) C57BL/6J mice. In WT mice, both compounds caused increased liver weight, hepatocellular hypertrophy, and cell proliferation. Both compounds caused alterations to a number of cell cycle genes consistent with induction of cell proliferation in WT mice. However, these gene expression changes did not occur in PXRKO/CARKO or huPXR/huCAR mice. Liver hypertrophy without hyperplasia was demonstrated in the huPXR/huCAR animals in response to both compounds. Induction of the CAR and PXR target genes, Cyp2b10 and Cyp3a11, was observed in both WT and huPXR/huCAR mouse lines following treatment with PB or chlordane. In the PXRKO/CARKO mice, neither liver growth nor induction of Cyp2b10 and Cyp3a11 was seen following PB or chlordane treatment, indicating that these effects are CAR/PXR dependent. These data suggest that the human receptors are able to support the chemically induced hypertrophic responses but not the hyperplastic (cell proliferation) responses. At this time, we cannot be certain that hCAR and hPXR when expressed in the mouse can function exactly as the genes do when they are expressed in human cells. However, all parameters investigated to date suggest that much of their functionality is maintained.


Subject(s)
Chlordan/toxicity , Liver Neoplasms, Experimental/chemically induced , Phenobarbital/toxicity , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cell Proliferation/drug effects , Constitutive Androstane Receptor , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P450 Family 2 , Humans , Hyperplasia , Hypertrophy , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/pathology , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Pregnane X Receptor , Species Specificity , Steroid Hydroxylases/biosynthesis
16.
Arch Toxicol ; 84(3): 233-43, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20069279

ABSTRACT

Short-chain chlorinated paraffins (SCCPs) cause kidney tumours in male rats, but not in female rats or mice of either sex. Male rat-specific tumours also occur in rats dosed with a range of compounds including 1,4- dichlorobenzene (DCB) and d-limonene (DL). These compounds bind to a male rat-specific hepatic protein, alpha-2-urinary globulin (α2u), and form degradationresistant complexes in the kidney. The resulting accumulation of α2u causes cell death and sustained regenerative cell proliferation, which in turn leads to the formation of renal tumours. To investigate whether the SCCP, Chlorowax 500C (C500C), causes tumours via the accumulation of α2u male rats were orally dosed with either C500C (625 mg/kg of body weight), DCB (300 mg/kg of body weight), or DL (150 mg/kg of body weight) for 28 consecutive days. An increase in renal α2u and cell proliferation was observed in DCB- and DL-treated rats but not in C500C-treated rats. C500C caused peroxisome proliferation and a down-regulation of α2u synthesis in male rat liver. This down-regulation occurred at the transcriptional level. Since less α2u was produced in C500C-treated rats, there was less available for accumulation in the kidney hence a typical α2u nephropathy did not appear. However, the administration of a radiolabelled SCCP, [14C]polychlorotridecane (PCTD), to male rats demonstrated its binding to renal α2u. Thus, it is possible that SCCPs bind to α2u and cause a slow accumulation of the protein in the kidney followed by delayed onset of α2u nephropathy. As a consequence of these findings in the current experiments, while evidence exists implicating α2u-globulin in the molecular mechanism of action of the C500C, the classic profile of a α2u-globulin nephropathy seen with other chemicals such as DCB and DL was not reproduced during this experimental protocol.


Subject(s)
Alpha-Globulins/metabolism , Carcinogens/metabolism , Hydrocarbons, Chlorinated/metabolism , Kidney Neoplasms/metabolism , Alpha-Globulins/genetics , Animals , Carcinogens/toxicity , Cell Proliferation/drug effects , Chlorobenzenes/metabolism , Chlorobenzenes/toxicity , Cyclohexenes/metabolism , Cyclohexenes/toxicity , Disease Models, Animal , Down-Regulation/drug effects , Female , Hydrocarbons, Chlorinated/toxicity , Kidney/drug effects , Kidney/metabolism , Kidney Neoplasms/chemically induced , Kidney Neoplasms/pathology , Limonene , Liver/drug effects , Liver/metabolism , Male , Peroxisomes/drug effects , Protein Binding , Rats , Rats, Inbred F344 , Terpenes/metabolism , Terpenes/toxicity , Transcription, Genetic/drug effects
17.
Toxicology ; 210(2-3): 147-57, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15840428

ABSTRACT

p-Phenylenediamine (PPD) is a widely used ingredient in permanent hair dyes; however, little has been published on its metabolism, especially with respect to hepatic cytochrome P450 (CYP)-mediated oxidation. This is regarded as a key step in the activation of carcinogenic arylamines that ultimately leads to the development of bladder cancer. Most epidemiology studies show no significant association between personal use of hair dyes and bladder cancer, but one recent study reported an increased risk of bladder cancer in women who were frequent users of permanent hair dyes. The aim of the present study was to use intact human hepatocytes, human liver microsomes, and heterologously expressed human CYPs to determine whether PPD is metabolised by hepatic CYPs to form an N-hydroxylamine. p-Phenylenediamine was N-acetylated by human hepatocytes to form N-acetylated metabolites, but there was no evidence for the formation of mono-oxygenated metabolites or for enzyme-mediated covalent binding of 14C-PPD to microsomal protein. In contrast, 2-aminofluorene underwent CYP-mediated metabolism to > or = 4 different hydroxylated metabolites. The lack of evidence for hepatic CYP-mediated metabolism of PPD is inconsistent with the hypothesis that this compound plays a causal role in the development of bladder cancer via a mode of action involving hepatic metabolism to an N-hydroxyarylamine.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/enzymology , Microsomes, Liver/enzymology , Phenylenediamines/metabolism , Acetylation , Cells, Cultured , Chromatography, High Pressure Liquid , Fluorenes/metabolism , Humans , Male , Mass Spectrometry , Protein Binding , Recombinant Proteins/metabolism
18.
J Histochem Cytochem ; 52(5): 653-62, 2004 May.
Article in English | MEDLINE | ID: mdl-15100242

ABSTRACT

Class kappa glutathione S-transferases are a poorly characterized family of detoxication enzymes whose localization has not been defined. In this study we investigated the tissue, cellular, and subcellular distribution of mouse glutathione S-transferase class kappa 1 (mGSTK1) protein using a variety of immunolocalization techniques. Western blotting analysis of mouse tissue homogenates demonstrated that mGSTK1 is expressed at relatively high levels in liver and stomach. Moderate expression was observed in kidney, heart, large intestine, testis, and lung, whereas sparse or essentially no mGSTK1 protein was detected in small intestine, brain, spleen, and skeletal muscle. Immunohistochemical (IHC) analysis for mGSTK1 revealed granular staining of hepatocytes throughout the liver, consistent with organelle staining. IHC analysis of murine kidney localized GSTK1 to the straight portion of the proximal convoluted tubule (pars recta). Staining was consistent with regions rich in mitochondria. Electron microscopy, using indirect immunocolloidal gold staining, clearly showed that mGSTK1 was localized in mitochondria in both mouse liver and kidney. These results are consistent with a role for mGST K1-1 in detoxification, and the confirmation of the intramitochondrial localization of this enzyme implies a unique role for GST class kappa as an antioxidant enzyme.


Subject(s)
Glutathione Transferase/metabolism , Subcellular Fractions/enzymology , Animals , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/metabolism , Glutathione Transferase/biosynthesis , Kidney/enzymology , Kidney/metabolism , Kidney/ultrastructure , Liver/enzymology , Liver/metabolism , Liver/ultrastructure , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mitochondria/enzymology , Organ Specificity , Protein Subunits/metabolism
19.
Biochem J ; 373(Pt 2): 559-69, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12720545

ABSTRACT

The class Kappa family of glutathione S-transferases (GSTs) currently comprises a single rat subunit (rGSTK1), originally isolated from the matrix of liver mitochondria [Harris, Meyer, Coles and Ketterer (1991) Biochem. J. 278, 137-141; Pemble, Wardle and Taylor (1996) Biochem. J. 319, 749-754]. In the present study, an expressed sequence tag (EST) clone has been identified which encodes a mouse class Kappa GST (designated mGSTK1). The EST clone contains an open reading frame of 678 bp, encoding a protein composed of 226 amino acid residues with 86% sequence identity with the rGSTK1 polypeptide. The mGSTK1 and rGSTK1 proteins have been heterologously expressed in Escherichia coli and purified by affinity chromatography. Both mouse and rat transferases were found to exhibit GSH-conjugating and GSH-peroxidase activities towards model substrates. Analysis of expression levels in a range of mouse and rat tissues revealed that the mRNA encoding these enzymes is expressed predominantly in heart, kidney, liver and skeletal muscle. Although other soluble GST isoenzymes are believed to reside primarily within the cytosol, subcellular fractionation of mouse liver demonstrates that this novel murine class Kappa GST is associated with mitochondrial fractions. Through the use of bioinformatics, the genes encoding the mouse and rat class Kappa GSTs have been identified. Both genes comprise eight exons, the protein coding region of which spans approx. 4.3 kb and 4.1 kb of DNA for mGSTK1 and rGSTK1 respectively. This conservation in primary structure, catalytic properties, tissue-specific expression, subcellular localization and gene structure between mouse and rat class Kappa GSTs indicates that they perform similar physiological functions. Furthermore, the association of these enzymes with mitochondrial fractions is consistent with them performing a specific conserved biological role within this organelle.


Subject(s)
Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chromatography, Affinity , Cloning, Molecular , Cytosol , Escherichia coli/enzymology , Expressed Sequence Tags , Glutathione Transferase/classification , Isoenzymes , Kidney/enzymology , Liver/enzymology , Mice , Mitochondria, Liver/enzymology , Molecular Sequence Data , Muscle, Skeletal/enzymology , Myocardium/enzymology , Open Reading Frames , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Subcellular Fractions
20.
Biochem J ; 365(Pt 2): 405-16, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-11991805

ABSTRACT

Mice that lack the Nrf2 basic-region leucine-zipper transcription factor are more sensitive than wild-type (WT) animals to the cytotoxic and genotoxic effects of foreign chemicals and oxidants. To determine the basis for the decrease in tolerance of the Nrf2 homozygous null mice to xenobiotics, enzyme assay, Western blotting and gene-specific real-time PCR (TaqMan) have been used to examine the extent to which hepatic expression of GSH-dependent enzymes is influenced by the transcription factor. The amounts of protein and mRNA for class Alpha, Mu and Pi glutathione S-transferases were compared between WT and Nrf2 knockout (KO) mice of both sexes under both constitutive and inducible conditions. Among the class Alpha and class Mu transferases, constitutive expression of Gsta1, Gsta2, Gstm1, Gstm2, Gstm3, Gstm4 and Gstm6 subunits was reduced in the livers of Nrf2 mutant mice to between 3% and 60% of that observed in WT mice. Induction of these subunits by butylated hydroxyanisole (BHA) was more marked in WT female mice than in WT male mice. TaqMan analyses showed the increase in transferase mRNA caused by BHA was attenuated in Nrf2(-/-) mice, with the effect being most apparent in the case of Gsta1, Gstm1 and Gstm3. Amongst class Pi transferase subunits, the constitutive hepatic level of mRNA for Gstp1 and Gstp2 was not substantially affected in the KO mice, but their induction by BHA was dependent on Nrf2; this was more obvious in female mutant mice than in male mice. Nrf2 KO mice exhibited reduced constitutive expression of the glutamate cysteine ligase catalytic subunit, and, to a lesser extent, the expression of glutamate cysteine ligase modifier subunit. Little variation was observed in the levels of glutathione synthase in the different mouse lines. Thus the increased sensitivity of Nrf2(-/-) mice to xenobiotics can be partly attributed to a loss in constitutive expression of multiple GSH-dependent enzymes, which causes a reduction in intrinsic detoxification capacity in the KO animal. These data also indicate that attenuated induction of GSH-dependent enzymes in Nrf2(-/-) mice probably accounts for their failure to adapt to chronic exposure to chemical and oxidative stress.


Subject(s)
DNA-Binding Proteins/physiology , Glutathione Transferase/genetics , Liver/enzymology , Trans-Activators/physiology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cloning, Molecular , DNA Primers , DNA Probes , DNA-Binding Proteins/genetics , Female , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Male , Mice , Mice, Knockout , Molecular Sequence Data , NF-E2-Related Factor 2 , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...