27.07.2013 Views

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

1103 THE ROLE OF 3, 3’, 4, 4’, 5-<br />

PENTACHLOROBIPHENYL (PCB 126) IN COPPER<br />

DISPOSITION IN RODENT LIVER.<br />

M. Li 1, 2 , I. Lai 1, 2 , B. Wels 3 , D. Simmons 3 , G. Ludewig 1, 2 and L. Robertson 1, 2 .<br />

1 Interdisciplinary Program in Human <strong>Toxicology</strong>, University <strong>of</strong> Iowa, Iowa City, IA,<br />

2 Occupation and Environmental Health, University <strong>of</strong> Iowa, Iowa City, IA and 3 State<br />

Hygienic Laboratory, University <strong>of</strong> Iowa, Iowa City, IA.<br />

Copper, an essential trace element and ubiquitous in animals and humans, is essential<br />

for the proper functioning <strong>of</strong> the electron transport chain in the mitochondria<br />

as well as an antioxidant in copper-zinc superoxide dismutase (CuZnSOD).<br />

However, in its free form, copper can also participate in Fenton-like reactions that<br />

result in the production <strong>of</strong> reactive hydroxyl radicals. In previous studies copper<br />

levels in rodent liver were increased following exposure to aryl-hydrocarbon receptor<br />

agonists, including the most potent polychlorinated biphenyl (PCB) congener,<br />

PCB 126. Increased hepatic copper was accompanied by several histological<br />

changes, including steatosis. In an effort to assess the effect <strong>of</strong> dietary copper on<br />

these changes, male Sprague-Dawley rats were fed an AIN-93G diet with one <strong>of</strong><br />

three dietary copper levels: low (2 ppm), adequate (6 ppm), and high (10 ppm).<br />

After three weeks, rats from each dietary group were given a single ip injection <strong>of</strong><br />

corn oil (control), 1, or 5 μmol/kg body weight PCB126 in corn oil, followed two<br />

weeks later by euthanization. Growth rate was slowed by PCB 126 in a dose-dependent<br />

manner, significantly at the highest dose (40-75%). Relative liver weight<br />

was increased in a dose-dependent manner (30-65%) by PCB 126, while there was<br />

no effect on growth or liver weight by dietary copper. Hepatic cytochrome P450 activity<br />

was maximally-induced by 1 μmol/kg PCB 126. Increasing dietary copper<br />

and increasing dose <strong>of</strong> PCB 126 both increased hepatic copper levels. Blood copper<br />

and serum ceruloplasmin levels were biphasic, diminishing with increasing PCB<br />

126 dose at low dietary copper, while increasing in rats fed high dietary copper.<br />

Hepatic CuZnSOD activity was not significantly affected by dietary copper. We<br />

conclude that while reducing dietary copper reduced hepatic copper levels, the<br />

function <strong>of</strong> essential copper enzymes were not negatively affected. (Supported by<br />

NIEHS P42 ES 013661)<br />

1104 ENTEROHEPATIC CIRCADIAN RHYTHM IN MICE:<br />

CONTRIBUTIONS OF BILE ACIDS.<br />

Y. J. Zhang, G. L. Guo and C. D. Klaassen. Pharmacology, <strong>Toxicology</strong>, and<br />

<strong>The</strong>rapeutics, University <strong>of</strong> Kansas Medical Center, Kansas City, KS.<br />

Diurnal fluctuation <strong>of</strong> bile acid (BA) concentration in the enterohepatic system <strong>of</strong><br />

mammals has been known for a long time. Recently, BAs have been shown as signaling<br />

molecules beyond their well-established roles in dietary lipid absorption and<br />

cholesterol homeostasis. <strong>The</strong> current study depicts diurnal variation <strong>of</strong> individual<br />

BAs detected by ultra-performance liquid chromatography/mass spectrometry<br />

(UPLC/MS) in serum and livers collected from C57BL/6 mice fed regular chow or<br />

chow containing cholestyramine (resin). Circadian rhythms <strong>of</strong> mRNA <strong>of</strong> vital BArelated<br />

nuclear receptors, enzymes, and transporters in livers and ilea were determined<br />

in control- and resin-fed mice, as well as in farnesoid X receptor (FXR) null<br />

mice. <strong>The</strong> circadian pr<strong>of</strong>iles <strong>of</strong> BAs show enhanced bacterial dehydroxylation during<br />

the fasting phase and efficient hepatic reconjugation <strong>of</strong> BAs in the fed phase.<br />

<strong>The</strong> resin removes more than 90% <strong>of</strong> BAs with β-hydroxy groups, such as muricholic<br />

acids and ursodeoxycholic acid, from serum and livers, but markedly increases<br />

deoxycholic acid and lithocholic acid in both compartments. Data from<br />

both resin-fed and FXR-null mouse models indicate that BAs regulate their own<br />

biosynthesis through the FXR-regulated ileal fibroblast growth factor 15. BA flux<br />

also influences the daily mRNA levels <strong>of</strong> multiple BA transporters. In conclusion:<br />

BA concentration and composition exhibit circadian variations in mouse liver and<br />

serum, which influences the circadian rhythms <strong>of</strong> BA metabolizing genes in liver<br />

and ileum. <strong>The</strong> diurnal variations <strong>of</strong> BAs appear to serve as a signal that coordinates<br />

daily nutrient metabolism in mammals. This study is supported by NIH grants<br />

DK-081461, ES-009716, ES-009649, ES-013714, and RR-021940.<br />

1105 MYELOPEROXIDASE-DEPENDENT METABOLISM OF<br />

P-CRESOL: A NOVEL PATHWAY FOR XENOBIOTIC<br />

AND ENDOBIOTIC BIOTRANSFORMATION DURING<br />

INFLAMMATION.<br />

J. Houghton 1 , K. Hayakawa 1 , D. DeGroot 2 , B. Zhao 2 , M. Denison 2 and J. P.<br />

Eiserich 1 . 1 Department <strong>of</strong> Internal Medicine, University <strong>of</strong> California, Davis, Davis,<br />

CA and 2 Department <strong>of</strong> Environmental <strong>Toxicology</strong>, University <strong>of</strong> California, Davis,<br />

Davis, CA.<br />

Myeloperoxidase (MPO) is a heme protein abundantly expressed in neutrophils<br />

that is traditionally regarded as a critical host defense enzyme that facilitates bacterial<br />

killing. Based upon its oxidizing P450-like behavior, we speculated that MPO<br />

236 SOT 2011 ANNUAL MEETING<br />

could play an unexpected role in xenobiotic and endobiotic metabolism. p-Cresol<br />

(4-methylphenol) is a natural product that humans are exposed to both exogenously<br />

(ie. cigarette smoke) and endogenously (ie. tyrosine metabolism by gut micr<strong>of</strong>lora).<br />

Herein we demonstrate that MPO is capable <strong>of</strong> converting p-cresol into<br />

nitrated and oxidized metabolites, the latter with bioactivity not shared with the<br />

parent compound. Enzymatic experiments with MPO revealed that p-cresol was<br />

converted primarily to 4α,9β-dihydro-8,9β-dimethyl-3(4H)-dibenz<strong>of</strong>uranone,<br />

otherwise known as “Pummerer’s ketone” (PK), with smaller yields <strong>of</strong> 2-nitro-4methylphenol,<br />

and 2,2’-dihydroxy-5,5’-dimethyldiphenyl but no chlorinated products.<br />

In vitro and cell culture approaches revealed that PK is a ligand for the AhR<br />

and activates its ability to bind to the dioxin-response element (DRE), and induces<br />

a 5-fold induction in the expression <strong>of</strong> cytochrome P450 1A1 mRNA.<br />

Additionally, PK induces the activation and nuclear translocation <strong>of</strong> the transcription<br />

factor Nrf-2, and consequently induces the expression <strong>of</strong> heme oxygenase-1<br />

(HO-1) and glutathione cysteine ligase catalytic (GCLC) subunit. Chemical reactions,<br />

coupled with LC/MS analyses, revealed that PK can react with the model<br />

thiol compounds glutathione (GSH) and N-acetylcysteine (NAC) to form covalent<br />

Michael addition products, suggesting that the cellular bioactivity <strong>of</strong> PK may be<br />

dictated by the formation <strong>of</strong> adducts with cysteine residues in critical proteins essential<br />

to transcriptional responses. Our data illustrate that MPO may play a previously<br />

unrecognized and important function in xenobiotic and endobiotic metabolism<br />

during inflammation.<br />

1106 TOXICITY, TOXICOGENOMIC, AND METABOLOMIC<br />

STUDY OF PENTAMETHYLCHROMANOL.<br />

T. Parman 1 , D. Bunin 1 , H. H. Ng 1 , T. Harrison 1 , J. McDunn 2 , I. M.<br />

Kapetanovic 3 and C. E. Green 1 . 1 Biosciences, SRI International, Menlo Park, CA,<br />

2 Metabolon, Inc., Durham, NC and 3 National Cancer Institute, Bethesda, MD.<br />

Pentamethyl-6-chromanol (PMCol) is a candidate chemopreventive agent against<br />

androgen dependent cancers. PMCol has been shown to cause hepatotoxicity,<br />

nephrotoxicity and hematological effects. <strong>The</strong> objectives <strong>of</strong> this study were to determine<br />

the mechanisms <strong>of</strong> toxicity and identify sensitive early markers <strong>of</strong> hepatic<br />

and renal injury using renal biomarkers (albumin, TIM-1, lipocalin-2, osteopontin,<br />

clusterin), toxicogenomics and metabolomic approaches. PMCol was administered<br />

to male Sprague-Dawley rats, 200 and 2000 mg/kg daily for 7 or 28 days. Changes<br />

in clinical chemistry included elevated alanine aminotransferase, total bilirubin,<br />

cholesterol and triglycerides, indicative <strong>of</strong> liver toxicity that was confirmed by microscopic<br />

findings (periportal hepatocellular hydropic degeneration and cytomegaly)<br />

in treated rats. Metabolomic evaluations <strong>of</strong> liver tissue revealed time and<br />

dose dependent changes including depletion <strong>of</strong> glutathione and glutathione conjugates,<br />

decreased methionine, and increased S-adenosylhomocysteine, cysteine, and<br />

cystine. PMCol treatment also decreased c<strong>of</strong>actor levels, e.g. FAD+ and NAD(P)+.<br />

Cholesterol and sphingomyelin were markedly higher in plasma from treated rats.<br />

Microarray analysis found that differentially expressed genes were enriched in the<br />

glutathione and cytochrome P450 pathways in liver by PMcol treatment. RTqPCR<br />

<strong>of</strong> six upregulated genes (Akr7a3, Cyp1a1, Gpx2, Gsr, Gstp1, and Hmox1)<br />

and one down regulated gene (Acnat2) confirmed the microarray results.<br />

Metabolomic, urine biomarkers and histopathologic changes in kidneys were also<br />

observed but were minor compared to hepatic effects. In conclusion, metabolomics<br />

and toxicogenomics demonstrate that chronic exposure to high doses <strong>of</strong> PMCol induces<br />

liver damage and dysfunction, probably due to both inhibition <strong>of</strong> synthesis<br />

and depletion <strong>of</strong> liver glutathione, as well as modification <strong>of</strong> other drug metabolism<br />

pathways. (Supported by NCI Contract HHSSN261200433005C)<br />

1107 EVIDENCE OF PEPPERMINT OIL SKIN<br />

PENETRATION AND CYTOTOXICITY ON RAT<br />

HEPATOCYTES IN VITRO.<br />

M. Dong 1 , O. Weber 1 , E. Di Lenarda 1 , P. End 2 , S. Chibout 1 , A. Wolf 1 and F.<br />

Pognan 1 . 1 Preclinical Safety, Novartis, Basel, Switzerland and 2 Drug Metabolism and<br />

Pharmacokinetics, Novartis, Basel, Switzerland.<br />

Peppermint oil is commonly used for digestive disorders taken orally and the relief<br />

<strong>of</strong> muscle pain by topical application. However, it has been suspected <strong>of</strong> potential<br />

adverse effects, including liver toxicity. <strong>The</strong> aim <strong>of</strong> the present study was to examine<br />

the acute effects <strong>of</strong> peppermint oil on rat hepatocytes in vitro.<br />

Freshly isolated rat hepatocytes were cultured in collagen-sandwich configuration.<br />

Cells were treated daily with peppermint oil (China Oil) at the concentrations<br />

ranging from 0.001% to 0.3% (v/v) for up to three days. GC/MS analysis determined<br />

that this peppermint oil contained 1.18 % <strong>of</strong> pulegone and 0.78% <strong>of</strong> its<br />

main metabolite, ment<strong>of</strong>urane. Both have been described as hepatotoxicants in<br />

vivo in rat. Cellular ATP content, total glutathione, as well as ALT and AST release<br />

were determined.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!