27.07.2013 Views

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

events in the liver. AZD9056 was evaluated in vitro in the Ames test and the mouse<br />

lymphoma assay and in vivo in a rat micronucleus study and UDS study. <strong>The</strong>re was<br />

no evidence <strong>of</strong> genotoxicity seen in any <strong>of</strong> these studies. In further studies rats<br />

dosed with AZD9056 were evaluated for cellular proliferation in hepatic foci by co<br />

administration <strong>of</strong> BrdU via osmotic minipumps. A marginal increase in hepatocyte<br />

proliferation, as determined by BrdU incorporation was detected in animals dosed<br />

at 125 mg/kg/day. <strong>The</strong>re were no effects on hepatocyte cell proliferation at doses up<br />

to 75 mg/kg/day. Foci <strong>of</strong> altered hepatocyte morphology and histopathological<br />

staining are a naturally occurring age related finding in the rat. AZD9056 at doses<br />

<strong>of</strong> 75 mg/kg/day for 6 months exacerbates the formation <strong>of</strong> these foci in a dose dependent<br />

manner. <strong>The</strong>re is no evidence <strong>of</strong> a genotoxic change that may be related to<br />

these findings and cellular proliferation has a clear no effect level. It was concluded<br />

that these findings have no impact on clinical safety <strong>of</strong> AZD9056.<br />

1099 THE EFFECT OF VITAMIN E DEFICIENCY ON DCA-<br />

AND TCA-INDUCED OXIDATIVE STRESS IN THE<br />

LIVERS OF MALE B6C3F1 MICE FOLLOWING<br />

SUBCHRONIC EXPOSURE.<br />

J. Cearfoss and E. Hassoun. Pharmacology, University <strong>of</strong> Toledo, Toledo, OH.<br />

Dichloroacetate (DCA) and Trichloroacetate (TCA) are water disinfection by products<br />

that are known to induce hepatotoxicity/ hepatocarcinogenicity in rodents.<br />

Previous studies in our lab have indicated induction <strong>of</strong> various biomarkers <strong>of</strong> oxidative<br />

stress (OS) in the livers <strong>of</strong> mice after subchronic exposure. To assess the effect<br />

<strong>of</strong> marginal Vitamin E deficiency on TCA- and DCA-induced OS, two groups<br />

<strong>of</strong> male B6C3F1 mice were fed either normal diet supplemented with vitamin E<br />

(E-normal group), or diet that was not supplemented with the vitamin( E-deficient).<br />

<strong>The</strong> two groups were subdivided into subgroups that were treated subchronically,<br />

by gavage, with the following: water (control), 77 mg DCA/kg/day, or 77 mg<br />

TCA/kg/day. Mice were sacrificed and the livers were collected and assayed for biomarkers<br />

<strong>of</strong> OS, such as production <strong>of</strong> superoxide anion (SA), lipid peroxidation<br />

(LP) and DNA single strand breaks (SSBs), as well as for antioxidant enzyme activities,<br />

including the activities <strong>of</strong> superoxide dismutase (SOD), catalase (CAT) and<br />

glutathione peroxidase (GSH-PX). DCA and TCA produced significant increases<br />

in the production <strong>of</strong> SA, LP and DNA-SSBs in the two groups when compared<br />

with the corresponding controls <strong>of</strong> each groups. However, the increases in those<br />

biomarkers were less significant in the E-deficient group when compared with the<br />

E-normal group. While DCA and TCA treatment resulted in SOD and GSH-Px<br />

inhibition in the E-normal group, they induced those enzyme activities in the Edeficient<br />

group. CAT activity on the other hand was not significantly changed<br />

when comparing various treatments in the E-deficient with the E-normal group.<br />

<strong>The</strong>se results suggest that marginal vitamin E deficiency in mice results in compensatory<br />

increases in antioxidant enzyme activities to provide partial protection<br />

against DCA- and TCA-induced OS. (Supported by NIH/NIEHS grant #<br />

R15ES013706-01A2)<br />

1100 O- AND N-DEMETHYLATION OF LEVOMETHORPHAN<br />

BY HUMAN CYTOCHROME P450 ENZYMES.<br />

M. Sunouchi 1 , A. Miyajima-Tabata 2 , R. Kikura-Hanajiri 3 and Y. Goda 3 .<br />

1 Division <strong>of</strong> Pharmacology, National Institute <strong>of</strong> Health Sciences, Tokyo 158-8501,<br />

Japan, 2 Division <strong>of</strong> Medical Devices, National Institute <strong>of</strong> Health Sciences, Tokyo<br />

158-8501, Japan and 3 Division <strong>of</strong> Pharmacognosy and Phytochemistry, National<br />

Institute <strong>of</strong> Health Sciences, Tokyo 158-8501, Japan. Sponsor: A. Hirose.<br />

Levomethorphan is the enantiomer <strong>of</strong> dextromethorphan. Dextromethorphan is an<br />

antitussive medicine and its metabolism has been reported well, whereas levomethorphan<br />

is an opioid analgesic with few metabolism studies. We have established<br />

the chiral analytical method for dextromethorphan, levomethorphan and<br />

their metabolites in biological samples using LC-MS/MS. In this study, we investigated<br />

the metabolic properties <strong>of</strong> levomethorphan by human and rat drug metabolizing<br />

enzymes and compared with the metabolism <strong>of</strong> dextromethorphan.<br />

<strong>The</strong>se drugs were incubated with rat or pooled human liver microsomes in<br />

NADPH generating system. Recombinant cytochrome P450 (CYP) 2D6 and<br />

CYP3A4 were also used for the particular analyses <strong>of</strong> the enantioselective metabolisms.<br />

<strong>The</strong> separation <strong>of</strong> these drugs and their metabolites was achieved on a Chiral<br />

CD-Ph column in a 0.1% formic acid-acetonitrile by a linear gradient program.<br />

MRM was used in the positive mode <strong>of</strong> an ESI-MS/MS for the quantitative analysis.<br />

After 20-min incubation with liver microsomes, levomethorphan was metabolized<br />

to O-demethyl and N-demethyl form by 63% and 15% (rat), 11% and 7.1%<br />

(human), respectively. <strong>The</strong> total amount <strong>of</strong> these metabolites from levomethorphan<br />

was higher than that from dextromethorphan both in rat (2.0 times) and human<br />

(3.1 times) microsomes. In the case <strong>of</strong> the incubation with CYP2D6, O-demethylation<br />

was the major metabolism pathway from levomethorphan as well as from<br />

dextromethorphan. O-demethylation <strong>of</strong> levomethorphan by CYP2D6 showed 2.3<br />

times higher than dextromethorphan. In the case <strong>of</strong> CYP3A4, only N-demethyled<br />

metabolites <strong>of</strong> these drugs were detected and N-demethylation <strong>of</strong> these drugs was<br />

showed almost similar degree. <strong>The</strong>se results suggest that the enantioselective metabolism<br />

<strong>of</strong> levomethorphan is mainly related to the O-demethylation by CYP2D6.<br />

1101 EFFECT OF ACARBOSE ON ALANINE<br />

AMINOTRANSFERASE 1 AND 2 PROTEINS AND<br />

GLUCONEOGENESIS IN RAT TISSUES.<br />

K. Kumagai, S. Arakawa, T. Matsuyama, K. Kai, M. Teranishi and A.<br />

Sanbuissho. Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd.,<br />

Shizuoka, Japan.<br />

Acarbose, an inhibitor <strong>of</strong> α-glucosidase and α-amylase, causes the elevation <strong>of</strong><br />

serum alanine aminotransferase (ALT) activity without any histopathological<br />

changes in rats. <strong>The</strong> mechanism for serum ALT elevation is still unclear, although it<br />

is assumed to be due to increased gluconeogenesis by the pharmacological effect <strong>of</strong><br />

acarbose. Rat ALT isozymes, ALT1 and ALT2, have been recently identified and<br />

have been reported that the two isozymes are expressed differently in tissues. <strong>The</strong><br />

present study investigated the effect <strong>of</strong> acarbose on rat ALT1 and ALT2 proteins in<br />

the serum and in the isozyme-expressed tissues: liver, heart, skeletal muscle, jejunum,<br />

and white and brown adipose tissues. <strong>The</strong> effect <strong>of</strong> acarbose on gluconeogenesis<br />

in the liver was also evaluated. Acarbose dissolved in distilled water was administered<br />

by gavage to male F344 rats at dose levels <strong>of</strong> 0 or 2000 mg/kg/day for 7<br />

days. <strong>The</strong> acarbose-treated rats had a significant increase in serum ALT activity<br />

compared with the control. Acarbose also induced increased serum levels in aspartate<br />

transferase and non-esterified fatty acid, and decreased serum levels in triacylglycerol<br />

and glucose. No histopathological changes by the acarbose treatment were<br />

observed in the liver, heart, skeletal muscle, jejunum, white or brown adipose tissues.<br />

Western blot analysis revealed that ALT1 protein levels in the serum and liver<br />

<strong>of</strong> acarbose-treated rats were significantly higher than those in the control. On the<br />

other hand, compared to the control, acarbose-administered rats had no increases<br />

in ALT1 proteins in other tissues or ALT2 proteins in any tissues. Moreover, serum<br />

ALT2 protein was not detected in either control or acarbose-administered rats.<br />

Phosphoenolpyruvate carboxykinase protein in the acarbose-treated rat livers was<br />

significantly increased compared to the control. In conclusion, the results <strong>of</strong> the<br />

present study suggest that acarbose causes increases in gluconeogenesis and ALT1<br />

protein in the liver, resulting in the elevation <strong>of</strong> ALT1 protein and ALT activity in<br />

the serum.<br />

1102 ENZYMES AND OXIDATIVE DAMAGE INDICATORS IN<br />

RATS FOLLOWING SUBACUTE OR SUBCHRONIC<br />

MANGANESE CHLORIDE EXPOSURE.<br />

P. Huang 1 , G. Li 2 and Z. Sun 1 . 1 Department <strong>of</strong> <strong>Toxicology</strong> and Sanitary Chemistry,<br />

School <strong>of</strong> Public Health and Family Medicine, Capital Medical University, Beijing,<br />

China and 2 Beijing Research Center for Preventive Medicine, Beijing, China.<br />

Sponsor: W. Zheng.<br />

Manganese (Mn) toxicity is most <strong>of</strong>ten found in mining and welding industry<br />

workers. Accumulation <strong>of</strong> manganese in the brain can result in a syndrome similar<br />

to that <strong>of</strong> Parkinson’s disease. Observations on former Mn-alloy workers suggested<br />

that residual effects could last for years after exposure. <strong>The</strong> objective <strong>of</strong> this study<br />

was to assess effects <strong>of</strong> Mn in the liver <strong>of</strong> rats following subacute or subchronic exposure<br />

and after recovery. Male Sprague-Dawley rats weighing 110 to 120 g were<br />

exposed to 6.0 mg Mn/kg b.w. using MnCl2 for 30 days, 90 days, or for 90 days<br />

followed by a 30-day post-exposure recovery period. Results showed that serum<br />

AST and ALT increased compared to controls. Mn concentrations in the liver <strong>of</strong><br />

treated animals increased 1.5 to 2.5-fold compared to controls. Similarly, concentrations<br />

<strong>of</strong> Ca and Cu increased in treated animals, while concentrations <strong>of</strong> Fe, Mg<br />

and Zn decreased in liver tissues after subacute or subchronic exposure. Subacute or<br />

subchronic Mn exposure also inhibited GPx activity, decreased GSH levels and increased<br />

MDA levels in liver tissues. All <strong>of</strong> the indicators returned to normal after<br />

the recovery period. We further evaluated the effects <strong>of</strong> manganese on rat hepatocyte<br />

mitochondria, membranes, and nuclei in the three periods. <strong>The</strong> results showed<br />

significant inhibition <strong>of</strong> mitochondrial SDH, cellular Na+,K+-ATPase,<br />

Ca2+,Mg2+-ATPase, Cu2+-ATPase, and GPx activities in hepatocytes. In addition,<br />

GSH levels decreased while MDA levels increased in hepatocyte mitochondria,<br />

membranes, and nuclei. All <strong>of</strong> these markers were normal after the recovery period.<br />

Taken together, these data suggest that MnCl2 exposure injured rat liver tissue, that<br />

the extent <strong>of</strong> injury is positively correlated with exposure time, and that the effect<br />

on mitochondria is presumably more serious than that on nuclei and membranes.<br />

Most biomarker alterations returned to pre-exposure levels by themselves after stopping<br />

Mn exposure.<br />

SOT 2011 ANNUAL MEETING 235

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

Saved successfully!

Ooh no, something went wrong!