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.

period. We have identified proteins implicated in bioactivation and detoxification<br />

pathways <strong>of</strong> acetaminophen and other relevant pharmaceutical agents that follow a<br />

diurnal variation in expression (e.g. cytochrome b5, cytokeratin-18,<br />

peroxiredoxin-6, aromatic-L-amino-acid decarboxylase and regucalcin). This<br />

dataset reveals new areas <strong>of</strong> investigation and testable hypotheses with which to explore<br />

the temporal relationship between circadian physiology and DILI.<br />

447 ACUTE LIVER TOXICITY OF ACETAMINOPHEN IS<br />

NOT POTENTIATED IN HCV TRANSGENIC MICE.<br />

T. Uehara 1 , O. Kosyk 1 , E. Jeannot 1 , B. Bradford 1 , J. Grimes 2 , T. O’Connell 2 ,<br />

G. Boorman 3 , S. Melnyk 4 , S. Weinman 5 and I. Rusyn 1 . 1 Department <strong>of</strong><br />

Environmental Sciences and Engineering, UNC, Chapel Hill, NC, 2 <strong>The</strong> Hamner<br />

Institutes, Research Triangle Park, NC, 3 Covance, Princeton, NJ, 4 University <strong>of</strong><br />

Arkansas for Medical Sciences, Little Rock, AR and 5 University <strong>of</strong> Kansas Medical<br />

center, Kansas City, KS.<br />

<strong>The</strong> global burden <strong>of</strong> hepatitis C virus (HCV) infection is increasing and chronic<br />

liver disease in subjects positive for HCV is becoming a significant public health<br />

concern in the developed and developing countries alike. While HCV infection<br />

may result in steatohepatitis, cirrhosis and hepatocellular carcinoma, it has been<br />

shown recently that it also potentiates the hepatotoxicity <strong>of</strong> acetaminophen<br />

(APAP), a drug which is a leading cause <strong>of</strong> acute liver toxicity. Since the mechanisms<br />

<strong>of</strong> such potentiation are not known, we hypothesized that HCV-Tg mice<br />

may be more susceptible to APAP hepatotoxicity and that an animal model may be<br />

established to elucidate the mechanisms for co-morbidity. Mice expressing core, E1,<br />

E2 and p7 proteins (HCV-Tg) and wild-type C57BL/6J mice were treated with a<br />

single dose (300 mg/kg) <strong>of</strong> APAP and liver toxicity was evaluated at 4 and 24 hrs<br />

after dosing. Acute liver injury due to APAP was not exacerbated in HCV-Tg mice<br />

and there were few differences between wild type and HCV-Tg in markers <strong>of</strong> oxidative<br />

stress, inflammation, or ER-stress in the liver. Interestingly, the amount <strong>of</strong><br />

mitochondrial total and reduced glutathione was elevated in HCV-Tg mice, and<br />

was decreased upon APAP treatment, an effect not observed in wild-type mice. This<br />

study shows that the extent <strong>of</strong> liver disease in HCV-Tg mice over-expressing core,<br />

E1, E2 and p7 alone are not sufficient to produce the sensitivity to APAP that has<br />

been observed clinically. However, our findings move the field forward by narrowing<br />

down the possibilities <strong>of</strong> how HCV affects drug toxicity. Our data suggest that<br />

either the nonstructural viral proteins are critical for the hyper-sensitivity to druginduced<br />

liver injury in humans, or that it may be due to an immune component<br />

that is difficult to reproduce in HCV-Tg mice.<br />

448 INTRAVITAL IMAGING OF ACETAMINOPHEN (APAP)<br />

HEPATOTOXICITY.<br />

J. Hu 1 , V. K. Ramshesh 1 , H. Jaeschke 2 and J. J. Lemasters 1 . 1 Medical University<br />

<strong>of</strong> South Carolina, Charleston, SC and 2 University <strong>of</strong> Kansas Medical Center, Kansas<br />

City, KS.<br />

APAP overdose causes liver injury involving mitochondrial dysfunction and onset<br />

<strong>of</strong> the mitochondrial permeability transition (MPT). NIM811 (N-methyl-4isoleucine<br />

cyclosporin) is a nonimmunosuppressive derivative <strong>of</strong> cyclosporin A that<br />

inhibits the MPT in vitro and in vivo. Monitoring mitochondrial function in living<br />

animals has been difficult. Recent developments in intravital confocal/multiphoton<br />

microscopy provide a novel approach to visualize mitochondrial dysfunction and<br />

cell death in vivo. Here we have two aims. <strong>The</strong> first is to investigate whether<br />

NIM811 decreases APAP-induced liver toxicity. Our second aim is to understand<br />

the mechanisms <strong>of</strong> APAP hepatotoxicity and the effect <strong>of</strong> APAP on mitochondrial<br />

polarization and cell death in vivo. Male C57BL/6 mice were fasted overnight and<br />

then administered APAP (300 mg/kg, i.p.) or vehicle. NIM811 was gavaged (10<br />

mg/kg) 1 h before APAP. For imaging, the abdomen <strong>of</strong> each mouse was opened,<br />

and the exposed liver was positioned on a #1.5 glass coverslip mounted on the inverted<br />

stage <strong>of</strong> an Olympus FV1000 multiphoton microscope. Mitochondrial polarization<br />

and cell death were assessed from the respective green and red fluorescence<br />

<strong>of</strong> rhodamine 123 (Rh123) and propidium iodide(PI) using 820-nm<br />

multiphoton excitation and a 25X, 1.1 NA water-immersion objective lens. After<br />

vehicle, green Rh123 fluorescence was punctate in most hepatocytes, indicating mitochondrial<br />

polarization, and nuclear PI staining signifying cell death was absent.<br />

At 6 h after APAP, loss <strong>of</strong> mitochondrial Rh123 fluorescence occurred in pericentral<br />

hepatocyes <strong>of</strong>ten accompanied by PI labeling. NIM811 decreased both mitochondrial<br />

depolarization and cell death. In conclusion, NIM811 attenuates APAP-induced<br />

mitochondrial depolarization and necrotic cell death in mouse liver in vivo.<br />

<strong>The</strong>se results also illustrate the utility <strong>of</strong> intravital confocal/multiphoton microscopy<br />

as a powerful technology to study disruption <strong>of</strong> liver function in living animals<br />

due to APAP.<br />

96 SOT 2011 ANNUAL MEETING<br />

449 THE ALPHA MUPA MICE, A MODEL OF CALORIC<br />

RESTRICTION, ARE MORE SUSCEPTIBLE TO<br />

ACETAMINOPHEN HEPATOTOXICITY.<br />

Z. Fu 1 , Y. Zhang 1 , R. Miskin 2 and C. D. Klaassen 1 . 1 Department <strong>of</strong><br />

Pharmacology, <strong>Toxicology</strong> and <strong>The</strong>rapeutics, University <strong>of</strong> Kansas Medical Center,<br />

Kansas City, KS and 2 Department <strong>of</strong> Biological Chemistry, <strong>The</strong> Weizmann Institute <strong>of</strong><br />

Science, Rehovot, Israel.<br />

Murine urokinase plasminogen activator (αMUPA) transgenic mice spontaneously<br />

eat less and live longer (approximately 20%), therefore, they have been considered<br />

a caloric restriction (CR) model. Previous studies indicate that CR protects against<br />

the hepatotoxic effects <strong>of</strong> bleomycin and thioacetamide, whereas two other longlived<br />

models, GHR-KO and Snell dwarf mice, are more susceptible to acetaminophen<br />

hepatotoxicity. <strong>The</strong> goal <strong>of</strong> the present study was to examine the resistance <strong>of</strong><br />

αMUPA mice to acetaminophen (APAP). Male αMUPA mice and wild-type (WT)<br />

mice (14-months <strong>of</strong> age) were given a single dose <strong>of</strong> APAP (375 mg/kg, ip).<br />

Whereas 92% <strong>of</strong> WT mice were alive at 48h, the survival <strong>of</strong> αMUPA mice was only<br />

58% at 24h, and 29% at 48h. In αMUPA mice, APAP caused more severe liver injury,<br />

as indicated by higher serum levels <strong>of</strong> alanine aminotransferase (4.3-fold) and<br />

lactate dehydrogenase (5.7-fold), as well as more severe liver necrosis. In unchallenged<br />

αMUPA and WT mice, the mRNAs <strong>of</strong> Cyp2e1 and Cyp3a11, two enzymes<br />

important for converting APAP to its toxic intermediate, NAPQI, were similar.<br />

However, αMUPA mice exhibited higher mRNAs <strong>of</strong> some other phase-I drug metabolizing<br />

enzymes, such as Cyp1a1 (2-fold), Cyp1a2 (1.6-fold), and Cyp4a14 (2fold).<br />

In addition, αMUPA mice exhibited higher mRNAs <strong>of</strong> phase-II enzymes<br />

that detoxify APAP, Ugt1a1 (1.6-fold) and Ugt1a6 (1.6-fold); that detoxifies<br />

NAPQI, Gstp (2.9-fold); and hepatic transporters that transport xenobiotics and<br />

bile acids into liver, such as Oatp1a1 (1.6-fold) and Ntcp (1.7-fold). In summary,<br />

the present study shows that αMUPA mice exhibit similar expression <strong>of</strong> key enzymes<br />

for APAP bioactivation, and higher expression <strong>of</strong> enzymes for detoxifying<br />

APAP and NAPQI. <strong>The</strong>refore, the higher susceptibility <strong>of</strong> αMUPA mice to APAP<br />

hepatotoxicity is likely due to differences in toxicodynamics, rather than toxicokinetics.<br />

(Supported by NIH grants ES-09649, ES-09716, ES-07079, DK-081461,<br />

and RR-021940)<br />

450 ACTIVATION OF CASPASES DURING<br />

ACETAMINOPHEN TOXICITY IS A STRAIN<br />

DEPENDENT PHENOMENON.<br />

H. Jaeschke, M. Koerner and C. D. Williams. University <strong>of</strong> Kansas Medical Center,<br />

Kansas City, KS.<br />

Acetaminophen (APAP)-induced liver injury resulting from overdose is the most<br />

frequent cause <strong>of</strong> acute hepatic failure in the US. <strong>The</strong> mechanisms <strong>of</strong> APAP-mediated<br />

cell death have been extensively characterized in both laboratory animals and<br />

man and it is generally accepted that liver cells die by oncotic necrosis (Gujral et al.,<br />

Toxicol Sci. 2002;67:322-8). Recently it was demonstrated in fed but not in fasted<br />

CD-1 mice (outbred strain) that there is transient caspase activation after APAP<br />

overdose. It was speculated that apoptosis is limited in APAP hepatotoxicity due to<br />

the use <strong>of</strong> fasted animals (Antoine et al., Toxicol Sci. 2009;112:521-31). To evaluate<br />

these findings in more detail, APAP-induced liver injury in an outbred strain<br />

(Swiss Webster, SW) was compared with the more commonly used inbred mouse<br />

strain (C57BL/6). In these studies fed and fasted mice were treated with 530 mg/kg<br />

APAP for 3, 5 and 24h with or without pan-caspase inhibitor (10 mg/kg Z-VDfmk).<br />

As a positive control for caspase-dependent apoptosis, mice were treated with<br />

700 mg/kg galactosamine (GalN) and 100 μg/kg endotoxin (ET). Fasted or fed<br />

APAP-treated C57BL/6 mice showed no evidence <strong>of</strong> caspase-3 processing by western<br />

blot or caspase-3 activity by Ac-DEVD-AFC fluorogenic assay at any timepoint.<br />

Interestingly, a minor, temporary increase in caspase-3 processing and enzyme<br />

activity (250% above baseline) was observed after APAP treatment in SW<br />

mice, but this could be observed in both fed and fasted animals. Z-VD-fmk in vivo<br />

partially attenuated this caspase activation but did not alter injury (plasma ALT,<br />

area <strong>of</strong> necrosis). <strong>The</strong> degree <strong>of</strong> caspase-3 processing and activation in SW mice<br />

after APAP was much less than that observed in the GalN/ET-treated mice (1700%<br />

above baseline). Conclusion: Caspase-3 processing and activation after APAP-overdose<br />

can be observed only in some outbred mouse strains independent <strong>of</strong> the fed or<br />

fasted state. In contrast to the GalN/ET-induced apoptosis, this transient, minor<br />

caspase activation does not seem to impact the overall liver injury and toxicity during<br />

APAP overdose.

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

Saved successfully!

Ooh no, something went wrong!