27.09.2014 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.

1963 CHARACTERIZATION OF HEPATOTOXICITY<br />

INDUCED BY 1-FURAN-2-YL-3-PYRIDIN-2-YL-<br />

PROPENONE, A NEW ANTI-INFLAMMATORY AGENT.<br />

H. W. Ha 1 , T. W. Jeon 2 , G. S. Ko 1 , J. W. Yoo 1 , S. K. Lee 1 , M. J. Kang 1 , E. S.<br />

Lee 1 and T. Jeong 1 . 1 Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk,<br />

Republic <strong>of</strong> Korea and 2 BioToxtech Incorporation, Ochang, Republic <strong>of</strong> Korea.<br />

1-Furan-2-yl-3-pyridin-2-yl-propenone (FPP-3) has recently been synthesized as<br />

an anti-inflammatory activity through the inhibition <strong>of</strong> the production <strong>of</strong> nitric<br />

oxide. In the present study, adverse effects <strong>of</strong> FPP-3 on hepatic functions were determined<br />

in female BALB/c mice. When mice were administered with FPP-3 at<br />

125, 250 or 500 mg/kg for 7 consecutive days orally, FPP-3 significantly increased<br />

absolute and relative weights <strong>of</strong> liver with a dose-dependent manner. In addition,<br />

FPP-3 administration dramatically increased the hepatotoxicity parameters in<br />

serum at 500 mg/kg, in association <strong>of</strong> hepatic necrosis. FPP-3 significantly induced<br />

several phase I enzyme activities. To elucidate the possible mechanism(s) involved<br />

in FPP-3 induced hepatotoxicity, we investigated the hepatic activities <strong>of</strong> free radical<br />

generating and scavenging enzymes and the level <strong>of</strong> hepatic lipid peroxidation.<br />

FPP-3 treatment significantly elevated the hepatic lipid peroxidation, measured as<br />

the thiobarbituric acid-reactive substance, and the activity <strong>of</strong> superoxide dismutase.<br />

Taken together, the present data indicated that reactive oxygen species might be involved<br />

in FPP-3-induced hepatotoxicity.<br />

1964 ALGINATE SPONGES AS PHYSIOLOGICALLY<br />

RELEVANT CULTURE ENVIRONMENTS FOR PRIMARY<br />

RAT HEPATOCYTES.<br />

A. Sams, Z. Li, M. Gonzalez, J. Jackson, J. Hill and M. Powers. PSCS R&D,<br />

Invitrogen Corporation, Frederick, MD.<br />

We have developed a lyophilized porous alginate matrix (AlgiMatrix) to establish<br />

a physiologically relevant environment that facilitates the formation <strong>of</strong> more in<br />

vivo-like tissue structures. <strong>The</strong> addition <strong>of</strong> divalent cationic salts (e.g. calcium, barium)<br />

to a sodium alginate solution produces a highly porous (>90%) and highly interconnected<br />

biocompatible scaffold. Using multiple cell types we have shown that<br />

the matrix pores (50-150μm) act as localized compartments in which cells are able<br />

to interact with one another (because cells do not significantly interact with the alginate)<br />

to form 3D multicellular spheroidal structures. <strong>The</strong> technology is <strong>of</strong> particular<br />

interest in the field <strong>of</strong> liver biology; hepatocellular spheroids have been shown<br />

to maintain expression <strong>of</strong> key hepatic markers. However, these spheroids are traditionally<br />

somewhat cumbersome to culture and maintain. Phase contrast microscopy<br />

reveals that spheroid formation is faster and more efficient in<br />

AlgiMatrix cultures compared to other currently available platforms (e.g. lowbind<br />

polystyrene). Live/dead staining and intracellular ATP levels confirm that<br />

AlgiMatrix cultures <strong>of</strong> primary rat hepatocytes retain a high viability through at<br />

least 14 days in culture. Cellular toxicity measurements, including intracellular reduced<br />

glutathione levels and caspase-3 activity, demonstrate that AlgiMatrix<br />

provides a more suitable culture environment for primary hepatocytes in comparison<br />

to conventional sandwich culture. AlgiMatrix cultures <strong>of</strong> primary rat hepatocytes<br />

demonstrate enhanced basal cytochrome P450 (e.g. CYP1A, 2B, 3A) activity<br />

and gene expression compared to conventional sandwich culture. Collectively,<br />

these results suggest that AlgiMatrix is a potentially useful predictive tool in<br />

xenobiotic metabolism and toxicology research. We conclude that 3D cell culture<br />

scaffolds such as AlgiMatrix serve as a foundation for the creation <strong>of</strong> more physiologically<br />

relevant culture systems.<br />

1965 CHARACTERIZATION OF AN IMMORTALIZED<br />

MURINE KUPFFER CELL LINE FOR USE IN<br />

TOXICOLOGICAL STUDIES.<br />

Z. Wang, J. E. Klaunig, B. Hocevar and L. M. Kamendulis. Pharmacology &<br />

Toxicolgy, Center for Environmental Health, Indiana University School <strong>of</strong> Medicine,<br />

Indianapolis, IN.<br />

Kupffer cells play a critical role in both liver physiology and the pathogenesis <strong>of</strong> various<br />

liver diseases. Isolated primary Kupffer cells have a limited lifespan in culture,<br />

and due to the relatively low number obtained, limit their study in vitro. Here, an<br />

immortalized murine Kupffer cell line was established from transgenic mice that express<br />

the thermolabile mutant tsA58 <strong>of</strong> the Simian virus 40 large T antigen under<br />

the control <strong>of</strong> the H-2kb promoter. Primary Kupffer cells were obtained using a<br />

three step procedure: liver perfusion, centrifugal elutriation, and sorting for F4/80+<br />

cells. A subpopulation <strong>of</strong> cells (ImKC) was identified within the small-intermediate<br />

population <strong>of</strong> sorted Kupffer cells that maintained stable expression <strong>of</strong> the F4/80<br />

and the surface antigens CD11b, CD14 and CD80. ImKCs maintained proliferative<br />

capacity at 37C and exhibited a doubling time <strong>of</strong> ~24 hours when cultured in<br />

RPMI 1640 with 5% FBS. ImKC cells maintained a high capacity to phagocytose<br />

FITC-latex beads, in response to lipopolysaccharide (LPS; 0.1-1μg/ml). In addition,<br />

similar to primary Kupffer cells, LPS treatment <strong>of</strong> ImKCs caused upregulation<br />

<strong>of</strong> TNFα (23-fold), IL-6 (28-fold), IL-1β (1459-fold), IL-10 (14-fold), inducible<br />

nitric oxide synthase (iNOS) (11-fold) mRNA levels as measured by qRT-PCR.<br />

Protein levels <strong>of</strong> TNFα were also increased 10-fold; and ROS and TLR4 expression<br />

were enhanced 2-fold and 2.6- fold, respectively. Furthermore, LPS caused a<br />

marked increase in mitogen activated protein kinase (MAPK) phospho- (ERK1/2,<br />

JNK) and NFkB p50 with decreased IκBα as assessed by Western blot. Moreover,<br />

ethanol-pretreatment sensitized ImKCs to LPS resulting in enhanced TNFα production<br />

through p38 activation, a finding not seen in the RAW 246.7 macrophage<br />

cell line. Collectively, the ImKC line retains critical characteristics <strong>of</strong> primary<br />

Kupffer cells, and thus provide a useful cell line to assess the role <strong>of</strong> Kupffer cells in<br />

liver injury and chronic diseases (Supported in part by NIH CA100908).<br />

1966 IN VITRO 3-D LIVER CO-CULTURES FOR ADMET<br />

ASSESSMENT OF NEW COMPOUNDS.<br />

D. R. Applegate, L. New and B. A. Naughton. RegeneMed Inc., San Diego, CA.<br />

Sponsor: R. Thomas.<br />

Many pharmaceutical companies are screening toxicity <strong>of</strong> drug candidates earlier in<br />

the discovery process to increase success in later stages <strong>of</strong> drug development, <strong>of</strong>ten<br />

using primary hepatocytes as an in vitro model <strong>of</strong> in vivo liver function. However,<br />

primary hepatocytes lose P450 activity and albumin synthesis within a few days <strong>of</strong><br />

culture, and thus better in vitro models are needed. Three-dimensional (3D) tissue<br />

technology was developed that provides a more physiologically relevant in vitro<br />

liver by culturing all liver cells on a 3D scaffold to form a tissue with long-term<br />

function. <strong>The</strong>se cultures maintain liver specific function in culture for months, including<br />

liver-specific protein expression, extracellular matrix protein deposition, receptor<br />

activity, cytochrome P450 metabolism, induction and inhibition, and biliary<br />

clearance. <strong>The</strong>se functions have been previously published to 3 months in<br />

culture and are now verified to 6 months including incorporation <strong>of</strong> recent assay<br />

methods including genomics, proteomics, metabolomics, 3D imaging through the<br />

tissue depth, and multiplexed assays. <strong>The</strong>se 3D liver co-cultures have been grown<br />

from a variety <strong>of</strong> species including mouse, rat, dog, nonhuman primate and human<br />

liver donors in multiwell plates from 1 to 96 well, with 384 and 1536 well formats<br />

in development. <strong>The</strong> liver co-cultures and assay methods serve as animal alternatives<br />

in accelerating drug discovery.<br />

1967 PHENOBARBITAL INDUCES TRANSCRIPTIONAL AND<br />

FUNCTIONAL CHANGES WITH TIME IN 3-D LIVER<br />

CO-CULTURES CONSISTENT WITH<br />

HEPATOCARCINOGENESIS.<br />

R. Brennan 3 , L. New 1 , R. S. Thomas 2 , C. I. Pearson 1 , A. H. Roter 4 , B. A.<br />

Naughton 1 and D. R. Applegate 1 . 1 Research, RegeneMed Inc., San Diego, CA,<br />

2<br />

Genomic Biology and Bioinformatics, <strong>The</strong> Hamner Institutes for Health Sciences,<br />

Research Triangle Park, NC, 3 Research, GeneGo, Inc., Encinitas, CA and 4 Research,<br />

Entelos, Inc., Foster City, CA.<br />

Many non-genotoxic compounds are tested for carcinogenic potential using the<br />

“gold stardard” two year rodent bioassays. Because <strong>of</strong> its high cost and lengthy time,<br />

many chemicals <strong>of</strong> concern have not been tested. Moreover, while rodent carcinogens<br />

are generally considered human carcinogens, there are exceptions. Less costly,<br />

high throughput assays are needed. A three-dimensional (3D) in vitro organotypic<br />

liver co-culture provides a physiologically relevant model <strong>of</strong> intact liver by culturing<br />

all hepatic cells on a 3D scaffold. <strong>The</strong>se co-cultures form a tissue that maintains<br />

liver functions including cytochrome P450 induction for months. To test their potential<br />

in carcinogenicity assessment, 3D co-cultures were exposed to<br />

Phenobarbital, a non-genotoxic hepatocarcinogen, for three weeks. Cell proliferation<br />

measured by tritiated thymidine uptake occurred in mouse, rat, and human<br />

co-cultures. Microarray data showed upregulation <strong>of</strong> Cyp2B and Cyp3A transcripts<br />

and other CAR-inducible transcripts within the first 24 hours <strong>of</strong> exposure, and<br />

maintained for at least 3 days. Transcriptional pr<strong>of</strong>iles <strong>of</strong> cell cycle pathways indicated<br />

cell cycle progression and DNA replication at early time points in male rats,<br />

whereas by 18 days, cell cycle arrest and oxidative stress were observed. Remodeling<br />

<strong>of</strong> the male 3D co-cultures by day 18 was indicated by up-regulation <strong>of</strong> genes associated<br />

with embryonic development, nervous system development, cell differentiation,<br />

and GPCR activity, and down-regulation <strong>of</strong> collagens, CAR-inducible genes,<br />

and acute phase response genes. <strong>The</strong>se functional and transcriptional studies indicate<br />

that 3D liver mirrors known in vivo effects leading to tumor formation, such as<br />

CAR activation, cell proliferation, and oxidative stress, providing a much-needed in<br />

vitro model suited for HTS assessment <strong>of</strong> hepatocarcinogenic potential.<br />

418 SOT 2010 ANNUAL MEETING

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

Saved successfully!

Ooh no, something went wrong!