27.09.2014 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.

1203 ARYL HYDROCARBON RECEPTOR-DNA BINDING<br />

AND FUNCTION INVOLVING A NOVEL REGULATORY<br />

ELEMENT INDEPENDENT OF THE ARNT PROTEIN.<br />

G. Huang and C. Elferink. Pharmacology and <strong>Toxicology</strong>, University <strong>of</strong> Texas<br />

Medical Branch, Galveston, TX.<br />

<strong>The</strong> aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor capable<br />

<strong>of</strong> modulating gene expression by binding to a well characterized xenobiotic<br />

response element (XRE), in response to agonists such as tetrachlorodibenzo-pdioxin<br />

(TCDD). However, DNA microarray experiments performed in several laboratories<br />

including ours, identified numerous AhR target genes lacking an obvious<br />

XRE in the promoter or flanking regulatory regions. Our examination <strong>of</strong> one such<br />

gene, encoding plasminogen activator inhibitor type 1, identified and characterized<br />

a new non-consensus XRE (NC-XRE) that binds the AhR independently <strong>of</strong> the<br />

canonical Arnt protein dimerization partner. Luciferase reporter expression studies<br />

and electrophoretic mobility shift assays confirmed AhR involvement and defined<br />

key nucleotides within the NC-XRE necessary for binding and function.<br />

Chromatin immunoprecipitation and shRNA-mediated Arnt protein knock-down<br />

studies demonstrated that AhR-mediated activity through the NC-XRE does not<br />

require the Arnt protein. Collectively, the evidence suggests that the NC-XRE represents<br />

a novel AhR binding site that defines a subset <strong>of</strong> target genes responsive to<br />

TCDD, distinct from the classical AhR/Arnt-regulated genes. Supported by the<br />

NIEHS grants ES007800 and ES012018.<br />

1204 TCDD INDUCED PERICARDIAL EDEMA AND<br />

RELATIVE COX-2 EXPRESSION IN MEDAKA EMBRYOS.<br />

W. Dong 1, 2 , F. Matsumura 3 and S. W. Kullman 1 . 1 Environmental and Molecular<br />

<strong>Toxicology</strong>, North Carolina State University, Raleigh, NC, 2 College <strong>of</strong> Marine<br />

Science, HuaiHai Institute <strong>of</strong> Technology, LianyunGang, Jiangsu, China and<br />

3<br />

Environmental <strong>Toxicology</strong>, University <strong>of</strong> California Davis, Davis, CA.<br />

Dioxin and other AhR ligands are well known to result in multiple defined developmental<br />

phenotypes including pericardial edema and circulation failure in small<br />

aquarium fish models. While these phenotypes are well describe, the mechanistic<br />

underpinning behind these toxicities manly remain elusive. To date, much <strong>of</strong><br />

2,3,7,8-tetrachlorodibenzo- p -dioxin (TCDD) toxicity is thought to be due to the<br />

classical genomic actions <strong>of</strong> AhR/ARNT transcription factor complex. Here we<br />

suggest that AhR additionally functions through a non-classical, non-genomic<br />

mechanism(s) involving activation <strong>of</strong> inflammation and eicosanoid pathways. We<br />

demonstrate that medaka embryos exposed to TCDD (0.5-1 ppb) during early development<br />

result in a dose related increase in the prevalence <strong>of</strong> pericardial edema<br />

and that this phenotype correlates with modification <strong>of</strong> both Cyclooxygenase-2<br />

(Cox-2) activity and gene expression in exposed medaka embryos. TCDD exposure<br />

significantly induces Cox-2 and CYP1A1 mRNA in all treated embryos. Those individuals<br />

exhibiting the edema phenotype were found to have significantly greater<br />

abundance <strong>of</strong> Cox-2 mRNA than the non-edema cohort. Biochemical inhibition <strong>of</strong><br />

Cox-2 with NS-398 a selective Cox-2 inhibitor, significantly attenuates the prevalence<br />

and severity <strong>of</strong> edema phenotype. Subsequently, exposures <strong>of</strong> medaka embryos<br />

to Arachidonic Acid (AA) resulted in recapitulation <strong>of</strong> the pericardial edema<br />

phenotype and significantly increased Cox-2 expression only in those individuals<br />

exhibiting pericardial edema compared to the non-edema cohort. Interestingly, AA<br />

exposure does not result in any significant induction <strong>of</strong> CYP1A expression. Taken<br />

together, these results indicate a putative correlation between TCDD induced pericardial<br />

edema and relative cox-2 mediated prostaglandin pathway in the medaka<br />

embryos. This work was supported by NCARS (02225), NCET-04-0262 and<br />

NSFC (30360090)<br />

1205 A RAPID, CELL-BASED BIOLUMINESCENT ARYL<br />

HYDROCARBON RECEPTOR ACTIVATION ASSAY<br />

THAT USES CYP1A1 ENZYME ACTIVITY AS A<br />

SELECTIVE MARKER ACTIVITY.<br />

J. Cali, D. Ma and P. Meisenheimer. Research and Development, Promega<br />

Corporation, Madison, WI. Sponsor: A. Li.<br />

<strong>The</strong> aryl hydrocarbon nuclear receptor is activated for positive transcriptional regulation<br />

by small molecule drugs and toxins. An example <strong>of</strong> an AHR responsive gene<br />

is cytochrome P450 1A1 and this gene can be used as a marker for AHR activity.<br />

CYP1A1 expression can be measured at the levels <strong>of</strong> mRNA, protein expression and<br />

enzyme activity. Here we describe a rapid, non-destructive bioluminescent cell<br />

based assay for monitoring CYP1A1 enzyme activity as a marker <strong>of</strong> AHR activity.<br />

<strong>The</strong> assay uses a cell permeable bioluminogenic CYP1A1 substrate.<br />

Bioluminogenic substrates are proluciferins that are converted by the enzymes <strong>of</strong><br />

interest to a luciferin that is detected in a bioluminescent reaction with luciferase,<br />

correlating light output with target enzyme activity. <strong>The</strong> bioluminogenic CYP1A1<br />

substrate is a novel luciferin derivative that showed exquisite CYP1A1 selectivity<br />

over all other CYPs from a panel <strong>of</strong> 21 that were tested, including CYP1A2 and<br />

CYP1B1. <strong>The</strong> assay detected robust activations by known AHR agonists in a 96<br />

well format that left cells intact for further analysis. While induction data with this<br />

assay correlated well with conventional mRNA and protein analysis methods, the<br />

luminescent assays were simpler, more rapid and by leaving cells intact allowed for<br />

a multiplexed assay that measured cell viability and AHR activity from single culture<br />

well. This bioluminescent assay enables a high throughput cell-based method<br />

for screening <strong>of</strong> AHR agonists.<br />

1206 SYNERGY BETWEEN ARYL HYDROCARBON<br />

RECEPTOR AND CONSTITUTIVE ANDROSTANE<br />

RECEPTOR ACTIVATION PROMOTES MURINE LIVER<br />

HYPERPLASIA.<br />

S. Wilson 1 , K. Mitchell 2 and C. Elferink 1 . 1 Pharmacology and <strong>Toxicology</strong>,<br />

University <strong>of</strong> Texas Medical Branch, Galveston, TX and 2 Biological Sciences, Boise<br />

State University, New London, CT.<br />

Mechanisms <strong>of</strong> hepatocyte proliferation triggered by tissue loss are distinguishable<br />

from those that promote proliferation in the intact liver in response to mitogens.<br />

Previous studies demonstrate that exogenous activation <strong>of</strong> the aryl hydrocarbon receptor<br />

(AhR), a soluble ligand-activated transcription factor in the basic helix-loophelix<br />

family <strong>of</strong> proteins, suppresses compensatory liver regeneration elicited by surgical<br />

partial hepatectomy (PH). <strong>The</strong> goal <strong>of</strong> the present study was to determine how<br />

AhR activation modulates hepatocyte cell cycle progression in the intact liver following<br />

treatment with the hepatomitogen, 1,4-bis[2-(3,5-dichloropyridyloxy)]<br />

benzene (TCPOBOP). Mice were pretreated with the exogenous AhR agonist<br />

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) 24 hours prior to treatment with<br />

TCPOBOP (3 mg/kg), a potent agonist for the constitutive androstane receptor<br />

(CAR), which is known to induce hepatocyte proliferation. In contrast to the suppressive<br />

effects <strong>of</strong> AhR activation observed during compensatory regeneration,<br />

TCDD pretreatment resulted in a 30-50% increase in hepatocyte proliferation in<br />

the intact liver <strong>of</strong> TCPOBOP-treated mice. Pretreatment with TCDD suppressed<br />

CDK2 kinase activity and increased the association <strong>of</strong> CDK2 with negative regulatory<br />

proteins p21Cip1 and p27Kip1, yet increased CDK4/cyclin D1 association<br />

and CDK4 activity, which culminated in enhanced phosphorylation <strong>of</strong> retinoblastoma<br />

protein. Moreover, pretreatment with TCDD transiently increased levels <strong>of</strong><br />

CAR transcript and activity, which coincided with elevated levels <strong>of</strong> CAR protein<br />

and cyclin D1 expression. Based on these findings, we conclude that AhR activation<br />

potentiates TCPOBOP-stimulated hepatocyte proliferation through a mechanism<br />

that may include increased CDK4 kinase activity as well as elevated CAR expression<br />

and activity. Supported by the NIEHS Grant ES007800 (C. E.),<br />

F32ES013588 (K. M.), and F30ES016490 (S. W.).<br />

1207 TELOMERASE INHIBITION BY ARYL HYDROCARBON<br />

RECEPTOR (AHR) AGONISTS IN HUMAN MAMMARY<br />

EPITHELIAL CANCER CELLS.<br />

S. Pittet and M. Charbonneau. INRS-Institut-Armand-Frappier, Université du<br />

Québec, Laval, QC, Canada.<br />

Persistent organic pollutants (POP) are ubiquitous lipophilic chemicals. POP exposure<br />

has been suggested as a risk factor for breast cancer development based on<br />

some epidemiological and rodents studies. Other data suggest, however, that AhR<br />

agonists can cause anticarcinogenic effects. Telomerase expression is a key pro-carcinogenic<br />

component responsible for increased cancer cell longevity. This work<br />

aimed to test the effect <strong>of</strong> 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and other<br />

AhR agonists on cell proliferation and telomerase expression in human epithelial<br />

mammary cancer cells. TCDD (5.3 nM, 6 d) decreased cell proliferation in two<br />

AhR-positive mammary cell lines, the estrogen receptor (ER)-positive T47D (36 %<br />

lower) and the ER-negative MDA-MB-231 (26 % lower). Telomerase activity,<br />

measured with the TRAPeze® XL Telomerase Detection Kit, was significantly reduced<br />

in both cell lines, T47D (34 % lower) and MDA-MB-231 (26 % lower).<br />

Similar results were observed with mixtures <strong>of</strong> PCBs, PCDFs and PCDDs. TCDD<br />

exposure <strong>of</strong> the AhR-negative neuronal cell line, SH-SY5Y (ER-), did not affect cell<br />

proliferation rate nor telomerase activity. <strong>The</strong>se data suggest an AhR-dependent<br />

pathway. Western Blot analyses showed a significant 2-fold decrease in the cytoplasmic<br />

AhR level in TCDD-treated (5.3 nM, 24 h) T47D cells compared to controls<br />

(DMSO), suggesting AhR activation and its nuclear translocation. Real-Time PCR<br />

quantification <strong>of</strong> the mRNA levels for the catalytic subunit <strong>of</strong> telomerase, hTERT,<br />

showed a down-regulation in gene expression consistent with the presence <strong>of</strong> inhibitory<br />

Dioxin Response Elements (iDRE) in the hTERT gene sequence. Overall,<br />

these results demonstrate that AhR agonists cause a receptor-dependent down-regulation<br />

<strong>of</strong> telomerase transcription and subsequent reduction in cell proliferation,<br />

suggesting anticarcinogenic effects in human mammary cancer cells. Inhibition <strong>of</strong><br />

telomerase expression by AhR agonists may represent a therapeutic pathway for<br />

breast cancer treatment<br />

SOT 2010 ANNUAL MEETING 257

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

Saved successfully!

Ooh no, something went wrong!