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.

that CCNG2 is positively regulated by TCDD via AHR activation, suggesting that<br />

AHR might utilize this pathway to suppress tumor growth. However, the mechanism<br />

by which AHR regulates CCNG2 is unknown. Using chromatin immunoprecipitation<br />

in T-47D human breast cancer cells we report that the prototypical AHR<br />

ligand TCDD (10nM) is able to recruit AHR and nuclear coactivator 3 (NCoA3)<br />

to the enhancer region <strong>of</strong> CCNG2 (60% and 10% compared to a 5% total) leading<br />

to a 2-fold increase in gene expression after 6h <strong>of</strong> treatment with TCDD(10nM).<br />

Furthermore, our data reveal that the transcription factor Forkhead box A1<br />

(FOXA1) is recruited to CCNG2 in a TCDD-dependent manner. FOXA1 is<br />

known to be an important pioneer factor controlling the binding <strong>of</strong> ERα to its target<br />

genes. RNAi-mediated knockdown <strong>of</strong> FOXA1 abolished TCDD-dependent recruitment<br />

<strong>of</strong> AHR to CCNG2 and reduced CCNG2 expression levels. Interestingly,<br />

knockdown <strong>of</strong> FOXA1 also caused a marked decrease in ERα, but not AHR protein<br />

levels. However, RNAi-mediated knockdown <strong>of</strong> ERα had no effect on TCDDdependent<br />

AHR recruitment to or expression <strong>of</strong> CCNG2. Our data show that<br />

FOXA1, but not ERα is essential for AHR-dependent regulation <strong>of</strong> CCNG2, assigning<br />

a role for FOXA1 in AHR action.<br />

372 DIVERGENT ROLES FOR THE ARYL HYDROCARBON<br />

RECEPTOR AND ARYL HYDROCARBON RECEPTOR<br />

NUCLEAR TRANSLOCATOR IN ESTROGEN<br />

SIGNALING.<br />

M. P. Labrecque 1 , K. J. Tam 1 , B. D. Hollingshead 2 , G. G. Prefontaine 1 , G. H.<br />

Perdew 2 and T. V. Beischlag 1 . 1 Faculty <strong>of</strong> Health Sciences, Simon Fraser University,<br />

Burnaby, BC, Canada and 2 Veterinary and Biomedical Sciences, <strong>The</strong> Pennsylvania<br />

State University, University Park, PA.<br />

<strong>The</strong> aryl hydrocarbon receptor (AHR) and the aryl hydrocarbon receptor nuclear<br />

translocator (ARNT) form a heterodimeric transcription factor upon binding a variety<br />

<strong>of</strong> environmental contaminants, including 2,3,7,8-tetrachlorodibenzo-pdioxin<br />

(TCDD). <strong>The</strong> activated AHR/ARNT complex (AHRC) regulates the expression<br />

<strong>of</strong> xenobiotic target genes to combat the effects <strong>of</strong> environmental<br />

contaminants and evidence has shown that TCDD represses estrogen receptor (ER)<br />

target gene activation through the AHRC. Our data indicates that AHR and<br />

ARNT may act independently <strong>of</strong> each other at <strong>of</strong>f-target sites so we set out to determine<br />

the specific functions <strong>of</strong> AHR and ARNT in estrogen dependent signalling.<br />

Human MCF7 breast cancer cells and human ECC-1 endometrial carcinoma<br />

cells were transfected with short inhibitory RNA’s (siRNA) to AHR or<br />

ARNT. Cells were treated with vehicle, 17β-estradiol (10nM), TCDD (2nM) or a<br />

combination <strong>of</strong> ligands for 16 h and target gene expression and protein levels were<br />

assayed by real-time PCR and Western blotting, respectively. Knockdown <strong>of</strong> AHR<br />

with siRNA abrogates dioxin-inducible repression <strong>of</strong> estrogen-dependent gene<br />

transcription. Intriguingly, knockdown <strong>of</strong> ARNT does not effect TCDD-mediated<br />

repression <strong>of</strong> estrogen-regulated transcription. Furthermore, basal and activated<br />

levels <strong>of</strong> cathepsin-D and progesterone receptor mRNA are downregulated in<br />

MCF7 cells but upregulated in ECC-1 cells in response to loss <strong>of</strong> ARNT. <strong>The</strong>se responses<br />

are mirrored at the protein level with cathepsin-D but progesterone receptor<br />

seems to be refractory. We have obtained experimental evidence demonstrating<br />

a dioxin-dependent repressor function for AHR and a dioxin-independent coactivator/corepressor<br />

function for ARNT in estrogen signalling. <strong>The</strong>se results provide<br />

us with further insight into the mechanisms <strong>of</strong> transcription factor crosstalk and<br />

putative therapeutic targets in estrogen positive cancers.<br />

373 THE ARYL HYDROCARBON RECEPTOR INTERACTS<br />

WITH ATP5α1, A SUBUNIT OF THE ATP SYNTHASE<br />

COMPLEX, AND MODULATES MITOCHONDRIAL<br />

FUNCTION.<br />

D. M. Tappenden 1, 2 , S. Lynn 1, 2 , R.B.Crawford 3 , N. E. Kamniski 2, 3 , R. S.<br />

Thomas 4 and J. J. LaPres 1, 2 . 1 Biochemistry, Michigan State University, East<br />

Lansing, MI, 2 Center for Integrative <strong>Toxicology</strong>, Michigan State University, East<br />

Lansing, MI, 3 Department <strong>of</strong> Pharmacology and <strong>Toxicology</strong>, Michigan State<br />

University, East Lansing, MI and 4 <strong>The</strong> Hamner Institutes for Health Sciences,<br />

Research Triangle Park, NC.<br />

Polyaromatic hydrocarbons (PAHs), such as dioxins are prevalent environmental<br />

contaminates produced by both natural and man-made activities. 2,3,7,8 tetrachloro-dibenzo-p-dioxin<br />

(TCDD) is the canonical molecule in this family <strong>of</strong> compounds.<br />

<strong>The</strong> aryl hydrocarbon receptor (AHR), a ligand activated transcription factor<br />

and a member <strong>of</strong> the PAS super-family <strong>of</strong> environmental sensors, mediates<br />

cellular responses to PAHs. A complete understanding <strong>of</strong> the signaling pathways involved<br />

in the toxicity induced by ligand activated AHR has not been elucidated. To<br />

explore the potential influences <strong>of</strong> protein-protein interactions in AHR-mediated<br />

80 SOT 2011 ANNUAL MEETING<br />

toxicity, a tandem affinity purification (TAP) and mass spectrometry strategy was<br />

utilized to isolate and identify proteins that co-purify with the AHR. <strong>The</strong> known<br />

protein interactions between the AHR, Hsp90, and ARA9 and the ligand-dependent<br />

interaction with ARNT were confirmed, demonstrating the validity <strong>of</strong> the approach.<br />

Using this method a novel interaction between the AHR and ATP5α1, a<br />

subunit <strong>of</strong> the ATP synthase complex, has been established. This interaction was<br />

confirmed by co-immunoprecipitation in wild type cells. <strong>The</strong> ATP synthase complex<br />

is localized to the inner membrane <strong>of</strong> the mitochondria and is essential for cellular<br />

energetics. Investigation <strong>of</strong> the interaction between the AHR and ATP5α1 has<br />

revealed that a fraction <strong>of</strong> the total cellular content <strong>of</strong> the AHR localizes to the mitochondria.<br />

Furthermore, exposure to TCDD leads to hyperpolarization <strong>of</strong> the mitochondrial<br />

inner membrane, in an AHR dependent manner, while cellular ATP<br />

levels are maintained. <strong>The</strong>se findings support a role for the AHR in mitochondrial<br />

function and cellular energy homeostasis that is disrupted in TCDD exposure.<br />

374 REGULATION OF CHOLESTEROL BIOSYNTHETIC<br />

GENE EXPRESSION BY AH RECEPTOR THROUGH A<br />

DNA-INDEPENDENT MECHANISM.<br />

R. Tanos, R.D.PatelandG. H. Perdew. Penn State University, University Park, PA.<br />

<strong>The</strong> aryl hydrocarbon receptor (AHR) is a ligand activated transcription factor.<br />

Activation <strong>of</strong> AHR by xenobiotic agonists such as TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin)<br />

is known to have toxic consequences through driving the<br />

expression <strong>of</strong> target genes, namely CYP1A1, by binding to dioxin response element<br />

sequences in their promoter region. Interestingly, Ahr-null mice exhibit a defined<br />

set <strong>of</strong> physiological phenotypes that indicate an endogenous role for AHR. Recent<br />

microarray studies performed in mice and rats treated with TCDD have shown a<br />

decrease in the expression <strong>of</strong> lipid metabolism genes. In our effort to understand<br />

the mechanisms <strong>of</strong> this regulation, we conducted a microarray analysis on liver<br />

samples from ligand-treated transgenic mice expressing a DNA-binding mutant<br />

AHR (A78D) on an Ahr-null background and showed that AHR DNA-binding is<br />

not essential for suppression <strong>of</strong> genes involved in cholesterol synthesis. To alleviate<br />

the phenotype <strong>of</strong> Ahr-null mice, we established a mouse line expressing a liver specific<br />

DNA-binding mutant AHR on a CreAlb/AhrFx/Fx background. Real-time<br />

qPCR performed on these mice as well as wild-type mice confirmed repression <strong>of</strong><br />

cholesterol biosynthesis genes namely HMGCR, SQLE and LSS following receptor<br />

activation in both mouse lines. Conversely, using real-time qPCR and western blot<br />

analysis, we showed that absence <strong>of</strong> the receptor in liver-specific CreAlb/AhrFx/Fx<br />

mice leads to upregulation <strong>of</strong> those genes in the absence <strong>of</strong> an exogenous ligand.<br />

<strong>The</strong>se data firmly establish the role <strong>of</strong> AHR as regulator <strong>of</strong> cholesterol biosynthesis<br />

independently <strong>of</strong> its DNA-binding ability and suggest that AHR may be a previously<br />

unrecognized therapeutic target.<br />

375 MULTI-SPECIES ANALYSES OF DIRECT ACTIVATORS<br />

OF THE CONSTITUTIVE ANDROSTANE RECEPTOR.<br />

C. Omiecinski 1 , D. M. Coslo 1 , T. Chen 1 and R. C. Peffer 2 . 1 Center for Molecular<br />

<strong>Toxicology</strong>, Penn State University, University Park, PA and 2 Syngenta Crop Protection,<br />

Inc., Greensboro, NC.<br />

<strong>The</strong> constitutive androstane receptor (CAR; NR1I3) is a member <strong>of</strong> the superfamily<br />

<strong>of</strong> nuclear receptors that functions as an important xenochemical sensor in<br />

mammalian cells. Upon chemical activation, CAR undergoes nuclear translocation<br />

and heterodimerization with the retinoid X receptor to subsequently interact with<br />

DNA targets to transcriptionally modulate gene expression. CAR is unusual among<br />

nuclear receptors in that it possesses a high level <strong>of</strong> constitutive activity in cell based<br />

assays, obscuring the detection <strong>of</strong> direct ligand activators. However a human splice<br />

variant <strong>of</strong> CAR, termed hCAR3, exists that exhibits negligible constitutive activity;<br />

rather, CAR3 is activated by ligands with similar specificity as the reference form <strong>of</strong><br />

the receptor, CAR1. In this study, we hypothesized that similar hCAR3 variant receptors<br />

could be constructed across various mammalian species’ forms <strong>of</strong> CAR,<br />

thereby preserving species specific ligand responses yet enabling their detection in a<br />

much more sensitive ligand-dependent mode. <strong>The</strong>refore, in this study, a battery <strong>of</strong><br />

CAR3 variant receptors were produced for the respective native CARs <strong>of</strong> mouse, rat<br />

and dog, and comparatively evaluated with selected ligands in mammalian cell reporter<br />

assays. <strong>The</strong> results demonstrate that the 5 amino acid insertion that typifies<br />

human CAR3 also creates ligand activated receptor function in other species’<br />

CARs, while maintaining signature responses in each species to select CAR ligands.<br />

<strong>The</strong>se variant constructs permit evaluation <strong>of</strong> differential effector chemical responses<br />

across species and facilitate, for example, comparative study <strong>of</strong> tumor promotional<br />

properties <strong>of</strong> suspected hepatocarcinogens on direct CAR activation responses<br />

in different species.

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

Saved successfully!

Ooh no, something went wrong!