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.

CYP1A transcription mediated by AHR-estrogen receptor α (ERα) crosstalk.<br />

Taken together, the function <strong>of</strong> AHRR is conserved in chicken in terms <strong>of</strong> the negative<br />

regulation <strong>of</strong> AHR and ERα activities, but its functional mechanism is likely<br />

distinct from those <strong>of</strong> the mammalian and fish homologues.<br />

354 STABLE OVER-EXPRESSION OF PPARβ/δ AND PPARγ<br />

TO EXAMINE RECEPTOR SIGNALING IN HUMAN<br />

HaCaT KERATINOCYTES.<br />

P. S. Palkar 1 , M. G. Borland 1 , C. Khozoie 1 , B. Zhu 1 , C. Lee 1 , F. J. Gonzalez 2<br />

and J. M. Peters 1 . 1 Veterinary Sciences, Pennsylvania State University, State College,<br />

PA and 2 Laboratory <strong>of</strong> Metabolism, National Cancer Institute, Bethesda, MD.<br />

Recent evidence suggests that peroxisome proliferator-activated receptor-β/δ<br />

(PPARβ/δ) can cross-talk with other nuclear receptor signaling, including PPARγ<br />

and the retinoic acid receptors (RARs). For example, it was suggested that retinoic<br />

acid may promote tumorigenesis by differentially targeting PPARβ/δ rather than<br />

RAR, or that PPARβ/δ can repress PPARγ activity and inhibit the chemopreventive<br />

properties <strong>of</strong> PPARγ agonists. To examine PPARβ/δ function and receptor crosstalk<br />

in greater detail, the Migr1 retroviral system was utilized to create stable human<br />

HaCaT keratinocyte cell lines over-expressing PPARβ/δ and PPARγ. Ligand activation<br />

<strong>of</strong> these receptors in HaCaT keratinocytes over-expressing PPARβ/δ or PPARγ<br />

led to enhanced dose-dependent transcriptional upregulation <strong>of</strong> a known PPAR<br />

target gene, angiopoietin-like protein 4 (ANGPTL4) as compared to controls. Alltrans<br />

retinoic acid (atRA) increased expression <strong>of</strong> a known RAR target gene in control<br />

and HaCaT keratinocytes over-expressing PPARβ/δ. However, despite a high<br />

ratio <strong>of</strong> fatty acid binding protein 5 (FABP5) to cellular retinoic acid binding protein<br />

II (CRABP-II), atRA did not increase expression <strong>of</strong> a PPARβ/δ target gene,<br />

even in HaCaT keratinocytes expressing markedly higher levels <strong>of</strong> PPARβ/δ.<br />

Further, while increased efficacy <strong>of</strong> rosiglitazone to increase ANGPTL4 expression<br />

in HaCaT keratinocytes over-expressing PPARγ was observed, over-expression <strong>of</strong><br />

PPARβ/δ did not modulate the efficacy <strong>of</strong> rosiglitazone activity in HaCaT keratinocytes<br />

as compared to controls. <strong>The</strong>se studies demonstrate that stable over-expression<br />

<strong>of</strong> PPARβ/δ or PPARγ in HaCaT keratinocytes significantly increases the<br />

efficacy <strong>of</strong> ligand activation. Despite overexpression <strong>of</strong> PPARβ/δ, no changes in<br />

retinoic acid-induced, or PPARγ-induced, modulation <strong>of</strong> target gene expression<br />

were observed in HaCaT cells. (Supported by CA124533, CA126826, CA141029,<br />

CA140369)<br />

355 PPARβ/δ MODULATES AHR SIGNALING IN MOUSE<br />

AND HUMAN KERATINOCYTES.<br />

J. Morales 1 , M. G. Borland 1 , C. Lee 1 , F. J. Gonzalez 2 and J. M. Peters 1 .<br />

1 Department <strong>of</strong> Veterinary and Biomedical Sciences, <strong>The</strong> Pennsylvania State<br />

University, University Park, PA and 2 Laboratory <strong>of</strong> Metabolism, National Cancer<br />

Institute, Bethesda, MD.<br />

Peroxisome proliferator-activated receptors (PPARs) are ligand activated transcription<br />

factors that modulate cell-specific biological functions. Previous evidence has<br />

shown that PPARβ/δ attenuates tumor promotion and skin carcinogenesis, but<br />

PPARβ/δ-dependent modulation <strong>of</strong> polycyclic aromatic hydrocarbon (PAH) metabolism<br />

or tumor initiation has not been specifically examined. <strong>The</strong> hypothesis<br />

that PPARβ/δ modulates aryl hydrocarbon receptor (AHR)-dependent signaling<br />

was examined. Surprisingly, Pparβ/δ-null primary keratinocytes exhibited reduced<br />

PAH-mediated induction <strong>of</strong> cytochrome P450 (Cyp) mRNAs, including Cyp1a1<br />

and Cyp1b1, as compared to wild-type cells. Further functional analyses revealed<br />

that Pparβ/δ-null keratinocytes exhibit decreased AHR occupancy and histone<br />

acetylation at the Cyp1a1 promoter in response to a PAH. To determine whether<br />

this change in occupancy <strong>of</strong> the Cyp1a1 promoter was due to alterations in promoter<br />

structure, differential DNA methylation was examined in mouse keratinocytes.<br />

Bisulfite sequencing <strong>of</strong> the Cyp1a1 promoter revealed that Pparβ/δ-null<br />

keratinocytes had increased DNA methylation, which may explain the lower basal<br />

Cyp mRNA expression observed in Pparβ/δ-null keratinocytes. Moreover, studies<br />

using a DNA methylation inhibitor demonstrated that PPARβ/δ modulates AHR<br />

signaling, in part, through DNA methylation. Similar modulation <strong>of</strong> PAH-mediated<br />

induction <strong>of</strong> Cyp mRNAs was observed in a human HaCaT keratinocyte cell<br />

line stably expressing shRNA against PPARβ/δ. However, inhibition <strong>of</strong> DNA<br />

methylation did not alter PAH-induced expression <strong>of</strong> Cyp mRNAs in this model.<br />

Collectively, these findings demonstrate a novel mechanism in mouse and human<br />

keratinocytes where PPARβ/δ modulates AHR-dependent signaling. Furthermore,<br />

these data suggest that PPARβ/δ may alter the carcinogenic effect <strong>of</strong> PAHs by modifying<br />

the balance between carcinogen bioactivation and clearance. (Supported by<br />

CA124533, CA126826, CA141029, CA149369, and the NSF GRFP)<br />

76 SOT 2011 ANNUAL MEETING<br />

356 ROLE OF 2, 3, 7, 8-TETRACHLORODIBENZO-P-<br />

DIOXIN-INDUCIBLE POLY(ADP-RIBOSE)<br />

POLYMERASE IN DIOXIN-INDUCED ARYL<br />

HYDROCARBON RECEPTOR SIGNALING.<br />

L. M. MacPherson and J. Matthews. Pharmacology and <strong>Toxicology</strong>, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada.<br />

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

mediates the toxic effects <strong>of</strong> 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). AHR<br />

regulates the expression a hundreds <strong>of</strong> genes including TCDD-inducible<br />

poly(ADP-ribose) polymerase (TiPARP, PARP-7). TiPARP is a member <strong>of</strong> the<br />

PARP superfamily, which is an enzyme family that mediates poly(ADPribosyl)ation<br />

<strong>of</strong> protein targets. Poly(ADP-ribosyl)ation is post-translational modification<br />

associated with a number <strong>of</strong> biological functions including DNA repair,<br />

transcription, apoptosis and cell division. TiPARP contains a C-terminal PARP catalytic<br />

domain, a conserved WWE (tryptophan-tryptophan-glutamic acid) domain<br />

and a zinc-finger domain. However the biological role <strong>of</strong> TiPARP and whether<br />

TiPARP plays a functional role in AHR signalling is unknown. <strong>The</strong> aim <strong>of</strong> the present<br />

study was to investigate modulation <strong>of</strong> TCDD-induced AHR activity by<br />

TiPARP. We investigated the effects <strong>of</strong> loss and overexpression <strong>of</strong> TiPARP on<br />

TCDD-induced cytochrome P450 1A1 (CYP1A1) and CYP1B1 gene expression<br />

in T-47D human breast carcinoma and HuH-7 hepatoma cell lines. RNAi-mediated<br />

knockdown <strong>of</strong> TiPARP significantly increased TCDD-induced CYP1A1 and<br />

CYP1B1 expression. TiPARP overexpression decreased TCDD-induced CYP1A1and<br />

CYP1B1-regulated reporter activity in a dose-dependent manner. Chromatin<br />

immunoprecipitation (ChIP) assays showed increased TCDD-dependent recruitment<br />

<strong>of</strong> AHR to CYP1A1 and CYP1B1 enhancer regions following TiPARP<br />

knockdown. TiPARP knockdown also reduced TCDD-induced AHR protein<br />

degradation following 24 h treatment. TiPARP truncation mutants revealed<br />

TCDD-induced inhibition required the N-terminal (1-234 amino acid residues) as<br />

well as the PARP catalytic domain. Single point mutations <strong>of</strong> two highly conserved<br />

residues <strong>of</strong> the PARP catalytic core motif abolished inhibition <strong>of</strong> TCDD-induced<br />

reporter gene activity. Collectively, these results implicate TiPARP as a negative regulator<br />

<strong>of</strong> AHR signalling.<br />

357 PPARβ/δ INHIBITS VIRAL HRAS1 (V-HRAS1)-INDUCED<br />

NEOPLASTIC/MALIGNANT TRANSFORMATION OF<br />

MOUSE PRIMARY KERATINOCYTES.<br />

B. Zhu 1 , C. Khozoie 1 , M. T. Bility 1 , N. Blazanin 1 , A. B. Glick 1 , F. J. Gonzalez 2<br />

and J. M. Peters 1 . 1 Molecular <strong>Toxicology</strong>, University Park, PA and 2 Laboratory <strong>of</strong><br />

Metabolism, National Cancer Institute, Bethesda, MD.<br />

Ligand activation <strong>of</strong> PPARβ/δ inhibits chemically-induced skin tumorigenesis.<br />

Since Harvey sarcoma ras (Hras1) mutation is a critical event in chemical carcinogenesis,<br />

the role <strong>of</strong> PPARβ/δ in viral Hras1 (v-Hras1)-induced neoplastic transformation<br />

was examined in mouse primary keratinocytes. v-Hras1 infected wild-type<br />

keratinocytes were protected from malignant transformation, while v-Hras1 infected<br />

Pparβ/δ-null keratinocytes underwent malignant transformation. This protection<br />

was due in part to the induction <strong>of</strong> G2/M arrest by ligand activation <strong>of</strong><br />

PPARβ/δ in wild-type keratinocytes. Flow cytometric analysis revealed that<br />

Pparβ/δ-null keratinocytes underwent more active mitosis and adapted to mitotic<br />

stress better than wild-type keratinocytes. Further, while ligand activation <strong>of</strong><br />

PPARβ/δ exerts a negative selection against cells expressing higher levels <strong>of</strong> the v-<br />

Hras1 oncogene by inducing mitosis block in wild-type keratinocytes, Pparβ/δnull<br />

keratinocytes showed a growth advantage in the presence <strong>of</strong> higher level <strong>of</strong> v-<br />

Hras1 oncogene. To further investigate the mechanism <strong>of</strong> G2/M induction by<br />

ligand activation <strong>of</strong> PPARβ/δ, microarray analysis was performed. Microarray<br />

analysis revealed 62 genes involved in mitosis were induced to a higher level by v-<br />

HRAS1 in Pparβ/δ-null keratinocytes as compared to v-Hras1 infected wild-type<br />

keratinocytes and ligand activation <strong>of</strong> PPARβ/δ repressed these changes in wildtype<br />

but not Pparβ/δ-null keratinocytes. 33 genes out <strong>of</strong> these 62 are known to be<br />

regulated by the E2F transcription factors. Interestingly, other known E2F regulated<br />

genes, notably those involved in G1/S phase <strong>of</strong> the cell cycle, exhibited a similar<br />

pattern <strong>of</strong> expression as these 62 mitosis-related genes. <strong>The</strong>se results suggest<br />

that PPARβ/δ attenuates Hras1-induced neoplastic/malignant transformation by<br />

inducing G2/M arrest by interfering with E2F transcription factors in primary keratinocytes.<br />

(Supported by CA124533, CA126826, CA141029, CA140369)

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

Saved successfully!

Ooh no, something went wrong!