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.

2497 TRANSCRIPTIONAL REGULATION OF THE HUMAN<br />

FERRITIN GENE THROUGH AN ANTI-OXIDANT<br />

RESPONSIVE ELEMENT BY CORE HISTONE<br />

MODIFICATIONS.<br />

B. Huang, K. Iwasaki and Y. Tsuji. Environmental and Molecular <strong>Toxicology</strong>, North<br />

Carolina State University, Raleigh, NC. Sponsor: J. Ninomiya-Tsuji.<br />

Oxidative stress is implicated in various diseases such as cancer and neurodegeneration;<br />

therefore, gaining insight into the molecular mechanism through which antioxidant<br />

detoxification genes are regulated is important to understand the pathogenesis<br />

and prevention <strong>of</strong> oxidative stress-related diseases. Ferritin, the major iron<br />

storage protein that sequesters intracellular excess iron to prevent production <strong>of</strong> reactive<br />

oxygen species, is transcriptionally regulated via an ARE (anti-oxidant responsive<br />

element) under oxidative stress. We recently observed that Nrf2 and ATF1<br />

bind to the ferritin ARE. Single knockdown <strong>of</strong> ATF1 or Nrf2 showed only partial<br />

effect, whereas ATF1 and Nrf2 double-knockdown in K562 human erythroleukemic<br />

cells showed a complete inhibition <strong>of</strong> ferritin mRNA induction following<br />

hemin treatment. Hemin-mediated transcriptional activation <strong>of</strong> the ferritin<br />

gene was associated with p38 MAPK phosphorylation along with ATF1 phosphorylation<br />

at Ser-63, in which SB203580, a p38 inhibitor, inhibited all these events.<br />

Concomitantly, both recruitment <strong>of</strong> protein arginine methyltransferase 1<br />

(PRMT1) and its target methylation <strong>of</strong> histone H4 Arg-3 around the ferritin ARE<br />

were increased after hemin treatment. PRMT1 expression activated ARE-dependent<br />

ferritin transcription, and AMI-1, a PRMT inhibitor, inhibited hemin-mediated<br />

ferritin transcription. <strong>The</strong>se results suggest that hemin-mediated ferritin transcription<br />

through the ARE is regulated by p38-mediated ATF1 Ser-63<br />

phosphorylation along with PRMT1-mediated histone H4 Arg-3 methylation.<br />

Furthermore, ChIP assays demonstrated that acetylation <strong>of</strong> histone H3 Lys-9 and -<br />

14, and recruitment <strong>of</strong> histone acetyltransferases p300 and CBP around the ferritin<br />

ARE were induced following hemin treatment. We are currently investigating<br />

whether phosphorylation <strong>of</strong> ATF1 at Ser-63 and recruitment <strong>of</strong> PRMT1 orchestrate<br />

ferritin ARE activation by recruiting chromatin remodeling proteins such as<br />

p300 and CBP and subsequent histone acetylation.<br />

2498 ACTIVATION OF NRF2 BY INTRINSIC AND EXTRINSIC<br />

LIGANDS.<br />

A. Chia, R. Megherbi, I. Copple, N. R. Kitteringham, L. E. Randle, C. E.<br />

Goldring and K. Park. Molecular & Clinical Pharmacology, University <strong>of</strong> Liverpool,<br />

Liverpool, Merseyside, United Kingdom. Sponsor: D. Mendrick.<br />

Nrf2 is a key activator <strong>of</strong> cytoprotective genes in mammalian cells. Cysteine<br />

residues in the inhibitor <strong>of</strong> Nrf2, Keap1, are targets for electrophilic and oxidative<br />

modifications, which may result in uncoupling <strong>of</strong> Keap1 to Nrf2 resulting in Nrf2<br />

stabilization. <strong>The</strong> work <strong>of</strong> a number <strong>of</strong> groups, including our own, has shown that<br />

patterns <strong>of</strong> cysteine adduction in Keap1 in vitro and in living cells are electrophiledependent.<br />

<strong>The</strong> aim <strong>of</strong> the present work was to assess the relevance <strong>of</strong> covalent<br />

modification <strong>of</strong> cysteines in Keap1 in the activation <strong>of</strong> Nrf2-dependent genes<br />

through the use <strong>of</strong> a model compound, DNCB, which targets only one cysteine in<br />

Keap1 expressed in cells, i.e. C257. LC-MS/MS analysis was performed on Keap1<br />

intracellularly modified by DNCB and its analogues (dinitrohalobenzenes), which<br />

retain similar chemistry to DNCB, deplete glutathione with similar potencies, but<br />

differ in their overall reactivity with intracellular protein targets. We detected modification<br />

<strong>of</strong> intracellular Keap1 by all <strong>of</strong> these chemicals only at C257, and found<br />

that this remarkable cysteine residue selectivity occurs despite the fact that all <strong>of</strong> the<br />

cysteines <strong>of</strong> Keap1 are potentially available for modification by other sulfhydryl-reactive<br />

chemical probes. Pre-treatment <strong>of</strong> Keap1-expressing cells with buthionine<br />

sulfoximine (an inhibitor <strong>of</strong> glutathione synthesis), resulted in a reduced ability <strong>of</strong><br />

DNCB to adduct C257. Nevertheless, mutation <strong>of</strong> this cysteine did not affect the<br />

ability <strong>of</strong> DNCB to induce Nrf2 accumulation and Nrf2-dependent gene transcription.<br />

<strong>The</strong> hypothesis that reversible and/or irreversible oxidation <strong>of</strong> other cysteine<br />

residues in Keap1 may provide a signal for activation <strong>of</strong> the Nrf2 pathway is<br />

currently being pursued. In summary, covalent binding to specific reactive cysteine<br />

residues in Keap1 may not necessarily be the sole trigger for activation <strong>of</strong> Nrf2-dependent<br />

cell defence, rather that other forms <strong>of</strong> modification <strong>of</strong> Keap1, and possibly<br />

other proteins in this pathway, may also serve as sensors, to enable the system to<br />

be chemically highly versatile.<br />

2499 KEAP1-KNOCKDOWN AND HEPATOCYTE-SPECIFIC<br />

SIRT1 DELETION CONFER RESISTANCE TO FASTING-<br />

INDUCED DOWN-REGULATION OF NRF2-TARGET<br />

GENE EXPRESSION.<br />

S. Kulkarni1 , J. Xu1 , W. Wei1 , X. Li2 , M. Yamamoto3 and A. Slitt1 . 1Biomedical and Pharmaceutical Sciences, University <strong>of</strong> Rhode Island, Kingston, RI, 2National Institute <strong>of</strong> Environmental Health Sciences, National Institutes <strong>of</strong> Health, Research<br />

Triangle Park, NC and 3Department <strong>of</strong> Medical Biochemistry, Tohoku University<br />

Graduate School <strong>of</strong> Medicine, Sendai, Japan.<br />

Food deprivation (fasting) causes a coordinated whole body response to enhance<br />

gluconeogenesis. Fasting increases serum glucagon levels; this activates the protein<br />

kinase A (PKA)/cAMP signal transduction pathway and enhances liver glucose production.<br />

Fasting and caloric restriction also increase Sirtuin 1 (Sirt1) activity, which<br />

also enhances gluconeogenesis. Both pathways enhance the expression and activity<br />

<strong>of</strong> the transcriptional co-activator Pgc-1α. Nuclear factor-E2 related factor 2 (Nrf2,<br />

Nfe2l2) is a transcription factor, well known to be responsive to cellular oxidative<br />

stress, but may be also sensitive to cellular nutrient status. First, C57Bl/6 (lean) and<br />

ob/ob (obese) mice were food withheld (fasted) for 30 hours and Nrf2 target gene<br />

(e.g. Nqo-1, gclc, gclm, gsta-1, Mrp2) expression was quantified. Fasting for 30<br />

hours decreased Nrf2 target gene expression in liver by 40-75% compared to fed<br />

C57BL/6 and ob/ob mice. Second, increased constitutive expression <strong>of</strong> Nrf2 diminished<br />

the fasting effects on Nrf2 target genes (lower fold decrease), as observed<br />

in Keap1-KD fasted mice compared to C57Bl/6 fasted controls. Third, Liver-specific<br />

deletion <strong>of</strong> Sirt1 (Sirt1LKO), a deacetylase known to be activated upon nutrient<br />

deprivation, also diminished the fold decrease in expression <strong>of</strong> Nrf2 target genes<br />

upon fasting. In general, the obese, Keap1-KD, and Sirt1LKO mice were resistant<br />

to fasting-induced changes in Nrf2 target gene expression, as observed by the attenuated<br />

expression <strong>of</strong> Pgc-1α in these livers. <strong>The</strong> above data directs towards a novel<br />

role for nutrient status affecting Nrf2 activity, the antioxidant response, and potential<br />

novel interplay between Sirt1 and the antioxidant response (NIH<br />

3R01ES016042).<br />

2500 A BIOCHEMICAL AND PROTEOMIC ANALYSIS OF<br />

THE EFFECT OF NRF2 GENE DELETION ON ACUTE<br />

ACETAMINOPHEN-INDUCED HEPATOTOXICITY.<br />

C. E. Goldring, L. E. Randle, R. L. Sison, N. R. Kitteringham, D. Denk, R. E.<br />

Jenkins, J. Walsh, B. Lane, A. Kipar and K. Park. Molecular & Clinical<br />

Pharmacology, University <strong>of</strong> Liverpool, Liverpool, Merseyside, United Kingdom.<br />

Sponsor: D. Mendrick.<br />

<strong>The</strong> Keap1-Nrf2-ARE signalling pathway has emerged as a key protective mechanism<br />

in the response to chemical stress. To date, a number <strong>of</strong> studies have investigated<br />

Nrf2-dependent changes at the RNA level, however these may not necessarily<br />

be translated into changes at the protein level and function. We have now sought to<br />

determine the role <strong>of</strong> Nrf2 in the early mechanisms <strong>of</strong> adaptation to acute DILI<br />

through a shotgun proteomics approach and targeted metabolite analysis, in a<br />

murine model <strong>of</strong> acetaminophen (APAP)-induced hepatotoxicity. Clinical chemistry<br />

and histopathology confirmed the increased sensitivity <strong>of</strong> Nrf2-null mice towards<br />

APAP-induced liver damage. Metabolomic analysis <strong>of</strong> the glutathione system<br />

indicated adaptation to Nrf2 deletion through an enhancement <strong>of</strong> cellular defence<br />

through conservation and/or increase <strong>of</strong> intermediates in this pathway, in an apparent<br />

attempt to maintain redox homeostasis. Proteomic analysis revealed Nrf2-mediated<br />

efforts by the liver to maintain energy production. Following acute APAPmediated<br />

injury, it appears that an early Nrf2-mediated attempt is made to source<br />

alternative substrates for the citric acid cycle by upregulating enzymes within the<br />

hepatoproteome. Liver carboxylesterase 31 and propionyl-coA carboxylase were<br />

identified as novel Nrf2-dependent, APAP-responsive proteins. Several other<br />

APAP-sensitive proteins were also identified, and these may warrant further investigation<br />

into their suitability as biomarkers <strong>of</strong> APAP intoxication. In summary, we<br />

have demonstrated novel APAP-mediated, Nrf2-dependent changes in the liver<br />

proteome and metabolome that provides new areas <strong>of</strong> study for delineating the molecular<br />

events that are implicated in this important form <strong>of</strong> liver toxicity.<br />

2501 PROFILING ENVIRONMENTAL CHEMICALS IN THE<br />

ANTIOXIDANT RESPONSE ELEMENT PATHWAY<br />

USING QUANTITATIVE HIGH-THROUGHPUT<br />

SCREENING (QHTS).<br />

S. J. Shukla 1 , R. Huang 1 , S. O. Simmons 2 , R. R. Tice 3 , K. L. Witt 3 and M.<br />

Xia 1 . 1 NIH Chemical Genomics Center, Rockville, MD, 2 U.S. EPA, Research<br />

Triangle Park, NC and 3 National <strong>Toxicology</strong> Program, Research Triangle Park, NC.<br />

<strong>The</strong> antioxidant response element (ARE) signaling pathway plays an important role<br />

in the amelioration <strong>of</strong> oxidative stress, which can contribute to a number <strong>of</strong> diseases,<br />

including cancer. We screened 1408 NTP-provided substances in 1536-well<br />

SOT 2011 ANNUAL MEETING 535

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

Saved successfully!

Ooh no, something went wrong!