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.

215 HUMAN INDUCED PLURIPOTENT STEM CELL<br />

DERIVED CARDIOMYOCYTES FOR ASSESSING DRUG<br />

INDUCED CARDIAC ARRHYTHMIAS.<br />

B. Anson 1 , J. Ma 1 , L. Guo 2 , S. Fiene 1 , R. Adams 2 , T. Weiser 3 , T. Singer 3 and<br />

K. Kolaja 2 . 1 Cellular Dynamics International, Madison, WI, 2 Non-Clinical Safety, F.<br />

H<strong>of</strong>fman-La Roche, Nutley, NJ and 3 Pharmacology Research and Early Development,<br />

F. H<strong>of</strong>fman-La Roche AG, Basel, Switzerland.<br />

<strong>The</strong> potential for drug-induced cardiac arrhythmias is a vital component <strong>of</strong> the toxicological<br />

and safety pharmacological pr<strong>of</strong>ile <strong>of</strong> new chemical entities (NCEs).<br />

Cardiomyocytes contain multiple ion channels and ion transporters and cardiac<br />

electrical activity arises from the temporal and spatial activity <strong>of</strong> these proteins.<br />

Commonly employed methods fail to recapitulate the human condition as the predominant<br />

systems typically express a single channel without ancillary proteins and<br />

may therefore miss exacerbating or compensatory protein interactions, while animal<br />

models may ultimately miss species-specific effects. Human induced pluripotent<br />

stem cell (hIPSC) -derived cardiomyocytes provide a relevant human system in<br />

which to assess the arrhythmogenic nature <strong>of</strong> an NCE. We present data characterizing<br />

the electrophysiological pr<strong>of</strong>ile <strong>of</strong> hIPSC-derived cardiomyocytes and demonstrating<br />

their suitability for assessing NCE-induced arrhythmogenicity. Single cell<br />

voltage and current clamp experiments demonstrated the presence <strong>of</strong> multiple endogenous<br />

ionic currents present in human cardiomyocytes including I Na , I Ca , I to ,<br />

I Kr , I f , and I K1 while drug application produced the expected changes in action potential<br />

phenotypes including an approximate 70% increase in duration in response<br />

to hERG channel block (100nM <strong>of</strong> E-4031), 80% decrease in duration in response<br />

to Ca 2+ channel block (100nM nifedipine), and a 60% slowing <strong>of</strong> the action potential<br />

upstroke in response to Na + channel block (10μM TTX). <strong>The</strong>se results were<br />

verified through organotypic extracellular recordings made with microelectrode<br />

array (MEA) techniques that also demonstrated a 35% increase in field potential<br />

duration in response to block <strong>of</strong> IKs by 10μM chromanol 293B. This dataset<br />

demonstrates that hIPSC-derived cardiomyocytes are a suitable and novel humanbased<br />

model for assessing NCE arrhythmogenic potential.<br />

216 ISIS 416858, AN ANTISENSE INHIBITOR OF FACTOR<br />

XI, IS WELL TOLERATED AND PRODUCES NO RISK<br />

OF BLEEDING IN THE CYNOMOLGUS MONKEY FOR<br />

UP TO 13 WEEKS OF TREATMENT.<br />

H. S. Younis 1 , J. Crosby 2 and S. P. Henry 1 . 1 Preclinical Development, ISIS<br />

Pharmaceuticals, Carlsbad, CA and 2 Drug Discovery, ISIS Pharmaceuticals,<br />

Carlsbad, CA.<br />

Complications associated with increased bleeding risk are the main limitations with<br />

current anticoagulant therapy. A strategy to produce sufficient anticoagulant properties<br />

with reduced bleeding risk may be the inhibition <strong>of</strong> Factor XI in the intrinsic<br />

coagulation cascade. <strong>The</strong> objective <strong>of</strong> this work was to determine the safety pr<strong>of</strong>ile<br />

<strong>of</strong> ISIS 416858, a 2’-methoxyethoxy antisense oligonucleotide inhibitor <strong>of</strong> Factor<br />

XI, with special focus on assessing bleeding risk. Cynomolgus monkeys were administered<br />

ISIS 416858 (4, 8, 12 and 40 mg/kg/wk, SC) for up to 13 weeks. ISIS<br />

416858 produced a dose-dependent reduction in systemic Factor XI concentrations<br />

and a concomitant increase in aPTT. Plasma Factor XI was reduced by 80% at 4<br />

weeks <strong>of</strong> treatment (40mg/kg/wk) that resulted in a 33% increase in aPTT by 13<br />

weeks. No effects on PT or platelets were observed during this time period confirming<br />

that the effects <strong>of</strong> ISIS 416858 on Factor XI inhibition were limited to the<br />

intrinsic coagulation cascade. Skin laceration bleeding time, a measure <strong>of</strong> bleeding<br />

risk, was unchanged following 13 weeks <strong>of</strong> treatment. ISIS 416858 produced typical<br />

changes anticipated for an antisense oligonucleotide in the monkey including<br />

acute and transient complement activation, basophilic granules and mononuclear<br />

cell infiltrates in multiple tissues, which increased in incidence and severity in a<br />

dose-dependent manner. Collectively, these results suggest that ISIS 416858 produces<br />

antisense inhibition <strong>of</strong> Factor XI, is well tolerated and produces no risk <strong>of</strong><br />

bleeding at clinically relevant doses.<br />

217 BENZO[A]PYRENE AUGMENTS INFLAMMATORY AND<br />

PROTEOLYTIC CHANGES IN MOUSE MODEL OF<br />

ABDOMINAL AORTIC ANEURYSM.<br />

P. A. Prins 1 , P. R. Perati 1 , A. Ramesh 2 , Z. Guo 2 and U. K. Sampson 1 .<br />

1 Cardiovascular Medicine, Vanderbilt University, Nashville, TN and 2 Meharry<br />

Medical College, Nashville, TN.<br />

Background: Benzo[a]pyrene [B(a)P] is an abundant environmental polycyclic aromatic<br />

hydrocarbon (PAH) known to promote atherosclerosis—a predisposing factor<br />

for the development <strong>of</strong> abdominal aortic aneurysms (AAA). Since experimental<br />

46 SOT 2011 ANNUAL MEETING<br />

evidence suggests that inflammation and enzymatic degradation <strong>of</strong> the vascular wall<br />

are central to the pathobiology <strong>of</strong> AAA, we sought to determine the effects <strong>of</strong> BaP<br />

exposure on these processes. Methods: A total <strong>of</strong> 112 male, 5 weeks <strong>of</strong> age, Apo E-<br />

/- mice were evaluated in 4 experimental groups: B(a)P (n=39), AngII (n= 30),<br />

B(a)P+AngII (n=29) and control (n=24). Exposure to B(a)P (0.71mg/kg/day x<br />

60days) was via oral gavage and AngII exposure (1000ng/kg/min x 2 wks) occurred<br />

via subcutaneous osmotic mini pumps. In the B(a)P+AngII group, AngII exposure<br />

overlapped with the last two weeks <strong>of</strong> B(a)P treatment. Results: <strong>The</strong>re were no incidence<br />

<strong>of</strong> AAA in either control or B(a)P groups. <strong>The</strong>re was no difference in the<br />

number <strong>of</strong> intact (Ni) and ruptured (Nr) AAA between the AngII (Ni = 10, Nr = 7)<br />

and B(a)P + AngII (Ni = 8, Nr = 8) groups. Intact AAA from mice in the<br />

B(a)P+AngII group were significantly larger in cross-sectional diameter (2.30 ±<br />

0.09, N=8) compared to those <strong>of</strong> the AngII group (1.9 ± 0.09, N=10) (p < 0.05).<br />

Immunohistochemical and molecular evaluation <strong>of</strong> aortic tissue from B(a)P+AngII,<br />

and AngII treatment groups demonstrated higher levels <strong>of</strong> tumor necrosis factor<br />

alpha (TNF-α) and cyclophilin A (Cyp A) proteins, and <strong>of</strong> matrix metalloproteinase<br />

(MMP9) in the B(a)P+AngII group. Accordingly, serum inflammatory cytokine<br />

levels (TNF-α) and interleukin (IL-6) were higher in the B(a)P+AngII<br />

group. Conclusions: <strong>The</strong>se findings suggest that B(a)P potentiates the inflammatory<br />

and proteolytic processes that attend AAA formation. Given the abundance <strong>of</strong><br />

B(a)P (e.g. in charcoaled foods and smoke), the possible role <strong>of</strong> B(a)P in AAA evolution<br />

merits further evaluation for mechanistic insights.<br />

218 ARYL HYDROCARBON RECEPTOR AGONIST - PCB 77<br />

- INCREASES THE INCIDENCE AND SEVERITY OF<br />

ABDOMINAL AORTIC ANEURYSM.<br />

V. Arsenescu, R. Arsenescu and L. Cassis. GCNS, University <strong>of</strong> Kentucky,<br />

Lexington, KY. Sponsor: H. Swanson.<br />

Rationale: Male sex and smoking are the main risk factors for abdominal aortic<br />

aneurysm (AAA). PCB 77, a dioxin-like component <strong>of</strong> cigarette smoke, triggers<br />

AhR dependent inflammatory response in the cardiovascular system. We hypothesized<br />

that PCB77 will induce inflammation in the perianeurysmal adipose tissue<br />

and promote the development <strong>of</strong> Angiotensin II (AngII) - induced AAA. Methods:<br />

We injected male, apoE-/- mice with vehicle or PCB 77 (150mmol/Kg, i.p) at the<br />

beginning and at the end <strong>of</strong> 1 month AngII infusion. We monitored body weight,<br />

blood pressure and the aneurysm formation and progression using ultrasonography.<br />

<strong>The</strong> incidence <strong>of</strong> abdominal aortic aneurysm was analyzed in a cohort <strong>of</strong> patients<br />

with/without exposure to cigarette smoke. CYP1A1 induction was determined in<br />

aortic wall specimens from patients that underwent AAA surgical repair. Results:<br />

PCB 77 increased body weight by expanding visceral adipose tissue and liver mass<br />

in AngII + PCB 77 - infused mice. In addition, total cholesterol and free fatty acids<br />

(NEFA) were increased in mice receiving PCB 77. AAA incidence and severity were<br />

increased in mice exposed to PCB 77 (9 out <strong>of</strong> 10) compared to AngII group (5 out<br />

<strong>of</strong> 11). PCB77 was preferentially accumulated in the mesenteric fat. Furthermore,<br />

PCB 77 treatment induced proinflammatory adipokines, Angiotensin, AT1 and<br />

macrophage accumulation in the perianeurysmal adipose tissue while decreasing<br />

the antiinflammatory adiponectin. Patients exposed to cigarette smoke had twice<br />

the incidence <strong>of</strong> AAA and it was correlated with high CYP1A1 expression in the<br />

aneurysmal aortic wall. Conclusions: Persistent exposure to a dioxin like toxicant<br />

was associated with increase AAA severity and mortality. Metabolic syndrome and<br />

inflammatory response in the perivascular fat could explain this association. <strong>The</strong><br />

strong correlation between smoking and AAA in human subjects further supports<br />

our observations. Smoking cessation and avoidance <strong>of</strong> fatty foods containing<br />

dioxin-like toxicants represent viable approaches for prevention and early, non-surgical<br />

management <strong>of</strong> patients with AAA.<br />

219 NNK, A TOBACCO-SPECIFIC CARCINOGEN, INHIBITS<br />

THE EXPRESSION OF LYSYL OXIDASE: A TUMOR<br />

SUPPRESSOR GENE.<br />

Y. Zhao, S. Gao, J. Zhou, P. Toselli and W. Li. Biochemistry, Boston University<br />

School <strong>of</strong> Medicine, Boston, MA.<br />

4-(methyl nitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a tobacco-specific carcinogen,<br />

is believed to contribute to the cancer burden in cigarette smokers. To<br />

evaluate NNK effects on the expression <strong>of</strong> lysyl oxidase (LO), a tumor suppressor<br />

gene, we examined this enzyme at promoter, mRNA, protein and catalytic levels in<br />

NNK-treated rat fetal lung fibroblasts (RFL6). Exposure <strong>of</strong> cells to NNK at 10, 30,<br />

100, and 300 μM for 48 h reduced LO transcript levels to 79.3, 56.4, 8.6 and<br />

1.8% <strong>of</strong> the control, respectively. NNK decreased levels <strong>of</strong> all LO protein species<br />

including the 46 kDa preproenzyme, the 50 kDa proenzyme and the 32 kDa mature<br />

enzyme, in a dose-dependent manner. Notably, the 50 kDa and the 32 kDa<br />

species <strong>of</strong> LO were more sensitive to NNK. Although 300 μM NNK, markedly de-

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

Saved successfully!

Ooh no, something went wrong!