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.

1168 DIFFERENTIAL PAH EFFECTS MEDIATED BY CYP1A1<br />

AND CYP1B1 IN THE BONE MARROW SPECIFICALLY<br />

TARGET HEMATOPOIETIC PROGENITORS.<br />

M. L. Larsen 1 , A. U. N’Jai 3 , C. J. Czuprynski 3, 2 and C. R. Jefcoate 1, 2 .<br />

1 Pharmacology, University <strong>of</strong> Wisconsin, Madison, WI, 2 Molecular and<br />

Environmental <strong>Toxicology</strong> Center, University <strong>of</strong> Wisconsin, Madison, WI and 3 School<br />

<strong>of</strong> Veterinary Medicine, University <strong>of</strong> Wisconsin, Madison, WI.<br />

<strong>The</strong> polycyclic aromatic hydrocarbons (PAHs), benzo(a)pyrene (BP) and 7,12-dimethylbenzanthracene<br />

(DMBA), produce very different effects in a Min-mouse<br />

cancer model and differential suppression <strong>of</strong> hematopoietic progenitors in mouse<br />

bone marrow (BM). In each model, DMBA is much more active than BP and the<br />

effects are dependent on Cyp1b1. Colony forming unit assays show that DMBA<br />

suppresses lymphoid progenitor activity within 6 h. BP is much less effective, due<br />

to an Ah receptor (AhR)-mediated recovery process. Microarray analyses show few<br />

DMBA responses, but numerous BP responses (25 versus 600). Nine gene responses,<br />

which were similar for each PAH and for TCDD, are attributed to direct<br />

AhR stimulation. <strong>The</strong>se include Cyp1a1, Cyp1b1, and AhRR. Many BP responses,<br />

like the recovery process, were AhR dependent, but resistant to DMBA. <strong>The</strong> time<br />

dependence <strong>of</strong> BP-responses demonstrated an inflammatory group that responded<br />

within 6 h (chemokines, cytokines, COX-2) and decayed within 24 h, and a second<br />

group (Nqo1 muGSTs), which showed the inverse response. Gene expression<br />

changes in BM <strong>of</strong> Cyp1b1-/- mice demonstrated the substantial altered expression<br />

<strong>of</strong> many developmental regulators. Remarkably, inflammatory gene responses to<br />

DMBA, which were absent in WT mice, were increased to BP levels. Thus, removal<br />

<strong>of</strong> BM Cyp1b1 metabolism appears to enhance local DMBA responses. In Cyp1a1-<br />

/- mice, many BP-inflammatory responses declined and a cluster <strong>of</strong> other gene responses<br />

increased. We hypothesize that Cyp1a1 generates BP quinones in BM that<br />

initiate selective oxidative stress responses that contribute to protection, while endogenous<br />

Cyp1b1 metabolites control developmental processes, consistent with a<br />

role in embryogenesis.<br />

1169 IMMUNOMODULATORY RESPONSE OF MOUSE BONE<br />

MARROW-DERIVED MESENCHYMAL STROMAL<br />

CELLS (MBMSCS) TO STIMULATION WITH<br />

LIPOPOLYSACCHARIDES (LPS).<br />

N. V. Gorbunov, B. R. Garrison, G. Ledney and J. G. Kiang. Radiation Combined<br />

Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD.<br />

Mortality induced by gram-negative bacterial sepsis remains a serious problem <strong>of</strong><br />

complicated injury due to penetrating wounds, burns, blunt trauma, radiation,<br />

chemical exposures, or combinations there<strong>of</strong>. A promising therapeutic tool for<br />

management <strong>of</strong> trauma and sepsis is transfusion with bone marrow-derived mesenchymal<br />

stem cells (BMSCs) that exhibit immunomodulatory capacity. However,<br />

the molecular mechanisms underlying BMSC action in septic conditions are poorly<br />

understood. Thus, this report focuses on in vitro investigations <strong>of</strong> the response <strong>of</strong><br />

mouse BMSCs (mBMSCs) to stimulation with products <strong>of</strong> gram-negative bacteria,<br />

LPS. mBMSCs were obtained from the femur bone marrow <strong>of</strong> B6D2F1/J female<br />

mice. <strong>The</strong> cells were expanded and cultivated in hypoxic conditions for 28 days in<br />

MESENCULT medium. <strong>The</strong> cell phenotype, proliferative and colony-forming activity<br />

were analyzed with flow cytometry and immun<strong>of</strong>luorescence imaging; the<br />

cells were identified by BMSC-positive markers: CD44, STRO1, SCA1. mBMSCs<br />

were exposed to 0.05 - 2.5 μg/ml LPS for 1-3 h and analyzed at different timepoints.<br />

LPS-induced gene and protein expression <strong>of</strong> immunomodulatory effectors<br />

(IL-1, IL-6, IL-8, and iNOS) were determined by qRT-PCR and immunoblotting<br />

techniques. Nuclear translocation <strong>of</strong> p65-NFkB and nitric oxide (NO) production<br />

in the cells was assessed with fluorescent imaging. Treatment <strong>of</strong> mBMSCs with LPS<br />

resulted in a substantial increase in gene expression <strong>of</strong> IL-1α, IL-1β, and iNOS that<br />

occurred in a dose- and time-dependent manner. At 24 h following exposure to 0.5<br />

μg/ml LPS, expression <strong>of</strong> NFkB, IL-1β, and iNOS proteins, and NO production<br />

in the cells was increased 2-, 12-, 10-, and 14-fold, respectively. <strong>The</strong>se data are consistent<br />

with the idea that LPS-induced alterations in mBMSCs are mediated by the<br />

Toll-like receptor 4/NFkB axes that, in turn, modultate the host adaptive responses<br />

during septic conditions. (Supported by NIH/NIAID YI-AI-5045-04).<br />

1170 FINE PARTICULATE MATTER (PM 2.5 ) EXPOSURE<br />

AFFECTS CIRCULATING AND BONE MARROW<br />

ENDOTHELIAL PROGENITOR CELLS.<br />

P. Haberzettl, J. Lee, D. Duggineni, J. McCracken, A. Bhatnagar and D. J.<br />

Conklin. Diabetes and Obesity Center, University <strong>of</strong> Louisville, Louisville, KY.<br />

Inhalation <strong>of</strong> fine particulate matter (PM 2.5 ) induces endothelial dysfunction and<br />

increases the risk for cardiovascular disease and mortality. Endothelial progenitor<br />

cells (EPC) contribute to endothelium health and we reported that exposure to am-<br />

250 SOT 2011 ANNUAL MEETING<br />

bient or concentrated ambient PM 2.5 (CAPs) decreases the number <strong>of</strong> circulating<br />

EPC in humans and mice. Thus, we performed experiments to assess effects <strong>of</strong><br />

PM 2.5 exposure on the recruitment/mobilization <strong>of</strong> EPC. Mice (male; C57BL/6;<br />

12-wks) were exposed up to 30 consecutive days (6h/day) to HEPA-filtered air or<br />

CAPs (≈8-fold conc.; 80-100 μg/m 3 ) <strong>of</strong> urban Louisville air using a versatile aerosol<br />

concentration enrichment system (VACES). CAPs exposure for 4, 9 or 30 days significantly<br />

decreased the number <strong>of</strong> circulating EPC measured as Flk-1 + /Sca-1 + -cells<br />

by FACS by 32.6±13.8, 35.8±10.1 or 46±16.9 (% control), respectively. EPC levels<br />

returned to baseline within 1 week following a 9day CAPs exposure, indicating<br />

a reversible PM effect. CAPs exposure (9-day) significantly increased bone marrowderived<br />

Flk-1 + /Sca-1 + -cells by 1.75±0.1-fold when compared with controls.<br />

Similarly, CAPs exposure (9-day) significantly increased colony forming units<br />

(1.6±0.1-fold) and the number <strong>of</strong> Flk-1 + /Sca-1 + or acLDL + /UE-lectin + cells (1.8 ±<br />

0.1 or 1.6 ± 0.1 fold) <strong>of</strong> bone marrow-derived cells in culture compared with controls.<br />

<strong>The</strong>se results indicated that PM blocked EPC mobilization. To test this, mice<br />

exposed for 9 days to air or CAPs were subjected to an EPC mobilization protocol<br />

(VEGF, 100μg/kg/d * 4d and AMD3100, 5mg/kg) and blood levels <strong>of</strong> EPC were<br />

analyzed. VEGF/AMD3100 treatment doubled the number <strong>of</strong> circulating Flk-<br />

1 + /Sca-1 + -cells in air controls but not in CAPs-exposed mice (-48±14 % EPC).<br />

<strong>The</strong>se results indicate that inhalation <strong>of</strong> PM 2.5 affects EPC mobilization leading to<br />

suppressed blood EPC level, which implicates EPC as a potential mechanistic link<br />

between PM 2.5 exposure and CVD.<br />

1171 ALDH1B1 IS A MARKER FOR HUMAN COLON<br />

CANCER.<br />

Y. Chen 1 , A. Matsumoto 1 , D. Orlicky 2 , S. Singh 1 and V. Vasiliou 1 .<br />

1 Pharmaceutical Sciences, University <strong>of</strong> Colorado Denver, Aurora, CO and 2 Pathology,<br />

University <strong>of</strong> Colorado Denver, Aurora, CO.<br />

Aldehyde dehydrogenases (ALDHs) belong to a superfamily <strong>of</strong> NAD(P) + -dependent<br />

enzymes, which catalyze the oxidation <strong>of</strong> various endogenous and exogenous<br />

aldehydes to their corresponding acids. Increased expression and/or activity <strong>of</strong><br />

ALDHs, particularly ALDH1A1, have been reported to occur in human cancers <strong>of</strong><br />

various origins. It is proposed that ALDH1A1 may play a critical role in cancer<br />

stem cells due to its high affinity for the oxidation <strong>of</strong> retinal. Nevertheless, the identity<br />

<strong>of</strong> ALDH isozymes in contributing to the enhanced ALDH activity in a specific<br />

type <strong>of</strong> human cancers remains to be elucidated. ALDH1B1 has recently been<br />

characterized as a mitochondrial ALDH that actively metabolizes acetaldehyde. In<br />

this study, we examined the expression <strong>of</strong> ALDH1A1 and ALDH1B1 in human tumors<br />

<strong>of</strong> colon, lung, breast and ovary using tissue array (NIH) by immunohistochemistry.<br />

We observed a much higher percentage <strong>of</strong> immunopositivity for<br />

ALDH1B1 (>60%) in these cancerous tissues than that for ALDH1A1 ( ALDH1B1 in ovary<br />

> ALDH1B1 in lung or breast > ALDH1A1 in lung > ALDH1A1 in colon or ovary<br />

> ALDH1A1 in breast. This result demonstrates that ALDH1B1 isozyme is more<br />

pr<strong>of</strong>oundly expressed in studied human cancer tissues than ALDH1A1, particularly<br />

in colon cancer. We further examined the mRNA and protein levels <strong>of</strong> the two<br />

ALDHs in six human colon cancer cell lines. ALDH1A1 was found to be abundant<br />

in three cell lines, whereas ALDH1B1 was highly expressed in all six cell lines. In<br />

conclusion, our study suggests that ALDH1B1 is preferably upregulated in human<br />

colon cancer tissues and cell lines, making it a potential marker for human colon<br />

cancer. Supported in part by NIH grant R21AA017754-01.<br />

1172 DIFFERENTIAL TOXICITIES OF KINASE INHIBITORS<br />

(KI) ON BONE MARROW PROGENITORS FROM<br />

DIFFERENT SPECIES.<br />

E. Clarke and G. dos Santos. ReachBio, Seattle, WA. Sponsor: R. Steigerwalt.<br />

KIs represent a new class <strong>of</strong> rationally designed drugs. <strong>The</strong> success <strong>of</strong> the initial<br />

therapeutic KIs in treating leukemia has spurred the development <strong>of</strong> new KIs for<br />

treating various other cancers and inflammation. However, since myelotoxicity is<br />

<strong>of</strong>ten associated with administering these agents, and as the use <strong>of</strong> KIs extends to<br />

non-oncology applications, there is a need to develop compounds with lower levels<br />

<strong>of</strong> toxicity than previously associated with this drug class. We previously showed<br />

that the IC50 value for 6 KIs using the human colony-forming unit-granulocyte<br />

macrophage (CFU-GM) assay correlated with clinical neutropenia and could be<br />

used to accurately rank the myelotoxic potential <strong>of</strong> compounds to other members<br />

<strong>of</strong> the same chemical class. Interestingly, studies have revealed significant differences<br />

between human, dog, rat and mouse CFU-GM sensitivities to certain pharmaceuticals.<br />

Since multiple animal models are <strong>of</strong>ten used in toxicity testing, we

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

Saved successfully!

Ooh no, something went wrong!