.4 Urdu. ..( ..V . .J .t.) xv fidnuwflram{m=1mnwvmhu . u. 1.“. L?!) 7 nun . 59...? . , ,umw . I . \‘v. n... ma. 4», , VHF-I . . "Emfiuéé vflufiflfir. . 2.. 3 w... ‘ spa l‘ «:11 Jflmmmfin . “w”: flat :3 3%: fig? .2. .1152594 2.5;. Ifar. ‘5be . Y Wu! +5 a s! u “If... Mm” .1 I. fix fimw f ‘ ,1. 3. . n. an; 5: | J r ‘3; $3 , '3 tr. LIBRARY Michigan State UDIVG rsuty This is to certify that the dissertation entitled MEMBRANE ASSOCIATED EFFECTS AND ALTERATIONS IN GENE EXPRESSION RESULTING FROM PERFLUOROOCTANE SULFONIC ACID EXPOSURE presented by WENYUE HU has been accepted towards fulfillment of the requirements for the Ph.D. degree in ZOOLOGY / IET Major Professor nature /7/1§103C7 Date MSU is an Affirmative Action/Equal Opportunity Institution PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 6/01 c:/CIRC/DateDue.p65-p. 15 MEMBRANE ASSOCIATED EFFECTS AND ALTERATIONS IN GENE EXPRESSION RESULTING FROM PERFLUOROOCTAN E SULFONIC ACID EXPOSURE By Wenyue Hu A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Zoology Institute of Environmental Toxicology 2003 ABSTRACT MEMBRANE ASSOCIATED EFFECTS AND ALTERATIONS IN GENE EXPRESSION RESULTING FROM PERFLUOROOCTANE SULFONIC ACID EXPOSURE By Wenyue Hu The recent detection of perfluorinated fatty acids (PFFAS) in wildlife from even remote locations has spurred interest in the environmental occurrence and effects of these chemicals. Among them, perfluorooctane sulfonic acid (PFOS) is the end metabolite of a number of perfluorinated fatty acid analogues extensively used in industrial materials and commercial applications. Few studies have been conducted on this novel compound, and its mechanism of action still remains unclear. The amphiphillic nature of PFOS suggests its cell membrane related effects. In the current study, effects of PFOS on membrane fluidity and mitochondrial membrane potential were examined using flow cytometry and effects on membrane permeability were tested using cell bioassay procedures (H4IIE, MCF-7, PLHC-l). PFOS increased plasma membrane fluidity and decreased mitochondrial membrane potential in fish leukocytes in a dose-dependent fashion. The lowest effective concentrations for both membrane fluidity and mitochondrial membrane potential effects of PFOS were 5 to 15 mg/L. This suggests that membrane properties could be used as sensitive biomarkers for PFOS related adverse effects. Besides membrane related effects, studies were also designed to examine modulations of gene expression by PFOS exposure using states of art molecular toxicology techniques. In the current study, the restriction fragment differential display (RFDD-PCR), a mRNA fingerprinting technique, and Affymetrix rat genome U34A chips, the high throughput genomic technique with 8790 genes, were used to identify alterations in gene expression level due to PFOS exposure in vitro and in viva. RNA samples were extracted from H4IIE rat hepatoma cells and rat livers exposed to PFOS, and prepared for subsequent analysis. Following the RFDD-PCR procedure, 55 bands on sequencing gel were identified as different across treatment groups. All these candidate genes were sub-cloned and sequenced. Gene chip analysis was conducted by hybridizing U34A chips with experimental samples. Approximately 5% of the genes on the chip were identified as differentially expressed in response to PFOS exposure, and clustered as genes coding for fatty acid metabolizing enzymes, drug and xenobiotic metabolizing enzymes, and proteins involved in signal transduction pathways and hormone regulation. Consistent results were obtained from replicate exposures, however, expression profiles of samples prepared in vitro and in vivo showed only limited similarity. The major pathway affected by PFOS is postulated to be peroxisomal fatty acid beta-oxidation, which could be explained by the structural similarity between PFOS and endogenous fatty acids. Comparisons were made between differential display and gene chip techniques based on their specificity, sensitivity, and scope of applications. The mechanistic interpretation derived from these two methods was in agreement, although the results were not directly comparative. To my dear husband Michael Gu and my parents, Hua Hu and XiaoRu Liu iv ACKNOWLEDGEMENT Funding for this project was provided by the 3M Company at St. Paul, MN. I sincerely thank my academic supervisors Dr. John Giesy and Dr. Paul Jones for their continuous support and tremendous help. Only with their never ceased encouragement could I make it through all the difficulties during the process of doing this project and writing the dissertation. I am honored to have Dr. Don Hall, Dr. Will Kopachik, and Dr. Robert Roth on my graduate committee, and to receive invaluable advices from them. Special thanks are owed to Dr. Wim DeCoen and Dr. Trina Celius for their help on the differential display and gene chip experiments. The assistance from staffs at the Michigan State University Macromolecular Structure Facility is gratefully acknowledged. The assistance of Dr. Chris Lau and associates from the Reproductive Toxicology Laboratory at US-EPA, Research Triangle Park, NC, in carrying out the in viva exposures was invaluable to this project. I am also thankful to Dr. Louis King from the Biochemistry department at Michigan State University for kindly providing the flow cytometry instrument and helping out with sample measurement. I greatly appreciate the friendship and supports from all my friends at Michigan State University, especially my colleagues at the Aquatic Toxicology Laboratory. TABLE OF CONTENTS LIST OF TABLES ............................................................................... viii LIST OF FIGURES ............................................................................... ix LIST OF SYMBOLS OR ABBREVIATIONS .............................................. xiv INTRODUCTION ................................................................................... 1 Acute Toxicity .............................................................................. 2 Subchronic and Chronic Toxicity ......................................................... 3 Tissue Distribution, Metabolism and Excretion ........................................ 3 Induction of Fatty Acid and Lipid Metabolizing Enzyme Activities ............... 5 Effect on Hepatic Microsomal Cytochrome P450 Enzyme Activity ............... 7 Effect on Leydi g Cell Function ............................................................ 7 Non-genotoxic Tumor Promoter ......................................................... 8 Previous Findings ........................................................................... 8 Study Objectives and Rationale ........................................................... 10 CHAPTER 1. MEMBRANE EFFECTS OF PFOS ............................................................. 14 Abstract ..................................................................................... 15 Introduction ................................................................................ 16 Material and Methods ..................................................................... 17 Chemicals ......................................................................... 17 Cell Biaassays ................................................................... 17 F law Cytametry Membrane F luidity Assay .................................. 20 Results ....................................................................................... 24 PF 0S Effects on Membrane F luidity and Membrane Potential ............ 24 Eflects of PF OS on EROD and Luciferase Activities ........................ 25 Discussion .................................................................................. 27 CHAPTER 2. GENE PROFILING IN RESPONSE TO PFOS ............................................... 41 Abstract .................................................................................... 42 Introduction ................................................................................ 43 Material and Methods ..................................................................... 45 Chemicals ........................................................................ 45 Cell Culture and Treatment ................................................... 45 In viva Treatment ................................................................ 46 Gene Chip Array Experimental Procedure .................................. 46 Gene Chip Data Analysis ........................................................ 48 Results ....................................................................................... 50 Overall Changes ................................................................. 50 vi Scatter Plats ....................................................................... 52 Gene Tree .......................................................................... 53 Gene List .......................................................................... 53 Discussions .................................................................................. 54 CHAPTER 3. COMPARISON OF GENE EXPRESSION TECHNIQUES ................................. 72 Abstract .................................................................................... .73 Introduction ................................................................................ 74 Material and Methods ..................................................................... 77 Chemicals ......................................................................... 77 Cell Culture and Treatment ................................................... 77 RNA Extraction and Purification .............................................. 78 Differential Display Procedure ............................................... 78 Subclaning of PA GE bands .................................................... 80 Gene Array Analysis ............................................................ 80 Results ....................................................................................... 81 Identifi/ and Subclone Differentially Expressed Genes ...................... 81 Comparison with Gene Chip Results .......................................... 83 Discussions ................................................................................. 84 CONCLUSION ..................................................................................... 94 APPENDICES ..................................................................................... 98 Appendix A ................................................................................. 99 Appendix B ............................................................................... 101 Appendix C .............................................................................. 112 REFERENCES ................................................................................... 120 vii LIST OF TABLES Table 2.1. List of genes induced significantly (p<0.0025) by PFOS in viva exposure. Long-term values are the mean fold-change and standard deviation (s.d.) for the four possible control/exposed comparisons ............................................................ 62 Table 2.2. List of genes suppressed significantly (p<0.0025) by PFOS in viva exposure. Long-term values are the mean fold-change and standard deviation (s.d.) for the four possible control/exposed comparisons ........................................................... 63 Table 3.1. Genes identified to be differentially expressed due to PFOS exposure in H4IIE cells in vitra. The “Clone seqs” indicates the number of clones sequenced from each original gel band. The “majority sequence”, if present, is the sequence that was present in 50% or more of the sequenced clones. The “proportion majority” is the number of clones out of the total that were the majority sequence ........................... 91 Table 3.2. Alterations in gene expression (as fold change from control) assessed by differential display and Genechip analysis. “dd” and “gc” refer to differential display or GeneChip analysis at 2 or 50 mg/L as indicated ................................................. 92 Table C.l. Comparison of differential display and gene chip methods .................. 97 viii LIST OF FIGURES Figure 0.1. Molecular structure of perfluorooctane sulfonic acid (PFOS), a synthetic, fully fluorinated, eight-carbon chain fatty acid analogue ...................................... 13 Figure 1.1. Density Plot of Fluorescence Emission of Pyrene Labeled Leukocytes that were treated with Pentanol and PFOS. Normal membrane fluidity is represented by ‘cells+pyrene’. The potent membrane affecting reagent pentanol increases cellular fluorescence at 450nm as it increases membrane fluidity. PFOS at 33 and 100 uM (equivalent to 16.5 and 50 mg/L) also increase membrane fluidity .......................... 34 Figure 1.2. Effects of 1% pentanol (positive control) and PFOS on fish leukocyte membrane fluidity. Cells were labeled with pyrene decanoic acid and then exposed to test chemicals as described in the text. Cells were analyzed by flow cytometry as described in text. Error bars represent standard deviation of 3 determinations. Results were analyzed by ANOVA with Dunnett's test using DMSO exposed cells as control; * p<0.05; ** p< 0.01; *** P< 0.001 ................................................................. 35 Figure 1.3. Effects of PFOS on mitochondrial membrane potential. Cells were labeled with the mitochondrial membrane potential dye J C-1 and were exposed to test chemicals. Cells were analyzed by flow cytometry as described in the text. Error bars represent standard deviation for three determinations. Valinomycin (100 nM), a reagent known to decrease mitochondrial membrane potential, was used as a positive control. Values were ix compared to the WBC control by ANOVA followed by Dunnett's test * p 90% humidity. For bioassays cells were plated in 96-well culture ViewPlates (Packard Instruments, Meriden, CT) at a density of approximately 15,000 cells in 250 pl media. Cells were dosed 24 hr after plating and were exposed for another 72 hr. E2 was dissolved in acetonitrile and PFOS was dissolved in methanol. Each exposure concentration was dosed in triplicate with 2.5 ul of extract solution, the final concentration of solvents was 0.5 % v/v or less. At least three replicate standard calibration curves ranging from 0.15 to 500 pM E2 were used with each assay. Each sample was dosed in six serial dilutions (1:3 diluting step) with 3-4 replicates per dilution. The exposure time for all bioassays was 72 hr. In competition experiments the concentration of E2 added was 10 pM, equivalent to an EC20. There were at least three blank and solvent control replicates on each plate. Cell viability for MVLN cells was assessed using the same method as for the H4IIE and PLHC-l cells. Luciferase activity was determined as described for the H4IIE cells. F low Cytometty Membrane Fluidity Assay Pyrenedecanoic acid (Molecular Probes, Eugene, OR) was dissolved in 0.03% ethanol and 0.1 M phosphate buffer (pH=7.4) to a concentration of 300 pM. JC-l (5,5',6,6'- tetrachloro—l,l',3,3'-tetraethylbenzimidazolylcarbocyanine iodide, Molecular Probes, Eugene, OR) was dissolved in DMSO to a concentration of 150 pM. The stock solutions were stored in the dark at —20°C after flushing the headspace with N2. Working solutions of pyrenedecanoic acid and JC-l were prepared freshly on the day of assay by diluting stock solution ten times to the concentration of 30 uM and 15 pM, respectively. 20 The effects of PFOS and related chemicals on membrane fluidity were investigated by use of flow cytometry. Fish blood cells were used as a model membrane system and membrane fluidity was measured using the excimer-forming lipid technique with pyrenedecanoic acid (Pownall and Smith, 1989). Fish blood cells were chosen rather than cultured cells as the membranes of cultured cells must be perturbed to release the cells from the culture dishes. In addition most cultured cells have been 'immortalized' and so cannot be considered normal. Fish blood cells represented a readily available source of cells that could be easily manipulated in the laboratory without causing undue stress to the cells. The excimer-forming lipid method is based on the formation of excimers (dimer of excited monomer and ground state monomer) of fluorescent pyrene molecules. The emission spectrum of pyrene is composed of two parts: one due to the excited pyrene monomers, and the other, at longer wavelength, originating from excimers formed upon collision of an excited pyrene with a ground-state pyrene. The rate of the excimer formation is dependent on the translational diffusion rate of pyrene molecules, which are incorporated into the cell membrane. Therefore, the ratio of excimer fluorescence to monomer fluorescence intensities (IE/1M) is proportional to membrane fluidity (Masuda et al., 1987). JC-l (5,5',6,6'-tetrachloro-1,1',3,3'- tetraethylbenzimidazolylcarbocyanine iodide) is a novel cationic carbocyanine dye that accumulates in mitochondria. The dye exists as a monomer at low concentrations and yields green fluorescence, similar to fluorescein. At higher concentrations, the dye forms J-aggregates that exhibit a broad excitation spectrum and an emission maximum at ~590 nrn. These characteristics make JO] a sensitive marker for mitochondrial membrane potential (Cossarizza et al., 1993). 21 Pyrenedecanoic acid and JC-l were excited with 365nm and 488nm argon lasers respectively. The fluorescence intensities of monomer and excimer pyrenedecanoic acid were determined using a FACS Vantage flow cytometer (Becton Dickinson, San Jose, CA) equipped with bypass filters of 400:15 nm and 450:30 nm, respectively. JC-l fluorescence was determined at 530i30 nm and 590:42 nm for monomer and J- aggregate respectively. At least 10,000 cells were examined in each sample. Cell scattering was shown as contour plot for FCS and SSC. Fluorescence intensities were recorded as histograms with event number (cell count) vs. channel number (fluorescence intensity). Preparation of Carp Leukocytes Carp were anaesthetized with MS-222 (250 mg/L in water). Blood was collected from the caudal vein into a heparinized syringe, an average 2.5 ml blood per fish can be collected in this way. During the course of these experiments blood was collected on 3 to 4 occasions and the whole blood of 3 to 4 individual fish was collected and pooled on each occasion. Three ml of Histopaque-1077 (Sigma, St Louis MO), was added to a 15 ml centrifuge tube and allowed to warm to room temperature. Three ml of the collected fish blood was carefully laid on top of the histopaque before centrifugation at 400 x g for 30 min at room temperature. After centrifugation, the upper serum layer was removed with a Pasteur pipette and discarded. The opaque interface (white blood cells and histopaque) was transferred to clean centrifuge tube, 10 ml PBS was added and the mixture was mixed gently. The cells were centrifuged at 250 x g for 10 min at room 22 temperature, the supernatant was discarded and the cell pellet was resuspended in 5 ml PBS before another centrifugation at 250 x g for 10 min. The final cell pellet was resuspended in 0.5 ml PBS (or McCoy’s 5A medium). Cell numbers were determined in a hemocytometer and the final cell concentration was adjusted to 1 x 105 ~ 1.5 x 106 cells per 200 pl of suspension. Labeling afCam Leukocytes Labeling was performed by adding 100 pl of 30 uM pyrenedecanoic acid solution, 100 pl of 15 DM JC-l solution and 200 pl of the cell suspension to a 5 ml round-bottom tube and gently mixing for 15 min at 25°C, excess label was removed by two washes with PBS before the final volume was adjusted to 1 ml with PBS. Chemical treatments including blanks, solvent controls, and positive controls (1% pentanol for membrane fluidity and 100 nM valinomycin for mitochondria membrane potential), and test compounds in serial dilutions were carried out by incubating the labeled cells with test chemicals for 15 min at 25°C. For PFOS each treatment was performed in triplicate, for other chemicals single determination was sufficient to demonstrate their inactivity in the assays as performed. The concentrations of positive controls were based on previously published data (Pownall & Smith, 1989; Cossarizza et al., 1993). F low Cvtametrv Data Analysis Flow cytometry data were acquired and analyzed using CellQuest software (Becton Dickinson, San Jose, CA) interfaced to the flow cytometer. The raw data from each histogram were extracted, and copied to a Microsoft Excel spreadsheets for subsequent 23 analysis. Total fluorescence intensity for each wavelength was calculated as sum of event number times channel number. Fluorescence ratios were calculated as the ratio of the total fluorescence intensities. Appropriate statistics were performed on multiple determinations of the fluorescence ratio. Statistical Analysis All cell bioassay data were collected electronically and converted into Excel spreadsheet format. Dose response curves were analyzed using Microsoft Excel 98, ANOVAs and Tukey’s test were conducted using SYSTAT 10 (SPSS, Chicago IL). RESULTS PF 0S Effects on Membrane F luidity and Membrane Potential Exposure to PFOS significantly increased membrane fluidity of fish leukocytes (Figure 1.1) at 33 and 100 pM (16.5 — 50 mg/L). The degree of the maximal response observed was similar to that observed for 1% pentanol, the positive control for the experiment. In subsequent experiment the response was determined to be dose-dependent (Figure 1.2). In two independent experiments the least dose significantly different (p<0.05) from the control were 15 mg/L (30 pM) and 16.5 mg/L PFOS. Therefore, the threshold for effects on membrane fluidity in vitra is approximately 15 mg/L. PFHS and PFBS, compounds that have similar structures to PFOS but with shorter carbon chain lengths, had no effect on membrane fluidity in the same concentration range used for the PFOS exposures (results not shown). Therefore, as with other effects 24 observed for perfluorinated compounds the response seems to be related to the length of the carbon chain (Hu et al., 2002b). The effects of PFOS on mitochondrial membrane potential were also determined by flow cytometry (Figure 1.3). Mitochondrial membrane potential was inversely related to the PFOS concentration. The maximum decrease observed was similar in magnitude to that observed for 100 nM valinomycin, the positive control. The variability in determination of mitochondrial membrane potential was greater than that for the membrane fluidity. Statistical analysis of the membrane potential data revealed that the first dose significantly different (p=0.0018) from control was 30 11M (15 mg/L), which is similar to the threshold concentration for effects on membrane fluidity. Effects of PF OS on EROD and Luciferase Activities PFOS alone did not induce cytochrome P450 1A1 (CYP1A1), as measured by EROD activity, compared to solvent-exposed cell culture controls (Figure 1.4A). TCDD induced EROD activity in a dose-dependent manner, with the greatest induction being 17 fold with an EDSO of approximately 0.01 11ng (Figure 1.4A). To assess the interactive effects between TCDD and PFOS, cells were exposed to the two chemicals in combination. Cells were dosed with TCDD alone, or with TCDD in combination with PFOS at concentrations ranging from 0.0001 to 10 mg PFOS/L. Co-exposure of cells to PFOS and TCDD increased the CYP1A1 activity induced by TCDD (Figure 1.5A). Compared to the TCDD standard dose-response curve, the addition of PFOS increased both the slope of the curve and the magnitude of maximum response, with PFOS at 0.1 25 mg/L causing the greatest increase in the TCDD response. The interactive effects observed were statistically significant at 0.2 ug/L TCDD plus 0.1 mg/L PFOS (p<0.05), 1 ug/L TCDD plus 0.01 mg/L PFOS (p<0.05), and 1 ug/L TCDD plus 0.1mg/L PFOS (p<0.01) (Figure 1.5B). In the last combination, the addition of PFOS increased the effect of TCDD by 40%. Results were similar for PFOS exposure to PLHC cells. PFOS alone exhibited no detectable effect on CYP1A1 induction (Figure 1.4B). For PLHC-1 cells the standard TCDD dose-response curve had a slightly different shape compared to that of the H4IIE- luc cells, however the general trend of interactive effects was similar to that of the H4IIE- luc cells. The most significant interactive effects in the PLHC cells were observed at a TCDD concentration of 0.2 ug/L and at a PFOS concentration of 0.1 mg/L (p<0.01), which increased the effect of TCDD by approximately 40% (Figure 1.6). To determine whether the PFOS related increase was specific to the CYP450 enzyme assay used, the luciferase assay was also conducted with H4IIE-luc cells dosed with PFOS and/or TCDD. In H4IIE-luc cells PFOS alone did not induce AhR-mediated luciferase activity relative to that of the control. In contrast, TCDD induced luciferase activity in a dose-dependent manner (Figure 1.4C). Exposure to l ug/L TCDD plus 0.1 mg/L PFOS (ANOVA, p<0.05), and 0.2 ug/L TCDD plus 0.1 mg/L PFOS (ANOVA, p<0.05), significantly increased induction over TCDD alone, with the maximum of increase by 40% (data not shown). 26 MVLN cell treated with PFOS showed no indication of induction of estrogen receptor- controlled genes at concentrations as high as 10 mg/L. In contrast, 17B-estradiol (E2) added to the cells strongly induced the production of luciferase in a dose dependent manner with maximal activity observed between 25 and 100 nM E2 (Figure 1.7). To determine whether the interactive effects observed between PFOS and TCDD in the AhR reporter gene system were also acting in the ER reporter system, experiments were conducted with mixtures of PFOS and E2 and TCDD (Figure 1.7). As in the AhR bioassay system a moderate (approximately 40%) PFOS dependent increase in the E2- mediated expression of luciferase was observed at the three higher doses of E2. The additional increase in luciferase activity was dependent on the dose of PFOS, with a PFOS concentration of 0.1 mg/L resulting in the greatest increase in expression at all E2 concentrations. In addition, in cells treated with PFOS alone, concentrations as high as 10 mg/L, did not adversely affect MVLN cell viability or the responsiveness of the ER- mediated pathway (results not shown). In contrast TCDD at 0.5 ug/L caused a significant decrease in the activity of the ER-mediated pathway and cell viability in the MVLN cells (Figure 1.7). DISCUSSION These studies indicated that of the chemicals tested only PFOS significantly altered membrane properties in the concentration range tested. The effects observed were subtle and different effects occurred at distinctly different concentrations. It is our hypothesis that these effects represent a series of concentration dependent changes in specific 27 membrane properties brought about by the detergent like effects of PF OS on membrane lipids and/or proteins. It has become apparent over recent years that the physical structure of all cellular membranes is tightly controlled, and that the physical properties of different membranes are important for their function. There is evidence of extensive differentiation of lipid components between the two sides of many biological lipid bilayer membranes. As well as these ‘vertical’ differences in membrane composition, it has been demonstrated that lateral domains exist within membrane layers. In particular, cholesterol forms ‘raft’ like structures that are characterized by their low detergent solubility (Galbiati et al., 2001). It has also been demonstrated that outside these rafts the lateral movement of individual lipid molecules within the ‘bulk’ phase of the membrane appears to be limited. Lateral movement of these molecules appears to progress as a series of transitions between distinct lipid compartments within the ‘bulk’ membrane phase (Kawasaki et al., 2001). It is clear fiom these observations that any alterations in cellular membrane properties caused by xenobiotics could have a considerable impact on the various fimctions of the membrane and its substructures. Alterations in membrane fluidity have been measured as a consequence of decreased cell proliferation (Beguinot et al., 1987) and obesity (Beguinot et al., 1985). In addition to the direct physical effects of PFOS on cellular lipid components it is also possible that alterations in cholesterol metabolism may have consequences for membrane function. Alterations in membrane fluidity have been associated with alterations in cellular or membrane cholesterol by a number of studies (Beguinot et al., 1985; Beguinot et al., 1987; Jefferson et al., 1990). PFOS has been demonstrated to be 28 hypocholesterolaemic in primates during long-term sub-chronic exposure (Seacat et al., 2002b). Therefore, the observed increases in membrane fluidity due to PFOS exposure could be compounded during in viva exposures by decreases in the cholesterol content of the membranes resulting in further increases in membrane fluidity. While alterations in the permeability of the membrane may have more directly observable effects, our understanding of the consequences of changes in specific aspects of membrane fluidity and function is less clear. The in vitra systems in these studies were used as means of probing effects of PFOS on membrane permeability. While PFOS itself was inactive in AhR and ER receptor- mediated pathways, it was able to increase the amount of TCDD and Estradiol reaching the cell signaling pathways. Thus, PFOS was hypothesized to be able to increase the permeability of the cell membranes to these two ligands. The fact that these responses are neither ligand nor biochemical pathway specific yet occur at essentially the same PFOS concentrations suggests that the effect is at the level of the cell membrane rather than effects on specific transporter protein systems. In addition PFOS at concentrations of 15 mg/L and greater was able to decrease mitochondrial membrane potential in exposed cells. These results are in agreement with functional assessments of the effects of PFOS on mitochondrial energy production, which demonstrate that at 10 uM PFOS (equivalent to 5 mg/L) only weakly affected energy production (Starkov and Wallace, 2002). These alterations are attributed to increased membrane permeability resulting in dissipation of the mitochondrial proton gradient. These effects were distinct from the more potent effects of other perfluorinated chemicals that acted as either classical 29 protonophoric uncouplers (Starkov & Wallace, 2002) or chemicals capable of inducing the mitochondrial membrane permeability transition (Sokol et al., 2001). Since the optimal PFOS concentrations for the ligand permeability effects observed were similar (approximately 0.1 mg/L), as were the extents of the increase (approximately 40%) in effect, we hypothesize that the increase in permeability is non-specific and so is most likely a result of lipid/PFOS interactions. Alternatively, it is possible that the amphiphilic nature of PF OS acts to improve penetration of hydrophobic ligands (TCDD and E2) through the cell membrane or the ligands may be involved in a co-transportation mechanism, although it is generally assumed that at least TCDD crosses the membrane by simple diffusion. These studies have also demonstrated the ability of PF OS to modulate membrane fluidity in vitra. The threshold concentrations of PFOS which elicited these effects were in the range of 5 to 15 mg/L which is similar to the threshold concentrations that have been observed for cell permeability effects and for other PFOS-mediated cellular responses, such as gap-junction intercellular communication (Hu et al., 2002b). Together these results suggest a range of responses linked by a common mode of action. From these studies on membrane fluidity it appears that the mechanism of action is the interaction of PFOS with membrane lipids since the regulator of the association of the pyrene dimer is the horizontal fluidity of the lipid bilayers that form the cellular membrane. This fluidity should not be interpreted as indicating that the cell membranes are any more ‘leaky’ than unexposed membranes. The transport or translocation of compounds across the cell 30 membrane is a different physiological process, not directly related to the horizontal fluidity of the membrane. The kinetics of occurrence for the effects observed in these experiments indicate that the interaction of PFOS with cell membranes is rapid, with effects observed after only 15 minutes of incubation. Similarly, rapid effects and rapid recovery have previously been described for the effects of PFOS on Gap Junction Intercellular Communication (GJIC) (Hu et al., 2002b). The short time period before the onset of the effects preclude the possibility of direct incorporation of these fatty acid analogues into membrane lipid, which would normally require a timeframe of hours. Indeed, the relatively short chain length of PFOS compared to the normal 16 and 18 carbon chain fatty acids present in phospholipids would suggest that PFOS is unlikely to be covalently inserted into phospholipids. While the effects observed here on membrane fluidity are suggestive of lipid/PFOS interactions previous work on the effects of PFOS on GJIC suggest a mechanism more related to protein/lipid interactions (Hu et al., 2002b). It seems most probable given the highly hydrophobic nature of the fluorocarbon chain of PFOS that this compound may be most active at lipid/protein interfaces within membranes. This mechanism of action is supported by the highly surface active nature of PFOS and other perfluorinated compounds. It is clear that the effects on membrane fluidity (15 mg/L) are observed at different concentrations from those observed on membrane permeability (0.1 mg/L). 31 It is difficult to interpret what the observed changes in membrane fluidity and permeability mean in viva. We are aware of no studies which have linked membrane fluidity effects to other toxic endpoints. Those studies that are available suggest rather that alterations in membrane fluidity are a consequence of diseased or abnormal conditions (Beguinot et al., 1987; Beguinot et al., 1985). The experiments described here and those of other investigators clearly demonstrate that the alterations in membrane properties caused by PFOS do not result in the classical mitochondrial membrane permeability transition, which leads to apoptotic cell death (Sokol et al., 2001). It is therefore unclear what, if any, would be the likely consequences of the subtle membrane fluidity alterations at the whole organism level. We have however previously demonstrated that the effects of PFOS on GJIC observed in in vitra exposures also occurred in viva (Hu et al., 2002b). All the assay systems used here were in vitra and results cannot be expected to directly reflect in vivo conditions. Notably PFOS has been shown to bind to a variety of proteins both intracellular (Luebker et al., 2002) and extracellular (Jones et al., 2002). It is possible that binding of PFOS to proteins could significantly ameliorate the membrane related effects observed here if the affinity for protein is greater than that for membranes. Additional studies will need to be conducted to determine whether the observed effects actually occur in viva. Studies on the inhibition of gap junctions by PFOS have indicated that effects observed in cell culture also occur in viva albeit at different concentrations (Hu et al., 2002b). Given that the tissue concentrations of PFOS measured in some organisms can reach 1-10 mg/kg (Giesy and Kannan, 2001; Kannan et al., 2001a; 2001b) 32 we would expect that to some extent alterations in membrane fluidity and permeability might occur providing that there are no other factors, which might ameliorate these effects. If the suggested alterations in membrane fluidity do occur there is little evidence to indicate whether adverse whole organism effects are likely to occur. 33 Fig 1.1 A B 53 I. V 2 .. .2 S 'a In Emission at 400 nm Emission at 400 nm Cells+Pyrene Cells+Pentanol C D O V) v ‘5 s: .52 S '5 LL} Emission at 400 nm Emission at 400 nm Cells+PFOS(15 mg/L) Cells+PFOS(50 mg/L) 34 Fluorescent Ratio 450/400 Figure 1.2 1.3 1.1~--— H, ___ w--. a W I I I I I r—l—I Fl... l--l I (WIT-I—M—{ITE‘M i. *-I- __ 0.8 ~— 0.7 w 0.6 I T T T T 1 . e . ¢ $©Qo$08D~r°§Doe°W$ (9&3) 55$ 69¢? 5w"? ‘0 e e 0 Exposure Type and Concentration 35 Figure 1.3 an: al— 0.95 "TIT . T I 1 _ Til—HI. i *T||_H||. — L .I —i u . T I _ _ I _ . _ P _ m. m m .7.. O O O 0 Did“ oofioomqu—ATH Exposure Type and Concentration 36 Figure 1.4 2000 A 1800 ~ 1600 . 9mm) 1400 ~ E" PFOS 1200 - 1000 ‘ 800 “ 600 I 400 “ 200 " 0 . 1200 13 1000 “ ty % control 1 EROD Activ OO O O 600 ‘ 400 “ 200 “ r 0. . . . , i 1000 900 800 I e-TCDD 700 " 600 500 400 300 200 100 EROD Activity % control Luciferase Activity % control 0 1 i 1 0.0001 0.001 0.01 0.1 1 10 Concentration PFOS (mg/l)/ TCDD (pg/l) I l fi 37 Figure 1.5 S mm WP A O I mmy mom DMD .. . www ‘ n: 52/- .A./ /:M Ao- A 5 4 3 2 1 _ob:oo 8 2528 b_>uom QOMm 10 1 0.001 0.01 0.1 0.0001 TCDD (ng/ml) 38 Figure 1.6 EROD relative to control *1: 0.0001 0.001 0.01 0.1 1 10 PFOS (mg/L) 39 Figure 1.7 Luciferase Activity (RLU) 0.15 0.10 0.05 0.00 , ’1 z ' ’E2+PFOS E2+TCDD \ I I I I J 20 40 60 80 100 120 E2 (nM) 40 CHAPTER TWO Hu, W.Y., Jones, P.D., Celius, T, and Giesy, JP. (2003) Identification of genes responsive to perfluorooctane sulfonic acid using gene expression profiling. European Journal of Toxicology and Pharmacology (submitted) 41 ABSTRACT Perfluorooctane sulfonic acid (PF OS) is widely distributed in the environment including in the tissues of wildlife and humans, however its mechanism of action remains unclear. In the current study, the Affymetrix rat genome U34A gene chip was used to identify alterations in gene expression due to PFOS exposure. Rat hepatoma cells were treated with PFOS at 2 or 50 mg/L in culture medium for 96 hr, and Sprague-Dawley rats were orally dosed with PFOS at 5mg/kg/day for 3 d or 3 wk. Genes that were significantly (p<0.0025) induced were primarily genes for fatty acid metabolizing enzymes, cytochrome P4505, or genes involved in hormone regulation. The significantly down regulated genes were mostly involved in signal transduction pathways. Consistent expression profiles were obtained for replicate exposures within treatment, and for short- terrn and long-term in viva exposures. Limited similarity in gene expression profile was observed between the in viva and in vitra exposure systems. The structural similarity between PFOS and endogenous fatty acids may explain why the major pathway affected by PFOS is postulated to be peroxisomal fatty acid beta-oxidation. Key words: PFOS, gene expression, fatty acids metabolism 42 INTRODUCTION Perfluorinated fatty acids (PFFAS) are synthetic, fully fluorinated, fatty acid analogues. Recent studies indicate that perfluorooctane sulfonic acid (PFOS) is the most commonly found compound in the tissues of wildlife, perfluorooctane sulfonamide (PFOSA), perfluorooctanoic acid (PFOA), and perfluorohexane sulfonate (PFHS) have also been detected in the tissues of several species (Giesy and Kannan, 2001). To date, most toxicological studies have been conducted on PFFAs such as PFOA and perfluorodecanoic acid (PF DA), rather than the more environmentally prevalent PFOS. PFOS appears to be the ultimate degradation product of a number of commercially used perfluorinated compounds (Giesy and Kannan, 2002). The concentrations of PFOS found in wildlife are greater than other perfluorinated compounds (Giesy and Kannan, 2002; Kannan et al., 2001a, 2001b). The mechanisms by which some PF FAS elicit their toxic effects are not well known. For example several PF FAS have been reported to be peroxisome proliferators. PF FAs, such as PFOA and PFDA can interfere with lipid metabolism by increasing peroxisomal fatty acid B-oxidation, and inducing several hepatic enzyme activities (Sohlenius et al., 1996). Both in viva and in vitra exposures to PFOA result in increased activities of peroxisomal acyl-CoA oxidase, which catalyzes the first and rate-limiting step in fatty acid oxidation (Sohlenius et al., 1994). Fatty acid oxidation is also a process known to produce hydrogen peroxide, an oxidative radical, such that PFF As can lead to oxidative stress and could possibly result in DNA damage (Sohlenius et al., 1994). 43 Some PFF As, including PFOA and PF DA, have been shown to be involved in regulating tissue fatty acid composition and content. PFOA can reduce cholesterol and triacylglycerol concentrations in serum, increase liver triacylglycerol concentration, and reduce hepatic lipid output (Haughom and Spydevold, 1992). Treatment with PF DA can inhibit acyl-CoA synthetase activity and result in an increase in the level of free fatty acids, which are known to be able to activate protein kinase C (PKC), and lead to a signaling cascade that is important for normal cell function, cell proliferation and gene expression (Reo et al., 1996). Hepatic microsomal cytochrome P450 enzymes were induced in rats treated with PFOA (Permadi et al., 1992). In the CYP4A sub-family, nine enzymes specific for fatty acid w-hydroxylation, were significantly induced with exposure to 500 11M PFOA for 7 d. Recent studies have also demonstrated effects of PFOS in vitro and/or in viva on Gap Junctional Intercellular Communication (Hu et al., 2002b), membrane fluidity (Hu et al., 2002a) and serum steroid binding globulins (Jones et al., 2002). Chronic exposure of rodents and primates to PFOS resulted in significantly altered concentrations of cholesterol in blood (Seacat et al., 2002a, 2003). In the current study, the effects of PFOS on gene expression were determined using the Affimetrix GeneChip array, a genome-wide expression analysis method based on the rat genome. Our null hypothesis is that PFOS exposure does not cause mechanism specific modulations of gene expression. This screening approach was used to identify genes responsive to PF OS with the intent of identifying critical target pathways for the biological effects of PFOS. In vitro and in viva exposures were used to allow comparison of gene expression profiles between in vitro and in viva models, and long-term verses short-term exposure. MATERIALS AND METHODS Chemicals Perfluorooctane sulfonic acid (PFOS) was obtained from 3M company (St. Paul, MN). The PFOS (potassium salt) used for in vivo experiments was purchased from F luka Chemicals (Buchs, Switzerland), chemical analysis of the isomer patterns revealed that it was essentially the same as the product obtained from 3M. Cell Culture and Treatment H4IIE rat hepatoma cells, between passages 5 and 15, were cultured in 100 mm disposable tissue culture dishes at 37°C under sterile conditions (pH=7.4) in a humidified 5/95% C02/air incubator (Forma Scientific, Model 8173). Cells were grown in Dulbecco’s Modified Eagle Medium (DMEM, Sigma D-2902, Sigma, St. Louis MO), supplemented with 10% fetal bovine serum (FBS, Hyclone, Logan, UT). At confluence, cells were removed from the dish with trypsin/EDTA (Hyclone, Logan, UT), and split into four tissue culture plates. The cells were given 24 h after splitting to allow for attachment, the medium was then replaced and cells were dosed with PFOS to achieve final concentrations of 2 mg/L and 50 mg/L, methanol was used as solvent control, and the blank control received no dose. Cells were incubated for 72 hr after exposure. 45 In vivo Treatment Sixty-day old Sprague-Dawley rats (males 294:4 g; females 209:2 g) were obtained from Charles River Laboratories (Raleigh, NC), and housed at 20-24°C in humidity- controlled (40-60%) facilities at the US-EPA Reproductive Toxicology Laboratory (Research Triangle Park NC). Estrous cycle was not determined in female rats and breeding was not a targeted endpoint for these studies either. Rats were randomly assigned to two blocks, block one with six males and six females, and block two with four males and four females. Block one was exposed to PFOS for 21 d, block two was exposed for 3 d. Within each block, half of the males and half of the females were randomly assigned to treatment or control groups. Rats received PFOS (5 mg/kg) or vehicle control (0.5% Tween-20) daily by oral gavage at a rate of 1 ml/kg body weight. At the end of exposure, animals were sacrificed, livers were removed within 1-2 minutes of sacrifice and portions of liver were removed and placed in TriReagent. Liver samples were processed for RNA isolation on the same day as collection. GeneChip Array Experimental Procedure The Affymetrix rat genome U34A gene chip array was purchased from Affymetrix Inc. (Santa Clara, CA). The oligonucleotide probes on U34A array cover approximately 8800 known genes and Expressed Sequence Tags (ESTS) in the Rat genome. Transcripts were selected from Genebank Unigene build 34 and the dbEST database. Total RNA from cell cultures and rat liver samples were extracted using TriPure Isolation Reagent (Boehringer Ingelheim, Germany) using the manufacturers recommended 46 procedures. The optical densities of RNA samples were measured at 260 nm and 280 nm, and the 260 nm/ 280 nm ratio was evaluated as a measurement of nucleic acid purity. RNA concentration was quantified using optical density at 260 nm. The quality of RNA was evaluated by the appearance of distinct of 183 and 28s ribosomal RNA bands on 1% agarose gel. First and second strand cDNA was synthesized from total RNA samples using the SuperScript Choice System (Gibco BRL life Technologies, Cheshire, UK). High quality total RNA (16 ug) was used as the starting material and lul 100 pmol/ul T7-(dT)24 primer (5’ — GGC CAG TGA ATT GTA ATA CGA CTC ACT ATA GGG AGG CGG — (dT)24 —3’; Genset Crop. San Diego, CA) was used to prime the reaction. After double standed cDNA clean up and quality check, an in vitra transcription reaction was conducted to produce biotin-labeled cRNA from the cDNA. The cRNA was then purified and fragmented for hybridization analysis. Following hybridization and washing, staining and scanning procedures were performed in the Genomics facility on Michigan State University campus (Fluidics Station 400 and Hybridization Oven 640 from Affymetrix, Santa Clara, CA). Briefly the biotin-labeled cRNA was combined with probe array control, BSA, and herring sperm DNA into a hybridization cocktail, and applied to the probe array after a cleanup procedure. It was then allowed to hybridize on the array for 16 hr at 45°C. Following the hybridization, the arrays underwent an automated washing, staining and scanning protocol on the fluidics station (Affymetrix, Santa Clara, CA). Each complete probe array image was stored in a separate data file identified by experimental name. 47 A total of nine chips were used in this study. Three were used to analyze samples from the in vitro exposure (solvent control, PFOS at 2mg/L and 50mg/L), while four chips were used for examining the long-term in viva samples (2 animals exposed to solvent controls and 2 animals exposed to PFOS at 5 mg/kg/day for 21d). Finally, two chips were used for the in viva short-term sample (1 animal treated with solvent control and 1 animal exposed to PFOS at 5 mg/kgd for 3 d). The PFOS concentration was measured in each rat liver samples with an average of 600 mg/kg—body weight for 21d exposure, and 90 mg/kg-body weight for 3 (1 exposure. Data collected from the nine chips was transferred to a Microsoft Access database. GeneChip Array Data Analysis Each image file was analyzed and data was retrieved using the Affymetrix “data mining” tool (Affymetrix Santa Clara, CA). Initial data normalization and filtering was also conducted. The output file was then stored in a Microsoft Access database and the initial data analysis was conducted using Microsoft Access query design. Cluster analysis, Genetree construction and pathway analysis were conducted using GeneSpring software (SiliconGenetics, Redwood City, CA). Scatter plot The scatter plot is useful for examining the expression levels of genes in two distinct conditions, samples, or normalization schemes. To evaluate the reproducibility of results for individual animals and cell cultures, scatter plots based on a correlation analysis were prepared for the different samples. 48 Gene tree Genes can be classified in a manner similar to classification of organisms into phylogenic dendrograms or trees. As organisms sharing evolutionary properties tend to be clustered together, genes sharing similar expression pattern can be used to determine the similarity in responses of species, doses, or duration of exposure. The vertical distance along the branches of such a tree represents a measure of degree of similarity. The Genetree algorithm of GeneSpring was used to draw a hierarchical dendrogram of clustered genes according to their expression profiles among treatments. The algorithm calculated the correlation for each gene with every other genes in the set. Then pairs of genes exhibiting the greatest correlations were merged and their expression profiles averaged. The new composite 'gene' was then compared with all the unpaired genes. This was repeated until all of the genes had been paired. Based on the way their expression was altered across the nine samples, the 8800 genes were grouped on the horizontal axis with the nine treatment group clustered based on how they affect gene profiles on the vertical axis. Pathway analysis and Target path way Based on the genes that were identified as having their expression modulated by exposure to PF OS, a pathway analysis was performed by linking genes via their Enzyme Commision (EC) numbers to their positions in known metabolic pathways as described in metabolic pathway maps in the Kyoto Encyclopedia of Genes and Genomes (http://wwwgenomeadjp/keggI. 49 Statistical Analysis Statistical analysis of gene array data is complicated by the large amount of data generated, the different arraying technologies used by different manufacturers and the generally small sample sizes used in experiments involving Gene Arrays. In addition, many of the software programs used to analyze capture and analyze array data perform at least initial background corrections and some statistical analysis using proprietary algorithms. For example GeneChip probe arrays are manufactured such that each gene is represented with a series of 11-20 probes pairs (each probe is 25 bp in length). Each probe pair is composed of a perfect match probe and a mismatch probe, the mismatch probe has almost the same sequence as that of the “perfect match”, except one nucleotide difference. This so-called “tiling” design serves as an internal control for hybridization specificity, which allows consistent discrimination between closely related target sequences. While the number of chips used in the current study was limited to 9, these chips represented exposures at a variety of concentrations both in vitra and in viva. RESULTS Overall changes To determine the overall gene expression changes associated with PFOS exposure, genes were classified by their fold change in expression. Expression of only about 5% of the genes analyzed were significantly (p<0.0025) induced or suppressed beyond a 3-fold change in expression in samples treated with PFOS relative to the control. However, this still represents some 400 genes whose expression was altered by exposure to PF OS. The use of a 3-fold cutoff for significance is based on a variety of previous studies (Wan and 50 Nordeen 2002; Gerhold et a1. 2002) and the desire to identify the most dramatically alterations in gene expression. The Affymetrix GeneChip system utilizes extensive statistical analysis of the array image before a gene is reported as induced or suppressed. The detection of a single gene product is based on analysis of between 11 and 20 oligonucleotide ‘probe pairs’. Each probe pair consists of two 25-mer oligonucleotides, one ‘perfect match’ for the target sequence and one with a base mismatch at nucleotide13. The relative spot intensities between all perfect match and mismatch probe pairs give these chips extraordinary sensitivity and specificity. In ‘comparison analysis’ mode the software compares the arrays for two samples analyzed on two different chips. One array is designated as the baseline (control) and the other the experiment (exposed). Before comparing the two arrays scaling and normalization are carried out automatically to correct for variations in overall signal intensity and heterogeneity between the two arrays. During a comparison analysis, each probe set on the experiment array is compared to its counterpart on the baseline array and, using Wilcoxon’s Signed Rank Test, a ‘Change p-value’ is calculated indicating an ‘increase’, ‘decrease’, or ‘no change’ in gene expression (GeneChip® Expression Analysis Technical Manual). The degree of significance for the change call is user specified, for chip comparisons conducted in the current study default probability cut-off values were used so in all cases when a gene expression is reported as altered, p<0.0025. For the determination of the degree of alteration in gene expression the software determines the “Signal Log Ratio” using a one-step Tukey’s Biweight method by taking a mean of the log (base 2) ratios of probe pair intensities across the two arrays 51 (Affymetrix® Microarray Suite 5.0 User’s Guide). This approach helps to cancel out differences in individual probe intensities, since ratios are derived at the probe level, before computing the Signal Log Ratio. Since log base 2 is used to determine the signal log ratio “Fold Change” = 25‘3"" “’8 km). From this discussion it is clear that although the analysis system reports only a ‘fold change’ in gene expression there is a high degree of statistical rigor in the determination of these changes in gene expression. Scatter Plots Scatter plots were first constructed by comparing the duplicates in control or 21 day in viva PFOS exposed samples. In each case, the majority of the data points fell into the reference space representing the 95% confidence interval for a correlation coefficient of 1.0 (Figures 2.1A and 2.18). This demonstrated the degree of reproducibility of the biological responses as well as the reproducibility of the analysis method. In contrast, when a sample from the long-term in viva exposure was plotted against a control sample from the same experiment a greater degree of scatter was observed (Figure 2.1C). Scatter plots comparing in vitra and in viva exposure systems showed differences in gene expression between two controls from the rat liver cells in viva and the genetically modified rat hepatoma cells in culture. A significant degree of data scattering and deviation from the reference lines were observed across all expression levels (Figure 2.1D). This demonstrated the relatively large difference in gene expression between the in vitro and in viva systems even without considering the impact of chemical exposure. 52 Gene Tree When the patterns of responses for all samples were examined using a 'Gene Tree' analysis, the in vitro samples exhibited a clearly different profile than did the in viva samples (Figure 2.2), consistent with the result observed in the scatter plot analysis. In all three in vitra samples, genes separated into two nodes according to their expression level. No distinguishing patterns were recognizable among the six in viva samples, since subtle differences among these samples were masked by the greater differences between the in vitro and in viva exposure systems. When a similar gene tree analysis was conducted using solely the in viva data, a group of genes exhibiting a distinct pattern among treatments was discemable (Figure 2.3). All of the genes in this group were expressed at a significantly (p<0.0025) greater level in the two long-term PF OS treated samples than in the controls. The short-term sample exhibited a pattern similar to that of the long-term exposure although lack of replicate analyses in the short term exposure prohibited statistical comparison. am A list of genes whose expression level was significantly (p<0.0025) altered by PFOS exposure was identified (Tables 2.1 and 2.2; Figures 2.4 and 2.5). For the long-term exposure two exposed and two control rats were available for comparison. Since the Affymetrix system is limited to comparing two chips at a time, each of the exposed rats was compared to each of the control rats providing a total of four estimates of the fold change in gene expression. This approach was taken to ensure that only consistent 53 alterations in gene expression were identified. The largest grouping of genes induced by PFOS exposure in viva were the cytochrome P4508 and genes that coded for lipid metabolizing enzymes. Several genes involved in hormone regulation and other regulatory processes were also induced significantly (p<0.0025). Several genes encoding factors involved in signal transduction pathways were suppressed by PFOS exposure as were genes involved in regulating neuro-system functions. Of the pathways represented by these altered genes the peroxisomal fatty acid 0 oxidation pathway seems to be most affected by exposure to PFOS (Figure 2.6). The gene expression for the enzymes involved in peroxisomal lipid metabolism were altered but those for the same pathway in mitochondria were not. DISCUSSION The results of this study illustrate the utility of high-throughput toxicogenomics methods to study the effects of a compound for which the mechanism(s) of action are still unclear. Gene expression analysis is useful in identifying chemical-specific alterations in gene expression to allow classification of toxicants and provide important insights into mechanisms of action (Hamadeh et al., 2002a, 2002b). Alterations in expression profiles were used to determine potential critical pathways affected by exposure to PFOS. It is clear that alterations in the concentrations of mRNA species do not necessarily translate to alterations in the corresponding enzyme concentration or activity. However, coordinated alterations in mRN A concentrations for a particular biochemical pathway provide strong evidence for an effect of PFOS on that pathway. Confirmation of 54 alterations at the protein/enzyme level is the next step in the assessment, but is beyond the scope of this study. Of the 8,800 functionally annotated genes and ESTs present on the array, only about 5% responded to PFOS with a more than three-fold change in expression. Differences between gene expression profiles were observed between the in vitro and in viva control samples. All three in vitra exposures exhibited an expression pattern distinct from that of the in viva samples. This could be explained by the differences in exposure system, dosage, toxicokinetics, toxicodynamics, levels of organization and functional integration, which make in viva exposure more complicated than in vitra exposure. Concentrations of PF OS were measured in the rat liver samples (Hu et al., 2002b), however the exposure concentration provided for the in vitro samples was the dose applied to the culture medium not the dose internalized by the cells. The genetic profiles of the transformed rat hepatoma cell line and the freshly isolated normal rat liver tissue would also be expected to be different. Also, rat liver tissue is composed of a variety of cell types, including hepatocytes, Kupffer cells, fibroblasts and stellate cells. Thus, the response of liver tissue to chemicals is different from a monoculture of cells such as the H4IIE cells that are composed solely of hepatocytes derived from a limited population of progenitor cells. While this finding may not be surprising the data provided demonstrates just how significant those differences, previously unknown, can be. Based on our results, interpretation of in vitro data and implication on in vivo response should be conducted with caution. No direct extrapolation of in vitro data should be made without further in vivo testing. 55 Previous studies have indicated that PF OS can be incorporated into cell membranes and elicits physical membrane effects both in vitra and in viva (Hu et al., 2002a; 2002b). PFOS inhibited GJIC in a dose-dependent fashion, which occurred rapidly and reversibly. PF OS increased fish leukocyte membrane fluidity and decreased mitochondria membrane potential in a dose-dependent fashion. The results from these studies established that PF OS could alter membrane structure and function, but they could not determine if these physical effects were also accompanied by effects on gene expression. In this study we have confirmed that the physical effects on membrane processes, such as alterations in GJIC (Hu et al., 2002b), are accompanied by alterations in lipid metabolizing enzymes. In mammalian cells, both mitochondria and peroxisomes are involved in the B-oxidation of fatty acids and the substrate specificity of the two systems overlap. Mitochondria oxidize mainly short, medium, and long, straight-chain fatty acids, while peroxisomes are capable of oxidizing very long straight-chain and branched-chain fatty acids. Short-chain fatty acids (2-6 carbons) are poor substrates for peroxisomes because of the low affinity of the peroxisomal B-oxidation enzymes for short-chain substrates. Results obtained through this study of gene expression indicated that PFOS specifically enhanced the peroxisomal but not mitochondrial B-oxidation. Fatty acid B-oxidation in peroxisomes is carried out in four consecutive steps. The enzymes involved in these processes were increased from 2- tolO-fold by in viva exposure to PFOS. Enzymes responsible for the equivalent functions in mitochondria were not significantly affected by PFOS exposure (Figure 2.6). 56 Other PF FAs have been reported to cause peroxisome proliferation (Berthiaume & Wallace, 2002). However, the response observed for PFOS was not characteristic of a 'classic' peroxisome proliferator. Genes which are indicative of peroxisome proliferation and other xenobiotic responses have recently been identified (Hamedah et al., 2002a). In the present study PFOS induced the gene expression level of camitine palmitoyl transferase (CPT I) in a manner similar to the architypal peroxisome proliferator Wyeth 14,643. However PFOS also increased the activities of carboxyesterase and CYP2B1, a response characteristic of phenobarbital inducible systems (Figure 2.7). PFOS exposure resulted in increases in the activity of thiolase and enoyl-CoA isomerase, enzymes not increased significantly (p<0.0025) by either of the above xenobiotics. Clofibrate, a classic peroxisome proliferator, was found to modulate expression of genes involved in fatty acid B-oxidation in both peroxisome and mitochondria. Finally, while CYP4A is strongly induced by other peroxisome proliferators it was not increased by exposure to PFOS in either the in viva or in vitro exposures. Therefore, it seems that while other PFFAs function mainly through peroxisome proliferation, PFOS results in additional alterations to gene expression and so may exert its biological effects via other mechanisms of action. There is evidence of cross talk between peroxisome proliferator and lipid metabolism pathways, however the exact mechanism for this cross talk is unclear (Duplus et al., 2000). While there is evidence that free fatty acids are capable of altering gene expression, the mechanism by which fatty acids can act as signaling molecules is unknown (Duplus et al., 2000). It has been suggested that liver X receptors (LXR) are involved in regulation of both fatty acid and sterol metabolism (Tobin et al., 2002). Indeed the fact that LXR is responsive to fatty acids could provide a clear 57 mechanism for the cross talk observed between cholesterol and fatty acid metabolism (Duplus et al., 2000). The implication of the LXR receptor in the mode of action of PFOS would provide plausible explanation for the hypocholesterolaemic effects observed in primates chronically exposed to PF OS (Seacat et al., 2002a). The possibility of PFOS acting on more than one metabolic or regulatory pathway is plausible because commercial mixtures of PFOS contain both straight chain and branched chain homologues. There are several possible ways that PFOS could alter peroxisome function. The simplest explanation could be that, due to the structural similarity of PFOS to endogenous fatty acids, PFOS could be mistaken by the fatty acid metabolism machinery as a substrate. However, due to the non-degradable nature of this compound, the B-oxidation process would fail to oxidize PF OS. To compensate, the major enzymes involved in this pathway could be induced. However, this hypothesis does not explain the lack of increase in the more energetically important mitochondrial pathway, which provides most of the cell’s energy. Another possible explanation of the effects of PFOS on peroxisomal fitnction is that PFOS alters peroxisomal membrane permeability in a way that allows fatty acid influx, requiring greater activity of the oxidation enzymes. This mechanism of action may be less relevant to the mitochondrial pathway since fatty acid entry into mitochondria is a three-step enzymatic transport process. It is also possible that the increase in peroxisomal metabolism is a response to partial uncoupling of the mitochondrial membrane potential resulting in an increase in energy 58 production from peroxisomes. The fact that studies have indicated that PFOS can act as weak protonophoric uncouplers of mitochondrial respiration (Starkov and Wallace, 2002) and is able to alter mitochondrial membrane potential (Hu et al., 2002a) support this hypothesis. Peroxisomal B—oxidation is a process that generates hydrogen peroxide (H202) that can cause oxidative stress and oxidative damage to proteins and DNA. While peroxisome [3- oxidation enzymes were induced up to lO-fold, catalase and glutathione peroxidase, two of the enzymes involved in detoxification of hydrogen peroxide, were relatively unchanged. If catalase or GPX were limiting steps in the removal of peroxide this could result in an increase in hydrogen peroxide, which could induce responses including lipid peroxidation, membrane damage, accumulation of lipofuscin, and DNA damage. The one cytosolic enzyme that was dramatically induced (90 fold) by PFOS treatment was long chain acyl-CoA hydrolase, which cleaves acyl-CoA to free fatty acid and CoA. The counterparts of acyl-CoA hydrolase found in microsomes and mitochondria are carboxylesterase and long chain acyl-CoA thioesterase, in viva these enzymes were induced 5.9 fold and 10.6 fold, respectively. Acyl-CoA hydrolases and related enzymes are important in the regulation of fatty acid metabolism. They have been proposed to maintain CoASH pools for both oxidation and synthesis of fatty acids and to regulate the B-oxidation of fatty acids by controlling the level of acyl-CoA. Induction of acyl-CoA hydrolase would increase cytosolic free fatty acid concentrations. Rodents and primates, when exposed to PFOS exhibit hepatocellular hypertrophy and lipid vacuolation, which 59 could be caused by accumulation of free fatty acids (Seacat et al., 2003). Another significant finding from those studies was a lowered serum total cholesterol level. Cholesterol production is controlled by HMG CoA reductase, the expression of which was reduced 2.5-fold in the current gene expression study. This is consistent with previous studies that suggest that the hypolipemic effect of PFOS may, at least partly, be mediated via a common mechanism; impaired production of lipoprotein particles due to reduced synthesis and esterification of cholesterol together with enhanced oxidation of fatty acids in the liver (Haughom and Spydevold, 1992). The effects on peroxisome fatty acid B-oxidation does not seem to be receptor mediated, since PPAR 0t mRNA expression was not affected. This is consistent with previous studies conducted in our laboratory investigating alterations in expression of PPARor and y in PFOS exposed fat head minnows (Celius et al., unpublished results). Another observation that supports this hypothesis is that even though PFOS has been classified as a peroxisome proliferator, it did not induce P450 4A1, as did most peroxisome proliferators (Figure 2.7). Two groups of P450s that were up-regulated by PFOS exposure were the P450 28 and P450 3A families. Both P450 2B1 and P450 232 were significantly (p<0.0025) induced (9-fold and 22-fold respectively) by exposure to PFOS. These two cytochrome P450 enzymes are phenobarbital inducible, which is a response mediated by the constitutive androstane receptor (CAR). It is known that cytochrome P450 gene expressions are regulated at gene transcriptional level and mRNA processing and stabilization stages, which are mediated through various nuclear factors and co- regulators. Each family of the major CYP genes is under control of distinct nuclear factor 60 pathway, however, cross-talk does occur between different pathways through substrate or co-factor sharing. Therefore the induction of CYP ZB family by PFOS could be explained as cross-talk between CAR and PPAR mediated pathways. In fact, one classic peroxisome proliferator, Clofibrate, was found to induce both CYP 4A1 and CYP 2B1 genes. Gene expression data is useful in identifying affected pathways and possible mechanism of action, but should not be used to develop dose-response relationship. Furthermore the degree of toxicity can not be inferred from these results. Once “significant” genes or pathways have been identified, changes in more toxicologically relevant parameters such as proteins or substrate should be measured subsequently. Also the comparison between in vitro and in viva results indicates that while in vitra studies can be used to focus on response of specific pathway to compounds such as PFOS, the in vitro system is not a substitute of in viva studies. The greatest utility of the in vitra studies is to determine the effect of different structured compounds on a specific pathway and their responses once the critical pathway has been determined. 61 Table 2.1. List of genes induced significantly (p<0.0025) by PFOS in viva exposure. Long-term values are the mean fold-change and standard deviation (s.d.) for the four possible control/exposed comparisons. Gene ID Long-term Long-term Short- Mean s.d. term cytochrome P450 2B15 gene 7.10 0.16 2.6 P450 6beta-2 6.15 0.79 -3.1 cytochrome p-450e 22.57 6.33 6.5 P-450(l) variant 9.09 2.72 5.5 cytochrome P-450b 21.55 5.03 6.7 acyl-COA hydrolase 90.25 36.80 1.7 carboxylesterase precursor 5.89 0.44 - l .l mitochondrial acyl-CoA thioesterase 10.61 3.88 20.1 delta2-enoyl-COA isomerase 6.02 1.25 2.5 stearyl-COA desaturase 2 mRNA 12.88 2.43 5.3 peroxisomal 3-ketoacyl-COA thiolase 9.78 2.05 1.1 peroxisomal enoyl-CoA-hydrotase/3 -hydroxyacyl- 6.5 1.05 2.3 CoA dehydrogenase bifunctional protein peroxisomal enoyl hydratase-like protein (PXEL) 5.11 0.34 2.7 aldehyde dehydrogenase (ALDH) 6.04 0.53 1 17-alpha-hydoxylase cytochrome P-450 19.30 0.36 -2.5 neuroendocrine-specific protein (RESP18) 6.44 0.99 2.6 androgen binding protein (ABP) 12.49 0.57 2.6 Tsx gene 3.50 0.63 2.9 testosterone 6-beta-hydroxylase (CYP3A1) 5.19 0.29 1.4 multidrug resistance-associated protein 5.50 2.22 1.6 DNA polymerase alpha 13.54 0.60 2.2 G-protein coupled receptor RAlc 3.96 0.45 2.8 cytochrome P450 PCNl, NADPH mono-oxygenase 7.04 1.93 17.1 62 Table 2.2. List of genes suppressed significantly (p<0.0025) by PFOS in vivo exposure. Long-term values are the mean fold-change and standard deviation (s.d.) for the four possible control/exposed comparisons. Gene Name Long-Term Long Term Short- Mean s.d. Term B regulatory subunit of protein phosphatase 2A -5.81 0.79 -l .9 Ca++ independent phospholipase A2 -2.45 0.23 -3 protein tyrosine phosphatase -5.76 2.47 -2.7 postsynaptic protein CRIPT mRNA -3.26 0.46 -1.2 tyrosine kinase p72 -2.79 0.33 -3 phosphorylase kinase catalytic subunit -3.37 0.42 -2.8 rat CELF mRNA -O.51 2.03 -4.6 Na+/K+ ATPase alpha2 subunit -11.14 4.69 -2 liver Na+/Cl- betaine/GABA transporter -3.25 0.13 -1.5 R3109 (brain specific gene) -1.83 1.06 -4.1 synaphin 2 -0.16 2.61 -5.5 skeletal muscle selenoprotein W (SelW) -3. 12 0.52 -1.6 apolipoprotein A-IV mRNA -5.90 3.11 -1.2 peripherin mRNA -2.65 1.79 -2.3 cholesterol 7-alpha-hydroxylase -1 .83 0.97 -4 peptidylarginine deiminase type 111 -1.76 0.83 -11.9 mRNA for RT] 975 4.11 -1.1 MHC class II A-beta RTl -16.39 3.97 -2.8 DNA binding protein (GATA-GT2) -5.03 1.41 -1.2 63 Figure 2.1 A Control 1 l llllll PFOS l .4. + + 1 2 + * + q d + .. + it: 1 4"” *4- -1 + I + + ++ 1 + + q + t+ 4- -I + .2“. + + E + 'I'+ I ‘0’ 4+4. 4 + + + r + .. + + + + ++ + ++- + 1 1 .1 I V V 1 "fi' ' ' 'Y'7 W V V ' "Trr‘ Y Y I ' ""1 I' 1 ' MMTj‘ Control 2 II # + +1- 5 4' + ? + a + -1 + at 4. g + + +"*' -4 .h- i ‘ + 3.7.. +:' .. + + W + + ++ ‘t .I fi+ ++ 1 + + * d + + 1 + + + + ~ + 2 1 + "' + + a i 1 111111 PFOS 2 64 Figure 2.1 v vvv“ v vvvwv' v v v v rvrvrw' fir v vvvtvv' vvv-I vvvv v v vvvvvv' vrv v rvvvv‘ I PFOS] Contro 1 ‘_ MA... ' vvv I I'VVV‘I A J_JLLAA-- - 'v'V“ V "Y' 1 V D In Vivo Control In Vitro-Control 65 Figure 2.2 Grouped by gene expression pattern H in vitro control III-I e IIIIIIIIIIIIIIII I-IIII in vitro PFOS at2 mg/L - IIIIIIIII IIII III—I II “WWI 5:1 . §_ 5' ‘ I; I I: II iEHENE ”H invivo long-term control 0 , I . I If; .1 III III" IIIIIIIIII III —o I -~ IIIIIIII IIIIIII IIIIIIII . I” III IIIIIIIII IIIIIII III IIIIIIIIII I I I t {I I H“ “U in vivo short-term control vi 1. II ,. III III II I ,1 "i t I IIIIIIIII III I ‘I I IIIIIIII I invasiontampms 66 Figure 2.3 Grouped by gene expression pattern long-term controll long-term controlZ g I ll long-term PFOSl “O : I ~ 0 .E‘, _'i' . ;' I 5 - II : i I i I III WEI I E . I I short-term PFOS __ . I . . - I I: - .I . ' 67 I7ignlre:2254 A honnone regulator lipid metabolism V 4 CYPs V 41 cytochrome P450 PCN1. NADPH mono- _ i oxygenase DNA polymerase alpha multidrug resistance-associated protein testosterone 6-beta-hydroxylase (CYP3A1 ) androgen binding protein (ABP) neuroendocrine-specific protein (RESP18) y l l l 17-alpha-hydoxylase cytochrome P450 é . Short-term { I D long- -term : aldehyde dehydrogenase (ALDH) 1;. ' ‘A ' peroxisomal enoyl hydratase-like protein b 3 , (PXEL) i peroxisomal 3oketoacyl-CoAthiolase L'FH ? ; l r stearyl- -CoA desaturase 2 mRNA detta2-enoy4~CoAisomerase mitochondrial acyt-CoAthioesterase carboxylesterase precursor acyl-CoAhydrolase .fi cytochrome P-4SOb _= P-4SO(1 ) variant -_ 1; cytochrome p-4SOe _.._. l : P450 6beta-2 I I cytochrome P45028159ene E , i 0 30 60 90 68 Figure 2.5 l ll short-term l D “9949'? ‘ ___ l 9— k rLIH-TLrlrll minim .i‘i. i1 1 r DNA binding protein (GATA-GT2) MHC class II A—beta RT1 mRNA for RT1 peptidylarginine deiminase type III cholesterol 7-alpha—hydroxylase peripherin mRNA apolipoprotein A-IV mRNA skeletal muscle selenoprotein W (SelW) synaphin 2 A R8109 brain s ific ene ( pee g ) neurosysten Ii\er Na+/Cl- betaine/GABA transporter regulation mRNA Na+lK+ ATPase alpha2 subunit V rat CELF mRNA phosphorylase kinase catalytic subunit tyrosine kinase p72 postsynaptic protein CRIPT mRNA signal transduction protein tyrosine phosphatase Ca++ independent phospholipase A2 8 regulatory subunit of protein phosphatase 2A * 69 Figure 2.6 fatty acid CoASH, ATP, Mg2+ fatty acyl-CoA synthetase . D9 0 I 09 AMP+PP1 (I .5 fold) acyl-CoA membrane fatty acid CoASH, fatty acyl-CoA ATP, Mg2+ synthetase D90109 (1 .5 fold) AMP+PPi acyl-CoA outer memm Carnitine { camitine acyltransferase I D43623 (1 fold) é. CoA-SH i acyl-CoA FAD H acyI-CoA oxidase J02 753 (2 fold) FADH2 O enoyl-CoA enoyl- C 0A hydratas K03249 (6.5 fold) hydroxyacyl-COA hydroxyacyl—COA NAD+ dehydrogenase K03249 (6.5 fold) NADH ketoacyl-COA 3-Ket0acyl—COA T hiolase J02 749 (1 0 fold) acetyl-CoA + acyl-CoA shortened by 2 carbons Peroxisomal Fatty Acid B-Oxidation 7O Fatty acyl-carnitine inner membrane Carnitine r COA'SH acyltransferase II 202 J054 70 ( I .2 fold) b‘ camitine acyl-CoA 2 acyI-CoA FAD dehydrogenases M35826 (1.1f01d) FADH? ATP enoyl-CoA enoyl-CoA hydratase D16478 (I .2 fold) hydroxyacyl-CoA h - ydroxyacyl CoA NAD+ dehydrogenase DI 64 78 (1.2 fold) NADH ket acyl-CoA 3 -Ket0acyl- C 0A T hiolase D164 79 (1.5 fold) acetyl-CoA + acyl-CoA shortened by 2 carbons Mitochondrial Fatty Acid B-Oxidation Figure 2.7 0 W 2 $0/ v.0 9045 71 CHAPTER THREE Hu, W.Y., Jones, P.D., DeCoen, W., and Giesy, J.P. (2003b) Comparison of gene expression methods to identify genes responsive to perfluorooctane sulfonic acid. Environmental Toxicology and Pharmacology (Submitted) 72 ABSTRACT Genome-wide expression techniques are being increasingly used to assess the toxic effects of environmental contaminants. Oligonucleotide or cDNA microarray methods make possible the screening of large numbers of known sequences for a given species, while differential display analysis makes possible analysis of the expression of all the genes from any species. We report a comparison of two currently popular methods for genome-wide expression analysis in rat hepatoma cells treated with perfluorooctane sulfonic acid. The two analyses provided 'complimentary’ information. Approximately 5% of the 8800 genes analyzed by the GeneChip array, were altered by a factor of three or greater. Differential display results were more difficult to interpret since multiple gene products were present in most gel bands. A probabilistic approach was required in this analysis. The mechanistic interpretation derived from these two methods was in agreement, both showed similar alterations in a specific set of genes. Keywords: PFOS, Gene expression, Differential display, GeneChip array 73 INTRODUCTION A variety of techniques have been developed to examine alterations in gene expression as a result of exposure to chemical agents or other stressors (Higuchi et al., 2003; Mong et al., 2002; Matsuba et al., 1998). Of particular significance are those techniques which allow examination of the gene modulation in the entire genome. These techniques represent new approaches in predictive toxicology and allow identification of unknown modes of action, screening for potential toxicity and grouping of chemicals on the basis of the mode of action. Each of the techniques used has specific advantages and disadvantages and, more significantly, produces information of a different nature. Differential display, first introduced by Liang and Pardee (1992), is a useful tool that permits the identification of numbers of differentially expressed genes at the whole genome level. The basic principle of differential display is the separation of total mRNA into many subsets of approximately four hundred gene products each produced by choosing appropriate primer pairs for reverse transcriptional polymerase chain reaction (RT-PCR). Each of the subsets can then be ‘displayed’ using a denaturing polyacrylamide gel. Differential display is an mRNA “finger-printing” technique that facilitates the identification of altered functions resulting from changes in mRNA transcription and/or degradation rates. Differential display has been successfully employed by many research groups to compare the gene expression patterns in different organisms and tissues, collected at different developmental stages, in normal and diseased states, in exposed and unexposed individuals or under different in vitro cell culture conditions (Liang and Pardee, 1992; Zhang et al., 1993, Douglass et al., 1995; 74 Green et al., 1996; Gao, 1998). In the field of environmental toxicology, differential display is becoming a useful and powerful too], especially when the mode of action of a stressor is unknown. However, the original design of the differential display technique is not without limitations. Specifically the technique is labor intensive, time consuming and preferentially amplifies the 3’-ends of transcripts. Therefore, a modified version of the original technique which is termed restriction fragment differential display PCR (RFDD- PCR) was developed (www.displaysystems.com). Instead of directly amplifying the first-strand cDNA, the RF DD-PCR approach adds specially designed adaptors to the ends of cDNA fragments obtained from Tan digestion. The subsequent PCR amplification uses a series of designed arbitrary primers selectively annealed to the adaptor junctions. This approach avoids the problem of 3’-end bias and limits the number of primers required to screen the complete genome of eukaryotic organisms to 64 pairs (Liang & Pardee, 1992; Liang et al., 1994; Guimaraes et al., 1995; Liang, 1998). Differential display works via the systematic amplification of selected sub-populations of mRNA using arbitrary primers and resolution and visualization of those fragments on denaturing polyacrylamide sequencing gel. It thus allows for side-by—side comparison of potentially all expressed genes in a systematic and sequence-dependent manner among related cells. Since most cellular processes and responses to toxicants are driven by the temporal and spatial expression of mRN A, differential display is a very useful technique in examining and comparing mRNA expression profiles under given conditions or 75 treatments. Due to this unique feature, differential display has been utilized in a wide range of applications including developmental biology, cancer research, neuroscience, endocrinology and many other fields, including toxicological studies (Higuchi et al., 2003; Mong etal., 2002; Matsuba et al., 1998). More recently, methods of screening gene expression based on DNA microarrays have been developed. In these methods, target cDNA sequences or oligonucleotides are spotted or synthesized in situ onto a glass slide or ‘array’. Fluorescently labeled cDNA fragments generated from control and exposed tissues or organisms are hybridized to the array and differences in gene expression are visualized as differences in the fluorescent signal intensity of the specific target ‘spots’ or ‘features’. The automation of array production, hybridization and analysis has lead to the development of arrays with in excess of 100,000 discrete features per slide. Similarly, automation and analysis software have greatly reduced the complexity of the data produced. With the use of specific arrays, it is a relatively simple and fast procedure to assess the expression of over 10,000 genes with considerable statistical rigor. While the speed and accuracy of microarray methods is generally unquestioned, these methods are limited to the analysis of gene expression in the specific species for which the array was designed. In contrast, the differential display technique can be used on any species in which the investigator is interested. However, the process of analyzing all possible gene transcripts on polyacrylamide gels is labor intensive. In addition, many of 76 the bands isolated from the gels contain multiple gene products making unequivocal identification of the altered genes difficult and additionally time consuming. Of considerable significance to interpretation of data derived from these two methods is the question of data comparability. Specifically how does sequence specific gene array data compare to data generated from the entire genome by differential display given that the ‘pools’ of gene products analyzed are different and the analytical and statistical methods applied generate distinctly different data. In this study we compared the two gene expression techniques to determine the degree of ‘comparability’ or ‘agreement’ when the same mRN A samples were analyzed by the two methods. MATERIALS AND METHODS Chemicals Perfluorooctane sulfonic acid (PFOS) used in the in vitro experiments was obtained from 3M (St. Paul, MN). The PFOS (potassium salt) used for in vivo experiments was purchased from Fluka Chemicals (Switzerland), chemical analysis revealed that it was essentially the same as the product obtained from 3M. Cell Culture and Treatment H4IIE rat hepatoma cells were cultured in 100 mm disposable tissue culture dishes at 37°C under sterile conditions (pH=7.4) in a humidified 5/95% C02/air incubator (Forma Scientific, Model 8173). Cells were grown in Dulbecco’s Modified Eagle Medium (DMEM, Sigma D-2902, Sigma, St. Louis MO), supplemented with 10% fetal bovine 77 serum (FBS, Hyclone, Logan, UT). At confluence, cells were removed from the dish with trypsin/EDTA (Hyclone, Logan, UT), then split into four tissue culture plates. Twenty four hours after splitting, cells were dosed with PFOS to achieve final concentrations of 2 mg/L and 50 mg/L, methanol was used as solvent control, and the blank control received no dose. Cells were incubated for 72 hr after exposure. RNA extraction and purification Total RNA from cell culture samples was extracted using TriPure Isolation Reagent (Boehringer Ingelheim, Germany) using the manufacturers recommended procedures. The optical densities of RNA samples were measured at 260 nm and 280 nm on spectrophotometer, and the 260/280 ratio was calculated. RNA concentration was quantified using optical density at 260 nm. The quality of RNA was evaluated by the appearance of distinct 185 and 28s ribosomal RNA bands on 1% agarose gels. Differential Display Procedure Differential display was conducted using reagents and procedures from the Display System Profile Kit (Buena Vista, CA). Total RNA was used as a template for cDNA synthesis using reverse Transcriptase and an oligo-dT primer. After synthesis from total RNA, double-stranded cDNA was digested with Taq I restriction enzyme, which is a 4- base-cutter that leaves a 5'-overhanging end. Following digestion, two different specifically constructed DNA adaptors are ligated to the ends of the cDNA fragments. One of the adaptors, the EP adaptor, has a 5'-overhang and an "extension-protection group" on the 3'—end, which prevents 3' to 5' synthesis filling in the overhang. Each 78 reaction uses a labeled O-extension 5'-primer that anneals to the ligated EP adaptor, and a specific displayPROBE 3'-primer that anneals to the junction between the standard adaptor and the cDNA insert. The extension-protection group on the EP adaptor prevents amplification of cDNA fragments that have EP adaptors on both ends. Three bases of the displayPROBE primer extend into the cDNA sequence. It is these three bases that make the displayPROBE specific for certain cDNA sequences, and also, prevents amplification of cDNA with standard adaptors on both ends since both ends would need to have the same three bases to be amplified. To amplify all variations, 43 or 64 different display probe primers are required, each for a separate PCR reaction that amplifies a different set of cDNA fragments. For less complex prokaryotic cDNA, the 64 display probe primers were combined in pairs reducing the number of reactions to 32. Each PCR reaction amplifies 400 or more fragments, which is referred to as an "expression window." The 64 reactions, used for a eukaryotic sample, produce approximately 25,000 distinct cDNA fragments. Because of the design, each amplified fragment should be represented in two different expression windows in the RFDD-PCR analysis (Display Systems manual 2.1, 1999). RF DD-PCR was conducted using procedures recommended by the Manufacturer (Display Systems Biotech Inc., Buena Vista CA). After amplification and labeling with 33F, fragments were separated on polyacrylamide sequencing gels. After gel drying, amplicons were visualized by autoradiography at -80°C over night. Differentially displayed bands were identified, excised from the gel by overlaying the film on the gel, 79 and DNA was extracted by placing the gel slice in buffer overnight. DNA was then amplified by PCR with the same primers used for the initial amplification. Subcloning of PA GE Bands Direct sequencing of the PCR products extracted from gels was attempted but demonstrated the presence of more than one amplicon sequence in each gel band. Cloning of target genes was conducted using pGEM-T easy vector system (Promega, Madison, WI) with the T-A cloning technique. PCR products were ligated into plasmids and then transformed into E. coli JM109 competent cells. Positively transformed cells were selected via blue-white screening, 6 colonies were picked from each plate. Plasmids were purified using Wizard plus SV minipreps from Promega (Madison, WI). DNA sequencing was conducted using dideoxynucleotide labeling technique at the Michigan State University Macromolecular Structure Facility. The sequences obtained were used to interrogate the Gene Bank database (http://www.ncbi.nlm.nih.gov/) using the multiple sequence alignment BLOSUM 62 BLAST program (Altschul et al. 1997). GeneArray Analysis The RNA samples described above were also analyzed by GeneChip Array technology. The Affymetrix rat genome U34A gene chip array was purchased from Affymetrix Inc. (Santa Clara CA). The oligonucleotide probes on U34A array cover approximately 8800 known genes and Expressed Sequence Tags (ESTS) in the rat genome. Transcripts were selected from Genebank Unigene build 34 and the dBEST database. The methods and 80 results of that study are reported extensively elsewhere (Hu et al 2003a) and will not be discussed here. RESULTS Identify and Subclone Diflerentiallv Expressed Genes To determine gene expression changes associated with PFOS exposure, H4IIE cells treated with PFOS at 2 or 50 mg/L were compared with untreated cells and solvent control treated cells. The differential display technique was able to identify distinct gel bands, which represented differentially expressed genes (Figure 3.1). Gene products that were either induced or inhibited by exposure to PFOS could be identified as increase or decrease of gel band intensities. RT-PCR differential display was conducted, in duplicate, using 32 of the possible 64 primer combinations from the Display Systems kit. Theoretically, since each primer set covers approximately 400 amplicons in its expression window, 32 primer sets should result in a sum of 12,500 mRNA fragments. Since less than 11,000 mRNAs from the rat genome have been sequenced and functionally annotated, the 32 primer sets should be able to give a fairly broad, though not complete, coverage of the rat genome. The number of genes analyzed is comparable to the number analyzed by Gene Chip analysis (Hu et al., 2003a). After separation of the reaction products on sequencing gels and examining the gel band intensities carefully, 55 amplicons were identified that were altered consistently in duplicates based on comparison between the controls and the treated groups. Of these 55 amplicons, 34 appeared to be affected in a completely present/absent fashion (the bands 81 appeared only in control or treatment group, but not both). The remaining 21 amplicons were partially increased or decreased (the bands were present in all treatment groups but with different intensities). The 55 bands were excised from the sequencing gels and re-amplified using the short arbitrary primers used in the original amplification step. Direct sequencing of the PCR products was attempted, but was not successful due to the short length of the amplicons and the presence of multiple sequences in each band. Therefore, each amplicon was sub- cloned into a plasmid vector which was then used to transform E. coli host cells. After sub-cloning amplicon DNAs, 29 of the original gel bands were able to be prepared in sufficient quality and quantity to permit direct sequencing. Six bacterial colonies were selected from the transformed E. coli hosts containing plasmids from each gel band, and the inserted DNAs were sequenced yielding 154 sequences suitable for sequence comparison analysis. There were generally two or more different sequences present in each gel band (Table 3.1). Clones where 6 out of six colonies contained the same DNA insert were considered to be unambiguously identified. Clones for which 4 or 5 colonies contained the same amplicon were considered to be identified with relatively great certainty (Table 3.1). After sequencing the putatively differentially expressed amplicons, the sequences were used to search the Gene Bank database with the BLAST algorithm (Altschul et al., 1997). The searches were first conducted against all Rattus norvegicus sequences since the 82 exposures were conducted with a rat cell line. If the initial searches did not match known rat sequences searches were conducted against all mammalian sequences in the data base. Of the 154 sequences searched, 120 matched rat sequences, 13 matched mouse sequences and 1 matched a human sequence. Thirteen sequences did not match any known mammalian sequences in the database. As expected, sequence similarity ratings were highest for comparison to rat sequences and were lower for comparisons to mouse and human sequences. Of the six clones isolated from band 5_8, none showed homology to any known mammalian sequence although all of the six clones contained the same 89 bp amplicon. When compared to the entire Gene Bank database, the highest degree of homology for the 5_8 band sequence was to a plant ferredoxin gene but that homology only extended over a 23 bp region. Since the cells used in this study were of rat origin, this weak homology probably represented a random coincidence. Of the original 29 gel bands 12 contained a clear ‘majority sequence’ as indicated by greater than 50% of the sequenced clones representing one gene product (Table 3.1). Comparison with GeneChip Results The results of a GeneChip analysis of the same samples used in the current analysis have been previously reported (Hu et al., 2003a). Due to the different biases inherent in the two gene expression profiling methods a comparison of results from the two methods was conducted. To permit a numerical comparison with the fold induction values determined in the GeneChip analysis, each gene altered in the differential display data set was assigned a score ranging from -2 to 2 based on the degree of inhibition (-2 = strong inhibition of the band, -1 = moderate decrease in band intensity) or induction (2 = strong 83 induction of band, 1 = moderate increase in band intensity) under the different treatment conditions. There was little correspondence in the estimated degree of alteration of expression determined by the two methods (Table 3.2). This is not surprising as the differential display analysis and band selection are based on the subjective assessment of the intensity of the specific gel bands. The GeneChip analysis reported that the expression of over 400 genes, from the 8790 genes and ESTs on the array, were significantly altered by treatment with PFOS (Hu et al., 2003a). Of the genes whose expression was altered 161 increased at 2 mg/L PFOS; 74 decreased at 2 mg/L PFOS; 76 increased at 50 mg/L PFOS and 99 decreased at 50 mg/L PF OS. Of the gene whose expression increased 32 were induced in both treatments while only 5 were identified as being decreased at both treatment concentrations. Concordance of results between the two treatment concentrations was higher for the differential display results. This is not surprising as a common response for the two adjacent samples would be more likely to be visually identified as a definite alteration in gene expression when differentially expressed gel bands were being identified. DISCUSSIONS PF OS has been shown to alter a variety of properties of the biological membranes (Hu et al., 2002a, 2002b). It is therefore of interest that the differential display method demonstrated alterations in the expression of several genes related to membrane structure and function. Specifically, differential display indicated a decrease in the expression of 84 desmoplakin rabin 3, profilin and guanine nucleotide exchange factor. Desmoplakin are important proteins in desmosomes which serve as intercellular junctions and attachment sites for intermediate filaments (Meng et al., 1997). They are thus important in the maintenance of the intercellular ‘skeleton’ of tissues. Rabin3 is an inhibitor of Rab3A, a small Ras-like GTPase expressed in neuroendocrine cells where it is associated with secretory vesicle membranes and controls exocytosis (Brondyk et al., 1995). Although Rab3A appears to be limited to neuroendocrine cells Rabin3 is expressed in a wide range of tissues (Brondyk et al., 1995) where it presumably interacts with proteins whose fimction is homologous to Rab3A. Guanine nucleotide exchange factors are also involved in the formation of the cell skeleton and in membrane trafficking in various ways particularly in regulating the activity of Rab and Ras type proteins (Ganesan et al., 1999) While PFOS does have many effects at the level of the cell membrane it has also been demonstrated to alter the expression of a number of genes involved in lipid metabolism (Hu et al., 2003a) and has been shown to alter the lipid status in rodents and primates in vivo (Seacat et al., 2002a, 2003). Genechip analysis of the same samples used in the current study indicated that PFOS specifically induced the peroxisomal fatty acid oxidation pathway but not the same enzyme systems in the mitochondria (Hu et al., 2003a). Interestingly, the differential display and gene chip analyses demonstrated a decrease in the expression of complex I NADqubiquinone oxidoreductase. This complex is the first membrane bound electron transport complex of the mitochondrial respiratory chain and so accounts for up to 40% of the proton-translocating capacity of 85 the respiratory chain. Loss of activity in this proton-translocating complex could result in a need to increase peroxisomal fatty acid oxidation as observed or, alternatively, increased peroxisomal fatty acid oxidation could result in a lower energy demand on the mitochondria and so result in a down regulation of the mitochondrial electron transport chain. Complex I has been shown to be susceptible to hydrophobic inhibitors (Okun er al., 1999) and the highly electronegative nature of the PFOS molecule suggests that it would have an propensity to modulate electron transport and translocation. Interference with complex I could also explain the observation that PFOS is a weak non-ionophoric mitochondrial uncoupler (Starkov and Wallace 2002). Both methods of expression analysis used have inherent biases. For example, the need to visually identify gel bands whose intensity is different across treatments biases the differential display data set towards those genes whose expression is altered the greatest. In contrast the Genechip data analysis treats all gene expression levels equally. However, the genes represented on the GeneChip are strictly limited to those which have been isolated and identified and sequenced by other researchers. In contrast the differential display method is capable of identifying any gene product whose expression is altered. Therefore the data sets generated by these two methods tend to be diametrically opposed. One data set (GeneChip) is biased towards sequences of toxicological interest while the other (differential display) attempts to identify every possible gene but is mechanistically biased towards the greatest alterations in expression level. 86 The differential display method relies on the ability to separate and visually identify gel bands whose intensity varied with different treatments. As such this procedure is open to interpretation by the individual investigator. Therefore the critical step in this process is one of observation by the individual conducting the analysis. As such, this method is limited by the resolving power of the PAGE system used and by ability of the human eye to detect ‘significant’ changes in expression profiles. After determination and excision of bands which are differentially expressed, a series of procedures are required to unequivocally determine which of the possible multiple cDNAs present in the band is the gene product which is differentially expressed. This is achieved by amplifying and subcloning the sequences in the band such that only a single sequence is inserted into each bacterial host. In the simplest case where all bacterial cells contain the same insert we can assume that the original gel band contains only a single amplicon. In those cases where the majority of the amplicons represent one gene product it can also be safely assumed that the most abundant amplicon represents the differentially expressed gene. In cases where several amplicons are present in similar proportions it is not possible to identify the gene product which is differentially expressed. In summary the differential display method is labor intensive and requires application of considerable professional judgment. This makes the method less than ideal for rapid screening and assessment of large numbers of stressors. However, the method is of use in determining critical modes of action and is an ‘open platform’ method suitable for use in species and tissues where significant DNA sequence information is not available. 87 The differential display method was able to detect alterations in expression of genes not present on the microarray used. In some instances, for example desmoplakin, the differential display method also allowed a tentative identification of the function of the product as it was homologous to genes in other species. In the case of one gene identified by differential display, the sequence was not homologous to any known mammalian genes, this product would be an obvious candidate for further investigation to determine the nature and function of the gene product. In contrast to the differential display method, the gene array technique provides a statistically robust identification of each amplicon and the degree to which it is altered relative to the control conditions. Essentially all the amplicon identification procedures are carried out during the preparation of the array. Based on the quality of the data including the sensitivity, specificity and reproducibility of the results, gene chip arrays appear to provide a greater wealth of information than differential display (Yuen et al., 2002). Because of the current need for visual detection of differences in gel band patterns differential display is limited to only abundantly expressed messenger RNAs. Theoretically, separation of RNA fragments on a well-cast polyacrylamide sequencing gel can identify one single nucleotide difference, in practice, however, this is rarely the case. The separation of gene fragments is limited by several factors, including the quality of the denatured polyacrylamide gel and the means of detection. In this study we used 33P—labeled NTP to assist visualization, and differentially expressed genes were identified by visual examination. Therefore, the results are not 88 quantitative. In contrast, the gene chip technique utilizes fluorescent dye labeling and computer image analysis, which makes gene chip results specific and quantitative. Another limitation of differential display method as discussed previously is the relatively great incidence of false positives. The ambiguity in determining the gene identities using similarity search of sequence databases add another level of complication. Whereas, the gene chip array has well-defined gene identities, and using the specially designed perfect/miss match strategy serving as internal control allows for unambiguous identification of significant alterations. Even with these limitations, over-all there was still a general agreement between the results obtained by use of the differential display and gene chip methods. It is somewhat naive to assume that the two techniques would provide identical information. Rather they provide complimentary information, this explains the difficulty encountered in trying to obtain a direct comparison of the results. The gene chip technology provides precise definition of relatively small changes in gene expression in a biased sample of genes present in the genome. In contrast the visual identification in the differential display analysis provides evidence of large changes in specific gene products from the entire genome but gene identification is equivocal and relies on probabilistic approaches to identification. It is probably most significant that this comparison of the two techniques provides evidence of a general agreement in mechanistic interpretation rather than a direct comparison. In general genes whose expression was apparently altered in one technique was similarly altered in the other. Both gene expression techniques clearly deliver a wealth of information. In the case of the gene array the amplification process and data reduction software delivers higher 89 'quality’ information. In essence a great deal of the probabilistic interpretation of the data is done before the data reaches the researcher. In contrast the differential display method requires that the researcher filters data to remove false positives and interpret sequence matches to identify genes of interest. In the gene array technique the gene identity is known exactly for each probe set on the array. These basic differences in the two methodologies result in data that should be seen as complimentary. As to economic and practical consideration, differential display is less expensive than gene chips. All the equipment and reagents needed for differential display technique can be easily obtained in most molecular biology laboratories. On the other hand, gene chips are expensive themselves and the technique requires a specially designed work station, and scanner, furthermore the reagents used for labeling cRNA and the software used for analyzing the results are also expensive. However, when we look at the cost efficiency aspects, differential display is relatively more time and labor intensive. The choice of which technique is more appropriate will be based on considerations of the research question at hand. 90 Table 3.1. Genes identified to be differentially expressed due to PFOS exposure in H4IIE cells in vitro. “Clone seqs” indicates the number of clones sequenced from each original gel band. The “majority sequence”, if present, is the sequence that was present in 50% or more of the sequenced clones. The “Proportion Majority” is the number of clones out of the total that were the majority sequence. Gel clone majority Proportion Name Band seqs sequence Majority l_l 6 None l_10 3 None l_ll 6 None l_12 6 NM_017313 6/6 rabin 3 (RABIN3) l_2 7 XM_225259 4/7 similar to (Desmoplakin I (DPI)) l_3 8 None Mk1 protein (Mk1), mRNA l_4 6 NM_134399 3/6 (homologous to profilin) similar to RIKEN cDNA l_5 5 XM_213242 4/5 0610011B04 Ribosomal DNA external l_7 6 X16321 4/6 transcribed spacer 1 (ETSI) 4_2 6 None 4_3 3 None 5_1 3 None mitogen-activated protein kinase- 5_lO 6 AY197741 3/6 activated protein kinase2 5_11 6 None 5_12 3 None 5_13 10 None 5_2 3 None 5_3 6 None 5_4 6 None 5_5 6 None 5_7 7 NM_080907 5/7 protein phosphatase 4 5_8 6 No mammalian equivalent 5_9 6 AF 476964 5/6 Transferring-like mRNA 6_2 5 None 6_3 6 NM_053556 6/6 maternal G10 transcript (G10 similar to mouse guanine nucleotide- 7_l 4 XM_225548 2/4 exchange factor (LOC307098) 7 ;2 2 NM_017245 2/2 eukaryotic elongation factor 2 7_4 6 V01270 3/6 Ribosomal 188, 5.88, 288 91 Table 3.2. Alterations in gene expression (as fold change from control) assessed by differential display and Genechip analysis. “dd” and “go” refer to differential display or GeneChip analysis at 2 or 50 mg/L as indicated. Band majority sequence (5d (513 gzc g3 Gene l_12 NM_017313 -1 -1 0 -1.2 Rabin3 l_2 XM_225259 0 -l desmoplakin l_4 NM_134399 -l -l -1.1 -1.1 profilin l_5 XM_213242 0 -1 NADqubiquinone oxidoreductase l_7 X16321 O 1 ribosomal ETSl 5_10 AY197741 -l -1 1.1 1.3 protein protein kinase 2 5_7 NM_080907 -1 -1 protein phosphatase 4 5_9 AF47 6964 1 1 1.8 -l .9 transferrin like 6;3 NM_053556 1 2 -1.1 -l.l G10 protein 7_1 XM_225548 0 -1 -1.1 -1.2 guanine nucleotide exchange factor 7_2 NM_017245 0 -l 1.5 1.4 elongation factor 7_4 V01270 -1 -2 1 -l .2 ribosomal genes 92 Figure 3.1 BC SC P2 P50 BC SC P2 P50 93 CONCLUSION The results from gene expression analysis and cell bioassays can be summarized as follows: Overall genome-wide gene expression analysis revealed the modulation of several critical metabolic and regulatory pathways by PFOS exposure. Genes that were significantly up regulated by PFOS were involved in fatty acid metabolism, drug and xenobiotic metabolism, and hormone regulation. Down regulated genes were mostly coding for key factors involved in signal transduction pathways and the regulation of neural system function. Among the pathways regulated by PFOS exposure in the GeneChip experiment, fatty acid B-oxidation appeared to be the major pathway affected. Further interpretation of the data revealed that most key enzymes involved this pathway were consistently and significantly induced in the peroxisomes but not in the mitochondria. GeneChip expression analysis also indicated that distinct patterns of gene regulation were resulted from the in vitro vs. in vivo exposure. However, when gene expression profiles derived from long-term and short-terrn in vivo exposure were compared, a much greater level of consistency in gene regulation was observed. Comparison of differential display and gene chip technique based on PFOS in vitro treatment results indicated that the mechanistic interpretation derived from these two methods was in general agreement, although the list of significantly modulated genes generated from the two methods was more complimentary than directly comparative. A contrast of the advantages and disadvantages with differential display and gene chip was listed in table C. l. 94 0 Results from cell bioassays using model ligands in combination of PFOS indicated that PF OS increased the membrane permeability selectively to those ligands tested. 0 Membrane related bioassays using fluorescent probes and flowcytometer revealed that PFOS treatment significantly increased the membrane fluidity and decreased the mitochondrial membrane potential in a dose dependent fashion. PFHS and PFBS, with shorter carbon chain length, showed no effects on those membrane-associated properties within the concentration range tested. Results from the current study provide useful information on various aspects of the molecular and biochemical toxicity of PFOS. A structure activity relationship can be derived with the evidence provided in this study in combination with previous findings in regarding to PFFAs. Fatty acids with different chain lengths have different cellular functions and are metabolized by different systems in the cell. Fatty acids can be incorporated into cellular membrane phopholipids or can be utilized to synthesize other lipids or steroids. Fatty acids can also be broken down for energy generation. Some fatty acids are known to affect protein kinase pathways, thus modulating cell signaling cascades. Although PFOS has a similar structure with endogenous fatty acids, short chain fatty acids are generally not present in normal adult rat tissues. PFOS is therefore ‘foreign’ to the cell and the sulfonic acid functional group is relatively non-reactive making PFOS metabolically non-degradable. Therefore, PFOS should exhibit membrane- related effects as well as alter fatty acid metabolism. Results fiom the current study support this prediction. PF OS was proven to affect membrane-associated properties, such as membrane permeability, plasma membrane fluidity, mitochondria membrane potential, 95 and previously reported gap junctional intercellular communication. PFOS treatment also up regulated fatty acid B-oxidation, altered hormone and cholesterol synthesis, and down regulated signal transduction pathways. Furthermore, studies conducted on PFFAs with different chain lengths, including PFOS, PFHS and PFBS, indicated at least some of the observed effects were associated with the molecules chain length. Results from this study confirmed and elaborated on the gene expression modulation by PFOS in the rat. The results also supported using toxicogenomic evaluation for the detection of molecular mechanisms of toxicity. One issue of concern with genome wide gene expression analysis is whether it is feasible to extrapolate these mRNA level effects to protein expression and functional responses at the cellular or whole animal level. This has been a point of discussion within the field of toxicogenomics. Predicting protein or cellular level function and toxicity from alterations in gene expression profiles must be done with caution. One should not assume a direct correlation between RNA and protein concentration or enzyme activity without further testing. Gene expression profiles exhibit strong temporal variation, and are complicated with numerous factors, such as genetic background, age, gender, nutrient conditions, as well as social behavior. Furthermore, modulation at the post-translational level, such as protein phosphorylation or changes in protein 3-D conformations, would never be predicted based solely on genomic data. Since changes at the transcriptional level occur rapidly after exposure, and respond sensitively to relative low level treatment, gene expression analysis should be treated as an early-stage screening tool, which can provide hypotheses and predictive models for further investigation. Especially with the high throughput nature of this technology, 96 genome wide gene expression analysis provides a theoretically unlimited source of information, which keeps growing with the increasing amount of genomic sequence annotation. When coupled with other “omics” technology, such as proteomics and metabonomics, genomics technology, including the two examples discussed in this study, will become much more powerful in applications such as predictive toxicology, drug discovery and development, biomarker identification, and risk assessment. Table C 1. Comparison between differential display and genechip techniques Differential display GeneChips 'Open Platform - Very sensitive and reproducible Advantages 0No need for sequence infor. - Gene specific -Not lrmrted by specres - Quantitative -Relatively cheaper - Labor intensive -Close Platform Disadvantages - Time consuming -Less flexible . . - Frequent “false- ~Need sequence rnforrnatron positive” -Only available for several species 0 not quantitative ~More expensive 97 APPENDICES METHODS AND PROTOCOLS 98 A. RNA Extraction and Purification . Cell culture medium was aspirated and cells were washed with 5 ml PBS twice. 1.5 ml tri-reagent was added into each cell culture dish, and allowed to immerse the cells for 3 min. A sterile policeman was used to scrape the cells off the bottom of the plate, and cell suspension was then passed through a pipette tip several times before it was transferred to 2 m1 RNase free Eppendorf tube. Rat liver samples were rinsed with PBS solution, then cut into approximately 100 mg per piece and transferred to a typhoon tube. 1.5 ml of tri-reagent was added into each tube, and the liver samples were homogenized. The tissue homogenate was then transferred to 2 ml RNAse free Eppendorf tube. . The cell and tissue lysates were incubated for 5 min at room temperature to ensure complete dissociation of nucleoprotein complexes. 150 pl of chloroform was added to each tube. The tubes were shaken vigorously for 15 sec and allowed to stand at room temperature for 15 min. . Samples were centrifuged at 12,000 x g for 15 min at 4°C. Three layers were formed in the tube: the upper colorless aqueous phase contains RNA, the interphase and lower red organic phase contain protein and DNA. The upper phase was carefully transferred to a new Eppendorf tube. . 400 pl of isopropanol was added into each tube, and mixed thoroughly by inversion. The tube was incubated for 10 min at —80°C to allow RNA precipitation. . Samples were spun at 12,000 x g for 10 min at 4°C. The supernatant was discarded, and 750 pl 75% Ethanol was added in the tube, and the pellet was washed by vortexing in Ethanol. Samples were then centrifuged at 7500 x g for 5 min at 4°C, 99 and the supernatant was discarded. Excess ethanol was removed fi'om the RNA pellet under vacuum. . The RNA pellet was re-suspended with 20 pl DEPC treated water, and dissolved completely by incubating in a 55°C-60°C water-bath for 15 min. . The optical density of RNA samples were measured at 260 nm and 280 nm on spectrophotometer, and the 260/280 ratio was calculated. This value for pure RNA sample should be as close to 2 as possible. RNA concentration was estimated using the OD260 value based on the observation that OD260 of l is equivalent to an RNA concentration of 40 pg/ml. . The quality of RNA samples was tested on a 1% agarose gel. Two distinct bands at 188 and 28s appeared after electrophoresis, verifying for pure and undegraded samples. 100 B. Restriction Fragment Differential Display Polymerase Chain Reaction (RFDD-PCR) Outline Cell culture (H4IIE) l Exposure to PF OS (for 72 hrs) Total RNA extraction Jr RT reaction and template preparation l PCR amplification l Separation on sequencing gel Isolation of differentially expressed gene l Re-amplification of gene fragments of interest Subcloning of isolated genes l Sequencing Determination of the identity and bio-significance of the genes l Confirming results Differential display experiment was conducted using kit from display systems, which is part of Azign Bioscience (Copenhagen, Denmark). RT reaction and template preparation 1. Based on the concentration of RNA samples determined using spectrophotometer, dilute appropriate volume of RNA sample in DEPC-water to make up the final concentration of 1 pg total RNA per tube. 2. Set up the first strand cDNA synthesis reaction as following: Total RNA (1.0 pg) 10.0 pl 101 Anchored primer 1.5 pl 10 x cDNA buffer] 2.5 pl dNTP mix 5.0 pl display Therrno-RT 1.0 pl sterile water 5.0 pl Incubate at 42 °C for 2 hr. 3. Prepare the second strand synthesis reaction mixture as following: First-strand mixture 25 pl 10 x cDNA buffer 2 7.5 pl dNTP mix 2.5 pl DNA polymerase I 1.2 pl RNAse H 0.8 pl Sterile water 38.0 pl Incubate at 16 °C for 2 hr. 4. Perform a phenol/chloroform extraction by adding to each 75 pl reaction mixture 125 pl water and 200 pl phenolzchloroform, and vortexing, then centrifuge for 5 min at 12,000 x g at 4°C. 5. Transfer the aqueous phase to a 1.5 ml eppendorf tube containing 20 pl 3 M sodium acetate (pH 5.2) and 400 pl 96% ethanol, and precipitate at -80°C overnight. 6. Centrifuge the precipitated samples at 12,000 x g for 15 min at 4°C. Discard supernatant, and wash the pellet in 50 pl 70% ethanol. After centrifugation at 12,000 x g for 5 min at 4 °C, air-dry the pellet, and dissolve it in 20 pl sterile water. 7. Confirm the efficiency of cDNA synthesis by running 10 pl of the re-suspended cDNA on a 0.8% agarose gel. A cDNA smear should be visible from approximately 100 to 200 base pair. 8. Prepare template by a endonuclease digestion reaction followed by adaptor ligation reaction: 102 10 x display PROFILE buffer 2.0 pl cDNA sample 10.0 pl Taq I restriction enzyme 0.5 p] Sterile water 7.5 pl After incubation at 65 °C for 2 hr, add the following: 10 x displayPROFILE buffer 0.75 pl adaptor mix 0.75 pl ATP 1.25 pl T4 DNA ligase 0.3 pl Sterile water 4.45 pl Incubated at 37 °C for 3 hr. 9. Confirm the quality of template by running control PCR reaction as following: DisplayTAQ FL 10 x reaction buffer 2.0 pl dNTP mix 0.8 pl control primer 8.0 pl display TAQ FL 0.3 pl template 0.2 pl sterile water 8.7 pl Perform PCR reaction for 30 cycles using the following condition: 94 °C, 30 sec 55°C, 30 sec 72°C, 1 min When running 10 pl reaction mix on a 1.5 % agarose gel, a DNA smear from approximately 50 to 1000 base pair should be visible. PCR amplification 1. End-labeling for radioactive reaction Set up reaction mix as following: 10 x display PROFILE buffer 0.10 pl O-extension primer 0.40 pl [y33P]-ATP 0.20 pl T4 polynucleotide kinase 0.02 pl 103 Sterile water 0.28 pl Incubate at 37 °C for 30 min. 2. Amplification of template Set up PCR reaction mix as following: DisplayTAQ FL 10 x reaction buffer 2.0 pl dNTP mix 0.8 pl labeled primer 1.0 pl display TAQ FL 0.3 pl display PROBE 4.0 pl template 0.2 pl sterile water 11.7 pl Amplify each template using the following PCR profile: 94°C, 1 min the first 10 cycles 94°C, 30 sec 60°C, 30 sec (reduce by 05°C for each cycle until 55°C is reached) 72°C, 1 min continue with another 30 cycles 94°C, 30 sec 55°C, 30 sec 72°C, 1 min Expression on the sequencing gel 1. Casting 5 % poly acrylamide sequencing gel 1.) make up 10 x TBE buffer as following: Tris base 108 g Boric acid 55 g EDTA 97g Add distilled water up to 1 liter, autoclave, pH 8.3 2.) make up gel solution as following: Urea 63 g 10 x TBE buffer 15 ml 30 % acrylamide/bis 25 ml add distilled water up to 150 ml 104 dissolve with low heat, filter sterilized, de-gas for 15 min. 3.) Case gel using sequensing gel unit from Biorad (Hercules, CA). Wash sequencing gel plates thoroughly using soap and hot water, rinse with distilled water followed by spray with ethanol. 4.) Assemble gel cell and holders following the instruction on the manual. 5.) After de-gas, add 165 p1 TEMED and 165 pl 25 % ammonium persulfate, mix gently. Fill up the big syringe with gel solution, carefully get rid of air bubbles. Insert the tip of the tube connected to the syringe to the small hole at the bottom of the gel cell, push syringe slowly. Take extreme care while casting the gel, no air bubble should form between the plates. Insert the comb on top of the gel, with the flat edge down. Hold the top of the gel with four clamps, let the gel stand for about 1 hr to become solid. . After gel is cast, take out the comb, revert it and insert back, this forms 49 wells on the top of the gel. Pre-run the gel with 1 x TBE buffer at 75 W for 30 min. Loading buffer can be loaded in pre-run to check the quality of the gel. . Take out 4 p1 of PCR reaction, add 3 pl loading buffer to each tube, incubate at 85 °C for 5 min. Load 5 pl of sample to each well on the gel. Run the sequencing gel at 80 W for 3 hr. . After run-off, discard running buffer, carefully separate the plate, use filter paper to pick up the gel, put pre-wet membrane on top of the gel. Dry in the gel dryer for 3 hr. . When the gel is completely dry, in the dark room, put the gel in the exposure cassette. Mark one comer of the gel with alignment marker. Fit film on top, expose for 48 hr. 105 Put exposure cassette in —80°C for best exposure result. Develop film after 2 days exposure. Isolating the differentially expressed gene Compare the DNA profile among samples, especially the PFOS treated ones and the solvent control. Identify differentially expressed gene, both the density of the band and the presence of specific band. Overlay the film on top of the gel, pin at the band of interest. Use steriled razor blade cut off the both the band on the film and the underlying gel, and re-dissolve it in 50 pl sterile water. Heat up samples at 95°C for 15 min, and spin down for 10min at 14,000 x g. Transfer the supernatant to a fresh microcentrifuge tube. Re-amplification of gene fragments of interest To re-amplify the gene fragments of interest, set up the PCR reactions as the following: display 10X reaction buffer 4.0 pl dNTP mix (SmM each) 1.6 pl O-extension primer (10 pM) 0.8 pl display probe (1 pM) 8.0 pl gene fragment solution 5.0 pl display TAQ 0.6 pl sterile water 20 pl Amplify the reaction with 30 cycles of PCR using the following profile: 94°C, 30 sec for strand separation; 55°C, 30 sec for anneal; 72°C, 1 min for amplification. Separate PCR products on 1 % low melting agarose gel at 75 V for 45 min. Cut out gel slices containing single band, and extract DNA using Ultrafree-DA devices from Amicon (Charlotte, NC). Place gel slices into the pro-assembled filtering unit and spin at 5,000 xg for 10 min at room temperature. Store DNA samples at —20°C. 106 Subclaning of isolated genes Due to the short length of the differential display probes, subcloning of isolated DNA fragments is necessary before they can be subjected to sequencing. Cloning of target genes is conducted using pGEM-T easy vector system with T-A cloning technique from Invitrogen (Carlsbad, CA). The pGEM-T easy vector is constructed by cutting normal vector using EcoR-V restriction enzyme and adding a 3’ terminal thymidine to both ends. These single 3’-T overhangs at the insertion site greatly improve the efficiency of ligation of a PCR product into the plasmid, since PCR product generated by Taq DNA polymerase often have a single 3’-A overhangs in a template-independent fashion. Purify DNA fragments isolated from sequencing gel using Qiagen RNeasy kit (Valencia, CA), and concentrate for subsequent ligation into plasmid. Set up the ligation reactions along with appropriate controls in the following way: Target reaction Positive control Background control 2X ligation buffer 5 pl 5 pl 5 pl PGEM-T easy vector 1 pl 1 pl 1 pl PCR product X pl 0 pl 0 p1 Control insert DNA 0 pl 2 pl 0 pl T4 DNA ligase 1 pl 1 pl 1 pl Distilled water (lO-x) pl 1 pl 3 pl Mix the reaction components by pipetting and incubate for 1hr at room temperature. 107 *Positive control of insert DNA is used to determine ligation efficiency, typically 100 colonies should be observed. Background control with no insert DNA is used to determine the number of background blue colonies resulting from undigested vectors. Transformation control with uncut plasmid was used to check the transformation efficiency with competent cells. Transformation is conducted using JM109 high efficiency competent cells from Promega (Madison, WI). Thaw one vial of competent cell on ice for 5 min, and label four 5 ml round bottom culture tubes. Add 2 pl of each ligation product, 50 pl of competent cell suspension to each tube, and mix gently. Incubate on ice for 20 min and heat shock on 42°C heat block for exact 45 sec. Transfer to ice and add 950 pl SOC medium, shake for 1.5 hr in 37°C water-bath at about 150 RPM. During the meanwhile, take out 8 LB plate, on each plate add 100 pl IPTG and 20 pl X-Gal solution, spread over using metal policeman (rinse with ethanol and burn on fire, cool down before each use), and warm up the plate to room temperature. After incubation, add 100 pl of each transformation solution to the plate, spread with metal policeman, incubate at 37°C over night. Successful cloning of an insert into the pGEM-T vector interrupts the coding sequence of B-galactosidase, so positive recombinant clones can be identified with color screening. After overnight incubation, plates containing positive control should show a great amount of white colonies, and very few blue colonies for background control. Pick 6 white colonies from target plate using sterilized tooth pick and insert it into 5 ml culture tube containing 2 ml LB medium. Incubate the tube in 37°C water-bath for 12-15 hr, while shaking at 125 RPM. 108 After overnight incubation, competent cells grow to an intensity by which tooth-pick could not be seen through the cell suspension. Transfer cell suspension to 2 ml centrifugation tube and purify plasmid using Wizard plus SV minipreps from Promega (Madison, WI). Harvest 2 ml of bacterial culture by centrifugation for 5 min at 10,000 x g. Discard the supernatant and blot the inverted tube on a paper towel to remove excess media. Add 250 pl of cell suspension solution and completely resuspend cell pellet by vortexing. Add 250 pl of cell lysis solution and mix by inverting the tube four times, and incubate for 5 min. Add 10 pl of alkaline protease solution and mix by inverting the tube four times. Incubate for 5 min at room temperature. Add 350 pl of neutralization solution and immediately mix by inverting the tube four times. Centrifuge the cell lysate at 14,000 x g for 10 min at room temperature. Transfer the clear lysate to the spin column, avoid disturbing or transferring any precipitate. Centrifuge the supernatant at 14,000 x g for 1 min at room temperature. Remove the spin column and discard the flow through from the collecting tube. Add 750 pl of wash solution, previously diluted with 95 % ethanol to spin column. Spin at 14,000 x g for l min, and discard flow through. Repeat the wash procedure using 250 pl wash solution. Centrifuge at 14,000 x g for 2 min. Transfer the spin column to a fresh microcentrifuge tube and elute the plasmid DNA by adding 100 pl of nuclease-free water to spin column. Let it sit for 10 min, then centrifuge at 14,000 x g for 1 min. Store sample at -20°C. Reagents used for cloning: 109 LB medium: in a 2 L flask add Bacto-trypton 10 g, Bacto-yeast extract 5g, NaCl 5 g, 800 ml distilled water, adjust pH to 7.0 with NaOH and add volume up to l L. Sterilize with autoclaving. LB plate: add 15 g agar to l L LB medium and autoclave. Allow the medium to cooldown to 50°C then add 1 ml of ampicillin stock solution and mix well. Pour approximately 25 ml of the medium into eath petri-dish, let it harden. Store at 4°C. Ampicillin stock: add 1 g ampicillin in 10 ml distilled water to make 100 mg/ml stock, split into 10 tubes with 1 ml in each, and store at -20°C. IPTG stock: add 1.2 g IPTG in 50 ml distilled water, filter sterilize, and store at 4°C. X-Gal stock: dissolve 100 mg X-Gal in 2 ml dimethyl-formamide, and cover the bottle with aluminum foil, store at -20°C. SOC medium: add 2 g bacto-tryptone, 0.5 g bacto-yeast extract, 1 m1 1 M NaCl, and 0.25 ml 1 M KCl in 90 ml distilled water, autoclave and cool down to room temperature. Add 1 ml 2 M megnisium stock (10.17 g Mng and 12.33 g MgSO4 in 50 ml water) andlml 2 M glucose, bring volume to 100 ml, and filter sterilize. Sequencing and Sequence comparison Transfer 10 pl of each cloning sample (concentrate to 50 ng/pl) to 96 round bottom well plate, and submit the plate to Genomics facility on MSU campus for sequence analysis. DNA sequencing is conducted through dideoxynucleotide labeling technique using the ABI 3730 Genetic Analyzer from Applied Biosystems (Foster City, CA). Obtained sequences can then be submitted to Genebank (th://ncbi.nlm.nih.gov) for sequence comparison. Since different gene sequences with the same length could co-migrate to the 110 same position, typically 2-3 different sequences could be obtained from each of the 6 white colonies picked from original plate. Multiple sequence alignment BLAST program using BLOSUM62 was used to make sequence comparison, the top 10 significant hits were examined, and sequence identity determined. lll C. Affymetrix GeneChip® Array Overview of GeneChips method Animal Exposure l Total RNA extraction l cDNA synthesis l Biotin-labeled cRNA synthesis Target-grobe hybridization Probe array washing and staining Probe array scan l Data analysis A flfvmetrix GeneChip Array build-up GeneChip probe arrays are manufactured using technology that combines photolithographic methods and combinational chemistry. A glass substrate is coated with linkers containing photo liable-protecting groups. Then, a mask is applied on top of glass surface so that only selected portions of the probe array are exposed to ultraviolet light. Illumination removes the protecting groups enabling selective nucleoside to be added on the end of linker compound by chemical coupling reaction. Next a different mask is applied and the cycle of illumination and chemical coupling is performed again. By repeating this process, a specific set of oligonucleotide probes is synthesized at known positions. This 1 cm2 glass substrate is then mounted in a plastic cartridge, which then serves as hybridization chamber as well as protecting shield. 112 cDNA synthesis Synthesis of cDNA from total RNA was performed using the SuperScript Choice System obtained from Gibco BRL life Technologies (Cheshire, UK). The amount of 16 pg high quality total RNA dissolved in 10 pl DEPC water was incubated at 70°C with 1 pl 100 pmol/pl T7-(dT)24 primer for 10 min. T7-(dT)24 primer (5’ — GGC CAG TGA ATT GTA ATA CGA CTC ACT ATA GGG AGG CGG — (dT)24 —3’) was obtained from Genset Corp. (Boulder CO). 4 pl of the 5 X first strand cDNA buffer, 2 pl of 0.1 M DTT and 10 mM dNTP mixture were added to the tube, and allowed to equilibrate to 42°C for 2 min, before a 2 pl of S811 reverse transcriptase was added in the tube. The first strand cDNA synthesis reaction took 1 hr at 42°C. At the end of reaction the samples were placed on ice and centrifuged briefly. The following reagent were added to each tube: DEPC-treated water 91 pl 5 X second strand reaction buffer 30 pl 10 mM dNTP mix 3 pl 10 U/pl DNA ligase 1 pl 10 U/ pl DNA polymerase I 4 pl 2 U/ pl Rnase H 1 pl The tube was gently tapped to mix and incubated at 16°C for 2 hr. At the end of incubation, 2 pl T4 DNA polymerase was added and incubation went on for another 5mins. A 10 pl 0.5 M EDTA was added to the reaction mixture. Double stranded cDNA was cleaned up using phase-lock-gel (PLG) phenol/chloroform extraction tube from Eppendorf (W estbury, NY). A volume of 162 pl phenolzchloroformzisoamyl alcohol (saturated with 10 mM Tris-HCl and 1 mM EDTA, pH 8.0) was added to the cDNA synthesis preparation, and vortexed for 2 min. Then the mixture was transferred to PLG 113 tube, which was centrifuged at 12,000 x g for 2 min. The aqueous upper phase was transferred to a flesh tube and 0.5 volumes of 7.5 M ammonia acetate and 2.5 volumes of absolute ethanol (prechilled) were added to each tube, and vortexed, followed by immediate centrifugation at 12,000 x g for 20 min. The supernatant was removed and the pellet was washed with 0.5 ml 80 % prechilled ethanol. After centrifugation at 12,000 x g for 5 min, the washing step was repeated one additional time. The pellet was air dried and resuspended in 12 pl DEPC water. Synthesis ofbiotin-labeled cRNA An in vitro transcription reaction was performed to produce biotin-labeled cRNA from the cDNA obtained from previous step using the BioArray Higtheld RNA transcript labeling kit from Affymetrix Inc. (Santa Clara, CA). To an Eppendorf tube the following were added; CDNA template 5 p1 DEPC water 17 pl 10 X reaction buffer 4 pl 10 X biotin labeled ribonucleotides 4 pl 10 X DTT 4 pl 10 X Rnase Inhibitor 4 pl 20 X T7 RNA polymerase 2 pl The reagents were mixed and the tube was transferred to a 37°C water-bath incubating for 4.5 hr. The contents of the tube were mixed every 30 min by gently tapping on the tube. The cRNA samples were cleaned up using an Rneasy spin column from Qiagen (Valencia, CA) to remove unincorporated NTPs and followed by ethanol precipitation. Each sample was split into two tubes, and the volume was adjusted to 100 pl with Rnase- 114 free water. The 350 pl of the RLT lysis buffer was added to each tube. Samples were mixed with vigorous shaking and a volume of 250 pl 100 % pure ethanol was added into the lysate, and mixed well by pipetting. The mixtures were applied directly to the Rneasy mini spin column sitting on a collection tube. The set was then centrifuged at 8000 x g for 15 sec. The collection tube was discarded and the column was transferred to a fresh tube. An aliquot of 500 pl of salt buffer RPE was added in each column and the set was spun for another 15 sec at 8000 x g. The washing step was repeated once more, and the column was centrifuged for 2 min at maximum speed (14,000 x g) to dry the Rneasy membrane. The column was then transferred to a new collection tube, and 40 pl DEPC water was pipetted onto the membrane and allowed to sit at RT for 5 min. The RNA samples were eluted by centrifugation for 1min at 10,000 x g, this step was repeated one more time to allow for complete elution. A 0.5 volume of 7.5 M ammonia acetate and 2.5 volumes of absolute ethanol (prechilled) were added to each tube, and vortexed. RNA samples were allowed to precipitate at —20°C overnight. The samples were centrifuged at 12,000 x g for 30 min at 4°C. Supernatant was removed and pellet was washed with 0.5 ml 80 % prechilled ethanol. After centrifugation at 12,000 x g for 5 min, the washing step was repeated one additional time. The pellet was air dried and resuspended in 30 pl DEPC water. The yield of Biotin-labeled cRNA was determined using spectrophotometric analysis. The optical density (OD) value at 260 nm and 280 nm was measured to determine sample concentration and purity, applying the convention that lOD at 260nm equals 40 pg/ml RNA. Only samples with OD 260/280 ratio between 1.9 and 2.1 are acceptable. Since the 115 cRNA was obtained using total RNA as starting material, an adjusted cRNA yield was calculated using the following formula to reflect carryover of unlabeled total RNA: Adjusted cRNA yield = RNAm — (RNAmml) (Y) RN Am = amount of cRNA measured after the in vitro transcription reaction (pg) RNAtom. = starting amount of total RNA (pg) Y = fraction of cDNA reaction used in the in vitro transcription reaction For efficient hybridization the cRNA was fragmented using 20 pg cRNA sample and 8 pl of 5 X fragmentation buffer diluted with RNase-free water up to the volume of 40 pl. The mixture was incubated at 94°C for 35 min, and transferred to ice following the incubation. 5 X fragmentation buffer: 4.0 ml 1M Tris acetate pH 8.1 0.64 g MgOAc 0.98 g KOAc DEPC-treated water to 20 ml Reaction mixture was mixed thoroughly and filtered through a 0.2 pm vacuum filter unit. Gel electrophoresis of the cRNA product was conducted to estimate the yield and size distribution of labeled transcripts. Samples were loaded into the 1 % agarose gel, and electrophoresize for 45 min at 75 V. Gel was stained using ethidium bromide for 10 min and destained with destilled water for 30 min. Sample distribution was visualized using GelDoc system from BioRad (Hercules, CA). T he following steps were performed in the Gemonics facility on MS U campus. 116 Target Hybridization The hybridization cocktail was prepared following the table, and the volume was scaled up for multiple chips. Component Amount Final Concentration Source Fragmented cRNA 15 pg 0.05 pg / pl from previous steps Control Oligonucleotide 5 pl 50 pM Affymetrix 20 X Eukaryotic 15 pl 100 pM Affymetrix Hybridization Controls Herring Sperm DNA 3 p] 0.1 mg/ml Promega Acetylated BSA 3 p] 0.5 mg/ml Gibco BRL 2 X Hybridization 150 pl 1 X Affymetrix Buffer Rnase-free water To a final volume of 300 pl The hybridization cocktail was heated to 99°C for 5 min in a heated block and was then transferred to 45°C heat block for 5 min for equilibration. The hybridization mixture was centrifuged for 5 min to remove any insoluble material. Meanwhile, the chip array was warmed up to room temperature, and wetted with 250 pl 1 X hybridization buffer, and incubated at 45°C for 10 min with rotation. After the buffer solution was removed from the probe array cartridge, it was filled with 250 pl volume of the clarified hybridization cocktail solution. The probe array was then placed in rotating incubator at 45°C and allowed to hybridize for 16hrs while rotating at 60 RPM. Washing, Staining and Scanning Probe arrays The GeneChip Fluidics Station 400 from Affymetrix was used to wash and stain the probe arrays. It was operated using GeneChip software. Refer to the GeneChip Fluidics Station 400 user’s manual for instructions on connecting and addressing multiple fluidics 117 stations. The experiment was first defined by entering experiment name and parameters in GeneChip software. Priming the fluidics station was conducted by turning on appropriate modules and filling in the buffer reservoir with non-stringent wash buffer. After 16hrs of hybridization, the hybridization cocktail was removed from the probe array, and the array was refilled with non-stringent wash buffer and allowed to equilibrate to room temperature. Streptavidin phycoerythrin working solution obtained from Molecular Probes (Eugene, OR) was prepared immediately before usage. The following components was mixed in a microcentrifuge tube: 2X stain buffer 300 pl DEPC water 270 pl 50 mg/ml acetylated BSA (Gibco BRL) 24 pl 1 mg/ml streptavidin phycoerythrin 6 pl The antibody amplification solution was prepared by mixing the following components in a microcentrifuge tube: 2 X stain buffer 300 p1 DEPC water 266.4 pl 50 mg/ml acetylated BSA 24 pl 10 mg/ml normal goat IgG (Sigma chemical) 6 pl 0.5 mg/ml biotinylated antibody (Vector Laboratories) 3.6 pl The probe array was inserted into the designated module of the fluidics station, and microcentrifuge tubes containing the staining buffer and antibody staining buffer were place in appropriate sample holder. In the fluidics station protocol drop-down list, the appropriate control protocol for washing and staining steps were selected and following the instruction on LCD window display to start. Eject message would be displayed on 118 LCD window when the whole protocol was completed. The probe array was then removed from the module and kept at 4°C in the dark until ready for scanning. The scanner was also connected with the station and controlled by the GeneChip software. Prior to scanning, make sure the scanner option was set to pixel value = 3 pm, wavelength = 570 nm, and the laser was warmed up for at least 15 min. The probe array was inserted into the sample holder. From the control drop-down manual, the experiment name corresponded to the probe array was selected, and the image scanning was performed by clicking the start button. Each probe array was scanned twice with each time approximately 5 min. The computer work station automatically overlaied the two scanned images and averaged the intensities of each probe cell for the greatest assay sensitivity. Each complete image was then saved as data image file with .dat extension. GeneChips Data Analysis Each image file was analyzed and data was retrieved using Affymetrix data mining tool. The outcome file was then stored in a database built using Microsoft Access. Initial data analysis was also conducted using Microsoft Access query design. Further Cluster analysis, Genetree construct and pathway analysis were conducted using GeneSpring software obtained from SiliconGenetics (Redwood City, CA). 119 REFERENCES 120 REFERENCES . Affymetrix® Microarray Suite 5.0 User’s Guide. . Altschul, S.F., Madden, T.L., Schiiffer, A.A., Zhang, J., Zhang, Z., Miller, W., and Lipman, DJ. (1997) Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25: 3389-3402. . Beguinot, F., Tramontano, D., Duilio, C., Formisano, S., Beguinot, L., Mattioli, P., Mancini, M., and Aloj, SM. (1985) Alteration of erythrocyte membrane lipid fluidity in human obesity. J. Clin. Endocrinol. Metab 60: 1226-1230. . Beguinot, F., Beguinot, L., Tramontano, D., Duilio, C., Formisano, S., Bifulco, M., Ambesi-Impiombato, RS, and Aloj, SM. (1987) Thyrotropin regulation of membrane lipid fluidity in the FRTL-S thyroid cell line. Its relationship to cell grth and functional activity. J. Biol. Chem. 262:1575-1582. . Berthiaume, J., and Wallace, KB. (2002) Perfluorooctanoate, perfluorooctane sulfonate, and N-ethyl perfluorooctane sulfonamide ethanol; peroxisome proliferation and mitochondrial biogenesis. T oxicol. Lett. 129: 23-32. . Biegel, L.B., Liu, R.C. M., Hurtt, M.E., and Cook, J.C. (1995) Effects of ammonium perfluorooctanoate on Leydig cell function: in vitro, in vivo, and ex vivo studies. T oxicol. Appl. Pharmacol. 134: 18-25. . Boujrad, N., Vidic, B., Gazouli, M., Culty, M., and Papadopoulos, V. (2000) The peroxisome proliferator perfluorodecanoic acid inhibits the peripheral-type benzodiazepine receptor (PBR) expression and hormone-stimulated mitochondrial cholesterol transport and steroid formation in Leydig cells. Endocrinology 141(9): 3137-3148. . Brondyk, W.H., McKiernan, C.J., Fortner, K.A., Stabila, P., Holz, R.W., and Macara, LG. (1995) Interaction cloning of Rabin3, a novel protein that associates with the Ras-like GTPase Rab3A. Mol. Cell Biol. 15(3): 1137-1143. 121 10. 11. 12. 13. 14. 15. 16. 17. 18. Cook, J.C., Murray, S.M., Frame, SR, and Hurtt, ME. (1992) Induction of Leydig cell adenomas by ammonium perfluorooctanoate: a possible endocrine- related mechanism. T oxicol. Appl. Pharmacol. 113: 209-217. Cossarizza, A., Baccarani-Contri, M., Kalashnikova, G., and Franceschi, C. (1993). A new method for the cytofluorimetric analysis of mitochondrial membrane potential using the J-aggregate forming lipophilic cation 5,5’,6,6’- tetrachloro-1,l ’,3,3’-tetraethy1 benzimidazol carbocyanine iodide (JC-l). Biochem. Biophy. Res. Comm. 197: 40-45. Diaz, M.J., Chinje, E., Kentish, P., Jamot, B., George, M., and Gibson, G. (1994) Induction of cytochrome P450 4A by the peroxisome proliferator perfluoro-n- octanoic acid. Toxicology. 86: 109-122. Display Systems Biotech Inc. (1999) Restriction Fragment Differential Display. Kit manual version 2.1. Display Systems Biotech Inc., Buena Vista, CA. pp19. Douglass et al., (1995) PCR differential display identifies a rat brain mRNA that is transcriptionally regulated by cocaine and amphetamine. J. Neurosciences. 15: 2471-2481. Duplus, E., Glorian, M., and Forest, C. (2000) Fatty acid regulation of gene transcription. J. Biol. Chem. 275: 30749-30752. Galbiati, F., Razani, B., and Lisanti, MP. (2001) Emerging themes in lipid rafts and caveolae. Cell 106: 403-411. Ganesan, A.K., Vincent, T.S., Olson, J .C., and Barbieri, J .T. (1999) Pseudomonas aeruginosa exoenzyme S. disrupts Ras-mediated signal transduction by inhibiting Guanine nucleotide exchange factor-catalyzed nucleotide exchange. J. Biol. Chem. 274: 21823-21829. Gao, L. et al., (1998) A novel response to dioxin. Induction of ecto-ATPase gene expression. J. Biol. Chem. 273:15358-15365. GeneChip® Expression Analysis Technical Manual. 122 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. Gerhold, D.L., Liu, F., Jiang, G., Li, Z., Xu, J., Lu, M., Sachs, J.R., Bagchi, A., Fridman, A., Holder, D.J., Doebber, T.W., Berger, J ., Elbrecht, A., Moller, DE, and Zhang, BB. (2002) Gene expression profile of adipocyte differentiation and its regulation by peroxisome proliferator-activated receptor-gamma agonists. Endocrinology 143: 2106-21 18. Giesy, J. P., and Kannan, K. (2001) Global distribution of perfluorooctane sulfonate in wildlife. Environ. Sci. T echnol. 35: 1339-1342. Giesy, J. P., and Kannan, K. (2002) Perfluorochemical surfactants in the environment. Environ. Sci. T echnol. 35: 1339-1342. Goldenthal, E.I., Jessup, D.C., Geil, R.G., Jefferson, N.D., Arceo, R.J., and Ruecker, FA. (1978) 90-day subacute rat study. Study No. 137-085, International Research and Development Corp. Mattawan, MI. Available on USEPA public docket AR-226. Green, S. (1992) Commentary: receptor-mediated mechanisms of peroxisome proliferators. Biochemical Pharmacology 43: 393-401. Green, C.B. et al., (1996) Identification of a novel vertebrate circadian clock- regulated gene encoding the protein noctumin. Proc. Natl. Acad. Sci. USA. 93: 14884-14888. Guimaraes, M.J. et al., (1995) Differential Display by PCR: novel findings and applications. Nucleic Acids Res. 23: 1832-1833. Hahn, M.E., and Chandran, K. (1996) Uroporphyrin accumulation associated with cytochrome P4501A induction in fish hepatoma cells exposed to aryl hydrocarbon receptor agonists, including 2,3,7,8-tetrachlorodibenzo-p-dioxin and planar chlorobiphenyls. Arch. Biochem. Biophys. 329: 163-174. Hahn, M.E., Lamb, T.M., Schultz, M.E., Smolowitz, R.M., and Stegeman, JJ. (1993) Cytochrome P4501A induction and inhibition by 3,3’,4,4’- tetrachlorobiphenyl in an Ah receptor-containing fish hepatoma cell line (PLHC- 1). Aquat. T oxicol. 26: 185-208. Hamadeh, H.K., Bushel, P.R., Jayadev, S., Martin, K., DiSorbo, 0., Siegber, S., Bennett, L., Tennant, R., Stoll, R., Barrett, C., Blanchard, K., Paules, RS, 123 29. 30. 31. 32. 33. 34. 35. 36. 37. Afshari, C.A., (2002a) Gene expression analysis reveals chemical-specific profiles. T oxico]. Sci. 67: 219-231. Hamadeh, H.K., Bushel, P.R., Jayadev, S., Martin, K., DiSorbo, 0., Bennett, L., Lee, L., Tennant, R., Stoll, R., Barrett, C., Barrett, K., Paules, R.S., Blanchard, K., and Afshari, C.A., (2002b) Prediction of compound signature using high density gene expression profiling. T oxico]. Sci. 67 : 232-240. Hanhijarvi, H., Ophaug, RH, and Singer, L. (1982) The sex-related difference in perfluorooctanoate excretion in the rat. Proc. Soc. Exp. Biol. Med. 171: 50-55. Hansen, K.J., Clemen, L.A., Ellefson, M.E., and Johnson, H0. (2001) Compound-specific, quantitative characterization of organic fluorochemicals in biological matrices. Environ. Sci. T echno]. 35: 766-770. Haughom, B., and Spydevold, O. (1992) The mechanism underlying the hypolipemic effect of perfluorooctanoic acid (PFOA), perfluorooctane sulphonic acid (PFOSA) and clofibric acid. Biochimica et Biophysica Acta. 1128: 65-72. Hightower, LE. and Renfro, J .L. (1988) Recent applications of fish cell culture to biomedical research. J. Exp. Zoo]. 248: 290-302. Higuchi, E., Oridate, N., Furuta, Y., Suzuki, S., Hatakeyama, H., Sawa, H., Sunayashiki-Kusuzaki, K., Yamazaki, K., Inuyama, Y., and Fukuda, S. (2003). Differentially expressed genes associated with CIS-diarnminedichloroplatinum (II) resistance in head and neck cancer using differential display and CDNA microarray. Head Neck 25: 187-193. I-lu, W.Y. (2000) In vitro studies of the biochemical toxicity of perfluorooctane sulfonic acid and its possible interaction with 2,3,7,8 — tetrachlorodibenzo-p- dioxin. MSc Thesis, Michigan State University, East Lansing, MI 48824. Hu, W.Y., Jones, P.D., DeCoen, W., King, L.B., Fraker, P., Newsted, J.L., and Giesy, J .P. (2002a) Alterations in cell membrane properties caused by perfluorinated compounds. Comp. Biochem. Physio]. 135: 77-88. Hu, W.Y., Jones, P.D., Upham, B.L., Trosko, J.E., Lau, C., and Giesy, J.P. (2002b) Inhibition of gap junctional intercellular communication by 124 38. 39. 40. 41. 42. 43. 45. perfluorinated compounds in rat liver and dolphin kidney epithelial cell lines in vitro and Sprague-Dawley rats in vivo. T oxicol. Sci. 68: 429-436. Hu, W.Y., Jones, P.D., Celius, T., and Giesy, J .P. (2003a) Identification of genes responsive to perfluorooctane sulfonic acid using gene expression profiling. European J. T oxicol. Pharmacol. (In Press). Hu, W.Y., Jones, P.D., DeCoen, W., and Giesy, J .P. (2003b) Comparison of gene expression methods to identify genes responsive to perfluorooctane sulfonic acid. Environmental Toxicology and Pharmacology (In Press). Ikeda, T., Aiba, K., Fukuda, K., and Tanaka, M. (1985) The induction of peroxisome proliferation in rat liver by perfluorinated fatty acids, metabolically inert derivatives of fatty acids. J. Biochem. 98: 475-482. Jefferson, J.R., Powell, D.M., Rymaszewski, Z., Kukowska-Latallo, J., Lowe, J .B., and Schroeder, F. (1990) Altered membrane structure in transfected mouse L-cell fibroblasts expressing rat liver fatty acid-binding protein. J. Biol. Chem. 265: 11062-11068. Jones, P.D., Hu, W.Y., DeCoen, W., Newsted, J .L., and Giesy, JP. (2002) The binding of perfluorinated chemicals to serum proteins. Environ. T oxico]. Chem. (In Press). Just, W.W., Gorgas, K., Hartl, F .U., Heinemann, P., Salzer, M., and Schimassek, H. (1989) Biochemical and zonal heterogeneity of peroxisome proliferation induced by perfluorocarboxylic acids in rat liver. Hepatology 9: 570-581. . Kannan, K., Koistinen, J ., Beckmen, K., Evans, T., Gorzelany, J .F., Hansen, K.J., Jones, P.D., Helle, E., Nyrnan, M., and Giesy, J.P. (2001a). Accumulation of perfluorooctane sulfonate in marine mammals. Environ. Sci. T echnol. 35: 1593- 1598. Kannan, K., Franson, J.C., Bowerman, W.W., Hansen, K.J., Jones, P.D., and Giesy, J .P. (2001b) Perfluorooctane sulfonate in fish-eating water birds including bald eagles and albatrosses. Environ. Sci. T echnol. 35: 3065-3070. 125 46. 47. 48. 49. 50. 51. 52. 53. 54. Kawashima, Y., Kobayashi, H., Miura, H., and Kozuka, H. (1995) Characterization of hepatic response of rat to administration of perfluorooctanoic acid and perfluorodecanoic acid at low level. Toxicology 99: 169-178. Kawasaki, K., Yin, J.J., Subczynski, W.K., Hyde, J.S., and Kusumi, A. (2001) Pulse EPR detection of lipid exchange between protein-rich raft and bulk domains in the membrane: methodology development and its application to studies of influenza viral membrane. Biophys. J. 80: 738-748. Kramer, V.J., Helferich, W.G., Bergman, A., Klasson-Wehler, E., and Giesy, J .P. (1997) Hydroxylated polychlorinated biphenyl metabolites are anti-estrogenic in a stably transfected human breast adenocarcinoma (MCF7) cell line. T oxicol. Appl. Pharmacol. 144: 363-376. Kudo, N., Bandai, N., Suzuki, E., Katakura, M., and Kawashima, Y. (2000) Induction by perfluorinated fatty acids with different carbon chain length of peroxisomal B-oxidation in the liver of rats. Chemico-Biologica] Interaction 124: 119-132. Kudo, N., Suzuki, E., Katakura, M., Ohmori, K., Noshiro, R., and Kawashima, Y. (2001) Comparison of the elimination between perfluorinated fatty acids with different carbon chain length in rats. Chemico-Biologica] Interactions 134: 203- 216. Lau, C., Thibodeaux, J .R., Hanson, R.G., Rogers, J .M., Grey, B.E., Stanton, M.E., Butenhoff, J .L., and Stevenson, LA. (2003) Exposure to perfluorooctane sulfonate during pregnancy in rat and mouse. 11: postnatal evaluation. T oxico]. Sci. 74: 382-392. Liang, P., and Pardee, AB. (1992) Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science 257: 967-971. Liang, P. et al., (1992) Differential display and cloning of messenger RNAs from human breast cancer versus mammary epithelial cells. Cancer Research. 52: 6966-6968. Liang, P. et al., (1994) Differential display using one-base anchored oligo-dT primers. Nucleic Acids Res. 22: 5763-5764. 126 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. Liang, P. (1998) Factors ensuring successful use of differential display. Methods in Enzymology. 16: 361-364. Liu, R.C.M., Hahn, C., and Hurtt, ME. (1996) The direct effect of hepatic peroxisome proliferators on rat Leydig cell firnction in vitro. T oxico]. Appl. Pharmacol. 30: 102-108. Liu, R.C., Hurtt, M.E., Cook, BC, and Biegel, L.B. (1996b) Effect of peroxisome proliferator, ammonium perfluorooctanoate (C8), on hepatic Aromatase activity in adult male Cr1:CD BR rats. F undam. Appl. T oxico]. 30(2): 220-228. Lockhart, D.J., and Winzeler, EA. (2000) Genomics, gene expression and DNA arrays. Nature 405: 827-836. Luebker, D.J., Hansen, K.J., Bass, N.M., Butenhoff, J .L., and Seacat, A.M. (2002) Interactions of fluorochemicals with rat liver fatty acid-binding protein. Toxicology 176: 175-185. Masuda, M., Kuriki, H., Komiyama, Y. et al. (1987) Measurement of membrane fluidity of polyrnorphonuclear leukocytes by flow cytometry. J. Immunol. Meth. 96: 225-231. Maniatis, T., and Reed, R. (2002) An extensive network of coupling among gene expression machines. Nature 416: 499-505. Matsuba, T., Keicho, N., Higashimoto, Y., Granleese, S., Hogg, J .C. Hayashi, S., and Bondy, GP. (1998) Identification of glucocorticoid- and adenovirus ElA- regulated genes in lung epithelial cells by differential display. Am. J. Respir. Cell Mo] Bio]. 18: 243-254. Matz, M.V., and Lukyanov, SA. (1998) Different strategies of differential display: areas of application. Nucleic Acids Research 26: 5537-5543. Meng, J.J., Bornslaeger, E.A., Green, K.J., Steinert, P.M., and 1p, W. (1997) Two- hybrid analysis reveals fundamental differences in direct interactions between desmoplakin and cell type-specific intermediate filaments. J. Biol. Chem. 272 (34): 21495-21503. 127 65. 66. 67. 68. 69. 70. 71. 72. 73. Mong, J.A., Krebs, C., and Pfaff, D.W. (2002) Perspective: micoarrays and differential display PCR-tools for studying transcript levels of genes in neuroendocrine systems. Endocrinology 143: 2002-2006. Oboum, J.D., Frame, S.R., Bell, R.H., Longnecker, D.S., Elliott, G.S., and Cook, J .C. (1997) Mechanisms for the pancreatic oncogenic effects of the peroxisome proliferator wyeth-14,643. T oxico]. Appl. Pharmacol. 145: 425-436. Okun, J .G., Lummen, P., and Brandt, U. (1999) Three classes of inhibitors share a common binding domain in mitochondrial complex I (N ADqubiquinone oxidoreductase). J. Biol. Chem. 274: 2625-2630. Olson, C. T., and Andersen, M. E. (1983) The acute toxicity of perfluorooctanoic and perfluorodecanoic acids in male rats and effects on tissue fatty acids. T oxicol. Appl. Pharmacol. 70: 362-372. Permadi, H., Lundgren, B., Anderson, K., and Depierre, J. W. (1992) Effects of perfluoro fatty acids on xenobiotic-metabolizing enzymes, enzymes which detoxify reactive forms of oxygen and lipid peroxidation in mouse liver. Biochemical Pharmacology. 44: 1 183-1 191. Pons, M., Gagne, D., Nicolas, J .C., Mehtali, M. (1990) A new cellular model of response to estrogens: a bioluminescent test to characterize anti-estrogen molecules. Biotechniques 9: 450-459. Pownall, H.J., and Smith, LC. (1989) Pyrene-labeled lipids: versatile probes of membrane dynamics in vitro and in living cells. Chemistry and Physics of Lipids. 50: 191-211. Ravis, W.R., Hoke, J .F ., and Parsons, D.L. (1991) Perfluorochemical erythrocyte substitutes: disposition and effects on drug distribution and elimination. Drug Metabolism Reviews. 23: 375-411. Reo, N.V., Narayanan, L., Kling, KB, and Adinehzadeh, M. (1996) Perfluorodecanoic acid, a peroxisome proliferator, activates phospholipase C, inhibits CTP: phosphocholine cytidylyltransferase, and elevates diacylglycerol in rat liver. T oxicologv Letters. 86: 1-11. 128 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. Richter, C.A., Tieber, V.L., Denison, MS, and Giesy, J.P. (1997) An in vitro rainbow trout cell bioassay for aryl hydrocarbon receptor-mediated toxins. Environ. T oxico]. Chem. 3: 543-550. Sanderson, J.T., Aarts, J.M., Brouwer, A., Froese, K.L., Denison, MS, and Giesy, J .P. (1996) Comparison of Ah receptor-mediated luciferase and ethoxyresorufin-O- deethylase induction in H4IIE cells: implications for their use as bioanalytical tools for the detection of polyhalogenated aromatic hydrocarbons. T oxico]. Appl. Pharmacol. 137: 316-325. Sargent, J., and Seffl, R. (1970) Properties of perfluorinated liquids. Fed. Proc. 29: 1699-1703. Seacat, A.M., Thomford, P.J., Hansen, K.J., Olsen, G.W., Case, M.T., and Butenhoff, J .L. (2002a) Subchronic toxicity studies on perfluorooctane sulfonate potassium salt in cynomolgus monkeys. T oxicol. Sci. 68: 249-264. Seacat, A.M., Thomford, P.J., and Butenhoff, J .L. (2002b) Terminal observations in Sprague-Dawley rats after lifetime dietary exposure to potassium perfluorooctane sulfonate. T oxicologist 66: 185 (Abstract ID: 906). Seacat, A.M., Thomford, P.J., Hansen, K.J., Clemen, L.A., Eldridge, S.R., Elcombe, CR, and Butenhoff, J.L. (2003) Sub-chronic dietary toxicity of potassium perfluorooctane sulfonate in rats. T oxicol. Sci. 183: 117-131. Shinoda, K., and Nomura, T. (1980) Miscibility of fluorocarbon and hydrocarbon surfactants in micelles and liquid mixtures. Basic studies of oil repellent and fire extinguishing agents. J. Phys. Chem. 84: 365-369. Simpson, E.R., Mahendroo, M.S., Means, G.D., Kilgore, M.W., et a]. (1994) Aromatase cytochrome P450, the enzyme responsible for estrogen biosynthesis. Endocrine Rev. 15: 342-355. Sohlenius, A.K., et a]. (1994) Effects of perfluorooctanoic acid, a potent peroxisome proliferator in rat, on morris hepatoma 7800Cl cells, a rat cell line. Biochimica et Biophysica Acta. 1213: 63-74. Sohlenius, A. K., Reinfeldt, M., Backstrom, K., Bergstrand, A., and Depierre, J. W. (1996). Hepatic peroxisome proliferation in vitamin A deficient mice without 129 84. 85. 86. 87. 88. 89. 90. 91. a simultaneous increase in peroxisomal Acyl-CoA oxidase activity. Biochemical Pharmacology. 51: 821-827. Sokol, R.J., Straka, M.S., Dahl, R., Devereaux, M.W., Yerushalmi, B., Gumpricht, E., Elkins, N., and Everson, G. (2001) Role of oxidant stress in the permeability transition induced in rat hepatic mitochondria by hydrophobic bile acids. Pediatr Res 49: 519-531. Starkov, A.A., and Wallace, KB. (2002) Structural determinants of fluorochemical-induced mitochondrial dysfunction. T oxico]. Sci. 66: 244-252. Takagi, A., Sai, K., Umemura, T., Hasegawa, R., and Kurokawa, Y. (1991) Short- terrn exposure to the peroxisome proliferators, perfluorooctanoic acid and perfluorodecanoic acid, causes significant increase of 8-hydroxydeoxyguanosine in liver DNA of rats. Cancer Lett. 57(1): 55-60. Thibodeaux, J.R., Hanson, R.G., Rogers, J.M., Grey, B.E., Barbee, B.D., Richards, J.H., Butenhoff, J.L., and Stevenson, LA. (2003) Exposure to perfluorooctane sulfonate during pregnancy in rat and mouse. 1: maternal and prenatal evaluations. T oxicol. Sci. 74: 369-381. Tobin, K.A.R., Ulven, S.M., Schuster, G.U., Hermansen Steineger, H., Andresen, S.M., Gustafsson, J .A., Nebb, H1 (2002) Liver X receptors as insulin-mediating factors in fatty acid and cholesterol biosynthesis. J. Biol. Chem. 277: 10691- 10697. Upham, B.L., Deocampo, N.D., Wurl, B., and Trosko, J.E. (1998) Inhibition of gap junctional intercellular communication by perfluorinated fatty acids is dependent on the chain length of the fluorinated tail. Int. J. Cancer. 78: 491-495. Vanden Heuvel, J .P., Davis, J .W., Sommers, R., and Peterson, R.E. (1992a) Renal excretion of perfluorooctanoic acid in male rats: inhibitory effect of testosterone. J. Biochem T oxico]. 7(1): 31-36. Vanden Heuvel, J .P., Kuslikis, BL, and Peterson, R.E. (1992b) Covalent binding of perfluorinated fatty acids to proteins in the plasma, liver and testes of rats. Chem. Biol. Interact. 82(3): 317-328. 130 92. 93. 94. 95. 96. 97. 98. 99. 100. Vanden Heuvel, J.P., Sterchele, P.F., Nesbit, D.J., and Peterson, RE. (1993) Coordinate induction of acyl-CoA binding protein, fatty acid binding protein and peroxisomal B-oxidation by peroxisome proliferators. Biochim. Biophys. Acta. 1177(2): 183-190. Wallace, K.B., Luebker, D.J., Butenhoff, J.L., and Seacat, A.M. (2001) Perfluorooctane sulfonate and 2-(N-ethylperfluorooctanesulfonamido)-ethy1 alcohol are peroxisome proliferators in rats, but not guinea pigs. T oxicologist 60: 348 (Abstract ID: 1657). Wan, Y., and Nordeen, SK, (2002) Overlapping but distinct gene regulation profiles by glucocorticoids and progestins in human breast cancer cells. Mo]. Endocrinol. 16: 1204-1214. Ylinen, M., Hanhijarvi, H., Jaakonado, J., and Peura, P. (1989) Stimulation by oestradiol of the urinary excretion of perfluorooctanoic acid in the male rat. Pharmacol. T oxico]. 65: 274-277. Ylinen, M., and Auriola, S. (1990) Tissue distribution and elimination of perfluorooctanoic acid in the rat after single intraperitoneal administration. Pharmacol. T oxico]. 66: 45-48. Youssef, J., and Badr, M. (1998) Extraperoxisomal targets of peroxisome proliferators: mitochondrial, microsomal, and cytosolic effects. Implications for health and disease. Crit. Rev. T oxicol. 28(1): 1-33. Yuen, T., Wurmbach, E., Pfeffer, R.L., Ebersole, B.J. and Sealfon, SC. (2002) Accuracy and calibration of commercial oligonucleotide and custom cDNA microarrays. Nucl. Acids. Res. 30: e48. Zhang, L., and Medina, D. (1993) Gene expression screening for specific genes associated with mouse mammary tumor development. Mo]. Carcinog. 8: 123-26. Zoeteweij, J .P., Water, B.V., DeBont, H.J., Mulder, G.J., and Nagelkerke, J .F . (1992) Involvement of intracellular Ca2+ and K+ in dissipation of the mitochondrial membrane potential and cell death induced by extracellular ATP in hepatocytes. Biochem. J. 288: 207-213. 131 Iiiilinllin llilil “ W 3 1293 ll