Hun!“ ‘ A': .5333. 7.. . .... ix“). A A .u... _ v.12: .3 ‘ 3 .4.‘ .3 1L: ~ra‘ J“... .12 . :. p 7. :5. 1.. t z . . t .5...) ’93 ~¥... .5 .. ‘11... 1 . .4 .r'nig. .O- .|'4->‘ (1.,- TAhssx 2. ~ l. :31...’ . . .2 “A an #5111: . a... 2.1%.; .. II." II - ‘l8 .‘J lllll‘llllll lllll it till This is to certify that the thesis entitled Development of an in vitro Rainbow Trout Cell Bioassay For AhR—mediated Toxins presented by Catherine Ann Richter has been accepted towards fulfillment of the requirements for Master of Science degree in Fish. & Wildl. (MA I914“! Major professor 0-7639 MS U is an Affirmative Action/Equal Opportum'ty Institution _ .— "— __ ——4——-— 4—.J—" LIBRARY . littlchigan mate I L'né'vereity PLACE ll RETURN BOXtoromavothb chockomfrom yourrocord. TO AVOID FINES mum on or before date duo. DATE DUE DATE DUE DATE DUE MSU Is An Affirmative Adlai/Emu! Opportunlty lmtltulon Wins-9.1 DEVELOPMENT OF AN IN VITRO RAINBOW TROUT CELL BIOASSAY FOR AHR-MEDIATED TOXINS By Catherine Ann Richter A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE Department of Fisheries and Wildlife 1995 ABSTRACT DEVELOPMENT OF AN IN VITRO RAINBOW TROUT CELL BIOASSAY FOR AHR-MEDIATED TOXINS By Catherine Ann Richter Halogenated aromatic hydrocarbons (HAHs) and other chemicals, which act as aromatic hydrocarbon receptor (AhR) agonists, cause a variety of toxic effects. In sac fry of many fish species, these effects include blue-sac disease and mortality. Because HAHs occur in complex mixtures, their toxicity in the environment is difficult to predict. A bioassay useful in predicting AhR- mediated toxicity to fish was developed using the RTH-149 rainbow trout hepatoma cell line. Stable transfection of this cell line with the pGudLuc1.1 plasmid, which contains a firefly luciferase reporter gene under the transcriptional regulation of dioxin responsive enhancers (DREs), has produced a recombinant cell line designated Remodulated Lightning Trout (RLT 1.0). The RLT 1.0 bioassay method detection limit for TCDD was estimated to be 22 pM. The responses of the RLT 1.0 bioassay to TCDD and several PCB congeners closely matched the responses observed in in viva fish bioassays. The RLT 1.0 bioassay can provide an integrative measure of the total AhR-mediated toxicity of a sample to fish. ACKNOWLEDGMENTS I would first like to thank my committee, Dr. Frank M. D'ltri, Dr. William Helferich, and Dr. John P. Giesy, for their guidance and support. Dr. Giesy has devoted much time and attention to my development as a scientist, and has provided many opportunities for me to expand my professional experiences. Dr. Mike Denison of the University of California at Davis developed the pGudLuc1.1 plasmid which made this project possible, and taught me the basics of working with cell culture and plasmids. Dr. Dave DeWitt and Dr. Bill Smith generously allowed me to work in their laboratories and provided valuable advice. Stacey Kraemer shared her knowledge of molecular biology and transfection techniques with me, and reviewed a much earlier draft of this thesis. The students and technicians of the Aquatic Toxicology Lab provided support, advice, and assistance. In particular, Virginia Leykam was indispensable in helping me make the cells grow and make the transfections work. I am also indebted to Bob Crawford, Dave Verbrugge, and John Besser. Finally, I would like to thank my family for teaching me to love the water, and listening to me talk about science stuff. This research was supported, in part, by National Institutes of Health Grant NIH-ES-O4911, and by National Institute of Environmental Health Sciences training grant T32—ES-07255. University sources of support included the Institute for Environmental Toxicology, the Department of Biochemistry, the Department of Fisheries and Wildlife, and the Pesticide Research Center. TABLE OF CONTENTS LIST OF TABLES ....................................... vii LIST OF FIGURES ...................................... ix INTRODUCTION ........................................ 1 Halogenated aromatic hydrocarbons ...................... 1 Ah receptor mechanism .............................. 4 TCDD equivalence factors ............................. 7 Rainbow trout cell bioassay: advantages and utility ............ 8 OBJECTIVES ......................................... 1 2 METHODS ........................................... 14 Cell culture, plasmids, and transfections .................. 14 Luciferase assays .................................. 17 EROD assays ..................................... 17 Calculations ..................................... 18 RESULTS ............................................ 19 Transient transfections .............................. 19 Stable transfections ................................ 20 Characterization of the RLT 1.0 cell line .................. 21 Comparison of luciferase and EROD induction .............. 24 Comparison of TCDD and PCBs ........................ 25 Comparison of RLT 1.0 TEFs with in viva fish TEFs .......... 29 DISCUSSION ......................................... 33 Transient transfections .............................. 33 Stable transfections ................................ 34 Characterization of the RLT 1.0 cell line .................. 34 Comparison of luciferase and EROD induction .............. 36 Comparison of TCDD and PCBs ........................ 37 Comparison of RLT 1.0 TEFs with in viva fish TEFs .......... 38 Utility of the RLT bioassay ........................... 38 CONCLUSIONS ....................................... 42 APPENDIX A ......................................... 44 APPENDIX B ......................................... 55 APPENDIX C ......................................... 58 APPENDIX D ......................................... 60 LIST OF REFERENCES ................................... 62 vi LIST OF TABLES Table 1. Transient transfection of RTH-149 with pGL2—Control and pGudLuc1.1. Cells were dosed for 24 hr, 2 replicates per treatment. Luciferase activity is expressed as arbitrary light units. .................. 19 Table 2. TEF values calculated from the RLT 1.0 bioassay compared with bioassays using rainbow trout. ......... 32 Table 3. Growth rates of RTH-149 cells seeded in DMEM ......... 55 Table 4. Comparison of transient transfection methods. Cells were transfected with pGL2—Control. Luminescence is in arbitrary units per 20 pL sample. ............... 58 Table 5. Transient transfections of RTG-2 and RTL with the TCDD-inducible plasmid, pGudLuc1.1. Treatments each had two replicates. Luminescence is in arbitrary units per 20 pL sample. ................... 59 Table 6. Stable transfection of RTH-149 produced seven clones. Each was tested for inducible luciferase expression by dosing with 0.1 % DMSO or 1 nM TCDD in DMSO for 24 hr. T6 was exposed for 3 d, and its luminescence is expressed in Iuminometer units/pL. ................................... 60 vii LIST OF FIGURES Figure 1. Structures of several halogenated aromatic hydrocarbons (HAHs). ........................... 3 Figure 2. Proposed Ah receptor mechanism .................... 5 Figure 3. pGudLuc1.1 plasmid. ........................... 15 Figure 4. Relationship between mass of cells and luciferase induction. Protein concentration is used as an index of total cell mass. Cells were treated for 24 hr with 0.1 % DMSO or 1 nM TCDD in DMSO. Bovine serum albumin was used as a protein standard. ......... 22 Figure 5. Relationship between duration of exposure and luciferase induction. Cells were treated with 1 nM TCDD in DMSO. Medium containing 1 nM TCDD in DMSO was renewed 3 d and 7 d before harvesting ................................... 23 Figure 6. Effects of solvent on luciferase induction. Cells were treated with TCDD in DMSO (0.1 %) or isooctane (1 %) for 4 d. Error bars represent standard deviation from the mean. ........................ 27 Figure 7. Comparison of (a) luciferase induction in RLT cells and (b) EROD induction in RTH-149 cells. Duration of exposure to TCDD was 4 d. RTH—149 cells were exposed to TCDD in DMSO. RLT cells were exposed to TCDD in isooctane. .......................... 28 Figure 8. TCDD and PCB dose response curves. Cells were exposed to the test compounds in isooctane for 4 d. ...................................... 31 viii Figure 9. Figure 10. Figure 1 1. Scatchard plots of dose-response curves. Points represent the mean of three samples at each dose. Points at the high and low doses which do not fall in the linear range were not included in calculations, and are shown as open diamonds. .................. 54 Growth rate of RTH-149 cells is dependent on seeding density. Cells were grown in 25 cm2 flasks. ..................................... 57 Changes in the response to TCDD in RLT 1.0 cells. ...... 61 ix INTRODUCTION Halagena ted aroma tic hydrocarbons Halogenated aromatic hydrocarbons (HAHs) are a diverse group of chemicals. Some representative classes of HAHs are the polychlorinated or polybrominated dibenzo-p-dioxins, dibenzofurans, biphenyls, naphthalenes, azobenzenes, and diphenyl ethers (Figure 1). Some were produced for industrial processes, while others are byproducts of combustion or chlorination reactions [1-3]. This chemical family includes many congeners which are Iipophilic, persistent, and toxic [3]. HAHs tend to partition from water into aquatic organisms and become bioconcentrated, because they are not readily metabolized or excreted [3]. HAHs transfer efficiently from prey to predators, causing concentrations of HAHs in organisms to increase with trophic level, or biomagnify. HAHs produce a wide range of toxic effects. Most of these effects involve epidermal tissues, and many effects seem to be the result of altered regulation of cell growth and differentiation. Commonly studied effects include wasting syndrome, immunotoxicity, hepatotoxicity, carcinogenesis, chloracne, reproductive impairment, and mortality [1]. 10 Cl 9 1 CI Cl 9 0 ‘ 2 CI 8 2 .. o e .. Cl 6 0 , Cl 6 o 4 5 5 2,3,7,8—Tetrachlorodibenzo-p-dioxin 2,3,7,8-Tetrachlorodibenzofuran CI Cl 1 2 2 3 CI 7 a 2 Cl .. O O .. 00 Cl ' 5 4 CI 5 6 6 5 3,3',4,4'-Tetrachlorobiphenyl 2,3,6,7-Tetrachloronaphthalene \‘N 1 It 1 3 5 o 2 Cl CI 2 O 2 Cl 3.3'.4.4'-Tetrach|oroazobenzene 3,3',4,4'-Tetrachlorodiphenylether Figure 1. Structures of several halogenated aromatic hydrocarbons (HAHs). 3 These types and intensities of effects of HAHs vary among species. In fish, HAH exposure may cause mortality, wasting syndrome, fin and gill lesions, hepatotoxicity, immunotoxicity, or reproductive impairment [4—7]. Fish eggs exposed to HAHs exhibit increased mortality at hatching and blue sac disease, which results in mortality before the swim-up stage [7-9]. Ah recep tor mechanism HAHs which assume a planar configuration share a common mechanism of action (Figure 2; [10, 11]). Some non-halogenated planar compounds, such as the polycyclic aromatic hydrocarbons (PAHs), also share this mechanism. These compounds bind with varying affinities to a cytosolic protein, the aromatic hydrocarbon receptor (AhR). The AhR tertiary structure is currently unknown, but three regions important to function have been identified. A basic helix-loop-helix (bHLH) domain is involved in DNA binding and dimerization [10, 12]. A PER-ARNT-SIM (PAS) homology region is involved in ligand binding and dimerization [10, 12] A glutamine-rich region is involved in activation of transcription [10, 12]. Once an HAH is bound by the AhR, the liganded receptor complex undergoes a transformation process involving the dissociation of at least a 90 kDa heat shock protein (hsp90l [10]. The activated AhR then translocates to the nucleus [10]. An as yet unidentified protein may be involved in this translocation [10]. Once in the nucleus, the activated AhR forms a dimer with the Ah receptor nuclear translocator (ARNT) protein [10, 13]. Despite being named a translocator protein, ARNT is not required for phosphorylation (7) translocator (? protein phosphorylation changes in gene expression pathway e.g. P4501A1 \ . \ changes In _~ protein activity \ \‘ \ . . \ changes In gene expressuon \ e.g. AP-1 \ \ ¥ \ \ - — -y toxicity Figure 2. Proposed Ah receptor mechanism [10, 11]. 5 translocation, but is required for DNA binding [10]. Phosphorylation of either AhR, or ARNT, or both may be necessary for dimerization and/or DNA binding [10, 14, 15]. The high affinity binding of HAH:AhR:ARNT complex to specific DNA sequences, known as dioxin responsive enhancers (DREs), results in enhanced transcription of the adjacent genes [10]. Mammalian DREs consist of a core sequence, TNGCGTG, surrounded by flanking regions which are also functionally important [16, 17]. Some of the genes induced through the AhR mechanism are involved in metabolism of xenobiotics. One such gene encodes the monooxygenase cytochrome P4501A1, which catalyses the addition of hydroxyl groups to aromatic substrates [18]. This process makes the compounds more water soluble and, thus, more easily conjugated and excreted [18]. Induction of cytochrome P4501A1 can be measured by an increase in 7- ethoxyresorufin-o-deethylase (EROD) activity. An additional mechanism involving the AhR, designated the protein phosphorylation pathway, has recently been proposed [11]. This pathway requires functional AhR but does not require ARNT, and leads to an increase in the phosphorylation of proteins [11, 19]. The protein phosphorylation pathway may interact with growth factor signal transduction pathways [1 1]. The AP-1 transcriptional enhancer appears to be induced through this mechanism [1 1, 19]. The steps in the mechanism of toxicity between activation of the AhR and physiological effects have not been identified. The involvement of the AhR in toxicity is supported by two lines of evidence. First, inbred mouse strains 6 selected for insensitivity to HAHs are deficient in functional AhR [1 , 3, 10]. Second, structure-activity relationships reveal that the potency of an HAH is correlated with the binding affinity of the AhR for that compound [1, 10]. However, evidence has been found for at least two pathways involving AhR: the DRE pathway and the protein phosphorylation pathway [10, 11]. Both pathways may lead to toxicity, and their relative contributions to toxicity are unknown. Most in vitro bioassays for AhR-mediated toxicity, including the RLT 1.0 bioassay, measure the activity of the DRE pathway only. Thus, these bioassays may provide an incomplete measure of the potency of AhR agonists. Future studies are needed to elucidate the relative contributions of the two pathways to toxicity, and to develop methods for measuring the activity of the protein phosphorylation pathway. TCDD equivalence factors TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) is used as a prototypic AhR agonist because it is among the most potent HAHs known, in both induction of AhR-controlled genes and toxicity. The potency of other HAH congeners or mixtures can be related to the potency of TCDD through TCDD equivalence factors (TEFs) [3]. The response of interest, which can be anything from enzyme induction to mortality, is measured at several levels of TCDD exposure. The resulting dose-response relationship is used to calculate an effective dose to produce 50% of the maximal response (EC50). Other levels of response, such as the EC10 or EC90 may also be used, however the EC50 is used most 7 commonly because it can be determined most precisely. The TEF is the ratio of the EC50 for TCDD to the EC50 for the HAH congener of interest. TEFs are useful because HAHs occur in complex mixtures of hundreds of compounds, each with different potencies and occurring at different concentrations. The toxicity of a given mixture can be expressed in TCDD equivalents (TEO), which are calculated by summing the molar concentration of each congener multiplied by its TEF. Rainbow trout cell biaassa y: advantages and utility Here we report on the development and characterization of a bioassay for AhR-mediated toxins. The bioassay uses a genetically engineered rainbow trout liver tumor cell line containing the firefly luciferase reporter gene, the transcription of which is under the control of DREs. The cell line is designated Remodulated Lightning Trout (RLT 1.0). When the cells are exposed to HAHs, they express the luciferase gene. Luciferase activity can be detected as light emission in the presence of substrate. Several methods for studying HAHs are available, including instrumental analysis of the chemicals present in a sample, in viva assays of early life stage mortality [20] or EROD induction in fish [21], and in vitra assays such as the rat H4lle cell EROD assay [22] and the recombinant mouse T13 cell assay [23]. The RLT 1.0 bioassay can provide unique information which is not available through other methods. Instrumental analysis, in viva bioassays and in vitra bioassays each provide information complementary to the others. 8 Together, these methods make up a powerful set of tools for analysis of AhR- mediated toxicity. In vitro bioassays have several advantages relative to in viva bioassays. In vitra bioassays are generally more convenient than in viva bioassays, because cell culture is less labor-intensive than fish culture. Dosing cells with the compound of interest is generally less labor-intensive than dosing fish or fish eggs. In vitra bioassays often yield results more rapidly than in viva bioassays, because the responses of cells generally occur more rapidly than the responses of fish. In vitra bioassays can be designed to measure an endpoint which is a specific indicator of the presence of AhR agonists. Endpoints measured in in viva bioassays, such as mortality, are generally more toxicologically relevant but less specific to AhR-mediated toxins. These characteristics make in vitra bioassays appropriate for screening compounds and environmental samples for AhR-mediated toxins. Instrumental analysis of samples can also be enhanced by in vitra bioassays. Instrumental analysis alone requires the assumption that the toxicity of the mixture of compounds present is additive and that all potentially toxic compounds were included in the analysis. In contrast, bioassays provide an integrative measure of the total toxicity of a sample. They include non-additive interactions between compounds, and compounds which are not measured by conventional techniques. These qualities make bioassays appropriate for studying interactions among toxins, for identifying novel sources of toxicity, and for guiding chemical analysis in fractionation studies. 9 The RTH-149 cell line was chosen for this bioassay for several reasons. The Ah receptor has been detected in this cell line [24]. A functional AhR is a prerequisite for a successful bioassay. Because the RTH-149 cell line is derived from rainbow trout, the results of the RLT 1.0 bioassay will be more relevant to fish than the results of bioassays based on mammalian systems such as the rat H4lle EROD bioassay. Also, the responses of rainbow trout to AhR agonists have been well studied in viva and this data is available for validation of the RLT bioassay [4, 7, 20]. Measuring induction of a reporter gene offers several advantages over measuring the induction of a native HAH-responsive gene. The plasmid used in the RLT cell line can be transfected into cell lines from any species. This allows direct comparisons of the responses of different species. The reporter gene used can be chosen on the basis of the availability of a sensitive, convenient, rapid assay for its expression. Finally, the regulatory region controlling transcription of the reporter gene can be chosen to respond only to activation of AhR. This makes the bioassay useful in studies of the regulation of genes controlled by DREs. Firefly luciferase was chosen as a reporter gene in the RLT bioassay. The assay for luciferase activity is sensitive, convenient, rapid, and can be performed in 96-well plates [25]. The unique reaction, light production, catalyzed by luciferase reduces the chance of problems with high background activity in a cell line. Assays which measure luciferase activity have several advantages over EROD assays. Because cytochrome P4501A1 is involved in 10 xenobiotic metabolism, it recognizes HAHs as substrates. This allows EROD activity to be competitively inhibited by HAHs, which can complicate interpretation of dose-response curves [26]. In addition, it is often useful to inhibit metabolism of HAHs by cytochrome P4501 A1 in order to determine the biologically active form of a compound. Because cytochrome P4501A1 activity cannot be measured in these studies, reporter genes such as luciferase are useful. l1) (2) (3) (4) OBJECTIVES Develop a rainbow trout cell bioassay for AhR-mediated toxins by performing a stable transfection of the RTH-149 rainbow trout hepatoma cell line with the pGudLuc1.1 plasmid, which contains the luciferase reporter gene under the transcriptional regulation of DREs. Characterize the recombinant cell line by estimating the discrimination power of the bioassay and investigating three variables which may affect luciferase induction: degree of confluence of the cells, duration of exposure to the test compound, and solvents used to deliver the test compound. Establish the relationship between reporter gene induction and native gene induction by comparing EROD induction in the parent cell line with luciferase induction in the recombinant cell line. Establish the relevance of the reporter gene bioassay to HAH toxicity in fish by comparing the responses of the bioassay to TCDD and several PCB congeners with previously reported in viva responses of fish. 11 METHODS Cell culture, plasmids, and transfec tians RTH-149 cells (ATCC 1710, American Type Culture Collection, Rockville, MD) were grown in basal Eagle’s medium (Gibco/BRL, Grand Island, NY) supplemented with 10 % fetal bovine serum (HyClone Laboratories, Logan, UT) in an enriched CO2 atmosphere at 21°C. For details of cell culture and other methods, see Appendix A. RTH-149 growth characteristics are shown in Appendix B. The pGL2-Control plasmid (Promega, Madison, WI) contains the firefly luciferase gene driven by an SV40 promoter and enhancer. It was used as a positive control in transient transfections. The plasmid pGudLuc1.1 was a gift of Mike Denison, University of California at Davis (Figure 3). This plasmid was constructed by inserting the 1810 bp Hindlll fragment found in pMpap1.1 and pMcat5.9 [23, 27] into the Hindlll site of pGL2-Basic (Promega, Madison, WI) upstream of the luciferase coding region. The insert contains a 1.3 kb fragment of the long terminal repeat from the mouse mammary tumor virus (MMTV-LTR), which contains the viral promoter but not the glucocorticoid responsive element (GRE) normally present in the MMTV-LTR [28]. A 482 bp fragment of the regulatory region of the mouse cytochrome P4501A1 gene containing four 12 13 Ampr polylA) signal ori MMTV promoter l A ' I DO Yl IS'Qna pGudLu01.1 SV40 intron 7380 bp DREs 'MTV promoter firefly luciferase Figure 3. pGudLuc1.1 plasmid. 14 dioxin responsive enhancers (DREs) is inserted 100 bp upstream of the MMTV- LTR promoter start site [27, 29, 30]. Previous studies have shown that activity of a reporter gene controlled by this MMTV-LTR/DRE insert is dependent on the presence of TCDD, functional AhR in the cell line, and DRE sequences in the regulatory region [23]. Plasmids were introduced into the RTH-149 cells by a polybrene transfection with a DMSO shock to increase efficiency [31, 32]. Polybrene was the most effective method tested with this cell line (Appendix C). DEAE- dextran, calcium phosphate, and Iipofectin® were also tested (Appendix C). In transient transfections, cells were transfected with 10 pg vector, dosed with the compound of interest 48 hr after the DMSO shock, and harvested for luciferase assays after 24 hr exposure to the test compound. Cells were dosed with the compound of interest dissolved in DMSO or isooctane. DMSO was used at a final concentration of 0.1 % in medium; isooctane was used at a final concentration of 1 % in medium. In stable transfections, cells were cotransfected with 7 pg pGudLuc1.1 and 3 pg pSVzneo per 60 mm plate [33]. Cells were subcultured at a 1:30 dilution in medium containing 500 mg/L geneticin (G418) 3 d after transfection. The G418 concentration was raised to 1000 mg/L after 10 d. Colonies of surviving cells were isolated, grown and screened for HAH-inducible luciferase activity. 15 Luciferase assays Cells were rinsed twice with phosphate buffered saline (PBS), then harvested with 100 pL cell lysis buffer (Promega, Madison, WI) per 60 mm plate or 30 pL per well in 96 well plates. 20 pL samples of the cell lysates were analyzed for luciferase activity using a Iuminometer and luciferase assay reagent (Promega, Madison, WI). In the transient transfection, protein, and time course experiments, a TD-20e Iuminometer (Turner Designs, Sunnyvale, CA) was used. For all other experiments, an ML 3000 Microtiter® plate Iuminometer (Dynatech Laboratories, Chantilly, VA) was used. Light production was expressed in arbitrary luminescence units. For the time course experiment, light production was normalized to protein concentration, as an index of total cell mass. Protein concentration was measured using a Bradford dye-binding assay (Bio-Rad, Melville, NY) [34]. EROD assays The EROD assay was performed using the methods of Tillitt et al. [22]. with modifications. RTH-149 cells were grown and exposed to TCDD in DMSO (0.1 % in medium) in 60 mm plates. Plates were washed twice with PBS and harvested by scraping. Harvested cells were centrifuged, and the pellets were frozen at -70°C. Cells were thawed in TRIS (trislhydroxymethyl)aminomethane) buffer containing 40 pM dicumerol, an inhibitor of diaphorase, which degrades resorufin [35]. Cells were then disrupted by sonication (25 pulses, 60 % duty cycle, output control 7). The disrupted cell solutions were transferred to 16 individual wells of a 96-well plate. The samples were warmed to 30°C after 7-ethoxyresorufin was added to the cell lysate. NADPH was added to initiate the EROD reaction, which was allowed to proceed for 60 min at 30°C. Resorufin production was quantified by fluorescence (excitation 530 nm, emission 590 nm) on a CytoFluor 2300 fluorometer (Millipore, Bedford, MA). Calculations Dose-response curves were analyzed using Scatchard plots to calculate EC505. The EC505 were used to calculate TEF values, by taking the ratio of the EC50 for TCDD to the EC50 for the compound of interest. For compounds which did not produce the same maximum induction as TCDD, the TEF was based on the EC25 (Appendix A). Power analysis was used to estimate the ability of the bioassay to distinguish between induced and control treatments [36]. Variances of control (002) and treatment (0,2) groups were measured in a single experiment. The type I error rate, a, was set at 0.05 and the type II error rate, 3, at 0.20. The sample size per treatment (n,) was 3. The smallest detectable difference in means (6) was calculated using equation (1) [36]. 2 2 2 O=J (2°.os+zo.2o) (0043:) (1) n u l RESULTS Transient transfec tians The functions of the luciferase reporter gene, the MMTV-LTR and SV40 promoters, and the mouse DREs in RTH-149 cells were investigated by transient transfection experiments. Cells were transfected with one of the following treatments: no DNA, pGL2-Control, or pGudLuc1.1. Transfected cells were dosed with 0.1 % DMSO or 1 nM TCDD in DMSO (Table 1). Cells transfected with no DNA had no luciferase expression. Cells transfected with pGL2-Control expressed luciferase. There was no difference between the DMSO and TCDD treatments in the pGL2-Control-transfected cells. Cells transfected with pGudLuc1.1 expressed luciferase in both DMSO and TCDD treatment groups. Table 1. Transient transfection of RTH-149 with pGLZ-Control and pGudLuc1.1. Cells were dosed for 24 hr, 2 replicates per treatment. Luciferase activity is expressed as arbitrary light units. treatment plasmid DMSO (0.1 %l 1 nM TCDD in DMSO :0 treatment 0.035 :I: 0.013 NT' pGL2-Control 0.268 :I: 0.136 0.309 :t 0.216 JGudLuc1.1 0.255 :t 0.182 0.798 :t 0.317 ' not tested 17 18 However, the TCDD treated cells exhibited two-fold greater luciferase activity than cells treated with DMSO only. Stab/e transfections RTH-149 cells were cotransfected with pSVZneo and pGudLuc1.1. One month after the transfection, ten colonies of cells survived G418 selection. Because approximately 20 to 25 million cells were transfected, the chance of a cell taking up the G418 resistance gene and inserting it into the genome in an interruptible region was estimated to be approximately 1 in 2 million. Seven of the colonies survived to be tested for luciferase expression and induction (Appendix D). Three clones exhibited relatively great constitutive luciferase expression and some induction. Three clones did not have detectable background luciferase expression, and two of these clones exhibited detectable induced luciferase expression. One clone (RTH-149-T1) exhibited relatively little background luciferase expression and approximately 2.5 fold induction after exposure to 1 nM TCDD for 24 hr. RTH-149-T1 was designated Remodulated Lightning Trout (RLT 1.0) and was used in all subsequent luciferase bioassays. The RLT 1.0 cell line exhibited changes over time in the shape of its dose response curve for the luciferase reporter gene and in the maximum luciferase induction produced by TCDD treatment (Appendix D). Because these changes were likely to be the result of a mixed cell population, the cell line is currently being reisolated to correct this problem. The cell line reisolated from RLT 1.0 will be designated RLT 2.0. 19 Characterization of the RL T 1.0 cell line Characterization of the RLT 1.0 cell line included investigation of the relationship between total cell mass and induction, duration of exposure and induction, and solvents and induction. Protein concentration was used as an index of total cell mass. Luciferase activity increased in a linear relationship I with protein concentration for both TCDD and DMSO treated cells, over the entire range tested (Figure 4). The time course for luciferase induction in these cells with 1 nM TCDD is shown in Figure 5. Induction increases in a linear relationship with time up to 4 d of exposure. After 4 d, induction begins to level off and variability increases. Thus, cells were exposed to test compounds for 4 d in all subsequent experiments. DMSO and isooctane were compared as solvents to deliver the test compounds to the cells. RLT 1.0 cells treated with TCDD delivered in these two solvents produced similar dose response curves (Figure 6). Comparison of luciferase and EROD induction In order to compare the activity of the new recombinant cell line reporter gene with the activity of a well characterized native inducible gene, luciferase activity and induction in the RLT 1.0 cell line were compared to EROD activity and induction in the parent cell line, RTH-149 (Figure 7). Background EROD activity and induction above background are small in the RTH-149 cell line; in fact, EROD activity was undetectable at TCDD concentrations below 0.1 nM. EROD activity was close to the detection limit of the assay and variable 20 200 O 180 " v 160 P 140 ' 120 " 100 " luminescence 80' 60' 40' 20* 0 I I I I I I I I I 0 10 20 3O 40 50 60 70 80 90100 protein (ug) Figure 4. Relationship between mass of cells and luciferase induction. Protein concentration is used as an index of total cell mass. Cells were treated for 24 hr with 0.1 % DMSO or 1 nM TCDD in DMSO. Bovine serum albumin was used as a protein standard. 21 14 . i' 12 - I 1” o/l .5 ./l m . / .h E 10 0 a. / 3’ o \ 8 " .- G) 2 :/ Q) 8 a: 6 ' / .E o E /1 _ 4 _ O 2. ./ 9/ O I I I J I I I I I I I 012345678910 time (clays) Figure 5. Relationship between duration of exposure and luciferase induction. Cells were treated with 1 nM TCDD in DMSO. Medium containing 1 nM TCDD in DMSO was renewed 3 d and 7 d before harvesting. 22 0.012 —0- DMSO 0.010 -V- isooctane 0.008 ' /'-:'- 0.006 r I 0£m4'- ¥///1 - 3 i4. luminescence 10'4 10'3 10'2 10‘1 10° 101 1O2 TCDD(nM) Figure 6. Effects of solvent on luciferase induction. Cells were treated with TCDD in DMSO (0.1 %) or isooctane (1 %) for 4 d. Error bars represent standard deviation from the mean. 23 a) 0.016 0.014 " .IL 0.012 - / o o d o I H\ IOI luminescence O O o co l 0.006 '- 0.004 - I E/J. 0.002 - o 000 L Lanna“! . a nnlnnnl A 444.“1L A n lllml j A 1...“! J . AAAAJ 0.0001 0.001 0.01 0.1 1 10 100 b) 1.0 I 0 A08 r 3 h g 0.6 - o 2 3 . go 4 T J D o 0.2 - 0 (I Lu 0.0"- _ 0.0001 0.001 0.01 0.1 1 10 100 TCDD concentration (nM) Figure 7. Comparison of (a) luciferase induction in RLT cells and (b) EROD induction in RTH-149 cells. Duration of exposure to TCDD was 4 d. RTH-149 cells were exposed to TCDD in DMSO. RLT cells were exposed to TCDD in isooctane. 24 throughout the dose response curve. In contrast, luciferase activity in the RLT 1.0 cell line was well above the detection limit of the assay with little variability. The luciferase assay was more convenient than the EROD assay, because the luciferase assay could be performed on cells grown and lysed in a 96-well plate. Luciferase and EROD induction also showed differences in the shapes of their dose-response curves. Both responses had thresholds between 0.01 and 0.1 nM TCDD. However, EROD induction reached a maximum at 1 nM, while luciferase induction reached a maximum at 10 nM. EROD induction declined slightly after the maximum, while luciferase induction levelled off after the maximum. Thus, the dynamic range of the luciferase assay was approximately 10 fold greater than that of the EROD assay in RTH-149 cells. Comparison of TCDD and P683 In order to assess the effects of TCDD and other AhR agonists in the RLT bioassay, TCDD and four PCB congeners were tested (Figure 8). The PCB congeners included two coplanar congeners (3,3’,4,4',5-pentachlorobiphenyl, IUPAC number 126, and 3,3’,4,4’-tetrachlorobiphenyl, IUPAC number 77), which are generally strong inducers of AhR-mediated responses, and two mono- artha- substituted congeners (2,3’,4,4’,5-pentachlorobiphenyl, IUPAC number 118, and 2,3,3',4,4’-pentachlorobiphenyl, IUPAC number 105) which are weak inducers because they do not readily assume a planar conformation. PCB 126 induced luciferase activity to the same maximum induced by TCDD. PCB 77 induced luciferase activity to only half the maximum induced by TCDD. PCB 25 0.016 -o- TCDD 0.014 ' -I- PC8126 -o- PCB 77 0.012 - -A- PCB118 m 0.010 ' 3 T 8 o 3 0.008 ' .E '5' — 0.006 ' 0.004 ' I e s/L 0.002 ' 0.000 * 10'3 10‘2 10‘1 10° 101 102 103 104 concentration (nM) Figure 8. TCDD and PCB dose response curves. Cells were exposed to the test compounds in isooctane for 4 d. 26 1 18 and PCB 105 did not induce luciferase activity at any concentration tested (Figure 8; PCB 105 data not shown.) The discrimination power of the RLT 1.0 bioassay was determined by a power analysis [36]. Because the RLT 1.0 cell line is actually a mixed population, data from a single experiment was used. The mean of 35 blank and solvent control treatments was 0.0031 luminescence units per 20 pL sample, and the variance was 2.6 x 10". The mean variance of the 22 treatment groups was 7.9 x 10". The least detectable difference in means was 0.0015 luminescence units. This is equivalent to 1.5 times the control. A TCDD concentration of 22 pM is predicted to produce this response. Comparison of RLT 7.0 TEFs with in vivo fish TEFs TEF values were calculated from the dose response curves for TCDD and the PCB congeners (Table 2). The relative potencies of TCDD and PCB congeners were similar for the RLT 1.0 bioassay and two measures of responses in whole fish, EROD induction and early life stage mortality [7, 21]. In all three assays, PCB 118 and PCB 105 failed to induce any response. PCB 126 was approximately 10 times more potent than PCB 77. However, the TEFs calculated from the RLT bioassay for PCB 126 and PCB 77 were approximately 10 fold greater than those calculated from the whole fish bioassays. 27 Table 2. TEF values calculated from the RLT 1.0 bioassay compared with bioassays using rainbow trout. juvenfle EC50 rainbow trout early life stage (nM) (pg/L) TEF EROD TEFa mortality TEF" TCDD 0.142 ' 0.046 , 1.0 1.0 1.0 PCB 126 5.34 1.8 0.027 0.0014 0.005 PCB 77 14.7 4.3 0.0032c 0.0006 0.00016 PCB 1 18 > 4740 > 1550 <0.00003 <0.00006 <0.00007 PCB 105 > 6230 > 2040 < 0.00002 < 0.00005 < 0.00007 ' from Newsted et al. [21] b from Walker and Peterson [7] ccalculated from EC25 DISCUSSION Transient transfec tians The function of the pGudLuc1.1 reporter system in the RTH-149 cell line was first determined by transient transfection studies. The cells were able to produce functional luciferase, and their growth was not obviously affected (Table 1). The SV40 and MMTV-LTR promoters both supported transcription of the luciferase gene in RTH-149 cells (Table 1). The fragment of the MMTV- LTR promoter used in pGudLuc1.1 was chosen because it lacks the endogenous MMTV glucocorticoid response element (GRE) [28]. Assuming it contains no cryptic regulatory elements, it should allow transcription of the reporter gene to depend only on binding of the DREs by liganded AhR complex. Thus, bioassays based on the reporter gene may be used in studies of the mechanism of regulation of AhR-induced genes. Differences in induction of the reporter gene from native genes reflects differences in their regulation. The DREs from the regulatory region of the mouse cytochrome P4501 A1 gene were recognized by the fish AhR (Table 1). This demonstrates that the RTH-149 cell line expresses AhR, as previously shown by Lorenzen and Okey [24]. These results also imply that the DRE sequence, and the DNA binding 28 29 region of the AthARNT complex are conserved among species as distantly related as trout and mouse. The transient transfections produced small, variable luciferase expression (Table 1). This could be caused by either poor efficiency of luciferase expression in these cells, or poor transfection efficiency. Thus, transient transfections are not appropriate for routine assays using this cell line. Stable transfec tians Uptake and stable incorporation of introduced DNA into the RTH-149 cell genome is a rare event. The differences in luciferase expression among the isolated clones illustrates the importance of factors unique to each clone in influencing reporter gene expression. These factors could include the number of plasmids inserted, their orientation relative to each other and to host sequences, and the surrounding region of host DNA. The changes in RLT 1.0 must result from one or more recombination events which changed one or more of these factors. The differences among clones also point out that the reporter gene is not always free from regulation by mechanisms other than DREs. However, it should be relatively free from other regulation compared with native genes. Characteriza tian of the RL T 1. 0 cell line Characterization of the newly isolated recombinant cell line established information needed in planning the comparisons of luciferase and EROD activity 30 and the TCDD and PCB dose-response relationships. Protein concentration was used as an index of total cell mass. If the average cell mass is not affected by the test compounds, protein is also correlated with cell density and the degree of confluence of the cells. The linear relationship between protein concentration and luciferase induction demonstrated that the degree of confluence of the cells does not affect induction, and that induction can be normalized to protein in order to reduce variation (Figure 4). However, the protein assay was not readily adaptable for use with the 96—well plate reading Iuminometer. This problem was minimized by seeding the wells with aliquots from a well-mixed cell suspension in order to reduce the variation in cell density among walls. This approach allows variation in cell mass to affect results. Future studies are needed to develop a measure of cell mass appropriate for use with the luciferase assay in 96 well plates. The time course was used to determine the duration of exposure used in subsequent experiments. A duration of 4 d was chosen because it was in the linear range of the curve, with relatively great induction, but little variation (Figure 5). The relative induction and variation are important in determining the method detection limit of the bioassay. The solvents used to deliver test compounds to the cells did not affect induction (Figure 6). Neither solvent caused induction or obvious effects on cell growth at the concentrations used. Solvents could effect induction either by changing the kinetics of delivery of the test compound to the cells, or by 31 directly affecting the cells. Effects of solvent on induction have been observed in assays of EROD activity in rat H4lle cells [22]. Comparison of luciferase and EROD induction The RLT bioassay was compared with an assay of EROD induction in the parent cell line, RTH-149 (Figure 7). The two induction curves have similar thresholds between 0.01 and 0.1 nM. This suggests that the threshold of induction for both genes is dependent on activation of the AhR. EROD induction reaches a maximum at a TCDD concentration of 1 nM, then declines slightly. However, luciferase induction continues to increase until it reaches a maximum at 10 nM TCDD. This increase is consistent over many dose- response relationships measured for the RLT 1.0 cell line (Appendix D). The increase in luciferase activity between 1 and 10 nM TCDD can be interpreted as an increase in the level of liganded AhR. The lack of a response in EROD activity to this increase in liganded AhR indicates the presence of another regulatory mechanism affecting EROD activity. Previous studies have shown that EROD activity is inhibited by HAHs [26]. This inhibition could be the reason for the maximum and decline in EROD activity. This comparison between luciferase and EROD induction illustrates the utility of the RLT bioassay in mechanistic studies to detect differences in the regulation of genes induced by HAHs. 32 Comparison of TCDD and PCBs The dose response relationships for TCDD and various PCB congeners were similar to those seen in tests of early life stage mortality in rainbow trout (Figure 8; [7]). As expected, TCDD and the non-ortho-substituted PCBs induced luciferase activity. However, there was no response to the mono- ortho—substituted PCBs, which do not produce early life stage mortality in rainbow trout [7]. This is an important difference between fish and mammalian systems. Mono-ortho-substituted PCBs can induce responses in mammalian systems [22]. Given the abundance of these congeners in environmental mixtures, they may be a significant source of AhR-mediated toxicity to mammals, and a significant source of error in using TEFs derived for mammals to predict toxicity to fish. Walker et al. [7] compared the potency of various HAHs calculated from assays of early life stage mortality in rainbow trout to measures of potency calculated from a rat H4lle cell bioassay. They found that the rat cell bioassay underestimated the potency of polychlorinated dioxins to fish, and overestimated the potency of PCBs [7]. PCB 77 did not produce the same maximum response as TCDD (Figure 8). This suggests that PCB 77 is a partial AhR agonist. Another possibility is that in the cells, PCB 77 is metabolized to a form that does not bind AhR or that partitions out of the cells into the medium. However, the RTH-149 cells have low EROD activity, which suggests that they would not efficiently metabolize HAHs. Future studies are needed to address this question. 33 Comparison of RL T 1.0 TEFs with in vivo fish TEFs The RLT 1.0 bioassay is not an appropriate tool for the development of TEFs for use in ecological risk assessment. The bioassay does not account for the pharmacokinetics and distribution of HAHs in fish, or for fish physiology. Presumably, there are several steps between induction of AhR-activated genes and mortality. These steps are currently undefined, and are certainly not modelled by the RLT bioassay. These differences between fish and fish cell cultures probably account for the differences in TEF values calculated using the two systems (Table 2). The comparison of TEFs calculated from the RLT bioassay with TEFs calculated from in viva fish bioassays was used to validate the RLT bioassay. Because the pattern of HAH potencies was the same for the RLT bioassay as for fish, the bioassay can be used to predict the responses of fish. Future studies including tests of a wider range of HAH congeners are needed to further define the relationship between potency predicted by the RLT bioassay, and actual potency in fish. Utility of the RLT bioassay Because the RLT bioassay can predict AhR-mediated toxicity to fish, it can be useful in a variety of applications. The bioassay can be used to test compounds or extracts in situations in which in viva testing is impractical. It can also be used as a supplement to chemical analysis. Finally, it can be used to study the mechanism of action and the interactions of AhR-mediated toxins. 34 The RLT bioassay is an appropriate alternative to in viva testing. The rainbow trout egg injection early life stage mortality assay developed by Walker et al. [20] offers the advantages of realism, measurement of a toxicologically relevant endpoint, and delivery of a known amount of the test compound to the test organism. Early life stage mortality refers to the syndrome exhibited by rainbow trout and other salmonids when exposed to TCDD as eggs [7]. TCDD exposure causes increased mortality at hatching and development of blue-sac disease, which results in mortality before the swim-up stage [7-9]. Early life stage mortality is very sensitive to TCDD, with an LD50 of 400 pg/g. However, this test is more labor-intensive than the RLT bioassay. The egg injection assay requires 4 hr of work by two investigators to inject the eggs for one dose- response study [20], while the RLT bioassay requires less than 15 min to dose the cells for a study. Also, results from the egg injection study are obtained 45 to 60 d after dosing [20], while results from the RLT bioassay are obtained 4 d after dosing. The RLT bioassay is appropriate for use as a screening tool which could test large numbers of samples for their potential toxicity to fish, and identify samples and compounds to be further tested in the egg injection assay. Instrumental analysis of extracts and effluents can be enhanced by the RLT bioassay. Prediction of total AhR-mediated toxicity of a sample by chemical analysis can be confounded by two factors. First, interactions among HAHs may lead to greater or lesser toxicity than predicted from an additive model. Second, some compounds which act through the AhR are not measured 35 in routine analysis, because standard are unavailable or because they have not yet been identified. The RLT bioassay can act as a check to insure that chemical analyses have identified all the relevant components of the mixture, that is, it can be used to check for a mass balance of toxicity units. It can also be used to test defined mixtures in order to identify and characterize interactions involving AhR-mediated toxins. In fractionation studies, the components of a complex mixture are separated according to their chemical properties. The RLT bioassay can be used to screen the separated fractions and direct further chemical analysis to identify sources of AhR-mediated toxicity. For example, pulp and paper mill effluents have recently been found to contain components which induce EROD activity and disrupt reproductive cycles in fish but are neither persistent nor chlorinated [37-40]. The RLT bioassay could be useful in identifying the compoundls) responsible for these effects. It could be used to screen effluents, fractions of effluent, and putative AhR ligands found in the effluent through chemical analysis. The RLT bioassay is also well suited for use in studies of the mechanism of action of AhR-mediated toxins to fishes. Because it is relatively free from regulation compared with native genes, the RLT bioassay can be used to detect regulatory mechanisms other than the AhR which act on native induced genes. For instance, in this study the RLT bioassay was able to show that EROD activity is inhibited at high TCDD concentrations, and that this inhibition is not due to saturation of the AhR. Because the RLT bioassay measures induction of the luciferase reporter gene, it can be used in studies with metabolism 36 inhibitors which inhibit EROD activity. These studies can be used to determine which form of a compound, the parent form or a metabolite, is active, and to determine if a compound is readily metabolized in fish liver cells. In summary, the RLT bioassay is useful as a screening tool which provides an integrative measure of the total AhR-mediated toxicity of a sample to fish. It can be used in conjunction with in viva bioassays and with chemical analysis. In mechanism studies, it provides an index of the activation of the Ah receptor. Finally, the same plasmid can be used to develop recombinant cell lines from different species, to allow direct comparisons of different species sensitivities to AhR-mediated toxins. (1) (2) (3) CONCLUSIONS The RTH-149 cell line was stably transfected with pGudLuc1.1. The new recombinant cell line is designated Remodulated Lightning Trout (RLT 1.0). The luciferase reporter gene, driven by either the MMTV or SV40 promoter, is functional in RTH-149 cells. DREs from the 5’ regulatory region of the mouse cytochrome P4501A1 gene are recognized by fish AhR. This demonstrates the conservation among species of the DRE and DRE binding region of the AhR:ARNT complex. Characterization of the RLT 1.0 cell line revealed that the estimated detection limit of the RLT 1.0 bioassay for TCDD was 22 pM, induction of luciferase in RLT 1.0 cells was not dependent on degree of confluence of the cells or on the presence of DMSO or isooctane, and a duration of exposure to TCDD of 4 d produced relatively great induction with little variation. Induction of luciferase and induction of EROD by TCDD exhibited similar threshold concentrations, but differed in the shapes of their dose- 37 (4) 38 response relationships. This reveals differences in the regulation of the two enzymes. The pattern of potencies among TCDD and four PCB congeners measured by the RLT bioassay was similar to the pattern measured by in viva fish bioassays. The RLT bioassay is a useful tool for screening environmental extracts, guiding chemical analysis, and studying the AhR mechanism. APPENDICES APPENDIX A Detailed methods Cell culture This project used three cell lines derived from rainbow trout: RTH-149 (rainbow trout hepatoma), RTG-2 (rainbow trout gonad), and RTL (rainbow trout liver). RTH-149 (ATCC 1710) was derived from an aflatoxin-induced trout liver parenchymal cell tumor in an adult female [41, 42]. RTG-2 (ATCC CCL55) was derived from embryonic trout gonad [43]. The Ah receptor has been detected in the RTH-149 and RTG-2 cell lines [24, 44]. RTH-149 cells were grown in Dulbecco’s modified Eagle’s medium (Sigma Chemical Company, St. Louis, MO) or basal Eagle’s medium (Gibco/BRL, Grand Island, NY) supplemented with 10 % fetal bovine serum (HyClone Laboratories, Logan, UT). RTG-2 cells were grown in basal Eagle’s medium supplemented with nonessential amino acids (Gibco/BRL, Grand Island, NY) and 10 % fetal bovine serum. RTL cells were grown in Eagle’s minimum essential medium (Gibco/BRL, Grand Island, NY) buffered with HEPES (4—(2- hydroxyethyI)-1-piperazineethanesulfonic acid) and supplemented with MEM vitamin solution (Gibco/BRL, Grand Island, NY) and 10 % fetal bovine serum. All cells were grown at 21°C. RTH-149 and RTG-2 cells were grown in 39 40 medium with bicarbonate buffer, and were kept in an enriched CO2 atmosphere. All cell culture reagents were sterilized in an autoclave or by filtration before use. Plates of confluent cells were split by treatment with trypsin. Plates were first rinsed twice with PBS (phosphate buffered saline - 137 mM NaCl, 2.7 mM KCI, 12 mM NaZHPO4, 1.5 mM KHZPO4), then incubated for 10 to 15 min in 0.05 % trypsin-EDTA (Gibco/BRL, Grand Island, NY). An equivalent volume of medium was added to the trypsin solution immediately after the incubation. The cells were suspended and mixed, then the cells were added to the new plate(s) and the medium was renewed. Cells were stored in liquid nitrogen for future use. Cells were first treated with trypsin as described above. The cell suspension was centrifuged and the cell pellet was resuspended in 1 mL freezing medium (80 % fetal bovine serum, 20 % DMSO) and transferred to a cryogenic vial. The vials were packed in cotton and stored at -70°C for 2 d before transfer to liquid nitrogen. Cells were thawed by warming the vial in a 37°C water bath for less than 1 min, than allowing the frozen contents to drop into a flask which contained 24 mL fresh medium at room temperature. Medium was changed within 24 hr after thawing. Cell densities were determined by counting cells in a hemacytometer. Cells were treated with trypsin and suspended in culture medium. A 100 pL sample of the cell suspension was mixed with 20 pL trypan blue and added to the counting chamber. The trypan blue dye is excluded from viable cells. Cells 41 were counted in 5 squares of the hemacytometer. The average number of viable cells/square was multiplied by 1.2 to correct for the volume of trypan blue. The volume of one square is 0.1 pL, because the area of a square is 1 mm2 and the depth of the chamber is 0.1 mm. The corrected average number of cells per square was multiplied by 104 to convert from cells/0.1 pL to cells/mL. Plating efficiency of seeded cells was calculated by taking the ratio of the number of cells counted 17 to 24 hr after seeding to the number of cells seeded. The growth rate (k’) and doubling time (G) of the cells were calculated by equations (2) and (3) [45]: _ (logN - IogNo) ‘ (2) N0 = initial cell number, N = final call number, t = time kl .'£9_2_ G k’ (3) Plasmids Plasmids were grown in E. coli DH5 cells, and isolated by either a polyethylene glycol (PEG) precipitation method or a cesium chloride gradient method. Identity and purity of the isolated plasmids were confirmed using restriction enzyme digestion and agarose gel electrophoresis. In the PEG precipitation method, cells were lysed with lysozyme. Proteins and linear DNA were then denatured with NaOH-SDS (sodium dodecyl 42 sulfate) [46]. The denatured molecules were precipitated by neutralizing in high osmolality (sodium acetate) solution [46]. The supernatant was treated with RNase to remove RNA. Protein was removed by extraction with a 1:1 phenolzchloroform mixture. The aqueous phase, containing plasmid DNA and remaining genomic DNA, was precipitated with ethanol. The pellet was resuspended and plasmid DNA was precipitated with 13 % PEG (MW 8,000) [47]. Remaining impurities were removed by washing the plasmid DNA with 70 % ethanol. In the cesium chloride gradient method, calls were lysed, and proteins and linear DNA denatured with NaOH-SDS [46]. The denatured protein and DNA were precipitated by neutralizing in high osmolality solution [46]. Nucleic acids in the supernatant were precipitated with isopropanol, then separated on a continuous CsCl gradient containing the intercalating dye ethidium bromide [48]. The supercoiled plasmid DNA band was isolated and ethidium bromide removed by extraction with isopropanol saturated with NaCI and water. CsCl was removed by dialysis with TE buffer, and salt was removed by ethanol precipitation. Plasmid identity, concentration and purity were determined by measuring optical density and by agarose gel electrophoresis. Concentration was determined by measuring the optical density of the DNA solution at 260 nm. The extinction coefficient of nucleic acids is 0.02 absorbance unit/pg/mL. Purity of the plasmid preparation was assessed by agarose gel electrophoresis 43 and visualization with ethidium bromide [49, 50]. Identity of the isolated plasmid was confirmed by digestion with restriction enzymes [50]. Transfections Several transfection methods were investigated, including polybrene, DEAE-dextran, calcium phosphate, and liposome. The polybrene method is simple, reliable, and easily manipulated [31]. Cells were first incubated with a mixture of polybrene and the plasmid of interest. Polybrene's positive charges apparently attract the phosphate groups on DNA and the cell membrane. After an incubation of 6 hr, the DNA solution was removed and the cells were exposed to 25 % DMSO in culture medium for 4 min. The DMSO shock increases the efficiency of transfections, apparently by causing disruptions of the cell membrane through which membrane-associated DNA may enter the cell [32]. The cells were then rinsed twice in PBS and returned to regular medium. Liposome transfections were performed using Iipofectin® reagent (Gibco/BRL, Grand Island, NY), and lipofectin® protocols. DNAzliposome complexes were formed by mixing 20 pL lipofectin® reagent in 100 pL serum- free medium with 10 pg plasmid DNA in 100 pL serum-free medium and incubating for 15 min. Lipofectin® reagent forms complexes with DNA spontaneously and with high efficiency [51]. The complexes were then added to 0.8 mL serum-free medium and layered onto PBS-washed 60 mm plates of cells. Cells were incubated with the DNAzliposome complexes for 6 hr. The 44 liposomes become incorporated into the cell membranes [51]. After the incubation, the DNA solution was removed and regular medium was added to the plates. DEAE—dextran transfections used 5 pg/mL plasmid DNA and 250 pg/mL DEAE-dextran in serum-free medium [32]. 1.5 mL of the DNA-DEAE-dextran solution was added to each 60 mm plate of cells. After a 1 hr incubation, 3.5 mL of 52 pg/mL chloroquine in medium with serum was added to the plates. Chloroquine may inhibit the degradation of plasmid DNA by transfected cells [52]. After an additional 5 hr incubation, the transfected cells were washed, and returned to regular medium. DEAE-dextran is not a suitable method for producing stably transfected cell lines [32]. For calcium phosphate transfections, a DNAzcalcium phosphate precipitate was formed by mixing 880 pL of 40 pg/mL plasmid DNA in TE", 1 mL 2X HBS (HEPES buffered saline - 280 mM NaCl, 10 mM KCI, 1.5 mM NazHPO4, 12 mM dextrose, 50 mM HEPES), and 124 pL 2M CaCIz. The precipitate was allowed to form for 30 min, then the mixture was resuspended and added to 60 mm plates of cells (0.5 mL precipitate in 5 mL medium). After a 5 hr incubation, the medium was removed and 1.5 mL 15 % glycerol in 1X HBS was added [32]. After a 1 hr glycerol shock, the cells were washed and returned to normal medium. 45 Luciferase assays Firefly luciferase is a 550 amino-acid protein with a predicted molecular weight of 60,746 Da and an apparent molecular weight of 62,000 Da [25]. Luciferase catalyzes a reaction which requires Mg“, and uses luciferin, ATP or coenzyme A (CoA), and 02 to produce oxyluciferin, C02, and light [25, 53, 54]. The light produced is yellow-green, with an emission maximum at 560 nm [25]. Luciferase alone is a relatively unstable enzyme, probably because it contains a high percentage of hydrophobic residues; however, its stability is increased by adding surfactants and other hydrophobic proteins in solution [55]. Luciferase activity was assayed in cells after harvesting by rinsing twice with phosphate buffered saline (PBS), then adding 1X cell lysis buffer (Promega, Madison, WI). The cell lysis buffer contains triton detergent, which disrupts cell membranes, and buffers which stabilize the luciferase enzyme. Samples (20 pL) of the cell lysates were then analyzed on a Iuminometer. Luciferase assay reagent (Promega, Madison, WI) was added and light emmision was measured. Luciferase assay reagent contains the luciferase substrates luciferin and coenzyme A. Using coenzyme A as a substrate produces steady production of light which lasts several minutes instead of the short flash of light observed in assays using ATP [53, 54]. One of two luminometers was used in these experiments. The TD-20e Iuminometer (Turner Designs, Sunnyvale, CA) reads samples in cuvettes. After a 30 s delay, it integrates light production for 60 s. The ML 3000 Microtiter® plate Iuminometer (Dynatech Laboratories, Chantilly, 46 VA) reads samples in opaque 96-well plates. After a 5 s delay, it integrates light production for 30 s. It maintains samples at a constant temperature of 30°C, which is greater than the optimal temperature for efficient luciferase activity of 20-25°C. However, because luciferase activity is temperature dependent, it is important to conduct assays at a constant temperature [56]. Luciferase denatures at 40°C [55]. Stable Transfec tians In order to allow propagation of exogenous DNA sequences, the plasmid must become stably integrated into the cellular genome in a non-essential (i.e. interruptible) region. Cells were cotransfected with the inducible plasmid, pGudLuc1.1, and a plasmid (pSVZneo) containing the aminoglycoside phosphotransferase 3'(l) (APH3'(I)) gene, which confers antibiotic (G418) resistance [33, 57, 58]. G418 is a neomycin analog that kills eukaryotic cells by inhibiting protein synthesis [58] Cells without the APH3’(I) gene die after 7 to 10 d [57]. Calculations Scatchard plots were used to determine the dose-response relationships. Scatchard plots model saturable binding of a receptor and ligand. The ratio of bound ligand to free ligand is plotted against the concentration of bound ligand to give a linear relationship. The slope of the line is -1/kD, where kD is the dissociation constant. The x-intercept is 8",”, the maximum concentration of 47 ligand which can be bound. In these experiments, change in luminescence units relative to the control was used as an index of concentration of bound ligand. Dose of the compound tested in pM was used as a measure of free ligand. In this case, the k0 is analogous to the EC50, or effective concentration for 50 % of the maximum response. Only points between the threshold and the maximum response are expected to fall within the linear range of the Scatchard plot. Points outside this range were not included in calculations (Figure 9). Because experiments were designed to investigate the dose- response relationship over a wide range of concentrations, only 3 to 5 doses of each test compound fell between the threshold and maximum. Future studies which include more doses in this range are needed to improve the accuracy of the Scatchard analysis. TEFs were used to validate the rainbow trout cell bioassay, by comparing the potency of PCBs predicted by the cell bioassay with the potency observed in in viva fish bioassays. TEFs are calculated be taking the ratio of the E050 for TCDD to the EC50 for the compound tested. Because PCB 77 did not produce the same maximum response as TCDD, this calculation could not be used. Instead, its TEF was based on the TCDD EC25. First, the response equal to 25 % of the maximum induced by TCDD was calculated. Next, the concentrations of TCDD or PCB 77 which produce this response were calculated from the dose-response curves. The TCDD EC25 was then divided by the concentration of PCB 77 which produces a response equivalent to the TCDD 25 % maximal response to give the TEF. This approach was validated 48 by performing the same calculations on the PCB 126 response. The TEFs calculated for PCB 126 by the EC50 and EC25 methods were identical. 49 *— l 7.0 ~ . TCDD 6.0 r . 5.0 - R2-0.845 4.0 , E050-142 pM 3.0 , max-0.011 2.0 l 1.0 ~ 0.0<> . s a 1 «M 0.000 0.002 0.004 0.006 0.008 0.010 é o x “Si P08126 ’2‘ e 0.4 r 0 R’-0.999 0 g 0.3 - E050-5340 pM E max-0.011 o 0.2 F\-\ 3 \\\ e 0.1 l- \.\ .E c [ ‘ \A.\ g 0.0 ' L ‘ 1 ‘ v 1 g 0.000 0.002 0.004 0.006 0.000 0.010 0 ___ M l PCB 77 o °~4 l R’-0.999 0.3 . 5050-14000 pM max-0.005 0.2 ~ 0.1 » NMR 00 1, 0““ I l L 0.0000 0.0020 0.0040 0.0060 0.0080 0.0100 change in response (luminescence units) Figure 9. Scatchard plots of dose-response curves. Points represent the mean of three samples at each dose. Points at the high and low doses which do not fall in the linear range were not included in calculations, and are shown as open diamonds. APPENDIX B Growth of RTH- 149 cells RTH-149 cells were originally grown in Dulbecco’s Modified Eagle's Medium (DMEM) for growth rate experiments (Table 3; Figure 10). However, it was discovered that this medium is too rich for this cell line. The RTH-149 cells used in this experiment were accidentally selected for their ability to grow in DMEM. After the growth rate experiments, new RTH-149 stocks were purchased from ATCC (American Type Culture Collection, Rockville, MD) and grown in basal Eagle's medium, which better supports the growth of RTH-149 cells. The doubling times for the three growth curves illustrate the contact- dependence of RTH-149 cells (Table 3). The cells grow faster when cell Table 3. Growth rates of RTH-149 cells seeded in DMEM. seeding density doubling time (hr) (cells/cm’) plating efficiency over log phase 0.1x10° 65 96 42 0.05x10a 33 % 53 0.03x10° 54 % 61 5O 51 density is greater, until they reach confluence. In the 0.1x10° cells/cm2 experiment, cell growth was measured until over a week past confluence. After confluence, the cells grew more slowly, but their viability was not affected. Plating efficiency was apparently not dependent on seeding density. However, in the 0.03x10" cells/cm2 experiment, about half the cells which originally plated died before the next sampling. cells/flask (log base 2) 24 23 18 52 I I ' i i t / 0 seed 1.25 x 10‘ cells ’ . seed 0.8 x 10° cells .,. / ° / I seed 2.6 x 10" cells T O L I n I n I n I n I 4 I n I 4681012141618 time (days) Figure 10. Growth rate of RTH-149 cells is dependent on seeding density. Cells were grown in 25 cm2 flasks. 53 APPENDIX C Transient transfection me thads The RTH-149 cell line was tested with four transient transfection methods: polybrene, calcium phosphate, DEAE-dextran, and lipofectin (Table 4). The RTG-2 cell line was tested with polybrene and lipofectin. For both cell lines, polybrene was the most effective method (Table 4). Polybrene was used in all subsequent transient transfections of inducible plasmids, and in the stable transfections. Table 4. Comparison of transient transfection methods. Cells were transfected with pGL2- Control. Luminescence is in arbitrary units per 20 pL sample. cell line method n luminescence W lipofectin 4 0.171 :I: 0.031 RTH-149 Calcium phosphate 2 0.060 d: 0.006 DEAE-dextran 2 0.297 :I: 0.024 polybrene 4 2.803 :I: 2.541 ”6'2 lipofectin 4 0.137 :I: 0.048 pooled blank measurements 8 0.033 1:0.016 54 Transfec tian experiments using RTG-Z and RT]. The RTG-2 and RTL cell lines were tested for inducibility by transiently transfecting with the pGudLuc1.1 plasmid (Table 5). Both cell lines showed inducibility. Induction and background activity were both much greater in RTG-2 cells than in RTL cells. Stable transfections of RTL and RTG-2 were unsuccessful. Table 5. Transient transfections of RTG-2 and RTL with the TCDD-inducible plasmid, pGudLuc1.1. Treatments each had two replicates. Luminescence is in arbitrary units per 20 pL sample. pGudLuc1.1 cell line no treatment DMSO TCDD 0.041 :I: RTL 0.042 :t 0.011 0.062 :t 0.030 0.438 :I: 0.454 APPENDIX D Changes in the RLT 1.0 cell line over time RTH-149 cells were stably transfected with pSV2neo and pGudLucl .1. Ten colonies were isolated, and seven of these survived to be tested for inducible luciferase expression (Table 6). RTH-149-T1 was chosen as the most suitable stable cell line, based on its relatively low background activity, low variability, and high inducibility. However, this clone has exhibited changes in its dose-response curve over time (Figure 1 1). The cell line is being reisolated in order to correct this problem. Table 6. Stable transfection of RTH-149 produced seven clones. Each was tested for inducible luciferase expression by dosing with 0.1 % DMSO or 1 nM TCDD in DMSO for 24 hr. T6 was exposed for 3 d, and its luminescence is expressed in Iuminometer units/pL. clone no treatment DMSO TCDD T1 __ __ I I 96.8 8.1 _ _ 9 95 2323. d: 2.4 T2 2478 a: 100 3138 :1: 308 3654 :1: 991 T3 3305 x 37 2941 :I: 580 5534 1 631 T4 0.020 :I: 0.004 0.049 :1: 0.016 0.202 :1: 0.216 T5 0.021 :t 0.008 0.047 :t 0.003 0.240 :I: 0.304 ' T6 436.6 :I: 15.4 559.2 :1: 28.4 1848 :I: 434 T8 0.034 :I: 0.027 0.029 :I: 0.001 0.006 :1: 0.002 56 —o- 4-1-94 9 20 ‘ —<>~ 6-3-94 18 1- ““ 6-9-94 -4- 6-30-94 16 - -v- 7-18-94 * —v— 8‘26‘94 —10-4-94 A — / <> 7 —A-— 10-12-94 w //;:;/ 4 ' u/jf/ —-A—— 10-13-94 ' ' //4/o:f:,——;. .. :3/ 2 _ —v— 10-24-94 " , 10'4 10‘3 10’2 10‘1 100 101 TCDD(nM) Figure 11. Changes in the response to TCDD in RLT 1.0 cells. LIST OF REFERENCES LIST OF REFERENCES Poland, A. and J.C. Knutson. 1982. 2,3,7,8-Tetrachl0rodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: examination of the mechanism of toxicity. Ann. Rev. Pharamacol. Toxicol. 22:517-554. Safe, S.H. 1986. Comparative Toxicology and Mechanism of Action of Polychlorinated Dibenzo-p-Dioxins and Dibenzofurans. Ann. Rev. Pharamacol. Toxicol. 26:371-399. Safe, 8. 1990. Polychorinated Biphenyls (PCBs), Dibenzo-p-Dioxins (PCDDs), Dibenzofurans (PCDFs), and Related Compounds: Environmental and Mechanistic Considerations Which Support the Development of Toxic Equivalency Factors (TEFs). Crit. Rev. Toxicol. 21:51-88. Spitsbergen. J.M., J.M. Kleeman and R.E. Peterson. 1988. Morphologic lesions and acute toxicity in rainbow trout (Salmo gairdnen) treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. J. Toxicol. Environ. Health 23:333- 358. Spitsbergen, J.M., K.A. Schat, J.M. Kleeman and R.E. Peterson. 1988. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or Aroclor 1254 on the resistance of rainbow trout, SaImo gairdneri Richardson, to infectious haematopoietic necrosis virus. J. Fish Dis. 11:73-83. Thomas, P. 1989. Effects of Aroclor 1254 and cadmium on reproductive endocrine function and ovarian growth in Atlantic croaker. Marine Environ. Res. 28:499-503. Walker, M.K. and R.E. Peterson. 1991. Potencies of polychlorinated dibenzo-p-dioxin, dibenzofuran, and biphenyl congeners, relative to 2,3,7,8-tetrachlorodibenzo-p-dioxin, for producing early life stage mortality in rainbow trout (Oncorhynchus mykiss). Aquat. Toxicol. 21:219-238. 57 10. 11. 12. 13. 14. 15. 16. 17. 18. 58 Walker, M.W. and R.E. Peterson. 1994. Toxicity of 2,3,7,8- tetrachlorodibenzo-p-dioxin to brook trout (Salve/inus fontinalis) during early development. Environ. Toxicol. Chem. 13:817-820. Walker, M.K., J.M. Spitsbergen, J.R. Olson and R.E. Peterson. 1991. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) toxicity during early life stage development of lake trout (Salvelinus namaycush). Can. J. Fish. Aquat. Sci. 48:875-883. Whitlock, J.P., Jr. 1993. Mechanistic aspects of dioxin action. Chem. Res. Toxicol. 6:754-763. Matsumura, F. 1994. How important is the protein phosphorylation pathway in the toxic expression of dioxin-type chemicals? Biochem. Pharmacol. 48(2):21 5-224. Dolwick, K.M., H.I. Swanson and C.A. Bradfield. 1993. In vitra analysis of Ah receptor domains involved in ligand-activated DNA recognition. Proc. Natl. Acad. Sci. USA 90:8566-8570. Hoffman, E.C., H. Reyes, C. Fang-Fang, F. Sander, L.H. Conley, B.A. Brooks and O. Hankinson. 1991. Cloning of a Factor Required for Activity of the Ah (Dioxin) Receptor. Science 252:954—958. Carrier, F., R.A. Owens, D.W. Nebert and A. Puga. 1992. Dioxin- dependent activation of murine cyp la-l gene transcription requires protein kinase C-dependent phosphorylation. Mol. Cell. Biol. 12:1856— 1863. Reiners, J.J., Jr., A.R. Cantu and A. Schéller. 1992. Phorbol ester- mediated suppression of cytochrome P450 Cyp 1a-1 induction in murine skin: Involvement of protein kinase C. Biochem. Biophys. Res. Commun. 186:970-976. Yao, E.F. and M.S. Denison. 1992. DNA Sequence Determinants for Binding of Transformed Ah Receptor to a Dioxin-Responsive Enhancer. Biochemistry 31 :5060—5067. Denison, M.S., J.M. Fisher and J.P. Whitlock Jr. 1988. The DNA Recognition Site for the Dioxin-Ah Receptor Complex. J. Biol. Chem. 263: 1 7221-1 7224. Gonzalez, F.J. 1989. The Molecular Biology of Cytochrome P450s. Pharmacol. Rev. 40:243-288. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 59 Puga, A., D.W. Nebert and F. Carrier. 1992. Dioxin induces expression of c-fos and c-jun proto-oncogenes and a large increase in transcription factor AP-1. DNA. Cell. Biol. 11:269-281. Walker, M.K., L.C. Hufnagle Jr., M.K. Clayton and R.E. Peterson. 1992. An egg injection method for assessing early life stage mortality of polychlorinated dibenzo-p-dioxins, dibenzofurans, and biphenyls in rainbow trout, (Oncorhynchus mykiss). Aquat. Toxicol. 22:15-38. Newsted, J.L., J.P. Giesy, G.T. Ankley. D.E. Tillitt, B.A. Crawford, J.W. Gooch, P.D. Jones and M.S. Denison. submitted. Development of toxic equivalency factors for PCB congeners and the assessment of TCDD and PCB mixtures in rainbow trout. Environ. Toxicol. Chem. Tillitt, D.E., J.P. Giesy and G.T. Ankley. 1991. Characterization of the H4IIE Rat Hepatoma Cell Bioassay as a Tool for Assessing Toxic Potency of Planar Halogenated Hydrocarbons in Environmental Samples. Environ. Sci. Technol. 25:87-92. El-Fouly, M.H., C. Richter, J.P. Giesy and M.S. Denison. 1994. Production of a novel recombinant cell line for use as a bioassay system for detection of 2,3,7,8-tetrachlorodibenzo-p-dioxin-like chemicals. Environ. Toxicol. Chem. 13:1581-1588. Lorenzen, A. and A.B. Okay. 1990. Detection and Characterization of [3H]2,3,7,8-Tetrachlorodibenzo-p-dioxin Binding to Ah Receptor in a Rainbow Trout Hepatoma Cell Line. Toxicol. Appl. Pharmacol. 106:53- 62. de Wet, J.R., K.V. Wood, M. DeLuca, D.R. Helinski and S. Subramani. 1987. Firefly luciferase gene: Structure and expression in mammalian cells. Mol. Cell. Biol. 7:725-737. Hahn, M.E., T.M. Lamb, M.E. Schultz, R.M. Smolowitz and J.J. Stegeman. 1993. Cytochrome P4501A induction and inhibition by 3,3',4,4'-tetrachlorobiphenyl in an Ah receptor-containing fish hepatoma cell line (PLHC-1). Aquat. Toxicol. 26:185-208. Fisher, J.M., K.W. Jones and J.P. Whitlock Jr. 1989. Activation of transcription as a general mechanism of 2,3,7,8-tetrachlorodibenzo-p- dioxin action. Molec. Carcinogen. 1:216-221. Lee, F., C.V. Hall, G.M. Ringold, D.E. Dobson, J. Luh and PE. Jacob. 1984. Functional analysis of the steroid hormone control region of mouse mammary tumor virus. Nucleic Acids Res. 12:4191-4206. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 60 Jones, P.B.C., D.R. Galeazzi, J.M. Fisher and J.P. Whitlock Jr. 1985. Control of cytochrome P1-450 gene expression by dioxin. Science 227: 1 499-1 502. Jones, P.B.C., L.K. Durrin, D.R. Galeazzi and J.P. Whitlock Jr. 1986. Control of cytochrome P1-450 gene expression: Analysis of a dioxin- responsive enhancer system. Proc. Natl. Acad. Sci. USA 83:2802-2806. Kawai, S. and M. Nishizawa. 1984. New Procedure for DNA Transfection with Polycation and Dimethyl Sulfoxide. Mol. Cell. Biol. 4:1172-1174. Sussman, D.J. and G. Milman. 1984. Short-term, high-efficiency expression of transfected DNA. Mol. Cell. Biol. 4:1641-1643. Southern, F.J. and P. Berg. 1982. Transformation of mammalian cells to antibiotic resistance with a bacterial gene under control of the SV40 early region promoter. J. Mol. Appl. Gen. 1:327-341. Bradford, MM. 1976. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72:248-254. Lubet. B.A., R.W. Nims. R.T. Mayer. J.W. Cameron and L.M. Schechtman. 1985. Measurement of cytochrome P-450 dependent dealkylation of alkoxyphenoxazones in hepatic S98 and hepatocyte homogenates: effects of dicumerol. Mutation Res. 142:127-131. Oris, J.T. and J.A. Bailer. 1993. Statistical analysis of the Ceriodaphnia toxicity test: Sample size determination for reproductive effects. Environ. Toxicol. Chem. 12:85-90. Munkittrick, K.R., G.J. Van Der Kraak, M.E. McMaster and C.B. Portt. 1992. Response of hepatic MFO activity and plasma sex steroids to secondary treatment of bleached kraft pulp mill effluent and mill shutdown. Environ. Toxicol. Chem. 11:1427-1439. Munkittrick, K.R., G.J. Van Der Kraak. M.E. McMaster, C.B. Portt, M.R. van den Heuvel and M.R. Servos. 1994. Survey of receiving-water environmental impacts associated with discharges from pulp mills. 2. Gonad size, liver size, hepatic EROD activity and plasma sex steroid levels in white sucker. Environ. Toxicol. Chem. 13:1089-1101. Servos, M.R., S.Y. Huestis, D.M. Whittle, G.J. Van Der Kraak and K.R. Munkittrick. 1994. Survey of receiving-water environmental impacts associated with discharges from pulp mills. 3. Polychlorinated dioxins 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 61 and furans in muscle and liver of white sucker (Catastamus cammersom). Environ. Toxicol. Chem. 1 3:1 103-1 1 15. van den Heuvel, M.R., K.R. Munkittrick, G.J. Van Der Kraak, M.E. McMaster, C.B. Portt, M.R. Servos and D.G. Dixon. 1994. Survey of receiving-water environmental impacts associated with discharges from pulp mills. 4. Bioassay-derived 2,3,7,8-tetrachlorodibenzo-p-dioxin toxic equivalent concentration in white sucker (Catastamus commersom) in relation to biochemical indicators of impact. Environ. Toxicol. Chem. 13:1117-1126. Fryer, J.L., 8.8. McCain and J.C. Leong. 1981. A Cell Line Derived from Rainbow Trout (Salma gairdnen) Hepatoma. Fish Pathol. 15:193-200. Lannan, C.N., J.R. Winton and J.L. Fryer. 1984. Fish Cell Lines: Establishment and Characterization of Nine Cell Lines from Salmonids. In Vitra: Cell. Dev. Biol. 20:671-676. Wolf, K. and M.C. Ouimby. 1962. Established Eurythermic Line of Fish Cells in vitro. Science 135:1065-1066. Swanson, H.I. and G.H. Perdew. 1991. Detection of the Ah receptor in rainbow trout: use of 2-azido-3—[125lliodo-7,8-dibromodibenzo-p-dioxin in cell culture. Toxicol. Lett. 58(1):85-95. Wetzel, R.G. 1975. Limnology, p. 326. W.B. Saunders Company, Philadelphia, p. 326. Bimboim, H.C. 1983. A rapid alkaline extraction method for the isolation of plasmid DNA. Meth. EnzymoI. 100:243-255. Lis, J.T. 1980. Fractionation of DNA fragments by polyethylene glycol induced precipitation. Meth. Enzymol. 65:347-353. Radloff, R., W. Bauer and J. Vinograd. 1967. A dye-buoyant-density method for the detection and isolation of closed circular duplex DNA: The closed curcular DNA in HeLa cells. Proc. Natl. Acad. Sci. USA 57:1514-1521. Southern, E. 1979. Gel electrophoresis of restriction fragments. Meth. EnzymoI. 68:152. McDonell, M.W., M.N. Simon and F.W. Studier. 1977. Analysis of restriction fragments of T7 DNA and determination of molecular weights by electrophoresis in neutral and alkaline gels. J. Mol. Biol. 110:119-146. 51. 52. 53. 54. 55. 56. 57. 58. 62 Felgner, P.L., T.R. Gadek, M. Holm, R. Roman, H.W. Chan, M. Wenz, J.P. Northrop, G.M. Ringold and M. Danielsen. 1987. Lipofection: A highly efficient, lipid-mediated DNA-transfection procedure. Proc. Natl. Acad. Sci. USA 84:7413-7417. Luthman, H. and G. Magnusson. 1983. High efficiency polyoma DNA transfection of chloroquine treated cells. Nucleic Acids Res. 11:1295- 1308. Wood, K.V. 1991. The origin of beetle luciferases. In P.E. Stanley and L.J. Kricka, eds., Bioluminescence and Chemiluminescence Current Status. John Wiley 81 Sons, New York, pp. 11-14. Wood, K.V. 1991. Recent advances and prospects for use of beetle luciferases as genetic reporters. ln P.E. Stanley and L.J. Kricka, eds., Bioluminescence and Chemiluminescence Current Status. John Wiley & Sons, New York, pp. 543-546. Wang, C.Y. and JD. Andrade. 1991. Denaturation of firefly luciferase. In P.E. Stanley and L.J. Kricka, eds., Bioluminescence and Chemiluminescence Current Status. John Wiley & Sons, New York, pp. 427-432. Schram, E. 1991. Evolution of bioluminescent ATP assays. In P.E. Stanley and L.J. Kricka, eds., Bioluminescence and Chemiluminescence Current Status. John Wiley & Sons, New York, pp. 407-412. Colbere-Garapin, F., F. Horodniceanu, P. Kourilsky and A.-C. Garapin. 1981. A new dominant hybrid selective marker for higher eukaryotic cells. J. Mol. Biol. 150:1-14. Jimenez, A. and J. Davies. 1980. Expression of a transposable antibiotic resistance element in Saccharomyces. Nature 287:869-871. MICHIGAN STATE UNIV. LIBRARIES lllll11111111111I||I1111111111111111ll 31293014110559