THESIS I J IIIIIIIIII IIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIII <7 p.30 3 1293 02060 4090 K L/ This is to certify that the thesis entitled INTERACTION BETWEEN ARYL HYDROCARBON RECEPTOR (AHR) AND HYPOXIA SIGNALING PATHWAYS: A POTENTIAL MECHANISM FOR TCDD TOXICITY presented by MINGHUA NIE has been accepted towards fulfillment of the requirements for MASTER degree in ZOOLOGY 10ng Major p‘o/essor Date 07639 MS U is an Affirmative Action/Equal Opportunity Institution LIBRARY Michigan State University 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 11/00 chlRC/DateDuepSS—pu INTERACTION BETWEEN ARYL HYDROCARBON RECEPTOR (AHR) AND HYPOXIA SIGNALING PATHWAYS: A POTENTIAL MECHANISM FOR TCDD TOXICITY By Minghua Nie A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE Department of Zoology 1999 ABSTRACT INTERACTION BETWEEN ARYL HYDROCARBON RECEPTOR (AHR) AND HYPOXIA SIGNALING PATHWAYS: A POTENTIAL MECHANISM FOR TCDD TOXICITY By Minghua Nie Many of the toxic responses of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) are mediated through the AhR, which requires ARNT to regulate gene expression. ARNT is also required by HIP—1a to enhance the expression of various genes in response to hypoxia. Since both the AhR and hypoxia transcriptional pathways require ARNT, some of the effects of TCDD and similar types of ligands could be explained by interaction between the AhR and hypoxia pathways involving ARNT. The studies on which I report here were conducted to test the hypothesis that there is cross talk between AhR- and HIF- l-mediated transcription pathways. TCDD significantly reduced the hypoxia-mediated reporter gene activity in B-l cells. Reciprocally, the hypoxia response inducers desferrioxamine or CoClz inhibited AhR-mediated CYP1A1 enzyme activity in B-1 and Hepa 1 cells, and the AhR-mediated luciferase reporter gene activity in H1L1.102 cells. The inhibition of AhR-mediated transcription by hypoxia inducers, however, was not observed in H4IIE-luc cells. The interaction between AhR- and HIF-l-mediated transcription can be attributed to changes in DNA binding activities. TCDD-induced protein binding to the dioxin responsive element (DRE) was diminished by desferrioxamine, and TCDD reduced the binding activity to the HIF-l binding site in desferrioxamine-treated Hepa 1 cells. This mutual repression may provide an underlying mechanism for many TCDD-induced toxic responses. The results reported here indicate that there is cross talk between ARNT-requiring pathways. Since ARNT is possibly required by a number of pathways, this type of interaction may explain some of the pleiotropic effects caused by TCDD. T o my parents Guangmao Nie and Short F u for their unconditional love, and to my mentor Dr. Gordon Ultsch for his understanding and support. iv ACKNOWLEDGMENTS First of all, I would like to thank my major professor Dr. John P. Giesy for his training, support, and guidance during the past years. I wish to thank Dr. Larry Fischer, Dr. Tim Zacharewski, and Dr. Lynwood Clemens for serving as members of my graduate committee. I also want to thank the members of Dr. Giesy’s laboratory, Dr. Alan Blankenship, Dr. Alex Villalobos, Dr. Kurunthachalam Kannan, Dr. Paul Jones, Katie Kemler, Carolyn Duda, Stephanie Pastva, Wen-Yue Hu, Dan Villeneuve, Erin Snyder, Shane Snyder, Kevin Henry, and Tim Keith for their help and friendship, with especial appreciation to Dr. Alan Blankenship for his valuable advise and mentorship. I would like to acknowledge Dr. Jamie Caro, Dr. Oliver Hankinson, Dr. Michael Denison for providing cell lines used in my study, and Dr. Norbert E. Kaminski as well as the members of his laboratory for technical assistance and offering laboratory space for doing electrophoresis mobility shift assay. TABLE OF CONTENTS LIST OF FIGURES ............................................................................. viii LIST OF ABBREVIATIONS .................................................................. ix INTRODUCTION ................................................................................ 1 MATERIALS AND METHODS ............................................................... 7 Cell lines and cell culture conditions ................................................. 7 Chemicals and treatments .............................................................. 8 Luciferase assay ......................................................................... 8 EROD assay .............................................................................. 9 Nuclear protein extraction ............................................................ 10 Electrophoresis mobility shift assay and supershifi assay .......................... 11 Statistical analysis ........................................................................ 12 RESULTS .......................................................................................... 13 Effect of TCDD on HIF-l -mediated reporter gene activity ........................ 13 Effect of hypoxia response inducer on AhR-mediated CYP1A1 induction .................................................................................. 13 Effect of hypoxia response inducer on AhR-mediated reporter gene activity ............................................................................... 16 Cell line dependency of interaction between the AhR and hypoxia pathways ......................................................................... 21 vi Interaction of TCDD and Dfx on Protein Complex Binding Activities to DRE3 and HIF-l .................................................................................. 21 DISCUSSION ..................................................................................... 33 REFERENCES .................................................................................... 41 vii LIST OF FIGURES Figure 1. Schematic representation of the involvement of ARN T in AhR- and HIF-l-mediated transduction pathways ......................................... 5 Figure 2. TCDD suppresses the hypoxia-inducible enhancer in B-l cells . . . . . . . .. 15 Figure 3. Dfx inhibits the TCDD-induced EROD activity in 8-1 and Hepa 1 cells ....................................................................................... 18 Figure 4. Dfx and C002 suppress TCDD-inducible CYP1A1 enhancer in H1L1.1c2 cells .................................................................................... 20 Figure 5. Effect of hypoxia inducers on the AhR-mediated gene expression in H4IIE-luc cells. .................................................................................... 24 Figure 6A. Effect of Dfx on AhR:ARNT binding activity to DRE3 ....................... 26 Figure 6B. Presence of AhR in protein complexes binding to DRE3 ...................... 28 Figure 7A. Effect of TCDD on DNA-binding activity of HIF-l ........................... 30 Figure 7B. Presence of HIF-la in the protein complex that binds to HIF-l . .. . . . . 32 Figure 8. A potential mechanism of action for TCDD ........................................ 40 viii LIST OF ABBREVIATIONS AHH — Aryl hydrocarbon hydroxylase AhR — Aryl hydrocarbon receptor Ah receptor — Aryl hydrocarbon receptor ARNT (or Amt) — Ah receptor nuclear translocator bHLH —- Basic-Helix-Loop-Helix CYP1A1 — Cytochrome p450 1A1 CYP1A2 — Cytochrome p450 1A2 Dfx — Desferrioxamine DRE — Dioxin response element EROD — Ethoxyresorufin O-deethylase HIP-1a -- Hypoxia inducible factor 1 alpha HIF-l [3 -- Hypoxia inducible factor 1 beta HRE — Hypoxia responsive element Hsp 9O — Heat shock protein 90 PAS — Per-Sim-Arnt PCB — Polychlorinated biphenyl PCDD — Polychlorinated dibenzo-p-dioxin PCDF — Polychlorinated dibenzofuran PCDH — Polychlorinated diaromatic hydrocarbon TCDD (or 2,3,7,8-TCDD) — 2,3,7,8-tetrachlorodibenzo-p-dioxin ix INTRODUCTION Polychlorinated diaromatic hydrocarbons (PCDHS), such as polychlorinated biphenyls (PCBS), and polychlorinated naphthalenes (PCNs), polychlorinated dibenzo-p- dioxins (PCDDs), polychlorinated dibenzofurans (PCDFs), are industrial compounds or byproducts. PCDHS are widespread environmental contaminants that have been identified in air, sediment, domestic animals, wildlife, and human tissues. Environmental contamination by PCDHs has adversely affected wildlife, and has raised concern of its consequences for human health (Poland and Knutson, 1982; Safe, 1986 and 1990). For instance, some pathological defects observed in the colonial, fish-eating water birds of the North American Great Lakes have been attributed to exposure to PCDHs. These effects include embryonic deformities and lethality, immune suppression, edema, and porphyria, and were caused by eating contaminated fish (Giesy et al., 1994a and 1994b; Ludwig et al.,1996). As the most potent member of PCDHs, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has been studied more extensively than other structurally related compounds. In animals, TCDD elicits a wide range of biological effects, including alteration in metabolic pathways, immunological changes, cardiac dysfunction, hepatotoxicity, carcinogenicity, endocrine dysfunction, teratogenic effects, and neoplasia (Schmidt and Bradfield, 1996; Okey et al., 1994; Pohjanvirta and Tuomisto, 1994; Poland and Knutson, 1982). Diverse biochemical responses to TCDD have been observed, such as drug metabolizing enzyme induction (Whitlock, 1990 and 1993), modulation of cytokine levels, receptor function, and ligand binding (Abbott and Bimbaum, 1990; Choi et al., 1991; Clark et al., 1991), and activation of protein kinases (Enan and Matsumura, 1993; Bombick et al., 1985). The evidence supports the hypothesis that most, if not all, TCDD effects are mediated through the aryl hydrocarbon receptor (Ah receptor, or AhR; see Okey et al., 1994; Hankinson, 1995 for reviews), a cytosolic receptor protein that was first discovered by Poland and coworkers (1976). The AhR signaling transcription pathway is initiated by TCDD diffusion into the cell, where it binds with high affinity to the cytosolic AhR protein complex, which also includes heat shock protein 90 (Hsp90) and a 38-kDa, immunophilin-related protein (Ma and Whitlock, 1997; Carver and Bradfield, 1997). The ligand binding activates AhR and stimulates the dissociation of AhR-associated proteins. The ligand-receptor complex is subsequently translocated into the nucleus, where it dimerizes with AhR nuclear translocator (ARNT) (Hankinson, 1995; Probst et al., 1993). The heterodimers are capable of recognizing and binding DNA at the consensus sequence, GCGTG, of dioxin responsive elements (DREs) (Denison et al., 1989; Dong et al., 1996). This action either increases or decreases the transcription of target genes (Nebert et al., 1993; Schmidt and Bradfield, 1996), including cytochrome P-450 (CYP1A1, CYP1A2) (Whitlock, 1987; Quattrochi and Tukey, 1989), NAD(P)H:quinone reductase (Favreau and Pickett, 1991), class 3 aldehyde dehydrogenase (Asman et al., 1993), and glutathione S-transferase (Paulson et al., 1990). The ARNT protein, however, does not function uniquely in the AhR signaling pathway, but also pairs with HIP-1a (hypoxia inducible factor 10.) to regulate genes active in response to low oxygen stress (Guillemin and Krasnow, 1997; Semenza, 1994; Wenger and Gassmann, 1997). HIP-10L is continuously synthesized and degraded under normal oxygen tension. Hypoxic conditions inhibit the degradation of HIF-la by the ubiquitin proteasome system, and triggers the nuclear localization of HIP-10L (Huang et al., 1998; Kallio et al., 1997; Salceda and Caro., 1997). Transition metals, such as cobalt, and iron chelators, such as desferrioxamine (Dfx), elicit the same response in cells, which suggests that these stimuli work at different stages of a common oxygen sensing pathway (Guillemin and Krasnow, 1997). Inside the nucleus, HIF-la heterodimerizes with ARNT to form a transcription factor complex HIF-l (Jiang et al., 1996; Salceda et al., 1996; Wood et al., 1996). The binding of HIF-l to the core DNA sequence, TACGTG, of the hypoxia response enhancer (HRE) (Jiang et al., 1996; Semenza et al., 1991; Wang and Semenza, 1993) and subsequent transactivation enhence expression of genes such as Epo for erythropoiesis (Semenza, 1994), VEGF for angiogenesis (Forsythe et al., 1996; Goldberg and Schneider, 1994; Maxwell et al., 1997; Shweiki et al., 1992), and GLUT-l for glucose transport (Semenza et al., 1994; Wenger and Gassmann, 1997) (Figure 1). In vitro, ARNT can also form a homodimer or heterodimers with a number of PAS (Per-ARNT-Sim) proteins, which include AhR, ARNT, HIP-10L, and transcription factors involved in various gene regulation pathways (Hahn, 1998; Hogenesch et al., 1997). These results suggest ARNT might be a central regulator of many cellular pathways through its formation of functional transcription activators or repressors by dimerizing with other members of the PAS family. In adult animals, ARNT is ubiquitously expressed in all tissues. The essential role of ARNT is supported by studies on ARNT null mice, which are not viable beyond day 10.5 of gestation (Kozak et al., 1997; Maltepe et al., 1997). The importance of ARNT during development and the observation that ARNT dimerizes with various PAS proteins suggest that availability of ARNT could be critical for various cellular activities, and competitive recruitment of ARNT may repress the activation of certain genes. TCDD is a potent inducer of AhR-mediated gene activation and can cause prolonged activation of AhR. It is possible that exposure to TCDD and subsequent recruitment of ARNT through AhR may inhibit other signal transduction pathways depending on ARNT. We hypothesize that the pleiotropic effects of TCDD could be due to TCDD-induced ARNT deficiencies in multiple ARNT-requiring pathways. Because the AhR and HIF-l pathways are two of the most well understood pathways involving ARNT, they were chosen as a model to study the potential effect of TCDD on linked pathways. The interaction between the AhR and hypoxia pathways was investigated at the level of transcription, nuclear protein complex formation, and DNA- binding activities. Figure 1. Schematic representation of the involvement of ARNT in AhR- and HIF—l- mediated transduction pathways. The AhR pathway is initiated when AhR ligands, such as TCDD, diffuse into the cell (1), bind to and activate the cytosolic AhR complex, which also includes Hsp90s and a immunophilin-related protein (IRP) (2). Activation leads to dissociation of the AhR-associated proteins (3) and nuclear translocation of AhR- ligand complex (4). Inside the nucleus, the AhR-ligand complex heterodimerizes with ARNT (5), and binds to DREs to alter gene expression (6). The hypoxia response pathway is initiated by stimuli such as hypoxia, CoClz, or Dfx (7), which inhibit the degradation of cytosolic HIF -10L by the ubiquitin proteasome system (8), and trigger the nuclear localization of HIF-la (9). In the nucleus, HIP-10L forms HIF-l a complex with ARNT (10), then binds to the HIF-l binding site of hypoxia responsive enhancer (HRE) and enhances the transcription of genes regulated through HRE. 15.2 no 0.» ma? MATERIALS AND METHODS Cell Lines and Cell Culture Conditions Four cell lines were selected for study because of particular properties that were useful in investigating cross talk between the two pathways of interest. B-l cells were derived from the human hepatoma Hep 38 cell line, which has been stably transfected with an expression vector containing luciferase cDNA under the control of a minimal Epo promoter (300-base pair SfaNI-XbaIII fragment) and the hypoxia responsive enhancer from the human Epo gene (ISO-base pair ApaI/PstI fragment) (Salceda and Caro, 1997). H1L1.1c2 and H4IIE-luc cells are mouse and rat hepatoma cells, respectively, which are stably transfected with a luciferase reporter gene under control of dioxin-responsive elements (DRES) (Garrison et al., 1996). B-l cells were obtained from Dr. J. Caro (Department of Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA). Wild-type Hepa 1 cells were obtained from Dr. 0. Hankinson (Department of Pathology, University of California at Los Angeles, Los Angeles, CA). H1L1.102 cells were provided by Dr. M. Denison (Department of Environmental Toxicology, University of California at Davis, Davis, CA). 3-] cells were maintained in minimal essential medium. Hepa 1 and H1L1.102 cells were maintained in alpha-modified minimal essential medium. H4IIE-luc cells were grown in Dulbecco’s Modified Eagle Medium. All media were supplemented with 10% heat- inactivated fetal bovine serum, penicillin (100 units/ml), and streptomycin (100 ug/ml). Cells were grown under sterile conditions at 37 0C in a humidified atmosphere with 5% C02. Chemicals and Treatments Desferrioxamine (Dfx) or CoClz (in Millipore H20) was used as inducers of the hypoxia response pathway (Salceda and Caro, 1997), TCDD (in MeOH or DMSO) was used as AhR ligand. The concentrations of each treatment were selected in pilot studies to result in maximal responses in each cell line. The concentration ranges used were non- toxic to cells as determined by microscopic morphological examination, cell viability assays based on esterase activity and DNA integrity, and protein assays. Luciferase Assay The luciferase assay was based on the methods of Sanderson et al. (1996). In brief, cells were trypsinized from plates containing 80-100% confluent monolayers, resuspended in media, and diluted to a concentration of approximately 105 cells/ml and seeded into the 60 interior wells of 96-well culture ViewPlateTM (Packard Instruments, Meriden, CT) at 250 pl per well. The 36 exterior wells of each plate were filled with 250 pl culture media. Cells were incubated overnight, then dosed with 1.25 ul of the appropriate treatment chemical or vehicle solvent (methanol). Blank wells received no dose. Luciferase and protein assays (Kennedy and Jones, 1994) were conducted after 24 to 30 hr of exposure. Briefly, culture medium was removed by vacuum manifold and the cells were rinsed with Dulbecco’s phosphate-buffered saline (PBS, containing 0.02% (w/w) KCl, 0.02% (w/w) KHZPO4, 0.8% (w/w) NaCl, 10 mM NazHPO4, pH 7.4). Into each well, 75 111 PBS with Ca2+ and Mg2+ (0.01% (w/w) of CaCI2 and MgClz) and 75 pl LucLite reagent (from LucLiteTM Kit; Packard Instruments, Meriden, CT ) were added. The plates were incubated for 15 min at 30 oC and then scanned with an ML3000 microplate reading luminometer (Dynatech Laboratories, Chantilly, VA, USA). Following the luminometer scan a 1.08 mM solution of fluorescamine in acetonitrile was added to each well and plates were assayed for protein after a 15 min incubation at room temperature, using a Cytofluor 2300 (Millipore Corp., Bedford, MA) (excitation 400 nm, emission 460 nm). EROD Assay Assays for ethoxyresorufin-O—deethylase activity (EROD) of CYP1A1 were performed as described by Sanderson et al. (1996). Briefly, the cells were trypsinized and seeded as described in the description of the luciferase assay. At the end of the exposure period, medium was aspirated from each well, which was washed twice with PBS before adding 30 ul nanopure water. The plates were frozen at —80 °C for 10 min and thawed at room temperature, and then 100 pl HEPES-dicumoral buffer (50 mM HEPES and 40 uM dicumarol) and 50 ul ethoxyresorufin (20 uM in HEPES-dicumoral buffer) were added to the wells. The EROD reaction was started by adding 20 ul NADPH (1.25 mM in 40 mM HEPES). After a 60-min incubation at 30 0C, the reaction was stopped by adding 50 ul of 1.08 mM fluorescamine (in acetonitrile), followed by an additional 15-min incubation at room temperature. Fluorescence was then measured on a Cytofluor 2300. Resorufin was measured at excitation/emission wavelengths of 530/590 nm and protein was measured at excitation/emission wavelengths of 400/460 nm. BSA and resorufin standard curves were prepared on the day of assay. Nuclear Protein Extraction Hepa 1 cells from four 90% confluent 100 mm petri dishes were harvested into ice-cold PBS using a cell scraper. The cell suspension was centrifuged for 5 min at 500 g. After discarding the supernatant, the pellet was resuspended and incubated in 1 ml HM (10 mM HEPES, 3 mM MgC12, pH 7.5) on ice for 10 min. The Suspension was then homogenized with a tight fitting (B pestle) Dounce Homogenizger. The homogenate was centrifuged at 1000 g for 5 min at 4 OC. The supernatant was removed and the pellet was washed twice with HMK (25 mM HEPES, pH 7.5; 3mM MgClz, 100 mM KCl) and centrifuged to remove supernatant. The pellet was then resuspended in 2 volumes of HEKG (25 mM HEPES, pH 7.5; 1 mM EDTA, 400 mM KCl, 10% glycerol), vortexed, and incubated on ice for 30 to 40 min. At the end of the incubation, the suspension was centrifuged at 15,000 g for 15 min at 4 °C. The supernatant was collected as nuclear protein extract and stored at —80 0C. The protein content was measured using Bradford protein assay using BSA as a standard. All buffers for extraction were supplemented with 2 ug/ml aproptinin, 2 11ng leupeptin, 1 mM DTT, 0.1 mM Na3VO4, and 0.5 mM PMSF before each use. 10 Electrophoresis Mobility Shift Assay and Supershift Assay Binding sequences for electrophoresis mobility shift assays (EMSA) were DRE3 (5’-GAGCTCGGAGITMAGAAGAGCCT-3’) and W18 (5’-GCCCTACGTGCT GTCTCA-3’). The DNA probes (underlines indicate the protein binding sites) were made by the Macromolecular Facility at Michigan State University. The probes were end-labeled with EasyTide® [y 32P] ATP (NEN Life Science Products, Inc., Boston, MA) using Ready-To-Go T4 polynucleotide kinase (Pharmacia Biotech) and purified using NucTrap Push Column (Stratagene). Before the assay, 4% acrylamide gel was pre-run for l to 2 hr at 110 volts using TAE buffer (6.7 mM Tris, pH 8.0, 3.3 mM sodium acetate, 1 mM EDTA), and was supplied with fresh TAE before loading the samples. The final sample mixture contained 9 ug nuclear proteins, 25 mM HEPES (pH 7.5), 1 mM EDTA, 80 mM KCl, lmM DTT, 10% glycerol, 2 ug Poly (dI-dC), and 40000 to 60000 cpm radiolabeled oligonucleotides to make up a final volume of 30 ul. For the samples containing cold competitor, ZOO-fold excess of non-radiolabeled probe was added 5 min before adding 32P-labeled probe. For HRE binding assays, the samples were incubated on ice instead of at room temperature. For the samples used in the supershift assays, 1.5 ug antibody was incubated with the protein mixture (1:5 antibody to protein ratio) on ice for 1 hr before adding the probes. AhR antibody was purchased from Affinity Bioreagents (Golden, CO). HIF-lor antibody was purchased from Novus Biologicals, Inc. (Littleton, CO). After sample loading, the gel was run for 2.5 to 3.25 hr at 110 volts, dried on a gel dryer, and then exposed to Hyperfilm MP in a film cassette with intensifying screen overnight to two days at —80 0C. 11 Statistical Analysis The data were analyzed using SigmaStat software (J andel Scientific, Inc., Rafael, CA). Pair-wise t-tests were performed with the probability of type I error (0L) set to be less than 0.05. 12 RESULTS Eflect of T CDD on HIF -1 ~Mediated Reporter Gene Activity Effects of TCDD on HIF -1-mediated gene expression were determined by measuring changes in luciferase activity in response to factors (CoClz or Dfx) known to induce hypoxia-like responses in the presence or absence of TCDD in B-l cells. The luciferase activity in B-l cells was increased 12-fold by 125.0 uM Dfx and 7-fold by 40 uM CoClz after 24-hr exposure. To examine the effect of AhR-mediated induction on HIF-l-mediated transcriptional activation, B-l cells were treated with a range of concentrations of TCDD from 0.125 to 12.5 nM for 6 hr before the administration of hypoxia inducer. Pre-incubation with TCDD caused a concentration-related reduction of luciferase activity (Figure 2). At a concentration of 12.5 nM, TCDD reduced the reduced the maximal induction of the hypoxia response pathway by approximately 35% in both Dfx and CoClz-treated B-l cells. Methanol, the carrier solvent of TCDD had no significant effect on HIF- 1 -mediated luciferase induction. Effects of Inducers of the Hypoxia Response Pathway on AhR-Mediated CYP1A1 Induction To investigate whether AhR-mediated responses are affected by the activation of the hypoxia response signaling pathway, CYP1A1 induction was examined through the 13 Figure 2. TCDD suppresses the hypoxia-inducible enhancer in B-l cells. (A) B-l cells expressed luciferase activity in response to Dfx. Cells were treated with solvent control (methanol) or 0.0 to 12.5 nM TCDD for 30 hr and 125 uM Dfx for 24 hr before being assayed for luciferase activity. The responses to the treatments with or without TCDD were compared. The probability of type I error for comparison of each treatment with the appropriate control is labeled on the graph if it was less than 0.05. Confidence intervals represent one standard deviation of the mean. (B) In a parallel experiment, 40 uM CoClz was used to induce the hypoxia response in B-l cells. 14 3 50 — A _l I: 5 E 40 ‘ p<0.0001 E p<0.0001 E 30 ‘ 5 <1: 3 20 — 9 :8 g 10 - _J SolventorTCDD(nM) BLK 0 MeOH 0.125 1.25 12.5 Dfx(125.0uM) - + + + + + Luciferase Activity (1 OOXRLU) SOIVGI‘IIOI‘TCDD (nM)BLK O MeOH 0.125 1.25 12.5 CoCI2(40uM) - + + + + + measurement of its EROD activities in B-1 and Hepa 1 cells. EROD activity was maximally induced by exposure to 3.75 nM TCDD in B-l cells and by exposure to 375 pM TCDD in Hepa 1 cells after 24 hr exposure. To examine the effect of activation of the hypoxia response pathway on induction of AhR-mediated responses, various concentrations of Dfx were added 6 hr before the cells were treated with TCDD. A concentration-dependent reduction of TCDD-induced EROD activity was observed in both cell lines (Figure 3). Exposure to 250 uM Dfx reduced the maximal EROD activity by 67% in Hepa 1 cells. In B-l cells, 125 uM Dfx reduced the EROD activity induced by 3.75 nM TCDD to background levels. The concentrations of Dfx that caused the reduction of EROD activity in B-l cells were inversely related to the concentrations that induced luciferase activity as a measure of HIF-l-mediated transcriptional activation. Effects of Inducers of the Hypoxia Response Pathway on AhR-Mediated Reporter Gene Activity To confirm that the response of the endogenous reporter system, EROD, was not an artifact caused by the effects of Dfx on iron availability and the synthesis of cytochromes, the effect of HIF-l-mediated induction on the TCDD-inducible enhancer was studied using H1L1.1c2 cells. An increase of greater than 15-fold increase in luciferase activity was observed in H1L1.102 cells exposed to 5.0 nM TCDD for 24 hr. Luciferase induction was significantly inhibited by pre-incubation for 6 hr with 5 uM Dfx or by 10 uM CoClz (Figure 4). l6 Figure 3. Dfx inhibits the TCDD-induced EROD activity in B-1 and Hepa 1 cells. (A) B-l cells expressed EROD activity in response to TCDD. The cells were treated with concentrations of Dfx ranging from 0.0 to 125 uM for 30 hr and solvent (methanol) or 3.75 nM TCDD for 24 hr before being assayed for EROD activity. (B) Hepa 1 cells treated with concentrations of Dfx ranging from 0.0 to 125 uM for 30 hr and methanol or 375 pM TCDD for 24 hr before EROD assays. Pairwise comparisons of the responses to each of the Dfx treatments were compared to cells treated with TCDD alone by use of a one-tailed T-test. The probability of type I error is labeled on the graph if it was less than 0.05. Confidence intervals represent one standard deviation of the mean. 17 8O 5: 60 - o E £3: ,2 40 - g p<0.0001 S 0: 20 - P<°-°°°1p<0.0001 0— Solvent or Dfx (11M) MeOH 0 5 10 50 125 TCDD (3.75 nM) - + + + + + 100 80 - '6 E p=0.0018 3 60 “ I C G: 2 o 40 ‘ p<0.0001 (I) 0) II 20 ~ 0 ... Solvent or Dfx (uM) MeOH 0 125 250 TCDD (375 pM) - + + + + 18 Figure 4. Dfx and C002 suppress TCDD-inducible CYP1A1 enhancer in H1L1.1c2 cells. (A) H1L1.1c2 cells expressed luciferase activity in response to TCDD. The cells were treated with concentrations of Dfx ranging from 0.0 to 150 uM for 30 hr and methanol or 5 nM TCDD for 24 hr before being assayed for luciferase activity. (B) In a parallel experiment, luciferase activity induced by TCDD was studied in the presence of concentrations of CoClz ranging from 0.0 to 100 uM. The responses to TCDD with hypoxia response inducers were compared to the response to TCDD alone by use of a one-tailed T-test. The probability of type I error is labeled on the graph if it was less than 0.05. Confidence intervals represent one standard deviation of the mean. 19 —L O Luciferase Activity (1 OOOxRLU) O Solvent or Dfx (nM) MeOH TCDD(5.0 nM) .3 O p<0.0001 p<0.0001 No 5 50 150 -§ 0) 00 l l l Luciferase Activity (1 OOOxRLU) N OI Solvent or CoCl2 (uM) MeOH TCDD(5.0 nM) p=0.008 100 20 Cell Line Dependency of Interaction between the AhR and Hypoxia Pathways Responses of both endogenous and exogenous gene activities mediated through AhR in the presence of inducers of the hypoxia response pathway observed in B-l, Hepa 1, and H1L1.1c2 indicated that there is an interaction between AhR and hypoxia signaling pathways in these cell lines. However, the inhibitory effect of hypoxia response inducers on AhR-mediated gene expression was not observed in rat hepatoma H4IIE-luc cells. AhR-mediated transcriptional activation after treatment with TCDD can be measured either as endogenous enzyme activity (EROD) or via an exogenous reporter system (luciferase), which has been introduced into H4IIE-luc cells. When H4IIE-luc cells were exposed to Dfx or CoClz before being treated with TCDD, no reduction of either luciferase activity or EROD activity was observed (Figure 5). Interaction of T CDD and Dfx on Protein Complex Binding Activities to DRE3 and HIF-I Nuclear protein binding activity to DRE3 was examined in Hepa lcells (Figure 6A). Treating the cells for 1 hr with 5 nM TCDD increased the binding activity (lane 4), compared to that of untreated cells (lane 2) or carrier solvent control (DMSO) (lane3). There were two bands that are inducible by TCDD treatment, which is possible due to the presence of two isoforms of the AhR. The constitutive band is due to binding of the nuclear protein to single-stranded DRE (Denison and Yao, 1991). The DRE3 binding activity of TCDD-treated cells was reduced by 1 hr pre-treatment with 125 uM Dfx (lane 5). The specificity of DRE3 binding was examined using a ZOO-fold molar excess of 21 unlabelled DRE3 (lane 6). The presence of AhR in the DRE-binding complex was investigated using an AhR antibody (Figure 6B). The antibody to AhR caused an up-shift of the bands (lane 3), which demonstrates the presence of the AhR in the protein complex when it binds to DRE3 The DNA-binding activity to the HlF-l binding site was induced in Hepa 1 cells treated for 4 hr with 125 uM Dfx (Figure 7A, lane 3), relative to untreated cells (lane 2). One hour pre-incubation with 5 nM TCDD before Hepa 1 cells were treated with Dfx caused marked reduction of the Dfx-induced DNA-binding (lane 5 and 6). Carrier solvent (DMSO) alone and TCDD in DMSO had no effect on background HIF-l binding activity (lane 7 and 8). The specificity of nuclear protein binding to the HIF-l binding site was examined by using a ZOO-fold excess of unlabelled probe to competitively displace the labeled probe (lane 4). This demonstrates that the protein binding to the oligonucleotide was specific to the sequence of the probe used. To examine the identity of the DNA-binding protein complex, antibody to HIF-la was used. The antibody to HIP-10L caused the reduction of the protein complex binding to the probe (Figure 7B, lane 3), compared to the mouse IgG control (lane 4). This result demonstrates that HIF- 10L is necessary for binding of the nuclear protein complex with the HIF-l binding site. Furthermore, this demonstrates that the inducible bands contain the HIP-10L proteins. 22 Figure 5. Effect of hypoxia inducers on the AhR-mediated gene expression in H4IIE- luc cells. (A) H4IIE-luc cells increase DRE-mediated luciferase and EROD activity in response to TCDD. The cells were treated with concentrations of Dfx ranging from 0.0 to 125 uM for 30 hr and solvent control (methanol) or 1.25 nM TCDD for 24 hr before being assayed for luciferase or EROD activities. (B) H4IIE-luc cells were treated with concentrations of CoClz ranging from 0.0 to 100.0 uM instead of Dfx. Confidence intervals represent one standard deviation of the mean. 23 N 01 _8 O O ’5 — Luciferase A El :1 EROD o — 75 13 2 a S 15 - “a” .5 — 50 3’ <3? 10 6 ‘ 3 3 2 g — 25 V £3 5 ‘ 0 3 _l O J 0 Solvent or Dfx (uM) MeOH 0 1.25 12.5 125 TCDD (1.25 nM) - + + + + A 30 100 3 — Luciferase B (I 25 - : EROD c": — 75 an 0 <0 1: 20 - 8 3‘ E .5 15 — — 50 3’ :2 e _ 3 g 10 2 B — 25 v :93 5 - o 3 A I 0 . 0 Solvent or CoCl2 (uM) MeOH o 1 10 100 TCDD (1.25 nM) - + + + + 24 Figure 6A. Effect of Dfx on AhR:ARNT binding activity to DRE3. The electrophoresis mobility shift assay shows DRE probe alone (lane 1), nuclear protein from Hepa 1 cells with no treatment (lane 2), 1 hr carrier solvent (DMSO) treatment (Lane 3), and 1 hr 5 nM TCDD (lane 4). The effect of Dfx was examined in cells treated with 125 uM Dfx for 1 hr (lane 5). The specificity of DRE-binding was evaluated using ZOO-fold molar excess of unlabelled DRE probe (lane 6). IN 9: induced binding activity, C 9: constitutive binding, F 9: free probe. This figure is representative of more than three independent experiments. 25 Cold Competitor %‘ - - - - + 125 uM Dfx g - - - 1h - 5nMTCDD g - - 1h 1h 1h Solvent (DMSO) 0. - + + + + IN -> u '1 IN fl F-I Lane 26 Figure 6B. Presence of AhR in protein complexes binding to DRE3. Hepa 1 cells were either untreated (lane 1) or treated with 5 nM TCDD for 1 hr (lane 2 to 4). AhR antibody (lane 3) was used and compared to mouse IgG controls (lane 4). 27 Solvent control 5 nM TCDD 1h 1h 1h Antibody - AhR - IgG - - + lN-D on w- Lane 1 2 3 4 28 Figure 7A. Effect of TCDD on DNA-binding activity of HIF-l. Electrophoresis mobility shift assay with W18 probe, which contains the HIF-l binding site, alone (lane 1), with nuclear protein from untreated Hepa 1 cells (lane 2), or 4 hr 125 uM Dfx (lane 3). To study the effect of TCDD, cells were treated with 5 nM TCDD for 5hr (lane 5) or 8 hr (lane 6) and with 125 uM Dfx for 4 hr. DNA-binding activity in the cells treated with solvent (DMSO) alone (lane 7) was also examined. The specificity of HIF-l- binding was demonstrated using a ZOO-fold excess of unlabelled W18 probe (lane 4). IN 9: induced binding activity, NS 9: non-specific binding; F 9: free probe. This figure is representative of more than three independent experiments. 29 Cold Competitor 125 uM Dfx 5nMTCDD Solvent (DMSO) IN-t IMfi NS‘ r:-» Lane 30 Figure 7B. Presence of HIF-la in the protein complex that binds to HIF—l. Hepa 1 cells were untreated (lane 1) or exposed to 125 uM Dfx for 4 hr (lane 2 to 4). HIF-la antibody was used (lane 3) and compared to mouse IgG control (lanes 4). 31 125 uM Dfx Antibody IgG Lane 1 32 DISCUSSION Induction of the TCDD-activated AhR signaling pathway, which is represented by the induction of CYP1A1, alters gene expression controlled by the enhancer sequence containing DREs. One explanation of the pleiotropic responses of animals to exposure to TCDD is that there are multiple DRE-controlled genes which are regulated by AhR- ligand complex. This mechanism of action confines the effect of the AhR ligands to the regulation of genes specified by the DRE enhancer sequences. This limited scope of action, however, can not explain the wide spectrum of toxic responses, ranging from cell proliferation to apoptosis, caused by the AhR ligands, such as PCDHs. The correlation between CYP1A1 induction and species sensitivity to TCDD is poor. For example, guinea pigs, which are the most sensitive species to the lethal effects of TCDD, do not show induction of liver metabolizing enzymes (Poland and Knutson, 1982). It is, however, unlikely that all of the effects caused by T CDD are due to the direct response of DREs. The fact that many effects of TCDD can be described as alteration of cell growth and differentiation (Okey et al., 1994) suggests an interaction among multiple signaling pathways. Therefore, an alternative hypothesis that could explain the pleiotropic responses of animals to TCDD is cross talk among pathways with common mechanisms of signaling transduction. In this study, it has been demonstrated that the AhR and hypoxia pathways interact at the level of gene expression and such interaction is correlated with interference of DNA binding activity and nuclear complex formation. Reciprocal repression of gene expression of the two pathways was observed in B-l cells. 33 Inhibitory effects of the inducers of the hypoxia response pathway on AhR-mediated gene expression were also observed in Hepa 1 and H1L1.1c2 cells. This suggests that such an interaction is not unique to B-l cells. Although cytochrome p450, such as CYP1A1, are heme proteins whose production requires iron, repression of EROD activity in B 1 and Hepa 1 cells by Dfx was not caused by direct inhibition of protein synthesis because the luciferase reporter enzyme whose production is independent of iron was also reduced by Dfx in H1L1.102. Furthermore, the H4IIE-luc cells did not exhibit effect of Dfx on either EROD or luciferase activity when cells were treated with same concentrations of Dfx. These results suggest that the concentrations of Dfx used are sufficient to activate the hypoxia response pathway, yet did not affect CYP1A1 protein synthesis. AhR, ARNT, and HIP-10L belong to bHLH (basic-he]ix-loop-helix)/PAS protein family. The bHLH motif is characteristic of a family of proteins that function as modulators of cell proliferation and differentiation. PAS proteins are found in representative organisms of all five kingdoms and may play a role in determining target gene specificity (Hahn, 1998). PAS proteins are involved in development and differentiation (Sim group, trachealess) (Nambu et al., 1991; Isaac and Andrew, 1996), regulation of circadian clocks (Per, CLOCK) (Huang et al., 1995; King et al. 1997), sensing and responding to oxygen tension (HIP-la, EPAS-l/HLF) (Wenger and Gassmann, 1997; Tian et al., 1997), and steroid receptor signaling (SRC-l) (Yao et al., 1996). The myogenic bHLH proteins autoregulate their own expression and cross- regulate the expression of other family members (Olson, 1990). Since the secondary dimerization surface, PAS domain can provide the specificity for dimerization among 34 PAS/bHLH proteins (Pongratz et al., 1998). PAS proteins, therefore, may behave in a way similar to the myogenic bHLH proteins and interact with each other through the PAS domain. Since ARNT has been found to dimerize with many PAS proteins such as AhR, Siml, Sim2, HIF-la, and EPAS-l (Hogenesch et al., 1997; Jiang et al., 1996; Moffett et al., 1997; Sogawa et al., 1995; Tian et al., 1997), it may act as a central regulator of PAS protein-dependent pathways. Eukaryotic gene regulation can be viewed as an interplay between opposing activating and repressing influences. Transactivation of genes is controlled at many levels, but typically involves binding by transcription factors to specific regulatory cis- elements. While the cis-elements can be specific to a particular set of genes, the transcription factors may be less diverse and shared by several pathways. Sharing of a few common transcription factors by many transcription regulation pathways also provides the advantage of network regulation and more sophisticated, fine-tuned controls. This sharing also leads to the observed phenomenon known as “squelching”, which refers to alteration of transcription by sequestering limiting components required for transcriptional activation or repression away from the promoter in the affected gene (Cahill et al., 1994). For instance, the estrogen hormone receptor has been shown to inhibit the transcriptional activation mediated by the progesterone and glucocorticoid receptors (Meyer et al., 1989). Squelching could be a common mechanism employed by transcription factors to down-regulate promoters whose activity is governed by coactivators of low abundance. Reciprocal repression of the AhR- and HIF-l-mediated pathways was observed in B-l cells, but the degrees of interaction were different. Inhibition of AhR-mediated gene 35 expression by induction of the hypoxia response pathway was greater than the inhibition of the hypoxia response pathway by TCDD. HIP-lo: has been found to bind to ARNT with greater affinity than does AhR (Gradin et al., 1996). This differential binding affinity to ARNT may explain the stronger inhibition by hypoxia response inducers on the AhR pathway, and may serve as a regulatory mechanism in addition to dimerization specificity. Results of electrophoresis mobility shift assays provided further elucidation of the mechanism of interaction between the two pathways. The reduction of hypoxia-induced DNA-binding activity by TCDD (Figure 7A) and TCDD-induced DNA-binding activity by Dfx (Figure 6A) offers support for the hypothesis that interaction between the AhR- and HIF -1-mediated pathways is due to recruitment of ARNT (Hogenesch et al., 1997). Therefore, ARNT may serve as a nuclear integrator and modulator of various signal transduction pathways. Alternatively, these data do not exclude the possibility that other commonly involved transcription activators and coactivators are also recruited. For instance, the coactivator CBP/p300 has been found to be associated with both the AhR- and HIF -1-mediated pathways, acting synergistically with the transcription factors and basal transcription machinery (Bunn et al., 1998; Ebert and Bunn, 1998; Kallio et al., 1998; Kobayashi et al., 1997). Therefore, besides potential competition for ARNT, the AhR- and HIF -1-mediated pathways may also interact through squelching of CBP or other shared coactivators. Unlike in B-l, Hepa 1, and H1L1.102 cells, AhR-mediated gene expression in H4IIE-luc cells was not affected by the induction of hypoxia response pathway. This suggests that the interaction between the AhR and hypoxia pathways might be different 36 among cell lines, tissues, or species. The degree of interaction among ARNT-dependent pathways may depend on the abundance of ARNT in the cells. In H4IIE cells, the ratio of AhR to ARNT is 0.3, compared to 10 in Hepa 1 cells (Holmes and Pollenz, 1997), which indicates that ARNT exists in excess in H4IIE cells. However, to elucidate this hypothesis, more needs to be known about the abundance of HIF-la in cells. Another explanation for the lack of effect of hypoxia inducers on the induction by TCDD is that H4IIE-luc cells are defective in the hypoxia pathway, or insensitive to hypoxia inducers. In conclusion, our study provided evidence for a potential mechanism of action for T CDD toxicity. In this model, TCDD acts as a disrupter of multiple gene regulation pathways through its recruitment and sequestering of ARNT by activating AhR. TCDD, by altering the transcription regulation network, therefore can cause diverse toxicological effects by changing the relative and absolute rates of transcription of a variety of genes expressed under normal physiological conditions (Figure 9). This model offers a plausible explanation for the wide range of toxic effects of TCDD. For example, VEGF, a growth factor that regulates angiogenesis, is regulated by HIF-la and ARNT in response to oxygen availability. ARNT- and HIF-la-null mice can not survive gestation due to the defects in vasculature development (Maltepe et al., 1997; Iyer et al., 1998). TCDD has been reported to cause reduced vasculature in developing chicks and medaka (Orizias latipes) (Walker et al., 1997; Cantrell et al., 1996). TCDD is also known to elicit a wasting syndrome, which is partially attributed to its down regulation of glucose transporters (Matsumura, 1995). At least one of the glucose transporters, GLUT-l, has been found to be regulated through HIP-0L and ARNT (Ebert et al., 1995). Furthermore, this hypothesis could also explain the wide range of sensitivities among tissues and 37 species. As more genes regulated by ARNT to be discovered and more ARNT- interacting transcription factors to be characterized, it should be possible to offer better and more complete explanations of TCDD toxicity. 38 Figure 8. A potential mechanism of action for TCDD. In this model, TCDD acts through AhR to sequester ARNT and reduce the abundance of free ARNT that can be used by other ARNT-dependent pathways, such as the hypoxia pathway and other ones possibly involving the members of the PAS protein family. The activation of the AhR pathway by ligands like TCDD can also reduce the availability of common cofactors such as CBP, and therefore affect gene regulations involving non-PAS proteins. This disturbance of multiple gene regulation pathways at the cellular level may be manifested in vivo as the pleiotropic effects of TCDD. 39 530.5 mega” OmQOhOmDE 9.65935 Emu 9:. III co ceases. "no.2 maowoemoo Samoa 8:0st 8388M sonmEOmSub... .850 08.35 38%: .52 SEER WM Q % sonnaowgfi. .550 230800 55066 40 REFERENCES Abbott, B. D., and Birbaum, L. S. (1990). Effects of TCDD on embryonic ureteric epithelial EGF receptor expression and cell proliferation. T eratology 41, 71-84. Asman, D. C., Takimoto, K., Pitot, H. C., Dunn, T. J ., and Lindahl, R. (1993). Organization and characterization of the rat class 3 aldehyde dehydrogenase gene. J. Biol. Chem. 268, 12530-12536. Bombick, D. W., Madhukar, B. V., Brewster, D. W., and Matsumura, F. (1985). TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) causes increases in protein kinases, particularly protein kinase C, in the hepatic plasma membrane of the rat and the guinea pig. Biochem. Biophys. Res. Commun. 127, 296-302. Bunn, H. F., Gu, J ., Huang E., Park, J.-W., and Zhu, H. (1998). Erythropoietin: A model system for studying oxygen-dependent gene regulation. J. Exp. Biol. 201, 1197- 1201. Cahill, M. A., Ernst, W. H., Janknecht, R., and Nordheim, A. (1994). Regulatory squelching. FEBS Letter. 344, 105-108. Cantrell, S. M. Lutz, L. H., Tillitt, D. E., and Hannink, M. (1996). Embryotoxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD): The embryonic vasculature is a physiological target for TCDD-induced DNA damage and apoptotic cell death in Medaka (Orizias latipes). T oxicol. Appl. Pharmacol. 141, 23-34. 41 Carver, L. A., and Bradfield, C. A. (1997). Ligand-dependent interaction of the aryl hydrocarbon receptor with a novel immunophilin homolog in vivo. J. Biol. Chem. 272, 11452-11456. Choi, E., Toscano, D., Ryan, J., Riedel, N., and Toscano, W. (1991). Dioxin induces transforming growth factor-0t in human keratinocytes. J. Biol. Chem. 266, 9591- 9597. Clark, G. C., Taylor, M. J., Trischer, A. M., and Lucier, G. W. (1991). Tumor necrosis factor involvement in 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated endotoxin hypersensitivity in C57BL/6J mice congenic at the Ah locus. T oxicol. Appl. Pharmacol. 1 l l , 422-431 . Denison, M. S., Fisher, J. M., and Whitlock, J. P. Jr. (1989). Protein-DNA interactions at recognition sites for the dioxin-Ah receptor complex. J. Biol. Chem. 264, 16478- 16482. Denison, M. S., and Yao, E. F. (1991). Characterization of the interaction of transformed rat hepatic cytosolic Ah receptor with a dioxin responsive transcriptional enhancer. Arch. Biochem. Biophys. 284, 158-166. Dong, L., Ma, Q., and Whitlock, J. P. Jr. (1996). DNA binding by the heterodimeric Ah receptor: Relationship to dioxin-induced CYP1A1 transcription in vivo. J. Biol. Chem. 271, 7942-7948. Ebert, B. L., Firth, J. D., and Ratcliffe, P. J. (1995). Hypoxia and mitochondrial inhibitors regulate expression of glucose transporter-1 via distinct cis-acting sequences. J. Biol. Chem. 270, 29083-29089. 42 Ebert, B. L., and Bunn, H. F. (1998). Regulation of transcription by hypoxia requires a multiprotein complex that includes hypoxia-inducible factor 1, an adjacent transcription factor, and p300/CREB binding protein. Mol. Cell. Biol. 18, 4089- 4096. Enan, E., and Matsumura, F. (1993). 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD)- induced alterations in protein phosphorylation in guinea pig adipose tissue. J. Biochem. T oxicol. 8, 89-99. Favreau, L. V., and Pickett, C. B. (1991). Transcriptional regulation of the rat NAD(P)H:quinone reductase gene. Identification of regulatory elements controlling basal level expression and inducible expression by planar aromatic compounds and phenolic antioxidants. J. Biol. Chem. 266, 4556-4561. Forsythe, J. A., Jiang, B-H, Iyer, N. V., Agani, F., Leung, S.W., Koos, R. D., and Semenza, G. L. (1996). Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1.Mol. Cell. Biol. 16, 4604-4613. Garrison, P. M., Tullis, K., Aarts, J. M. M. J. G., Brouwer, A., Giesy, J. P., and Dension, M. S. (1996). Species-specific recombinant cell lines as bioassay systems for the detection of 2,3,7,8-tetrachlorodibenzo-p-dioxin-like chemicals. Fundam. Appl. T oxicol. 30, 194-203. Giesy, J. P., Ludwig, J. P., and Tillitt, D. E. (1994a). Dioxins, dibenzofurans, PCBs and colonial, fish-eating water birds. In Dioxins and Health. (A. Schecter, Ed), pp.249-307. Plenum Press, New York. Giesy, J. P., Ludwig, J. P., and Tillitt, D. E. (1994b). Deformities in birds of the Great Lakes regiond: Assigning causality. Environ. Sci. T echnol. 28, 128A-135A. 43 Goldberg, M. A., and Schneider, T. J. (1994). Similarities between the oxygen-sensing mechanisms regulating the expression of vascular endothelial grth factor and erythropoietin. J. Biol. Chem. 269, 4355-4359. Gradin, K., McGuire, J ., Wenger, R. H., Kvietikova, I., Whitelaw, M. L., Tofigard, R., Tora, L., Gassmann, M., and Poellinger, L. (1996). Functional interference between hypoxia and dioxin signal transduction pathways: Competition for recruitment of the Amt transcription factor. Mol. Cell. Biol. 16, 5221-5231. Guillemin, K., and Krasnow, M. A. (1997). The hypoxic response: Huffing and hifing. Cell 89, 9-12. Hahn, M. E. (1998). The aryl hydrocarbon receptor: A comparative perspective. Comp. Biochem. Physiol. 121C(3), 23-53. Hankinson, O. (1995). The aryl hydrocarbon receptor complex. Annu. Rev. Pharmacol. T oxicol. 35, 307-340. Hogenesch, J. H, Chan, W. K., Jackiw, V. H., Brown, R. C., Gu, Y.-Z., Pray-Grant, M., Perdew, G. H., and Bradfield, C. A. (1997). Characterization of a subset of the basci-helix-loop-helix-PAS superfamily that interacts with components of the dioxin signaling pathway. J. Biol. Chem. 272, 8581-8593. Holmes, J. L., and Pollenz, R. S. (1997). Determination of aryl hydrocarbon receptor nuclear translocator protein concentration and subcellular localization in hepatic and nonhepatic cell culture lines: Development of quantitative western blotting protocols for caculation of aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator protein in total cell lysates. Mol. Pharmacol. 52, 202-21 1. Huang, L. E., Gu, J., Schau, M., and Bunn, H. F. (1998). Regulation of hypoxia-inducible factor 10. is mediated by an Oz-dependent degradation domain via the ubiquitin- proteasome pathway. Proc. Natl. Acad. Sci. USA 95, 7987-7992. Huang, Z. J., Curtin, K. D., and Rosbash, M. (1995). PER protein interactions and temperature compensation of a circadian clock in Drosophila. Science 267, 1169- 1172. Isaac, D. D., and Andrew, D. J. (1996). Tubulogenesis in Drosophila: A requirement for the trachealess gene product. Gene Dev. 10, 103-107. Iyer, N. V., Kotch, L. E., Agani, F., Leung, S. W., Laughner, E., Wenger, R. H., Gassmann, M., Gearhart, J. D., Lawler, A. M., Yu., A. Y., and Semenza, G. L. (1998). Cellular and developmental control of Oz homostasis by hypoxia- inducible factor 10L. Genes Dev. 12, 149-162. Jiang, B.-H., Rue, B, Wang, G. L., Roe, R., and Semenza, G. L. (1996). Dimerization, DNA binding, and transaction properties of hypoxia-inducible factor 1. J. Biol. Chem. 271, 17771-17778. Kallio, P. J., Okamoto, K., O'Brien, 8., Carrero, P., Makino, Y., Tanaka, H., and Poellinger, L. (1998). Signal transduction in hypoxic cells: inducible nuclear translocation and recruitment of the CBP/p300 coactivator by the hypoxia- inducible factor-10L. EMBO J. 17, 6573-6586. Kallio, P. J ., Pongratz, I., Gradin, K., McGuire, J ., and Poellinger, L. (1997). Activation of hypoxia-inducible factor 10L: Posttranscriptional regulation and conformational change by recruitment of the Amt trancription factor. Proc. Natl. Acad. Sci. USA 94, 5667-5672. 45 Kennedy, S. W., and Jones, S. P. (1994). Simultaneous measurement of cytochrome P4501A catalytic activity and total protein concentration with a fluorescence plate reader. Anal. Biochem. 222, 217-223. King, D. P., Zhao, Y., Sangoram, A. M., Wilsbacher, L. D., Tanaka, M., Antoch, M. P., Steeves, T. D. L., Vitaterna, M. H., Komhauser, J. M., Lowrey, P. L., Turek, F. W., and Takahashi, J. S. (1997). Positional cloning of the mouse circadian clock gene. Cell 89, 641-653. Kobayashi, A., Numayama-Tsuruta, K., Sogawa, K., and Fujii-Kuriyama, Y. (1997). CBP/p300 functions as a possible transcriptional coactivator of Ah receptor nuclear translocator (Amt). J. Biochem. 122, 703-710. Kozak, K. R., Abbott, B. D., and Hankinson, O. (1997). ARNT-deficient mice and placental differentiation. Dev. Biol. 191, 297-305. Ludwig J. P., Kurita-Matsuba, H., Auman, H. J ., Ludwig, M. E., Summer, C. L., Giesy, J. P., Tillitt, D. E., and Jones, P. D. (1996). Deformities, PCBs, and TCDD- equivalents in double-crested cormorants (Phalacrocorax auritus) and Caspian terns (Hydroprogne caspia) of the Upper Great Lakes 1986-1991: Testing a cause-effect hypothesis. J. Great Lakes Res. 22, 172-197. Ma, Q., and Whitlock, J. P. Jr. (1997). A novel cytoplasmic protein that interacts with the Ah receptor, contains tetraicopeptide repeat motifs, and augments the transcriptional response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. J. Biol. Chem. 272, 8878-8884. 46 Maltepe, E., Schmidt, J. V., Baunoch, D., Bradfield, C. A., and Simon, M. C. (1997). Anormal angiogenesis and response to glucose and oxygen deprivation in mice lacking the protein ARNT. Nature 386, 403-407. Matsumura, F. (1995). Mechanism of action of dioxin-type chemicals, pesticides, and other xenobiotics affecting nutritional indexes. Am. J. Clin. Nutr. 61(suppl.), 6958-7018. Maxwell, P. H., Dachs, G. U., Gleadle, J. M., Nicholls, L. G., Harris, A. L., Stratford, I. J., Hankinson, O., Pugh, C. W., and Ratcliffe, P. J. (1997). Hypoxia-inducible factor-l modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. Proc. Natl. Acad. Sci. USA 94, 8104-8109. Meyer, M.-E., Gronemeyer, H., Turcotte, B., Bocquel, M.-T., Tasset, D., and Charnbon, P. (1989). Steroid hormone receptors compete for factors that mediate their enhancer function. Cell 57, 433-442. Moffett, P., Reece, M., and Pelletier, J. (1997). The murine Sim-2 gene product inhibits transcription by active repression and functional interference. Mol. Cell. Biol. 17, 4933-4947. Nambu, J. R., Lewis, J. O., Wharton, K. A. Jr., and Crews, S. T. (1991). The Drosophila single-minded gene encodes a helix-loop-helix protein that acts as a master regulator of CNS midline development. Cell 67, 1157-1167. Nebert, D. W., Puga, A., and Vasiliou, V. (1993). Role of the Ah receptor and the dioxin- inducible [Ah] gene battery in toxicity, cancer, and signal transduction Ann. NY Acad. Sci. 685, 624-640. 47 Olson, E. N. (1990). MyoD family: A paradigm for deve10pment? Genes Dev. 4, 1454- 1461. Okey, A. B., Riddick, D. S., and Harper, P. A. (1994). Molecular biology of the aromatic hydrocarbon (dioxin) receptor. Trends Pharmacol. Sci. 15, 226-232. Paulson, K. E., Darnell, J. E. Jr., Rushmore, T., and Pickett, C. B. (1990). Analysis of the upstream elements of the xenobiotic compound-inducible and positionally regulated glutathione S-transferase Ya gene. Mol. Cell. Biol. 10, 1841-1852. Pohjanvirta, R., and Tuomisto J. (1994). Short-term toxicity of 2,3,7,8- tetrachlorodibenzo-p-dioxin in laboratory animals: Effects, mechanisms, and animal models. Pharmacol. Rev. 46, 483-549. Poland, A., Glover, E., and Kende, A. S. (1976). Stereospecific, high affinity binding of 2,3,6,8-tetrachlorodibenzo-p-dioxin by hepatic cytosol. J. Biol. Chem. 251, 4936- 4946. Poland, A. and Knutson, J. C. (1982). 2,3,7,8-Tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: Examination of the mechanism of toxicity. Annu. Rev. Pharmacol. T oxicol. 22, 517-554. Pongratz, I., Antonsson, C., Whitelaw, M. L., and Poellinger, L. (1998). Role of the PAS domain in regulation of dimerization and DNA binding specificity of the dioxin receptor. Mol. Cell. Biol. 18, 4079-4088. Probst, M. R., Reisz-Porszasz, S., Agbunag. R. V., Ong, M. S., and Hankinson, O. (1993). Role of the aryl hydrocarbon receptor nuclear translocator protein in aryl hydrocarbon (dioxin) receptor action. Mol. Pharmacol. 44, 511-518. 48 Quattrochi, L. C., and Tukey, R. H. (1989). The human cytochrome Cyp1A2 gene contains regulatory elements responsive to 3-methylcholanthrene. Mol. Pharmacol. 36, 66-71. Safe, S. (1986). Comparative toxicology and mechanism of action of polychlorinated dibenzo-p-dioxins and dibenzofurans. Annu. Rev. Pharmacol. T oxicol. 26, 371- 399. Safe, S. (1990). Polychlorinated biphenyls (PCBS), dibenzo—p-dioxins (PCDDs), dibenzofiirans (PCDFs), and related compounds: environmental and mechanistic considerations which support the development of toxic equivalency factors (TEFS). Toxicol. 21, 51-88. Salceda, 8., Beck, 1., and Caro, J. (1996). Absolute requirement of aryl hydrocarbon receptor nuclear translocator protein for gene activation by hypoxia. Arch. Biochem. Biophys. 334, 389-394. Salceda, S., and Caro, J. (1997). Hypoxia-induced factor 10L (HIF -10t) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions. J. Biol. Chem. 272, 22642-22647. Sanderson, J. T., Aarts, J. M. M. J. G., Brouwer, A., Froese, K. L., Denison, M. S., 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 oxicol. Appl. Pharmacol. 137, 316-325. Schmidt, J. V., and Bradfield, C. A. (1996). Ah receptor signaling pathways. Annu. Rev. Cell Dev. Biol. 12, 55-89. 49 Semenza, G. L. (1994). Regulation of erythropoietin production. Hematol. Oncol. Clin. N. Am. 8, 863-884. Semenza, G. L., Nejfflt, M. K., Chi, S. M., and Antonarakis, S. E. (1991). Hypoxia- inducible nuclear factors bind to an enhancer element located 3‘ to the human erythropoietin gene. Proc. Natl. Acad. Sci. USA 88, 5680-5684. Semenza, G. L., Roth, P. H., Fang, H.-M., and Wang, G. L. (1994). Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J. Biol. Chem. 269, 23757-23763. Shweiki, D., Itin, A., Soffer, D., and Keshet, E. (1992). Vascular endothelial grth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 359, 843-845. Sogawa, K., Nakano, R., Kobayashi, A., Kikuchi, Y., Ohe, N., Matsushita, N., and Fujii- Kuriyama, Y. (1995). Possible function of Ah receptor nuclear translocator (Amt) homodimer in transcriptional regulation. Proc. Natl. Acad. Sci. USA 92, 1936- 1940. Tian, H., McKnight, S. L., and Russell, D. W. (1997). Endothelial PAS domain protein 1 (EPASl), a transcription factor selectively expressed in endothelial cells. Genes Dev. 11, 72-82. Walker, M. K., Pollenz, R. S., and Smith, S. M. (1997). Expression of the aryl hycarbon receptor (AhR) and AhR nuclear translocator during chick cardiogenesis is consistent with 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced heart defects. T oxicol. Appl. Pharmacol. 143, 407-419. 50 Wang, G. L., and Semenza, G. L. (1993). Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia. J. Biol. Chem. 268, 21513- 21518. Wenger, R. H., and Gassmann, M. (1997). Oxygen(es) and the hypoxia-inducible factor— 1. Biol. Chem. 378, 609-616. Whitlock, J. P. Jr. (1987). The regulation of gene expression by 2,3,7,8- tetrachlorodibenzo-p-dioxin. Pharmacol. Rev. 39, 147-161. Whitlock, J. P. Jr. (1990). Genetic and molecular aspects of 2,3,7,8-tetrachlorodibenzo-p- dioxin action. Annu. Rev. Pharmacol. T oxicol. 30, 25,1-277. Whitlock, J. P. Jr. (1993). Mechanistic aspects of dioxin action. Chem. Res. T oxicol. 6, 754-763. Wood, S. M., Gleadle, J. M, Pugh, C. W., Hankinson, O., and Ratcliffe, P. J. (1996). The role of the aryl hydrocarbon receptor nuclear translocator (ARNT) in hypoxic induction of gene expression. J. Biol. Chem. 271, 15117-15123. Yao, T.-P., Ku, G., Zhou, N., Scully, R., and Livingston D. M. (1996). The nuclear hormone receptor co-activator SRC-l is a specific target of p300. Proc. Natl. Acad. Sci. USA. 93, 10626-10631. 51 "IIIIILIIIIIIIIIIIIIIIII