1: Toxicol Sci. 2005 Nov 16; [Epub ahead of print] AhR-Agonist-Induced Transcriptional Changes of Genes Involved in Thyroid Function in Primary Porcine Thyrocytes. P P, T K, C B, K E, C F, C HV, Hombach-Klonisch S. Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany. The Ah receptor (AhR) is a ligand transcription factor mediating toxic effects of chemicals such as dioxins. The 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD) and the coplanar polychlorinated biphenyl 126 (PCB 126) are member of the polyhalogenated aromatic hydrocarbons family exerting a variety of toxic effects in tissue- and specie-specific manner including thyroid function. In the present study, we aimed to investigate the effects of TCDD (1 and 10 nM) and dioxin-like PCB 126 (306 nM) on AhR signaling pathway and on the gene expression profiles of key factors involved in thyroid function, including thyroglobulin (TG), thyroid peroxidase (TPO), sodium iodide symporter (NIS), TSH receptor (TSHR) and cathepsins (Cat B and L), using a primary porcine thyrocyte culture as experimental model. AhR and ARNT expression was detected both as mRNA and on the protein level. Expression did not vary upon treatment with either TCDD or PCB 126. However, treatment with TCDD and PCB 126 induced an AhR signaling response as indicated by the expression of the AhR-target gene cytochrome p450 1A1 (CYP1A1). Both 10 nM TCDD and PCB 126 treatment induce a significant down-regulation in the expression of NIS and cathepsin B without affecting any of the other parameters investigated. In conclusion, these data indicate that (a) thyrocytes are targets of TCDD and TCDD-like compounds and (b) there is evidence for two independent most likely AhR-mediated molecular mechanisms, by which these compounds negatively interfere with thyroid function. PMID: 16291828 [PubMed - as supplied by publisher] --------------------------------------------------------------- 2: Eur J Obstet Gynecol Reprod Biol. 2005 Nov 8; [Epub ahead of print] The association of endometriosis risk and genetic polymorphisms involving dioxin detoxification enzymes: A systematic review. Guo SW. Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, MS 756, Milwaukee, WI 53226-0509, USA. Genetic polymorphisms involving genes encoding for dioxin detoxification enzymes have been implicated as a risk factor for endometriosis, but individual studies have been equivocal and controversial. We therefore performed a systematic review of 10 studies on association of endometriosis and various genes involved in dioxin detoxification process excluding GSTM1/GSTT1. We found that almost all genetic variants involving CYP1A1, CYP2E1, EPHX1, AHR, ARNT, AHRR, and NAT1 that have been investigated by single studies show no association with endometriosis. Two genetic variants were reported to be associated with endometriosis, with each variant only investigated by a single study and there has been no independent confirmation so far. For CYP1A1 MspI polymorphisms, women with +/- and +/+ genotype have about 40% of increased risk of endometriosis as compared with women of -/- genotype. However, there is no strong indication that CYP1A1 MspI polymorphism is consistently associated with endometriosis. For NAT2 polymorphisms, there is no evidence that it is associated with endometriosis. PMID: 16289302 [PubMed - as supplied by publisher] --------------------------------------------------------------- 3: J Reprod Dev. 2005 Oct;51(5):593-605. Effects of exposure in utero to bisphenol a on the expression of aryl hydrocarbon receptor, related factors, and xenobiotic metabolizing enzymes in murine embryos. Nishizawa H, Imanishi S, Manabe N. Research Unit for Animal Life Sciences, Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo. To evaluate the effects of bisphenol A (BPA), a candidate endocrine disruptor (ED), on embryonic development, we examined the mRNA expression levels of the aryl hydrocarbon receptor (AhR; which binds with many EDs and plays crucial roles in their metabolism) and related factors [aryl hydrocarbon receptor repressor (AhRR) and AhR nuclear translocator (Arnt)], xenobiotic metabolizing enzymes [XMEs; cytochrome P450 1A1 (CYP1A1) and UDP-glucuronosyltransferase, and the glutathione S-transferase Ya subunit (GST)], in murine embryos exposed in utero to BPA (0.02, 2, 200, and 20,000 mug/kg/day) and 17beta-estradiol (E2; 5 mug/kg/day, used as a positive control) at 6.5-13.5 or 6.5-17.5 days post coitum (dpc) using the quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) method. Protein levels of CYP1A1 and GST in embryonic livers were estimated by Western immunoblotting. Exposure in utero to BPA [0.02 (1/100 dose of environmental exposure), 2, 200, and 20,000 mug/kg/day] increased AhR mRNA expression in the cerebra, cerebella, and gonads (testes and ovaries) of male and female mid-and late-developmental stage (14.5- and 18.5-dpc, respectively) embryos. BPA dose-independently up-regulated the expression of AhRR and Arnt in mid- and late-stage embryos. BPA had no remarkable effect on the mRNA levels of XMEs in mid-stage embryos, but dose-dependently up-regulated the expression in late-stage embryos. Moreover, the protein levels of these enzymes in the livers of late-stage embryos were increased. The present findings revealed that exposure to BPA in utero disrupts the expression of AhR and related factors and of xenobiotic metabolizing enzymes, and that mid-stage embryos, in the organogenic stage, are sensitive to BPA. PMID: 16284450 [PubMed - in process] --------------------------------------------------------------- 4: Biochem Pharmacol. 2005 Nov 25;70(11):1623-33. Epub 2005 Oct 13. Role of the carboxy-terminal transactivation domain and active transcription in the ligand-induced and ligand-independent degradation of the mouse Ah(b-1) receptor. Pollenz RS, Popat J, Dougherty EJ. Department of Biology, University of South Florida, BSF 110, 4202 E Fowler Ave, Tampa, FL 33620, USA. To assess the importance of transactivation domains (TAD), DNA binding and transcription on the degradation of the AH receptor (AHR), Hepa-1 cells were pre-treated with actinomycin D (AD) or cycloheximide (CHX) and exposed to 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD). AD or CHX did not affect nuclear localization or DNA binding of the AHR but inhibited ligand-induced degradation. In contrast, AD or CHX did not inhibit geldanamycin (GA) induced degradation of the AHR. To assess the role of the COOH-terminal TAD in AHR degradation, stop codons were placed at nucleotide 1501 and 1921 of the Ah(b-1) AHR coding region to generate AHR(500) and AHR(640). Stable cell lines were generated and exposed to TCDD. Cells expressing AHR(500) did not induce CYP1A1 protein, but exhibited significant degradation of AHR(500). Cells expressing AHR(640) induced CYP1A1 protein to 50% of the level of cells expressing wild type AHR and exhibited significant degradation of AHR(640). Importantly, AD and CHX did not inhibit the TCDD-induced degradation of either AHR(500) and AHR(640) and these receptors showed a more rapid profile of ligand-induced degradation compared to cells expressing wild type AHR. TCDD exposure to Hepa-1 cells with reduced aryl hydrocarbon receptor nuclear translocator (ARNT), showed ligand-induced degradation of the AHR that was not blocked by AD. However, AD inhibited TCDD-induced degradation when ARNT expression was restored. These results show that multiple mechanisms exist for the ligand and GA-induced degradation of the AHR and suggest that ligand-induced degradation can switch between two mechanisms depending on the presence of a functional TAD and the binding to DNA. PMID: 16226227 [PubMed - in process] --------------------------------------------------------------- 5: Mol Pharmacol. 2005 Oct 24; [Epub ahead of print] AHR ACTIVATION INHIBITS REGENERATIVE GROWTH. Mathew LK, Andreasen EA, Tanguay RL. Oregon State University. There is considerable literature supporting the conclusion that inappropriate activation of the aryl hydrocarbon receptor (AHR) alters cellular signaling. We have previously established that fin regeneration is specifically inhibited by TCDD in adult zebrafish and have used this in vivo end point to evaluate interactions between AHR and growth controlling pathways. Because there are experimental limitations in studying regeneration in adult animals, we have developed a larval model to evaluate the effect of AHR activation on tissue regeneration. Two day old zebrafish regenerate their amputated caudal fins within 3 days. Here we demonstrate that TCDD specifically blocks regenerative growth in larvae. The AHR pathway in zebrafish is considerably more complex than in mammals, with at least three zebrafish AHR genes (zfAHR1a, zfAHR1b and zfAHR2), and two ARNT genes (zfARNT1 and zfARNT2). Although it was presumed that the block in regeneration was mediated by AHR activation, it had not been experimentally demonstrated. Using antisense morpholinos and mutant fish lines, we report that zfAHR2 and zfARNT1 are the in vivo dimerization partners that are required for inhibition of regeneration by TCDD. Several pathways including FGF signaling are essential for fin regeneration. Even though impaired FGF signaling and TCDD exposure both inhibit fin regeneration, their morphometric response are distinct suggesting that the mechanisms of impairment are different. With the plethora of molecular and genetic techniques that can be applied to larval stage embryos, this in vivo regeneration system can be further exploited to understand cross talk between AHR and other signaling pathways. PMID: 16214955 [PubMed - as supplied by publisher] --------------------------------------------------------------- 6: Arch Toxicol. 2005 Oct 5;:1-6 [Epub ahead of print] Tissue distribution and function of the Aryl hydrocarbon receptor repressor (AhRR) in C57BL/6 and Aryl hydrocarbon receptor deficient mice. Bernshausen T, Jux B, Esser C, Abel J, Fritsche E. Arbeitsgruppe Toxikologie, IUF, Auf'm Hennekamp 50, 40225, Dusseldorf, Germany, josef.abel@uni-duesseldorf.de. The Aryl hydrocarbon receptor repressor (AhRR) is a new member of bHLH-PAS proteins which is important in the regulation of cell growth and differentiation. The AhRR shares structural similarities with Aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (ARNT). The AhRR is thought to be involved in transcriptional control of AhR-regulated genes by sequestering ARNT. Most of the knowledge of regulation and function of the AhRR is from studies in cell lines. Here, we report the tissue distribution of AhRR in AhR deficient and wild type C57BL/6 mice. In addition, the inducibility of the AhRR and Cytochrome P450 (CYP) 1A1 in response to benzo(a)pyrene (B(a)P) (10 mg/kg bw i.p.) was investigated. The results show that the AhRR mRNA expression pattern in untreated C57BL/6 mice varies across tissues with high levels in hearts and brains. In other tissues, AhRR mRNA expression was low. In contrast to wild-type animals, the tissue levels in AhR(-/-) mice were about two to three orders of magnitude lower. Treatment of wild-type animals with B(a)P resulted in an induced AhRR expression in liver, spleen, lung and ovary. No significant induction of AhRR mRNA was found in brain and heart tissues, which have a constitutively high level of AhRR expression. Simultaneous measurements of CYP1A1 and AhRR mRNA expression do not strongly support the view that the AhRR tissue pattern triggers the tissue specific responsiveness of AhR-regulated genes to B(a)P treatment. PMID: 16205913 [PubMed - as supplied by publisher] --------------------------------------------------------------- 7: J Biochem Mol Toxicol. 2005;19(4):244-55. Early steps in bilirubin-mediated apoptosis in murine hepatoma (Hepa 1c1c7) cells are characterized by aryl hydrocarbon receptor-independent oxidative stress and activation of the mitochondrial pathway. Oakes GH, Bend JR. Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1. Unconjugated bilirubin (UCB), the end product of heme catabolism, causes apoptosis in cells of the central nervous system, endothelial cells, and hepatotoma cells. However, the molecular mechanisms that contribute to UCB cytotoxicity remain unclear. The purpose of this study was to characterize the sequence of early events leading to UCB-mediated cytotoxicity in murine hepatoma Hepa 1c1c7 cells. In the present study, UCB (5-50 microM) was found to markedly increase the intracellular generation of reactive oxygen species (ROS) in a concentration-dependent manner, which is significantly elevated by 30 min post-treatment. This generation of ROS by UCB is not dependent on aryl hydrocarbon receptor (Ahr) signaling, as cells deficient in the Ahr (C12 cells) or the Ahr nuclear translocator protein (Arnt; C4 cells) were as efficient at generating ROS as wild type (WT) Hepa 1c1c7 cells. Mitochondrial membrane depolarization, evaluated with the lipophilic cationic dye, JC-1, occurred at least by 2 h after treatment with 50 muM UCB. Analysis of the caspase cascade demonstrated that activation of caspase-9 preceded activation of caspase-3. No conversion of procaspase-2 to active caspase-2 was detected in this study. These results demonstrate that UCB-mediated apoptosis in Hepa 1c1c7 cells is associated with increased oxidative stress and that caspase-9, and definitely not caspase-2, is the initiator caspase for apoptosis in UCB-treated Hepa 1c1c7 cells. Copyright 2005 Wiley Periodicals, Inc PMID: 16173058 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 8: Arch Biochem Biophys. 2005 Oct 1;442(1):59-71. Evidence that ligand binding is a key determinant of Ah receptor-mediated transcriptional activity. Murray IA, Reen RK, Leathery N, Ramadoss P, Bonati L, Gonzalez FJ, Peters JM, Perdew GH. Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary Science, The Pennsylvania State University, University Park, PA 16802, USA. The aryl hydrocarbon receptor (AhR) mediates the biological activity of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Whether the AhR can mediate enhanced transcriptional activity in the absence of ligand binding has not been established. Hepatocytes from AhR-null (AhR-KO) and wild-type (AhR-WT) neonatal mice were immortalized with Simian virus 40. Two point mutants of the AhR, A375I and A375F, were generated to test the hypothesis that the AhR requires ligand binding to exhibit significant transcriptional activity, both mutants fail to bind ligand or exhibit enhanced activity in cells exposed to AhR ligands. Upon transient, co-expression of ARNT with AhR-A375I or AhR-A375F in AhR-KO cells, these mutants exhibited significant ligand-independent transcriptional activity. However, in CV-1 cells, which others have previously shown to contain relatively high levels of AhR ligand(s), these AhR mutants exhibit essentially no constitutive activity. These results indicate that while the AhR can potentially exhibit activity in the absence of ligand binding, the high constitutive receptor activity observed in many cell lines appears to be due to the presence of endogenous AhR ligands. PMID: 16137638 [PubMed - in process] --------------------------------------------------------------- 9: Drug Metab Dispos. 2005 Aug 24; [Epub ahead of print] Modulation of human cytochrome P450 1B1 expression by 2,4,3',5'- tetramethoxystilbene. Chun YJ, Lee SK, Kim MY. College of Pharmacy, Chung-Ang University. We have previously shown that 2,4,3(' ),5(')-tetramethoxystilbene (TMS), a synthetic trans-stilbene analogue, is one of the most potently selective inhibitors of recombinant human cytochrome P450 (CYP) 1B1 in vitro. In the present studies, the effects of TMS on CYP1B1 expression were investigated in human cancer cells. TMS significantly inhibited CYP1-mediated 7-ethoxyresorufin O-deethylation (EROD) activity in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced MCF-7 cells or lung microsomes of Sprague Dawley rats treated with 7,12-dimethylbenz[a]anthracene (DMBA). TCDD-stimulated CYP1B1 protein and mRNA expression were significantly suppressed by TMS in a concentration-dependent manner in MCF-7, MCF-10A, and HL- 60 cells. While TMS down-regulated TCDD-induced CYP1B1 gene expression, the levels of AhR and ARNT mRNA expression were not changed by TMS treatment. In human cancer cells, TMS induced apoptotic cell death and the cytotoxic effects of TMS were significant when the cells were incubated with TCDD. CYP1B1 was able to convert TMS to a metabolite(s) when incubated with NADPH. Metabolic activation of TMS by CYP1B1 induced by TCDD may mediate cellular toxicity of TMS in human cancer cells because the sensitivity to TMS in MCF-7 cells treated with TCDD were more significant than in HL-60 cells treated with TCDD. Taken together, our results indicate that TMS acts as a strong modulator of CYP1B1 gene expression as well as a potent selective inhibitor in vitro. The ability of TMS to induce apoptotic cell death in tumor cells, as well as CYP1B1 inhibition, may contribute to its usefulness for cancer chemoprevention. PMID: 16120791 [PubMed - as supplied by publisher] --------------------------------------------------------------- 10: Clin Cancer Res. 2005 Aug 15;11(16):5793-801. Cytochrome P450 1B1 is overexpressed and regulated by hypomethylation in prostate cancer. Tokizane T, Shiina H, Igawa M, Enokida H, Urakami S, Kawakami T, Ogishima T, Okino ST, Li LC, Tanaka Y, Nonomura N, Okuyama A, Dahiya R. Department of Urology, Veterans Affairs Medical Center and University of California, San Francisco, California 94121, USA. PURPOSE: Cytochrome P450 1B1 (CYP1B1), a dioxin inducible member of the CYP supergene family, is overexpressed in various human malignancies including prostate cancer. We hypothesized that promoter/enhancer CpG methylation contributes to the regulation of CYP1B1 expression in human prostate tissue. EXPERIMENTAL DESIGN: Expression and induction of the CYP1B1 gene in clinical prostate tissues and prostate cancer cell lines were investigated. The methylation status of the CYP1B1 gene was analyzed in 175 prostate cancer and 96 benign prostatic hyperplasia samples using methylation-specific PCR (MSP) and bisulfite-modified DNA sequencing. MSP primers covered dioxin response elements (DRE) and Sp1 sites that are important for the expression of CYP1B1. RESULTS: Expressions of CYP1B1 mRNA and protein were increased in prostate cancer. The aryl hydrocarbon receptor (AhR)/AhR nuclear translocator (ARNT) heterodimer complex activates gene transcription by binding to the DREs of CYP1B1. In prostate cancer cells, CYP1B1 mRNA was induced by 2,3,7,8-tetrachlorodigenzo-p-dioxin (TCDD) and/or demethylation agent (5-aza-2-deoxycytidine). There was no change in the expressions of AhR and ARNT. Methylation of promoter/enhancer regions was significantly higher in benign prostatic hyperplasia compared with prostate cancer. MSP-positive patients had significantly lower risk for prostate cancer as compared with MSP-negative patients. There was no correlation between CYP1B1 methylation status and clinicopathologic features.CONCLUSIONS: CYP1B1 is overexpressed in prostate cancer and regulated by hypomethylation of its promoter/enhancer region. This is the first report about CYP1B1 regulation in human clinical prostate samples showing that hypomethylation of the CYP1B1 gene may play an important role in prostate cancer. PMID: 16115918 [PubMed - in process] --------------------------------------------------------------- 11: Comp Biochem Physiol C Toxicol Pharmacol. 2005 Jul;141(3):281-91. Aryl hydrocarbon receptor (AHR) and AHR nuclear translocator (ARNT) expression in Baikal seal (Pusa sibirica) and association with 2,3,7,8-TCDD toxic equivalents and CYP1 expression levels. Kim EY, Iwata H, Suda T, Tanabe S, Amano M, Miyazaki N, Petrov EA. Ehime Prefectural Institute of Public Health and Environmental Science, 8-234 Sanban-cho, Matsuyama 790-0003, Japan. Most toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related planar halogenated aromatic hydrocarbons (PHAHs) are mediated by ligand-activated aryl hydrocarbon receptor (AHR) signaling pathway. To understand the regulation mechanism of AHR and AHR nuclear translocator (ARNT) expression in wild Baikal seal (Pusa sibirica) population contaminated by PHAHs, the present study investigated hepatic mRNA expression levels of AHR and its heterodimer, ARNT genes, in association with biological index (age, gender and body weight), PHAH accumulation and expression levels of cytochrome P450 (CYP) 1A and 1B. While there was no gender difference, the AHR mRNA expression levels were increased with ages (p = 0.014) and body weights (p = 0.015), indicating that AHR expression might be affected by these biological factors. The AHR mRNA expression levels exhibited significant positive correlations with total TEQs and most of individual congener TEQs derived from polychorinated dibenzo-p-dioxins, dibenzofurans and non-ortho coplanar polychorinated biphenyls (PCBs), indicating the transcriptional up-regulation of AHR expression by these congeners. On the other hand, there was no significant correlation between individual TEQs from mono-ortho coplanar PCBs and AHR expression. These results imply the structure-related transcriptional activity of AHR among PHAHs congeners. AHR mRNA levels showed positive correlations with both CYP1A protein (p = 0.039) and CYP1A1 mRNA expression levels (p = 0.046). In contrast to AHR expression, neither the total nor individual congener TEQs influenced ARNT at the transcriptional level. ARNT mRNA showed significant negative correlations with CYP1A/1B protein (p = 0.027 and p = 0.006) and CYP1A1 mRNA expression levels (p = 0.039), implying the existence of different transcriptional regulation between AHR and ARNT genes and negative regulation by CYP1A/1B-mediated signaling pathways. The present findings may render significant insight on the basic mechanisms underlying regulation of AHR and ARNT expressions associated with biological factors and PHAH exposure in wild mammalian populations. PMID: 16111922 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 12: Biochemistry. 2005 Aug 23;44(33):11148-59. The transactivation domain of the Ah receptor is a key determinant of cellular localization and ligand-independent nucleocytoplasmic shuttling properties. Ramadoss P, Perdew GH. Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary Science, The Pennsylvania State University, University Park, Pennsylvania 16802, USA. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates transcription of a number of target genes upon binding ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Large intra- and interspecies variations exist with respect to sensitivity to TCDD, and this could, at least in part, be due to a considerable variation in the AhR amino acid sequence between species. The N-terminal half of the AhR is well-conserved across species, whereas the C-terminal half exhibits a considerable degree of degeneracy. It has previously been shown that there are differences between the mouse (mAhR) and human AhR (hAhR) in terms of cellular localization, nucleocytoplasmic shuttling, the effect of chaperone proteins on these properties, and differences in relative ligand affinity. In this study, two chimeras were generated such that each had the N-terminal half of one receptor and the C-terminal half of the other receptor. The C-terminal half of the receptor, containing the transactivation domain, determines the cellular localization of the transiently transfected receptor and regulates the ability of hepatitis B virus X-associated protein 2 (XAP2) to inhibit ligand-independent nuclear import of AhR. In addition, the transactivation domain (TAD) appears to determine the presence of XAP2 in the nuclear ligand-bound AhR/hsp90 complex prior to association with the AhR nuclear translocator protein (ARNT). However, the transactivation domain does not appear to play a role in determining relative ligand affinity of the receptor, and mAhR and hAhR have similar overall transactivation potential in a cell-based reporter system at a saturating dose of ligand. This study demonstrates for the first time that the transactivation domain of the AhR influences important biochemical properties of the N-terminal half of the AhR, and the degeneracy in the transactivation domain between the mAhR and the hAhR results in species-specific differences in receptor properties. PMID: 16101299 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 13: Gene. 2005 Sep 26;358:39-52. Regulation of an insect cytochrome P450 monooxygenase gene (CYP6B1) by aryl hydrocarbon and xanthotoxin response cascades. Brown RP, McDonnell CM, Berenbaum MR, Schuler MA. Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA. Many organisms respond to toxic compounds in their environment by inducing regulatory networks controlling the expression and activity of cytochrome P450 monooxygenase (P450s) detoxificative enzymes. In particular, black swallowtail (Papilio polyxenes) caterpillars respond to xanthotoxin, a toxic phytochemical in their hostplants, by activating transcription of the CYP6B1 promoter via several regions located within 150 nt of the transcription initiation site. One such element is the xenobiotic response element to xanthotoxin (XRE-Xan) that lies upstream of consensus XRE-AhR (xenobiotic response element to the aryl hydrocarbon receptor) and OCT-1 (octamer-1 binding site) element known to be utilized in mammalian aryl hydrocarbon response cascades. Two-plasmid transfections conducted in Sf9 cells have indicated that XRE-Xan, XRE-AhR and a number of other proximal elements, but not OCT-1, are critical for basal as well as xanthotoxin- and benzo[alpha]pyrene-induced transcription of the CYP6B1 promoter. Four-plasmid transfections with vectors co-expressing the spineless (Ss) and tango (Tgo) proteins, the Drosophila melanogaster homologues of mammalian AhR and ARNT, have indicated that these proteins enhance basal expression of the CYP6B1 promoter but not the magnitude of its xanthotoxin and benzo[alpha]pyrene induction. Based on these results, we propose that these Drosophila transcription factors modulate basal expression of this promoter in a ligand-independent manner and attenuate its subsequent responses to planar aryl hydrocarbons (benzo[alpha]pyrene) and allelochemicals (xanthotoxin). PMID: 16099607 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 14: Fertil Steril. 2005 Aug;84(2):454-8. Analysis of the AhR, ARNT, and AhRR gene polymorphisms: genetic contribution to endometriosis susceptibility and severity. Tsuchiya M, Katoh T, Motoyama H, Sasaki H, Tsugane S, Ikenoue T. Department of Public Health, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan. OBJECTIVE: To explore whether polymorphisms in AhR, ARNT, and AhRR contribute to endometriosis susceptibility and severity. DESIGN: Case control study. SETTING: Hospital. PATIENT(S): One hundred thirty-eight Japanese women with or without endometriosis, diagnosed endoscopically. INTERVENTION(S): Endoscopic laparoscopy, with blood samples for genotyping obtained before the laparoscopic examination for genomic DNA extraction from peripheral leukocytes. MAIN OUTCOME MEASURE(S): AhR, ARNT, and AhRR polymorphisms were genotyped using real-time polymerase chain reaction (PCR) analysis. Odds ratios and 95% confidence intervals were calculated for AhR, ARNT, and AhRR genotypes to evaluate the risk of endometriosis. Associations between these polymorphisms and stage of endometriosis were also examined. RESULT(S): The C/G + G/G genotypes at codon 185 of AhRR showed a statistically significant association with risk of endometriosis (adjusted odds ratio, 2.53; 95% confidence interval, 1.16-5.55). Furthermore, a statistically significant trend associated the C/G + G/G genotypes with the clinical stage of endometriosis. No statistically significant association was observed between AhR codon 554 or ARNT codon 189 polymorphisms and endometriosis. CONCLUSION(S): AhRR codon 185 polymorphism was associated with susceptibility to and severity of endometriosis in Japanese women. PMID: 16084889 [PubMed - in process] --------------------------------------------------------------- 15: Toxicol Lett. 2005 Dec 30;160(1):22-33. Epub 2005 Jul 28. Lack of CYP1A1 expression is involved in unresponsiveness of the human hepatoma cell line SK-HEP-1 to dioxin. Shiizaki K, Ohsako S, Koyama T, Nagata R, Yonemoto J, Tohyama C. Environmental Health Sciences Division, National Institute for Environmental Studies, Tsukuba 305-8506, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba 305-8577, Japan. The aryl hydrocarbon receptor (AhR) mediates a wide variety of toxic effects due to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The human hepatoma cell line SK-HEP-1 expresses AhR and ARNT. However, TCDD failed to induce CYP1A1 and XRE-dependent reporter genes in these cells. Although CYP1A1 was not induced by TCDD exposure, both CYP1B1 and AhR repressor (AhRR) were constitutively expressed. The AhR antagonist alpha-naphthoflavone altered the basal level of XRE-dependent reporter gene expression dose-dependently. As our results suggested the activation of AhR signals by putative endogenous ligands, we established SK-HEP-1-derived cell lines that stably expressed CYP1A1. The inducibility of XRE-dependent reporter genes and CYP1B1 by TCDD was restored in these cells. Our findings demonstrated the presence of endogenous ligands in SK-HEP-1 cells due to the absence of the metabolizing enzyme CYP1A1, but not CYP1B1, which allowed the constitutive expression of AhR target genes. PMID: 16054781 [PubMed - in process] --------------------------------------------------------------- 16: Toxicol Appl Pharmacol. 2005 Sep 1;207(2 Suppl):62-9. CYP2S1: A short review. Saarikoski ST, Rivera SP, Hankinson O, Husgafvel-Pursiainen K. Department of Industrial Hygiene and Toxicology, Finnish Institute of Occupational Health, FI-00250, Finland; Department of Mental Health and Alcohol Research, National Public Health Institute, FI-00250, Finland. A new member of the cytochrome P450 superfamily, CYP2S1, has recently been identified in human and mouse. In this paper, we review the data currently available for CYP2S1. The human CYP2S1 gene is located in chromosome 19q13.2 within a cluster including CYP2 family members CYP2A6, CYP2A13, CYP2B6, and CYP2F1. These genes also show the highest homology to the human CYP2S1. The gene has recently been found to harbor genetic polymorphism. CYP2S1 is inducible by dioxin, the induction being mediated by the Aryl Hydrocarbon Receptor (AHR) and Aryl Hydrocarbon Nuclear Translocator (ARNT) in a manner typical for CYP1 family members. In line with this, CYP2S1 has been shown to be inducible by coal tar, an abundant source of PAHs, and it was recently reported to metabolize naphthalene. This points to the involvement of CYP2S1 in the metabolism of toxic and carcinogenic compounds, similar to other dioxin-inducible CYPs. CYP2S1 is expressed in epithelial cells of a wide variety of extrahepatic tissues. The highest expression levels have been observed in the epithelial tissues frequently exposed to xenobiotics, e.g., the respiratory, gastrointestinal, and urinary tracts, and in the skin. The observed ubiquitous tissue distribution, as well as the expression of CYP2S1 throughout embryogenesis suggest that CYP2S1 is likely to metabolize important endogenous substrates; thus far, retinoic acid has been identified. In conclusion, CYP2S1 exhibits many features of interest for human health and thus warrants further investigation. PMID: 16054184 [PubMed - in process] --------------------------------------------------------------- 17: Comp Biochem Physiol C Toxicol Pharmacol. 2005 Jun;141(2):177-87. Molecular characterization of the aryl hydrocarbon receptors (AHR1 and AHR2) from red seabream (Pagrus major). Yamauchi M, Kim EY, Iwata H, Tanabe S. Ehime Prefectural Institute of Public Health and Environmental Science, 8-234 Sanban-cho, Matsuyama 790-0003, Japan. The aryl hydrocarbon receptor (AHR) mediates the toxic effects of planar halogenated aromatic hydrocarbons (PHAHs). Bony fishes exposed to PHAHs exhibit a wide range of developmental defects. However, functional roles of fish AHR are not yet fully understood, compared with those of mammalian AHRs. To investigate the potential sensitivity to PHAHs toxic effects, an AHR cDNA was initially cloned and sequenced from red seabream (Pagrus major), an important fishery resource in Japan. The present study succeeded in identifying two highly divergent red seabream AHR cDNA clones, which shared only 32% identity in full-length amino acid sequence. The phylogenetic analysis revealed that one belonged to AHR1 clade (rsAHR1) and another to AHR2 clade (rsAHR2). The rsAHR1 encoded a 846-residue protein with a predicted molecular mass of 93.2 kDa, and 990 amino acids and 108.9 kDa encoded rsAHR2. In the N-terminal half, both rsAHR genes included bHLH and PAS domains, which participate in ligand binding, AHR/ARNT dimerization and DNA binding. The C-terminal half, which is responsible for transactivation, was poorly conserved between rsAHRs. Quantitative analyses of both rsAHRs mRNAs revealed that their tissue expression profiles were isoform-specific; rsAHR1 mRNA expressed primarily in brain, heart, ovary and spleen, while rsAHR2 mRNA was observed in all tissues examined, indicating distinct roles of each rsAHR. Furthermore, there appeared to be species-differences in the tissue expression profiles of AHR isoforms between red seabream and other fish. These results suggest that there are isoform- and species-specific functions in piscine AHRs. PMID: 16006195 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 18: Toxicol Lett. 2005 Jul 28;158(1):50-60. Epub 2005 Apr 1. Comment in: Toxicol Lett. 2005 Aug 14;158(2):164-5. Sanguinarine activates polycyclic aromatic hydrocarbon associated metabolic pathways in human oral keratinocytes and tissues. Karp JM, Rodrigo KA, Pei P, Pavlick MD, Andersen JD, McTigue DJ, Fields HW, Mallery SR. The Ohio State University College of Dentistry, Department of Pediatric Dentistry, Columbus, OH 43210, USA. Sanguinarine's use in human clinical applications is currently controversial. While some studies have demonstrated sanguinarine's anti-inflammatory and anti-oxidant properties, other investigations reported sanguinarine's procarcinogenic effects. Like the tobacco-associated carcinogen, benzo(a)pyrene (B(a)P), sanguinarine is a polycyclic aromatic hydrocarbon (PAH). PAH exposure activates the aryl hydrocarbon transcription activating factor (AhR), resulting in nuclear translocation, binding to the aryl hydrocarbon nuclear translocator (ARNT), which thereby increases expression of a pool of carcinogen metabolizing enzymes. The goal of this study was to investigate whether sanguinarine activates this PAH-associated signaling cascade in human oral cells and tissues. Our results demonstrate that sanguinarine: (i) results in formation of the AhR-ARNT complex, (ii) induces AhR-associated gene expression, (iii) inhibits cytochrome P450 1A1 (CYP 1A1) microsomal oxidative activity and (iv) pretreatment upregulates CYP 1A1 function. Collectively, these data provide evidence that sanguinarine activates PAH-associated signaling and metabolic pathways. Notably, previous studies have demonstrated that mammalian hepatic microsomes metabolize sanguinarine to a mutagenic epoxide. Persons who respond to sanguinarine exposure with induction of primarily Phase I relative to Phase II enzymes are, therefore, at risk for sanguinarine bioactivation and its potential mutagenic effects. PMID: 15993743 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 19: Expert Opin Investig Drugs. 1999 Sep;8(9):1385-96. Development of selective aryl hydrocarbon receptor modulators for treatment of breast cancer. Safe S, Qin C, McDougal A. Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466 USA. ssafe@cvm.tamu.edu The aryl hydrocarbon receptor (AhR) is a basic helix-loop-helix DNA-binding protein that forms a transcriptionally-active heterodimer with the AhR nuclear translocator (Arnt) protein. The nuclear AhR complex is a ligand-induced transcription factor and the environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a high affinity ligand for the AhR. TCDD induces a diverse spectrum of tissue-, sex- and species-specific biochemical and toxic responses in Ah-responsive cells/tissues including the inhibition of 17beta-oestradiol (E2)-induced gene expression in the rodent uterus and mammary and in human breast cancer cell lines. TCDD also inhibits spontaneous and carcinogen-induced mammary tumour formation and growth in rodent models. Research in this laboratory has utilised the AhR as a target for developing anticancer drugs for treatment of breast cancer and two different structural classes of selective AhR modulators (SAhRMs) have been developed. Alternate-substituted (1,3,6,8- and 2,4,6,8-) alkyl polychlorinated dibenzofurans (PCDFs) and substituted diindolylmethanes (DIMs) bind the AhR and induce a pattern of AhR-oestrogen receptor (ER) inhibitory cross-talk similar to that observed for TCDD including inhibition of mammary tumour growth at doses < 1.0 mg/kg/day. In contrast, effective doses of these compounds do not induce hepatic CYP1A1-dependent activity or other AhR-mediated toxic responses induced by TCDD. These results indicate that SAhRMs may be an important new class of drugs for clinical treatment of breast cancer via AhR-ER inhibitory cross-talk. PMID: 15992156 [PubMed] --------------------------------------------------------------- 20: Toxicol Appl Pharmacol. 2005 Jun 24; [Epub ahead of print] 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces matrix metalloproteinase (MMP) expression and invasion in A2058 melanoma cells. Villano CM, Murphy KA, Akintobi A, White LA. Department of Biochemistry and Microbiology, 76 Lipman Dr., Rutgers, The State University of NJ, New Brunswick, NJ 08901, USA. There has been a 34% increase in melanoma related mortality in the United States from 1973 to 1992. Although few successful treatments for malignant melanoma exist, it is known that genetic susceptibility and environmental factors contribute to the initiation and progression of melanoma. Excessive UV exposure is considered the main etiological factor in melanoma initiation, however, epidemiological and experimental evidence suggests that exposure to environmental carcinogens contribute to melanoma. We propose that exposure to environmental chemicals that activate the aryl hydrocarbon receptor pathway contribute to melanoma progression, specifically through stimulation of the expression and activity of the matrix metalloproteinases (MMPs). Therefore, we investigated the effect of AhR activation on normal human melanocytes and several melanoma cell lines. The data presented here demonstrate that normal melanocytes and melanoma cells express the AhR and Arnt and are responsive to activation by TCDD. Furthermore, activation of this pathway in transformed melanoma cells (A2058) results in increased expression and activity of MMP-1, MMP-2 and MMP-9, as well as increased invasion using in vitro invasion assays. Furthermore, TCDD-induced expression of the MMP-1 promoter in melanoma cells appears to require different elements than those required in untransformed cells, indicating that this pathway may have multiple mechanisms for activation of MMP expression. PMID: 15982688 [PubMed - as supplied by publisher] --------------------------------------------------------------- 21: Med Hypotheses. 2005;65(4):760-5. Oestrogen-receptors (ER) are likely to be promiscuous: wider role for oestrogens and mimics. Wiseman A. School of Biomedical and Molecular Sciences, University of Surrey, Guildford GU2 7XH, UK. alan@tridgway.wanadoo.co.uk The anti-breast cancer drug tamoxifen that binds to ER is metabolised in human liver by CYP2D6 isoenzyme, whilst the metabolism of 17beta-oestradiol (by hydroxylation) is by phase I biotransformation in the liver to 2-hydroxyoestradiol and to 4-hydroxyoestradiol respectively by two isoenzymes of this mixed function oxidase CYP cytochromes P450 (EC 1.14.14.1); CYP1A2 and by CYP1B1. Nevertheless, it appears that the receptor (AhR) itself causes the expression of oestrogen-regulated target genes (studied by binding of dioxin). This is the result of an unknown signalling mechanism at the genome that is triggered directly by this receptor by binding promiscuously to ER (alpha or beta) sites. This has been observed even in the absence of oestrogens or mimics therefore in genome-binding investigations of target tissues such as uterus: oestrogen-receptor (ER) is likely to be promiscuous therefore. Furthermore, AhR (polycyclic aromatic hydrocarbon receptor), when activated by the binding of aromatic hydrocarbons (Ah) forms a complex with the aryl hydrocarbon nuclear-translocator chaperone protein (Arnt). It is this binding to xenobiotic response elements in DNA that initiates expression of the appropriate oestrogen-regulated target-genes in the uterus and other target tissues (including mammary, ovaries, and brain). The likely promiscuity of oestrogen receptors is proposed to be the cause of numerous side effects when oestrogen is involved in therapy, these can be manifest in hormone replacement therapy (HRT) and in the incorporation of synthetic oestrogens in the wide varieties of oral contraceptives now available. PMID: 15961252 [PubMed - in process] --------------------------------------------------------------- 22: Toxicol Sci. 2005 Aug;86(2):342-53. Epub 2005 May 11. Comparative microarray analysis of basal gene expression in mouse Hepa-1c1c7 wild-type and mutant cell lines. Fong CJ, Burgoon LD, Zacharewski TR. Department of Biochemistry and Molecular Biology, National Food Safety and Toxicology Center, Michigan State University, East Lansing, Michigan 48824, USA. Hepa-1c1c7 wild-type and benzo[a]pyrene-resistant derived mutant cell lines have been used to elucidate pathways and mechanisms involving the aryl hydrocarbon receptor (AhR). However, there has been little focus on other biological processes which may differ between the isolated lines. In this study, mouse cDNA microarrays representing 4858 genes were used to examine differences in basal gene expression between mouse Hepa-1c1c7 wild-type and c1 (truncated Cyp1a1 protein), c4 (AhR nuclear translocator, ARNT, deficient), and c12 (low AhR levels) mutant cell lines. Surprisingly, c1 mutants exhibited the greatest number of gene expression changes compared to wild-type cells, followed by c4 and c12 lines, respectively. Differences in basal gene expression were consistent with cell line specific variations in morphology, mitochondrial activity, and proliferation rate. MTT and direct cell count assays indicate both c4 and c12 mutants exhibit increased proliferative activity when compared to wild-type cells, while the c1 mutants exhibited decreased activity. This study further characterizes Hepa-1c1c7 wild-type and mutant cells and identifies significant differences in biological processes that should be considered when conducting comparative mechanistic studies with these lines. PMID: 15888666 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 23: Reprod Toxicol. 2005 Nov-Dec;20(4):521-30. Ontogenetic development, sexual differentiation, and effects of Aroclor 1254 exposure on expression of the arylhydrocarbon receptor and of the arylhydrocarbon receptor nuclear translocator in the rat hypothalamus. Pravettoni A, Colciago A, Negri-Cesi P, Villa S, Celotti F. Department of Endocrinology, University of Milano, Via Balzaretti, 9, 20133 Milano, Italy. Interaction of polychlorinated biphenyls (PCBs) with the aryl hydrocarbon receptor (AhR)/nuclear translocator (ARNT) system might interfere with the mechanisms controlling the sexual differentiation of the developing hypothalamus. The aim of this study was to evaluate the presence of AhR/ARNT in brain cells and the developmental profile of their expression in the hypothalamus of male and female rats during the perinatal period. Brain accumulation of the main PCB congeners after prenatal exposure to Aroclor 1254 and its influence on hypothalamic expression of AhR/ARNT was also assessed. The results show that: (a) AhR and ARNT are expressed both in neurons and in glia; (b) AhR expression progressively increases in the developing hypothalamus particularly in males, while ARNT is relatively constant in both sexes; (c) the prenatal administration of Aroclor to dams produces a differential accumulation of PCBs, depending on the chlorine atom number, and stimulates AhR expression only in the male hypothalamus. In conclusion, the developing male hypothalamus might be more sensitive to disrupting potential of PCBs. PMID: 15869859 [PubMed - in process] --------------------------------------------------------------- 24: Biochem Pharmacol. 2005 May 15;69(10):1403-8. Ah receptor- and TCDD-mediated liver tumor promotion: clonal selection and expansion of cells evading growth arrest and apoptosis. Bock KW, Kohle C. Department of Toxicology, Institute of Pharmacology and Toxicology, University of Tubingen, Tubingen, Germany. bock@uni-tuebingen.de The Ah receptor (AhR) has been characterized as a ligand-activated transcription factor which belongs to the bHLH/PAS (basic helix-loop-helix/Per-Arnt-Sim) family of chemosensors. Transgenic mouse models revealed adaptive and developmental functions of the AhR in the absence of exogenous ligands. Use of persistent agonists such as dioxins including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds demonstrated that the AhR mediates a plethora of species- and tissue-dependent toxicities, including chloracne, wasting, teratogenicity, immunotoxicity, liver tumor promotion and carcinogenicity. However, molecular mechanisms underlying most aspects of these toxic responses as well as biological functions of the AhR are currently unknown. Previous studies of liver tumor promotion in the two-stage hepatocarcinogenesis model indicated that TCDD mediates clonal expansion of 'initiated' preneoplastic hepatocytes, identified as enzyme-altered foci (EAF) by inhibiting apoptosis and bypassing AhR-mediated growth arrest. In contrast, the Ah receptor has been shown in cell models to stimulate growth arrest and apoptosis. Possible underlying mechanisms of these AhR responses are discussed, including enhanced metabolism of retinoic acid which attenuates TGFbeta-mediated apoptosis and interaction of the Ah receptor with the hypophosphorylated retinoblastoma tumor suppressor protein. The discrepancy between in vivo findings in EAF and AhR functions may be solved by hypothesizing that sustained activation of the Ah receptor generates a strong selective pressure in liver treated with genotoxic carcinogens leading to selection and expansion of clones evading growth arrest and apoptosis. Models are discussed which may facilitate verification of this hypothesis. Publication Types: Review Review, Tutorial PMID: 15857604 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 25: Eur J Gynaecol Oncol. 2005;26(2):175-80. Effects of dioxin and nutrition on cellular proliferation and dioxin- and estrogen-linked genes in ovarian cancer cell lines. Takahashi S, Hirano S, Ito N, Tamaya T. Department of Obstetrics and Gynaecology, Gifu University School of Medicine, Gifu, Japan. This study was undertaken to examine the effects of dioxin (TCDD) and nutrition on cellular proliferation and dioxin- and estrogen-linked gene expression in ovarian cancer cell lines. Caov-3 and SK-OV-3 cells were incubated in a medium supplemented with 0.5-10% fetal bovine serum (FBS). Cell proliferation was assayed with an MTT assay. Dioxin- and estrogen-linked genes (AhR, ERalpha, ERbeta, CYP1A1 and ARNT) expressed were determined with the RT-PCR method. Caov-3 cells, but not SK-OV-3 cells, were proliferated with TCDD alone with increased AhR and ERa mRNA expressions when incubated in the low FBS concentration. CYP1A1 and ARNT mRNA expressions of SK-OV-3, but not that of Caov-3, were suppressed in the low FBS (under 1.0%) concentration. In the low FBS concentration medium with dioxin, AhR and ERa expression were increased with the proliferation of Caov-3 cells; CYP1A1 and ARNT were stable. Each ovarian cancer cell line may have its own distinct responsiveness to dioxin depending on the nutritional state. PMID: 15857025 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 26: Toxicology. 2005 Jun 1;210(2-3):189-96. Gene expression of drug metabolizing enzymes in adult and aged mouse liver: a modulation by immobilization stress. Mikhailova ON, Gulyaeva LF, Filipenko ML. Laboratory of Molecular Mechanisms of Carcinogenesis, Institute of Molecular Biology and Biophysics, Timakov Str. 2, Novosibirsk 630117, Russia. pharmacogenomics@ngs.ru The role of stress in the regulation of enzymatic systems involved in the biotransformation of xenobiotics, as well as endogenous substrates in the liver was investigated using single immobilization stress as a model. Adult (3 months of age) and aged (26 months) C3H/a male mice were used. Cytochrome P450 1A1 and 1A2 (CYP1A1 and CYP1A2), glutathione S-transferase M1 (GSTM1), aryl hydrocarbon receptor (AHR), aryl hydrocarbon receptor nuclear translocator (ARNT) and catechol-O-methyltransferase (COMT) mRNA levels in the mouse liver were measured by a semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) method. Excluding CYP1A1, experiments revealed significant differences in the expression of these genes between adult- and aged-control animals. The influence of stress on the expression of genes studied was shown to be higher in adult mice than in aged ones. Our results clearly demonstrate the lack of response or even the attenuation of gene expression in aged animals that may play an important role in age-related pathologies and diseases. PMID: 15840432 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 27: J Biol Chem. 2005 Jun 3;280(22):21607-11. Epub 2005 Apr 18. ER alpha-AHR-ARNT protein-protein interactions mediate estradiol-dependent transrepression of dioxin-inducible gene transcription. Beischlag TV, Perdew GH. Center for Molecular Toxicology and Carcinogenesis and Department of Veterinary Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA. The aryl hydrocarbon receptor (AHR) and the aryl hydrocarbon receptor nuclear translocator (ARNT) form a heterodimeric transcription factor upon binding a wide variety of environmental pollutants, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). AHR target gene activation can be repressed by estrogen and estrogen-like compounds. In this study, we demonstrate that a significant component of TCDD-inducible Cyp1a1 transcription is the result of recruitment of estrogen receptor (ER)-alpha by AHR/ARNT as a transcriptional co-repressor. Both AHR and ARNT were capable of interacting directly with ER alpha, as ascertained by glutathione S-transferase pull-down. 17Beta-estradiol repressed TCDD-activated Cyp1a1 and Cyp1b1 gene transcription in MCF-7 cells in the presence of cycloheximide, as determined by reverse transcription/real-time PCR. Furthermore, chromatin immunoprecipitation (ChIP) assays have shown that ER alpha is present at the Cyp1a1 enhancer only after co-treatment with E2 and TCDD, in MCF-7 cells. Sequential two-step ChIP assays were performed which demonstrate that AHR and ER alpha are present together at the same time on the Cyp1a1 enhancer during transrepression. Taken together these data support a role for ER-mediated transrepression of AHR-dependent gene regulation. PMID: 15837795 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 28: Arch Pharm Res. 2005 Mar;28(3):249-68. Induction of phase I, II and III drug metabolism/transport by xenobiotics. Xu C, Li CY, Kong AN. Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA. Drug metabolizing enzymes (DMEs) play central roles in the metabolism, elimination and detoxification of xenobiotics and drugs introduced into the human body. Most of the tissues and organs in our body are well equipped with diverse and various DMEs including phase I, phase II metabolizing enzymes and phase III transporters, which are present in abundance either at the basal unstimulated level, and/or are inducible at elevated level after exposure to xenobiotics. Recently, many important advances have been made in the mechanisms that regulate the expression of these drug metabolism genes. Various nuclear receptors including the aryl hydrocarbon receptor (AhR), orphan nuclear receptors, and nuclear factor-erythoroid 2 p45-related factor 2 (Nrf2) have been shown to be the key mediators of drug-induced changes in phase I, phase II metabolizing enzymes as well as phase III transporters involved in efflux mechanisms. For instance, the expression of CYP1 genes can be induced by AhR, which dimerizes with the AhR nuclear translocator (Arnt), in response to many polycyclic aromatic hydrocarbon (PAHs). Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). The peroxisome proliferator activated receptor (PPAR), which is one of the first characterized members of the nuclear hormone receptor, also dimerizes with RXR and has been shown to be activated by lipid lowering agent fibrate-type of compounds leading to transcriptional activation of the promoters on CYP4A gene. CYP7A was recognized as the first target gene of the liver X receptor (LXR), in which the elimination of cholesterol depends on CYP7A. Farnesoid X receptor (FXR) was identified as a bile acid receptor, and its activation results in the inhibition of hepatic acid biosynthesis and increased transport of bile acids from intestinal lumen to the liver, and CYP7A is one of its target genes. The transcriptional activation by these receptors upon binding to the promoters located at the 5-flanking region of these CYP genes generally leads to the induction of their mRNA gene expression. The physiological and the pharmacological implications of common partner of RXR for CAR, PXR, PPAR, LXR and FXR receptors largely remain unknown and are under intense investigations. For the phase II DMEs, phase II gene inducers such as the phenolic compounds butylated hydroxyanisol (BHA), tert-butylhydroquinone (tBHQ), green tea polyphenol (GTP), (-)-epigallocatechin-3-gallate (EGCG) and the isothiocyanates (PEITC, sulforaphane) generally appear to be electrophiles. They generally possess electrophilic-mediated stress response, resulting in the activation of bZIP transcription factors Nrf2 which dimerizes with Mafs and binds to the antioxidant/electrophile response element (ARE/EpRE) promoter, which is located in many phase II DMEs as well as many cellular defensive enzymes such as heme oxygenase-1 (HO-1), with the subsequent induction of the expression of these genes. Phase III transporters, for example, P-glycoprotein (P-gp), multidrug resistance-associated proteins (MRPs), and organic anion transporting polypeptide 2 (OATP2) are expressed in many tissues such as the liver, intestine, kidney, and brain, and play crucial roles in drug absorption, distribution, and excretion. The orphan nuclear receptors PXR and CAR have been shown to be involved in the regulation of these transporters. Along with phase I and phase II enzyme induction, pretreatment with several kinds of inducers has been shown to alter the expression of phase III transporters, and alter the excretion of xenobiotics, which implies that phase III transporters may also be similarly regulated in a coordinated fashion, and provides an important mean to protect the body from xenobiotics insults. It appears that in general, exposure to phase I, phase II and phase III gene inducers may trigger cellular "stress" response leading to the increase in their gene expression, which ultimately enhance the elimination and clearance of these xenobiotics and/or other "cellular stresses" including harmful reactive intermediates such as reactive oxygen species (ROS), so that the body will remove the "stress" expeditiously. Consequently, this homeostatic response of the body plays a central role in the protection of the body against "environmental" insults such as those elicited by exposure to xenobiotics. Publication Types: Review PMID: 15832810 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 29: Reproduction. 2005 Apr;129(4):379-89. Molecular interactions of the aryl hydrocarbon receptor and its biological and toxicological relevance for reproduction. Pocar P, Fischer B, Klonisch T, Hombach-Klonisch S. Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, D-06097, Halle (Saale), Germany. paola.pocar@medizin.uni-halle.de The dioxin/aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor responsive to both natural and man-made environmental compounds. AhR and its nuclear partner ARNT are expressed in the female reproductive tract in a variety of species and several indications suggest that the AhR might play a pivotal role in the physiology of reproduction. Furthermore, it appears to be the mediator of most, if not all, the adverse effects on reproduction of a group of highly potent environmental pollutants collectively called aryl hydrocarbons (AHs), including the highly toxic compound 2,3,7,8-tetrachlor-odibenzo-p-dioxin (TCDD). Although a large body of recent literature has implicated AhR in multiple signal transduction pathways, the mechanisms of action resulting in a wide spectrum of effects on female reproduction are largely unknown. Here we summarize the major types of molecular cross-talks that have been identified for the AhR and linked cell signaling pathways and that are relevant for the understanding of the role of this transcription factor in female reproduction. Publication Types: Review PMID: 15798013 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 30: Curr Med Chem. 2005;12(5):599-616. Molecular actions of polyhalogenated arylhydrocarbons (PAHs) in female reproduction. Hombach-Klonisch S, Pocar P, Kietz S, Klonisch T. Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, R3E 0W3, Canada. klonisch@cc.umanitoba.ca Polyhalogenated aromatic arylhydrocarbons (PAHs) such as polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs), the polychlorinated biphenyls (PCBs) and polybrominated diphenyl ethers (PBDEs) are persistent lipophilic pollutants, which affect female fertility resulting in severe reproductive dysregulation, including anovulation, reduced conception rates, abortion, menstrual abnormalities and developmental defects of female reproductive tissues. Many PAHs exert their effects by activating a family of basic helix-loop-helix (bHLH) transcription factors, the arylhydrocarbon receptor (AhR) and the arylhydrocarbon receptor nuclear translocator (ARNT), which result in the expression of AhR target molecules. Complex interactions between PAH-mediated AhR activation and ER signalling pathways have been discovered which may contribute to the developmental malformations, impact on reproductive dysfunctions and promote carcinogenic dedifferentiation of tissues within the female reproductive tract. This review will focus on the multifaceted roles of PAHs in key organs of the female reproductive tract, the ovary, uterus/ endometrium and the mammary gland. The complexity and diversity of actions unleashed by PAHs in these female reproductive tissues identify these environmental pollutants as important endocrine disrupting toxicants impacting on female fertility. Publication Types: Review Review, Tutorial PMID: 15777215 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 31: Toxicol Sci. 2005 May;85(1):727-34. Epub 2005 Feb 23. Inhibitory effects of vitamin A on TCDD-induced cytochrome P-450 1A1 enzyme activity and expression. Yang YM, Huang DY, Liu GF, Zhong JC, Du K, Li YF, Song XH. Molecular Biology Center, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is an extremely potent environmental contaminant that produces a wide range of adverse biological effects, including the induction of cytochrome P450 1A1(CYP1A1) that may enhance the toxic effects of TCDD. Several studies indicated that concurrent supplementation of vitamin A could reduce the toxicity, and potentially inhibit CYP1A1 activity (measured as ethoxyresorufin-O-deethylase [EROD] activity). In the present study, we investigated the in vivo effects of vitamin A on EROD activities and the expression of CYP1A1 in the liver of TCDD-treated mice. In Experiment I, the mice were given a single oral dose of 40 mug TCDD/kg body weight with or without the continuous administration of 2500 IU vitamin A/kg body weight/day, and were killed on day 1, 3, 7, 14, or 28. In Experiment II, the mice were given daily an oral dose of 0.1 mug TCDD/kg body weight with or without supplement of 2000 IU vitamin A/kg body weight, and were killed on day 14, 28, or 42. In both experiments, TCDD caused liver damage and increase in relative liver weights, augmented the EROD activities and CYP1A1 expression, and increased the aryl hydrocarbon receptor (AhR) mRNA expression, but did not alter the AhR nuclear translocator (ARNT) mRNA expression. CYP1A1 mRNA expression and AhR mRNA expression showed a similar time course. The liver damage in TCDD + vitamin A-treated mice was less severe than that in TCDD-treated mice. EROD activities, CYP1A1 expression, and AhR mRNA expression in vitamin A + TCDD-treated mice were lower than those in TCDD-treated mice, indicating that supplementation of vitamin A might attenuate the liver damage caused by TCDD. PMID: 15728703 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 32: J Immunol. 2005 Mar 1;174(5):2770-7. Constitutively active aryl hydrocarbon receptor expressed specifically in T-lineage cells causes thymus involution and suppresses the immunization-induced increase in splenocytes. Nohara K, Pan X, Tsukumo S, Hida A, Ito T, Nagai H, Inouye K, Motohashi H, Yamamoto M, Fujii-Kuriyama Y, Tohyama C. Environmental Health Sciences Division, National Institute for Environmental Studies, University of Tsukuba, Tsukuba, Japan. keikon@nies.go.jp The aryl hydrocarbon receptor (AhR) is a transcription factor belonging to the basic helix-loop-helix-PER-ARNT-SIM superfamily. Xenobiotics, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin, bind the receptor and trigger diverse biological reactions. Thymocyte development and T cell-dependent immune reactions are sensitive targets of AhR-dependent 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicity. However, the exact role of the AhR in T cells in animals exposed to exogenous ligands has not been clarified because indirect effects of activated AhR in other cell types cannot be excluded. In this study, we generated transgenic (Tg) mice expressing a constitutively active mutant of AhR under the regulation of a T cell-specific CD2 promoter to examine AhR function in T cells. The mRNAs of the constitutively active mutant of AhR and an AhR-induced gene, CYP1A1, were expressed in the thymus and spleen of the Tg mice. The transgene expression was clearly detected in the thymocytes, CD4, and CD8 T cells, but not in the B cells or thymus stromal cells. These Tg mice had a decreased number of thymocytes and an increased percentage of CD8 single-positive thymocytes, but their splenocytes were much less affected. By contrast, the increase in number of T cells and B cells taking place in the spleen after immunization was significantly suppressed in the Tg mice. These results clearly show that AhR activation in the T-lineage cells is directly involved in thymocyte loss and skewed differentiation. They also indicate that AhR activation in T cells and not in B cells suppresses the immunization-induced increase in both T cells and B cells. PMID: 15728486 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 33: Bull Exp Biol Med. 2004 Sep;138(3):233-6. Transcriptional activation of cytochrome P450 1A1 with alpha-tocopherol. Sidorova YA, Grishanova AY, Lyakhovich VV. Laboratory for Biochemistry of Foreign Compounds, Institute of Molecular Biology and Biophysics, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk, Russia. Peroral administration of alpha-tocopherol in a daily dose of 150 mg/kg for 1, 4, 8, and 12 days leads to induction of cytochromes P450 1A in male rats. Activity of CYP1A1 and CYP1A2 increased most significantly one day after alpha-tocopherol administration (by 2.6 and 2.7 times, respectively). CYP1A1 was immunohistochemically detected in rat liver microsomes during this period. The content of CYP1A1 mRNA significantly increased in the liver. The amount of CYP1A2 mRNA and regulatory proteins for signal activation of CYP1A1 (AhR and Arnt) remained unchanged after treatment with alpha-tocopherol. PMID: 15665910 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 34: Toxicology. 2005 Mar 1;208(1):1-11. Effect of TCDD on mRNA expression of genes encoding bHLH/PAS proteins in rat hypothalamus. Korkalainen M, Linden J, Tuomisto J, Pohjanvirta R. National Public Health Institute, Department of Environmental Health, P.O. Box 95, FIN-70701 Kuopio, Finland. merja.korkalainen@ktl.fi 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) brings about a wide variety of toxic and biochemical effects via an AH receptor (AHR)-mediated signalling pathway. Wasting syndrome and acute lethality are TCDD-induced endpoints showing a striking sensitivity difference between two rat strains, TCDD-sensitive Long-Evans (Turku/AB) (L-E) and TCDD-resistant Han/Wistar (Kuopio) (H/W). These rat strains were used to study hypothalamic effects of TCDD on expression of genes encoding AHR-regulated bHLH/PAS proteins potentially involved in molecular pathogenesis of the wasting syndrome. In addition, two well-established target genes of TCDD, CYP1A1 and CYP1A2 were also examined. Quantitative RT-PCR was used to measure mRNA levels in hypothalamus, which is a major center of food intake and body weight regulation. At both 6 and 96 h after a single dose of 50 microg/kg TCDD, significant elevations were found in mRNA levels of AHR repressor (AHRR), CYP1A1 and CYP1A2, but not those of AHR, ARNT or ARNT2. Likewise, TCDD (100 microg/kg) did not alter the expression of SIM1, implicated in the suppressive impact of TCDD on food intake, nor that of PER2, involved in regulation of circadian rhythms. Differences between H/W and L-E rats appeared in constitutive levels of AHR and ARNT and in TCDD-induced levels of CYP1A2, AHRR, AHR and ARNT, which all were about two- to four-fold lower in H/W rats. Thus, although the changes found do not account for the wasting syndrome, expression of all principal genes of the AHR-signalling pathway in rat hypothalamus make it a candidate target for TCDD. PMID: 15664428 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 35: Toxicology. 2005 Feb 28;207(3):401-9. Identification of aldehyde oxidase 1 and aldehyde oxidase homologue 1 as dioxin-inducible genes. Rivera SP, Choi HH, Chapman B, Whitekus MJ, Terao M, Garattini E, Hankinson O. Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, University of California, 650 Charles E. Young Dr., Factor Bldg. 13-230, Los Angeles, CA 90095-1732, USA. Aldehyde oxidases are a family of highly related molybdo-flavoenzymes acting upon a variety of compounds of industrial and medical importance. We have identified aldehyde oxidase 1 (AOX1) as a 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin) inducible gene in the mouse hepatoma cell line Hepa-1. AOX1 mRNA levels were not increased by dioxin in mutant derivatives of the Hepa-1 cell line lacking either functional aryl hydrocarbon receptor (AHR) or aryl hydrocarbon receptor nuclear translocator (ARNT) proteins, thus demonstrating that transcriptional induction of AOX1 in response to dioxin occurs through the AHR pathway. Dioxin induction of AOX1 mRNA was also observed in mouse liver. In addition, levels of AOX1 protein as well as those of aldehyde oxidase homologue 1 (AOH1), a recently identified homolog of AOX1, were elevated in mouse liver in response to dioxin. Employing an aldehyde oxidase specific substrate, AOX1/AOH1 activity was shown to be induced by dioxin in mouse liver. This activity was inhibited by a known inhibitor of aldehyde oxidases, and eliminated by including tungstate in the mouse diet, which is known to lead to inactivation of molybdoflavoenzymes, thus confirming that the enzymatic activity was attributable to AOX1/AOH1. Our observations thus identify two additional xenobiotic metabolizing enzymes induced by dioxin. PMID: 15664268 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 36: Mol Pharmacol. 2005 Apr;67(4):1325-33. Epub 2005 Jan 18. Regulation of the Cyp2a5 gene involves an aryl hydrocarbon receptor-dependent pathway. Arpiainen S, Raffalli-Mathieu F, Lang MA, Pelkonen O, Hakkola J. Department of Pharmacology and Toxicology, P.O. Box 5000, University of Oulu, FIN-90014 Oulun yliopisto, Finland. We have investigated the role of the aryl hydrocarbon receptor (AHR) in the regulation of the Cyp2a5 gene. The C57BL/6 and DBA/2 mouse strains with a genetically determined difference in AHR function were used to study the CYP2A5 induction by typical AHR ligands, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and 3-methylcholanthrene. The CYP2A5 mRNA up-regulation in these mouse strains showed a difference in response, typical for AHR-regulated genes, both by TCDD in cultured primary hepatocytes and by 3-methylcholanthrene in vivo. In primary hepatocytes, TCDD caused a 3-fold elevation of the CYP2A5 protein level and a similar induction of the CYP2A5-catalyzed coumarin 7-hydroxylation activity. In reporter gene assays, the Cyp2a5 promoter region -3033 to +10 mediated a 2- to 5-fold induction of luciferase activity by TCDD treatment in primary hepatocytes and in Hepa-1 hepatoma cells with an intact AHR/AHR nuclear translocator (ARNT) complex. In Hepa-1 variant cell lines with deficiencies in the AHR/ARNT complex, the absence of ARNT abolished the induction. A putative AHR response element (XRE) was identified in the Cyp2a5 promoter at the position -2514 to -2492 and found to interact with the AHR/ARNT heterodimer. Transfection experiments combined with mutation of the XRE site indicated that the site partly mediates the TCDD induction of Cyp2a5. An additional AHR-dependent mechanism also regulates the proximal promoter of the Cyp2a5 gene. In conclusion, our studies showed that AHR ligands up-regulate Cyp2a5 transcriptionally by an AHR/ARNT-dependent mechanism and established Cyp2a5 as a novel AHR-regulated gene. PMID: 15657367 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 37: J Pharm Sci. 2005 Mar;94(3):493-506. Down-regulation of aryl hydrocarbon receptor-regulated genes by tumor necrosis factor-alpha and lipopolysaccharide in murine hepatoma Hepa 1c1c7 cells. Gharavi N, El-Kadi AO. Faculty of Pharmacy and Pharmaceutical Sciences, 3118 Dentistry/Pharmacy Centre, University of Alberta, Edmonton, AB, Canada T6G 2N8. Although much is known concerning the effects of inflammation and oxidative stress on the cytochrome P450 1A1 (CYP1A1), little is known about the modulation of other aryl hydrocarbon receptor (AHR)-regulated genes such as glutathione-S-transferase Ya (GST Ya) and NAD(P)H:quinone oxidoreductase (QOR) by inflammation. In the present study, the effect of tumor necrosis factor (TNF)-alpha and lipopolysaccharides (LPS) on the constitutive and inducible expression of the AHR-regulated genes cyp1a1, GST Ya, and QOR was determined in murine hepatoma Hepa 1c1c7 (WT), AHR-deficient (C12), and AHR nuclear translocator protein (ARNT)-deficient (C4) cells. We found that both TNF-alpha and LPS strongly repressed the constitutive expression and the beta-naphthoflavone-mediated induction of cyp1a1, GST Ya, and QOR in WT but not in C12 and C4 cells. The induction of GST Ya and QOR activities and mRNA levels by phenolic antioxidant, tert-butylhydroquinone, through the antioxidant response element was not significantly affected by TNF-alpha or LPS. In addition, a significant increase in reactive oxygen species was observed in WT, C12, and C4 cells treated with TNF-alpha or LPS which was completely prevented by tert-butylhydroquinone. These results show that the down-regulation of AHR-regulated genes by TNF-alpha and LPS is dependent on the presence of both heterodimeric transcription factors, AHR and ARNT. Furthermore, reactive oxygen species may be involved in the down-regulation of AHR-regulated genes. Copyright 2004 Wiley-Liss, Inc. and the American Pharmacists Association. PMID: 15627257 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 38: Toxicol Lett. 2005 Feb 15;155(2):253-8. Rat hepatic CYP1A1 and CYP1A2 induction by menadione. Sidorova YA, Grishanova AY, Lyakhovich VV. Institute of Molecular Biology and Biophysics, Siberian Division of Russian Academy of Medical Sciences, Timakova str. 2, Novosibirsk 630090, Russia. ysidorova@soramn.ru The effects of menadione on activities and expression of cytochrome P450 (CYP) 1A subfamily (CYP1A) isozymes in rat hepatic tissue were examined. When rats were treated orally with 15 mg/kg menadione for 4 days, the elevation of hepatic CYP1A1/1A2 specific activities in microsomal preparations was detected with approximately 5.4- and 2.5-fold increase over control values for ethoxyresorufin-O-deethylase (EROD, CYP1A1) and methoxyresorufin-O-demethylase (MROD, CYP1A2) activities, respectively. CYP1A1 and CYP1A2 mRNA levels in the liver of menadione-treated rats were approximately 11.8- and 1.8-fold higher than in controls, respectively, whereas the expression of the CYP1A regulatory proteins aryl hydrocarbon-receptor (AhR) and AhR nuclear translocator (Arnt) was not changed at the mRNA level. The result of this study demonstrates that menadione induces CYP1A1/1A2 expression in vivo through either transcriptional activation and/or mRNA stabilization. PMID: 15603920 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 39: J Toxicol Environ Health B Crit Rev. 2004 Nov-Dec;7(6):465-80. Immune systems, geographic information systems (GIS), environment and health impacts. Blanco GA, Cooper EL. Department of Immunology, IDEHU-National Research Council (CONICET), School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina. gblanco@ffyb.uba.ar Exposure to dioxins, polychlorinated biphenyls (PCBs), and polycyclic aromatic hydrocarbons (PAHs) has been related to alterations in cellular and humoral immune responses in both adaptive and innate immune systems of most animal species. These compounds share a common signaling mechanism to exert their effects on cells of the immune system, which includes the aryl-hydrocarbon receptor (AhR) and the AhR nuclear translocator (ARN). Recently, the interference of AhR-ARNT with the nuclear factor (NF)-kappaB signaling pathway has been proposed as a critical event in the adverse effects on the immune system. Studies on the effects of these AhR-ARNT-related toxicants on the immune system of higher and lower phylum animals and knowledge of intracellular mechanisms of toxicity may contribute to development of biomarkers of ecotoxicant exposure and effects. Biomarkers of this kind allow sampling over extended geographic areas, in several sentinel species, including wildlife animals, and facilitate the building of risk models and risk maps of environmentally induced diseases. On the basis of location, biomarker sampled data obtained through evaluation of ecotoxicant exposure and effects on the immune system in sentinel species can be further integrated and analyzed together with other sources of environmental geographic information, or human population health data, by means of geographic information systems (GIS). The spatial analysis capability of GIS can help to evaluate the complex relationships of overlaid information and to identify areas with high risk indices or "hot spots." This integrative approach can be useful in studies contributing to support environmental and health-related policies and regulations. Publication Types: Review Review, Tutorial PMID: 15586880 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 40: Toxicol Lett. 2005 Jan 15;155(1):151-9. Hypoxic inhibition of 3-methylcholanthrene-induced CYP1A1 expression is independent of HIF-1alpha. Allen JW, Johnson RS, Bhatia SN. Division of Biological Sciences, Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA. Hypoxia-inducible factor-1alpha (HIF-1alpha) and aryl hydrocarbon receptor (AhR) both require dimerization with AhR nuclear translocator (ARNT) to initiate transcription of their respective target genes. It has been proposed that competition for ARNT results in decreased targeting of AhR to cytochrome P450 1A1 (CYP1A1) under hypoxia. We established primary cultures of HIF-1alpha null hepatocytes to examine the interaction between HIF-1alpha and AhR signaling. Gene expression of known HIF targets phosphoglycerate kinase (PGK), vascular endothelial growth factor (VEGF) and glucose transporter-1 (GLUT-1) increased under hypoxia, but was reduced in the HIF null cultures. Concomitant treatment of cultures with hypoxia (1% O2) and 3-methylcholanthrene (an AhR ligand) did not significantly alter HIF target gene expression. Furthermore, enzymatic activity and transcription of CYP1A1 was inhibited by hypoxia in HIF-1alpha null cultures, indicating that HIF-1alpha is not directly involved in negative regulation of AhR signaling. PMID: 15585370 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 41: Proc Natl Acad Sci U S A. 2004 Nov 23;101(47):16677-82. Epub 2004 Nov 15. Gestational exposure of Ahr and Arnt hypomorphs to dioxin rescues vascular development. Walisser JA, Bunger MK, Glover E, Bradfield CA. McArdle Laboratory for Cancer Research, University of Wisconsin Medical School, Madison, WI 53706, USA. The aryl hydrocarbon receptor (AHR) is commonly known for its role in the adaptive metabolism of xenobiotics and in the toxic events that follow exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin). Previously, we have demonstrated that the AHR and its heterodimeric partner, the AHR nuclear translocator (ARNT), play a role in the developmental closure of a hepatic vascular shunt known as the ductus venosus (DV). To investigate the mechanism of DV closure, we generated hypomorphic alleles of the Ahr and Arnt loci. Using these models, we then asked whether this vascular defect could be rescued by receptor activation during late development. By manipulating gestational exposure, the patent DV in AHR or ARNT hypomorphs could be efficiently closed by dioxin exposure as early as embryonic day 12.5 and as late as embryonic day 18.5. These findings define the temporal regulation of receptor activation during normal ontogeny and provide evidence to support the idea that receptor activation and AHR-ARNT heterodimerization are essential for normal vascular development. Taken in the broader context, these data demonstrate that similar AHR signaling steps govern all major aspects of AHR biology. PMID: 15545609 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 42: Toxicol Sci. 2005 Feb;83(2):340-8. Epub 2004 Nov 10. Aryl hydrocarbon receptor expression and activity in cerebellar granule neuroblasts: implications for development and dioxin neurotoxicity. Williamson MA, Gasiewicz TA, Opanashuk LA. Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent teratogen that produces neurobehavioral abnormalities associated with both cognitive and locomotor systems, yet the precise regional and cellular targets of developmental neurotoxicity remain largely unknown. Most, if not all, TCDD-induced pathology is mediated via binding to the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that belongs to the basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) superfamily. Upon ligand binding, AhR translocates to the nucleus, dimerizes with the AhR nuclear translocator protein (Arnt), and regulates transcription by interaction with dioxin-response elements (DREs) in target genes, most notably specific cytochrome P450 (CYP) family members. To assess whether developing cerebellar granule neuroblasts are potential direct targets for TCDD toxicity, AhR expression and transcriptional activity were examined. AhR and Arnt proteins were present in mouse cerebellum from birth throughout postnatal development. AhR protein levels peaked between postnatal day (PND) 3-10, a critical period for granule neuroblast growth and maturation. Transcriptionally active AhR was detected in immature cerebellar granule cells in a transgenic dioxin-responsive lacZ mouse model after acute TCDD exposure. AhR and Arnt were also expressed in cerebellar granule neuroblast cultures. AhR localized to the nucleus in granule cells 15 min after TCDD treatment. TCCD elicited time-dependent and concentration-dependent increases in CYP1A1 and 1B1 mRNA and protein levels. Moreover, TCDD treatment reduced both thymidine incorporation and granule neuroblast survival in a concentration-dependent manner. These data suggest that (1) granule neuroblasts are direct targets for developmental AhR-mediated TCDD neurotoxicity and (2) TCDD exposure may disrupt granule cell neurogenesis. PMID: 15537747 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 43: Huan Jing Ke Xue. 2004 Jul;25(4):155-8. [Modification and application of recombinant yeast bioassay for measuring the AhR ligand activity] [Article in Chinese] Wang W, Hu JY, Gu WM, Liu YH. College of Environmental Science, Peking University, Beijing 100871, China. A recombinant yeast bioassay, a yeast Saccharomyces cerevisiae, in which the human AhR and ARNT complex are coexpressed, is one of the methods to screen the active AhR agonist. In this study, the original agonist test was modified. The exposure time was reduced from 18 to 8 hours when experiment was under the following conditions: (1) the yeast was cultured in 0.2% glucose medium for 24 hours; (2) chemical exposure was carried in 2% galactose medium in glass tube. Finally, the AhR acitivity of hexachlorobenzene and pentachlorobenzene were assessed, and their toxicity equivalent factors were found to be 0.018629 and 0.000294, respectively. PMID: 15515957 [PubMed - in process] --------------------------------------------------------------- 44: Chem Biol Interact. 2004 Oct 15;149(2-3):151-64. Metabolic fate of the Ah receptor ligand 6-formylindolo[3,2-b]carbazole. Bergander L, Wincent E, Rannug A, Foroozesh M, Alworth W, Rannug U. Department of Genetics, Microbiology and Toxicology, Stockholm University, S-10691 Stockholm, Sweden. The physiological role of the aryl hydrocarbon receptor (AhR), a member of the basic helix-loop-helix PER-ARNT-SIM (PAS) transcription factor family is not known. We have suggested that the AhR is involved in light signaling through binding of photoproducts with high AhR affinity. This suggestion is based on (i) the high AhR affinity of the tryptophan photoproduct formylindolo[3,2-b]carbazole (FICZ), (ii) the induction of rapid and transient expression of AhR-regulated genes by FICZ and by extracts of UV-irradiated tryptophan as well as (iii) the fact that light induces the AhR-regulated cytochrome P450s CYP1A1, CYP1B1 and CYP2S1. The transient mRNA expression caused by light and tryptophan photoproducts suggests that the biotransformation enzymes induced by AhR activation take part in a metabolic degradation of the natural AhR ligand. This study aimed at identifying the involvement of phase I and phase II enzymes in the metabolic degradation of FICZ. A cytochrome P450-dependent metabolism of FICZ giving rise to preferentially mono- and di-hydroxylated derivatives has earlier been reported. In the present study, rat and human hepatic S9 mixes were employed together with specific enzyme inhibitors and cofactors. Compared to the Aroclor-induced rat liver S9, the non-induced rat liver S9 and the human liver S9 caused a more complex metabolite profile of FICZ. The CYP1A1 enzyme was confirmed to be the most important enzyme for the first step in the metabolism. CYP1A2 was found to have overlapping specificity with CYP1A1 being able to form the same major metabolites although with different kinetics. CYP1B1 turned out to be preferentially involved in the further metabolism of dihydroxylated metabolites. Microsomal epoxide hydrolase, and as yet not identified forms of sulphotransferases and glucuronosyltransferases were also found to take part in the metabolic degradation of FICZ. Thus, tryptophan photoproducts fit into a model in which the ligand-activated AhR signaling is autoregulated by the induced metabolic enzymes. PMID: 15501436 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 45: Mol Pharmacol. 2005 Jan;67(1):88-96. Epub 2004 Oct 18. Multiple mechanisms are involved in Ah receptor-mediated cell cycle arrest. Huang G, Elferink CJ. Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA. The liver is the only solid organ that can respond to major tissue loss or damage by regeneration to restore liver biomass. The aryl hydrocarbon receptor (AhR) agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can disrupt the regenerative process, as evidenced by suppression of DNA synthesis in rat primary hepatocytes in culture and in vivo liver regeneration after partial hepatectomy. Independent observations demonstrated that AhR-mediated G(1) phase cell cycle arrest depends on an interaction with the retinoblastoma tumor suppressor protein (pRb), but differences exist regarding proposed mechanisms of action. Two distinct models have been proposed, one supporting the AhR-pRb interaction functioning in corepression of E2F activity and the other favoring an AhR-pRb interaction participating in transcriptional coactivation of genes encoding G(1) phase regulatory proteins. In the present study, experiments in rat hepatoma cells using dominant-negative DNA-binding-defective AhR and Ah receptor nuclear translocator (Arnt) mutants provided evidence that TCDD-induced AhR-mediated G(1) arrest is only partially regulated by direct AhR transcriptional activity, suggesting that both coactivation and corepression are involved. Studies using a small interfering RNA to down-regulate Arnt protein expression revealed that TCDD-induced G(1) arrest is absolutely dependent on the Arnt protein. PMID: 15492120 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 46: J Biol Chem. 2004 Dec 24;279(52):54620-8. Epub 2004 Oct 13. Recruitment of thyroid hormone receptor/retinoblastoma-interacting protein 230 by the aryl hydrocarbon receptor nuclear translocator is required for the transcriptional response to both dioxin and hypoxia. Beischlag TV, Taylor RT, Rose DW, Yoon D, Chen Y, Lee WH, Rosenfeld MG, Hankinson O. Department of Pathology and Laboratory Medicine, Molecular Biology Institute, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA. The aryl hydrocarbon receptor nuclear translocator/hypoxia-inducible factor (ARNT/HIF-1 beta) mediates an organism's response to various environmental cues, including those to chemical carcinogens, such as 2,3,7,8-tetrachlorodibenzo-rho-dioxin (TCDD or dioxin), via its formation of a functional transcription factor with the ligand activated aryl hydrocarbon receptor (AHR). Similarly, tissue responses to hypoxia are largely mediated through the HIF-1 heterodimeric transcription factor, comprising hypoxia-inducible factor-1 alpha (HIF-1 alpha) and ARNT. The latter response is essential for a metabolic switch from oxidative phosphorylation to glycolytic anaerobic metabolism as well as for angiogenesis and has been implicated as necessary for growth in many solid tumors. In this report, we demonstrate that the thyroid hormone receptor/retinoblastoma-interacting protein 230 (TRIP230) interacts directly with ARNT and is essential for both hypoxic and TCDD-mediated transcriptional responses. We initially identified TRIP230 as an ARNT-interacting protein in a yeast two-hybrid assay screen. This interaction was confirmed in mammalian cell systems using co-immunoprecipitation and in mammalian two-hybrid assays. Furthermore, TRIP230 could be recorded at sites of activated transcription of either TCDD- or hypoxia-inducible genes in a stimulus-dependent fashion by chromatin immunoprecipitation analysis. Finally, using single-cell microinjection and RNA interference assays, we demonstrate that TRIP230 is indispensable for TCDD- and hypoxia-dependent gene transcription. PMID: 15485806 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 47: Mutat Res. 2004 Oct 4;554(1-2):267-77. Identification of genetic polymorphisms of CYP2S1 in a Finnish Caucasian population. Saarikoski ST, Suitiala T, Holmila R, Impivaara O, Jarvisalo J, Hirvonen A, Husgafvel-Pursiainen K. Department of Industrial Hygiene and Toxicology, Finnish Institute of Occupational Health, Helsinki FI-00250. sirkku.saarikoski@ktl.fi CYP2S1 is a recently discovered member of the cytochrome P450 (CYP) gene superfamily. Interestingly, even though the DNA sequence identifies it as the sole member of the new CYP2S family, CYP2S1 exhibits many features typical to CYP1 family members, e.g. dioxin-inducibility mediated by the aryl hydrocarbon receptor (AHR) and the aryl hydrocarbon receptor nuclear translocator (ARNT). In addition, CYP2S1 metabolises some aromatic hydrocarbons as well as cellular substances. These characteristics, together with a wide extrahepatic tissue distribution, suggest that CYP2S1 may have an important role in both exogenous and endogenous metabolism. This is the first study characterising CYP2S1 alleles and naming them with the recommended CYP allele nomenclature. We used denaturing gradient gel electrophoresis (DGGE) and direct sequencing to investigate genetic variation of CYP2S1 in 100 male Finnish Caucasians. Those exons in which variation was found were examined in subsequent 100 subjects. The coding region of all of the nine exons, as well as a 449 bp fragment of the proximal promoter region, was analysed. This systematic investigation revealed eight single nucleotide polymorphisms (SNPs), which comprise nine different variant alleles (haplotypes), in addition to the wild-type allele. Seven of the SNPs occurred in the protein-coding areas and one in the proximal 3' untranslated region (3'UTR). Two of these sequence variations (10347C > T and 13106C > T) result in non-conservative amino acid substitutions, i.e. Arg380Cys and Pro466Leu, respectively. The respective allelic variants, CYP2S1*2 ([10347C > T]) and CYP2S1*3 (13106C > T; 13255A > G]), occurred in our study population at frequencies of 0.50 and 3.75%, respectively. The most common of the variant alleles was CYP2S1*1H (23.8%), harbouring a 13255A > G substitution located in the 3'UTR. PMID: 15450424 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 48: J Biol Chem. 2004 Nov 26;279(48):49842-8. Epub 2004 Sep 20. Role of the coiled-coil coactivator (CoCoA) in aryl hydrocarbon receptor-mediated transcription. Kim JH, Stallcup MR. Department of Pathology and Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California 90089, USA. The aryl hydrocarbon receptor (AHR) and AHR nuclear translocator (ARNT) are DNA binding transcription factors with basic helix-loop-helix/Per-Arnt-Sim (bHLH-PAS) domains. These two proteins form a heterodimer that mediates the toxic and biological effects of the environmental contaminant and AHR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin. The coiled-coil protein coiled-coil coactivator (Co-CoA) is a secondary coactivator for nuclear receptors and enhances nuclear receptor function by interacting with the bHLH-PAS domain of p160 coactivators. We report here that CoCoA also binds the bHLH-PAS domains of AHR and ARNT and functions as a potent primary coactivator for them; i.e. CoCoA does not require p160 coactivators for binding to and serving as a coactivator for AHR and ARNT. Endogenous CoCoA was recruited to a natural AHR target gene promoter in a 2,3,7,8-tetrachlorodibenzo-p-dioxin -dependent manner. Moreover, reduction of CoCoA mRNA levels by small interfering RNA inhibited the transcriptional activation by AHR and ARNT. Our data support a physiological role for CoCoA as a transcriptional coactivator in AHR/ARNT-mediated transcription. PMID: 15383530 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 49: Am J Med Genet A. 2004 Sep 15;130(1):40-4. Significant association between nonsyndromic oral clefts and arylhydrocarbon receptor nuclear translocator (ARNT). Kayano S, Suzuki Y, Kanno K, Aoki Y, Kure S, Yamada A, Matsubara Y. Department of Medical Genetics, Tohoku University School of Medicine, Sendai, Japan. The etiology of nonsyndromic oral clefts (cleft lip, cleft palate, or cleft lip and palate) is still controversial, but is considered to involve both genetic and environmental factors. One of suspected environmental factors is 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) found in tobacco, herbicides, contaminated soil, and food. TCDD administered during organogenesis in mice causes a high incidence of CP in fetuses. There is ample evidence that aryl hydrocarbon receptor (AHR), AHR nuclear translocator (ARNT), and cytochrome P450 1A1 (CYP1A1) are involved in TCDD metabolism. We assessed whether there is any association in the Japanese population of nonsyndromic oral clefts with single nucleotide polymorphisms (SNPs) in the AHR, ARNT, and CYP1A1 genes using transmission disequilibrium test (TDT) and case-control study. We identified and investigated three SNPs in ARNT; 567G/C (V189V), IVS12-19T/G, and 2117C/T (P706L). Two amino acid substitutions, R554L in AHR and I462V in CYP1A1, were also investigated. In the TDT, the C allele of ARNT 567G/C was preferentially transmitted to patients (P = 0.033). When a haplotype consisting of 567G/C and IVS12-19T/G in ARNT was considered, the preferential transmission of the CT (567C-IVS12-19T) haplotype was observed (P = 0.0012). In a case-control study, a significant association of IVS12-19T/G in ARNT was observed (P = 0.021). The SNPs studied in AHR and CYP1A1 were not associated with the disease. Our results suggest that ARNT is involved in the development of nonsyndromic oral clefts in the Japanese population. Copyright 2004 Wiley-Liss, Inc. PMID: 15368494 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 50: Toxicol Appl Pharmacol. 2004 Sep 15;199(3):220-38. Disruption of cell-cell contact maximally but transiently activates AhR-mediated transcription in 10T1/2 fibroblasts. Cho YC, Zheng W, Jefcoate CR. Molecular and Cellular Pharmacology Program, University of Wisconsin Medical School, Madison 53706, USA. The aryl hydrocarbon receptor (AhR) is activated by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), but activation without an exogenous ligand also occurs when normal cell-cell contact is prevented. Suspension of several C3H10T1/2 fibroblast clonal sub-lines that contain an integrated AhR-responsive reporter produced a time course and level of reporter activation and CYP1B1 induction that paralleled TCDD stimulation in confluent monolayer culture. Suspension activation was, however, more transient. Loss of cell-cell contact at low density also activated these reporters independent of cell cycle changes to levels comparable to TCDD stimulation of confluent cells. Loss of cell-cell contact may, therefore, activate AhR. Suspension and TCDD activations exhibited comparable nuclear translocation of AhR and then AhR/ARNT complex formation. Each AhR activation process was equally attenuated by inhibition of, respectively, HSP90 ATPase, the 26S proteosome, and by depletion of intracellular Ca2+. By contrast, the AhR antagonist alpha-naphthoflavone (alphaNF) blocked ligand-stimulated AhR activity, but not activation through loss of cell-cell contact. Suspension-induced reporter activation was selectively enhanced by LiCl, which prevented GSK-3beta effects on the simultaneously released beta-catenin. The effects of suspension and LiCl on reporters were reversed by Ro-31-8220, which did not affect beta-catenin, TCDD-activation processes, or AhR turnover. Neither LiCl nor Ro-31-8220 altered suspension-induced AhR/ARNT complex formation. Loss of cell-cell contact permits nuclear translocation and AhR activation that is largely replicated after TCDD binding, but with activity differences due to contact-sensitive factors functioning after AhR/ARNT complex formation. PMID: 15364539 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 51: Biochem Biophys Res Commun. 2004 Aug 27;321(3):707-15. Dioxin-responsive AHRE-II gene battery: identification by phylogenetic footprinting. Boutros PC, Moffat ID, Franc MA, Tijet N, Tuomisto J, Pohjanvirta R, Okey AB. Department of Pharmacology, University of Toronto, Toronto, Ont., Canada M5S 1A8. We identified a set of genes that respond to dioxins through the recently discovered AHRE-II ("XRE-II") enhancer element. A total of 36 genes containing AHRE-II motifs conserved across human, mouse, and rat gene orthologs were identified by genome-wide transcription-factor binding-site searches and phylogenetic footprinting. Microarray experiments on liver from rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin revealed statistically significant changes in mRNA levels for 13 of these 36 genes after three hours and 15 genes after 19h. The set of responsive genes was functionally characterized by ontological analysis and found to be enriched in ion-channels and transporters. Our identification of 36 putatively AHRE-II-regulated genes highlights the regulatory versatility of the aryl hydrocarbon receptor (AHR) and the ability of the AHR and its dimerization partner, ARNT, to act both as a ligand-activated transcription-factor (on AHRE-I) and as a ligand-activated coactivator (on AHRE-II). Collectively, these results demonstrate that the AHRE-II induction mechanism is employed by multiple genes and provide the first categorization of the gene battery of a ligand-activated coactivator. PMID: 15358164 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 52: Mol Pharmacol. 2004 Sep;66(3):512-21. 2,3,7,8-Tetrachlorodibenzo-p-dioxin activation of the aryl hydrocarbon receptor/aryl hydrocarbon receptor nuclear translocator pathway causes developmental toxicity through a CYP1A-independent mechanism in zebrafish. Carney SA, Peterson RE, Heideman W. Molecular and Environmental Toxicology Center, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, USA. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that dimerizes with ARNT to mediate responses to compounds such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). TCDD and other AHR agonists cause toxic responses in early life stages of fish, including the zebrafish, Danio rerio. The most well characterized target gene for the AHR/aryl hydrocarbon receptor nuclear translocator (ARNT) dimer is a cytochrome P450, CYP1A. Induction of CYP1A by TCDD has been correlated with certain toxic responses in developing zebrafish and has been postulated to mediate these responses. To determine whether CYP1A is the important downstream effector enzyme for the AHR/ARNT pathway, we used morpholino oligonucleotides (MOs) to block induction of CYP1A in response to TCDD in zebrafish embryos. Although the zfcyp1a-MO effectively prevented CYP1A up-regulation, it did not prevent the signs of developmental toxicity, including pericardial edema, slowed blood flow, craniofacial malformation, and defects in erythropoiesis. We conclude that the important target for the AHR/ARNT pathway in developing zebrafish exposed to TCDD is not zfcyp1a. This suggests an alternative model in which TCDD-activated AHR/ARNT disrupts the normal process of growth and development by altering programs of gene expression or function. PMID: 15322242 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 53: Biochem Biophys Res Commun. 2004 Sep 10;322(1):9-16. Inhibition of the MEK-1/p42 MAP kinase reduces aryl hydrocarbon receptor-DNA interactions. Yim S, Oh M, Choi SM, Park H. Department of Life Science, University of Seoul, 90 Cheonnong-dong, Tongdaemun-gu, Seoul 130-743, Republic of Korea. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces expression of the cytochrome P450 1A1 gene, cyp1a1, by binding to its receptor, aryl hydrocarbon receptor (AhR). TCDD-bound AhR translocates to the nucleus and forms a heterodimer with its partner protein, AhR nuclear translocator (Arnt). The AhR/Arnt heterodimer then binds to the dioxin-response elements (DREs) in the cyp1a1 enhancer and stimulates transcription of cyp1a1. We tested whether kinase pathways are involved in this process by treating Hepa1c1c7 cells with kinase inhibitors. The MEK-1 inhibitor PD98059 reduced TCDD-induced transcription of cyp1a1. TCDD treatment results in phosphorylation of p44/p42 mitogen-activated protein kinase (MAPK), a substrate of MEK-1. Overexpression of dominant negative form of p42 MAPK suppressed TCDD-dependent transcription of a reporter gene controlled by dioxin-response elements (DREs), and pretreatment with PD98059 also blocked this transcription. PD98059 pretreatment also inhibited TCDD-induced DRE binding of the AhR/Arnt heterodimer. Together these results indicate that TCDD activates the MEK-1/p44/p42 MAPK pathway, which in turn activates AhR and so facilitates binding of AhR to the cyp1a1 DRE. PMID: 15313166 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 54: Toxicol Sci. 2004 Nov;82(1):46-61. Epub 2004 Aug 5. Linked expression of Ah receptor, ARNT, CYP1A1, and CYP1B1 in rat mammary epithelia, in vitro, is each substantially elevated by specific extracellular matrix interactions that precede branching morphogenesis. Larsen MC, Brake PB, Pollenz RS, Jefcoate CR. Department of Pharmacology and the Environmental Toxicology Center, University of Wisconsin Medical School, 1300 University Avenue, Madison, WI 53706, USA. Cytochrome P4501B1 (CYP1B1), the major constitutively expressed CYP in the rat mammary gland, is induced by Ah-receptor (AhR) ligands, while CYP1A1 is predominantly expressed only after induction. These CYPs contribute to carcinogenic activation of polycyclic aromatic hydrocarbons (PAHs). AhR, ARNT, and CYP1B1 were only weakly expressed, even after 2,3,7,8-tetrachlorodibenzo-p-dioxin induction, when rat mammary epithelial cells (RMEC) were cultured on plastic. RMEC cultured on the extracellular matrix (ECM), Matrigel, or on a floating gel of collagen I demonstrated branching morphogenesis and substantially increased basal CYP1B1 and induced CYP1A1 expression, in parallel with large increases in AhR and ARNT expression. Branching was more pronounced in the Wistar Kyoto than in the Wistar Furth rat strain. Although EGF enhanced branching, neither strain nor growth factor treatment substantially impacted CYP expression. Increased AhR and ARNT expression is observed within 24 h of dispersal on Matrigel, substantially prior to branch formation. Culture on thin layers of collagen I, collagen IV, and laminin, respectively, failed to reproduce the branching morphogenesis or increases in AhR, ARNT, or CYP expression. However, adherent, gelled collagen I recapitulated the increased protein expression, without supporting branching. This increased protein expression was closely paralleled by enhanced expression of beta-catenin and E-cadherin, components of cell-cell adhesion complexes. A synthetic peptide that selectively antagonizes integrin-ECM interactions reduced branch formation, without diminishing AhR, ARNT, and CYP expression. These data demonstrate that early ECM surface adhesion interactions mediate AhR and ARNT expression, which enhances CYP expression, independent of branching morphogenesis. PMID: 15297627 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 55: Arch Biochem Biophys. 2004 Sep 1;429(1):42-9. CyP40, but not Hsp70, in rabbit reticulocyte lysate causes the aryl hydrocarbon receptor-DNA complex formation. Shetty PV, Wang X, Chan WK. Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, CA 95211, USA. Upon ligand binding, the aryl hydrocarbon receptor (AhR) translocates into the nucleus and dimerizes with its partner aryl hydrocarbon receptor nuclear translocator (Arnt). The AhR-Arnt heterodimer binds to the dioxin response element (DRE) to regulate target gene expression. Using baculovirus expressed human AhR and Arnt, we showed that the formation of the ligand-dependent AhR-Arnt-DRE complex requires protein factors in vitro. Recently, we provided evidence that p23, an Hsp90-associated protein, is involved in the complex formation. The aim of this study was to determine whether two other Hsp90-associated proteins present in rabbit reticulocyte lysate (RRL), namely CyP40 and Hsp70, play any role in forming the AhR-Arnt-DRE complex. Fractionation and immunodepletion experiments revealed that Hsp70 is not necessary for the formation of this complex. In contrast, CyP40 is involved in forming the complex since (1) immunodepletion of CyP40 from a RRL fraction reduces the intensity of the AhR-Arnt-DRE complex by 48% and (2) recombinant human CyP40 alone causes the formation of this complex. In addition, CyP40-interacting proteins appear to be essential for the full CyP40 effect on the AhR gel shift complex. PMID: 15288808 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 56: Toxicol Sci. 2004 Nov;82(1):80-7. Epub 2004 Jul 22. A critical role for MAP kinases in the control of Ah receptor complex activity. Tan Z, Huang M, Puga A, Xia Y. Center for Environmental Genetics and Department of Environmental Health, University of Cincinnati Medical Center, 123 E. Shields Street, Cincinnati, OH 45267-0056, USA. The heterodimeric complex of aromatic hydrocarbon receptor (AHR) and Ah receptor nuclear translocator (ARNT) plays a pivotal role in controlling the expression of drug metabolism genes, such as the cytochromes p450 (Cyp) 1a1 and 1b1, believed to be responsible for most toxic effects of the environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). In this study, we show that activation of Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK) modulates ARNT transcription activity and potentiates the transcriptional activity of AHR/ARNT complexes. Inhibition of ERK by chemical compounds and ablation of JNK caused significant decreases in CYP1A1 induction by TCDD. Compared to wild type, JNK2 ablation significantly reduced TCDD-stimulated CYP1A1 expression in mouse thymus and testis, but not in liver. In contrast, CYP1B1 expression was unaffected in all three tissues of the knockout mice. These data suggest that JNK and ERK modulate ARNT activity and AHR/ARNT-dependent gene expression, contributing to the gene-specific and tissue-specific toxicity of environmental contaminants. PMID: 15272135 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 57: Nucleic Acids Res. 2004 Jun 9;32(10):3169-79. Print 2004. Altered DNA binding specificity of Arnt by selection of partner bHLH-PAS proteins. Kinoshita K, Kikuchi Y, Sasakura Y, Suzuki M, Fujii-Kuriyama Y, Sogawa K. Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan. The Ah receptor (AhR) and HLF are transcription factors involved in xenobiotic metabolism and hypoxic response, respectively. AhR and HLF heterodimerize with Arnt as the common partner, and bind to asymmetric E-boxes termed XRE and HRE, respectively. In order to investigate nucleotide preference of the heterodimers, reporter plasmids with oligonucleotides for XREs or HREs with systematic mutations were constructed and their activity was determined. Comparison of the activity revealed that DNA length and nucleotide preference recognized by Arnt subunit in the two heterodimers were largely different between XRE and HRE. We expressed AhR-Arnt and HLF-Arnt in Escherichia coli and used them for DNA binding. The dissociation constant of HLF-Arnt-HRE was 10.4 +/- 1.6 nM. Competition activity of mutated XREs or HREs with wild type was consistent with their transcription activity. Bending of XRE and HRE induced by binding of the relevant heterodimers was observed with stronger bending of XRE than of HRE. By deletional and mutational analyses, an alanine and three arginine (Ala 8, Arg 9, Arg 11 and Arg 12) residues in the basic sequence of HLF were found to be indispensable for the transcriptional activity. PMID: 15190133 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 58: Biochem Pharmacol. 2004 Jul 1;68(1):63-71. Phenylthiourea as a weak activator of aryl hydrocarbon receptor inhibiting 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced CYP1A1 transcription in zebrafish embryo. Wang WD, Wang Y, Wen HJ, Buhler DR, Hu CH. Institute of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 202-24, Taiwan, ROC. The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that can be activated by a diverse synthetic and naturally-occurring chemicals, such as the halogenated aromatic hydrocarbons (HAHs) and the non-halogenated polycyclic aromatic hydrocarbons (PAHs). The liganded AHR modulates the genetic activity of a variety of xenobiotic-responsive genes, including cytochrome P4501A1 (CYP1A1). The tyrosinase inhibitor 1-phenyl-2-thiourea (PTU) is widely used in zebrafish research to suppress pigmentation in developing embryos/fry. Here we showed that 0.2 mM PTU induced a basal level of CYP1A1 transcription in zebrafish embryonic integument as early as 24 h postfertilization (hpf) stage. Subsequently, PTU induced CYP1A1 transcription in blood vessels at 36 hpf. During larval stage, the liver and all pharyngeal arch vessels of PTU-treated embryos exhibited CYP1A1 transcription as well. Comparing to TCDD, PTU induces CYP1A1 transcription with much lower efficacy in zebrafish embryos. Coincubating the embryos with PTU and TCDD led to repressing TCDD-induced CYP1A1 transcription. Mechanistic studies indicated that both of PTU- and TCDD-mediated CYP1A1 transcriptions are modulated by the same AHR-ARNT signaling pathway. PMID: 15183118 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 59: Mar Environ Res. 2004 Aug-Dec;58(2-5):119-24. Cloning and analysis of the CYP1A promoter from the atlantic killifish (Fundulus heteroclitus). Powell WH, Morrison HG, Weil EJ, Karchner SI, Sogin ML, Stegeman JJ, Hahn ME. Marine Biological Laboratory, Woods Hole, MA, USA. powellw@kenyon.edu Enzymes in the cytochrome P450 gene family 1 (CYP1) catalyze the metabolic activation of numerous hydrocarbon carcinogens and various natural compounds. CYP1 family members have been identified in several vertebrates, including fish, amphibians, birds, and mammals, and are inducible by aromatic hydrocarbons acting through the aryl hydrocarbon receptor (AHR). Together with its heterodimeric partner ARNT, the ligand-bound AHR binds conserved xenobiotic response elements (XREs) near the promoter of CYP1A and other genes. However, some populations of the Atlantic killifish Fundulus heteroclitus inhabiting highly contaminated sites are refractory to CYP1A induction by aromatic hydrocarbons. To better understand the mechanisms underlying this phenomenon, we are characterizing the AHR-CYP1A signaling pathway in this species. We report here the characterization of a genomic clone containing the 5(') end of the wild-type F. heteroclitus CYP1A gene. The 5(') coding sequence matches that of the F. heteroclitus CYP1A cDNA reported earlier [Comp. Biochem. Physiol. 121C (1998) 231]. Consistent with its inducibility by AHR agonists, the CYP1A gene contains three consensus XREs (5(')CACGC3(')) within 1.6 kb of the putative transcriptional start site. When oligonucleotides containing each of these sites were analyzed in an electrophoretic mobility shift assay, one of these showed a strong, TCDD-inducible mobility shift in the presence of in vitro expressed mouse AHR protein. These sequence data and initial functional characterization provide a valuable tool for the study of genetic variations in CYP1A expression and activity in sensitive and resistant populations. These studies may ultimately shed light on the importance of P4501A activity in xenobiotic toxicity. PMID: 15178023 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 60: Aquat Toxicol. 2004 Jul 14;68(4):301-13. Baltic salmon (Salmo salar) yolk-sac fry mortality is associated with disturbances in the function of hypoxia-inducible transcription factor (HIF-1alpha) and consecutive gene expression. Vuori KA, Soitamo A, Vuorinen PJ, Nikinmaa M. Laboratory of Animal Physiology, Department of Biology, University of Turku, FI-20014 Turku, Finland. Baltic salmon (Salmo salar) suffer from abnormally high yolk-sac fry mortality designated as M74-syndrome. In 1990s, 25-80% of salmon females, which ascended rivers to spawn, produced yolk-sac fry suffering from the syndrome. Symptoms of M74-affected fry include neurological disturbances, impaired vascular development and abnormal haemorrhages. The latter symptoms are observed in mammalian embryos if the function of hypoxia inducible transcription factor (HIF-1alpha), its dimerization partner aryl hydrocarbon nuclear translocator (ARNT) or target gene vascular endothelial growth factor (VEGF) is disturbed. To study the possible involvement of HIF-1alpha and its target gene VEGF in the development of the syndrome, we collected healthy and M74-affected wild Baltic salmon yolk-sac fry and analyzed HIF-1alpha mRNA and protein expression, HIF-1alpha DNA-binding, target gene VEGF protein expression, and blood vessel density in both groups at different stages of yolk-sac fry development. In addition, since Baltic salmon females contain organochlorine contaminants, which have been suggested to be the cause of M74 syndrome via the aryl hydrocarbon receptor (AhR)-dependent gene expression pathway, we studied AhR protein expression, AhR DNA-binding and target gene CYP1A protein expression. Since the parents of both healthy and M74-affected wild fry will have experienced the organochlorine load from the Baltic Sea, hatchery-reared fry were included in the studies as an additional control. The results show that the vascular defects observed in fry suffering from M74 are associated with reduced DNA-binding activity of HIF-1alpha and subsequent downregulation of its target gene vascular endothelial growth factor (VEGF). In addition, also AhR function is decreased in diseased fry making it unlikely that symptoms of M74-affected fry would be caused by an upregulation of xenobiotically induced AhR-dependent gene expression pathway. Copyright 2004 Elsevier B.V. PMID: 15177948 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 61: Aquat Toxicol. 2004 Jun 24;68(3):219-32. Unprecedented genomic diversity of AhR1 and AhR2 genes in Atlantic salmon (Salmo salar L.). Hansson MC, Wittzell H, Persson K, von Schantz T. Department of Animal Ecology, Lund University, Ecology Building, SE-22362 Lund, Sweden. maria.hansson@zooekol.lu.se Aryl hydrocarbon receptor (AhR) genes encode proteins involved in mediating the toxic responses induced by several environmental pollutants. Here, we describe the identification of the first two AhR1 (alpha and beta) genes and two additional AhR2 (alpha and beta) genes in the tetraploid species Atlantic salmon (Salmo salar L.) from a cosmid library screening. Cosmid clones containing genomic salmon AhR sequences were isolated using a cDNA clone containing the coding region of the Atlantic salmon AhR2gamma as a probe. Screening revealed 14 positive clones, from which four were chosen for further analyses. One of the cosmids contained genomic AhR sequences that were highly similar to the rainbow trout (Oncorhynchus mykiss) AhR2alpha and beta genes. SMART RACE amplified two complete, highly similar but not identical AhR type 2 sequences from salmon cDNA, which from phylogenetic analyses were determined as the rainbow trout AhR2alpha and beta orthologs. The salmon AhR2alpha and beta encode proteins of 1071 and 1058 residues, respectively, and encompass characteristic AhR sequence elements like a basic-helix-loop-helix (bHLH) and two PER-ARNT-SIM (PAS) domains. Both genes are transcribed in liver, spleen and muscle tissues of adult salmon. A second cosmid contained partial sequences, which were identical to the previously characterized AhR2gamma gene. The last two cosmids contained partial genomic AhR sequences, which were more similar to other AhR type 1 fish genes than the four characterized salmon AhR2 genes. However, attempts to amplify the corresponding complete cDNA sequences of the inserts proved very difficult, suggesting that these genes are non-functional or very weakly transcribed in the examined tissues. Phylogenetic analyses of the conserved regions did, however, clearly indicate that these two AhRs belong to the AhR type 1 clade and have been assigned as the Atlantic salmon AhR1alpha and AhR1beta genes. Taken together, these findings demonstrate that multiple AhR genes are present in Atlantic salmon genome, which likely is a consequence of previous genome duplications in the evolutionary past of salmonids. Plausible explanations for the high incidence of AhR genes in fish and more specifically in salmonids, like rapid divergences in specialized functions, are discussed. PMID: 15159049 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 62: Biochem Biophys Res Commun. 2004 Jun 4;318(3):746-55. A novel induction mechanism of the rat CYP1A2 gene mediated by Ah receptor-Arnt heterodimer. Sogawa K, Numayama-Tsuruta K, Takahashi T, Matsushita N, Miura C, Nikawa J, Gotoh O, Kikuchi Y, Fujii-Kuriyama Y. Department of Biomolecular Science, Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan. sogawa@mail.tains.tohoku.ac.jp We have identified an enhancer responsible for induction by 3-methylcholanthrene in the upstream region of the CYP1A2 gene. The enhancer does not contain the invariant core sequence of XREs that are binding sites for the Ah receptor (AhR) and Arnt heterodimer. The enhancer did not show any inducible expression in Hepa-1-derived cell lines, C4 and C12, deficient of Arnt and AhR, respectively. On the other hand, bacterially expressed AhR-Arnt heterodimer could not bind to the enhancer. Mutational analysis of the enhancer revealed that a repeated sequence separated by six nucleotides is important for expression. A factor binding specifically to the enhancer was found by using gel shift assays. Bacterially expressed AhR-Arnt heterodimer interacted with the factor. A dominant negative mutant of the AhR to XRE activated the enhancer. Collectively, these results demonstrate that a novel induction mechanism is present in which the AhR-Arnt heterodimer functions as a coactivator. PMID: 15144902 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 63: Biol Chem. 2004 Mar-Apr;385(3-4):291-4. Simultaneous exposure of rats to dioxin and carbon monoxide reduces the xenobiotic but not the hypoxic response. Hofer T, Pohjanvirta R, Spielmann P, Viluksela M, Buchmann DP, Wenger RH, Gassmann M. Institute of Veterinary Physiology, University of Zurich, CH-8057 Zurich, Switzerland. Aryl hydrocarbon receptor (AhR) and hypoxia-inducible factor-1alpha (HIF-1alpha) are conditionally regulated transcription factor subunits that form heterodimeric complexes with their common partner, AhR nuclear translocator (ARNT/HIF-1beta). Whereas the environmentally toxic compound 2,3,7,8-tetra-chlorodibenzo-p-dioxin (TCDD) initiates the trans-activation activity of AhR:ARNT/HIF-1beta, hypoxic exposure stabilizes HIF-1alpha and functionally activates the HIF-1alpha:ARNT/HIF-1beta complex. To analyze a possible crosstalk between these two pathways in vivo, rats were given dioxin orally and/or were exposed to carbon monoxide (CO), causing functional anemia. We found that exposure to CO inhibited the xenobiotic response while dioxin application had no significant negative impact on hypoxia-mediated gene transcription. PMID: 15134343 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 64: Med Sci Monit. 2004 May;10(5):BR135-8. Epub 2004 Apr 28. Expression of aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) mRNA expression in human spermatozoa. Khorram O, Garthwaite M, Jones J, Golos T. Department of Obstetrics and Gynecology, University of Wisconsin, and Wisconsin Regional Primate Research Center, Madison, WI, USA. okhorram@obgyn.humc.edu BACKGROUND: The objective of this study was to determine if human spermatozoa express AHR (dioxin receptor) and ARNT and if mRNA levels for these transcription factors correlate with sperm concentration, motility or morphology. MATERIAL/METHODS: This was a case-controlled cohort study in which AHR and ARNT mRNA levels in subjects with normal and abnormal semen analysis were compared. Semen analysis was performed by CASA. AHR/ARNT mRNA levels were determined by multiplex RT-PCR using Glyceryl aldehyde 3-phospho dehydrogenase (G3PDH) as an internal control. RESULTS: Human sperm expresses abundant amounts of AHR and ARNT mRNA. There were no differences in AHR and ARNT mRNA levels in subjects with normal and abnormal semen analysis. AHR and ARNT levels did not correlate with sperm concentration, morphology or motility. There was a trend (p=0.07) for lower AHR mRNA expression in sperm of men with severe oligospermia. There was a significant negative correlation (r=-0.42, p<0.05) between sperm AHR and ARNT mRNA. CONCLUSIONS: The presence of AHR in sperm provides a mechanism by which environmental dioxins, polycyclic aromatic hydrocarbons and polyhalogenated biphenyls could directly influence sperm function. PMID: 15114261 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 65: Toxicol Lett. 2004 Apr 1;149(1-3):287-93. Aryl-hydrocarbon receptor-dependent pathway and toxic effects of TCDD in humans: a population-based study in Seveso, Italy. Baccarelli A, Pesatori AC, Masten SA, Patterson DG Jr, Needham LL, Mocarelli P, Caporaso NE, Consonni D, Grassman JA, Bertazzi PA, Landi MT. Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD 20892-7236, USA. baccarea@mail.nih.gov Approximately 20 years after the Seveso, Italy accident, we conducted a population-based study to evaluate the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure upon immune and mechanistically based biomarkers of dioxin response in humans. TCDD toxic effects are known to be mediated by the aryl-hydrocarbon receptor (AhR). We randomly selected 62 study subjects from the highest exposed zones and 59 from the surrounding non-contaminated area. Current lipid-adjusted plasma TCDD concentrations in these subjects ranged from 3.5 to 90 ng/kg (or ppt) and were negatively associated with plasma IgG concentrations (r=-0.35; P = 0.0002). The expression of genes in the AhR-dependent pathway, including AhR, aryl-hydrocarbon receptor nuclear translocator (ARNT), CYP1A1, and CYP1B1 transcripts, and the CYP1A1-associated 7-ethoxyresorufin-O-deethylase (EROD) activity was measured in lymphocytes. AhR mRNA levels in uncultured lymphocytes were negatively associated with plasma TCDD (P=0.03). When mitogen-induced lymphocytes were cultured with 10nM TCDD, all AhR-dependent genes were induced 1.2- to 13-fold. In these cells, plasma TCDD was associated with decreased EROD activity. Markers within the AhR pathway were correlated with one another. Our findings suggest the presence of long-term effects in the subjects exposed to TCDD after the Seveso accident. PMID: 15093275 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 66: Int J Cancer. 2004 May 10;109(6):810-6. Inhibition of MUC1 expression by indole-3-carbinol. Lee IJ, Han F, Baek J, Hisatsune A, Kim KC. Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA. MUC1 is a large transmembrane glycoprotein overexpressed by a majority of carcinomas. High expression of MUC1 is associated with aggressive tumors, and MUC1 antigen is used as a marker to monitor disease progression in breast cancer patients. Several lines of evidence strongly suggest that the overexpression of MUC1 contributes to cancer progression and metastasis. In this report, we demonstrate that the naturally occurring cancer preventative, indole-3-carbinol (I3C), inhibits the expression of MUC1 in breast cancer cells. I3C inhibited both MUC1 mRNA and protein levels in a dose- and time-dependent manner. This inhibition was seen in the estrogen responsive MCF-7 cells as well as unresponsive MDA-MB-468 cells, indicating that the inhibitory pathway is independent of estrogen receptor. Gene expression studies using the human MUC1 gene promoter connected to a luciferase reporter demonstrated that I3C inhibits the transcription of the MUC1 gene. Promoter deletion studies indicate that the region containing up to 600 bp upstream (-600) of the initiation site is sufficient for inhibition by I3C. Furthermore, I3C represses the activation of transcription mediated by the region between -600 and -450 bp. A putative xenobiotic response element was located within this region but the binding of AhR/Arnt heterodimer to this site was undetectable by electrophoretic mobility shift assays. Our results may point to the existence of a novel pathway of transcriptional inhibition by I3C in cancer cells as well as a new mechanism of MUC1 gene inhibition. Our findings might have implications in the use of I3C as a preventative as well as a therapeutic agent for breast cancer. Copyright 2004 Wiley-Liss, Inc. PMID: 15027113 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 67: PLoS Biol. 2004 Mar;2(3):E65. Epub 2004 Mar 16. Interaction networks in yeast define and enumerate the signaling steps of the vertebrate aryl hydrocarbon receptor. Yao G, Craven M, Drinkwater N, Bradfield CA. McArdle Laboratory for Cancer Research, University of Wisconsin Medical School, Madison, Wisconsin, USA. The aryl hydrocarbon receptor (AHR) is a vertebrate protein that mediates the toxic and adaptive responses to dioxins and related environmental pollutants. In an effort to better understand the details of this signal transduction pathway, we employed the yeast S. cerevisiae as a model system. Through the use of arrayed yeast strains harboring ordered deletions of open reading frames, we determined that 54 out of the 4,507 yeast genes examined significantly influence AHR signal transduction. In an effort to describe the relationship between these modifying genes, we constructed a network map based upon their known protein and genetic interactions. Monte Carlo simulations demonstrated that this network represented a description of AHR signaling that was distinct from those generated by random chance. The network map was then explored with a number of computational and experimental annotations. These analyses revealed that the AHR signaling pathway is defined by at least five distinct signaling steps that are regulated by functional modules of interacting modifiers. These modules can be described as mediating receptor folding, nuclear translocation, transcriptional activation, receptor level, and a previously undescribed nuclear step related to the receptor's Per-Arnt-Sim domain. PMID: 15024417 [PubMed - in process] --------------------------------------------------------------- 68: J Exp Zoolog B Mol Dev Evol. 2003 Dec 15;300(1):48-57. ARNT gene multiplicity in amphibians: characterization of ARNT2 from the frog Xenopus laevis. Rowatt AJ, DePowell JJ, Powell WH. Kenyon College, Gambier, Ohio 43022, USA. The aryl hydrocarbon receptor nuclear translocator (ARNT) is a member of the Per-ARNT-Sim (PAS) protein superfamily, transcription factors that mediate the cellular responses to various developmental signals and environmental conditions. A beta-class ("partner") PAS protein, ARNT exhibits the capacity to form transcriptionally active heterodimers with several alpha-class ("sensor") proteins, including the aryl hydrocarbon receptors (AHRs), the hypoxia-inducible factors (HIFs), and the Single minded (Sim) proteins. Two genes encode different forms of ARNT in mammals: ARNT1, which is widely expressed, and ARNT2, which is limited to the brain and kidneys of adults and specific neural and branchial tissues of embryos. In contrast, fish apparently express only a single ARNT gene, although in different species, this may be either ARNT1 or ARNT2. In efforts to understand the evolution of ARNT proteins throughout the vertebrate lineage, we isolated an ARNT cDNA from early life stages of the amphibian Xenopus laevis. The encoded protein binds cognate DNA sequences in concert with mouse AHR. Phylogenetic analysis reveals that this sequence is orthologous to mammalian ARNT2 and paralogous to the recently reported X. laevis ARNT1. ARNT2 mRNA expression begins later than ARNT1 (stage 22 vs. stage 8), suggesting the two proteins play distinct roles during development. Hence, in the expression of two well-conserved ARNT paralogs with distinct expression patterns, X. laevis resembles mammals rather than fish. Diversity in the number and function of PAS proteins, including ARNT, may underlie significant species differences in developmental programming and biochemical response to environmental conditions. The identification of multiple amphibian ARNT paralogs represents an important step in the understanding of evolution and functional variation of ARNT in vertebrates. PMID: 14984034 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 69: J Biol Chem. 2004 May 14;279(20):20582-93. Epub 2004 Feb 20. The aryl hydrocarbon receptor (AhR) tyrosine 9, a residue that is essential for AhR DNA binding activity, is not a phosphoresidue but augments AhR phosphorylation. Minsavage GD, Park SK, Gasiewicz TA. Department of Environmental Medicine, School of Medicine, University of Rochester, 575 Elmwood Avenue, Rochester, NY 14642, USA. We delineate a mechanism by which dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin or TCDD)-mediated formation of the aryl hydrocarbon receptor (AhR) DNA binding complex is disrupted by a single mutation at the conserved AhR tyrosine 9. Replacement of tyrosine 9 with the structurally conservative phenylalanine (AhRY9F) abolished binding to dioxin response element (DRE) D, E, and A and abrogated DRE-driven gene induction mediated by the AhR with no effect on TCDD binding, TCDD-induced nuclear localization, or ARNT heterodimerization. The speculated role for phosphorylation at tyrosine 9 was also examined. Anti-phosphotyrosine immunoblotting could not detect a major difference between the AhRY9F mutant and wild-type AhR, but a basic isoelectric point shift was detected by two-dimensional gel electrophoresis of AhRY9F. However, an antibody raised to recognize only phosphorylated tyrosine 9 (anti-AhRpY9) confirmed that AhR tyrosine 9 is not a phosphorylated residue required for DRE binding. Kinase assays using synthetic peptides corresponding to the wild-type and mutant AhR residues 1-23 demonstrated that a tyrosine at position 9 is important for substrate recognition at serine(s)/threonine(s) within this sequence by purified protein kinase C (PKC). Also, compared with AhRY9F, immunopurified full-length wild-type receptor was more rapidly phosphorylated by PKC. Furthermore, co-treatment of AhR-deficient cells that expressed AhRY9F and a DRE-driven luciferase construct with phorbol 12-myristate 13-acetate and TCDD resulted in a 30% increase in luciferase activity compared with AhRY9F treated with TCDD alone. Overall, AhR tyrosine 9, which is not a phosphorylated residue itself but is required for DNA binding, appears to play a crucial role in AhR activity by permitting proper phosphorylation of the AhR. PMID: 14978034 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 70: J Mammary Gland Biol Neoplasia. 2000 Jul;5(3):295-306. Mechanisms of inhibitory aryl hydrocarbon receptor-estrogen receptor crosstalk in human breast cancer cells. Safe S, Wormke M, Samudio I. Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466, USA. ssafe@cvm.tamu.edu The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that forms a functional heterodimeric complex with the AhR nuclear translocator (Arnt) protein. The environmental toxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is a high affinity ligand for the AhR and has been extensively used to investigate AhR-mediated biochemical and toxic responses. TCDD modulates several endocrine pathways including inhibition of 17beta-estradiol-induced responses in the immature and ovariectomized rodent uterus and mammary gland and in human breast cancer cell lines. TCDD inhibits formation and growth of mammary tumors in carcinogen-induced rodent models and relatively nontoxic selective AhR modulators (SAhRMs) are being developed for treatment of breast cancer. The mechanisms of inhibitory AhR-estrogen receptor (ER) crosstalk have been investigated in MCF-7 breast cancer cells by analysis of promoter regions of genes induced by E2 and inhibited by TCDD. AhR-mediated inhibition of E2-induced cathepsin D, pS2, c-fos, and heat shock protein 27 gene expression involves direct interaction of the AhR complex with inhibitory pentanucleotide (GCGTG) dioxin responsive elements (iDREs) resulting in disruption of interactions between proteins binding DNA elements required for ER action and the basal transcription machinery. Mechanisms of inhibitory AhR-ER crosstalk indicate that functional iDREs are required for inhibition of some genes; however, results indicate that other interaction pathways are important including AhR-mediated proteasome-dependent degradation of the ER. Publication Types: Review Review, Tutorial PMID: 14973392 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 71: J Biol Chem. 2004 Apr 16;279(16):16326-31. Epub 2004 Feb 5. Patent ductus venosus and dioxin resistance in mice harboring a hypomorphic Arnt allele. Walisser JA, Bunger MK, Glover E, Harstad EB, Bradfield CA. McArdle Laboratory for Cancer Research, University of Wisconsin Medical School, Madison, Wisconsin 53706, USA. The Ah receptor nuclear translocator (ARNT) is the dimeric partner of hypoxia-inducible factors and thus plays a pivotal role in cellular adaptation to low oxygen environments. ARNT is also a dimeric partner for the Ah receptor (AHR), and this complex is essential in regulating the adaptive metabolic response to polycyclic aromatic hydrocarbons. Because of the essential role of ARNT in hypoxia-driven developmental events, it has been difficult to study the physiological significance of AHR.ARNT heterodimers in vivo. To address this issue, we developed a hypomorphic Arnt allele that displayed normal development and allowed the examination of the role of ARNT in AHR biology. In this regard, the AHR is also known to mediate two additional biological processes: the toxicological response to compounds such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin) and the developmental closure of a fetal vascular structure known as the ductus venosus. Although the mechanism of the adaptive pathway has been well described, the mechanism of AHR-mediated signal transduction in the toxic and developmental pathways is not well understood. Liver perfusion studies demonstrated that ARNT hypomorphs have a patent ductus venosus, identical to that observed in the Ahr null mice. Parallel dioxin toxicity studies demonstrated that the ARNT hypomorphs exhibited resistance to the end points of dioxin exposure. Moreover, we observed that toxicity could be segregated from the classical adaptive responses such as P4501A induction. Taken in sum, these experiments demonstrate that ARNT is an essential component of AHR developmental signaling and shed light on the mechanism of dioxin toxicity. PMID: 14764592 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 72: Development. 2004 Feb;131(4):819-28. The AHR-1 aryl hydrocarbon receptor and its co-factor the AHA-1 aryl hydrocarbon receptor nuclear translocator specify GABAergic neuron cell fate in C. elegans. Huang X, Powell-Coffman JA, Jin Y. Department of Molecular, Cellular and Developmental Biology, Howard Hughes Medical Institute, University of California, Santa Cruz, CA 95064, USA. The aryl hydrocarbon receptors (AHR) are bHLH-PAS domain containing transcription factors. In mammals, they mediate responses to environmental toxins such as 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD). Such functions of AHRs require a cofactor, the aryl hydrocarbon receptor nuclear translocator (ARNT), and the cytoplasmic chaperonins HSP90 and XAP2. AHR homologs have been identified throughout the animal kingdom. We report here that the C. elegans orthologs of AHR and ARNT, ahr-1 and aha-1, regulate GABAergic motor neuron fate specification. Four C. elegans neurons known as RMED, RMEV, RMEL and RMER express the neurotransmitter GABA and control head muscle movements. ahr-1 is expressed in RMEL and RMER neurons. Loss of function in ahr-1 causes RMEL and RMER neurons to adopt a RMED/RMEV-like fate, whereas the ectopic expression of ahr-1 in RMED and RMEV neurons can transform them into RMEL/RMER-like neurons. This function of ahr-1 requires aha-1, but not daf-21/hsp90. Our results demonstrate that C. elegans ahr-1 functions as a cell-type specific determinant. This study further supports the notion that the ancestral role of the AHR proteins is in regulating cellular differentiation in animal development. PMID: 14757639 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 73: Toxicol Sci. 2004 Mar;78(1):68-77. Epub 2003 Dec 22. Interactions between 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and hypoxia signaling pathways in zebrafish: hypoxia decreases responses to TCDD in zebrafish embryos. Prasch AL, Andreasen EA, Peterson RE, Heideman W. Molecular and Environmental Toxicology Center and dagger School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, USA. The aryl hydrocarbon receptor (AHR) interacts with the aryl hydrocarbon receptor nuclear translocator (ARNT) to form a heterodimer that binds to promoters in target genes to alter transcription in response to xenobiotics such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The ARNT protein also forms heterodimers with other proteins such as HIF-1alpha and HIF-2alpha to alter gene expression in response to low oxygen conditions. Because ARNT is shared between multiple signaling pathways it is possible that activation of one ARNT-requiring pathway could inhibit the activation of other pathways that depend on ARNT. One hypothesis to explain TCDD toxicity in early life stage fish is that TCDD activation of zfAHR2 sequesters zfARNT2 away from the hypoxia signaling pathway. To test this hypothesis we measured the ability of TCDD to prevent induction of heme oxygenase by hypoxia (40% saturation), as well as the ability of hypoxia to increase the sensitivity of zebrafish to the effects of TCDD during the first week of life. As a further test of the model we examined mutant zebrafish that lack zfARNT2 for phenotypes that resemble the effects of TCDD exposure. Our results demonstrate that sequestration of zfARNT2 is not causing TCDD toxicity. TCDD did not inhibit hypoxia induction of heme oxygenase, hypoxia and TCDD exposures were not additive in causing developmental toxicity, and mutant embryos that lack zfARNT2 do not develop defects mimicking TCDD toxicity. However, our results demonstrate some level of cross talk between the two pathways in the zebrafish embryo. Hypoxia decreased TCDD induction of zfCYP1A mRNA, and decreased the potency of TCDD in causing edema. It is not clear whether this is mediated through competition for zfARNT2, or through other mechanisms. PMID: 14691204 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 74: Life Sci. 2004 Jan 9;74(8):1039-49. Characteristic expression of aryl hydrocarbon receptor repressor gene in human tissues: organ-specific distribution and variable induction patterns in mononuclear cells. Yamamoto J, Ihara K, Nakayama H, Hikino S, Satoh K, Kubo N, Iida T, Fujii Y, Hara T. Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka 812-8582, Japan. jyamamoto@dr.hosp.kyushu-u.ac.jp To investigate the expression of aryl hydrocarbon receptor repressor (AhRR) and related molecules in various tissues and the effects of aromatic hydrocarbons (AHs) on their expression, we developed a reliable technique of quantification of human AhRR as well as aryl hydrocarbon receptor (AhR), AhR nuclear translocator (ARNT) and cytochrome P450 1A1 (CYP1A1) mRNA by real-time TaqMan PCR method. First, we examined the expression of these genes in human adult or fetal tissues. The levels of AhRR expression were extremely high in testis, very high in lung, ovary, spleen and pancreas from adults, whereas those were low in those from fetuses. On the other hand, CYP1A1 expression was extremely high in lung, and AhR and ARNT were ubiquitously expressed in almost all tissues. Second, we compared the expression levels of these genes in mononuclear cells (MNCs) from various sources. Comparison of the basal expression levels of these genes in MNCs demonstrated that MNCs from umbilical cord blood showed higher AhRR or CYP1A1 expression than those from adults. The induction of AhRR or CYP1A1 expression by 3-methylcholanthrene (3-MC) was observed in MNCs from adults but not from umbilical cord blood. Consequently, there existed characteristic differences in the basal levels of AhRR and CYP1A1 expression in MNCs, as well as in their inducibility by 3-MC among MNCs from various types of human bloods. These results will provide basic information for a possible application of AhRR and CYP1A1 measurements to evaluate AH exposure in vivo. PMID: 14672759 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 75: J Biol Chem. 2004 Mar 5;279(10):9306-12. Epub 2003 Dec 1. Regulatory interaction between arylhydrocarbon receptor and SIM1, two basic helix-loop-helix PAS proteins involved in the control of food intake. Yang C, Boucher F, Tremblay A, Michaud JL. Research Center, Hopital Sainte-Justine, 3175 Cote Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada. The basic helix-loop-helix PAS (bHLH-PAS) transcription factors SIM1 and arylhydrocarbon receptor (AHR) are involved in the control of feeding behavior. Sim1 haploinsufficiency causes hyperphagia in mice and humans, most likely by perturbing the hypothalamus function. The administration of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a ligand of AHR, causes severe anorexia, which also appears to be of central origin. Both SIM1 and AHR require heterodimerization either with ARNT or ARNT2 to function. Here, we characterize the promoter for Sim1 and show that a consensus AHR-ARNT/2 binding site positively regulates its activity in the context of transfection experiments in Neuro-2A cells. A gel shift assay indicated that AHR-ARNT/2 can bind its putative site in the Sim1 promoter. Overexpression of Arnt, Arnt2, or Ahr increased the activity of a reporter construct containing the Sim1 promoter by 1.8-, 1.5-, and 2.2-fold, respectively, but failed to do so when the AHR-ARNT/2 binding site was mutated. Similarly, TCDD increased the activity of the reporter construct by 1.8-fold but not that of its mutated version. Finally, we found that TCDD increased Sim1 expression in Neuro-2A cells and in mouse kidney and hypothalamus by 4-, 3-, and 2-fold, respectively. We conclude that Sim1 expression is regulated by AHR-ARNT/2. This result raises the possibility that Sim1 mediates the effect of TCDD on feeding and points to a complex network of regulatory interactions between bHLH-PAS proteins. PMID: 14660629 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 76: Toxicol Sci. 2004 Jan;77(1):109-16. Epub 2003 Dec 3. Role of aryl hydrocarbon receptor in mesencephalic circulation failure and apoptosis in zebrafish embryos exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Dong W, Teraoka H, Tsujimoto Y, Stegeman JJ, Hiraga T. Department of Toxicology, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent and potent developmental toxicant in various animals, with developing fish being the most sensitive organisms. Although the expression of aryl hydrocarbon receptor (AHR) as well as the partner molecule, AHR nuclear translocator (ARNT) in the brain has been reported, the effect of TCDD on the brain remains to be clarified in detail. Previously, we reported local circulation failure and apoptosis in dorsal midbrain caused by TCDD in developing zebrafish. In the present experiments, we investigated the effects of morpholino antisense oligos against aryl hydrocarbon receptor 2 (zfAHR2) (AHR2-MO) on toxicological endpoints caused by TCDD in developing zebrafish. AHR2-MO but not its negative homologue (4mis-AHR2-MO) improved TCDD-evoked circulation failure in mesencephalic vein and reduced the occurrence of apoptosis in dorsal midbrain, with concomitant inhibition of CYP1A induction in vascular endothelium. Injection of bovine serum albumin (BSA) into the general circulation, followed by immunohistochemistry with anti-BSA, showed that TCDD raised vascular permeability to albumin in dorsal midbrain, which was blocked by AHR2-MO and N-acetlycystein. In the absence of TCDD, development of embryos injected with AHR2-MO appeared normal at least until 60 h after fertilization. It is concluded that AHR2 activation in the vascular endothelium of the zebrafish embryo midbrain is involved in the mesencephalic circulation failure and apoptosis elicited by TCDD. This is the further evidence that vascular endothelium is the target of TCDD in relation to local circulation failure and apoptosis in dorsal midbrain. PMID: 14657521 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 77: Exp Mol Med. 2003 Oct 31;35(5):393-402. Genomic organization and expression of parkin in Drosophila melanogaster. Bae YJ, Park KS, Kang SJ. Department of Biological Science, College of Natural Science, Ewha Womans University, Seoul 120-750, Korea. We report here the isolation, characterization on genomic structure and expression of the D. melanogaster homolog of human parkin. The 2,122 bp parkin gene sequence contains six exons that form a 1,449 bp transcript encoding a protein of 482 amino acids. 151 bp of 5' and 112 bp of 3' untranslated regions were identified by a combination of 5'-RACE/primer extension and 3'-RACE, respectively. The 5' UTR contains three transcription initiation sites. Neither a classical TATA nor a CAAT box was found in the putative promoter sequence. However, binding sites for AhR-Arnt, AP4, NF1 and GATA transcription factors were identified. Transient transfection analysis of the 5' UTR confirmed its promoter activity in HEK 293 cells and SH-SY5Y neuronal cells using a dual luciferase reporting system. The amino acid sequence of D. melanogaster Parkin exhibits 42%, 43% and 43% identity to that of human, mouse and rat, respectively, representing a 54 kDa protein band via western blot analysis. It shows a high degree of conservation in the Ubiquitin-like domain at the N-terminus (34%), the In-Between RING finger domains (IBR, 65-69%), and the RING finger domains at the C-terminus (56-57%). The expression pattern of D. melanogaster parkin varies during the developmental stages, with the highest expression in the adult stage as measured by competitive RT-PCR. From immunostainings of the embryo, D. melanogaster parkin was expressed slightly higher in the central nervous system (brain and nerve cord) during the late embryonic stage. PMID: 14646593 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 78: J Clin Invest. 2003 Nov;112(9):1372-82. Alteration of the 4-sphingenine scaffolds of ceramides in keratinocyte-specific Arnt-deficient mice affects skin barrier function. Takagi S, Tojo H, Tomita S, Sano S, Itami S, Hara M, Inoue S, Horie K, Kondoh G, Hosokawa K, Gonzalez FJ, Takeda J. Department of Social and Environmental Medicine H3, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan. takeda@mr-envi.med.osaka-u.ac.jp Aryl hydrocarbon receptor nuclear translocator (ARNT), a transcription factor of the Per/AHR/ARNT/Sim family, regulates gene expression in response to environmental stimuli including xenobiotics and hypoxia. To examine its role in the epidermis, the Cre-loxP system was used to disrupt the Arnt gene in a keratinocyte-specific manner. Gene-targeted, newborn mice with almost normal appearance died neonatally of severe dehydration caused by water loss. Histology showed small changes in the architecture of cornified layers, with apparently preserved intercorneocyte lamellar structures responsible for the skin barrier function. In contrast, HPLC/ion-trap mass spectrometry revealed significant alterations in the compositions of ceramides, the major components of the lamellae. The murine epidermal ceramides normally contain 4-sphingenine and 4-hydroxysphinganine. In Arnt-null epidermis, 4-sphingenine was largely replaced by sphinganine and the amounts of ceramides with 4-hydroxysphinganine were greatly decreased, suggesting deficiency of dihydroceramide desaturases that catalyze the formation of both 4-sphingenyl and 4-hydroxysphinganyl moieties. A desaturase isoenzyme, DES-1, prefers desaturation, but DES-2 catalyzes both reactions to a similar extent. Transcript levels of Des-2, but not Des-1, were considerably decreased in cultured keratinocytes from Arnt-null epidermis. These results indicate that proper ceramide compositions through 4-desaturation regulated by ARNT are crucial for maintaining the epidermal barrier function. PMID: 14597763 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 79: Environ Toxicol Chem. 2003 Oct;22(10):2337-43. Expression and inducibility of aryl hydrocarbon receptor pathway genes in wild-caught killifish (Fundulus heteroclitus) with different contaminant-exposure histories. Meyer JN, Wassenberg DM, Karchner SI, Hahn ME, Di Giulio RT. Nicholas School of the Environment and Earth Sciences and Integrated Toxicology Program, Duke University, Durham, North Carolina 27708-0328, USA. Wildcaught killifish from a contaminated site on the Elizabeth River (VA, USA) are refractory to induction of cytochrome P4501A (CYP1A, measured as catalytic activity and immunodetectable CYP1A protein) after exposure to typical aryl hydrocarbon receptor (AHR) agonists, as has been reported for fish from other sites highly contaminated with these compounds. In an attempt to understand the molecular basis for the lack of inducibility of CYP1A protein expression and activity in Elizabeth River killifish, we analyzed the expression of CYP1A and four other members of the AHR signal transduction pathway: AHR1, AHR2, AHR repressor (AHRR), and AHR nuclear translocator (ARNT). Gene expression was measured by cycle-optimized reverse transcriptase-polymerase chain reaction (RT-PCR) analysis of messenger ribonucleic acid (mRNA) extracted from livers of killifish from the Elizabeth River and King's Creek (VA, USA) (reference site), 36 h after injection with beta-naphthoflavone (BNF, an AHR agonist) or corn oil (carrier control). Hepatic CYP1A mRNA was inducible in King's Creek killifish. However, in Elizabeth River killifish, no induction of CYP1A mRNA was observed, confirming and extending previous results showing no induction of CYP1A protein or catalytic activity in this population. Similarly, AHRR and AHR2 mRNA levels were induced by BNF in King's Creek but not Elizabeth River killifish. No population or treatment-related differences were observed in expression of AHR1 or ARNT mRNAs. The results reveal in Elizabeth River killifish a consistent lack of inducibility of genes that are normally inducible by AHR agonists (CYP1A, AHRR, AHR2). However, the expression of AHR1, AHR2, and AHRR in vehicle-treated fish did not differ between Elizabeth River and King's Creek killifish, suggesting that altered constitutive expression of AHRs or AHRR does not underlie the refractory CYP1A phenotype in Elizabeth River killifish. PMID: 14551997 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 80: Toxicol Appl Pharmacol. 2003 Oct 15;192(2):174-90. Ah receptor regulation of mouse Cyp1B1 is additionally modulated by a second novel complex that forms at two AhR response elements. Zhang L, Zheng W, Jefcoate CR. Department of Pharmacology, Medical Science Center, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA. Cyp1B1 is expressed constitutively in many extrahepatic cells and is induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). An enhancer region (AhER-810 to -1075 of the mouse Cyp1B1 promoter), which mediates aryl hydrocarbon receptor (AhR) regulation of transcription, contains three consensus XRE sequences (designated XRE1, XRE4, and XRE5) and a central Ebox. XRE5 is essential for both basal and induced activity in C3H10T1/2 cells. AhR/ARNT binding to XRE1, XRE4, and the Ebox complex function in combination to support the AhR/ARNT complex at XRE5. The identical 12 base cores of XRE1 and XRE4 differ from the core of XRE5 by two bases outside of the consensus XRE. These sites bind a constitutive complex slightly smaller than AhR/ARNT (anomalous complex; anC), which is not formed at XRE5 or six Cyp1A1 XREs. Exchange of these bases (m3 mutations) restores selective AhR/ARNT binding at XRE1/XRE4 and introduces anC binding at XRE5. The activities of multimeric XRE1 and XRE5 luciferase reporters responded in parallel to the extent of AhR/ARNT binding. The consensus anC binding sequence ((C/T)GCG(C/T)GCGC(C/A)GC) overlaps the XRE1/XRE4 AhR/ARNT element. Gel mobility analyses show that anC binds to XRE1/XRE4 under basal conditions, while AhR/ARNT partially displaces anC following TCDD induction. Selective depletion of anC with biotin-oligonucleotides increases AhR/ARNT binding. M3-mutations at, respectively, XRE1 and XRE4 of the AhER sequence, had opposite effects on luciferase reporters. Activities increased for the XRE1 mutation and decreased for the XRE4 mutation, but also depended on the level of AhR transfected into AhR -/- fibroblasts. AnC compete with AhR at XRE1 while playing an activating role at XRE4. This positive effect of constitutive anC binding at XRE4 may contribute to the characteristic basal Cyp1B1 expression in embryo fibroblasts, which is mediated by low constitutive activities of AhR. PMID: 14550751 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 81: J Immunol. 2003 Oct 15;171(8):4113-20. T cell-specific disruption of arylhydrocarbon receptor nuclear translocator (Arnt) gene causes resistance to 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced thymic involution. Tomita S, Jiang HB, Ueno T, Takagi S, Tohi K, Maekawa S, Miyatake A, Furukawa A, Gonzalez FJ, Takeda J, Ichikawa Y, Takahama Y. Department of Biochemistry, Kagawa Medical University, Kagawa, Japan. tomita@genome.tokushima-u.ac.jp The arylhydrocarbon receptor nuclear translocator (ARNT) is a member of the basic helix-loop-helix, PER-ARNT-SIM family of heterodimeric transcription factors, and serves as a dimerization partner for arylhydrocarbon receptor (AHR) and hypoxia-inducible factor-1alpha. To assess the function of ARNT in T cells, we disrupted the Arnt gene specifically in T cells of mice by conditional gene targeting using T cell-specific p56(lck)-Cre (Lck-Cre) transgenic Arnt-floxed mice. Thus generated, T cell-specific Arnt-disrupted mice (Lck-Cre;Arnt(flox/Delta) transgenic mice) exhibited complete loss of the expression of ARNT protein only in T cells, and were viable and appeared normal. The Arnt-disrupted T cells in the thymus were phenotypically and histologically normal. The Arnt-deficient T cells in the spleen were capable of responding to TCR stimulation in vitro. However, unlike normal mice in which exposure to the environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an AHR ligand, resulted in thymic involution, the thymus of Lck-Cre;Arnt(flox/Delta) mice were resistant to TCDD treatment in vivo. In contrast, benzo(a)pyrene, another AHR ligand, still caused thymic involution in Lck-Cre;Arnt(flox/Delta) mice. Finally, fetal thymus organ culture using Lck-Cre;Arnt(flox/Delta) and K5-Cre;Arnt(flox/Delta) (epithelial cell-specific Arnt-disrupted mice) showed that thymocytes rather than thymic epithelial cells are predominantly responsible for TCDD-induced thymic atrophy. Our results indicate that ARNT in T lineage cells is essential for TCDD-mediated thymic involution. PMID: 14530333 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 82: J Mol Biol. 2003 Oct 17;333(2):249-60. Proteasome inhibition induces nuclear translocation of the dioxin receptor through an Sp1 and protein kinase C-dependent pathway. Santiago-Josefat B, Fernandez-Salguero PM. Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias, Universidad de Extremadura, Avenida de Elvas s/n, 06071 Badajoz, Spain. The dioxin receptor (AhR), in addition to its role in xenobiotic-induced carcinogenesis, appears to participate in cell proliferation, differentiation and organ homeostasis. Understanding potential mechanisms of activation of this receptor in the absence of exogenous ligands is therefore important to study its contribution to endogenous cellular functions. Using mouse embryo primary fibroblasts, we have previously shown that proteasome inhibition increased AhR transcriptional activity in the absence of xenobiotics. We suggested that proteasome inhibition-dependent AhR activation could involve an increase in the expression of the partner protein dioxin receptor nuclear translocator (ARNT). Since ARNT over-expression induced nuclear translocation of the AhR, and ARNT-deficient cells were unable to translocate this receptor to the nucleus upon proteasome inhibition, we have analyzed the effect of proteasome inhibition on the expression of regulatory proteins controlling ARNT levels. Treatment with the proteasome inhibitor MG132 increased endogenous Sp1 phosphorylation and its DNA-binding activity to the ARNT promoter. Sp1 phosphorylation and binding to the ARNT promoter, ARNT over-expression and AhR nuclear translocation were inhibited by GF109203X, a protein kinase C-specific inhibitor. In addition, MG132 stimulated protein kinase C activity in MEF cells with a pattern similar to that observed for ARNT expression. These data suggest that cellular control of protein kinase C activity, through Sp1 and ARNT, could regulate AhR transcriptional activity in the absence of xenobiotics. PMID: 14529614 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 83: Biochem J. 2004 Jan 1;377(Pt 1):205-13. Induction of murine NAD(P)H:quinone oxidoreductase by 2,3,7,8-tetrachlorodibenzo-p-dioxin requires the CNC (cap 'n' collar) basic leucine zipper transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2): cross-interaction between AhR (aryl hydrocarbon receptor) and Nrf2 signal transduction. Ma Q, Kinneer K, Bi Y, Chan JY, Kan YW. Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA. qam1@cdc.gov TCDD (2,3,7,8-tetrachlorodibenzo- p -dixoin) induces phase II drug-metabolizing enzyme NQO1 [NAD(P)H:quinone oxidoreductase; EC 1.6.99.2; DT-diaphorase] in a wide range of mammalian tissues and cells. Here, we analysed the molecular pathway mediating NQO1 induction by TCDD in mouse hepatoma cells. Inhibition of protein synthesis with CHX (cycloheximide) completely blocks induction of NQO1 by TCDD as well as the basal expression and induction by phenolic antioxidant tBHQ (2-t-butylbenzene-1,4-diol), implicating a labile factor in NQO1 mRNA expression. The inhibition is both time- and concentration-dependent, requires inhibition of protein synthesis, and occurs at a transcriptional level. Inhibition of NQO1 transcription by CHX correlates with a rapid reduction of the CNC bZip (cap 'n' collar basic leucine zipper) transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) through the 26 S proteasome pathway. Moreover, blocking Nrf2 degradation with proteasome inhibitor MG132 increases the amount of Nrf2 and superinduces NQO1 in the presence of TCDD or tBHQ. Finally, genetic experiments using AhR (aryl hydrocarbon receptor)-, Arnt (aryl hydrocarbon receptor nuclear translocator)- or Nrf2-deficient cells reveal that, while induction of NQO1 by TCDD depends on the presence of AhR and Arnt, the basal and inducible expression of NQO1 by either TCDD or tBHQ requires functional Nrf2. The findings demonstrate a novel role of Nrf2 in the induction of NQO1 by TCDD and provide new insights into the mechanism by which Nrf2 regulates the induction of phase II enzymes by both phenolic antioxidants and AhR ligands. PMID: 14510636 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 84: Mutat Res. 2003 Sep 9;540(1):99-105. Construction of reporter yeasts for mouse aryl hydrocarbon receptor ligand activity. Kawanishi M, Sakamoto M, Ito A, Kishi K, Yagi T. Division of Radiobiology and Environmental Science, Research Institute for Advanced Science and Technology, Osaka Prefecture University, 1-2 Gakuen-cho, Sakai 599-8570, Japan. Aryl hydrocarbons such as dioxins, polychlorinated biphenyls and polyaromatic hydrocarbons bind to the cellular aryl hydrocarbon receptor (AhR) in the initial step of their metabolism. The activation of intracellular signaling subsequent to the AhR binding is highly correlated with the toxicity and carcinogenicity of these chemicals. We produced Saccharomyces cerevisiae coexpressing mouse AhR and aryl hydrocarbon receptor nuclear translocator (Arnt) protein in accordance with Miller III's method for constructing yeasts with human Ahr and Arnt [Toxicol. Appl. Pharmacol. 160 (1998) 297]. Ligand treatment induced a dose-dependent increase in beta-galactosidase activity from a reporter plasmid in the yeast. Then, we compared activities of several ligands in yeast having the mouse Ahr/Arnt genes with those in yeast having the human genes, both of which have the same genetic background. There was no significant difference in the EC50 values of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), benzo[a]pyrene, 3-methylcholanthrene and beta-naphthoflavone between the mouse and human genes. However, indirubin, which was recently found in human urine as a potent AhR ligand [J. Biol. Chem. 276 (2001) 31475], had a 35-140 times higher EC50 value in the yeast with human genes than mouse genes. This difference might reflect species-specificity between mouse and human AhR/Arnt. PMID: 12972062 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 85: Biochem Pharmacol. 2003 Sep 1;66(5):841-7. Contribution of the Ah receptor to the phenolic antioxidant-mediated expression of human and rat UDP-glucuronosyltransferase UGT1A6 in Caco-2 and rat hepatoma 5L cells. Munzel PA, Schmohl S, Buckler F, Jaehrling J, Raschko FT, Kohle C, Bock KW. Department of Toxicology, Institute of Pharmacology and Toxicology, University of Tubingen, Tubingen D-72074, Germany. UDP-glucuronosyltransferases (UGTs) represent major phase II enzymes of drug metabolism which are regulated in a tissue-specific manner by endogenous and environmental factors. Among the latter, aryl hydrocarbon receptor (AhR) agonists such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and phenolic antioxidants such as tert-butylhydroquinone (tBHQ) are known to induce the expression of human UGT1A6 in Caco-2 cells. While binding of the TCDD-activated AhR to one xenobiotic response element (XRE) in the 5'-flanking regulatory region of UGT1A6 was characterised previously, the mechanism responsible for tBHQ induction is unknown. Therefore, it was investigated whether antioxidant response elements (AREs) are involved in tBHQ induction of UGT1A6. Transfectants of 3 kb of its regulatory region and its deletion mutants were treated with tBHQ. These studies suggested a region with approximately 2-fold induction, including an ARE-like motif, 15 bp downstream of the previously characterised XRE. Transfectants of the point-mutated ARE-like motif showed marginally reduced response to tBHQ, but surprisingly, loss of response to TCDD, suggesting interference of flanking proteins with the AhR/Arnt complex. Coordinate responses of UGT activity after treatment with TCDD or tBHQ were also observed in rat hepatoma 5L cells, mutants without the AhR and with recomplemented AhR. The results suggest a contribution of the AhR pathway and of proteins binding to the XRE flanking region to the induction of human UGT1A6 by both AhR agonists and phenolic antioxidants. PMID: 12948865 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 86: J Biochem (Tokyo). 2003 Jul;134(1):83-90. Heterodimers of bHLH-PAS protein fragments derived from AhR, AhRR, and Arnt prepared by co-expression in Escherichia coli: characterization of their DNA binding activity and preparation of a DNA complex. Kikuchi Y, Ohsawa S, Mimura J, Ema M, Takasaki C, Sogawa K, Fujii-Kuriyama Y. Department of Biomolecular Sciences, Graduate School of Life Science, Tohoku University, Sendai 980-8578. ykikuchi@mail.cc.tohoku.ac.jp AhR (aryl hydrocarbon receptor), AhRR (AhR repressor), and Arnt (AhR nuclear translocator) are members of the bHLH (basic-helix-loop-helix)-PAS (Per-AhR/Arnt-Sim homology sequence) transcription factor superfamily. They associate with each other to form heterodimers, AhR/Arnt or AhRR/Arnt, and bind the XRE (xenobiotic responsive element) sequences in the promoter regions of the target genes to regulate their expression. Their basic regions and HLH motifs mediate DNA binding activity and protein dimerization, respectively. The PAS domain includes two incomplete repeats, PAS-A and PAS-B, and is considered to determine the specificity on protein dimerization. However, the three-dimensional structures of PAS folds reported so far are all monomeric, therefore, little is known about the structural basis of protein interaction through PAS domains. In the present study, we prepared heterodimeric bHLH-PAS domains derived from AhR and Arnt, and AhRR and Arnt by co-expressing each pair in E. coli, and showed that the heterodimers formed exhibited full DNA-binding activity, which was not apparently affected by deletion of the highly basic amino acid cluster most N-terminal as to the HLH region of AhR or AhRR. Methylation of the two CpG sites in the XRE core sequence reduced the binding affinity to heterodimeric proteins, with 5-methylcytosine in the AhR recognition site exhibiting a greater inhibitory effect than that in the Arnt recognition site. PMID: 12944374 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 87: Biochem Biophys Res Commun. 2003 Aug 1;307(3):595-9. Molecular cloning and expression analysis of the aryl hydrocarbon receptor of Xenopus laevis. Ohi H, Fujita Y, Miyao M, Saguchi K, Murayama N, Higuchi S. Laboratory of Biopharmaceutics, School of Pharmaceutical Sciences, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan. The aryl hydrocarbon receptor (AHR) is a member of the basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) family of transcription factors. Although this receptor has been known to mediate the toxic effects of environmental pollutants, its physiological functions remain elusive. Here, we describe the isolation and expression pattern of the Xenopus AHR gene. The predicted amino acid sequence contained regions characteristic of other vertebrate AHRs. However, in line with previously described fish AHR genes, no distinct Q-rich domain was found. Phylogenetic analysis demonstrated that Xenopus AHR was clustered within the AHR1 clade. As in the case of mammalian AHR genes, the Xenopus AHR gene was expressed in all the adult tissues tested. Xenopus AHR was also expressed during early development, in parallel with expression of the CYP1A7 gene, which is thought to be regulated by AHR. These results suggest that while frogs are relatively tolerant to TCDD toxicity, the AHR of frogs has characteristics similar to those of other vertebrate AHRs. PMID: 12893265 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 88: Arch Biochem Biophys. 2003 Aug 1;416(1):53-67. Cell selective cAMP induction of rat CYP1B1 in adrenal and testis cells. Identification of a novel cAMP-responsive far upstream enhancer and a second Ah receptor-dependent mechanism. Zheng W, Brake PB, Bhattacharyya KK, Zhang L, Zhao D, Jefcoate CR. Department of Pharmacology, Medical Science Center, University of Wisconsin, 1300, University Avenue, Madison, WI 53706, USA. CYP1B1 is unique among P450 cytochromes in exhibiting inductive responses mediated by both the Ah receptor (AhR) and cAMP. cAMP induction was mediated either by a 189bp far upstream enhancer region (FUER, -5110 to -5298) or by a 230bp AhR-responsive enhancer region (AhER) (-797 to -1026). CYP1B1 luciferase reporters respond selectively to cAMP and TCDD in adrenal Y-1 cells (only cAMP), testis MA10 cells (cAMP>TCDD), and C3H10T1/2 mouse embryo fibroblasts (only TCDD). In Y-1 cells, which lack AhR, cAMP induction is totally dependent on the FUER, including absolute requirements for upstream and downstream halves of this region, and for CREB activity at a CRE sequence located at the 3(')-end. cAMP stimulation of the FUER was remarkably high (27-fold) and equally effective when linked to an HSV-TK promoter, indicating direct cAMP activation of the FUER. Binding of CREB to the essential CRE was demonstrated along with dominant negative effects of functionally impaired mutants. cAMP induction in MA10 cells was partially mediated by the FUER mechanism but was regulated additionally by AhER through AhR activity. MA10 cells also exhibit cAMP-dependent AhR down-regulation and AhR/Arnt complex formation. Mutations in AhER including XRE5 were similarly inhibitory to cAMP stimulation in MA10 cells and to TCDD stimulation in C3H10T1/2 cells. Transfection of AhR into the AhR-deficient Y-1 cells did not introduce this second mechanism, which indicated a need for additional components that are present in MA10 cells. PMID: 12859982 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 89: J Biochem (Tokyo). 2003 May;133(5):583-92. Critical enhancer region to which AhR/ARNT and Sp1 bind in the human CYP1B1 gene. Tsuchiya Y, Nakajima M, Yokoi T. Division of Drug Metabolism, Faculty of Pharmaceutical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-0934, Japan. Cytochrome P450 (CYP) 1B1 is known to be induced by polycyclic aromatic hydrocarbons including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The constitutive and TCDD-inducible transcriptional expression of human CYP1B1 is known to be cell-specific. In order to identify the cis-elements that cell-specifically regulate the constitutive and TCDD-inducible transcription of CYP1B1, we constructed luciferase reporter plasmids containing a series of deletions of the XRE core sequence in the 5'-flanking region of the human CYP1B1 gene. Luciferase assays were performed with MCF-7 (breast carcinoma), HepG2 (hepatocellular carcinoma), LS-180 (colon carcinoma), and OMC-3 (ovarian carcinoma) cells. Although there were large differences in the relative luciferase activity and inducibility between these four cell lines, the contribution of each reporter construct was similar. Constitutive expression increased with the regulatory elements that are present at -910 to -852 and -1652 to -1243. Potential enhancer elements for TCDD-induction were located from -1022 to -852 including three XREs, XRE3 at -853, XRE4 at -940, and XRE5 at -989. Gel shift analyses revealed binding of the AhR/ARNT heterodimer to XRE2 at -834, XRE3 at -853, XRE6 at -1024, and XRE7 at -1490. In addition, the binding of a nuclear transcriptional factor, Sp1, near XRE2 and XRE8 was observed. It was suggested that mutual interaction of XRE2 and XRE3 is important for transcriptional regulation, and that the Sp1 binding to the Sp1-like motif (-824) enhances both the constitutive and inducible transcriptional activities of the human CYP1B1 gene. PMID: 12801909 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 90: Anat Embryol (Berl). 2003 Jul;207(1):9-18. Epub 2003 Jun 3. Epithelial cells in the oviduct and vagina and steroid-synthesizing cells in the rabbit ovary express AhR and ARNT. Hasan A, Fischer B. Department of Anatomy and Cell Biology, Martin Luther University Faculty of Medicine, Grosse Steinstrasse 52, 06097 Halle (Saale), Germany. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor. Beside exogenous ligands, an increasing list of endogenous ligands has recently been described. The AhR and its dimerization partner AhR nuclear translocator (ARNT) were found in embryos, fetuses and in genital tract tissue. Its role in reproduction and fertility is not known. In the current study, AhR and ARNT expression and co-localization were studied immunohistochemicaly during the pre-implantation period in various genital tract organs (ovary, oviduct, vagina) of the rabbit. In the ovary, the AhR was expressed in the steroid-secreting interstitial cells, in follicular and granulosa cells, and in lutein cells. The receptor was localized in the cytoplasm. A cytoplasmic localization was also found in the oviduct epithelium with a diffuse cytoplasmic staining in the ampulla and a localized cytoplasmic localization in the isthmus. In the vagina, AhR localization changed from cytoplasmic in the non-pregnant animal to nuclear staining in the basal layer of the vaginal epithelium on day 6 of pregnancy. The ARNT protein was found in all AhR expressing cells except for oocytes within primordial follicles. Its localization was in the cytoplasm and/or nuclei. Therefore, the full complement for AhR/ARNT transcriptional activity was found in the studied organs. AhR expression showed stage-specific changes in both the uterus (Hasan and Fischer 2001) and the vagina during the pre-implantation period, while the ARNT protein exhibited no change in expression during this period. In summary, these findings indicate a functional AhR/ARNT complex in the rabbit genital tract epithelia. Its precise role and activation mechanism in reproductive tissue is not clear. It is supposed, however, that this complex may be involved in both hormone production and hormone-induced cellular changes during early pregnancy of the rabbit. PMID: 12783319 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 91: Pharmacogenetics. 2003 Jun;13(6):339-47. Structure and polymorphisms of human aryl hydrocarbon receptor repressor (AhRR) gene in a French population: relationship with CYP1A1 inducibility and lung cancer. Cauchi S, Stucker I, Cenee S, Kremers P, Beaune P, Massaad-Massade L. Laboratoire de Toxicologie Moleculaire, U-490 INSERM, 45 rue des Saints-Peres, 75270 Paris Cedex, France. OBJECTIVE: The aryl hydrocarbon receptor repressor (AhRR) protein may dimerize with the AhR nuclear translocator (ARNT) and may compete with the aryl hydrocarbon receptor (AhR) to bind the xenobiotic responsive elements. The result is a negative feedback mechanism that involves a down regulation of all genes regulated by the AhR transcription factor which positively regulates the expression of the Cytochrome P-4501A1 gene (CYP1A1). METHODS: The structure of the AhRR gene was reconstituted, then the genetic polymorphisms of this gene including the promoter were investigated and the link between these polymorphisms, CYP1A1 inducibility and lung cancer incidence in a French population was examined. Four polymorphisms were found, two in the coding region (609G>C and 1977G>C) and two in the 5'-untranslated region (-96G>A and -869A>T). Among the four polymorphisms, only one, the 609G>C has been previously described. The 609G>C and 1977G>C are localized respectively in exon 6 and 12 and lead to Pro554Ala and Asp641His substitutions, respectively. To evaluate the frequency of these allelic variants, a DNA library of a case-control study of lung cancer (164 controls and 171 patients) was screened. These polymorphisms were detected at the same allele frequency (0.40 for 609C, 0.05 for 1977C, 0.24 for -96A and 0.17 for -869T) in both controls and patients. Statistical analysis did not show any relationship between all the mutations found and CYP1A1 inducibility and lung cancer incidence. CONCLUSION: None of the polymorphisms were found to play a key role in CYP1A1 inducibility or in the susceptibility to develop lung cancer. PMID: 12777964 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 92: Nature. 2003 May 29;423(6939):545-50. Comment in: Nature. 2003 May 29;423(6939):487-8. Modulation of oestrogen receptor signalling by association with the activated dioxin receptor. Ohtake F, Takeyama K, Matsumoto T, Kitagawa H, Yamamoto Y, Nohara K, Tohyama C, Krust A, Mimura J, Chambon P, Yanagisawa J, Fujii-Kuriyama Y, Kato S. The Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan. Environmental contaminants affect a wide variety of biological events in many species. Dioxins are typical environmental contaminants that exert adverse oestrogen-related effects. Although their anti-oestrogenic actions are well described, dioxins can also induce endometriosis and oestrogen-dependent tumours, implying possible oestrogenic effects. However, the molecular mechanism underlying oestrogen-related actions of dioxins remains largely unknown. A heterodimer of the dioxin receptor (AhR) and Arnt, which are basic helix-loop-helix/PAS-family transcription factors, mediates most of the toxic effects of dioxins. Here we show that the agonist-activated AhR/Arnt heterodimer directly associates with oestrogen receptors ER-alpha and ER-beta. This association results in the recruitment of unliganded ER and the co-activator p300 to oestrogen-responsive gene promoters, leading to activation of transcription and oestrogenic effects. The function of liganded ER is attenuated. Oestrogenic actions of AhR agonists were detected in wild-type ovariectomized mouse uteri, but were absent in AhR-/- or ER-alpha-/- ovariectomized mice. Our findings suggest a novel mechanism by which ER-mediated oestrogen signalling is modulated by a co-regulatory-like function of activated AhR/Arnt, giving rise to adverse oestrogen-related actions of dioxin-type environmental contaminants. PMID: 12774124 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 93: Mol Pharmacol. 2003 Jun;63(6):1373-81. Aryl hydrocarbon receptor gene silencing with small inhibitory RNA differentially modulates Ah-responsiveness in MCF-7 and HepG2 cancer cells. Abdelrahim M, Smith R 3rd, Safe S. Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, Veterinary Research Building 409, College Station, TX 77843-4466, USA. Sequence-specific small interfering RNA (siRNA) duplexes can be used for gene silencing in mammalian cells and as mechanistic probes for determining gene function. Transfection of siRNAs for the aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (ARNT) mRNAs in MCF-7 breast cancer cells resulted in a 60 to 80% decrease in levels of AhR and ARNT proteins in whole-cell extracts and decreased binding of nuclear extracts to 32P-labeled dioxin-responsive element. siRNA for the AhR also decreased 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1A1 protein, CYP1A1-dependent activity, and luciferase activity in cells transfected with an Ah-responsive construct. 17beta-estradiol (E2) induces proliferation of MCF-7 cells through enhanced G0/G1 --> S phase progression, and this response is inhibited in cells cotreated with E2 plus TCDD. The effects of TCDD on E2-induced cell-cycle progress were partially blocked in MCF-7 cells transfected with siRNA for AhR. The results also indicated that siRNA-dependent decreases in AhR protein in MCF-7 cells were accompanied by increased G0/G1 --> S phase progression, suggesting a growth-inhibitory role for the "endogenous" AhR. Surprisingly, TCDD alone induced G0/G1 --> S phase progression and exhibited estrogenic activity in MCF-7 cells transfected with siRNA for the AhR. In contrast, degradation of the AhR in HepG2 liver cancer cells resulted in decreased G0/G1 --> S phase progression, and this was accompanied by decreased expression of cyclin D1, cyclin E, cyclin-dependent kinase 2 (cdk2), and cdk4. In the absence of ligand, the AhR exhibits growth-inhibitory (MCF-7) and growth-promoting (HepG2) activity that is cell context-dependent. PMID: 12761348 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 94: Biochem Biophys Res Commun. 2003 Apr 18;303(4):1095-100. Identification of novel splice variants of ARNT and ARNT2 in the rat. Korkalainen M, Tuomisto J, Pohjanvirta R. National Public Health Institute, Laboratory of Toxicology, Department of Environmental Health, P.O. Box 95, Neulaniementie 4, FIN-70701 Kuopio, Finland. Merja.Korkalainen@ktl.fi Most of the biochemical and toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) are mediated by the bHLH/PAS protein AH receptor (AHR). For regulation of gene activities, AHR dimerizes with another member of the bHLH/PAS protein family, AHR nuclear translocator (ARNT). A substrain of Wistar rats, Han/Wistar (Kuopio) (H/W), is about 1000-fold more resistant to the acute lethality of TCDD than other strains, exemplified by Long-Evans (Turku/AB) (L-E); the LD50 values for these two strains are >9600 and 10-20 microg/kg, respectively. Previous studies have demonstrated that the major reason for the exceptional TCDD resistance of H/W rats lies in their AHR, which is remodeled at its C-terminal transactivation domain, but there appears to be another contributing gene product. The present study set out to compare the primary structure of ARNT and the closely related ARNT2 proteins in H/W and L-E rats by cDNA cloning. To our surprise, we found several isoforms of these proteins only one of which has previously been reported in rats. All of the isoforms appeared to arise from alternative splicing. For ARNT, isoforms with deletions at exon 5, 3(') end of exon 6 or 5(') end of exon 11, or with an insertion at 5(') end of exon 20 were discovered. There was also interindividual variation in the number of glutamine-encoding codons at 5(') end of exon 16. The most exciting new variant was revealed for ARNT2, because the insertion found at 5(') end of exon 19 disrupts the functionally critical transactivation domain in the protein, implying a dominant negative role for this isoform. The relative expression levels of the variants did not differ in the two rat strains, nor did TCDD modify the ratios, suggesting that the variants do not contribute to TCDD resistance. However, the regulation of ARNT and ARNT2 activities may be more intricate than previously assumed. PMID: 12684048 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 95: Cardiovasc Toxicol. 2002;2(4):263-74. Aryl hydrocarbon receptor null mice develop cardiac hypertrophy and increased hypoxia-inducible factor-1alpha in the absence of cardiac hypoxia. Thackaberry EA, Gabaldon DM, Walker MK, Smith SM. Molecular and Environmental Toxicology Center, Department of Nutritional Sciences, University of Wisconsin, Madison 53706, USA. The aryl hydrocarbon receptor (AhR) is a member of the basic helix loop helix PAS (Per-ARNT-SIM) transcription family, which also includes hypoxiainducible factor-1alpha (HIF-1alpha) and its common dimerization partner AhR nuclear translocator (ARNT). Following ligand activation or hypoxia, AhR or HIF-1alpha, respectively, translocate into the nucleus, dimerize with ARNT, and regulate gene expression. Mice lacking the AhR have been shown previously to develop cardiac enlargement. In cardiac hypertrophy, it has been suggested that the myocardium becomes hypoxic, increasing HIF-1alpha stabilization and inducing coronary neovascularization, however, this mechanism has not been demonstrated in vivo. The purpose of this study was to investigate the cardiac enlargement reported in AhR(-/-) mice and to determine if it was associated with myocardial hypoxia and subsequent activation of the HIF-1alpha pathway. We found that AhR(-/-) mice develop significant cardiac hypertrophy at 5 mo. However, this cardiac hypertrophy was not associated with myocardial hypoxia. Despite this finding, cardiac hypertrophy in AhR(-/-) mice was associated with increased cardiac HIF-1alpha protein expression and increased mRNA expression of the neovascularization factor vascular endothelial growth factor (VEGF). These data demonstrate that the development of cardiac hypertrophy in AhR(-/-) mice not associated with myocardial hypoxia, but is correlated with increased cardiac HIF-1alpha protein and VEGF mRNA expression. PMID: 12665660 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 96: Arch Biochem Biophys. 2003 Apr 1;412(1):95-105. Mutational analysis of the mouse aryl hydrocarbon receptor tyrosine residues necessary for recognition of dioxin response elements. Minsavage GD, Vorojeikina DP, Gasiewicz TA. Department of Environmental Medicine, School of Medicine, University of Rochester, Rochester, NY 14642, USA. Tyrosine phosphorylation of the aryl hydrocarbon receptor (AhR), a member of the basic helix-loop-helix/PER-ARNT-SIM transcription factor family, has been shown to regulate its dioxin response elements (DRE) binding ability, although no specific residues have been directly demonstrated to be phosphorylated. Of the 23 tyrosines in the mouse AhR, 19 are conserved across all mammalian species sequenced thus far. The studies presented here were conducted to examine tyrosine residue(s) that are both likely candidates of phosphorylation and necessary for DNA binding and/or transcriptional activity of the AhR. Two-dimensional gel electrophoresis of phosphatase-treated AhR indicated that the receptor is phosphorylated on serine/threonine and tyrosine residues. Computational analysis predicted several highly conserved tyrosine residues to be phosphorylated. Both the N terminus (amino acids 1-399) and the C terminus (amino acids 399-805) of the mouse receptor synthesized in vitro using a rabbit reticulocyte lysate system are tyrosine phosphorylated as detected by antiphosphotyrosine antibodies. Furthermore, the N-terminal AhR bound DRE in a ligand-dependent manner similar to that by the full-length receptor, suggesting that phosphorylated tyrosines involved in DNA binding are likely located in the region between residues 1 and 399. Mouse AhR tyrosine (Y) residues were evaluated by phenylalanine (F) mutational analysis for both DNA binding (electrophoretic mobility shift assays; EMSAs) and ability to induce a DRE-driven reporter gene in transiently transfected AhR-deficient cells. Of the 12 tyrosine residues in the N-terminal AhR, only a tyrosine 9 mutant (AhRY9F) significantly decreased DRE binding as determined by EMSA. Similarly, only the AhRY9F mutant decreased the DRE-driven luciferase expression in AhR-deficient cells. Overall, these data strongly suggest that the putative posttranslational modification at, or mediated by, tyrosine 9, and not any other individual mouse AhR tyrosine residue, is necessary for AhR DRE binding and transcriptional activity. PMID: 12646272 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 97: Mol Pharmacol. 2003 Apr;63(4):915-24. Species-specific transcriptional activity of synthetic flavonoids in guinea pig and mouse cells as a result of differential activation of the aryl hydrocarbon receptor to interact with dioxin-responsive elements. Zhou JG, Henry EC, Palermo CM, Dertinger SD, Gasiewicz TA. Molecular Toxicology and Environmental Medicine Program, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA. To investigate possible species-specificity of aryl hydrocarbon receptor (AhR)-mediated signal transduction pathways, activities of 2,3,7,8-tetrochlorodibenzo-p-dioxin (TCDD) and six synthetic flavonoids were evaluated in mouse hepatoma and guinea pig adenocarcinoma cells transfected with an AhR-responsive luciferase reporter. Rank order potency in these two cell lines was similar for the ability of these flavonoids to antagonize TCDD-induced reporter gene expression. However, in the presence of flavone alone, a species-specific difference in agonist activity was observed. In guinea pig cells, several flavonoids demonstrated agonist activity up to 50% of the maximum TCDD response. In mouse cells, however, no significant agonist activity was observed at the same concentrations based on luciferase enzyme activity, protein expression, and mRNA analysis. Moreover, competitive ligand-binding assays, using [(3)H]TCDD in cytosolic fractions, demonstrated that 3'-methoxy-4'-nitroflavone had a similar IC(50) in both recombinant cell lines, suggesting that the flavone has similar binding affinity to receptors from both species. However, electrophoretic mobility shift assay using the cytosolic fractions demonstrated that this flavone elicited binding to the DRE by guinea pig but not mouse AhR complex. The dependence of the AhR in this differential interaction was further demonstrated using in vitro synthesized guinea pig and mouse Ah receptors and mouse Arnt. Together, these data suggest that the differential agonist/antagonist activity of these flavone derivatives is caused by the efficacy of these flavonoids in eliciting an AhR conformation that recognizes regulatory response elements in a species-specific manner. PMID: 12644593 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 98: Biochem Pharmacol. 2003 Mar 15;65(6):941-8. P23 enhances the formation of the aryl hydrocarbon receptor-DNA complex. Shetty PV, Bhagwat BY, Chan WK. Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, CA 95211, USA. The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that requires heterodimerization with its partner, the Ah receptor nuclear translocator (Arnt), for activation of transcription. The heterodimer specifically recognizes the dioxin response element (DRE), which contains a core sequence (5'-TNGCGTG-3'). This AhR/Arnt/DRE complex has been well characterized and can be observed readily by the gel shift assay. Human AhR and Arnt with a C-terminal histidine tag have been expressed functionally using a baculovirus expression system. However, after purification of these proteins using the metal resin, they are not able to bind the response element in a ligand-dependent manner unless crude extracts, such as the rabbit reticulocyte lysate (RRL), are reconstituted with these proteins. Proteins in the RRL are responsible for this restoration of the gel shift complex because the activity is sensitive to both heat and proteolytic treatments. We have examined whether hsp90 and p23 are among the protein factors in the RRL that are responsible for this activity. By performing fractionation studies using filtration devices and immunodepletion studies, we have selectively fractionated these proteins. Among all the fractions, the centricon-10 retentate, which contains 100% of p23 but no hsp90, possessed the most enriched activity. Purified bacterial-expressed p23 restored the gel shift complex; the mechanism was mediated at the heterodimerization step and was hsp90-dependent. PMID: 12623125 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 99: Mol Pharmacol. 2003 Mar;63(3):597-606. Functional analysis of murine aryl hydrocarbon (AH) receptors defective in nuclear import: impact on AH receptor degradation and gene regulation. Song Z, Pollenz RS. Department of Biology, University of South Florida, Tampa, Florida 33620, USA. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that is also a substrate for the 26S proteasome. However, the subcellular location of the degradation events or the requirement for nuclear transport has not been resolved. To gain insight into both ligand-dependent and independent degradation of the AHR, studies were designed to evaluate the relationship between AHR localization, stability, and gene regulation in a defined cell culture model system. The strategy of these studies was to generate stable cell lines expressing murine AHR proteins that were defective in nuclear import and then to assess the location of the AHR, the time course of AHR degradation, and the level of induction of endogenous CYP1A1 protein after exposure to 2,3,7,8-tetrachlorodibezo-p-dioxin (TCDD), geldanamycin (GA), or the protease inhibitor carbobenzoxy-L-leucyl-L-leucyl-leucinal (MG-132). Mutation within the putative nuclear localization sequence (NLS) resulted in AHR mutants that were severely defective in nuclear import as evaluated by immunocytochemical staining after exposure to TCDD, GA, or MG-132. Importantly, the NLS mutants exhibited identical levels of degradation along a similar time course as wild-type AHR after exposure to TCDD or GA when stably expressed in either murine hepatoma cells (Hepa-1) or hamster lung cells (E36). In contrast, the NLS mutants were severely defective in ligand-mediated induction of CYP1A1 expression. These findings imply that the proteolytic machinery present in the cytoplasmic compartment is sufficient to degrade the AHR and that nuclear translocation, binding with ARNT, or DNA binding are not necessary for efficient degradation of the AHR. PMID: 12606767 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 100: Toxicol Sci. 2003 Mar;72(1):84-91. TCDD treatment eliminates the long-term reconstitution activity of hematopoietic stem cells. Sakai R, Kajiume T, Inoue H, Kanno R, Miyazaki M, Ninomiya Y, Kanno M. Department of Immunology, Graduate School of Biomedical Science, Hiroshima University, Minami-ku, Hiroshima 734-8551, Japan. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an endocrine disrupting chemical (EDC), can cause carcinogenesis, immunosuppression, and teratogenesis, through a ligand-activated transcription factor, the aryl hydrocarbon receptor (AhR). Despite remarkable recent advances in stem cell biology, the influence of TCDD on hematopoietic stem cells (HSCs), which possess the ability to reconstitute long-term multilineage hematopoiesis, has not been well investigated. In this study we examined the influence of TCDD on HSCs enriched for CD34(-), c-kit(+), Sca-1(+), lineage negative (CD34-KSL) cells. The number of the CD34-KSL cells was found to be increased about four-fold upon a single oral administration of TCDD (40 micro g/kg body weight). Surprisingly, we found that these TCDD-treated cells almost lost long-term reconstitution activity. This defect was not present in AhR(-/-) mice. These findings suggest that modulation of AhR/ARNT system activity may have an effect on HSC function or survival. PMID: 12604837 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 101: Endocrinology. 2003 Mar;144(3):767-76. Expression of aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator messenger ribonucleic acids and proteins in rat and human testis. Schultz R, Suominen J, Varre T, Hakovirta H, Parvinen M, Toppari J, Pelto-Huikko M. Department of Developmental Biology, Tampere University, FIN-33014 Tampere, Finland. Dioxins, e.g. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), use the aryl hydrocarbon receptor (AHR)/aryl hydrocarbon receptor nuclear translocator (ARNT) receptor complex to mediate their toxic actions. In addition to interaction with environmental pollutants, several transcription factors, steroid receptors, and growth factors are capable interacting with the AHR/ARNT complex, which suggests a constitutive role for the receptor complex. The testis has been reported to be among the most sensitive organs to TCDD exposure. Our experiments revealed a complex distribution of AHR and ARNT mRNAs and proteins in rat and human testis. AHR and ARNT immunoreactivities could be detected in the nuclei of interstitial and tubular cells. The incubation of seminiferous tubules in a serum-free culture medium resulted in up-regulation of AHR mRNA, which could be depressed by adding FSH to the culture medium. Furthermore, the incubation of tubular segments with a solution of 1 or 100 nM TCDD resulted in a 2- to 3-fold increase in apoptotic cells. Thus, up-regulation of AHR in cultured tubular segments and consecutive depression by FSH suggest a role for AHR in controlled cell death during spermatogenesis. We suggest that AHR and ARNT mediate effects by direct action on testicular cells in the rat and human testis. PMID: 12586752 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 102: Biochim Biophys Acta. 2003 Feb 17;1619(3):263-8. Functional role of AhR in the expression of toxic effects by TCDD. Mimura J, Fujii-Kuriyama Y. Department of Biomolecular Science, Graduate School of Life Science, Tohoku University, Aoba-ku, 980-8578, Sendai, Japan. Cytochrome P450 1A1 (CYP1A1) is one of the xenobiotic metabolizing enzymes (XMEs), which is induced by polycyclic aromatic hydrocarbons (PAHs). The most potent inducer of CYP1A1 is 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). In addition, TCDD induces a broad spectrum of biochemical and toxic effects, such as teratogenesis, immunosuppression and tumor promotion. Most, if not all, of the effects caused by TCDD and other PAHs are known to be mediated by AhR (aryl hydrocarbon receptor or dioxin receptor) which has a high binding affinity to TCDD. The liganded AhR translocates from cytoplasm to nuclei where it switches its partner molecule from Hsp90 to Arnt. Thus formed AhR/Arnt heterodimer binds a specific DNA sequence designated XRE in the promoter region of the target genes including CYP1A1, UDP-glucuronosyl transferase and others to enhance their expression. Although it remains to be studied how AhR is involved in the other TCDD-induced biological effects such as teratogenesis and immunosuppression than induction of XMEs, it is believed that these adverse TCDD effects are caused by untimely activation of gene expression by ligand-activated AhR in the biological process. We summarize the present knowledge about functional role of AhR in TCDD-induced biological effects. Publication Types: Review Review, Tutorial PMID: 12573486 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 103: Biochem Biophys Res Commun. 2003 Jan 24;300(4):943-9. Aryl hydrocarbon receptor ligands repress T-cadherin expression in vascular smooth muscle cells. Niermann T, Schmutz S, Erne P, Resink T. Department of Research, Cardiovascular Laboratories, ZLF 320, Basel University Hospital, Hebelstrasse 20, CH 4031 Basel, Switzerland. T-cadherin, a glycosylphosphatidylinositol-modified cadherin subtype, is highly expressed in cardiac and vascular tissues. Neither the functions nor regulation of T-cadherin in these tissues is understood. We have cloned rat T-cadherin cDNA encoding the full length amino acid sequence. The 5(') untranslated nucleotide sequences of rat, mouse, and human T-cadherin contain a conserved GCGTG motif which constitutes the invariant core sequence of dioxin- or xenobiotic-regulatory elements. These elements function as target sites for aryl hydrocarbon receptor/aryl hydrocarbon nuclear translocator (AhR/ARNT) in genes regulated by this transcription factor. Using cultures of rat aortic smooth muscle cells this study presents data revealing T-cadherin as a putative target gene for negative regulation of expression through AHR signalling. Prototypic AHR agonists benzo[a]pyrene (BaP) or 7,12-dimethylbenzanthracene (DMBA) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) repressed T-cadherin mRNA levels. Repression was antagonized by the cognate AHR antagonist alpha-naphthoflavone (alpha-NF). Repression was insensitive to inhibitors of gene transcription (actinomycin D) or de novo protein synthesis (cycloheximide), suggesting AHR/ARNT functions directly in transcriptional repression of T-cad. Regulation of adhesion proteins through the AHR pathway may represent a novel mechanism of action by atherogenic polycyclic aromatic hydrocarbons. PMID: 12559965 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 104: Gene. 2003 Jan 16;303:197-206. Characterization of two distinct aryl hydrocarbon receptor (AhR2) genes in Atlantic salmon (Salmo salar) and evidence for multiple AhR2 gene lineages in salmonid fish. Hansson MC, Wittzell H, Persson K, von Schantz T. Department of Animal Ecology, Lund University, Ecology Building, Solvegatan 37, Sweden. maria.hansson@zooekol.lu.se The aryl hydrocarbon receptor (AhR) mediates the toxicity of several environmental contaminants, e.g. 2,3,7,8-tetrachlorodibenzo-p-dioxin, and other halogenated hydrocarbons in vertebrates. This receptor initiates the transcription of several biotransformation enzymes, which in turn are responsible for causing severe harm to biological tissue. Here we describe the isolation and complete characterization of the first two AhR genes from the teleost fish Atlantic salmon (Salmo salar). The predicted amino acid sequences contain regions characteristic of other vertebrate AhRs including basic helix-loop-helix (bHLH) and PER-ARNT-SIM (PAS) domains but show little similarity to other vertebrate AhRs across the C-terminal half. Furthermore, they do not contain distinct Q-rich domains as found in the mammalian AhR, which is in line with previously described fish AhR genes. The salmon cDNAs encode 1106 and 1107 putative residues, respectively, approximately 50 amino acids longer than previously characterized AhR genes. Phylogenetic analyses demonstrated that the two salmon AhR sequences cluster within the AhR subfamily of the bHLH-PAS family, in a clade containing fish AhR2 genes. Although the two AhR2 forms are 92% identical at the amino acid level, the distribution of sequence differences and the presence of both forms in 30 tested individuals suggest that they are not allelic but derived from separate loci. Interestingly, they are not orthologs of the rainbow trout (Oncorhynchus mykiss) AhR2 alpha and beta genes and the new salmon loci are therefore here designated AhR2 gamma and AhR2 delta. In line with this, PCR with DNA from rainbow trout revealed a new trout AhR locus that was more similar to the two salmon genes than to the trout AhR2 alpha and beta genes, suggesting that the rainbow trout possesses at least three distinct AhR2 genes. The presence of multiple AhR genes in these species is probably a consequence of the genome duplications that occurred in the early evolution of fish and later also specifically in the salmonid lineage. Reverse transcription-PCR analyses revealed that both AhR2 gamma and AhR2 delta are transcribed in the liver, spleen and muscles of adult salmon. PMID: 12559581 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 105: Mol Pharmacol. 2003 Feb;63(2):392-400. Agonist but not antagonist ligands induce conformational change in the mouse aryl hydrocarbon receptor as detected by partial proteolysis. Henry EC, Gasiewicz TA. Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA. ellen_henry@urmc.rochester.edu The cytosolic transcription factor known as the aryl hydrocarbon receptor (AhR) undergoes transformation to a DNA-binding form by a series of processes initiated by binding of ligand. Subsequent steps include dissociation of several proteins that are complexed with the inactive receptor, nuclear translocation, and dimerization with Arnt. We have used limited proteolysis of the in vitro-translated mouse AhR to determine whether this technique can detect conformational change(s) associated with AhR transformation and whether the effect of agonist and antagonist ligands can be distinguished by this assay. Limited digestion of [(35)S]AhR/AhR nuclear translocator (Arnt) by trypsin produced a peptide of approximately 40 kDa that was more resistant to proteolysis in the presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) than vehicle and was also Arnt-dependent. This trypsin-resistant peptide was also elicited in the presence of other agonist ligands, but not with antagonist ligands that do not form the DNA-binding AhR/Arnt complex. Immunoblot of trypsin-treated AhR/Arnt +/- TCDD indicated that the trypsin-resistant peptide did not include the N-terminal portion of the AhR against which the antibody was made. Truncated AhRs were also subjected to limited trypsinization. From AhR(1-399), a TCDD-dependent peptide of approximately 35 kDa was observed; from the constitutively active AhR(1-348), a band of approximately 30 kDa was produced from vehicle- and TCDD-treated protein. From these observations, we hypothesize that the trypsin-resistant peptide from full-length AhR spans approximately from amino acid 80 to 440. We conclude that agonist ligands initiate structural alteration in AhR that is Arnt-dependent and at least partially involves the ligand-binding/Per-Arnt-Sim domain. PMID: 12527811 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 106: Neurotoxicology. 2002 Dec;23(6):783-93. Constitutive and TCDD-induced expression of Ah receptor-responsive genes in the pituitary. Huang P, Ceccatelli S, Hakansson H, Grandison L, Rannug A. Division of Toxicology and Neurotoxicology, Division of Occupational Toxicology, Institute of Environmental Medicine, Karolinska Institutet, S-171 77 Stockholm, Sweden. The 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related substances cause a wide variety of pathological alterations, with the most severe being progressive anorexia and body weight loss. These features suggest a possible involvement of the nervous system and endocrine organs, including the pituitary gland. TCDD-related toxicity is considered mainly to be mediated by the aryl hydrocarbon receptor (AHR) protein, which binds TCDD, and heterodimerizes with its partner protein, the aryl hydrocarbon receptor nuclear translocator (ARNT), and binds to xenobiotica responsive elements (XREs) in the promoter regions of biotransformation genes as well as genes involved in growth, differentiation and cellular homeostasis. In the present study, we have investigated the expression of AHR responsive genes in the pituitary of untreated and TCDD treated 129/SV/C57BL/6 mice in vivo and in pituitary cells in vitro. After TCDD or beta-naphthoflavone (beta NF) treatment, the relative levels of cytochrome P4501A1 (CYP1A1) mRNA and protein were dramatically increased in pituitary cells. The AHR repressor (AHRR) mRNA level was induced 7-13-fold by TCDD and beta NF. Furthermore, the expression of the adrenocorticotrophic hormone (ACTH) precursor, the proopiomelanocortin (POMC) gene, was investigated. A three-fold increase in POMC mRNA was observed in the pituitary of TCDD treated mice. POMC mRNA level was also increased in the pituitary cell line AtT-20 after TCDD treatment. The proteins encoded by POMC translational products, ACTH and beta-endorphin, were found with immunocytochemistry staining to be increased in AtT-20 cells after TCDD exposure. The presence of several XRE sequences in the promoter region and in the first intron of the human POMC gene suggest that the up-regulation of POMC expression in the pituitary may play a role in the endocrine alterations induced by TCDD. All together, the results point to the pituitary gland being a direct target for TCDD. PMID: 12520768 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 107: Toxicol Sci. 2003 Jan;71(1):20-6. Correlation between gene expression of aryl hydrocarbon receptor (AhR), hydrocarbon receptor nuclear translocator (Arnt), cytochromes P4501A1 (CYP1A1) and 1B1 (CYP1B1), and inducibility of CYP1A1 and CYP1B1 in human lymphocytes. Lin P, Hu SW, Chang TH. Institute of Toxicology, Chung Shan Medical University, Taichung, Taiwan. ppl@csmu.edu.tw The relationships between gene expression of aryl hydrocarbon receptor (AhR), aryl hydrocarbon receptor nuclear translocator (Arnt), cytochromes P4501A1 (CYP1A1), 1B1 (CYP1B1), CYP1A1, and the inducibility of CYP1A1 and CYP1B1 were determined in 32 cultivated human lymphocytes. Cytochrome P450 induction was performed by incubating lymphocytes with benzanthracene. The relative gene expression levels were determined by quantitative real-time RT-PCR assay. We found that gender is an important confounding factor for gene expression in cultivated lymphocytes. AhR, CYP1A1 and CYP1B1 levels in noninduced lymphocytes were significantly higher in female nonsmokers than in male nonsmokers (p < 0.05). Nevertheless, CYP1A1 and CYP1B1 inducibility was lower in female nonsmokers. CYP1A1 inducibility was higher in male smokers than in male nonsmokers (p < 0.05). After controlling for gender and cigarette smoking, AhR levels positively correlated with CYP1B1 levels and CYP1A1 inducibility (p < 0.01 and p = 0.03, respectively). Arnt levels also correlated with CYP1B1 levels in induced lymphocytes (p < 0.01). However, AhR levels were negatively correlated with CYP1B1 inducibility. These data indicate that AhR expression associates with individual variation of CYP1A1 inducibility and CYP1B1 expression in cultivated lymphocytes. Furthermore, gender and cigarette smoking are important confounding factors for gene expression levels in cultivated lymphocytes. PMID: 12520072 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 108: Mol Pharmacol. 2003 Jan;63(1):119-27. Mechanism of rat UDP-glucuronosyltransferase 1A6 induction by oltipraz: evidence for a contribution of the Aryl hydrocarbon receptor pathway. Auyeung DJ, Kessler FK, Ritter JK. Department of Pharmacology and Toxicology, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond, Virginia 23298-0613, USA. The utility of oltipraz as a cancer chemopreventive agent is thought to depend on the induction of enzymes involved in phase 2 xenobiotic detoxification. Although studies of some enzymes induced by oltipraz implicate a novel transcriptional activating pathway involving Nrf2 and antioxidant-response elements (AREs), the mechanism of phenol UGT induction has remained unclear. Previous work showed that UGT1A6 is transcribed from two promoters, P1 and P2, that are both induced by oltipraz in rat liver. The effect also occurs in rat hepatocytes treated with oltipraz (concentrations >3 microM). To investigate the mechanism, luciferase reporter plasmids under the control of P1 [p(-1078/+27)1A6P1-luc] or P2 [p(-1354/+65)1A6P2-luc] were transfected into rat hepatocytes and tested for inducibility. P1, but not P2, showed responsiveness to oltipraz (2- to 5-fold increase) and 3-methylcholanthrene (10- to 30-fold increase). Because P1 contained no visible AREs, the role of a xenobiotic response element (XRE) centered between bases -134 and -129 was evaluated. Mutation of the XRE core reduced the effects of both oltipraz and 3-methylcholanthrene on the P1 reporter. The 1A6 XRE conferred oltipraz responsiveness on the simian virus 40 promoter of pGL3-Promoter. Comparative effects of oltipraz and 3-methylcholanthrene on transfected cytochrome P4501A1 reporters support the general but relatively weak XRE-stimulating activity of oltipraz. The involvement of the aryl hydrocarbon receptor (AHR) and aryl hydrocarbon nuclear translocator (ARNT) in mediating the effects of oltipraz on the XRE is supported by electrophoretic mobility supershift data and AHR/ARNT overexpression studies. These data raise questions about the contribution of AHR and other secondary induction pathways in the mechanism of oltipraz. PMID: 12488544 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 109: J Biochem Mol Toxicol. 2002;16(6):317-25. A dynamic role for the Ah receptor in cell signaling? Insights from a diverse group of Ah receptor interacting proteins. Carlson DB, Perdew GH. Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary Science, Pennsylvania State University, University Park, PA 16802, USA. The aryl hydrocarbon (Ah) receptor (AhR) is a member of the basic helix-loop-helix PER-ARNT-SIM (PAS) transcription factor family. Consistent with the notion that PAS proteins are biological sensors, AhR binding to Ah toxicants induces or represses transcription of a wide range of genes and results in a cascade of toxic responses. However, an endogenous role for AhR in development and homeostasis is supported by (1) the discovery of low affinity, endogenous ligands; (2) studies demonstrating a role for the receptor in development of liver and vascular systems, that were established using mice lacking AhR expression; and (3) the presence of functional dioxin-responsive elements in promoter regions of genes involved in cellular growth and differentiation. A large body of recent literature has implicated AhR in multiple signal transduction pathways. AhR is known to interact with signaling pathways that are mediated by estrogen receptor and other hormone receptors, hypoxia, nuclear factor kappaB, and retinoblastoma protein. In addition, AhR complexes may affect cellular signaling through interactions with various other regulatory and signaling proteins, including PAS heterodimerization partners (ARNT), chaperone and immunophilin-like proteins (e.g. HSP90, XAP2/ARA9/AIP, p23), protein kinases and phosphatases (e.g. tyrosine kinases, casein kinase 2, protein kinase C), and coactivators (e.g. SRC-1, RIP 140, CBP/p300). Here we summarize the types of molecular cross talk that have been identified between AhR and cell signaling pathways. Copyright 2002 Wiley Periodicals, Inc. J Biochem Mol Toxicol 16:317-325, 2002; Published online in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/jbt.10051 Publication Types: Review PMID: 12481307 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 110: Virchows Arch. 2002 Nov;441(5):481-9. Epub 2002 Jun 27. The arylhydrocarbon receptor (AhR), but not the AhR-nuclear translocator (ARNT), is increased in hearts of patients with cardiomyopathy. Mehrabi MR, Steiner GE, Dellinger C, Kofler A, Schaufler K, Tamaddon F, Plesch K, Ekmekcioglu C, Maurer G, Glogar HD, Thalhammer T. Department of Cardiology, University of Vienna, Austria. mohammad.mehrabi@univie.ac.at The objective of this study was to investigate the expression of the arylhydrocarbon receptor (AhR) and its partner AhR-nuclear translocator (ARNT) in left ventricle specimens from explanted hearts from patients with cardiomyopathy (CMP). Explanted hearts from 16 patients with ischemic (n=9, age 63+/-12 years) and dilative (n=7, age 54+/-12 years) CMP, undergoing heart transplantation were examined. Healthy donor hearts from five accident victims served as controls. As these donors were of younger age (32+/-11 years), additionally, donor hearts from three older accident victims (age 48+/-15 years) without clinical symptoms but with signs of ventricular hyperthrophy (n=1) or atherosclerotic lesions (n=2) were included ("pathological controls"). Expression of AhR and ARNT was analyzed using semi-quantitative immunohistochemistry, and in selected samples, Western blot- and reverse-transcription polymerase chain reaction analysis were performed to confirm AhR and ARNT expression. Immunohistological analysis revealed weak to intermediate staining of anti-AhR in control, but weak to intense staining in CMP- and "pathologic control" specimens, indicating significantly increased AhR levels in the diseased heart. Moreover, in CMP specimens, the percentage of AhR-positive cells was strongly increased. Higher anti-AhR staining was also seen in two atherosclerotic "pathologic control" specimens. In all groups, the intensity of anti-ARNT staining was more pronounced than AhR staining, but significant differences or any age-related alterations were not observed. In conclusion, the increased cellular content of AhR in left ventricular specimens from CMP patients suggests a role for AhR in heart disease. PMID: 12447679 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 111: J Biochem (Tokyo). 2002 Nov;132(5):795-802. Overexpression of the aryl hydrocarbon receptor (AhR) accelerates the cell proliferation of A549 cells. Shimba S, Komiyama K, Moro I, Tezuka M. Department of Health Science, College of Pharmacy, Nihon University, Narashinodai, Funabashi, Chiba 274-8555. shimba@pha.nihon-u.ac.jp The arylhydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates a spectrum of toxic and biological effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds. Although the physiological ligand for the AhR has not yet been identified, several reports have suggested that the AhR may play important roles not only in the regulation of xenobiotic metabolism but also in the maintenance of homeostatic functions [Singh et al. (1996) Arch. Biochem. Biophys. 329, 47-55; Crawford et al. (1997) Mol. Pharmacol. 52, 921-927; Chang et al. (1998) Mol. Cell. Biol. 18, 525-535]. Several lines of evidence suggest that one of the possible physiological roles of the AhR is regulation of cell proliferation. In this study, we first showed that treatment of A549 cells with the AhR agonist stimulates cell proliferation. The effect was antagonized by co-treatment with alpha-naphthoflavone. To obtain direct evidence that the AhR regulates cell proliferation, we isolated the clones that overexpress the AhR. These clones grow faster than control cells, and the rate of growth is proportional to the amount of the AhR. Cell cycle analysis revealed that the acceleration of cell growth by overexpression of the AhR is most probably due to shortening of the late M to S phases. Studies on the expression profiles of cell cycle regulators showed that the AhR or AhR ligand induces the expression of DP2, PCNA, and RFC38. DP2 is the transcription factor that forms the functional dimer with E2F and regulates the expression of several genes involved in DNA synthesis. Interestingly, both PCNA and RFC38 are target genes of E2F and the DP complex. Also, both of these factors are involved in regulating DNA polymerase delta activity. E2F activity was substantially increased in both the AhR-overexpressing cells and the AhR-agonist treated cells, suggesting that AhR-activated E2F/DP2 may induce the expression of PCNA and RFC38 and subsequent DNA synthesis. Down-regulation of the expression of the Arnt by RNAi diminished the effects of the AhR on the cell proliferation of the A549 cells. Consequently, we conclude that the AhR, presumably in collaboration with the Arnt, activates the DNA synthesis and the subsequent cell proliferation in A549 cells. PMID: 12417031 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 112: Carcinogenesis. 2002 Oct;23(10):1625-30. The redox protein thioredoxin-1 regulates the constitutive and inducible expression of the estrogen metabolizing cytochromes P450 1B1 and 1A1 in MCF-7 human breast cancer cells. Husbeck B, Powis G. Arizona Cancer Center, University of Arizona, 1515 N. Campbell Avenue, Tucson, AZ 85724-5024, USA. The oxidative metabolites of estrogen have been proposed to play an important role in the development of some human cancers. The two major pathways of estrogen metabolism, to the carcinogenic 4-hydroxyestradiol (4-OHE2) and to the non-carcinogenic 2-hydroxyestradiol (2-OHE2), are mediated by cytochromes P450 CYP1B1 and CYP1A1, respectively. The expression of CYP1A1 and CYP1B1 is regulated by the aromatic hydrocarbon receptor/Ah receptor nuclear translocator (AhR/ARNT) transcription factor complex. CYP1B1 expression is elevated in a wide range of human cancers but is not found in corresponding normal tissue. Thioredoxin-1 (Trx-1) is a small redox protein that is overexpressed in a number of human cancers. We report that the expression of CYP1B1 mRNA and protein is increased by Trx-1 transfection of MCF-7 human breast cancer cells and decreased by a redox inactive mutant Trx-1. The Trx-1 inhibitor PX-12 inhibits CYP1B1 gene expression. Trx-1 transfected MCF-7 cells show increased AhR/ARNT DNA binding activity that is not due to altered AhR or ARNT protein expression. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, dioxin) induced expression of CYP1B1 in MCF-7 cells is increased by Trx-1. Trx-1 does not effect the basal expression of CYP1A1, but increases CYP1A1 mRNA in response to TCDD. The redox inactive mutant Trx-1 completely blocks the induction of both CYP1B1 and CYP1A1 by TCDD. Expression of CYP1A1 but not CYP1B1 has been linked to estrogen receptor (ERalpha) status. Trx-1 transfected MCF-7 cells have decreased ERalpha expression, which may account for the lack of CYP1A1 induction by Trx-1 in the absence of ligand. The results suggest that Trx-1 is involved in the constitutive expression of CYP1B1 and is required for the induction of CYP1B1 and CYP1A1 by TCDD in human MCF-7 breast cancer cells. PMID: 12376470 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 113: Curr Drug Metab. 2002 Oct;3(5):481-90. Pharmacogenomics, regulation and signaling pathways of phase I and II drug metabolizing enzymes. Rushmore TH, Kong AN. Department of Drug Metabolism, Merck Research Laboratory, West Point, PA, USA. rushmore@merck.com Drug or xenobiotics metabolizing enzymes (DMEs or XMEs) play central roles in the biotransformation, metabolism and/or detoxification of xenobiotics or foreign compounds, that are introduced to the human body. In general, DMEs protect or defend the body against the potential harmful insults from the environment. Once in the body, many xenobiotics may induce signal transduction events either specifically or non-specifically leading to various cellular, physiological and pharmacological responses including homeostasis, proliferation, differentiation, apoptosis, or necrosis. For the body to minimize the insults caused by these xenobiotics, various tissues/organs are well equipped with diverse DMEs including various Phase I and Phase II enzymes, which are present in abundance either at the basal level and/or increased/induced after exposure. To better understand the pharmacogenomic/gene expression profile of DMEs and the underlying molecular mechanisms after exposure to xenobiotics or drugs, we will review our current knowledge on DNA microarray technology in gene expression profiling and the signal transduction events elicited by various xenobiotics mediated by either specific receptors or non-specific signal transduction pathways. Pharmacogenomics is the study of genes and the gene products (proteins) essential for pharmacological or toxicological responses to pharmaceutical agents. In order to assess the battery of genes that are induced or repressed by xenobiotics and pharmaceutical agents, cDNA microarray or oligonucleotide-based DNA chip technology can be a powerful tool to analyze, simultaneously, the gene expression profiles that are induced or repressed by xenobiotics. The regulation of gene expression of the various phase I DMEs such as the cytochrome P450 (CYP) as well as phase II DMEs generally depends on the interaction of the xenobiotics with the receptors. For instance, the expression of CYP1 genes can be induced via the aryl hydrocarbon receptor (AhR) which dimerizes with the AhR nuclear translocator (ARNT), in response to many polycyclic aromatic hydrocarbon (PAHs). Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). The peroxisome proliferator activated receptor (PPAR) which is one of the first characterized members of the nuclear hormone receptor, also dimerizes with RXR and it has been shown to be activated by lipid lowering agent fibrate-type of compounds leading to transcriptional activation of the promoters on the CYP4A genes. The transcriptional activation of these promoters generally leads to the induction of their mRNA. The physiological and the pharmacological implications of common partner of RXR for CAR, PXR, and PPAR receptors largely remain unknown and are under intense investigations. For the phase II DMEs, phase II gene inducers such as phenolic compounds butylated hydroxyanisol (BHA), tert-butylhydroquinone (tBHQ), green tea polyphenol (GTP), (-)-epicatechin-3-gallate (EGCG) and the isothiocyanates (PEITC, sulforaphane) generally appear to be electrophiles. They can activate the mitogen-activated protein kinase (MAPK) pathway via electrophilic-mediated stress response, resulting in the activation of bZIP transcription factors Nrf2 which dimerizes with Mafs and binds to the antioxidant/electrophile response element (ARE/EpRE) enhancers which are found in many phase II DMEs as well as many cellular defensive enzymes such as thioredoxins, gammaGCS and HO-1, with the subsequent induction of gene expression of these genes. It appears that in general, exposure to phase I or phase II gene inducers or xenobiotics may trigger a cellular "stress" response leading to the increase in the gene expression of these DMEs, which ultimately enhance the elimination and clearance of the xenobiotics e xenobiotics and/or the "cellular stresses" including harmful reactive intermediates such as reactive oxygen species (ROS), so that the body will remove the "stress" expeditiously. Consequently, this homeostatic response of the body plays a central role in the protection of the organism against environmental insults such as xenobiotics. Advances in DNA microarray technologies and mammalian genome sequencing will soon allow quantitative assessment of expression profiles of all genes in the selected tissues. The ability to predict phenotypic outcomes from gene expression profiles is currently in its infancy, however, and will require additional bioinformatic tools. Such tools will facilitate information gathering from literature and gene databases as well as integration of expression data with animal physiology studies. The study of pharmacogenomic/gene expression profile and the understanding of the regulation and the signal transduction mechanisms elicited by pharmaceutical agents can be of potential importance during drug discovery and the drug development. Publication Types: Review Review, Tutorial PMID: 12369894 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 114: J Biol Chem. 2002 Nov 29;277(48):46576-85. Epub 2002 Sep 26. The aryl hydrocarbon receptor nuclear transporter is modulated by the SUMO-1 conjugation system. Tojo M, Matsuzaki K, Minami T, Honda Y, Yasuda H, Chiba T, Saya H, Fujii-Kuriyama Y, Nakao M. Department of Regeneration Medicine, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan. The aryl hydrocarbon receptor nuclear transporter (ARNT) is a member of the basic helix-loop-helix/PAS (Per-ARNT-Sim) family of transcription factors, which are important for cell regulation in response to environmental conditions. ARNT is an indispensable partner of the aryl hydrocarbon receptor (AHR) or hypoxia-inducible factor-1alpha. This protein is also able to form homodimers such as ARNT/ARNT. However, the molecular mechanism that regulates the transcriptional activity of ARNT remains to be elucidated. Here, we report that ARNT is modified by SUMO-1 chiefly at Lys(245) within the PAS domain of this protein, both in vivo and in vitro. Substitution of the target lysine with alanine enhanced the transcriptional potential of ARNT per se. Furthermore, green fluorescent protein-fused ARNT tended to form nuclear foci in approximately 20% of the transfected cells, and the foci partly colocalized with PML nuclear bodies. PML, one of the well known substrates for sumoylation, was found to augment the transcriptional activities of ARNT. ARNT bound AHR or PML, whereas the sumoylated form of ARNT associated with AHR, but not with PML, resulting in a reduced effect of PML on transactivation by ARNT. Our data suggest that the sumoylation of ARNT modulates its transcriptional role through affecting the ability of ARNT to interact with cooperative molecules such as PML. This exemplifies a crucial role of protein sumoylation in modulating protein-protein interactions. PMID: 12354770 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 115: Mol Pharmacol. 2002 Oct;62(4):806-16. Ligand-dependent and independent modulation of aryl hydrocarbon receptor localization, degradation, and gene regulation. Song Z, Pollenz RS. Department of Biology, University of South Florida, Tampa, Florida, USA. Changes in the concentration or subcellular location of the key proteins involved in signal transduction pathways have been shown to impact gene regulation. Studies were designed to evaluate the relationship between aryl hydrocarbon receptor (AHR) localization, stability, and gene regulation in a defined system where the endogenous AHR protein could be evaluated. The findings indicate that treatment of cells with geldanamycin (GA) or MG-132 (an inhibitor of the 26S proteasome) results in nuclear translocation of the endogenous AHR in both human HepG2 and murine Hepa-1 cells without induction of endogenous CYP1A1 protein. Exposure to GA resulted in the degradation of AHR by >90% in the nucleus via the 26S proteasome. Importantly, the reduced level of AHR resulted in a 50% reduction in the maximal level of CYP1A1 induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). In all treatments the concentration of the AHR nuclear translocator (ARNT) protein was unchanged and had no impact on the localization of the AHR. Thus, ligand-independent translocation of the AHR to the nucleus was not sufficient to induce CYP1A1 in the absence of ligand, but reductions in the level of the endogenous AHR protein pool shifted the dose-response curve for TCDD to the right. PMID: 12237327 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 116: Comp Biochem Physiol B Biochem Mol Biol. 2002 Sep;133(1):127-34. Estrous cycle-regulated expression of CYP1B1 mRNA in the rat ovary. Dasmahapatra AK, Trewin AL, Hutz RJ. Department of Biological Sciences, NIEHS Marine and Freshwater Biomedical Sciences Center, 308 Lapham Hall, 3209 North Maryland Avenue, University of Wisconsin, Milwaukee, WI, USA. CYP1B1, a member of the cytochrome p450 superfamily, is expressed constitutively in the steroidogenic tissues of mammals and is inducible by peptide hormones, cAMP and aromatic hydrocarbon receptor (AHR) ligands. The mechanism of induction of this cytochrome p450 is similar to that for CYP1A1, i.e. through the aromatic hydrocarbon receptor (AHR) signaling pathway. We have recently reported that CYP1B1, but not CYP1A1, is expressed in rat granulosa cells (GC) in the absence of any external stimulus. The induction of CYP1B1 mRNA in rat GC by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in vitro was followed by an increase in AHR and estrogen receptor (ER-beta) RNA levels. Estrous cycle-dependent expression of AHR, AHR-nuclear translocator (ARNT) and ER-mRNAs in the rat ovary was reported. We suggest that CYP1B1 may play a major role in the regulation of rat ovarian function/cycle but until now this has been unexplored experimentally. The present study was therefore aimed at examining the expression of CYP1A1, CYP1B1 and ER-mRNA in rat ovarian tissues throughout the estrous cycle to establish any correlation in the expressions of these mRNAs in rat ovary. Total RNA was extracted from the ovary and liver of cycling adult rats and the mRNAs were analyzed using relative RT-PCR with gene-specific primers for the target mRNA and for RPL 19 or S16 primers as an internal control. The results indicated that in the ovary, CYP1B1 mRNA increased significantly on the evening of proestrus and dramatically decreased on the morning of estrus, while ER-mRNA remained unaltered throughout the estrous cycle. CYP1A1 mRNA in the ovary and both CYP1A1 and CYP1B1 mRNAs in the liver were undetectable. That the sudden decrease of ovarian CYP1B1 mRNA on the morning of estrus was not an effect of the LH surge was verified in vitro using our short-term GC culture model. GC prepared from rats super-stimulated with equine chorionic gonadotropin (eCG) were cultured for 6 h with or without LH and TCDD. It was observed that both CYP1A1 and CYP1B1 mRNAs were induced by TCDD with no apparent effect of LH. It is suggested that the high level of CYP1B1 mRNA expression on the evening of proestrus in rat ovary might be involved in metabolism of estrogens to catecholestrogen (a known effect of CYP1B1), and that expression is unaffected in GC by LH. PMID: 12223220 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 117: Toxicol Sci. 2002 Sep;69(1):117-24. 2,3,7,8-tetrachlorodibenzo-p-dioxin causes alterations in lymphocyte development and thymic atrophy in hemopoietic chimeras generated from mice deficient in ARNT2. Laiosa MD, Lai ZW, Thurmond TS, Fiore NC, DeRossi C, Holdener BC, Gasiewicz TA, Silverstone AE. Department of Microbiology and Immunology, State University of New York, Upstate Medical University, 750 East Adams Street, Syracuse, New York 13210, USA. It is well established that dioxins cause a variety of toxic effects and syndromes including alterations of lymphocyte development. Exposure to the prototypical dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) leads to severe thymic atrophy in all species studied. It has been shown that most of this toxicity is due to TCDD binding to and activating the aryl hydrocarbon receptor (AHR). Upon activation, the AHR enters the nucleus, dimerizes with the AHR nuclear translocator (ARNT), and this heterodimer modulates a number of genes that mediate toxicity. The AHR and ARNT are members of the basic-helix-loop-helix-Per, ARNT, and Sim homology (bHLH-PAS) family of transcription factors. In this study, we wanted to determine if another bHLH-PAS transcription factor, ARNT2, which has high amino acid sequence identity to ARNT and has been shown to dimerize with the TCDD-activated AHR, is involved in mediating TCDD's effect on lymphocyte development. We determined by RT-PCR that ARNT2 is expressed at a low level in whole thymus, thymocytes, and bone marrow lymphocytes. We created hemopoietic chimeras by lethally irradiating C57BL/6 mice and reconstituting them with fetal liver stem cells that either have or are deficient in a portion of chromosome 7 that contains ARNT2. Regardless of whether chimeras possessed or lacked this chromosome fragment, equal sensitivity to TCDD-induced thymic atrophy was observed despite expression of ARNT2 in the thymus. Furthermore, the absence of ARNT2 (or any other genes found on this portion of chromosome 7) did not confer any protection against TCDD-induced alterations in bone marrow B-cell subsets. These data indicate that in this model system the effects of TCDD-induced thymic atrophy and alterations in B-cell maturation are not dependent on an AHR-ARNT2 heterodimer. PMID: 12215665 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 118: Chem Biol Interact. 2002 Sep 20;141(1-2):63-76. DNA binding and protein interactions of the AHR/ARNT heterodimer that facilitate gene activation. Swanson HI. Department of Molecular and Biomedical Pharmacology, University of Kentucky Medical Center, MS 303, Lexington 40536, USA. hswan@uky.edu Gene activation by the aryl hydrocarbon receptor (AHR) and its DNA binding partner, the aryl hydrocarbon receptor nuclear translocator (ARNT) requires a number of sequential steps that occur following the binding of ligand and entry of the AHR into the nuclear compartment. This includes heterodimerization of the AHR and ARNT, formation of the appropriate amino acid/nucleotide contacts at the GCGTG recognition site and interactions between either the AHR or ARNT with proteins that facilitate changes in chromatin structure. The majority of these steps are likely modulated by changes in both phosphorylation and oxidation status of the AHR, ARNT and associated proteins. Studies of both the basic helix-loop-helix transcription factors and the nuclear hormone receptor family can provide significant insights into how this unique signaling pathway activates its target genes. Publication Types: Review Review, Tutorial PMID: 12213385 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 119: Chem Biol Interact. 2002 Sep 20;141(1-2):41-61. The mechanism of AH receptor protein down-regulation (degradation) and its impact on AH receptor-mediated gene regulation. Pollenz RS. Department of Biology, University of South Florida, 4202 E Fowler Avenue, SCA 110, Tampa 33620-5200, USA. pollenz@chuma1.cas.usf.edu The proteolytic degradation of transcription factors is an established mechanism of regulating signal transduction pathways. Recent reports have suggested that the aryl hydrocarbon receptor (AHR) protein is rapidly downregulated (degraded) following ligand binding. The downregulation of AHR has been observed in nine distinct cells culture lines derived from human and rodent tissues and has also been observed in rodent models following exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The downregulation of AHR appears to be ubiquitin mediated and occurs via the 26S proteasome pathway following nuclear export of AHR. The consequence of blocking AHR degradation in cell culture appears to be an increase in both the magnitude and duration of gene regulation by the AHR.ARNT complex. Thus, the physiological role of AHR degradation may be to modulate AHR-mediated gene regulation. This review provides analysis of the studies that have focused on the degradation of AHR in vivo and in vitro and the hypothesis that the downregulation of AHR is critical in the attenuation of AHR-mediated gene regulation. Publication Types: Review Review, Tutorial PMID: 12213384 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 120: Chem Biol Interact. 2002 Sep 20;141(1-2):25-40. The role of chaperone proteins in the aryl hydrocarbon receptor core complex. Petrulis JR, Perdew GH. Department of Veterinary Science, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park 16802, USA. The aryl hydrocarbon receptor (AhR) exists in the absence of a ligand as a tetrameric complex composed of a 95-105 kDa ligand binding subunit, a dimer of hsp90, and the immunophilin-like X-associated protein 2 (XAP2). XAP2 has a highly conserved carboxy terminal tetratricopeptide repeat domain that is required for both hsp90 and AhR binding. Hsp 90 appears to be involved in the initial folding of newly synthesized AhR, stabilization of ligand binding conformation of the receptor, and inhibition of constitutive dimerization with ARNT. XAP2 is capable of stabilizing the AhR, as well as enhancing cytoplasmic localization of the receptor. XAP2 binds to both the AhR and hsp90 in the receptor complex, and is capable of independently binding to both hsp90 and the AhR. However, the exact functional role for XAP2 in the AhR complex remains to be fully established. Publication Types: Review Review, Tutorial PMID: 12213383 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 121: Arch Biochem Biophys. 2002 Aug 15;404(2):309-16. Erratum in: Arch Biochem Biophys. 2003 Sep 1;417(1):129. Induction and superinduction of 2,3,7,8-tetrachlorodibenzo-rho-dioxin-inducible poly(ADP-ribose) polymerase: role of the aryl hydrocarbon receptor/aryl hydrocarbon receptor nuclear translocator transcription activation domains and a labile transcription repressor. Ma Q. Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA. qam1@cdc.gov The environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces a novel poly(ADP-ribose) polymerase (TiPARP). In this study, the signaling pathway of the induction was analyzed. Induction of TiPARP by TCDD occurs in both hepa1c1c7 cells and C57 mouse liver. Induction is concentration and time dependent. Genetic analyses reveal that induction is abolished in aromatic hydrocarbon receptor (AhR)- or aromatic hydrocarbon receptor nuclear translocator (Arnt)-defective variants but restored upon reconstitution of the variant cells with cDNAs expressing functional AhR or Arnt. Moreover, induction is largely reduced in cells expressing a deletion mutant of AhR or Arnt lacking the transcription activation (TA) domain, thus implicating the TA activities of both AhR and Arnt in the induction. Inhibition of protein synthesis by cycloheximide enhances the induction of TiPARP in the presence of an AhR agonist. The superinduction is transcriptional and does not require pretreatment with TCDD. Finally, inhibition of the 26S proteasomes by MG132 superinduces TiPARP. These findings establish that induction of TiPARP by TCDD is mediated through an AhR and Arnt transcription activation-dependent signal transduction that is repressed by a labile factor through the ubiquitin-26S proteasome-mediated protein degradation. Copyright 2002 Elsevier Science (USA). PMID: 12147270 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 122: Arch Biochem Biophys. 2002 Jul 15;403(2):189-201. The silencing mediator of retinoic acid and thyroid hormone receptors can interact with the aryl hydrocarbon (Ah) receptor but fails to repress Ah receptor-dependent gene expression. Rushing SR, Denison MS. Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA. Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related chemicals causes a variety of tissue- and species-specific biological and toxicological effects, most of which are mediated by the aryl hydrocarbon receptor (AhR). The AhR complex is a ligand-dependent transcription factor that binds to its specific DNA recognition site as a dimer with the AhR nuclear translocator (ARNT) and activates gene transcription. Here, we have examined the ability of a nuclear corepressor, the silencing mediator of retinoic acid and thyroid hormone receptors (SMRT), to interact with and modulate AhR-dependent gene expression. Using glutathione S-transferase (GST) "pull-down" binding assays, we have mapped a major interaction between these factors to the silencing domain of SMRT and the PAS B ligand binding domain of AhR, and this interaction is unaffected by the addition of an AhR ligand. Association of SMRT with the AhR:ARNT:DNA complex was not detected by GST pull-down or gel retardation assays. Transient cotransfections of mammalian cells (Hepa1c1c7, MCF-7, and BG-1) with SMRT and a TCDD-inducible luciferase reporter containing the dioxin-responsive domain from the mouse CYP1A1 regulatory region revealed that SMRT does not repress, but enhances, AhR signaling. However, when a reporter containing a human CYP1A1 upstream region was cotransfected with SMRT into human MCF-7 cells, AhR-driven reporter activity was decreased by half, suggesting that SMRT acts on the human CYP1A1 promoter via a factor other than the AhR in MCF-7 cells. Furthermore, the interaction between SMRT and the AhR may have implications in pathways other than the AhR signaling pathway. PMID: 12139968 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 123: Reprod Toxicol. 2002 May-Jun;16(3):299-307. Alteration in ovarian gene expression in response to 2,3,7,8-tetrachlorodibenzo-p-dioxin: reduction of cyclooxygenase-2 in the blockage of ovulation. Mizuyachi K, Son DS, Rozman KK, Terranova PF. Center of Reproductive Sciences and Department of Molecular & Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160-7417, USA. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a reproductive toxicant and endocrine disrupter that is known to block ovulation. This study was designed to investigate alterations in relevant ovarian genes that may be involved in the blockage of ovulation by TCDD in immature intact rats primed with equine chorionic gonadotropin (eCG). In this ovulation model, rats were given either 32 microg/kg TCDD or corn oil by gavage on 25 days of age. The next day, eCG (5 IU) was injected subcutaneously (s.c.) to stimulate follicular development. Ovulation occurs 72 h after administration of eCG in controls of this model. TCDD blocked ovulation at the expected time and also reduced both ovarian and body weights. At 72 h after eCG (the morning after expected ovulation), TCDD did not alter significantly serum concentrations of progesterone (P4) and androstenedione (A4). However, estradiol (E2) was significantly higher at 72 h after eCG in TCDD-treated rats when compared with controls. Western blots revealed that ovarian CYP1A1 was induced by TCDD. In addition, the aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (ARNT) were down- and up-regulated by TCDD, respectively, indicating that AhR-mediated signal transduction was altered in the ovary. Ovarian estrogen receptor (ER)alpha, ER beta and progesterone receptor (PR) were not altered significantly by TCDD, but ovarian glucocorticoid receptor (GR) was increased at 24h after TCDD and decreased at 72 h after eCG when compared with controls. TCDD induced the early appearance of ovarian plasminogen activator inhibitor type-1 (PAI-1), plasminogen activator inhibitor type-2 (PAI-2), urokinase plasminogen activator (uPA), and tissue plasminogen activator (tPA) at 24h after dosing when compared with controls. On the morning after ovulation (72 h after eCG), no significant differences between control and TCDD-treated rats were observed except that TCDD had still increased tPA and decreased PAI-2 when compared with controls. Interestingly, ovarian COX-2 was induced on the morning after ovulation (72 h after eCG) in controls, but was greatly inhibited in TCDD-treated rats at that time. On the other hand, COX-1 was constitutively expressed throughout the ovulatory period and remained unaffected by TCDD. Immunolocalization of COX-2 in the ovary revealed that TCDD inhibited COX-2 expression in the granulosa cell layer when assessed in the morning of expected ovulation. In conclusion, AhR signaling is activated in the ovary by TCDD and inhibition of COX-2 appeared to be a critical step in the TCDD blockage of ovulation because blockage or reduction of COX-2 expression is well known to be associated with failure of ovulation. PMID: 12128104 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 124: Arch Toxicol. 2002 Jul;76(7):404-13. Epub 2002 May 16. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces plasminogen activator inhibitor-1 through an aryl hydrocarbon receptor-mediated pathway in mouse hepatoma cell lines. Son DS, Rozman KK. Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA. 2,3,7,8-Tetrachlorodibenzo- p-dioxin (TCDD), a ubiquitous environmental pollutant, elicits a variety of toxicities and is a well-known carcinogen. TCDD alters the expression of many genes including CYP1A1/2, CYP1B1, glutathione S-transferase Ya, aldehyde-3-dehydrogenase, NAD(P)H:quinone oxidoreductase, transforming growth factor (TGF)-alpha and TGF-beta. The present study was aimed at characterization of TCDD to induce plasminogen activator inhibitor-1 (PAI-1) in mouse hepatoma cell lines. A Hepa1c1c7 wild-type cell [H1(wt)], an aryl hydrocarbon receptor (AhR)-deficient mutant [H1(AhR(-))] and an AhR nuclear translocator (Arnt)-deficient mutant [H1(Arnt(-))] were used for this study. TCDD induced PAI-1 in H1(wt) cells, but not in H1(AhR(-)) and H1(Arnt(-)) mutants, indicating a functional role of the AhR-Arnt complex in this effect. Cycloheximide (CHX) treatment resulted in increased PAI-1 mRNA induction, indicating that this response to TCDD is a direct effect on transcription and not a secondary effect mediated by other TCDD-induced proteins. Transfection with PAI-1 promoter led to increased PAI-1 promoter activity in H1(wt) cells treated with TCDD, but no such effect occurred in H1(AhR(-)) or H1(Arnt(-)) cells, implying involvement of the AhR and Arnt. In addition, alpha-naphthoflavone and phenanthroline, two AhR antagonists, each blocked the enhancing effect of TCDD on PAI-1 promoter-coupled luciferase activity in H1(wt) cells. PAI-1 promoter deletion analysis indicated that TCDD-induced PAI-1 transcription was distinctly different from TGF-beta-dependent PAI-1 transcription, particularly in the region between -161 to +73. In summary, TCDD induced the PAI-1 gene directly via an AhR- and Arnt-dependent mechanism, which was distinctly different from TGF-beta-driven PAI-1 transcription. PMID: 12111005 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 125: Biochem Pharmacol. 2002 Jul 1;64(1):49-60. Analysis of rainbow trout Ah receptor protein isoforms in cell culture reveals conservation of function in Ah receptor-mediated signal transduction. Pollenz RS, Necela B, Marks-Sojka K. Department of Biology, University of South Florida, 4202 East Fowler Avenue, SCA 110, Tampa, FL 33620-5200, USA. Two distinct aryl hydrocarbon receptor (AHR) cDNAs have been isolated from rainbow trout. The encoded receptor protein products termed rtAHR2alpha and rtAHR2ss are 97% identical at the amino acid level but are reported to have distinct functions with regard to AHR-mediated gene regulation. To test this hypothesis, the two proteins were evaluated functionally both in vitro and in a Chinese hamster lung cell line, E36. To facilitate analysis, both rtAHR2 isoforms were tagged with the FLAG peptide and could be expressed and quantified in a rabbit reticulocyte lysate. However, both proteins failed to form functional complexes with mammalian or rainbow trout AHR nuclear translocator protein (ARNT) that could associate with xenobiotic response elements (XREs) in a ligand-dependent manner in vitro. In contrast, both proteins exhibited positive function on AHR-mediated signaling when expressed in the E36 cell line. Both rtAHR2 isoforms showed a cytoplasmic distribution in the unliganded state and could drive the expression of a reporter gene under control of the trout CYP1A3 promoter. Although both proteins induced reporter gene activity to the same magnitude, the EC(50) values of the two isoforms for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) differed by an order of magnitude, with the rtAHR2ss isoform less responsive to TCDD. When the functions of the rtAHR2 isoforms were tested in the context of the dominant negative rtARNT(a) protein, TCDD-mediated induction of reporter gene activity was reduced as the level of rtARNT(a) protein increased. In summary, both rtAHR2 isoforms appear to exhibit positive function in AHR-mediated signaling, suggesting conservation of function. PMID: 12106605 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 126: Int J Dev Biol. 2002 May;46(3):295-300. Expression of the mediators of dioxin toxicity, aryl hydrocarbon receptor (AHR) and the AHR nuclear translocator (ARNT), is developmentally regulated in mouse teeth. Sahlberg C, Pohjanvirta R, Gao Y, Alaluusua S, Tuomisto J, Lukinmaa PL. Department of Pedodontics and Orthodontics, Institute of Dentistry, University of Helsinki, Finland. carin.sahlberg@helsinki.fi Dioxins are persistent and ubiquitous environmental poisons that become enriched in the food chain. Besides being acutely lethal, the most toxic dioxin congener, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is developmentally toxic to many animal species. We have previously found that developing teeth of children may be sensitive to environmental dioxins via their mother's milk and that rat and mouse teeth are dioxin-sensitive throughout their development. The aryl hydrocarbon receptor (AHR) together with the AHR nuclear translocator (ARNT) protein is believed to mediate the toxic effects of dioxins. To study the potential involvement of the AHR-ARNT pathway in the dental toxicity of TCDD, we analysed the expression of AHR and ARNT by in situ hybridization and immunohistochemistry in developing mouse teeth. AHR mRNA first appeared in the epithelium of E12 first molar tooth buds and both proteins were weakly expressed in the bud. After cytodifferentiation the expression was up regulated and became intense in secretory odontoblasts and ameloblasts. The coexpression of AHR and ARNT during early tooth development as well as during the information and mineralization of the dental matrices is suggestive of the AHR-ARNT pathway as a mediator of dental toxicity of TCDD. PMID: 12068950 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 127: Aquat Toxicol. 2002 Jul;58(1-2):57-73. cDNA cloning and characterization of an aryl hydrocarbon receptor from the harbor seal (Phoca vitulina): a biomarker of dioxin susceptibility? Kim EY, Hahn ME. Biology Department, MS 32, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related planar halogenated aromatic hydrocarbons (PHAHs) are found at high concentrations in some marine mammals. Species differences in sensitivity to TCDD and PHAHs are a major limitation in assessing the ecological risk to these animals. Harbor seals accumulate high levels of PHAHs and are thought to be highly sensitive to the toxic effects of these compounds. To investigate the mechanistic basis for PHAH toxicity in harbor seals (Phoca vitulina), we sought to characterize the aryl hydrocarbon receptor (AHR), an intracellular protein that is responsible for PHAH effects. Here we report the cDNA cloning and characterization of a harbor seal AHR. The harbor seal AHR cDNA has an open reading frame of 2529 nucleotides that encodes a protein of 843 amino acids with a predicted molecular mass of 94.6 kDa. The harbor seal AHR protein possesses basic helix-loop-helix (bHLH) and Per-ARNT-Sim (PAS) domains. It is most closely related to the beluga AHR (82%) and human AHR (79%) in overall amino acid identity, indicating a high degree of conservation of AHR structure between terrestrial and some marine mammals. The ligand binding properties of the harbor seal AHR were determined using protein synthesized by in vitro transcription and translation from the cloned cDNA. Velocity sedimentation analysis on sucrose gradients showed that the harbor seal AHR exhibits specific binding of [(3)H]TCDD. The [(3)H]TCDD-binding affinity of the harbor seal AHR was compared with that of the AHR from a dioxin-sensitive mouse strain (C57BL/6) using a hydroxylapatite assay. The equilibrium dissociation constants of seal and mouse AHRs were 0.93+/-0.19 and 1.70+/-0.26 nM, respectively. Thus, the harbor seal AHR bound TCDD with an affinity that was at least as high as that of the mouse AHR, suggesting that this seal species may be sensitive to PHAH effects. The characteristics of the AHR potentially can be used as a biomarker of susceptibility to dioxin-like compounds, contributing to the assessment of the risk of these compounds to marine mammals and other protected animals. PMID: 12062155 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 128: Mol Biol Cell. 2002 Jun;13(6):2001-15. Recruitment of dioxin receptor to active transcription sites. Elbi C, Misteli T, Hager GL. Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-5055, USA. The aryl hydrocarbon receptor (AhR or dioxin receptor) is a ligand-activated transcription factor that heterodimerizes with the AhR nuclear translocator (ARNT/HIF-1beta) to form an AhR/ARNT transcription factor complex. This complex binds to specific DNA sites in the regulatory domains of numerous target genes and mediates the biological effects of exogenous ligands. Herein, we have investigated the subcellular distribution of the AhR/ARNT complex in response to ligand stimulation, by using live-cell confocal and high-resolution deconvolution microscopy. We found that unliganded AhR shows a predominantly cytoplasmic diffuse distribution in mouse hepatoma cells. On addition of ligand, AhR rapidly translocates to the nucleus and accumulates in multiple bright foci. Inhibition of transcription prevented the formation of AhR foci. Dual- and triple-immunolabeling experiments, combined with labeling of nascent RNA, showed that the foci are transcription sites, indicating that upon ligand stimulation, AhR is recruited to active transcription sites. The interaction of AhR with ARNT was both necessary and sufficient for the recruitment of AhR to transcription sites. These results indicate that AhR/ARNT complexes are recruited to specific subnuclear compartments in a ligand-dependent manner and that these foci represent the sites of AhR target genes. PMID: 12058065 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 129: J Hum Genet. 2002;47(5):217-24. Variability of the human aryl hydrocarbon receptor nuclear translocator (ARNT) gene. Scheel J, Hussong R, Schrenk D, Schmitz HJ. Food Chemistry and Environmental Toxicology, University of Kaiserslautern, Kaiserslautern, Germany. scheel@axaron.com The aryl hydrocarbon receptor nuclear translocator (ARNT) plays an essential role in vertebrate transcriptional regulation as the common subunit of transcriptionally active complexes like the aryl hydrocarbon receptor (AHR)/ARNT heterodimer and hypoxia-inducible factor 1, mediating cellular responses to certain xenobiotics and to hypoxia, respectively. A cohort of healthy Caucasian volunteers was screened for genetic variations of ARNT. Six polymorphic sites could be identified, a variation in a G-stretch upstream of the ATG translation start site, a frequent silent mutation (G567C), two polymorphic sites in intron 9, and two single nucleotide substitutions leading to amino acid exchanges, G1531A (D511N) and T1551G (D517E). The frequencies were 0.005 for the Asn-coding allele and for the Glu-coding allele, respectively, with no linkage between these two mutations. Although no significant correlation with activities of CYP1A2, which is under regulatory control of the AHR/ARNT transcription complex, could be established, metabolic or pathological phenotypes may be associated with these variations. PMID: 12032587 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 130: Mol Cell Biol. 2002 Jun;22(12):4319-33. Recruitment of the NCoA/SRC-1/p160 family of transcriptional coactivators by the aryl hydrocarbon receptor/aryl hydrocarbon receptor nuclear translocator complex. Beischlag TV, Wang S, Rose DW, Torchia J, Reisz-Porszasz S, Muhammad K, Nelson WE, Probst MR, Rosenfeld MG, Hankinson O. Department of Pathology and Laboratory Medicine, Molecular Biology Institute, and Jonsson Comprehensive Cancer Center, School of Medicine, University of California-Los Angeles, Los Angeles, California 90095, USA. The aryl hydrocarbon receptor complex heterodimeric transcription factor, comprising the basic helix-loop-helix-Per-ARNT-Sim (bHLH-PAS) domain aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) proteins, mediates the toxic effects of TCDD (2,3,7,8 tetrachlorodibenzo-p-dioxin). The molecular events underlying TCDD-inducible gene activation, beyond the activation of the AHRC, are poorly understood. The SRC-1/NCoA-1, NCoA-2/GRIP-1/TIF-2, and p/CIP/AIB/ACTR proteins have been shown to act as mediators of transcriptional activation. In this report, we demonstrate that SRC-1, NCoA-2, and p/CIP are capable of independently enhancing TCDD-dependent induction of a luciferase reporter gene by the AHR/ARNT dimer. Furthermore, injection of anti-SRC-1 or anti-p/CIP immunoglobulin G into mammalian cells abolishes the transcriptional activity of a TCDD-dependent reporter gene. We demonstrate by coimmunoprecipitation and by a reporter gene assay that SRC-1 and NCoA-2 but not p/CIP are capable of interacting with ARNT in vivo after transient transfection into mammalian cells, while AHR is capable of interacting with all three coactivators. We confirm the interactions of ARNT and AHR with SRC-1 with immunocytochemical techniques. Furthermore, SRC-1, NCoA-2, and p/CIP all associate with the CYP1A1 enhancer region in a TCDD-dependent fashion, as demonstrated by chromatin immunoprecipitation assays. We demonstrate by yeast two-hybrid, glutathione S-transferase pulldown, and mammalian reporter gene assays that ARNT requires its helix 2 domain but not its transactivation domain to interact with SRC-1. This indicates a novel mechanism of action for SRC-1. SRC-1 does not require its bHLH-PAS domain to interact with ARNT or AHR, but utilizes distinct domains proximal to its p300/CBP interaction domain. Taken together, these data support a role for the SRC family of transcriptional coactivators in TCDD-dependent gene regulation. PMID: 12024042 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 131: Chemosphere. 2002 Mar;46(9-10):1491-500. A cycloheximide-sensitive factor regulates TCDD-induced degradation of the aryl hydrocarbon receptor. Ma Q, Baldwin KT. Molecular Toxicology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA. qam1@cdc.gov 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), a prototype of environmental halogenated aromatic hydrocarbons, induces a rapid reduction in steady state aryl hydrocarbon receptor (AhR). Here, we analyzed the biochemical pathway and function of the downregulation. Our results reveal that TCDD downregulates the AhR protein by shortening the halflife of AhR. The TCDD-induced degradation of AhR is inhibited by MG132, a potent inhibitor of the 26S proteasome, indicating the ubiquitin-26S proteasome mediated proteolysis as a mechanism for the degradation of AhR. Furthermore, inhibition of protein synthesis by cycloheximide blocks the degradation of AhR by TCDD, suggesting a labile factor in controlling the stability of ligand-activated AhR (hence, designated as AhR degradation promoting factor, or ADPF). Analyses of nuclear AhR demonstrated that cycloheximide increases nuclear AhR protein and functional AhR/Arnt DNA-binding complex, resulting in superinduction of CYP1A1. Lastly, genetic analyses by using AhR- or Arnt-defective variant cells demonstrate that superinduction by cycloheximide requires the transcription activation (TA) domain of AhR, implicating the TA domain in the control of AhR turnover by ADPF. These findings provide new insights into the mechanism by which TCDD-activated AhR is regulated in nucleus through the 26S proteasome protein degradation pathway. PMID: 12002481 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 132: Toxicology. 2002 May 24;174(2):119-29. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on preimplantation mouse embryos. Wu Q, Ohsako S, Baba T, Miyamoto K, Tohyama C. Environmental Health Sciences Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent environmental contaminant that can exert developmental toxicity. To investigate the stage-specific effects of TCDD on preimplantation embryos, we exposed mouse embryos to TCDD at different stages (1-, 2-, and 8-cell) and collected them at different stages of development (the 1- or 2-, 8-cell, and blastocyst stage, respectively). Semiquantitative RT-PCR revealed increased constitutive gene expression of the arylhydrocarbon receptor (AhR) and AhR nuclear translocator (Arnt) at the 1-cell stage, decreased expression at the 2- to 8-cell stage, and increased expression again at the blastocyst stage, and addition of TCDD to media did not affect their mRNA levels. Interestingly, no cytochrome P4501A1 (CYP1A1) mRNA was detected in embryos at the 1-, 2-, and 8-cell stages after exposure to 10 nM TCDD for 12 or 24 h, whereas CYP1A1 mRNA was significantly increased at the blastocyst stage in response to TCDD, and its induction was found to be concentration-dependent on TCDD exposure from 0.01 to 10 nM for 24 h. In addition, no significant differences in development rate of preimplantation embryos, cell number of blastocyst embryos, or apoptotic indices, such as TUNEL-positive cell number or Bax/Bcl-2 expression ratios were observed at the blastocyst stage between TCDD-exposed groups and non-exposed group. These results suggest that the sensitivity to TCDD differs with the embryonic stage, which may reflect an ability of embryos to adapt to environmental stressors, such as dioxins. PMID: 11985889 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 133: Genesis. 2002 Mar;32(3):231-9. The aryl hydrocarbon receptor (AhR) and its nuclear translocator (Arnt) are dispensable for normal mammary gland development but are required for fertility. Le Provost F, Riedlinger G, Hee Yim S, Benedict J, Gonzalez FJ, Flaws J, Hennighausen L. Laboratory of Genetics and Physiology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0822, USA. The aryl hydrocarbon receptor (AhR) and its nuclear translocator (Arnt) are transcription factors that play a role in the detection of and adaptation to environmental signals. AhR-null mice are viable but show impaired lactation. Deletion of the Arnt gene from the mouse genome results in embryonic lethality. To determine the role of Arnt in mammary development and function, we inactivated the Arnt gene in mammary epithelium using Cre-loxP recombination. Inactivation of the Arnt gene during pregnancy did not disrupt alveolar development or the ability of dams to nurse their litters. In contrast, dams in which the Arnt gene had been inactivated during puberty and in ovaries were subfertile, exhibited retarded mammary development, and impaired mammary function. To distinguish defects autonomous to mammary epithelium from indirect effects controlled by ovarian hormones, we transplanted Arnt-null and AhR-null mammary epithelium into wild-type mice and evaluated development after one pregnancy. Normal mammary structures were observed in the absence of Arnt and AhR, demonstrating that neither transcription factor is necessary for mammary development. Copyright 2002 Wiley-Liss, Inc. PMID: 11892012 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 134: Int J Mol Med. 2002 Apr;9(4):411-6. The environmental toxin 2,3,7,8-tetrachlorodibenzo-p-dioxin induces cytochrome P450 activity in high passage PC 3 and DU 145 human prostate cancer cell lines. Schaufler K, Haslmayer P, Jager W, Pec M, Thalhammer T. Department of Pathophysiology, University of Vienna, A-1090 Vienna, Austria. The study was conducted to investigate whether 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces cytochrome P450 (CYP) 1A1 and CYP1B1 via the aryl hydrocarbon receptor (AhR) in the hormone-independent human prostate cancer cell lines PC 3 and DU 145. No quantitative differences in the expression of AhR and its partner transcription factor ARNT were seen in low and high passage number PC 3 and DU 145 cells in the absence and presence of TCDD as assessed by RT-PCR and Western blotting. However, CYP1A1/1B1 activity, measured by the 7-ethoxyresorufin-O-deethylase (EROD) assay, was induced by 10 and 100 nM TCDD only in high passage number PC 3 and DU 145 cells (PC 3, 7.7- and 2-fold stimulation; DU 145, 8.5- and 19.7-fold stimulation). Besides stimulation of EROD activity, induction of the expression of CYP1A1 and, to a lesser extend, of CYP1B1 by TCDD was also demonstrated by RT-PCR and Western blotting. However, 1-100 nM TCDD did not significantly alter cell cycle distribution and cell growth for up to five days. The induction of CYP1A1 and CYP1B1 by TCCD in the hormone-independent prostate cancer cell lines suggests that CYP induction should be considered in patients with advanced prostate cancer. This could result in higher elimination rates of concomitant drugs metabolized by these particular CYP isoenzymes. PMID: 11891538 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 135: Nippon Eiseigaku Zasshi. 2002 Jan;56(4):603-5. [Proteins in response to environmental stress] [Article in Japanese] Fujita H. Laboratory of Environmental Biology, Hokkaido University School of Medicine, North 15, West 7, Kita-ku, Sapporo 060-8638, Japan. alfujita@med.hokudai.ac.jp Last decade, remarkable progress was achieved in the field of environmental sciences, especially those related to biological functions against environmental stress. Recent progress has been focused on the transcriptional control of proteins, such as cytochrome P450, UDP-glucronosyltransferase, and metallothionein. Along with these studies, entirely new functions of some proteins were identified; for example heme oxygenase-1, originally isolated by the catalytic activity of heme, is nowadays recognized as one of the stress proteins. We are, therefore, interested in the relationship among proteins of body defense mechanisms; i.e., co-regulation of two (or more) distinct proteins, and why they should be under the same regulation. One possible idea is that they are members of the defense system against active oxygen, which will be described by Drs. Sato and Inoue in a future review. Alternatively, they might have evolved from an ancestor of the defense system that originally protected from oxygen toxicity. Findings on AhR/Arnt and HIF1 systems demonstrating that they are members of the same PAS family seem to support this hypothesis. One of the missing links of the hypothesis is the so called oxygen sensor, suggesting (a) hemoprotein in 1988. A review by Dr. Ogawa on Bach1, the first vertebrate transcription factor controlled by heme, will provide on answers to the missing links. Through this series of reviews, we have a rough map of the biological system against environmental stress to explore the new frontier of environmental science. Publication Types: Review Review, Tutorial PMID: 11868388 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 136: J Steroid Biochem Mol Biol. 2001 Dec;79(1-5):305-14. Differential tissue distribution, developmental programming, estrogen regulation and promoter characteristics of cyp19 genes in teleost fish. Callard GV, Tchoudakova AV, Kishida M, Wood E. Department of Biology, Boston University, 5 Cummington Street, Boston, MA 02215, USA. gvc@bio.bu.edu Teleost fish are characterized by exceptionally high levels of brain estrogen biosynthesis when compared to the brains of other vertebrates or to the ovaries of the same fish. Goldfish (Carassius auratus) and zebrafish (Danio rerio) have utility as complementary models for understanding the molecular basis and functional significance of exaggerated neural estrogen biosynthesis. Multiple cytochrome P450 aromatase (P450arom) cDNAs that derive from separate gene loci (cyp19a and cyp19b) are differentially expressed in brain (P450aromB>>A) and ovary (P450aromA>>B) and have a different developmental program (B>>A) and response to estrogen upregulation (B only). As measured by increased P450aromB mRNA, a functional estrogen response system is first detected 24-48 h post-fertilization (hpf), consistent with the onset of estrogen receptor (ER) expression (alpha, beta, and gamma). The 5'-flanking region of the cyp19b gene has a TATA box, two estrogen response elements (EREs), an ERE half-site (ERE1/2), a nerve growth factor inducible-B protein (NGFI-B)/Nur77 responsive element (NBRE) binding site, and a sequence identical to the zebrafish GATA-2 gene neural specific enhancer. The cyp19a promoter region has TATA and CAAT boxes, a steroidogenic factor-1 (SF-1) binding site, and two aryl hydrocarbon receptor (AhR)/AhR nuclear translocator factor (ARNT) binding motifs. Both genes have multiple potential SRY/SOX binding sites (16 and 8 in cyp19b and cyp19a, respectively). Luciferase reporters have basal promoter activity in GH3 cells, but differences (a>>b) are opposite to fish pituitary (b>>a). When microinjected into fertilized zebrafish eggs, a cyp19b promoter-driven green fluorescent protein (GFP) reporter (but not cyp19a) is expressed in neurons of 30-48 hpf embryos, most prominently in retinal ganglion cells (RGCs) and their projections to optic tectum. Further studies are required to identify functionally relevant cis-elements and cellular factors, and to determine the regulatory role of estrogen in neurodevelopment. Publication Types: Review Review, Tutorial PMID: 11850237 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 137: Teratology. 2002 Jan;65(1):10-8. Characterization of the aryl hydrocarbon receptor repressor gene and association of its Pro185Ala polymorphism with micropenis. Fujita H, Kosaki R, Yoshihashi H, Ogata T, Tomita M, Hasegawa T, Takahashi T, Matsuo N, Kosaki K. Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan. BACKGROUND: Genetic background of a fetus contributes to the abnormal development after teratogen exposure. In rodents, in utero exposure to dioxins affects male external genital development. The effects of dioxins are mediated via the aryl hydrocarbon receptor (AHR) and its binding protein, aryl hydrocarbon receptor nuclear translocator (ARNT). In mice, aryl hydrocarbon receptor repressor (AHRR), which binds to ARNT in competition with AHR, plays a critical negative regulatory role in AHR signaling. We attempt to characterize the human AHRR gene and investigate the relationship between AHRR polymorphisms and the incidence of micropenis, a phenotype of undermasculinization. METHODS: We identified and characterized the human homolog of mouse AHRR, taking advantage of the publicly available draft version of the human genome sequence. After detecting an AHRR protein polymorphism by the direct sequencing of pooled human genomic DNA, we evaluated the association between the polymorphism and the presence or absence of micropenis (< -2.5 SD) in patients with micropenis and control subjects. RESULTS: The deduced sequence for human AHRR (715 residues) and the mouse AHRR protein exhibited 81% sequence homology to each other. The Pro185Ala polymorphism was identified between the PAS-A region and the highly conserved arginine/cysteine-rich RCFRCRL/VRC region. Forty-six percent (27/59) of patients with micropenis and 27% (22/80) of the controls were homozygous for 185Pro; this difference in frequencies was significant (P = 0.03). CONCLUSIONS: Homozygosity for the 185Pro allele of AHRR may increase the susceptibility of a fetus to the undermasculinizing effects of dioxin exposure in utero, presumably through the diminished inhibition of AHR-mediated signaling. Copyright 2002 Wiley-Liss, Inc. PMID: 11835227 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 138: Mol Pharmacol. 2002 Feb;61(2):334-42. Unique property of some synthetic retinoids: activation of the aryl hydrocarbon receptor pathway. Gambone CJ, Hutcheson JM, Gabriel JL, Beard RL, Chandraratna RA, Soprano KJ, Soprano DR. Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA. Potential pharmacological applications in the areas of oncology, dermatology, diabetes, and atherosclerosis of synthetic analogs of retinoic acid that target a specific nuclear receptor and/or biological response have generated great interest in the development of new retinoid and rexinoid drugs. The pan-retinoic acid receptor antagonist AGN 193109 has been previously reported to elevate CYP1A1 levels, implicating the aryl hydrocarbon receptor (AhR) as an additional target for this retinoid. AhR is a cytosolic ligand-dependent transcription factor that, in conjunction with the AhR nuclear translocator (Arnt), binds to dioxin response elements (DREs) located in the promoter region of target genes, such as CYP1A1, and induces their transcription. The purpose of these studies was to determine whether additional synthetic retinoids were capable of elevating CYP1A1 levels and to examine the mechanism of this increase in CYP1A. Two additional retinoids, AGN 190730 and AGN 192837, were found to be potent inducers of DRE-driven transcriptional activity; AGN 190730 was the most potent. Moreover, electrophoretic mobility-shift assays demonstrate that AGN 190730 can transform AhR into its active DNA recognition form. In addition, trypsin digestion of AGN 190730-treated AhR reveals a conformational change in the protein similar to the conformational change of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-bound AhR. Finally, competitive binding studies demonstrate that AGN 190730 can inhibit the binding of TCDD to AhR. The sum of the data demonstrates that some synthetic retinoids in addition to activating the retinoic acid receptor/retinoid X receptor pathway are capable of binding to AhR and activating the AhR/Arnt pathway. PMID: 11809858 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 139: J Biol Chem. 2002 Apr 5;277(14):11821-7. Epub 2002 Jan 22. Functional involvement of the Brahma/SWI2-related gene 1 protein in cytochrome P4501A1 transcription mediated by the aryl hydrocarbon receptor complex. Wang S, Hankinson O. Department of Pathology and Laboratory Medicine, Center for the Health Sciences, University of California, Los Angeles, California 90095-1732, USA. Chromatin remodeling is a key step in overcoming the nucleosomal repression of active transcription in eukaryotes. The mammalian SWI/SNF ATP-dependent chromatin-remodeling complexes contain multiple subunits. The ATPase activities in these complexes are attributable to either BRG-1 or the related Brahma protein. The aryl hydrocarbon receptor (AHR), after binding xenobiotic ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), associates with the AHR nuclear translocator (ARNT), and the dimer so formed activates transcription of several genes, including the cytochrome P4501A1 (CYP1A1) gene. We show that BRG-1 potentiates AHR/ARNT-mediated reporter gene activity in a TCDD-dependent fashion in Hepa1c1c7 cells. Introduction of BRG-1 into the BRG-1- and hBrm-deficient SW13 and C33A human cell lines also enhances expression from a transiently transfected AHR/ARNT-dependent reporter gene. Replenishment of BRG-1 to SW13 cells also restores endogenous cytochrome P4501A1 (CYP1A1) gene expression, whereas an ATPase-deficient mutant of BRG-1 is unable to do so. Chromatin immunoprecipitation analysis demonstrated that BRG-1 associates with the enhancer region of the mouse CYP1A1 gene in vivo in a TCDD- and ARNT-dependent fashion, suggesting the specific recruitment of BRG-1 by AHR/ARNT. Finally, we demonstrate that the glutamine-rich subdomain of the transcriptional activation domain of AHR can interact with BRG-1. Together these studies reveal a functional involvement of BRG-1 in activating CYP1A1 gene transcription and implicate the importance of ATP-dependent chromatin remodeling activity on inducible gene expression mediated by AHR/ARNT. PMID: 11805098 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 140: Eur J Biochem. 2002 Jan;269(1):13-8. A model for recognition of polychlorinated dibenzo-p-dioxins by the aryl hydrocarbon receptor. Procopio M, Lahm A, Tramontano A, Bonati L, Pitea D. Dipartimento di Scienze dell'Ambiente e del Territorio, Universita degli Studi di Milano-Bicocca, Milano, Italy. Ligand binding by the aryl hydrocarbon receptor (AhR), a member of the bHLH-PAS family of transcriptional regulatory proteins, has been mapped to a region within the second 'PAS' domain, a conserved sequence motif first discovered in the Per-ARNT-Sim family of proteins. In addition to the bacterial photoactive yellow protein (PYP), which had been proposed as a structural prototype for the three dimensional fold of PAS domains, two crystal structures of the PAS domain have recently been determined: the human potassium channel HERG and the heme binding domain of the bacterial O(2) sensing FixL protein. The three structures reveal a highly conserved structural framework in evolutionary rather distant PAS domains, provide a more general view of how these domains can recognize their ligands and suggest a structure-function relationship that we exploited to build a three-dimensional model of the ligand binding domain (LBD) of the mouse aryl hydrocarbon receptor (mAhR). The model allowed us to putatively identify the residues responsible for the recognition of polychlorinated dibenzo-p-dioxins (PCDDs) by AhR receptors and to formulate an hypothesis on the signal transduction mechanism. PMID: 11784293 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 141: Mol Cell Biochem. 2001 Oct;226(1-2):107-18. Aryl hydrocarbon receptor/dioxin receptor in human monocytes and macrophages. Komura K, Hayashi S, Makino I, Poellinger L, Tanaka H. Second Department of Internal Medicine, Asahikawa Medical College, Midorigaoka Higashi, Japan. Aryl hydrocarbon receptor (AhR) belongs to the bHLH/PAS transcription factor family and is activated by various polycyclic or halogenated aromatic hydrocarbons, e.g. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 3-methylcholanthrene (3MC). In the present study, we showed that in U937 cells and human macrophages AhR, with its partner cofactor Arnt, is expressed and CYP1A1 mRNA expression is induced in the presence of AhR ligand 3MC. Moreover, we showed that AhR, associating with Arnt, binds to target DNA sequences and activates transcription. Since part of AhR is activated into DNA binding species in the absence of exogenous ligand and competitive AhR antagonist alpha-naphthoflavone inhibits this activation process with reducing CYP1A1 mRNA expression levels, the presence of endogenous ligand is indicated. PMID: 11768231 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 142: Mol Hum Reprod. 2002 Jan;8(1):75-80. Uterine and ovarian aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) mRNA expression in benign and malignant gynaecological conditions. Khorram O, Garthwaite M, Golos T. Department of Obstetrics and Gynecology, University of Wisconsin, and Wisconsin Regional Primate Research Center, Madison, WI, USA. okhorram@rei.edu The transcriptional regulators aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) modulate the transcription of genes involved in cellular differentiation and proliferation. In this study, we investigated the expression of these transcriptional regulators in the female reproductive tract. AHR and ARNT mRNA transcripts were readily detected by a ribonuclease protection assay in all reproductive tissues examined. The expression of these factors in the endometrium and myometrium did not vary during the menstrual cycle, and was not different in pre- versus post-menopausal women. However, post-menopausal women on continuous hormone replacement therapy had greater expression of AHR but not of ARNT in the endometrium and myometrium when compared with women not taking hormones. Leiomyomas expressed significantly less AHR and ARNT mRNA compared with normal myometrium. The ovaries expressed both AHR and ARNT mRNA, and expression was unaffected by age. Endometriotic ovarian cysts expressed more AHR but not more ARNT mRNA compared with healthy ovarian tissue. However, there were no changes in the expression of AHR or ARNT mRNA in ovarian cancer. In conclusion, the female reproductive tract expresses mRNA for the transcription factors AHR and ARNT, and changes in their expression at select target sites in specific pathological conditions such as endometriosis and uterine leiomyomas suggest a potential role for these factors in the pathogenesis of these conditions. PMID: 11756572 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 143: J Biochem (Tokyo). 2002 Jan;131(1):79-85. Characterization of the LxxLL motif in the aryl hydrocarbon receptor: effects on subcellular localization and transcriptional activity. Ikuta T, Watanabe J, Kawajiri K. Saitama Cancer Center Research Institute, Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan. togo@cancer-c.pref.saitama.jp The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that acts in concert with the AhR nuclear translocator (ARNT). Subcellular localization and transcriptional activation of target genes are mainly regulated by ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). We have previously reported that AhR migrates in cells as a nucleocytoplasmic shuttling protein mediated by its nuclear localization and export signals. A short sequence motif LxxLL (L is leucine and x is any amino acid) found in transcriptional co-activators has been reported to mediate the binding to liganded nuclear receptors. The role of the two LxxLL motifs, AhR[50-54] and [224-228], has now been analyzed by determining the localization of AhR and its transcriptional activity with Leu to Ala mutations in full-length AhR. Immunocytostaining revealed that mutation of the motif at AhR[50-54] promotes the efficiency of nuclear localization in the absence of ligand without altering HSP90 and ARA9 binding or nuclear export activity. Furthermore, this mutation decreases the transcriptional activity of the AhR/ARNT system, which is likely due to the suppression of AhR/ARNT/XRE complex formation. Another LxxLL motif at AhR[224-227] affects neither the subcellular localization nor transcriptional activity. These results indicate that the LxxLL motif at AhR[50-54] is important for the regulation of AhR activity. PMID: 11754738 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 144: J Biol Chem. 2002 Mar 1;277(9):6949-59. Epub 2001 Dec 12. Regulatory interactions among three members of the vertebrate aryl hydrocarbon receptor family: AHR repressor, AHR1, and AHR2. Karchner SI, Franks DG, Powell WH, Hahn ME. Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA. The effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds occur via the aryl hydrocarbon receptor (AHR), a member of the basic helix-loop-helix-Per-ARNT-Sim homology (bHLH-PAS) protein superfamily. A single AHR gene has been identified in mammals, whereas many fish species, including the Atlantic killifish (Fundulus heteroclitus) possess two distinct AHR genes (AHR1 and a novel form, AHR2). A mouse bHLH-PAS protein closely related to AHR and designated AHR repressor (AHRR) is induced by 3-methylcholanthrene and represses the transcriptional activity of the AHR. To determine whether AHRR is the mammalian ortholog of fish AHR2 and to investigate the mechanisms by which AHRR regulates AHR function, we cloned an AHRR ortholog in F. heteroclitus with high sequence identity to the mouse and human AHRRs. Killifish AHRR encodes a 680-residue protein with a predicted molecular mass of 75.2 kDa. We show that in vitro expressed AHRR proteins from human, mouse, and killifish all fail to bind [(3)H]TCDD or [(3)H]beta-naphthoflavone. In transient transfection experiments using a luciferase reporter gene under control of AHR response elements, killifish AHRR inhibited the TCDD-dependent transactivation function of both AHR1 and AHR2. AHRR mRNA is widely expressed in killifish tissues and is inducible by TCDD or polychlorinated biphenyls, but its expression is not altered in a population of fish exhibiting genetic resistance to these compounds. The F. heteroclitus AHRR promoter contains three putative AHR response elements. Both AHR1 and AHR2 activated transcription of luciferase driven by the AHRR promoter, and AHRR could repress its own promoter. Thus, AHRR is an evolutionarily conserved, TCDD-inducible repressor of AHR1 and AHR2 function. Phylogenetic analysis shows that AHRR, AHR1, and AHR2 are distinct genes, members of an AHR gene family; these three vertebrate AHR-like genes descended from a single invertebrate AHR. PMID: 11742002 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 145: J Nutr. 2001 Dec;131(12):3281-7. Soy and whey proteins downregulate DMBA-induced liver and mammary gland CYP1 expression in female rats. Rowlands JC, He L, Hakkak R, Ronis MJ, Badger TM. Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA. jcrowlands@salvitas.com One possible mechanism by which diet may reduce cancer risk is through enhancement of metabolic systems that prevent activation of carcinogens or accelerate carcinogen inactivation. We studied the effects of diet and 7,12-dimethylbenz-(a)anthracene (DMBA) on hepatic and mammary gland CYP1A1, CYP1A2 and CYP1B1 enzymes in female Sprague-Dawley rats. Diets (AIN-93G) were fed from conception to adulthood, and DMBA was given by oral gavage at age 48-50 d. The protein sources of diets were casein (CAS), soy protein isolate (SPI) or whey protein hydrolysate (WPH). The DMBA-induced hepatic ethoxyresorufin-O-deethylase and methoxyresorufin-O-demethylase activities and CYP1A1 protein and mRNA expression were lower (P < 0.05) in SPI-fed rats compared with those fed casein. Differences in mammary gland CYP1 expression were also observed with decreased DMBA induction (P < 0.05) of all three CYP1 proteins and mRNAs in rats fed either SPI or WPH compared with those fed CAS. Most notable were the decreased constitutive and DMBA-induced mammary gland expression of CYP1B1 protein of 93 and 96%, respectively, in the SPI-fed rats relative to the CAS-fed controls. The diet-induced changes in CYP1 enzyme expression were consistent with changes in the AhR and ARNT transcription factors that regulate them. Decreased (P < 0.05) mammary constitutive AhR and ARNT proteins were measured in SPI-fed rats. There was also a 100% increase in constitutive AhR protein in the WPH-fed rats that paralleled a 100% increase in constitutive CYP1B1 protein in the mammary gland. These results demonstrate the importance of diet in regulation of phase I metabolism in liver and mammary gland, and suggest a potential mechanism by which soy or whey proteins reduce DMBA-induced mammary tumor incidence. PMID: 11739881 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 146: J Steroid Biochem Mol Biol. 2001 Nov;78(5):427-39. Promoter characteristics of two cyp19 genes differentially expressed in the brain and ovary of teleost fish. Tchoudakova A, Kishida M, Wood E, Callard GV. Department of Biology, Boston University, 5 Cummington Street, Boston, MA 02215, USA. Teleost fish are characterized by exceptionally high levels of neural estrogen biosynthesis when compared with the brains of other vertebrates or to the ovaries of the same fish. Two P450arom mRNAs which derive from separate gene loci (cyp19a and cyp19b) are differentially expressed in brain (b>>a) and ovary (a>>b) and have a different developmental program (b>>a) and estrogen upregulation (b only). A polymerase chain reaction (PCR)-based genomic walking strategy was used to isolate the 5'-flanking regions of the goldfish (Carassius auratus) cyp19 genes. Sequence analysis of the cyp19b gene approximately 1.8 kb upstream of the transcription start site revealed a TATA box at nucleotide (nt) -30, two estrogen responsive elements (EREs; nt -351 and -211) and a consensus binding site (NBRE) for nerve growth factor inducible-B protein (NGFI-B/Nur77) at -286, which includes another ERE half-site. Also present were a sequence at nt -399 (CCCTCCT) required for neural specificity of the zebrafish GATA-2 gene, and 16 copies of an SRY/SOX binding motif. The 5'-flanking region ( approximately 1.0 kb) of the cyp19a gene had TATA (nt -48) and CAAT (nt -71) boxes, a steroidogenic factor-1 (SF-1) binding site (nt -265), eight copies of the SRY/SOX motif, and two copies of a recognition site for binding the arylhydrocarbon receptor (AhR)/AhR nuclear translocator factor (ARNT) heterodimer. Both genes had elements previously identified in the brain specific exon I promoter of the mouse aromatase gene. Cyp19a- and -b/luciferase constructs showed basal promoter activity in aromatase-expressing rodent pituitary (GH3) cells, but differences (a>>b) did not reflect expression in fish pituitary in vivo (b>>a), implying a lack of appropriate cell factors. Consistent with the onset of cyp19b expression in zebrafish embryos, microinjection of a green fluorescent protein (GFP) reporter plasmid into fertilized eggs revealed labeling in neural tissues at 30-48 h post-fertilization (hpf), most prominently in retinal ganglion cells (RGC) and axon-like projections to the optic tectum. Expression of a cyp19a/GFP reporter was not detectable up to 72 hpf. Tandem analysis of cyp19a and cyp19b promoters in living zebrafish embryos can be a useful approach for identifying cis-elements and cellular factors involved in the correct tissue-specific, spatial, temporal and estrogen regulated expression of aromatase genes during CNS and gonadal development. PMID: 11738553 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 147: Cancer Res. 2001 Dec 1;61(23):8534-9. Novel target genes of the Ah (dioxin) receptor: transcriptional induction of N-myristoyltransferase 2. Kolluri SK, Balduf C, Hofmann M, Gottlicher M. Forschungszentrum Karlsruhe, Institute of Toxicology and Genetics, D-76021 Karlsruhe, Germany. Dioxins are potent mammalian carcinogens and toxins affecting liver, skin, and immune and reproductive systems. The intracellular Ah receptor, a ligand-dependent transcription factor of the basic region/helix-loop-helix/Per-Ahr/Arnt-Sim homology domain (bHLH-PAS) protein family, mediates responses to dioxins. Target genes of the Ah receptor that mediate dioxin toxicity and carcinogenicity are, however, mostly unknown. We used 5L rat hepatoma cells to identify dioxin-inducible genes by suppression subtractive hybridization. Eleven cDNA fragments were identified that represented novel sequences or genes for which induction by dioxins had not been known. N-myristoyltransferase 2 (NMT2) is one of the later dioxin-inducible genes. Induction of NMT2 was confirmed in livers of mice in vivo. NMT2 induction was a direct consequence of Ah receptor activation in 5L cells. [(3)H]myristic acid incorporation into 5L cell proteins was inducible by dioxins, indicating that protein myristoylation is a regulated rather than a housekeeping function and that NMT activity is limiting in noninduced 5L cells. Here we show for the first time that expression of NMT2 and induced protein myristoyltransferase activity are direct responses to carcinogen exposure. Because inappropriate protein NH(2)-terminal myristoylation appears to play a role in carcinogenesis, induction of NMT2 may play a central role in dioxin carcinogenicity. PMID: 11731439 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 148: Gene. 2001 Oct 31;278(1-2):223-34. An aryl hydrocarbon receptor (AHR) homologue from the soft-shell clam, Mya arenaria: evidence that invertebrate AHR homologues lack 2,3,7,8-tetrachlorodibenzo-p-dioxin and beta-naphthoflavone binding. Butler RA, Kelley ML, Powell WH, Hahn ME, Van Beneden RJ. University of Maine, School of Marine Sciences, Orono, ME 04469, USA. The aryl hydrocarbon receptor (AHR) mediates numerous toxic effects following exposure of vertebrate animals to certain aromatic environmental contaminants, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). To investigate possible effects of TCDD on invertebrates, a cDNA encoding an AHR homologue was cloned from the soft-shell clam, Mya arenaria. The predicted amino acid sequence contains regions characteristic of vertebrate AHRs: basic helix-loop-helix (bHLH) and PER-ARNT-SIM (PAS) domains and a glutamine-rich region. Phylogenetic analysis shows that the clam AHR sequence groups within the AHR subfamily of the bHLH-PAS family, in a clade containing AHR homologues from Drosophila melanogaster and Caenorhabditis elegans. AHR mRNA expression was detected in all tissue types tested: adductor muscle, digestive gland, foot, gill, gonad, mantle, and siphon. The in vitro-expressed clam AHR exhibited sequence-specific interactions with a mammalian xenobiotic response element (XRE). Velocity sedimentation analysis using either in vitro-expressed clam AHR or clam cytosolic proteins showed that this AHR homologue binds neither [(3)H]TCDD nor [(3)H]beta-naphthoflavone (BNF). Similarly, in vitro-expressed D. melanogaster and C. elegans AHR homologues lacked specific binding of these compounds. Thus, the absence of specific, high-affinity binding of the prototypical AHR ligands TCDD and BNF, is a property shared by known invertebrate AHR homologues, distinguishing them from vertebrate AHRs. Comparative studies of phylogenetically diverse organisms may help identify an endogenous ligand(s) and the physiological role(s) for this protein. PMID: 11707340 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 149: DNA Seq. 2001 Jul;12(1):13-25. The human membrane progesterone receptor gene: genomic structure and promoter analysis. Bernauer S, Wehling M, Gerdes D, Falkenstein E. Institute of Clinical Pharmacology, Faculty of Clinical Medicine Mannheim, University of Heidelberg, 68135 Mannheim, Germany. Rapid, nongenomic effects of steroids are likely to be mediated by membrane receptors not by intracellular steroid receptors. We recently identified a progesterone membrane binding protein (mPR) from human liver. The corresponding hmpr gene is comprised of 3 exons and 2 introns. The promoter sequence of hmpr lacks a typical TATA box but contains instead a high homology to a transcription Initiatior consensus sequence, which overlaps the experimentally determined transcriptional start site. The major proximal promoter is GC-rich and sequence analysis revealed a CpG island spanning the transcriptional start site. Several putative cis-regulatory DNA-motifs, which represent possible binding sites for transcription factors like AP2, NF-AT, Ahr/Arnt and C/EBP were identified in the genomic upstream region by sequence homology. Functional analysis of differently deleted fragments of the hmpr upstream region in a GFP-reportergene assay in transiently transfected cultured cells indicates the general testability of the hmpr promoter in vivo. PMID: 11697142 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 150: Biochem Biophys Res Commun. 2001 Nov 2;288(3):503-8. The 5'-flanking regions of CYP19A1 and CYP19A2 in zebrafish. Kazeto Y, Ijiri S, Place AR, Zohar Y, Trant JM. Center of Marine Biotechnology, University of Maryland Biotechnology Institute, Baltimore, Maryland 21202, USA. This report describes the structure of the 5'-flanking regions of both the CYP19A1 and A2 genes that were isolated from the genome of the zebrafish (Danio rerio). Consensus sequences of three cAMP-responsive elements (CRE), an aryl hydrocarbon-responsive element (AhR/Arnt), a steroidogenic factor 1 (SF-1) site, and a TATA box were observed in the 5'-flanking region of CYP19A1. In contrast, the 5'-flanking region of CYP19A2 was located upstream of an untranslated exon and possessed consensus sequences of a single CRE, an estrogen-responsive element (ERE), a peroxisome proliferator-activated receptor alpha/retinoid X receptor alpha heterodimer-responsive element (PPARalpha/RXRalpha), and a TATA box. Primer extension analysis revealed that the predominant transcription initiation sites for CYP19A1 and A2 transcripts were 28 and 91 bp upstream from the putative translation initiation codon, respectively. These analyses indicate that substantially different regulators, including a variety of environmental xenobiotics, control the expression the two CYP19 genes. Copyright 2001 Academic Press. PMID: 11676471 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 151: Toxicol Sci. 2001 Nov;64(1):41-56. cDNA cloning and characterization of a high affinity aryl hydrocarbon receptor in a cetacean, the beluga, Delphinapterus leucas. Jensen BA, Hahn ME. Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543. Some cetaceans bioaccumulate substantial concentrations of planar halogenated aromatic hydrocarbons (PHAHs) in their tissues, but little is known about the effects of such burdens on cetacean health. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related PHAHs cause toxicity via activation of the aryl hydrocarbon receptor (AHR), a member of the bHLH-PAS family of transcription factors. Differences in AHR structure and function are known to contribute to species-specific differences in susceptibility to PHAH toxicity. To ascertain the potential for PHAH effects in a cetacean, we characterized an AHR from the beluga whale, Delphinapterus leucas. The 3.2 kb cDNA encodes an 845-amino acid protein with a predicted size of 95.5 kDa. Overall, the beluga AHR shares 85% amino acid sequence identity with the human AHR and 75% identity with the mouse AHR Ah(b-1) allele. Beluga AHR protein synthesized in a rabbit reticulocyte lysate system demonstrated specific, high-affinity [(3)H]TCDD binding. Saturation binding analysis was used to compare the [(3)H]TCDD binding affinity of the in vitro-expressed beluga AHR with affinities of in vitro-expressed AHRs from a dioxin-sensitive mouse strain (Ah(b-1) allele) and humans. The beluga AHR bound [(3)H]TCDD with an affinity (K(d)= 0.43 +/- 0.16 nM) that was at least as high as that of the mouse AHR (K(d)= 0.68 +/- 0.23 nM), and significantly greater than that of the human AHR (K(d)= 1.63 +/- 0.64 nM). In electrophoretic mobility shift assays, the beluga AHR exhibited sequence-specific, Arnt-dependent binding to a dioxin responsive enhancer (DRE). Upon transient transfection into mammalian cells, the beluga AHR activated transcription of a luciferase reporter under control of a DRE-containing fragment of the mouse Cyp1a1 promoter. These results show that in an in vitro system, the beluga AHR possesses characteristics similar to those of AHRs from other mammals that are considered sensitive to toxic effects of PHAHs. Together, these results demonstrate that the use of in vitro-expressed proteins is a promising approach for addressing molecular and biochemical questions concerning PHAH toxicity in endangered or protected species. PMID: 11606800 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 152: J Steroid Biochem Mol Biol. 2001 Sep;78(3):253-60. The effect of the arylhydrocarbon receptor on the human steroidogenic acute regulatory gene promoter activity. Sugawara T, Nomura E, Sakuragi N, Fujimoto S. Department of Biochemistry, Hokkaido University School of Medicine, Kita-ku, Kita 15, Nishi 7, 060-8638, Sapporo, Japan. terusuga@med.hokudai.ac.jp The steroidogenic acute regulatory (StAR) protein is a rate-limiting factor in steroid hormone production. The StAR protein plays a role in the movement of cholesterol from the outer membrane to the inner membrane, where cholesterol side chain cleavage enzyme exists. Dioxins, which may act as 'endocrine disruptors', mimic and antagonize endogenous hormone actions in vivo. Although the mechanism of endocrine disruption is not clear, the actions of dioxins are known to be mediated by binding to the arylhydrocarbon receptor (AhR), and it is known that dioxins act as transcription factors to endocrine-associated gene expression. In the present study, we examined the effect of the AhR on the human StAR gene promoter, and we clarified the action mechanisms of environmental endocrine disruptors. We transfected constructs containing the human StAR gene promoter sequences pGL(2) 1.3-kb StAR (nt -1293 to +39) into mouse Y-1 adrenal tumor cells and measured the promoter activity of the StAR gene. With the addition of beta-napthoflavone (betaNF), which is a ligand of AhR, to the culture medium, the activity of the StAR gene promoter increased significantly (P<0.05), and with the addition of 1 microM of betaNF, it became maximum (3.1+/-0.6-fold higher than the control value). When the AhR and ARNT were co-transfected together in Y-1 cells or human adrenocortical carcinoma H295R cells, the promoter activity of the StAR gene significantly (P<0.05) increased, to a level 1.4+/-0.01-fold higher in Y-1 cells and to a level 1.6+/-0.04-fold higher in H295R cells than the control level, when 1 microM of betaNF was added. We examined the effect of induction of cAMP with transfection with AhR or ARNT. With the addition of 1 mM 8-Br-cAMP, there were no differences between the StAR gene promoter activities in the group in which AhR and ARNT was introduced and in the group in which they were not introduced. The results suggest that AhR plays a role in the promoter activity of the human StAR gene and that the effect of AhR on StAR gene expression may cause a disturbance to the human endocrine system. PMID: 11595506 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 153: J Biol Chem. 2001 Nov 9;276(45):42302-10. Epub 2001 Sep 10. The Q-rich subdomain of the human Ah receptor transactivation domain is required for dioxin-mediated transcriptional activity. Kumar MB, Ramadoss P, Reen RK, Vanden Heuvel JP, Perdew GH. Department of Veterinary Science and the Center for Molecular Toxicology and Carcinogenesis and the Graduate Program in Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania 16802, USA. The aryl hydrocarbon receptor (AhR), a basic helix-loop-helix/Per-Arnt-Sim transcription factor, mediates many of the toxic and biological effects of the environmental contaminant, 2,3,7,8-tetrachlorodibenzo-p-dioxin, which include the transcriptional activation of dioxin-responsive genes such as CYP1A1. Many aspects of this process are known; however, the mechanism of transcriptional activation and the proteins that are key to this process remain to be determined. The hAhR has a complex transactivation domain, composed of three potentially distinct subdomains. Deletional analysis of the hAhR transactivation domain indicates that removal of the P/S/T-rich subdomain enhances transcriptional activity, whereas the Q-rich subdomain is critical for hAhR transactivation potential, and the acidic subdomain by itself fails to activate a dioxin response element-driven reporter gene. Deletional analysis of the Q-rich subdomain identified a critical stretch of 23 amino acids between residues 666 and 688 of the hAhR, which are required for transactivation potential. Alanine scanning mutagenesis of this region identified a leucine residue (Leu-678), which is required for hAhR activity. Functional analysis of this point mutant revealed that it is capable of binding ligand, heterodimerization, and subsequent binding to dioxin response elements. Further, when hAhR/L678A and hAhR containing only the acidic subdomain were overexpressed they acted as dominant negative receptors and repressed wild-type hAhR activity. In addition, the hAhR/L678A failed to activate CYP1A1 gene transcription in transfected BP-8 cells and exhibited reduced binding to RIP140 in vitro. Thus, Leu-678 appears to be critical for efficient transactivation activity of the hAhR and appears to disrupt recruitment of co-regulators. PMID: 11551916 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 154: Mol Cell Endocrinol. 2001 Aug 20;182(1):39-48. 2,3,7,8-Tetrachlorodibenzo-p-dioxin increases steady-state estrogen receptor-beta mRNA levels after CYP1A1 and CYP1B1 induction in rat granulosa cells in vitro. Dasmahapatra AK, Wimpee BA, Trewin AL, Hutz RJ. Department of Biological Sciences, 308 Lapham Hall, University of Wisconsin-Milwaukee, 3209 North Maryland Avenue, Milwaukee, WI 53211, USA. Previous in-vitro investigations of rat granulosa cells (GC) have shown that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) inhibits estrogen secretion and FSH-induced aromatase activity. Although TCDD exerted no effect on basal aromatase enzyme activity, TCDD did reduce steady-state aromatase mRNA levels in GC using competitive RT-PCR. TCDD is hypothesized to induce these changes through aromatic hydrocarbon receptor(AHR)-mediated gene transcription and the modulation of the estrogen receptor (ER)-signaling pathway. In this study we show that rat GC express mRNA for AHR and the AHR nuclear translocator (ARNT) as well as biomarkers of TCDD action, CYP1A1 and CYP1B1 mRNA. Basal CYP1A1 and ER-alpha mRNAs were present only in trace amounts. By relative RT-PCR analysis we showed that CYP1A1 and CYP1B1 mRNA were induced significantly by TCDD at 6 h and that induction of CYP1A1 was maintained throughout the experiment. Using competitive RT-PCR, we observed no significant change in the mRNA levels of ARNT between control and TCDD-treated GC. Both AHR and ER-beta mRNA levels increased significantly at 48 h with TCDD compared with controls. Since ER-beta mRNA was not increased significantly until 48 h in culture, we suggest that in rat GC, the observed ER-beta mRNA increase by TCDD might be a result of CYP1A1/CYP1B1 catalyzed estrogen metabolism and aromatase mRNA inhibition via AHR. PMID: 11500237 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 155: Curr Drug Metab. 2001 Jun;2(2):149-64. Induction of CYP1A1. The AhR/DRE paradigm: transcription, receptor regulation, and expanding biological roles. Ma Q. Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA. The CYP1A1 gene encodes microsomal cytochrome P4501A1 that catalyzes the metabolism of many xenobiotics, including the oxygenation of polycyclic aromatic hydrocarbons (PAH). Induction of CYP1A1 enhances the metabolism of PAHs, and therefore, represents an adaptive response to chemical exposure in mammalian cells. Mechanistic studies reveal an AhR/DRE paradigm for the induction, which involves activation of the aryl hydrocarbon receptor (AhR) by an agonist, dimerization of AhR with the Ah recceptor nuclear translocator (Arnt), followed by binding of the AhR/Arnt heterodimer to the dioxin-responsive enhancer (DRE) and transcription of the gene. The AhR mediated transcription is tightly regulated through, at least, two mechanisms: (a) the cytoplasmic AhR interacts with hsp90 and an immunophilin chaperone AIP for proper folding and receptivity, and (b) the agonist-activated, nuclear AhR is degraded through the ubiquitin-26S proteasome mediated protein turnover, such that the transcription by AhR is controlled at a physiologically adequate level. In addition to CYP1A1 induction, AhR mediates a broad range of biological responses to CYP1A1 inducers, typified by the environmental contaminant dioxin, via modulating gene expression. Thus, mechanistic studies of CYP1A1 induction have provided insights into P450 induction, PAH carcinogenesis, dioxin action, AhR function, and receptor-mediated mammalian gene expression. Publication Types: Review PMID: 11469723 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 156: Mar Environ Res. 2000 Jul-Dec;50(1-5):39-44. The evolution of aryl hydrocarbon signaling proteins: diversity of ARNT isoforms among fish species. Powell WH, Hahn ME. Biology Department, MS#32, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA. The aryl hydrocarbon receptor nuclear translocator (ARNT) mediates aryl hydrocarbon signaling and toxicity by dimerizing with the ligand-activated aryl hydrocarbon receptor (AHR), forming a complex that binds specific DNA elements and alters transcription of target genes. Two genes encode different forms of ARNT in rodents: ARNT1, which is widely expressed, and ARNT2, which exhibits a very restricted expression pattern. In an effort to characterize aryl hydrocarbon signaling mechanisms in fishes, we previously isolated an ARNT cDNA from Fundulus heteroclitus and discovered that this species expresses ARNT2 ubiquitously. This situation differs not only from mammals, but also from rainbow trout, which expresses a divergent ARNT gene that we hypothesized was peculiar to salmonids (rtARNTa/b). In this communication, we examine the ARNT sequences of multiple fish species, including a newly isolated cDNA from scup (Stenotomus chrysops). Our phylogenetic analysis demonstrates that zebrafish ARNT, like the Fundulus protein, is an ARNT2. Contrary to expectations, the scup ARNT is closely related to the rainbow trout protein, demonstrating that the existence of this ARNT isoform predates the divergence of salmonids from the other teleosts. Thus, different species of fish express distinct and highly conserved isoforms of ARNT. The number, type, and expression pattern of ARNT proteins may contribute to interspecies differences in aryl hydrocarbon toxicity, possibly through distinct interactions with additional PAS-family proteins. PMID: 11460724 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 157: Nat Genet. 2001 Aug;28(4):355-60. Comment in: Nat Genet. 2001 Aug;28(4):300-1. Aromatic hydrocarbon receptor-driven Bax gene expression is required for premature ovarian failure caused by biohazardous environmental chemicals. Matikainen T, Perez GI, Jurisicova A, Pru JK, Schlezinger JJ, Ryu HY, Laine J, Sakai T, Korsmeyer SJ, Casper RF, Sherr DH, Tilly JL. Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts 02114, USA. Polycyclic aromatic hydrocarbons (PAHs) are toxic chemicals released into the environment by fossil fuel combustion. Moreover, a primary route of human exposure to PAHs is tobacco smoke. Oocyte destruction and ovarian failure occur in PAH-treated mice, and cigarette smoking causes early menopause in women. In many cells, PAHs activate the aromatic hydrocarbon receptor (Ahr), a member of the Per-Arnt-Sim family of transcription factors. The Ahr is also activated by dioxin, one of the most intensively studied environmental contaminants. Here we show that an exposure of mice to PAHs induces the expression of Bax in oocytes, followed by apoptosis. Ovarian damage caused by PAHs is prevented by Ahr or Bax inactivation. Oocytes microinjected with a Bax promoter-reporter construct show Ahr-dependent transcriptional activation after PAH, but not dioxin, treatment, consistent with findings that dioxin is not cytotoxic to oocytes. This difference in the action of PAHs versus dioxin is conveyed by a single base pair flanking each Ahr response element in the Bax promoter. Oocytes in human ovarian biopsies grafted into immunodeficient mice also accumulate Bax and undergo apoptosis after PAH exposure in vivo. Thus, Ahr-driven Bax transcription is a novel and evolutionarily conserved cell-death signaling pathway responsible for environmental toxicant-induced ovarian failure. PMID: 11455387 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 158: Toxicol Sci. 2001 Aug;62(2):289-98. Expression of AhR and ARNT mRNA in cultured human endometrial explants exposed to TCDD. Pitt JA, Feng L, Abbott BD, Schmid J, Batt RE, Costich TG, Koury ST, Bofinger DP. Curriculum in Toxicology, University of North Carolina, Chapel Hill, NC 27599, USA. Endometriosis is a debilitating disease found in 10-15% of reproductive-age women and is characterized by the presence of endometrial tissue outside of the uterus. The present study characterizes the expression of AhR and ARNT mRNA in a human endometrial explant culture model in the absence and presence of TCDD exposure. In a parallel, companion study using this model, TCDD exposure was shown to induce CYP1A1 mRNA, CYP1B1 mRNA, EROD (7-ethoxyresorufin-O-deethylase) activity, and CYP1B1 protein in human endometrial explants. Explants were prepared from specimens obtained at laparoscopy or laparotomy from women undergoing surgery for tubal ligation, endometriosis, or pelvic pain unrelated to endometriosis. These specimens were a subset of the specimens used in the parallel study. The explants were cultured in medium containing 10 nM estradiol (E(2)) or 1 nM estradiol plus 500 nM progesterone (E(2) + P(4)) with or without TCDD (first 24 h). After culture, AhR and ARNT mRNA expression were quantified by RT-PCR. TCDD treatment significantly increased the expression of AhR mRNA, but not ARNT mRNA. The expression of both genes was similar for all individual explants and the ratio of AhR:ARNT mRNA expression across all samples was 1.7 to 1.8. Constitutive AhR mRNA expression was donor age dependent (increasing with age), while ARNT mRNA expression was donor age and tissue phase dependent (increased in older and proliferative phase specimens). Similar to results in the parallel study on expression of CYP1A1 mRNA, CYP1B1 mRNA, EROD activity, and CYP1B1 protein, the presence of endometriosis did not affect the expression of AhR or ARNT mRNA, either constitutively or following TCDD exposure. However, the detection of disease-specific change was limited by small sample size and variability in tissue cycle phase. The human endometrial explant culture model will be useful for future studies of the effects of dioxin-like compounds on human endometrium in relationship to cycle phase, hormonal exposure, and donor age. PMID: 11452142 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 159: Toxicol Appl Pharmacol. 2001 Jul 15;174(2):153-9. The synthetic retinoid AGN 193109 but not retinoic acid elevates CYP1A1 levels in mouse embryos and Hepa-1c1c7 cells. Soprano DR, Gambone CJ, Sheikh SN, Gabriel JL, Chandraratna RA, Soprano KJ, Kochhar DM. Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA. The synthetic retinoid AGN 193109 is a potent pan retinoic acid receptor (RAR) antagonist. Treatment of pregnant mice with a single oral 1 mg/kg dose of this antagonist on day 8 postcoitum results in severe craniofacial (median cleft face or frontonasal deficiency) and eye malformations in virtually all exposed fetuses. Using differential display analysis, we have determined that CYP1A1 mRNA levels are elevated in mouse embryos 6 h following treatment with AGN 193109. Similarly, an elevation in CYP1A1 mRNA levels, protein levels, and aryl hydrocarbon hydoxylase activity occurs in Hepa-1c1c7 cells, with the maximal elevation observed when the cells were treated with 10(-5) M AGN 193109 for 4 to 8 h. Elevation in CYP1A1 mRNA levels in mouse embryos and Hepa-1c1c7 cells does not occur upon treatment with the natural retinoid, all-trans-retinoic acid. Finally, elevation in CYP1A1 mRNA levels was not observed when mutant Hepa-1c1c7 cells, which are defective in either the aryl hydrocarbon receptor (AhR) or aryl hydrocarbon receptor nuclear translocator (ARNT), were treated with AGN 193109. This suggests that the AhR/ARNT pathway and not the RAR/RXR pathway is mediating the elevation of CYP1A1 mRNA levels by AGN 193109, at least in the Hepa-1c1c7 cells. This is the first example of a retinoid that displays the abililty to regulate both the RAR/RXR and AhR/ARNT transcriptional regulatory pathways. Copyright 2001 Academic Press. PMID: 11446831 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 160: J Biol Chem. 2001 Aug 31;276(35):33101-10. Epub 2001 Jun 21. Structure and expression of the Ah receptor repressor gene. Baba T, Mimura J, Gradin K, Kuroiwa A, Watanabe T, Matsuda Y, Inazawa J, Sogawa K, Fujii-Kuriyama Y. Department of Biomolecular Science, Graduate School of Life Science, Tohoku University, Sendai 980-8578, Japan. The aryl hydrocarbon receptor (AhR) repressor (AhRR) gene has been isolated and characterized from a mouse genomic library. The gene is distributed as 11 exons in a total length of about 60 kilobase pairs. Fluorescence in situ hybridization analysis has shown that the AhRR gene is located at mouse chromosome 13C2, at rat chromosome 1p11.2, and at human chromosome 5p15.3. The AhRR gene has a TATA-less promoter and several transcription start sites. In addition, putative regulatory DNA sequences such as xenobiotic responsive element (XRE), GC box, and NF-kappaB-binding sites have been identified in the 5'-upstream region of the AhRR gene. Transient transfection analyses of HeLa cells with reporter genes that contain deletions and point mutations in the AhRR promoter revealed that all three XREs mediated the inducible expression of the AhRR gene by 3-methylcholanthrene treatment, and furthermore, GC box sequences were indispensable for a high level of inducible expression and for constitutive expression. Moreover, by using gel mobility shift assays we were able to show that the AhR/Arnt heterodimer binds to the XREs with very low affinity, which is due to three varied nucleotides outside the XRE core sequence. We have also shown that Sp1 and Sp3 can bind to the GC boxes. Finally, both transient transfection analysis and gel mobility shift assay revealed that the AhRR gene is up-regulated by a p65/p50 heterodimer that binds to the NF-kappaB site when the cells has been exposed to 12-O-tetradecanoylphorbol-13-acetate, and this inducible expression was further enhanced by cotreatment of 12-O-tetradecanoylphorbol-13-acetate and 3-methylcholanthrene. PMID: 11423533 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 161: J Hum Genet. 2001;46(6):342-6. Human arylhydrocarbon receptor repressor (AHRR) gene: genomic structure and analysis of polymorphism in endometriosis. Watanabe T, Imoto I, Kosugi Y, Fukuda Y, Mimura J, Fujii Y, Isaka K, Takayama M, Sato A, Inazawa J. Department of Molecular Cytogenetics, Tokyo Medical and Dental University, Japan. The diversity of biological effects resulting from exposure to dioxin may reflect the ability of this environmental pollutant to alter gene expression by binding to the arylhydrocarbon receptor (AHR) gene and related genes. AHR function may be regulated by structural variations in AHR itself, in the AHR repressor (AHRR), in the AHR nuclear translocator (ARNT), or in AHR target molecules such as cytochrome P-4501A1 (CYP1A1) and glutathione S-transferase. Analysis of the genomic organization of AHRR revealed an open reading frame consisting of a 2094-bp mRNA encoded by ten exons. We found one novel polymorphism, a substitution of Ala by Pro at codon 185 (GCC to CCC), in exon 5 of the AHRR gene; among 108 healthy unrelated Japanese women, genotypes Ala/Ala, Ala/Pro, and Pro/Pro were represented, respectively, by 20 (18.5%), 49 (45.4%), and 39 (36.1%) individuals. We did not detect previously published polymorphisms of ARNT (D511N) or the CYP1A1 promoter (G-469A and C-459T) in our subjects, suggesting that these polymorphisms are rare in the Japanese population. No association was found between uterine endometriosis and any polymorphisms in the AHRR, AHR, ARNT, or CYP1A1 genes analyzed in the present study. PMID: 11393538 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 162: Proc Natl Acad Sci U S A. 2001 Jun 5;98(12):6692-7. Epub 2001 May 29. Targeted mutation of the murine arylhydrocarbon receptor nuclear translocator 2 (Arnt2) gene reveals partial redundancy with Arnt. Keith B, Adelman DM, Simon MC. Departments of Medicine and Cell and Developmental Biology, Abramson Family Cancer Research Institute, and Howard Hughes Medical Institute, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA 19104, USA. The ubiquitously expressed basic helix-loop-helix (bHLH)-PAS protein ARNT (arylhydrocarbon receptor nuclear transporter) forms transcriptionally active heterodimers with a variety of other bHLH-PAS proteins, including HIF-1alpha (hypoxia-inducible factor-1alpha) and AHR (arylhydrocarbon receptor). These complexes regulate gene expression in response to hypoxia and xenobiotics, respectively, and mutation of the murine Arnt locus results in embryonic death by day 10.5 associated with placental, vascular, and hematopoietic defects. The closely related protein ARNT2 is highly expressed in the central nervous system and kidney and also forms complexes with HIF-1alpha and AHR. To assess unique roles for ARNT2 in development, and reveal potential functional overlap with ARNT, we generated a targeted null mutation of the murine Arnt2 locus. Arnt2(-/-) embryos die perinatally and exhibit impaired hypothalamic development, phenotypes previously observed for a targeted mutation in the murine bHLH-PAS gene Sim1 (Single-minded 1), and consistent with the recent proposal that ARNT2 and SIM1 form an essential heterodimer in vivo [Michaud, J. L., DeRossi, C., May, N. R., Holdener, B. C. & Fan, C. (2000) Mech. Dev. 90, 253-261]. In addition, cultured Arnt2(-/-) neurons display decreased hypoxic induction of HIF-1 target genes, demonstrating formally that ARNT2/HIF-1alpha complexes regulate oxygen-responsive genes. Finally, a strong genetic interaction between Arnt and Arnt2 mutations was observed, indicating that either gene can fulfill essential functions in a dose-dependent manner before embryonic day 8.5. These results demonstrate that Arnt and Arnt2 have both unique and overlapping essential functions in embryonic development. PMID: 11381139 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 163: Environ. Toxicol. Pharmacol.. 2001 Jun;10(1-2):17-27. Interactions between aryl hydrocarbon receptor (AhR) and hypoxia signaling pathways. Nie M, Blankenship AL, Giesy JP. National Food Safety and Toxicology Center, Department of Zoology and Institute for Environmental Toxicology, Michigan State University, 48824-1311, East Lansing, MI, USA Most if not all 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 HIF-1alpha 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 we report here were conducted to test the hypothesis that there is cross talk between AhR- and HIF-1-mediated transcription pathways. TCDD significantly reduced the hypoxia-mediated reporter gene activity in B-1 cells. Reciprocally, the hypoxia response inducers desferrioxamine or CoCl(2) inhibited AhR-mediated CYP1A1 enzyme activity in B-1 and Hepa 1 cells, and the AhR-mediated luciferase reporter gene activity in H1L1.1c2 cells. The inhibition of AhR-mediated transcription by hypoxia inducers, however, was not observed in H4IIE-luc cells. The interaction between the AhR- and HIF-1-mediated transcription can be attributed to changes in DNA binding activities. TCDD-induced protein binding to dioxin responsive element (DRE) was diminished by desferrioxamine, and TCDD reduced the binding activity to HIF-1 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. PMID: 11382553 [PubMed - as supplied by publisher] --------------------------------------------------------------- 164: Arch Biochem Biophys. 2001 Jun 1;390(1):64-70. Short heterodimer partner (SHP) orphan nuclear receptor inhibits the transcriptional activity of aryl hydrocarbon receptor (AHR)/AHR nuclear translocator (ARNT). Klinge CM, Jernigan SC, Risinger KE, Lee JE, Tyulmenkov VV, Falkner KC, Prough RA. Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky 40292, USA. carolyn.klinge@louisville.edu SHP (short heterodimer partner) is an orphan nuclear receptor lacking a DNA binding domain that interacts with nuclear receptors (NR) including thyroid receptor (TR), retinoic acid receptors (RAR and RXR), and estrogen receptors alpha and beta (ERalpha and ERbeta). SHP acts as a negative regulator of these receptors by inhibiting DNA binding and transcriptional activation. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) binds to arylhydrocarbon receptor (AHR), activating the AHR/AHR nuclear translocator (ARNT) heterodimer. We investigated the physical and functional interaction of SHP with AHR/ARNT. In RL95-2 human endometrial carcinoma cells, SHP inhibited TCDD-stimulated reporter activity from the AHR-responsive CYP1A1 and UGT1A6 gene promoters in a concentration-dependent manner. In GST pull-down assays, ARNT interacted directly with SHP in vitro, but AHR did not interact with GST-SHP. SHP inhibited AHR/ARNT-DNA binding in vitro. These results identify ARNT as a novel SHP target. We speculate a role for SHP in the suppression of agonist-activated AHR/ARNT activity. Copyright 2001 Academic Press. PMID: 11368516 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 165: Toxicol Lett. 2001 Mar 31;120(1-3):1-7. Molecular biology of the Ah receptor and its role in carcinogenesis. Safe S. Department of Veterinary Physiology & Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA. ssafe@cvm.tamu.edu The aryl hydrocarbon receptor (AhR) is a ligand-activated nuclear transcription factor that mediates responses to toxic halogenated aromatic toxins such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), polynuclear aromatic hydrocarbons, combustion products, and numerous phytochemicals such as flavonoids and indole-3-carbinol (I3C). The nuclear AhR complex is a heterodimer containing the AhR and AhR nuclear translocator (Arnt) proteins, and the molecular mechanism of AhR action is associated with binding of the heterodimer to dioxin responsive elements (DREs) in regulatory regions of Ah-responsive genes. TCDD, a 'xenodioxin', is a multi-site carcinogen in several species and possibly in humans, whereas natural AhR ligands including I3C and flavonoids tend to protect against cancer. Both TCDD and phytochemicals inhibit estrogen-induced breast and endometrial cancer, and the molecular mechanisms of this common response will be described. Publication Types: Review Review, Tutorial PMID: 11323156 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 166: Clin Cancer Res. 2001 Apr;7(4):818-23. Association of ARNT splice variants with estrogen receptor-negative breast cancer, poor induction of vascular endothelial growth factor under hypoxia, and poor prognosis. Qin C, Wilson C, Blancher C, Taylor M, Safe S, Harris AL. Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843-4466, USA. The aryl hydrocarbon receptor nuclear translocator (ARNT) is a basic helix-loop-helix transcription factor that forms heterodimers with the aryl hydrocarbon receptor (AhR) or hypoxia inducible factor-1alpha to activate transcription via xenobiotic response element or hypoxia response element, respectively. Thus, it plays a major role in two key biochemical pathways involved in tumor growth. We previously showed that estrogen receptor (ER)-negative breast cancer cell lines expressed a splice variant of ARNT that was associated with Ah nonresponsiveness. We have now used a sensitive PCR method to analyze the expression of the variant in a series of 92 breast cancers to assess interactions with the ER and prognosis. The splice variant could be detected in all of the cases examined, with high ratios of variant:full-length ARNT (> or =10) characterized in 10 cases. When the patient group was split into quartiles by increasing splice variant ratios, there was an inverse relationship of ER status to ARNT splice-variant ratios (P = 0.01, chi(2)). Univariate analysis showed that cases with high ARNT splice-variant ratios > or =10 had a worse relapse-free and overall survival (P > or = 0.03; hazard ratio, 2.7; and P = 0.006; hazard ratio, 3.9, respectively). In multivariate analysis for relapse-free and overall survival, ARNT splice-variant ratio was the strongest independent factor and, although inversely related to ER, remained a separate risk factor. At least two potential mechanisms could explain this phenomenon: the loss of aryl hydrocarbon receptor-mediated antiestrogenic activity or the blockade of a proapoptotic pathway induced by hypoxia. Because several enzymes involved in drug resistance are induced through a xenobiotic response element, the tumors presenting high ARNT splice-variant ratios may be specifically targeted by drugs normally degraded or inactivated. This study shows the biological importance of ARNT splice variants in the behavior of human breast cancer and suggests that the breast cell lines in which the splice variant was discovered may be useful models for further investigation. PMID: 11309328 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 167: Toxicol Sci. 2001 May;61(1):187-96. Correlation of cardiotoxicity mediated by halogenated aromatic hydrocarbons to aryl hydrocarbon receptor activation. Heid SE, Walker MK, Swanson HI. Department of Pharmacology, University of Kentucky Medical Center, Lexington, Kentucky 40536, USA. In mammals, the toxicity of halogenated aromatic hydrocarbons (HAH) correlates with their ability to activate the aryl hydrocarbon receptor (AHR). To test this correlation in an avian model, we selected six HAHs based on their affinity for the mammalian AHR, including: 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD); 1,2,3,7,8-pentachlorodibenzo-p-dioxin (PCDD); 2,3,7,8-tetrachlorodibenzofuran (TCDF); 2,3,4,7,8-pentachlorodibenzofuran (PCDF); 3,3',4,4'-tetrachlorobiphenyl (PCB 77); and 2,2',4,4',5,5'-hexachlorobiphenyl (PCB 153). We determined the ability of these compounds to induce cardiotoxicity, as measured by an increase in heart wet weight on incubation day 10 in the chick embryo (Gallus gallus) and formation of the avian AHR/ARNT/DNA binding complex in chicken hepatoma cells. Relative potency values (RPs) were calculated by dividing the TCDD EC(50) (AHR/ARNT/DNA binding) or ED(50) (15% increase in day-10 heart wet weight) by the HAH congeners EC(50) or ED(50), respectively. The rank order of potencies for inducing cardiotoxicity were TCDD > PCDD = PCDF = TCDF > PCDF > PCB77, PCB 153, no effect. The RP values for inducing AHR/ARNT DNA binding were then correlated with those for inducing cardiotoxicity (the RP values of PCDD were determined to be statistical outliers). This correlation was found to be highly significant (r = 0.94, p = 0.017). The ability of PCDD to act as an AHR agonist was verified using luciferase reporter assays and analysis of cytochrome P4501A1 protein levels. These results indicate that the ability of HAHs to activate the avian AHR signaling pathway, in general, correlates with their ability to mediate cardiotoxicity in the chick embryo. PMID: 11294989 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 168: Mol Cell Biol Res Commun. 2000 Sep;4(3):174-80. Comparative effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on MCF-7, RL95-2, and LNCaP cells: role of target steroid hormones in cellular responsiveness to CYP1A1 induction. Jana NR, Sarkar S, Ishizuka M, Yonemoto J, Tohyama C, Sone H. Chemical Exposure and Health Effects Research Team, National Institute for Environmental Studies, 16-2 Onogawa,Tsukuba, Ibaraki 305-0053, Japan. A study was conducted to investigate whether target hormones affect 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible gene expression, using as an experimental model system three human cancer cell lines, breast (MCF-7), uterine (RL95-2), and prostate (LNCaP). Exposure to TCDD induced the CYP1A1 gene in all three cell lines. MCF-7 and RL95-2 cells showed more than 15- and 10-fold induction of EROD (7-ethoxyresorufin O-deethylase) activity, respectively, compared with the less responsive LNCaP cells. Surprisingly, however, TCDD-induced reporter gene activity driven by a single XRE element was similar in RL95-2 and LNCaP cells. The steady-state levels of expression of aryl hydrocarbon receptor (AhR) and aryl hydrocarbon receptor nuclear translocator (ARNT) were similar in all three cell lines. Expression of the CYP1B1 and PAI-2 genes was induced by TCDD in MCF-7 and RL95-2, but not in LNCaP, cells. Transient coexpression of estradiol receptor-alpha (ER-alpha) with a TCDD-responsive reporter plasmid and subsequent TCDD treatment increased responsiveness to TCDD in RL95-2 and LNCaP cells. Treatment with AZA-C, a DNA methyltransferase inhibitor, enhanced responsiveness to TCDD, in terms of EROD activity in LNCaP cells, but not in MCF-7 and RL95-2 cells, suggesting that DNA methylation in the CpG dinucleotide within the XRE core sequence is another factor involved in silencing of CYP1A1 in LNCaP cells. TCDD markedly inhibited E(2)- or testosterone-induced reporter gene activities in all three cell lines. Conversely, these target hormones inhibited TCDD-induced EROD activity in the three cell lines. These findings suggest that TCDD and the target steroid hormones negatively regulate each other's activity. Copyright 2000 Academic Press. PMID: 11281733 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 169: Drug Metab Rev. 2001 Feb;33(1):37-47. Competitive inhibition of the transcription of rabbit CYP1A1 gene by upstream stimulatory factor 1 (USF1). Takahashi Y, Kamataki T. Laboratory of Drug Metabolism, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan. The induction of CYP1A1 by 3-methylcholanthrene occurs in neonatal but not in adult rabbits. The expression of aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (Arnt) mRNAs is seen even in adult rabbits. The CYP1A1 inducibility does not seem to be regulated by DNA methylation, known to inhibit the transcription of a gene(s). Preliminary experiments suggest that a constitutive factor(s) in adult liver nuclear extracts is bound to the core sequence of rabbit xenobiotic-responsive element (XRE). The sequence of rabbit XRE overlaps with that of the upstream stimulatory factor 1 (USF1)-binding site. The AhR/Arnt-mediated activation of XRE-TK/Luc reporter gene in RK13 cells is blocked by transfection with a USF1 expression vector. These results indicate that the XRE of the rabbit CYP1A1 gene is recognized by the basic helix-loop-helix proteins to regulate the expression of CYP1A1 in both an agonistic (AhR/Arnt) and an antagonistic (USF1) manner. Publication Types: Review Review, Tutorial PMID: 11270661 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 170: Mol Pharmacol. 2001 Apr;59(4):664-73. Maximal aryl hydrocarbon receptor activity depends on an interaction with the retinoblastoma protein. Elferink CJ, Ge NL, Levine A. Institute of Chemical Toxicology, Wayne State University, Detroit, Michigan 48201, USA. cornelis_elferink@wayne.edu The aryl hydrocarbon receptor (AhR) belongs to the basic helix-loop-helix/periodicity/AhR nuclear translocator/simple-minded (Per-Arnt-Sim) family of transcription factors that regulate critical functions during development and tissue homeostasis. Within this family, the AhR is the only member conditionally activated in response to ligand binding, typified by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). We recently demonstrated that the AhR interacts with the retinoblastoma protein (pRb). This report presents evidence that a LXCXE motif in the AhR protein confers pRb binding, which is necessary for maximal TCDD induced G(1) arrest in rat 5L hepatoma cells. The data support a mechanism whereby pRb seems to regulate G(1) cell cycle progression distinct from the direct repression of E2F-mediated transcription. Furthermore, the results indicate that the AhR-pRb interaction regulates TCDD induction of CYP1A1, suggesting that pRb may be a general AhR coactivator. PMID: 11259609 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 171: Mol Cell Biol. 2001 Mar;21(5):1700-9. Proteasome inhibition induces nuclear translocation and transcriptional activation of the dioxin receptor in mouse embryo primary fibroblasts in the absence of xenobiotics. Santiago-Josefat B, Pozo-Guisado E, Mulero-Navarro S, Fernandez-Salguero PM. Departamento de Bioquimica y Biologia Molecular y Genetica, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain. The aryl hydrocarbon receptor (AHR) is a transcription factor that is highly conserved during evolution and shares important structural features with the Drosophila developmental regulators Sim and Per. Although much is known about the mechanism of AHR activation by xenobiotics, little information is available regarding its activation by endogenous stimuli in the absence of exogenous ligand. In this study, using embryonic primary fibroblasts, we have analyzed the role of proteasome inhibition on AHR transcriptional activation in the absence of xenobiotics. Proteasome inhibition markedly reduced cytosolic AHR without affecting its total cellular content. Cytosolic AHR depletion was the result of receptor translocation into the nuclear compartment, as shown by transient transfection of a green fluorescent protein-tagged AHR and by immunoblot analysis of nuclear extracts. Gel retardation experiments showed that proteasome inhibition induced transcriptionally active AHR-ARNT heterodimers able to bind to a consensus xenobiotic-responsive element. Furthermore, nuclear AHR was transcriptionally active in vivo, as shown by the induction of the endogenous target gene CYP1A2. Synchronized to AHR activation, proteasome inhibition also induced a transient increase in AHR nuclear translocator (ARNT) at the protein and mRNA levels. Since nuclear levels of AHR and ARNT are relevant for AHR transcriptional activation, our data suggest that proteasome inhibition, through a transient increase in ARNT expression, could promote AHR stabilization and accumulation into the nuclear compartment. An elevated content of nuclear AHR could favor AHR-ARNT heterodimers able to bind to xenobiotic-responsive elements and to induce gene transcription in the absence of xenobiotics. Thus, depending on the cellular context, physiologically regulated proteasome activity could participate in the control of endogenous AHR functions. PMID: 11238907 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 172: Biochem Pharmacol. 1999 May 15;57(10):1177-90. Functional analysis of activation and repression domains of the rainbow trout aryl hydrocarbon receptor nuclear translocator (rtARNT) protein isoforms. Necela B, Pollenz RS. Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA. The aryl hydrocarbon receptor nuclear translocator (ARNT) protein is involved in many signaling pathways. Rainbow trout express isoforms of ARNT protein that are divergent in their C-terminal domains due to alternative RNA splicing. Rainbow trout ARNT(b) (rtARNT(b)) contains a C-terminal domain rich in glutamine and asparagine (QN), whereas the C-terminal domain of rtARNT(a) is rich in proline, serine, and threonine (PST). rtARNT(b) functions positively in AH receptor-mediated signaling, whereas rtARNT(a) functions negatively. Studies were performed to understand how changes in the C-terminal domains of the two rtARNT isoforms affect function. Deletion of the QN-rich C-terminal domain of rtARNT(b) did not affect function in aryl hydrocarbon receptor (AHR)-mediated signaling, whereas deletion of the PST-rich domain of rtARNT(a) restored function. Expression of the PST-rich domain on truncated rtARNT(b) or mouse ARNT (mARNT) reduced function of this protein by 50-80%. Gel shift assays revealed that the PST-rich domain affected AHR-mediated signaling by inhibiting DNA binding of the AHR*ARNT heterodimer. Gal4 transactivation assays revealed a potent transactivation domain in the QN-rich domain of rtARNT(b). In contrast, Gal4 proteins containing the PST-rich domain of rtARNT(a) did not transactivate because the proteins did not bind to DNA. Secondary structure analysis of the PST-rich domain revealed hydrophilic and hydrophobic regions. Truncation of the hydrophobic domain that spanned the final 20-40 amino acids of the rtARNT(a) restored function to the protein, suggesting that repressor function was related to protein misfolding or masking of the basic DNA binding domain. Functional diversity within the C-terminal domain is consistent with other negatively acting transcription factors and illustrates a common biological theme. PMID: 11230806 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 173: Nippon Rinsho. 2000 Dec;58(12):2446-51. [Regulatory mechanism of genes by the Ah receptor which mediates toxic effects of dioxins] [Article in Japanese] Sogawa K. Department of Chemistry, Graduate School of Science, Tohoku University. The aryl hydrocarbon receptor(AhR) plays a central role in the metabolic pathways involved in the detoxification of important environmental carcinogens, most of which act as ligand for the receptor, although no endogeneous ligand has not yet been known. Activation of the AhR is responsible for a variety of toxic responses in animals and humans. The activation mechanisms become clear that include binding of ligand to receptor, transfer to the nucleus, formation of a ternary complex with Arnt, followed by binding to response elements upstream of the relevant target genes. However, the specific mechanisms responsible for the toxic responses of dioxins are unknown. Publication Types: Review Review, Tutorial PMID: 11187735 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 174: Pharmacogenetics. 2000 Nov;10(8):741-51. An uncommon phenotype of poor inducibility of CYP1A1 in human lung is not ascribable to polymorphisms in the AHR, ARNT, or CYP1A1 genes. Anttila S, Lei XD, Elovaara E, Karjalainen A, Sun W, Vainio H, Hankinson O. Department of Pathology and Laboratory Medicine and Johnson Comprehensive Cancer Center, University of California, Los Angeles, USA. Cigarette smoking can induce CYP1A1 in the lung. Induction requires the aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) proteins. Lung samples from seven of 75 Finnish patients who smoked until the time of surgery exhibited absent or low levels of CYP1A1 protein, mRNA and enzymatic activity, suggesting that these individuals might be genetically non or poorly inducible for CYP1A1. All seven lung samples expressed normal levels of AHR mRNA and ARNT mRNA, indicating that they did not carry inactivating polymorphisms in the 5' upstream regulatory regions of these genes. Sequencing of cDNAs encompassing the complete coding regions of AHR and ARNT identified a previously known codon 554 polymorphism in AHR, which was present in the homozygous state in one individual. This polymorphism, which leads to an amino acid substitution, has previously been reported either to have no effect or to enhance CYP1A1 induction. Previously unreported silent single nucleotide polymorphisms were identified in codon 44 of AHR and codon 189 of ARNT. 1500 bp of genomic sequence from the 5' upstream regulatory sequence of the CYP1A1 gene was also sequenced in the non-inducible individuals. A nucleotide substitution polymorphism at position -459 was detected in the heterozygous state in two individuals. This polymorphic site does not reside in any known regulatory sequence. The complete CYP1A1 coding sequence and intron/exon boundaries were then sequenced. None of the non or poorly inducible individuals exhibited any polymorphisms, either homozygous or heterozygous compared to representative inducible individuals or the previously published CYP1A1 sequence. Thus, no polymorphisms in the AHR, ARNT or CYP1A1 genes were identified that could be responsible for the non/low inducibility phenotype observed. Publication Types: Case Reports PMID: 11186136 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 175: Mol Pharmacol. 2001 Mar;59(3):557-66. Aryl hydrocarbon receptor (AhR)/AhR nuclear translocator (ARNT) activity is unaltered by phosphorylation of a periodicity/ARNT/single-minded (PAS)-region serine residue. Levine SL, Perdew GH. Department of Veterinary Science and Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania, USA. The aryl hydrocarbon nuclear translocator (ARNT) protein belongs to the family of basic helix-loop-helix (HLH)-periodicity/ARNT/single-minded [Per/ARNT/Sim (PAS)] transcription factors and regulates a range of cellular processes by either homodimerizing or heterodimerizing with other basic HLH-PAS proteins. To date, it has been shown that both the HLH and PAS domains are required for aryl hydrocarbon receptor (AhR) ARNT heterodimerization and that phosphorylation of ARNT is also required for this heterodimerization. Presently, regulation of ARNT with respect to phosphorylation is poorly understood. In an earlier study, murine ARNT was shown to be a phosphoprotein, to display charge heterogeneity, and to have a shift in its predominant isoforms after heterodimerization with the AhR. It was hypothesized that this shift may represent a change in ARNT phosphorylation status. Metabolic [(32)P]orthophosphate labeling of human ARNT-transfected COS-1 cells, in conjunction with phosphoamino acid analysis, Edman degradation, and phosphopeptide mapping, demonstrated that ARNT is predominantly phosphorylated on serine residues and that serine 348 (S348) in the PAS domain is phosphorylated. Alanine and glutamic acid substitutions were used to demonstrate that loss of phosphorylation at this site did not influence AhR-mediated xenobiotic response elements-driven or ARNT-mediated class B E-box-driven signaling. Additionally, the phosphorylation pattern of ARNT was unaltered after AhR heterodimerization. Although phosphorylation of S348 did not modulate AhR-ARNT or ARNT-ARNT signaling, phosphorylation of this PAS-region serine residue may be important in other ARNT-mediated gene expression systems. PMID: 11179451 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 176: Arch Biochem Biophys. 2000 Dec 1;384(1):190-8. Failure of Ah receptor to mediate induction of cytochromes P450 in the CYP1 family in the human hepatoma line SK-Hep-1. Roberts EA, Harper PA, Wong JM, Wang Y, Yang S. Metabolism Programme, The Hospital for Sick Children Research Institute, University of Toronto, Ontario, Canada. eve.roberts@sickkids.on.ca The Ah receptor mediates the induction of cytochrome P450 1A1 (CYP1A1) and toxicities of 2,3,7,8tetrachlorodibanzo-p-dioxin (TCDD). It has been detected in tissues of many species and in murine and human hepatoma lines. We show that the human hepatoma line SK-Hep-1 has cytosolic Ah receptor detectable by specific binding of [3H]TCDD. Concentrations of Ah receptor were low (mean = 43 +/- 3 fmol/mg cytosol protein compared to 430 fmol/mg protein in Hepa-1); the estimated number of receptor sites per cell is approximately 9,000, compared to 35,000 in Hepa-1. Ah receptor in SK-Hep-1 cells was physicochemically similar to Ah receptor in C57BL/6 mouse liver and in other human hepatoma lines studied to date except that binding affinity for TCDD, the most avidly bound ligand, was lower (estimated Kd was 14 nM by Woolf plot analysis). Translocation of the Ah receptor-ligand complex to the nucleus was shown; binding of the activated Ah receptor-ligand complex to an XRE in the 5'-upstream region of the CYP1A1 gene was demonstrated by gel-shift analysis. However, after SK-Hep-1 cells were incubated with typical PAHs including 3-methylcholanthrene, benzanthracene, and dibenz(a,h)anthracene, each over a wide range of concentrations, no induction of aryl hydrocarbon hydroxylase activity was detectable. On Northern analysis, no message for human CYP1A1 was detected in mRNA prepared from noninduced SK-Hep-1 cells or from cells treated for 24 h with 13 microM dibenz(a,h)anthracene. Further analysis by RT-PCR did not detect the induction of CYP1A1, CYP1A2, or CYP1B1 message in response to 10(-7) M TCDD, 10(-5) M benzanthracene, or 10(-5) M 3-methylcholanthrene. Transient transfection of reporter constructs containing either a minimal promoter or the CYP1A1 promoter fused to a reporter gene (luciferase) did not show any expression in response to increasing concentrations of TCDD up to 10(-8) M. Estimation of the size of the transcripts for AhR and ARNT protein revealed normal sizes, 2.7 and 2.4 kb, respectively. Together, these data suggest that SK-Hep-1 cells express an Ah receptor defective at the level of trans-activation of gene expression. SK-Hep-1 is the first human hepatoma line described with a demonstrable defect in CYP1A1 or its regulation. PMID: 11147830 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 177: Toxicol Appl Pharmacol. 2000 Dec 1;169(2):159-67. Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin on the expression of cytochrome P450 1A1, the aryl hydrocarbon receptor, and the aryl hydrocarbon receptor nuclear translocator in rat brain and pituitary. Huang P, Rannug A, Ahlbom E, Hakansson H, Ceccatelli S. Division of Toxicology and Neurotoxicology, National Institute of Environmental Medicine, Karolinska Institutet, S-171 77, Stockholm, Sweden. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related substances are ubiquitous environmental pollutants causing a wide variety of pathological alterations, with the most severe being progressive anorexia and body weight loss. These features suggest a possible involvement of the nervous system and neuroendocrine-related organs including the pituitary gland. However, so far there is little evidence for direct effects of TCDD on these areas. In the present study, male Sprague-Dawley rats were treated with a single oral dose of TCDD (10 microg/kg) and euthanized 1, 3, or 28 days after treatment. The expression of cytochrome P450 1A1 (CYP1A1), the aryl hydrocarbon receptor (AHR), and the aryl hydrocarbon receptor nuclear translocator (ARNT) were analyzed in different brain regions and pituitaries using semiquantitative RT-PCR and Western blotting. Relative levels of CYP1A1 mRNA and protein were dramatically increased in the pituitary. A significant increase in CYP1A1 mRNA was also detected in all the brain regions examined including olfactory bulb, striatum-caudate, hypothalamus, hippocampus, cortex, cerebellum, and substantia nigra. The increase in the expression was time-dependent with the highest level observed 1 day after TCDD treatment. The AHR and ARNT mRNAs were detected in the same areas but in contrast to CYP1A1 the changes in AHR and ARNT mRNA expression were limited to the 28-day time point. The present results provide evidence for the presence of CYP1A1, AHR, and ARNT in the central nervous system and in the pituitary, suggesting that TCDD may exert a direct effect on these regions. Copyright 2000 Academic Press. PMID: 11097868 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 178: J Biol Chem. 2001 Feb 16;276(7):4819-27. Epub 2000 Nov 28. Aromatic hydrocarbon receptor (AhR).AhR nuclear translocator- and p53-mediated induction of the murine multidrug resistance mdr1 gene by 3-methylcholanthrene and benzo(a)pyrene in hepatoma cells. Mathieu MC, Lapierre I, Brault K, Raymond M. Institut de Recherches Cliniques de Montreal, Montreal, Quebec H2W 1R7, Canada. The mouse multidrug resistance gene family consists of three genes (mdr1, mdr2, and mdr3) encoding P-glycoprotein. We show that the expression of mdr1 is increased at the transcriptional level upon treatment of the hepatoma cell line Hepa-1c1c7 with the polycyclic aromatic hydrocarbon 3-methylcholanthrene (3-MC). This increase is not observed in the aromatic hydrocarbon receptor (AhR)-defective TAOc1BP(r)c1 and the AhR nuclear translocator (Arnt)-defective BP(r)c1 variants, demonstrating that the induction of mdr1 by 3-MC requires AhR.Arnt. We show that the mdr1 promoter (-1165 to +84) is able to activate the expression of a reporter gene in response to 3-MC in Hepa-1c1c7 but not in BP(r)c1 cells. Deletion analysis indicated that the region from -245 to -141 contains cis-acting sequences mediating the induction, including a potential p53 binding sequence. 3-MC treatment of the cells increased the levels of p53 and induced p53 binding to the mdr1 promoter in an AhR.Arnt-dependent manner. Mutations in the p53 binding site abrogated induction of mdr1 by 3-MC, indicating that p53 binding to the mdr1 promoter is essential for the induction. Benzo(a)pyrene, a polycyclic aromatic hydrocarbon and AhR ligand, which, like 3-MC, is oxidized by metabolizing enzymes regulated by AhR.Arnt, also activated p53 and induced mdr1 transcription. 2,3,7,8-Tetrachlorodibenzo-p-dioxin, an AhR ligand resistant to metabolic breakdown, had no effect. These results indicate that the transcriptional induction of mdr1 by 3-MC and benzo(a)pyrene is directly mediated by p53 but that the metabolic activation of these compounds into reactive species is necessary to trigger p53 activation. The ability of the anticancer drug and potent genotoxic agent daunorubicin to induce mdr1 independently of AhR.Arnt further supports the proposition that mdr1 is transcriptionally up-regulated by p53 in response to DNA damage. PMID: 11096091 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 179: Toxicology. 2000 Nov 16;153(1-3):73-82. Is CYP1A1 induction always related to AHR signaling pathway? Delescluse C, Lemaire G, de Sousa G, Rahmani R. Laboratoire de Pharmaco-Toxicologie Cellulaire et Moleculaire, INRA, Centre de Recherches, 41 Bd du Cap, 06606, Antibes, France. Humans are daily subjected to ever increasing amounts of exogenous compounds. Some of them are capable of inducing cytochrome P450s, a process that allows the cell to adapt to changes in its chemical environment. One of the most widely CYP studied is CYP1A1 because it metabolises a large number of xenobiotics to cytotoxic and/or mutagenic derivatives. To date, results from the literature indicate that induction of CYP1A1 does not only involve the classical activation cascade of the Ah receptor, e.g. binding of the ligand to the AhR, heterodimerisation with Arnt protein, constitution of a complex with XRE responsive element and subsequent gene activation. Indeed, some xenobiotics do activate CYP1A1 gene expression in spite of their inability to compete with TCDD for binding to the AhR. Other signaling pathways must therefore also be considered. Firstly, the CYP1A1 inducer compounds could be very weak AhR ligands or may be metabolized into a form which is in turn capable of binding to the Ah receptor. A second hypothesis would be that these molecules could act through other signaling cascades. At this time, two of them seem to be implicated. One concerns the RARs signal transduction pathway, as already described for retinoic acid. The second may involve tyrosine kinase activation, but the precise relationship between this activation and CYPA1 induction remains yet to be established. For the moment there is still a black box which needs to be investigated. Publication Types: Review Review, Tutorial PMID: 11090948 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 180: J Biomol Struct Dyn. 2000 Oct;18(2):169-79. A model for the complex between the hypoxia-inducible factor-1 (HIF-1) and its consensus DNA sequence. Michel G, Minet E, Ernest I, Roland I, Durant F, Remacle J, Michiels C. Laboratoire de Chimie Moleculaire Structurale, Facultes Universitaires, Notre-Dame-de-la-Paix, Namur, Belgium. gaetan.michel@fundp.ac.be Hypoxia-inducible factor-1 (HIF-1) is a heterodimeric transcription factor activated by hypoxia. When activated, HIF-1 mediates the differential expression of genes such as erythropoietin and Vascular Endothelial Growth Factor (VEGF) during hypoxia. It is composed of two different subunits, HIF-1alpha and ARNT (Aryl Receptor Nuclear Translocator). These two subunits belong to the bHLH (basic Helix-Loop-Helix) PAS (Per, Ahr/ARNT, Sim) family. The bHLH domain of these factors is responsible for dimerization through the two helices and for DNA binding through their basic domain. In this work, we used various methods of molecular modeling in order to develop a 3D structure for the HIF-1 bHLH domain bound to its DNA consensus sequence. Firstly, the 3D structure of the bHLH domain of both subunits based on their amino acid sequence was defined. Secondly, we compared this model with data from known crystal structures of basic leucine zipper-DNA and bHLH-DNA complexes in order to determine a potential canvas for HIF-1. Thirdly, we performed a manual approach of the HIF-1 bHLH domain onto the DNA recognition site using this canvas. Finally, the protein-DNA complex 3D structure was optimized using a Monte Carlo program called MONTY. The model predicted a pattern of interactions between amino acids and DNA bases which reflect for ARNT what is experimentally observed among different X-ray structures of other bHLH transcription factors possessing the H (His), E (Glu), R (Arg) triad, as ARNT does. On the other hand, only the Arg residue is conserved in HIF- 1alpha. We propose from this model that a serine replaces the histidine while an alanine and a lysine also make contacts with DNA. From these results, we postulate that the specificity of HIF-1 toward its DNA sequence could be driven by the HIF-1alpha subunit. The predicted model will be verified by X-Ray currently ongoing. PMID: 11089639 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 181: Biochim Biophys Acta. 2000 Nov 15;1494(1-2):117-28. Identification and expression of alternatively spliced aryl hydrocarbon nuclear translocator 2 (ARNT2) cDNAs from zebrafish with distinct functions. Tanguay RL, Andreasen E, Heideman W, Peterson RE. Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Health Sciences Center, Denver 80262, USA. robert.tanguay@uchsc.edu In order to further establish zebrafish as a vertebrate model for studying the mechanism of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) toxicity it is necessary to characterize the aryl hydrocarbon receptor/aryl hydrocarbon receptor nuclear translocator (AhR/ARNT) signaling pathways in this species. In this study, three zfARNT2 cDNAs were isolated, expressed, and characterized and named zfARNT2b, zfARNT2c, and zfARNT2a. zfARNT2b, zfARNT2c, and zfARNT2a encode proteins with theoretical molecular weights of 81, 79, and 45 kDa, respectively. zfARNT2b and zfARNT2a proteins are identical over the first 403 amino acids but differ in their C-terminal domains as a result of alternative mRNA splicing. zfARNT2c is nearly identical to zfARNT2b, with the exception of an in frame 15 amino acid deletion adjacent to the basic region of zfARNT2c. Using quantitative RT-PCR methods the tissue distribution of each zfARNT2 isoform was determined. In COS-7 cells expressing zfARNT2b and zfAhR2, 10 nM TCDD causes a nine-fold induction of a dioxin responsive reporter gene. In COS-7 cells expressing zfARNT2a or zfARNT2c, TCDD does not induce reporter gene expression. In contrast, all three zfARNT2 proteins induce reporter gene activity under control of hypoxia responsive elements when cotransfected with the zebrafish endothelial specific PAS protein 1. DNA gel shift analysis suggests that the decreased function of zfARNT2a is due to inefficient binding of zfARNT2a/zfAhR2 complexes to dioxin responsive elements. These results also indicate that alternative mRNA splicing results in formation of ARNT proteins with distinct functional properties. PMID: 11072074 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 182: J Comp Neurol. 2000 Nov 20;427(3):428-39. Distribution of mRNAs encoding the arylhydrocarbon receptor, arylhydrocarbon receptor nuclear translocator, and arylhydrocarbon receptor nuclear translocator-2 in the rat brain and brainstem. Petersen SL, Curran MA, Marconi SA, Carpenter CD, Lubbers LS, McAbee MD. Department of Biology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003, USA. sandyp@bio.umass.edu Dioxin exposure alters a variety of neural functions, most likely through activation of the arylhydrocarbon receptor (AhR) pathway. Many of the adverse effects, including disruption of circadian changes in hormone release and depressed appetite, seem to be mediated by hypothalamic and/or brainstem neurons. However, it is unclear whether these effects are direct or indirect, because there have been no comprehensive studies mapping the expression of components of the AhR pathway in the brain. Therefore, we used a sensitive in situ hybridization histochemical (ISHH) method to map the neural expression of AhR mRNA, as well as those of the mRNAs encoding the AhR dimerization partners, arylhydrocarbon receptor nuclear translocator (ARNT) and ARNT2. We found that AhR, ARNT, and ARNT2 mRNAs were widely distributed throughout the brain and brainstem. There was no neuroanatomic evidence that AhR is preferentially colocalized with ARNT or ARNT2. However, ARNT2, unlike ARNT expression, was relatively high in most regions. The most noteworthy regions in which we found AhR, ARNT, and ARNT2 mRNA were several hypothalamic and brainstem regions involved in the regulation of appetite and circadian rhythms, functions that are disrupted by dioxin exposure. These regions included the arcuate nucleus (Arc), ventromedial hypothalamus (VMH), paraventricular nucleus (PVN), suprachiasmatic nucleus (SCN), nucleus of the solitary tract (NTS), and the dorsal and median raphe nuclei. This neuroanatomic information provides important clues as to the sites and mechanisms underlying the previously unexplained effects of dioxins in the central nervous system. Copyright 2000 Wiley-Liss, Inc. PMID: 11054704 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 183: Comp Biochem Physiol C Toxicol Pharmacol. 2000 Jul;126(3):305-19. Molecular characterization and developmental expression of the aryl hydrocarbon receptor from the chick embryo. Walker MK, Heid SE, Smith SM, Swanson HI. College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque 87131, USA. mkwalker@unm.edu The aryl hydrocarbon receptor (AhR) was cloned from the chick embryo and its function and developmental expression characterized. Chicken AhR cDNA coded for 858 amino acid protein and 396 bp of 3' UTR. The basic helix loop helix domain exhibited 87-100% amino acid identity to avian, mammalian, and amphibian AhR, and 69-74% to piscine AhR. The PAS (Per-ARNT-Sim) region was slightly less well conserved with (a) 97% identity to other avian sequences, (b) 81-86% to amphibian and mammalian AhR, and (c) 64-69% with piscine AhR. The carboxy terminus diverged the most among species with less than 53% amino acid identity between chicken and any available mammalian and piscine AhR sequences. The chicken AhR RNA and protein were 6.1 kb and 103 kDa, respectively. Chicken AhR dimerized with human AhR nuclear translocator and bound the mammalian dioxin-response element in a ligand-dependent manner. AhR protein was detected in neural ganglia; smooth, cardiac, and skeletal muscle; and epithelium involved in epithelial-to-mesenchymal transformations, such as pituitary, gastrointestinal tract, limb apical-ectodermal ridge, and kidney collecting ducts. AhR mRNA was detected in all tissues expressing protein, except myocardium. Cytochrome P4501A4 mRNA was highly induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in a subset of tissues expressing AhR, including small intestine, liver, kidney, blood vessels, and outflow tract myocardium. In conclusion, the AhR sequence and function is highly conserved between birds and mammals, and although many tissues express AhR during chick embryo development, only a subset are responsive to TCDD induction of CYP1A4. PMID: 11048681 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 184: Mol Endocrinol. 2000 Oct;14(10):1674-81. Conditional disruption of the aryl hydrocarbon receptor nuclear translocator (Arnt) gene leads to loss of target gene induction by the aryl hydrocarbon receptor and hypoxia-inducible factor 1alpha. Tomita S, Sinal CJ, Yim SH, Gonzalez FJ. Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland 20892, USA. To determine the function of the aryl hydrocarbon receptor nuclear translocator (ARNT), a conditional gene knockout mouse was made using the Cre-loxP system. Exon 6, encoding the conserved basic-helix-loop-helix domain of the protein, was flanked by loxP sites and introduced into the Arnt gene by standard gene disruption techniques using embryonic stem cells. Mice homozygous for the floxed allele were viable and had no readily observable phenotype. The Mx1-Cre transgene, in which Cre is under control of the interferon-gamma promoter, was introduced into the Arnt-floxed mouse line. Treatment with polyinosinic-polycytidylic acid to induce expression of Cre resulted in complete disruption of the Arnt gene and loss of ARNT messenger RNA (mRNA) expression in liver. To determine the role of ARNT in gene control in the intact animal mouse liver, expression of target genes under control of an ARNT dimerization partner, the aryl hydrocarbon receptor (AHR), was monitored. Induction of CYP1A1, CYP1A2, and UGT1*06 mRNAs by the AHR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin was absent in livers of Arnt-floxed/Mx1-Cre mice treated with polyinosinic-polycytidylic. These data demonstrate that ARNT is required for AHR function in the intact animal. Partial deletion of the Arnt allele was found in kidney, heart, intestine, and lung. Despite more than 80% loss of the ARNT expression in lung, maximal induction of CYP1A1 was found, indicating that the expression level of ARNT is not limiting to AHR signaling. Cobalt chloride induction of the glucose transporter-1 and heme oxygenase-1 mRNAs was also markedly abrogated in mice lacking ARNT expression, suggesting an inhibition of HIF-1alpha activity. These studies establish a critical role for ARNT in AHR and HIF-1alpha signal transduction in the intact mouse. PMID: 11043581 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 185: Arch Biochem Biophys. 2000 Sep 15;381(2):302-12. Regulation of DNA binding activity of the ligand-activated aryl hydrocarbon receptor by tyrosine phosphorylation. Park S, Henry EC, Gasiewicz TA. Department of Environmental Medicine, School of Medicine, University of Rochester, New York 14642, USA. Aryl hydrocarbon receptor (AhR), a member of the bHLH-PAS family, is a ligand-activated transcription factor which plays an important role in normal liver development and in mediating the toxicity of polycyclic and halogenated aromatic hydrocarbon pollutants such as 2,3,7,8-tetrachlorodibenzo-p-dioxin. Phosphorylation is known to regulate the transformation process of unliganded AhR into functionally active AhR/ARNT heterodimer that has high affinity for dioxin-responsive elements (DRE) and transactivation activity. Here, we report that DRE binding activity of the AhR is regulated by phosphorylation on the AhR/ARNT complex itself. Studies with specific protein phosphatases indicated that tyrosine phosphorylation is involved in this modulation. In addition, the AhR is phosphorylated at tyrosine residue(s) as determined by anti-phosphotyrosine immunoblot analysis. These results suggest that tyrosine phosphorylation on the AhR is required for its DNA binding activity and may provide mammalian cells with another layer of control mechanism that allows cell type specific and developmental stage specific induction of the AhR target genes. PMID: 11032419 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 186: Mol Cell Endocrinol. 2000 Jun;164(1-2):5-18. Demonstration of 2,3,7,8-tetrachlorodibenzo-p-dioxin attenuation of P450 steroidogenic enzyme mRNAs in rat granulosa cell in vitro by competitive reverse transcriptase-polymerase chain reaction assay. Dasmahapatra AK, Wimpee BA, Trewin AL, Wimpee CF, Ghorai JK, Hutz RJ. Department of Biological Sciences, University of Wisconsin-Milwaukee, 53211, USA. We investigated the effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), in prepubertal (PP) and adult (A) rat granulosa cells (GC) in vitro by examining the changes in estrogen secretion, aromatase enzyme activity and mRNAs for steroidogenic enzymes P450scc, 3beta-HSDI, P450arom; and for components of the AHR signaling pathway-CYP1A1, aromatic hydrocarbon receptor (AHR), and the AHR nuclear translocator protein (ARNT). In PP and A rat GC, TCDD (3.1 nM) reduced estrogen secretion at 48 h without altering aromatase enzyme activity. Addition of FSH (50 ng/ml) increased aromatase activity in GC with or without TCDD. FSH-induced aromatase activity was significantly reduced by TCDD (3.1 nM) at 48 h. Semi-quantitative RT-PCR showed a significant increase in CYP1A1 mRNA both at 24 and 48 h with TCAP, while a significant reduction in P450scc and P450arom mRNA was observed with competitive RT-PCR. All steroidogenic enzyme mRNAs were significantly lower in adults than in PP GC. We conclude that in rat GC, TCDD modulates the level of cytochrome P450 enzymes involved in the steroid biosynthetic cascade. This effect may be attributable to AHR interaction with dioxin-responsive elements present in the genes encoding these enzymes. PMID: 11026553 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 187: Gene Expr. 1999;8(5-6):273-86. Nuclear receptor coactivator SRC-1 interacts with the Q-rich subdomain of the AhR and modulates its transactivation potential. Kumar MB, Perdew GH. Graduate Program in Biochemistry and Molecular Biology, The Pennsylvania State University, University Park 16802, USA. The aryl hydrocarbon receptor (AhR), a soluble cytosolic protein, mediates many of the toxic effects of TCDD and related chemicals. The toxic effects are largely cell, tissue, and promoter context dependent. Although many details of the overall dioxin signal transduction have been elucidated, the transcriptional regulation of dioxin-induced genes like cyp1A1 is not yet completely understood. Previously, we have shown that the co-regulator RIP140 is a potential AhR coactivator. In this report, the role of coactivator, SRC-1, in AhR-mediated transcriptional regulation was examined. SRC-1 increased AhR-mediated, TCDD-dependent reporter gene activity three-fold in Hepa-1 and COS-1 cells. In in vitro interaction assays, SRC-1 was found to interact with AhR but not with ARNT. SRC-1 interacted weakly with AhR in the absence of TCDD and the addition of ligand further increased SRC-1 binding to AhR. Deletional mapping studies of the AhR revealed that SRC-1 binds to the AhR transactivation domain. Finer mapping of the SRC-1-interacting subdomains in the AhR transactivation domain suggested that the Q-rich subdomain was necessary and sufficient for interaction, similar to that seen with RIP140. Using GFP-tagged constructs, SRC-1 was shown to interact with AhR in cells. Unlike RIP140, LXXLL motifs in SRC-1 were necessary for interaction with AhR in vitro and for coactivation in Hepa-1 cells. The recruitment of certain coactivators by a variety of receptors suggests possible common coactivator pools and competition among receptors for limiting coactivators. Examination of the role of SRC-1 in AhR/ARNT transactivation in ARNT-deficient mutant Hepa-1 c4 cells demonstrates that the AhR transactivation domain is sufficient for enhanced coactivation mediated by SRC-1 in the presence of a transactivation domain deleted ARNT protein. PMID: 10947077 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 188: Mol Cell Biol. 2000 Aug;20(16):6095-104. Analysis of the complex relationship between nuclear export and aryl hydrocarbon receptor-mediated gene regulation. Pollenz RS, Barbour ER. Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA. rpollen@chuma.cas.usf.edu The aryl hydrocarbon receptor (AHR) contains signals for both nuclear import and nuclear export (NES). The purpose of the studies in this report was to determine the relationship between the nuclear export of the AHR and AHR-mediated gene regulation. Blockage of nuclear export in HepG2 cells with leptomycin B (LMB) resulted in increased levels of AHR-AHR nuclear translocator (ARNT) complex in the nucleus and correlative reductions in agonist-stimulated AHR degradation. However, LMB exposure inhibited agonist-mediated induction of numerous AHR-responsive reporter genes by 75 to 89% and also inhibited induction of endogenous CYP1A1. LMB did not transform the AHR to a ligand binding species or affect activation by TCDD (2, 3,7,8-tetrachlorodibenzo-p-dioxin). Mutagenesis of leucines 66 and 71 of the putative AHR NES resulted in a protein with reduced function in dimerization to ARNT and binding to DNA, while alanine substitution at leucine 69 (AHR(A69)) resulted in an AHR that bound with ARNT and associated with DNA. AHR(A69) protein injected directly into the nuclei of E36 cells remained nuclear following 6 h of agonist stimulation. In transient-transfection assays, AHR(A69) accumulated within the nucleus was not degraded efficiently following agonist exposure. Finally, AHR(A69) supported induction of AHR-responsive reporter genes in an agonist-dependent manner. These findings show that it is possible to generate an AHR protein defective in nuclear export that is functional in agonist-mediated gene induction. This implies that the negative effect of LMB on agonist-mediated gene induction is independent of the nuclear export of the AHR. PMID: 10913191 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 189: Nucleic Acids Res. 2000 Jun 15;28(12):2286-91. An aryl hydrocarbon receptor conformation acts as the functional core of nuclear dioxin signaling. Kronenberg S, Esser C, Carlberg C. Abteilung Immunologie des Medizinischen Instituts fur Umwelthygiene and Institut fur Physiologische Chemie I, Heinrich-Heine-Universitat Dusseldorf, Postfach 10 10 07, D-40001 Dusseldorf, Germany. DNA-complexed heterodimers of the aryl hydrocarbon receptor (AhR) with the Ah receptor nuclear translocator (Arnt) are the molecular switches for nuclear signaling of 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD). AhR-Arnt heterodimers regulate genes involved in the metabolism of xenobiotics or fatty acids and various genes important for growth and differentiation. In this report several potent methods, such as the limited protease digestion, gel shift and gel shift clipping assays, allowed the investigation of ligand-stabilized conformations of AhR monomers in comparison to that of AhR-Arnt heterodimers. Interestingly, the ligand sensitivity of monomeric AhR was found to be very low at 25 nM, whereas DNA-dependent methods consistently provided EC(50) values between 0.12 and 0.6 nM for AhR in a heterodimeric complex, i. e. an approximate 100-fold higher ligand sensitivity. This indicates that complex formation of AhR with Arnt on DNA is an important and critical step in transforming AhR into a high affinity receptor for TCDD. A comparison of wild-type AhR with different C-terminal receptor truncations suggests that the PAS-B subregion of its PAS domain is of central importance for stabilization of a functional, i. e. ligand-sensitive, AhR-Arnt conformation, whereas the PAS-A subregion appears to be critical for dimerization of AhR and Arnt. In conclusion, the results of this study provide important information on the ligand sensitivity of AhR and AhR-Arnt heterodimer conformations. PMID: 10871357 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 190: Proc Natl Acad Sci U S A. 2000 Jun 6;97(12):6757-62. The t(1;12)(q21;p13) translocation of human acute myeloblastic leukemia results in a TEL-ARNT fusion. Salomon-Nguyen F, Della-Valle V, Mauchauffe M, Busson-Le Coniat M, Ghysdael J, Berger R, Bernard OA. U434 Institut National de la Sante et de la Recherche Medicale-Centre d'Etude du Polymorphisme Humain-Fondation Jean Dausset, 27 rue Juliette Dodu, 75010, Paris, France. The TEL/ETV6 gene is located at 12p13 and encodes a member of the ETS family of transcription factors. Translocated ETS leukemia (TEL) is frequently involved in chromosomal translocations in human malignancies, usually resulting in the expression of fusion proteins between the amino-terminal part of TEL and either unrelated transcription factors or protein tyrosine kinases. We have characterized a t(1;12)(q21;p13) translocation in an acute myeloblastic leukemia (AML-M2). At the protein level, the untranslocated TEL copy and, as a result of the t(1;12) translocation, a fusion protein between TEL and essentially all of aryl hydrocarbon receptor nuclear translocator (ARNT) are expressed. The involvement of ARNT in human leukemogenesis has not been previously described. The ARNT protein belongs to a subfamily of the "basic region helix-loop-helix" (bHLH) protein that shares an additional region of similarity called the PAS (Per, ARNT, SIM) domain. ARNT is the central partner of several heterodimeric transcription factors, including those containing the aryl hydrocarbon (dioxin) receptor (AhR) and the hypoxia-inducible factor 1alpha (HIF1alpha). Our results show that the TEL-ARNT fusion protein is the crucial product of the translocation and suggest that interference with the activity of AhR or HIF1alpha can contribute to leukemogenesis. PMID: 10829078 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 191: J Steroid Biochem Mol Biol. 2000 Apr;72(5):197-207. Estrogen and aryl hydrocarbon receptor expression and crosstalk in human Ishikawa endometrial cancer cells. Wormke M, Castro-Rivera E, Chen I, Safe S. Department of Veterinary Physiology and Pharmacology, Texas A & M University, TX 77843-4466, College Station, USA. Ishikawa endometrial cancer cells express the estrogen receptor (ER), and this study investigates aryl hydrocarbon receptor (AhR) expression and inhibitory AhR-ER crosstalk in this cell line. Treatment of Ishikawa cells with the AhR agonist [3H]2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) gave a radiolabeled nuclear complex that sedimented at 6.0 S in sucrose density gradients, and Western blot analysis confirmed that Ishikawa cells expressed human AhR and AhR nuclear translocator (Arnt) proteins. Treatment of Ishikawa cells with 10 nM TCDD induced a 9.7-fold increase in CYP1A1-dependent ethoxyresorufin O-deethylase (EROD) activity and a 10.5-fold increase in chloramphenicol acetyltransferase (CAT) activity in cells transfected with pRNH11c containing an Ah-responsive human CYP1A1 gene promoter insert (-1142 to +2434). Inhibitory AhR-ER crosstalk was investigated in Ishikawa cells using E2-induced cell proliferation and transcriptional activation assays in cells transfected with E2-responsive constructs containing promoter inserts from the progesterone receptor and vitellogenin A2 genes. AhR agonists including TCDD, benzo[a]pyrene (BaP) and 6-methyl-1,3,8-trichlorodibenzofuran, inhibited 32-47% of the E2-induced responses. In contrast, neither estrogen nor progesterone inhibited EROD activity induced by TCDD in Ishikawa cells, whereas inhibitory ER-AhR crosstalk was observed in ECC-1 endometrial cells suggesting that these interactions were cell context-dependent. PMID: 10822009 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 192: Biochem Pharmacol. 2000 Jun 15;59(12):1549-56. Inhibition of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-stimulated Cyp1a1 promoter activity by hypoxic agents. Kim JE, Sheen YY. College of Pharmacy, Ewha Womans University, # 11-1, Daehyun-dong, Sudaemun-ku, Seoul, South Korea. Since hypoxia-inducible factor-1alpha (HIF-1alpha) and the arylhydrocarbon receptor (AhR) shared the AhR nuclear translocator (Arnt) for hypoxia- and AhR-mediated signaling, respectively, it was possible to establish the hypothesis that hypoxia could regulate cytochrome P450 1a1 (Cyp1a1) expression. In order to test this hypothesis, we undertook to examine the effect of hypoxia on Cyp1a1 transcription in Hepa-I cells. Mouse Cyp1a1 5'-flanking DNA, 1.6 kb was cloned into pGL3 expression vector in order to construct pmCyp1a1-Luc. Hepa-I cells were transfected with pmCyp1a1-Luc and treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the presence or absence of various hypoxic agents such as 1-100 microM cobalt chloride, 1-100 microM picolinic acid, and 1-100 microM desferrioxamine. Luciferase activity of the reporter gene was measured from pmCyp1a1-Luc-transfected Hepa-I cell lysate which contains 2 microgram total protein using luciferin as a substrate. Hypoxic agents such as cobalt chloride, picolinic acid, and desferrioxamine showed inhibition of luciferase activity that was induced by 1-nM TCDD treatment in a dose-and time-dependent manner. Concomitant treatment of 150 microM ferrous sulfate with 1-100 microM desferrioxamine or 1-100 microM picolinic acid recovered luciferase activity from that inhibited by hypoxic agents or induced by TCDD. These data demonstrated that iron-chelating and hypoxic agents inhibited dioxin-induced Cyp1a1 transcription in Hepa-I cells. Thus, we might suggest that hypoxia inhibits TCDD-induced Cyp1a1 expression due to the competition between HIF-1alpha and the AhR for the Arnt in Hepa-I cells. PMID: 10799651 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 193: J Biol Chem. 2000 Apr 28;275(17):12676-83. Superinduction of CYP1A1 gene expression. Regulation of 2,3,7, 8-tetrachlorodibenzo-p-dioxin-induced degradation of Ah receptor by cycloheximide. Ma Q, Renzelli AJ, Baldwin KT, Antonini JM. Molecular Toxicology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, NIOSH, National Institutes of Health, Centers for Disease Control and Prevention, Morgantown, West Virginia 26505, USA. Cycloheximide superinduces the transcription of CYP1A1 in the presence of an agonist for the Ah receptor (AhR). To investigate the molecular target for "superinduction," we analyzed the agonist-induced degradation of AhR. Whereas 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD), a potent agonist of AhR, induces a rapid reduction of the AhR protein, cycloheximide blocks the down-regulation of steady state AhR. Analyses of the turnover of AhR reveal that cycloheximide blocks the shortening of the half-life of AhR by TCDD. Blocking of the TCDD-induced AhR degradation requires inhibition of protein synthesis, because (a) cycloheximide inhibits protein synthesis at the concentration at which it causes superinduction and inhibition of AhR degradation; and (b) puromycin, an inhibitor of protein synthesis by mimicking aminoacyl-tRNA, also blocks the TCDD-induced AhR degradation. The blocking of the TCDD-induced AhR degradation correlates with the superinduction of CYP1A1 gene expression in a time- and dose-dependent manner. Furthermore, cycloheximide is shown to increase the accumulation of the TCDD-activated AhR and the functional AhR x Arnt complex in nucleus. Collectively, our results reveal a mechanism of superinduction by cycloheximide by enhancing the stability of agonist-activated AhR. The finding that inhibition of protein synthesis blocks the TCDD-induced AhR turnover implicates a cycloheximide-sensitive, labile factor (designated as AhR degradation promoting factor, or ADPF) in controlling the removal of agonist-activated AhR in nucleus. PMID: 10777561 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 194: J Biochem (Tokyo). 2000 Mar;127(3):503-9. Nucleocytoplasmic shuttling of the aryl hydrocarbon receptor. Ikuta T, Tachibana T, Watanabe J, Yoshida M, Yoneda Y, Kawajiri K. Saitama Cancer Center Research Institute, Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan. togo@cancer-c.pref.saitama.jp The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that acts in concert with the AhR nuclear translocator (ARNT), and alters gene expression in response to environmental contaminants such as 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD). We have previously shown that AhR contains both a nuclear localization signal (NLS), AhR(13-39), and a nuclear export signal (NES), AhR(55-75), in its NH(2)-terminal region. In this study, we obtained direct evidence for the nucleocytoplasmic shuttling of AhR and show the biological significance of the shuttling in terms of the transcriptional activation of its target gene, CYP1A1. When AhR(13-75) fused with glutathione S-transferase (GST)-green fluorescent protein (GFP) was microinjected into the nucleus of a polykaryotic of BHK21 cell, the GST-AhR(13-75)-GFP migrated from one nucleus to the other. This event, nucleocytoplasmic shuttling, was completely inhibited in the presence of leptomycin B (LMB). The interaction between chromosome region maintenance 1 (CRM1) and endogenous AhR was shown by immunoprecipitation with antibodies to AhR followed by immunoblot analysis with antibodies to CRM1. The inhibition of the nuclear export of AhR by LMB repressed the transcriptional activation of the CYP1A1 gene. The findings suggest that nuclear-cytoplasmic shuttling of AhR is essential for the inducible expression of the CYP1A1 protein. PMID: 10731723 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 195: Chem Biol Interact. 2000 Feb 1;124(3):205-16. Estrous cycle-dependent changes in the expression of aromatic hydrocarbon receptor (AHR) and AHR-nuclear translocator (ARNT) mRNAs in the rat ovary and liver. Chaffin CL, Trewin AL, Hutz RJ. Department of Biological Sciences, University of Wisconsin-Milwaukee, 53211, USA. The aromatic hydrocarbon receptor (AHR) and AHR nuclear translocator protein (ARNT) mediate the toxic effects of a wide variety of halogenated and polycyclic aromatic hydrocarbons. While it can be assumed that AHR has an endogenous function, its role in reproduction is currently undefined. The present study seeks to examine the regulation of AHR and ARNT mRNAs in liver and ovarian tissues across the rat estrous cycle. Message for hepatic AHR was increased significantly on the morning of proestrus, and decreased dramatically by the evening of proestrus; while hepatic ARNT mRNA was significantly decreased between diestrus and the morning of proestrus, and between the evening of proestrus and the morning of estrus. Ovarian AHR mRNA was unchanged from diestrus to proestrus, and was decreased on the evening of proestrus. Changes in the expression of ARNT mRNA mirrored changes in the liver. To assess interaction between the AHR- and estrogen-receptor (ER)-signaling pathways and to test the hypothesis that estrogen regulates AHR mRNA, 25-day-old female rats were injected with either 17beta-estradiol, the ER antagonist ICI 182 780, or with vehicle, and hepatic AHR mRNA was measured. Treatment with estrogen or the estrogen antagonist did not alter the abundance of AHR mRNA in the liver. These data suggest that while estrogen may not be the key regulator of AHR mRNA expression, a factor associated with the rat reproductive cycle may be important in regulating the expression of both the AHR and ARNT genes in the ovary and liver. PMID: 10728779 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 196: Endocr J. 1999 Dec;46(6):765-72. Expression of Ah receptor and dioxin-related genes in human uterine endometrium in women with or without endometriosis. Igarashi T, Osuga U, Tsutsumi O, Momoeda M, Ando K, Matsumi H, Takai Y, Okagaki R, Hiroi H, Fujiwara O, Yano T, Taketani Y. Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Japan. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been suggested as a possible etiologic factor for endometriosis, a condition in which endometrium-like tissues are present outside the uterus. The prevailing view pertaining to the origin of endometriotic cells is that they are from eutopic endometrial cells which regurgitate through fallopian tubes. In order to get insight into the possible involvement of TCDD in the pathogenesis of endometriosis, we suspected that TCDD may act differently on the endometrium with or without endometriosis. To address this, we examined the presence of messenger RNAs of arylhydrocarbon receptor (AhR), AhR nuclear translocator (Arnt) and two dioxin-responsive genes, cytochrome P-450 1B1 (CYP1B1) and downstream of tyrosine kinases (p62(dok)), in the endometrium of women with or without endometriosis using semi-quantitative reverse transcription-polymerase chain reaction. All the genes were expressed throughout the menstrual cycle. The expression level of p62(dok) was higher in the proliferative phase than in the secretory phase. In contrast, the expression levels of AhR, Arnt and CYP1B1 seemed to be constant during the cycle. In terms of the comparison between non-endometriosis and endometriosis group, the mRNA levels of AhR, Arnt, CYP1B1 and p62(dok) were essentially similar. Interestingly, AhR mRNA level was significantly lower in smokers than in non-smokers. Based on the regression analysis, significant linear and positive correlations were observed between AhR and Arnt mRNA levels, and between Arnt and p62(dok) mRNA levels. In summary, expression of AhR and dioxin-related genes in the endometrium did not differ in women with or without endometriosis. PMID: 10724351 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 197: J Biol Chem. 2000 Mar 24;275(12):8432-8. 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced degradation of aryl hydrocarbon receptor (AhR) by the ubiquitin-proteasome pathway. Role of the transcription activaton and DNA binding of AhR. Ma Q, Baldwin KT. Molecular Toxicology Laboratory, Toxicology and Molecular Biology Branch, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia 26505, USA. qam1@cdc.gov Activation of the aryl hydrocarbon receptor (AhR) by 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD), a potent agonist of AhR, induces a marked reduction in steady state AhR. To analyze the mechanism of regulation of ligand-activated AhR, we examined the biochemical pathway and function of the down-regulation of the receptor by TCDD. Pulse-chase experiments reveal that TCDD shortens the half-life (t1/2) of AhR from 28 to 3 h in mouse hepatoma cells. Inhibitors of the 26 S proteasome, lactacystin and MG132, block the TCDD-induced turnover of AhR. The TCDD-induced degradation of AhR involves ubiquitination of the AhR protein, because (a) TCDD induces formation of high molecular weight, ubiquitinated AhR and (b) degradation of AhR is inhibited in ts20 cells, which bear a temperature-sensitive mutation in the ubiquitin-activating enzyme E1, at a nonpermissive temperature. Inhibition of proteasomal degradation of AhR increases the amount of the nuclear AhR.Arnt complex and "superinduces" the expression of endogenous CYP1A1 gene by TCDD, indicating that the proteasomal degradation of AhR serves as a mechanism for controlling the activity of the activated receptor. We also show that deletion of the transcription activation domain of AhR abolishes the degradation, whereas a mutation in the DNA-binding region of AhR or Arnt reduces the degradation; these data implicate the transcription activation domain and DNA binding in AhR degradation. Our findings provide new insights into the regulation of TCDD-activated AhR through ubiquitin-mediated protein degradation. PMID: 10722677 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 198: Toxicol Sci. 1999 Dec;52(2):217-25. Differential gene expression in wild-type and arnt-defective mouse hepatoma (Hepa1c1c7) cells. Seidel SD, Denison MS. Department of Environmental Toxicology, University of California, Davis 95616, USA. The aryl hydrocarbon nuclear translocator (Arnt) is a basic helix-loop-helix (per/Arnt/Ahr/sim) PAS-containing protein that can heterodimerize with the aryl hydrocarbon receptor (AhR), the hypoxia-inducible factor-1 alpha, and other PAS-containing proteins to form transcriptionally active complexes. To identify the genes whose expression is modulated by Arnt, we used the technique of differential display to compare the expression of genes in wild-type and Arnt-defective (BPRc1) mouse hepatoma (Hepa1c1c7) cells. Here we report two gene products whose expression was reduced in BPRc1 cells (a WW domain-binding, protein-like factor and one unknown gene product) when compared to wild-type cells, and two that were elevated (Steel factor and a serpin-like protein). Comparison of the relative expression of these gene products between two independently-derived, Arnt-defective cell lines, as well as in BPRc1 cells in which Arnt expression was restored by a stably integrated Arnt-expression plasmid, revealed that each gene was expressed in an Arnt-independent manner. Our results clearly demonstrate that gene expression in the variant cell clones is distinctly different from that of the parental wild-type Hepa1c1c7 cells from which they were derived and involves genes in addition to, and unrelated to, that of Arnt. The identification of these differentially expressed gene products suggests that caution should be exercised when using these variant cell lines to confirm the role of the AhR/Arnt-signaling pathway in a given cellular response. PMID: 10630574 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 199: Arch Biochem Biophys. 2000 Jan 1;373(1):163-74. The aryl hydrocarbon receptor interacts with estrogen receptor alpha and orphan receptors COUP-TFI and ERRalpha1. Klinge CM, Kaur K, Swanson HI. Department of Biochemistry, University of Louisville School of Medicine, Louisville, Kentucky, 40292, USA. carolyn.klinge@louisville.edu The molecular mechanisms underlying the apparent "cross-talk" between estrogen receptor (ER)- and arylhydrocarbon receptor (AHR)-mediated activities are unknown. To determine how AHR ligand 2, 3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) may inhibit ER action and, conversely, to examine how 17-beta-estradiol (E(2)) affects AHR activity, we examined discrete activities of each receptor, i.e., protein-protein interactions, DNA binding, and transcriptional activation. We report that AHR interacts directly with ERalpha, COUP-TF, and ERRalpha1, in a ligand-specific manner in vitro. Unoccupied or beta-napthoflavone (beta-NF)-occupied AHR showed stronger interaction with ERalpha, COUP-TF, and ERRalpha1 than when AHR was occupied by the partial antagonist alpha-naphthoflavone (alpha-NF), indicating a role for ligand in AHR interaction with these proteins. We also report that AHR interacts with COUP-TF in transfected CV-1 cells. In contrast, the AHR nuclear translocator protein (ARNT) did not interact with COUP-TF, ERRalpha1, or ERalpha. We next examined the interaction of either ERalpha or COUP-TF with a consensus xenobiotic response element (XRE). Purified ERalpha did not bind the consensus XRE, but COUP-TFI bound the consensus XRE, suggesting a role for COUP-TF as a AHR/ARNT competitor for XRE binding. In transiently transfected MCF-7 human breast cancer cells, overexpression of COUP-TFI inhibited TCDD-activated reporter gene activity from the CYP1A1 promoter. TCDD inhibited estradiol (E(2))-activated reporter gene activity from a consensus ERE and from the EREs in the pS2 and Fos genes, and COUP-TFI did not block the antiestrogenic activity of TCDD. The specific interaction of COUP-TF with XREs and AHR together with the inhibition of TCDD-induced gene expression by COUP-TF suggests that COUP-TF may regulate AHR action both by direct DNA binding competition and through protein-protein interactions. Copyright 2000 Academic Press. PMID: 10620335 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 200: Mol Cell Endocrinol. 1999 Nov 25;157(1-2):105-19. The aryl hydrocarbon receptor (AHR)/AHR nuclear translocator (ARNT) heterodimer interacts with naturally occurring estrogen response elements. Klinge CM, Bowers JL, Kulakosky PC, Kamboj KK, Swanson HI. Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, KY 40292, USA. carolyn.klinge@louisville.edu To determine the molecular mechanisms underlying the "cross talk" between the activity of 2,3,7,8-tetra-chlorodibenzo-p-dioxin (TCDD), which binds to arylhydrocarbon receptor (AHR) and estradiol (E2)-liganded estrogen receptor (ER), we first examined the initial step of estrogen action, ligand binding to ER. None of the AHR ligands tested, i.e. TCDD, benzo[a]pyrene, 3,3',4,4',5-pentachlorobiphenyl, beta-naphthoflavone, or alpha-naphthoflavone, bound to ER alpha. We report the first examination of TCDD interaction with ER beta: TCDD did not displace E2 from ER beta. We then examined a second possible mechanism, i.e. direct inhibition of ER alpha binding to estrogen response elements (EREs) by the AHR/AHR nuclear translocator (ARNT) complex. The AHR/ARNT heterodimer did not bind either a full or half-site ERE. However, AHR/ARNT bound specifically to oligomers containing naturally occurring EREs derived from the human c-fos, pS2, and progesterone receptor (PR) gene promoters that include xenobiotic response element (XRE)-like sequences. In contrast, neither purified E2-liganded-ER from calf uterus or recombinant human ER alpha bound a consensus XRE. TCDD inhibited E2-activated reporter gene activity from a consensus ERE and from EREs in the pS2, PR, and Fos genes in transiently transfected MCF-7 human breast cancer cells. However, this inhibition was not reciprocal since E2 did not inhibit TCDD-stimulated luciferase activity from the CYP1A1 promoter in transiently transfected MCF-7 or human endometrial carcinoma HEC-1A cells. We propose that at least part of the mechanism by which the AHR/ARNT complex inhibits estrogen action is by competitively inhibiting ER alpha binding to imperfect ERE sites, adjacent to or overlapping XREs. PMID: 10619402 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 201: Mol Pharmacol. 2000 Jan;57(1):82-92. Role of heat shock protein 90 dissociation in mediating agonist-induced activation of the aryl hydrocarbon receptor. Heid SE, Pollenz RS, Swanson HI. Department of Pharmacology, University of Kentucky Medical Center, Lexington, Kentucky, USA. The aryl hydrocarbon receptor (AhR) is a cytosolic basic helix-loop-helix protein that associates with a chaperone complex that includes two molecules of heat shock protein 90 (HSP90). It has been hypothesized that after ligand binding, the AhR dissociates from its chaperone complex and translocates into the nucleus, where it heterodimerizes with its DNA binding partner, the AhR nuclear translocator (ARNT), and activates specific genes. However, it remains unclear whether nuclear translocation of the AhR occurs before or after dissociation of the HSP90/chaperone complex. Because sodium molybdate stabilizes the AhR-HSP90 interaction and inhibits the gene activation of a number of steroid receptors, we reasoned that molybdate would be a useful tool in delineating the role of HSP90 dissociation in AhR nuclear translocation. In this study, we demonstrate that molybdate inhibits AhR gene activation in both HepG2 and Hepa-1 cells in a concentration-dependent manner and protects the AhR against agonist-induced proteolysis. In addition, we demonstrate that AhR/ARNT dimerization, but not nuclear translocation of the AhR, is inhibited by molybdate. This indicates that 1) HSP90 dissociation is not required for nuclear translocation of the AhR, 2) HSP90 dissociation is essential for formation of the AhR/ARNT heterodimer, and 3) an additional undefined regulatory step is required for AhR/ARNT dimerization in the nucleus. PMID: 10617682 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 202: Endocrinology. 2000 Jan;141(1):450-3. The aryl hydrocarbon receptor, a basic helix-loop-helix transcription factor of the PAS gene family, is required for normal ovarian germ cell dynamics in the mouse. Robles R, Morita Y, Mann KK, Perez GI, Yang S, Matikainen T, Sherr DH, Tilly JL. Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital/Harvard Medical School, Boston 02114, USA. The aryl hydrocarbon receptor (AhR), so-designated based on the ability of the protein to bind with and be activated by polycyclic aromatic hydrocarbons (PAH) and related halogenated hydrocarbons, is part of an emerging family of ligand-activated transcriptional regulators that are distinct from the steroid-thyroid hormone receptor superfamily. Once bound by ligand, the AhR interacts with the AhR nuclear translocator (ARNT) protein to form the aryl hydrocarbon receptor complex (AHRC). Both subunits of the AHRC contain sequences corresponding to basic helix-loop-helix domains, a motif that is shared by a number of other dimeric transcription factors. Although the natural ligand(s) for the AhR remains to be elucidated, to date over fifteen genes, including enzymes, growth factors and other transcription factors, have been identified as potential targets for transcriptional regulation by the chemically-activated AHRC. In the ovary, PAH exposure is known to cause destruction of oocytes within immature follicles, implying that one function of the AhR is to mediate cell death signaling in the female germ line. To assess this possibility, we explored AhR expression patterns in the murine ovary, and then determined the impact of AhR-deficiency (gene knockout) on female germ cell dynamics. Immunohistochemical analysis of ovaries of wild-type female mice indicated that AhR protein was abundantly and exclusively expressed in oocytes and granulosa cells of follicles at all stages of development. Histomorphometric analysis of serial ovarian sections revealed a two-fold higher number of primordial follicles in Ahr-null versus wild-type females at day 4 postpartum. This phenotype likely results from a cell-intrinsic death defect in the developing germ line since AhR-deficiency attenuated the magnitude of oocyte apoptosis in fetal ovaries cultured without hormonal support for 72 h. We propose that the AhR, activated by an as yet unknown endogenous ligand(s), serves to regulate the size of the oocyte reserve endowed at birth by affecting germ cell death during female gametogenesis. PMID: 10614669 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 203: Toxicol Appl Pharmacol. 1999 Dec 1;161(2):123-39. Metabolism-based polycyclic aromatic acetylene inhibition of CYP1B1 in 10T1/2 cells potentiates aryl hydrocarbon receptor activity. Alexander DL, Zhang L, Foroozesh M, Alworth WL, Jefcoate CR. Environmental Toxicology Center, University of Wisconsin, Madison, Wisconsin, 53705, USA. We have used polycyclic aromatic hydrocarbon (PAH) alkyne metabolism-based inhibitors to test whether CYP1B1 metabolism is linked to aryl hydrocarbon receptor (AhR) activation in mouse embryo fibroblasts (MEF). 1-ethynylpyrene (1EP) selectively inactivated CYP1B1 dimethylbenzanthracene (DMBA) metabolism in C3H10T1/2 MEFs; whereas 1-(1-propynyl)pyrene (1PP) preferentially inhibited CYP1A1 activity in Hepa-1c1c7 mouse hepatoma cells (Hepa). In each cell type >90% inhibition of DMBA metabolism after 1 h treatment with each inhibitor (0.1 microM) was progressively reversed and then increased to levels seen with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induction (fourfold stimulation). It was found that 0.1 microM 1EP and 1PP maximally induce CYP1B1 and CYP1A1 mRNA levels in10T1/2 and Hepa cells, respectively, after 6 h. 1-Ethylpyrene (EtP), which lacks the activatable acetylene moiety, was far less effective as an inhibitor and as an inducer. AhR activation is essential for 1EP induction as evidenced by the use of AhR antagonists and AhR-deficient MEFs and absence of induction following inhibition of DMBA metabolism with carbon monoxide (CO). Inhibition of CYP1B1 was linked to enhanced AhR activation even at early stages prior to significant ligand depletion. 1EP and EtP were similarly effective in stimulating AhR nuclear translocation, though 5-10 times slower compared with TCDD, and produced no significant down-regulation of the AhR. TCDD activated AhR/Arnt complex formation with an oligonucleotide xenobiotic response element far more extensively than 1EP or EtP, even at concentrations of 1EP that increased CYP1B1 mRNA to similar levels. CO did not influence these responses to EtP, event hough CO treatment potentiated EtP induction of CYP1B1 mRNA. These differences suggest a fundamental difference between PAH/AhR and TCDD/AhR complexes where CYP1B1 metabolic activity regulates the potency, rather than the formation of the AhR/Arnt complex. Copyright 1999 Academic Press. PMID: 10581206 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 204: Mol Pharmacol. 1999 Dec;56(6):1127-37. Analysis of aryl hydrocarbon receptor-mediated signaling during physiological hypoxia reveals lack of competition for the aryl hydrocarbon nuclear translocator transcription factor. Pollenz RS, Davarinos NA, Shearer TP. Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29403, USA. pollenzr@musc.edu The aryl hydrocarbon nuclear translocator (ARNT) protein functions as a transcription factor after dimerization with other basic helix-loop-helix proteins. Thus, dimerization of ARNT within one pathway may limit the availability of this protein to others. To investigate this issue, aryl hydrocarbon receptor (AHR)-mediated signaling was investigated in mouse (Hepa-1), rat (H4IIE), and human (HepG2) hepatoma cell lines undergoing physiologically induced hypoxia (<1% O(2)). Basal levels of ARNT protein were not affected by hypoxia in any cell line, and ARNT remained exclusively nuclear. Furthermore, quantitative Western blotting revealed that the concentration of ARNT sequestered during hypoxia represented a small fraction of the total ARNT protein pool (12 and 15% in Hepa-1 and H4 cells, respectively). When the AHR-mediated signaling pathway was activated during hypoxia by 2,3,7,8-tetrachlorodibenzo-p-dioxin, the induction of P4501A1 protein was reduced by 55% without changes in the level of mRNA in Hepa-1 cells, whereas the levels of induction of both P4501A1 protein and CYP1A1 mRNA were reduced by >80% in the H4 cell line. Importantly, gel mobility shift analysis and Western blotting showed that the same level of AHR/ARNT complexes could be detected in cells treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin during hypoxia and normoxia. These data suggest that the effects of hypoxia on AHR-mediated gene regulation occur distal to the formation of AHR/ARNT complexes and imply that functional interference between hypoxia and AHR-mediated signaling does not occur through competition for ARNT protein. PMID: 10570039 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 205: J Biol Chem. 1999 Nov 19;274(47):33814-24. Identification and functional characterization of two highly divergent aryl hydrocarbon receptors (AHR1 and AHR2) in the teleost Fundulus heteroclitus. Evidence for a novel subfamily of ligand-binding basic helix loop helix-Per-ARNT-Sim (bHLH-PAS) factors. Karchner SI, Powell WH, Hahn ME. Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor through which 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds cause altered gene expression and toxicity. The AHR belongs to an emerging multigene family of transcription factors possessing basic helix loop helix (bHLH) and Per-ARNT-Sim (PAS) domains. Most bHLH-PAS proteins occur as duplicates or "paralog groups" in mammals, but only a single mammalian AHR has been identified. Here we report the cDNA cloning of two distinct AHRs, designated FhAHR1 and FhAHR2, from a single vertebrate species, the teleost Fundulus heteroclitus (Atlantic killifish). Both Fundulus AHR proteins possess bHLH and PAS domains that are closely related to those of the mammalian AHR. FhAHR1 and FhAHR2 are highly divergent (40% overall amino acid identity; 61% identity in the N-terminal half), suggesting that they arose from a gene duplication predating the divergence of mammals and fish. Photoaffinity labeling with 2-azido-3-[(125)I]iodo-7, 8-dibromodibenzo-p-dioxin and velocity sedimentation analysis using 2,3,7,8-[1,6-(3)H]TCDD showed that both FhAHR1 and FhAHR2 exhibit specific, high-affinity binding of dioxins. Both AHRs also showed specific, TCDD- and ARNT-dependent interactions with a mammalian xenobiotic response element. The two Fundulus AHR genes displayed different tissue-specific patterns of expression; FhAHR1 transcripts were primarily expressed in brain, heart, ovary, and testis, while FhAHR2 transcripts were equally abundant in many tissues. Phylogenetic analysis demonstrated that Fundulus AHR1 is an ortholog of mammalian AHRs, while AHR2 forms in Fundulus and other fish are paralogous to Fundulus AHR1 and the mammalian AHRs and thus represent a novel vertebrate subfamily of ligand-binding AHRs. PMID: 10559277 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 206: Arch Biochem Biophys. 1999 Nov 15;371(2):246-59. Lack of an absolute requirement for the native aryl hydrocarbon receptor (AhR) and AhR nuclear translocator transactivation domains in protein kinase C-mediated modulation of the AhR pathway. Long WP, Perdew GH. Center for Molecular Toxicology, Pennsylvania State University, University Park, Pennsylvania 16802, USA. Protein kinase C (PKC)-mediated modulation of the aryl hydrocarbon receptor (AhR) pathway was examined in CHOK1-derived L10.I cells stably transfected with the pGUDLUC6.1 reporter; pGUDLUC6.1 is solely controlled by four dioxin-responsive enhancer elements. Co treatment of L10.I cells with 10 nM 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) and 81 nM phorbol 12-myristate 13-acetate (PMA), an activator of sn-1,2-diacylglyerol binding PKCs, enhanced transactivation of the reporter construct several-fold relative to cells treated with a saturating 10 nM TCDD dose alone; this effect was dubbed the "PMA effect." A domain swapping and deletional analysis of the native AhR and AhR nuclear translocator (ARNT) protein transactivation domains (TADs) was performed to determine if these domains are absolutely required for the AhR x ARNT dimer-mediated PMA effect in the L10.I model system; controls demonstrate the suitability of the L10.I model for these analyses and that endogenous AhR and ARNT levels are extremely low in this model. Transient coexpression of the AhR and ARNT-474-FLAG, an ARNT protein lacking the native ARNT TAD, in L10.I cells reveals the native ARNT TAD is not absolutely required for the AhR x ARNT-474-FLAG dimer to mediate the PMA effect. Transient coexpression of AhRDeltaCVP, a chimeric AhR protein in which the native AhR TAD has been replaced with the VP16 (herpes simplex virus protein 16) TAD (which control experiments demonstrate is unaffected by PMA), and ARNT in L10.I cells indicates that the native AhR TAD is not absolutely required for this AhRDeltaCVP x ARNT dimer to mediate the PMA effect. These observations strongly suggest that PKC-mediated modulation of the AhR pathway is not absolutely dependent on coactivators recruited to the AhR. ARNT dimer by the native TADs of the AhR and its heterodimerization partner ARNT. Copyright 1999 Academic Press. PMID: 10545212 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 207: FEBS Lett. 1999 Oct 29;460(2):251-6. Hypoxia-induced activation of HIF-1: role of HIF-1alpha-Hsp90 interaction. Minet E, Mottet D, Michel G, Roland I, Raes M, Remacle J, Michiels C. Laboratoire de Biochimie et Biologie Cellulaire, Facultes Universitaires de la Paix, 61 rue de Bruxelles, 5000, Namur, Belgium. emmanuel.minet@fundp.ac.be The protein chaperone heat shock protein 90 (Hsp90) is a major regulator of different transcription factors such as MyoD, a basic helix loop helix (bHLH) protein, and the bHLH-Per-aryl hydrocarbon nuclear translocator (ARNT)-Sim (PAS) factors Sim and aryl hydrocarbon receptor (Ahr). The transcription factor hypoxia-inducible factor-1alpha (HIF-1alpha), involved in the response to hypoxia, also belongs to the bHLH-PAS family. This work was aimed to investigate the putative role of Hsp90 in HIF-1 activation by hypoxia. Using a EGFP-HIF-1alpha fusion protein, co-immunoprecipitation experiments evidenced that the chimeric protein expressed in COS-7 cells interacts with Hsp90 in normoxia but not in hypoxia. We also demonstrated that Hsp90 interacts with the bHLH-PAS domain of HIF-1alpha. Moreover, Hsp90 is not co-translocated with HIF-1alpha into the nucleus. At last, we showed that Hsp90 activity is essential for HIF-1 activation in hypoxia since it is inhibited in the presence of geldanamycin. These results indicate that Hsp90 is a major regulator in HIF-1alpha activation. PMID: 10544245 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 208: Toxicol Appl Pharmacol. 1999 Nov 1;160(3):231-7. Expression of the arylhydrocarbon receptor and the arylhydrocarbon receptor nuclear translocator during early gestation in the rabbit uterus. Tscheudschilsuren G, Hombach-Klonisch S, Kuchenhoff A, Fischer B, Klonisch T. Department of Anatomy, Martin Luther University Faculty of Medicine, Halle (Saale), D-06097, Germany. The arylhydrocarbon receptor (AhR) and the arylhydrocarbon receptor nuclear translocator (ARNT) are members of the PAS gene family mediating toxic effects of xenobiotics such as dioxin and polychlorinated biphenyls. We have analyzed the expression and cellular distribution of rabbit AhR and ARNT mRNA and protein level in the nonpregnant uterus and the pregnant and pseudopregnant uterus at Days 6 to 12 of gestation. In the preimplantation uterus at Day 6 of gestation and in the interimplantation and pseudopregnant uterus at Days 7, 8, 9, and 12 of gestation, low levels of AhR transcripts were detected in the glandular uterine epithelium. Upon attachment of the blastocyst at Day 7 of gestation, a strong expression of AhR and ARNT mRNA was observed in the luminal and glandular epithelium of the antimesometrial uterine compartment. In contrast, AhR and ARNT expression was low in the luminal epithelium of the paraplacental and the mesometrial placental fold. AhR mRNA was also detected in the trophoblast cells. During early placentation at Day 9 of gestation, expression of AhR and ARNT was first observed in the perivascular decidualized stromal cells and, at Day 12, extended to the decidualized stromal cells of the placental bed. Within the placenta, the syncytiotrophoblast expressed only low levels of AhR and ARNT mRNA and no protein. The specific expression patterns of AhR and ARNT during early gestation suggest functional roles for both transcription factors during feto-maternal interactions in the rabbit. Copyright 1999 Academic Press. PMID: 10544057 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 209: Life Sci. 1999;65(13):1339-49. Differential response to benzo[A]pyrene in human lung adenocarcinoma cell lines: the absence of aryl hydrocarbon receptor activation. Chang KW, Lee H, Wang HJ, Chen SY, Lin P. Institute of Toxicology, Chung-Shan Medical and Dental College, Taichung, Taiwan, ROC. Benzo[a]pyrene (B[a]P) has been shown to produce DNA adducts and to initiate pulmonary carcinogenesis in animals. We observed differential susceptibility to B[a]P in two human lung adenocarcinoma cell lines, A427 and CL3. DNA adducts were induced by B[a]P treatment in CL3 cells, however, A427 cells were much less responsive to B[a]P treatment. Cytochrome P450 1A1 (CYP1A1) is involved in bioactivation of B[a]P in nonhepatic tissues. Cotreatment with alpha-naphthoflavone, a CYP1A1 inhibitor, abolished DNA adduct formation by B[a]P in CL3 cells. Nevertheless, CYP1A1 inducer beta-naphthoflavone, enhanced DNA adduct formation by B[a]P in both A427 and CL3 cells. Both enzyme activity and mRNA levels of CYP1A1 were highly induced by 1 or 10 microM B[a]P treatment for 24 hr in CL3 cells but not in A427 cells. Protein levels of AhR and aryl hydrocarbon receptor nuclear translocator (Arnt) were similar in A427 and CL3 cells before B[a]P treatment. However, B[a]P induced a retarded band with the [32P]-dioxin responsive element in CL3 cells, but not in A427 cells. This study demonstrated that variation in AhR-mediated CYP1A1 induction contributes to differential susceptibility to B[a]P-DNA adduct formation in human lung cells. Since AhR and/or Arnt function is impaired in A427 cells, this cell line offers a model for investigating the repression mechanisms of CYP1A1 induction by B[a]P in lung cells. PMID: 10503953 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 210: J Hum Genet. 1999;44(5):293-9. Transitional change in interaction between HIF-1 and HNF-4 in response to hypoxia. Zhang W, Tsuchiya T, Yasukochi Y. Department of Molecular Genetics, Tokyo Medical and Dental University, Japan. yyasmgen@mri.tmd.ac.jp The roles of the erythropoietin (Epo) 3' enhancer in the activation of gene expression in response to hypoxia were investigated. The enhancer contains hypoxia-inducible enhancer binding site 1 (HIF-1 element) and two direct repeats of hexanucleotide consensus nuclear receptor half site (HNF-4 element). HIF-1, which is a heterodimeric complex of HIF-1 alpha and aryl hydrocarbon receptor nuclear translecator (ARNT), binds to HIF-1 element. HNF-4 binds to HNF-4 element as a homodimeric complex. Studies on mutant reporter plasmids demonstrated that both HIF-1 alpha and HNF-4 elements were necessary for augmentation of the enhancer activity, since mutation of either the HIF-1 or the HNF-4 element caused loss of inducibility under hypoxic conditions. Mammalian two-hybrid experiments in vivo revealed that transitional change took place from the interaction of HNF-4 with ARNT to that with HIF-1 alpha in response to hypoxia. Such interactive domains were identified in amino acids 369-465 containing the C-terminal of HNF-4 and amino acids 1-458 containing basic helix-loop-helix (bHLH) and Per-ARNT-AHR-Sim (PAS) domains of ARNT in normoxia. Also, an extended sequence containing ligand and dimerization domains, and the C-terminal of HNF-4 (amino acids 135-465), and the PAS domain (amino acids 106-526) of HIF-1 alpha were used for the interaction between the two transcription factors in hypoxia. From these data, the functional significance of the transitional change in the augmentation of gene expression by the Epo enhancer in hypoxia is discussed. PMID: 10496070 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 211: IARC Sci Publ. 1999;(148):149-57. Chapter 14. Ah receptor gene polymorphisms and human cancer susceptibility. Garte S, Sogawa K. Environmental and Occupational Health Sciences Institute, UMDNJ, Piscataway, NJ, USA. The Ah receptor (Ahr) gene occupies a central role in the metabolic pathways involved in the detoxification of important environmental carcinogens. The structures of the rodent and human genes have been elucidated, and the molecular details of the receptor function, including its interaction with other proteins such as Arnt and hsp90, have been thoroughly investigated. The Ahr gene is polymorphic in mice and in humans. In mice, good correlations have been found between structural polymorphisms in the gene and functional variants in various genetic strains. In humans, work on polymorphisms and their possible role in gene function as well as cancer susceptibility is just beginning. PMID: 10493256 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 212: Biochemistry. 1999 Aug 31;38(35):11490-500. Regulation of constitutive gene expression through interactions of Sp1 protein with the nuclear aryl hydrocarbon receptor complex. Wang F, Wang W, Safe S. Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466, USA. The region of residues -145 to -119 (CD/L) of the cathepsin D gene promoter contains a GC-rich motif that binds Sp1 protein and an adjacent pentanucleotide (CACGC) that corresponds to the core sequence of a dioxin responsive element (DRE) and binds the aryl hydrocarbon receptor (AhR)-AhR nuclear translocator (Arnt) complex. This Sp1(N)(4)DRE(core) motif has been identified in promoters of several genes in which Sp1 plays an important role in basal gene expression. In transient transfection assays with MCF-7 human breast cancer cells using wild-type pCD/L and constructs mutated in the core DRE (pCD/L(m1)) and Sp1 (pCD/L(m2)) sites, it was shown that both motifs were required for maximal basal activity. The requirements for AhR-Arnt interactions with Sp1 protein for maximal activity of pCD/L were confirmed in wild-type MCF-7 and Hepa 1c1c7 cells and Arnt-deficient Hepa 1c1c7 cells using antisense Arnt and Arnt expression plasmids. The functional interactions of Sp1 with AhR-Arnt were paralleled by physical interactions showing that AhR-Arnt and Sp1 proteins were co-immunoprecipitated and AhR-Arnt enhanced Sp1-[(32)P]CD/L binding in electrophoretic mobility shift assays. The physical and functional interactions of Sp1 with AhR-Arnt proteins bound to the Sp1(N)(4)DRE(core) motif were also dependent on the proximity of these sites, and both the activity and the extent of Sp1-DNA binding decreased as the number of intervening nucleotides increased from 4 to 20. These studies show that regulation of basal expression of some genes by Sp1 may also require interactions with AhR-Arnt. PMID: 10471301 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 213: Toxicol Appl Pharmacol. 1999 Aug 15;159(1):41-51. Transactivation activity of human, zebrafish, and rainbow trout aryl hydrocarbon receptors expressed in COS-7 cells: greater insight into species differences in toxic potency of polychlorinated dibenzo-p-dioxin, dibenzofuran, and biphenyl congeners. Abnet CC, Tanguay RL, Heideman W, Peterson RE. School of Pharmacy and Environmental Toxicology Center, University of Wisconsin, Madison, Wisconsin, 53706, USA. Transactivation assays were used to compare the potency and efficacy of polychlorinated dibenzo-p-dioxin (PCDD), dibenzofuran (PCDF), and biphenyl (PCB) congeners in activating aryl hydrocarbon receptors (AhRs) from rainbow trout (rtAhR2alpha and rtAhR2beta), zebrafish (zfAhR2), and human (huAhR), respectively. All AhRs were expressed with their species-specific AhR nuclear translocator (ARNT) in COS-7 cells. Transactivation activity was determined for two PCDD, two PCDF, and seven PCB congeners with each of the four AhR/ARNT pairs using prt1Aluc, a luciferase reporter driven by two dioxin-responsive enhancer elements (DREs) from the rainbow trout cyp1A gene. Maximal-fold induction, EC50, and relative potency values were calculated for congeners that exhibited dose-related activity in the assay. Of the four AhR/ARNT pairs tested with PCDD, PCDF, and non-ortho PCB congeners, three exhibited high activity (rainbow trout AhR2alpha, zebrafish AhR2, and human AhR), while rainbow trout AhR2beta had very weak or no activity. Comparisons between these AhRs showed that while mono-ortho PCBs were able to activate the human AhR, they were generally ineffective in activating rainbow trout and zebrafish AhR2s. This supports the hypothesis that structural differences between mammalian and fish AhRs may account for differences in relative potencies of the mono-ortho PCBs between mammals and fish. Another important finding was a significant difference in transactivation activity between the two rainbow trout AhR2 isoforms despite the fact that they are 95% identical at the amino acid level. For all PCDD, PCDF, and PCB agonists tested, rainbow trout AhR2alpha was significantly more active than AhR2beta. However, rainbow trout AhR2beta is active as a 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-activated transcription factor, with enhancer elements from the mouse cyp1A gene. This suggests that AhR2beta may have evolved to serve a different physiological function than AhR2alpha in salmonid fish species. Copyright 1999 Academic Press. PMID: 10448124 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 214: Development. 1999 Sep;126(17):3937-45. The spineless-aristapedia and tango bHLH-PAS proteins interact to control antennal and tarsal development in Drosophila. Emmons RB, Duncan D, Estes PA, Kiefel P, Mosher JT, Sonnenfeld M, Ward MP, Duncan I, Crews ST. Department of Biology, Washington University, St. Louis, MO, USA. The Drosophila spineless (ss) gene encodes a basic-helix-loop-helix-PAS transcription factor that is required for proper specification of distal antennal identity, establishment of the tarsal regions of the legs, and normal bristle growth. ss is the closest known homolog of the mammalian aryl hydrocarbon receptor (Ahr), also known as the dioxin receptor. Dioxin and other aryl hydrocarbons bind to the PAS domain of Ahr, causing Ahr to translocate to the nucleus, where it dimerizes with another bHLH-PAS protein, the aryl hydrocarbon receptor nuclear translocator (Arnt). Ahr:Arnt heterodimers then activate transcription of target genes that encode enzymes involved in metabolizing aryl hydrocarbons. In this report, we present evidence that Ss functions as a heterodimer with the Drosophila ortholog of Arnt, Tango (Tgo). We show that the ss and tgo genes have a close functional relationship: loss-of-function alleles of tgo were recovered as dominant enhancers of a ss mutation, and tgo-mutant somatic clones show antennal, leg, and bristle defects almost identical to those caused by ss(-) mutations. The results of yeast two-hybrid assays indicate that the Ss and Tgo proteins interact directly, presumably by forming heterodimers. Coexpression of Ss and Tgo in Drosophila SL2 cells causes transcriptional activation of reporters containing mammalian Ahr:Arnt response elements, indicating that Ss:Tgo heterodimers are very similar to Ahr:Arnt heterodimers in DNA-binding specificity and transcriptional activation ability. During embryogenesis, Tgo is localized to the nucleus at sites of ss expression. This localization is lost in a ss null mutant, suggesting that Tgo requires heterodimerization for translocation to the nucleus. Ectopic expression of ss causes coincident ectopic nuclear localization of Tgo, independent of cell type or developmental stage. This suggests that the interaction of Ss and Tgo does not require additional signals, unlike the ligand-dependent interaction of Ahr and Arnt. Despite the very different biological roles of Ahr and Arnt in insects and mammals, the molecular mechanisms by which these proteins function appear to be largely conserved. PMID: 10433921 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 215: J Biol Chem. 1999 Aug 6;274(32):22155-64. Differential recruitment of coactivator RIP140 by Ah and estrogen receptors. Absence of a role for LXXLL motifs. Kumar MB, Tarpey RW, Perdew GH. Center for Molecular Toxicology, Pennsylvania State University, University Park, Pennsylvania 16802, USA. The Ah receptor (AhR), a soluble cytosolic protein, mediates most of the toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related environmental contaminants. The mechanism of ligand-mediated AhR activation has been, in part, elucidated. The sequence of events following the binding of the AhR/AhR nuclear translocator protein (ARNT) heterodimer to dioxin response elements has yet to be completely understood. The role of coactivator, RIP140, in the modulation of transcriptional activity of AhR/ARNT heterodimer was examined. RIP140 enhanced TCDD-mediated, dioxin response element-driven reporter gene activity in three cell lines. Co-immunoprecipitation and co-localization assays revealed that RIP140 interacted with AhR, but not with ARNT, both in vitro and in cells. Mapping of the interaction sites revealed that RIP140 was recruited by the AhR transactivation domain via the Q-rich subdomain. The RIP140 domain that interacts with the AhR was mapped to a location between amino acid residues 154 and 350, which is distinct from those involved in estrogen receptor binding. The signature motif, LXXLL, which is responsible for binding of several coactivators to nuclear receptors, is not required for RIP140 binding to AhR. These results demonstrate that the AhR recruits coactivators that are capable of enhancing transcription and, thus, the AhR may compete with steroid receptors for a common coactivator pool. In addition, the data suggest that there are distinct motif(s) for the recruitment of RIP140 to AhR and possibly other non-steroid receptors/transcription factors. PMID: 10428779 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 216: Arch Biochem Biophys. 1999 Aug 1;368(1):31-9. Role of estradiol receptor-alpha in differential expression of 2,3,7, 8-tetrachlorodibenzo-p-dioxin-inducible genes in the RL95-2 and KLE human endometrial cancer cell lines. Jana NR, Sarkar S, Ishizuka M, Yonemoto J, Tohyama C, Sone H. Chemical Exposure and Health Effects Research Team, Regional Environment Division, Environmental Health Sciences Division, National Institute for Environmental Studies, 16-2 Onogawa, Ibaraki, Tsukuba, 305 0053, Japan. The present study was conducted to investigate the mechanism of the response of human uterine endometrial carcinoma cells, RL95-2 and KLE, to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). RL95-2 cells were highly responsive to TCDD in terms of cytochrome P4501A1 (CYP1A1), cytochrome P4501B1 (CYP1B1), and plasminogen activator inhibitor-2 (PAI-2), whereas KLE cells showed little stimulatory effects only at high doses. Neither showed any growth inhibition upon exposure to TCDD. KLE cells expressed higher levels of aryl hydrocarbon receptor (AhR) than RL95-2 and gel mobility shift assay also identified more liganded AhR-ARNT complex bound to xenobiotic response elements (XRE). TCDD had no downregulatory effects on the expression of either AhR or the estradiol receptor (ER). Though both cell types expressed ER-alpha almost equally, immunofluorescence demonstrated a defect in its nuclear translocation in KLE cells where ER-alpha was mainly cytoplasmic and estradiol-17beta (E(2)) was unable to translocate it to the nucleus. However, both cells were nonresponsive to E(2) in terms of transcriptional activation and transient expression of normal ER-alpha restored the E(2) responsiveness. Transient expression of ER-alpha in KLE cells also restored its responsiveness to TCDD on transcriptional activation. Collectively, these results indicate that ER-alpha acts as a positive modulator in regulation of the TCDD-inducible genes. Copyright 1999 Academic Press. PMID: 10415108 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 217: Biochemistry. 1999 Jul 13;38(28):8907-17. Characterization of the AhR-hsp90-XAP2 core complex and the role of the immunophilin-related protein XAP2 in AhR stabilization. Meyer BK, Perdew GH. Graduate Program in Biochemistry and Molecular Biology, Center for Molecular Toxicology, The Pennsylvania State University, University Park 16802, USA. The unliganded aryl hydrocarbon receptor (AhR) exists in the cytoplasm in a tetrameric 9S core complex, consisting of the AhR ligand-binding subunit, a dimer of hsp90, and the hepatitis B virus X-associated protein 2 (XAP2), an immunophilin-related protein sharing homologous regions with FKBP12 and FKBP52. Interactions between the recently identified XAP2 subunit and other members of the unliganded AhR complex and its precise role in the AhR signal transduction pathway are presently unknown. Mapping studies indicate that XAP2 requires the PAS, hsp90, and ligand binding domain(s) of the AhR for binding, and that both proteins directly interact in the absence of hsp90. XAP2 is also able to interact with hsp90 complexes in the absence of the AhR, and C-terminal sequences of XAP2 are required for this interaction. XAP2 binds to the C-terminal end of hsp90, which contains a tetratricopeptide repeat domain acceptor site, whereas the AhR binds to a domain in the middle of hsp90. XAP2 was not found to be associated with the AhR-Arnt heterocomplex either in vitro or in nuclear extracts isolated from Hepa 1 cells treated with TCDD. Transient expression of XAP2 in COS-1 cells resulted in enhanced cytosolic AhR levels, suggesting a role for XAP2 in regulating the rate of AhR turnover. PMID: 10413464 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 218: Biochem Biophys Res Commun. 1999 Jul 14;260(3):760-7. Characterization of three splice variants and genomic organization of the mouse BMAL1 gene. Yu W, Ikeda M, Abe H, Honma S, Ebisawa T, Yamauchi T, Honma K, Nomura M. Department of Physiology, Saitama Medical School, Moroyama, Saitama, 350-0495, Japan. The BMAL1 gene encodes a member of the basic helix-loop-helix/PER-ARNT-SIM (bHLH/PAS) family of transcription factors. It is a key regulator of circadian rhythms. Using sequence information from human BMAL1 (hBMAL1) cDNAs previously reported by our laboratory, we have isolated and characterized cDNAs encoding three splice variants of the mouse BMAL1 (mBMAL1) gene. Of the three splice variants, mBMAL1b extends for 1878 bp in the coding sequence, which is 91% identical to that of hBMAL1b; its deduced amino acid sequence is 626 residues long and is 98% identical to that of hBMAL1b, and sequence identities in the bHLH, PAS-A, and PAS-B regions are 98, 100, and 100%, respectively. mBMAL1b' arises from alternative usage of exon 2, which results in a 7-amino-acid insertion and alternative splice acceptor usage at the intron 9/exon 10 splice junction, which causes an alanine residue deletion. mBMAL1b' encodes 632 amino acids and contains the bHLH/PAS domains. mBMAL1g' is generated by alternative splice acceptor usage at the intron 6/exon 7 splice junction, which results in a 28-bp deletion adjacent to the 5' end of the PAS domain. Since the 28-bp deletion shifts the reading frame, mBMAL1g' is predicted to encode a product of only 222 amino acids that lacks the PAS domain. The tissue distributions of the three splice variants showed some variation. The variations in the tissue distributions and predicted amino acid sequences suggest that the three splice variants may have different functions. Direct sequencing of the genomic mBMAL1 clones indicated that the coding sequence of mBMAL1 spans 32 kb and includes 17 exons. An unusual exon/intron donor sequence was found in intron 14, which begins with GC at the 5' end. Comparison with the bHLH/PAS family genes revealed that the intron/exon splice pattern of mBMAL1 most closely matches that of the mAhr, which suggests that BMAL1 and Ahr belong to the same subclass and may be derived from a common primordial gene. Copyright 1999 Academic Press. PMID: 10403839 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 219: Arch Biochem Biophys. 1999 Jul 15;367(2):250-7. Interactions of nuclear receptor coactivator/corepressor proteins with the aryl hydrocarbon receptor complex. Nguyen TA, Hoivik D, Lee JE, Safe S. Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas, 77843-4466, USA. MCF-7 human breast cancer cells express the aryl hydrocarbon receptor (AhR), and treatment with AhR agonists such as 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) inhibits estrogen receptor (ER)-mediated responses. This study investigates physical and functional interactions of the AhR complex with a prototypical coactivator (estrogen receptor associating protein 140, ERAP 140) and corepressor (silencing mediator for retinoic acid and thyroid hormone receptor, SMRT) for ER and other members of the nuclear receptor superfamily. The AhR, AhR nuclear translocator (Arnt), and AhR/Arnt proteins were coimmunoprecipitated with 35S-ERAP 140 and 35S-SMRT and, in gel mobility shift assays, AhR/Arnt binding to 32P-dioxin response element (DRE) was enhanced by ERAP-140 and inhibited by SMRT; supershifted bands were not observed. In transactivation assays, coactivator and corepressor proteins enhanced or inhibited AhR-mediated gene expression; however, these responses varied with the amount of coactivator/corepressor expression. These results confirmed functional and physical interactions of AhR/Arnt with ERAP 140 and SMRT in breast cancer cells. Copyright 1999 Academic Press. PMID: 10395741 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 220: Mar Biotechnol (NY). 1999 Mar;1(2):155-166. Trout CYP1A3 Gene: Recognition of Fish DNA Motifs by Mouse Regulatory Proteins. Carvan MJ 3rd, Ponomareva LV, Solis WA, Matlib RS, Puga A, Nebert DW. Center for Environmental Genetics and Department of Environmental Health, University of Cincinnati Medical Center, P.O. Box 670056, Cincinnati, OH 45267-0056, U.S.A. : Transcriptional up-regulation of mammalian CYP1A1 genes by dioxin is known to require binding of dioxin to the Ah receptor (AHR), subsequent interaction of this ligand-receptor complex with the AHR nuclear translocator (ARNT), and binding of this heterodimer to aromatic hydrocarbon response elements (AHREs) located in the 5' flanking sequences. From the rainbow trout (Oncorhyncus mykiss), we have isolated and sequenced the CYP1A3 gene-spanning 4.0 kb and containing seven exons and six introns-and 1897 bp of the 5' flanking region. The transcription start site was determined by primer extension analysis. Five putative AHREs were found between -451 and -1820, with an overlap of AHRE3 and AHRE4 sharing 1 bp. The 5' flanking region of the trout CYP1A3 gene was fused to the firefly luciferase (luc) reporter gene and transiently transfected into mouse hepatoma Hepa-1c1c7 wild-type (wt) cell cultures and three benzo[a]pyrene-resistant mutant lines: c2, containing less than 10% levels of functional AHR; c4, defective in ARNT; and c37, deficient in CYP1A1 metabolism. We compared the trout CYP1A3 promoter-luc constructs with mouse and human CYP1A1 promoter-luc constructs. All of our trout CYP1A3 promoter data are consistent with dioxin-inducible luciferase activity being controlled by two or more AHREs via cooperativity with a GC-rich region (-1852)-as has previously been demonstrated for AHREs in mammalian CYP1A1 promoters. The dependence of trout CYP1A3 promoter activity on the AHR and on the ARNT, and the enhancement of CYP1A3 promoter activity in the absence of CYP1A1 metabolic capacity, are all similar to that with mammalian CYP1A promoters. These findings indicate that the DNA motifs in trout, and the mouse liver proteins that bind to these motifs, are evolutionarily conserved elements. PMID: 10373624 [PubMed - as supplied by publisher] --------------------------------------------------------------- 221: Toxicol Appl Pharmacol. 1999 Jun 1;157(2):125-33. Induction of arylhydrocarbon receptor expression in embryoblast cells of rabbit preimplantation blastocysts upon degeneration of Rauber's polar trophoblast. Tscheudschilsuren G, Kuchenhoff A, Klonisch T, Tetens F, Fischer B. Department of Anatomy and Cell Biology, Martin Luther University Faculty of Medicine, Halle (Saale), D-06097, Germany. The arylhydrocarbon receptor (AhR) is a ligand-activated transcription factor and mediates carcinogenic, teratogenic, and toxic effects of xenobiotics such as dioxin and coplanar polychlorinated biphenyls. The AhR nuclear translocator (ARNT) is involved in AhR signal transduction. We have analyzed the expression of AhR and ARNT mRNA and AhR protein in Day 3 pc (postcoitum) rabbit morulae and Days 4 and 6 pc blastocysts using RT-PCR, nested PCR, whole mount in situ hybridization, and whole mount immunohistochemistry with subsequent confocal laser scanning analysis. AhR and ARNT transcripts were detected in all stages investigated, indicating coexpression of both transcription factors. AhR protein was localized in the cytoplasm. It was detected in Day 3 pc morulae and in blastocysts. In Day 4 pc blastocysts, only trophoblast cells but not embryoblast cells were immunopositive. However, at Day 6 pc, the embryoblast cells also expressed AhR protein and this expression was correlated with the degeneration of Rauber's trophoblast layer. Copyright 1999 Academic Press. PMID: 10366545 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 222: Carcinogenesis. 1999 Jun;20(6):947-55. Expression of CYP1A1 and CYP1B1 depends on cell-specific factors in human breast cancer cell lines: role of estrogen receptor status. Angus WG, Larsen MC, Jefcoate CR. Department of Pharmacology and Environmental Toxicology Center, University of Wisconsin, 3770 Medical Sciences Center, 1300 University Avenue, Madison, WI 53706, USA. The impact of estrogen receptor (ER) was examined for expression and activity of cytochrome P4501B1 (CYP1B1) and cytochrome P4501A1 (CYP1A1) in two pairs of ER+/ER- human breast epithelial cell lines derived from single lineages, and representing earlier (T47D) or later (MDA-MB-231) stages of tumorigenesis. Acute loss of ER was evaluated using the anti-estrogen ICI 182,780 (ICI). In all lines, CYP1B1 was expressed constitutively and was induced by 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD), whereas CYP1A1 was expressed only following induction. Expression of each CYP (with or without TCDD) was greater in T47D cells than MDA cells. The ER impacted expression of these genes in opposite directions. The ER- phenotype was associated with less TCDD-induced CYP1A1 expression, but greater basal and induced CYP1B1 expression. A 48 h treatment of ER+ cells with ICI did not revert the P450 expression pattern to that of ER- cells. Based on activities of recombinant enzyme and expression levels, differences in 7,2-dimethylbenz [a]anthracene (DMBA) metabolism between the cell lines were consistent with differences in CYP1A1 and CYP1B1 expression. In T47D lines, basal microsomal DMBA metabolism was primarily due to CYP1B1, based on regioselective metabolite distribution and inhibition by anti-CYP1B1 antibodies (>80%). Metabolism in TCDD-induced microsomes was mostly due to CYP1A1 and was inhibited by anti-CYP1A1 antibody (>50%). TCDD-induced MDA+ cells demonstrated CYP1A1 activity, whereas TCDD-induced MDA- cells displayed CYP1B1 activity. Aryl hydrocarbon receptor (AhR) levels, but not AhR nuclear translocator protein (ARNT) levels were highly dependent on cell type; AhR was high and ER-independent in MDA, and low and ER-linked in T47D. AhR levels were insensitive to ICI. ER does not directly modulate the expression of CYP1A1, CYP1B1 or AhR. Indeed, factors that have replaced ER in growth regulation during clonal selection predominate in this regulation. Characteristics unique to each cell line, including ER status, determine CYP1A1 and CYP1B1 expression. PMID: 10357772 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 223: Mol Cells. 1999 Apr 30;9(2):172-8. Aromatic hydrocarbon nuclear translocator as a common component for the hypoxia- and dioxin-induced gene expression. Park H. Department of Life Science, University of Seoul, Korea. hspark@uoscc.uos.ac.kr Aromatic hydrocarbon nuclear translocator (Arnt) is an ubiquitously expressed protein that contains basic helix-loop-helix (bHLH) and Per-AhR-Arnt-Sim (PAS) motifs. Other bHLH-PAS proteins, hypoxia-inducible factor-1alpha (HIF-1alpha) and aromatic hydrocarbon receptor (AhR) mediate hypoxia- and dioxin-signal pathway, respectively. Arnt has been identified as a heterodimerization partner for AhR. AhR/Arnt heterodimer binds the regulatory region of xenobiotic-induced genes and activates their transcription. Here, in vivo results provide evidence that Arnt is involved in not only xenobiotic- but also hypoxia-induced transcriptional activation. In hypoxic condition, Arnt dimerizes with HIF-1alpha to make HIF-1alpha/Arnt heterodimer which is able to bind hypoxia-responsive DNA elements. The HIF-1alpha/Arnt heterodimer functions as a transactivator for hypoxia-inducible genes. Given that the expression of Arnt is limited, HIF-1alpha may compete with AhR for recruiting Arnt as a heteromeric partner. Consistent with this idea, the results indicate that the hypoxic activation of HIF-1alpha reduces dioxin-induced AhR's function on the dioxin-responsive reporter gene and the endogenous gene. PMID: 10340472 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 224: Annu Rev Pharmacol Toxicol. 1999;39:103-25. Induction of cytochrome P4501A1. Whitlock JP Jr. Department of Molecular Pharmacology, Stanford University School of Medicine, California 94305-5332, USA. jpwhit@leland.stanford.edu Cytochrome P4501A1 is a substrate-inducible microsomal enzyme that oxygenates polycyclic aromatic hydrocarbons, such as the carcinogen benzo(a)pyrene, as the initial step in their metabolic processing to water-soluble derivatives. Enzyme induction reflects increased transcription of the cognate CYP1A1 gene. The environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin is the most potent known cytochrome P4501A1 inducer. Two regulatory proteins, the aromatic (aryl) hydrocarbon receptor (AhR) and the AhR nuclear translocator (Arnt), mediate induction. AhR and Arnt are prototypical members of the basic helix-loop-helix/Per-Arnt-Sim class of transcription factors. Mechanistic analyses of cytochrome P4501A1 induction provide insights into ligand-dependent mammalian gene expression, basic helix-loop-helix/Per-Arnt-Sim protein function, and dioxin action; such studies also impact public health issues concerned with molecular epidemiology, carcinogenesis, and risk assessment. Publication Types: Review PMID: 10331078 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 225: J Biol Chem. 1999 Apr 23;274(17):12115-23. Cross-talk between the aryl hydrocarbon receptor and hypoxia inducible factor signaling pathways. Demonstration of competition and compensation. Chan WK, Yao G, Gu YZ, Bradfield CA. Department of Pharmaceutics and Medicinal Chemistry, School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California 95211, USA. The aryl hydrocarbon receptor (AHR) and the alpha-class hypoxia inducible factors (HIF1alpha, HIF2alpha, and HIF3alpha) are basic helix-loop-helix PAS (bHLH-PAS) proteins that heterodimerize with ARNT. In response to 2,3,7,8-tetrachlorodibenzo-p-dioxin, the AHR. ARNT complex binds to "dioxin responsive enhancers" (DREs) and activates genes involved in the metabolism of xenobiotics, e.g. cytochrome P4501A1 (Cyp1a1). The HIF1alpha.ARNT complex binds to "hypoxia responsive enhancers" and activates the transcription of genes that regulate adaptation to low oxygen, e.g. erythropoietin (Epo). We postulated that activation of one pathway would inhibit the other due to competition for ARNT or other limiting cellular factors. Using pathway specific reporters in transient transfection assays, we observed that DRE driven transcription was markedly inhibited by hypoxia and that hypoxia responsive enhancer driven transcription was inhibited by AHR agonists. When we attempted to support this cross-talk model using endogenous loci, we observed that activation of the hypoxia pathway inhibited Cyp1a1 up-regulation, but that activation of the AHR actually enhanced the induction of Epo by hypoxia. To explain this unexpected additivity, we examined the Epo gene and found that its promoter harbors DREs immediately upstream of its transcriptional start site. These experiments outline conditions where inhibitory and additive cross-talk occur between the hypoxia and dioxin signal transduction pathways and identify Epo as an AHR-regulated gene. PMID: 10207038 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 226: Biochem Biophys Res Commun. 1999 Apr 13;257(2):259-63. Expression of dioxin-responsive genes in human endometrial cells in culture. Yang JH. School of Medicine, Catholic University of Taegu-Hyosung, Taegu, Republic of Korea. yangjh@cuth.cataegu.ac.kr To investigate expression of dioxin-responsive genes in human endometrial cells with exposure to 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD), human endometrial stromal cells immortalized with temperature-sensitive SV40 T antigen were used for the experiments. Cells were treated with 0.1% DMSO or 0.1, 1, 10, or 100 nM TCDD for 24 h. Induction of interleukin-1beta (IL-1beta) and plasminogen activator inhibitor-2 (PAI-2) mRNAs was analyzed by reverse-transcription polymerase chain reaction. Expression of IL-1beta or PAI-2 mRNA in response to TCDD was increased in a dose-dependent fashion. The maximum increases of PAI-2 and IL-1beta mRNAs were observed at 100 and 10 nM TCDD, respectively. While cycloheximide treatment did not show a significant difference of PAI-2 mRNA levels between control and TCDD-treated cells, mRNA stability assay using actinomycin D showed that PAI-2 mRNA in TCDD-treated cells was about twofold more stable than the control cells. While expression of CYP1A1 mRNA was not detected and levels of ARNT mRNA were not altered by TCDD exposure, the amount of AhR mRNA was decreased dose dependently. The present study represents an initial attempt to determine the responses of dioxin-responsive genes in human endometrial cells following TCDD exposure. The results demonstrated that IL-1beta and PAI-2 genes are induced dose dependently in human endometrial cells with exposure to TCDD and expression of PAI-2 mRNA is controlled at the posttranscriptional level. Copyright 1999 Academic Press. PMID: 10198199 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 227: Mol Pharmacol. 1999 Apr;55(4):716-25. Flavone antagonists bind competitively with 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) to the aryl hydrocarbon receptor but inhibit nuclear uptake and transformation. Henry EC, Kende AS, Rucci G, Totleben MJ, Willey JJ, Dertinger SD, Pollenz RS, Jones JP, Gasiewicz TA. Department of Environmental Medicine, University of Rochester, Rochester, New York 14642, USA. henrye@envmd.rochester.edu Previous analyses suggested that potent aryl hydrocarbon receptor (AhR) antagonists were planar, with a lateral electron-rich center. To further define structural requirements and mechanism for antagonism, ten additional flavone derivatives were synthesized. Based on their ability to 1) compete with 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) for binding to the AhR; 2) inhibit TCDD-elicited binding of AhR to dioxin-responsive elements (DRE) in vitro; and 3) inhibit TCDD-induced transcription of DRE-dependent luciferase in stably transfected hepatoma cells, the most potent flavones contained a 3'-methoxy group and a 4'-substituent having one or more terminal atoms of high electron density (-N3, -NO2, or -NCS). Furthermore, these had low agonist activity as assessed by their inability to elicit AhR. DRE binding or to induce luciferase. Compounds containing bulkier 3' or 4'-substituents, or a 3'-OH group were less potent antagonists, and some were partial agonists. In rat liver cytosol, 3'-methoxy-4'-azido- and 3'-methoxy-4'-nitroflavones bound competitively (with TCDD) to the AhR, indicating that they bind to the TCDD-binding site. When hepatoma cells were exposed to these flavones, AhR complexes were primarily immunoprecipitable from the cytosol and contained 90 kDa heat shock protein. In contrast, AhR in TCDD-treated cells was primarily immunoprecipitated from nuclear extracts and was associated with Arnt but not 90 kDa heat shock protein. Immunocytofluorescence analysis in intact cells further indicated that the potent antagonist inhibited nuclear uptake of AhR and blocked TCDD-dependent down-regulation of AhR. Together, these data indicate that the most potent antagonists bind the AhR with high affinity but cannot initiate receptor transformation and nuclear localization. PMID: 10101030 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 228: Biochem Biophys Res Commun. 1999 Mar 24;256(3):462-8. Cross-talk between 2,3,7,8-tetrachlorodibenzo-p-dioxin and testosterone signal transduction pathways in LNCaP prostate cancer cells. Jana NR, Sarkar S, Ishizuka M, Yonemoto J, Tohyama C, Sone H. Regional Environment Division, Environmental Health Sciences Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki, 305 0053, Japan. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related compounds modulate various endocrine functions by enhancing ligand metabolism, altering hormone synthesis, down regulating receptor levels, and interfering with gene transcription. In the present study, we investigated the effects of TCDD on testosterone signal transduction pathways and vice versa in the androgen receptor (AR) positive LNCaP prostate cancer cell line. TCDD induced CYP1A1 mRNA and related enzyme activity in these cells, with dose and time-dependence. Both normal and testosterone-stimulated cell growth was inhibited by TCDD. The expression levels of the aryl hydrocarbon receptor (AhR), the aryl hydrocarbon receptor nuclear translocator (ARNT), and AR were not affected by exposure to TCDD at a dose of 10 nM for a 24 hr time period. Testosterone treatment dose-dependently inhibited the TCDD-induced CYP1A1 mRNA accumulation and related enzyme activity. Reciprocally, TCDD also dose-dependently inhibited testosterone-dependent transcriptional activity and testosterone-regulated prostate specific antigen (PSA) expression. Taken together, these results demonstrate antiandrogenic functions of TCDD and a specific ligand-induced bilateral transcriptional interference between TCDD and testosterone mediated signal transduction pathways. Copyright 1999 Academic Press. PMID: 10080920 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 229: Toxicol Appl Pharmacol. 1999 Mar 1;155(2):177-89. Ah receptor and ARNT protein and mRNA concentrations in rat prostate: effects of stage of development and 2,3,7, 8-tetrachlorodibenzo-p-dioxin treatment. Sommer RJ, Sojka KM, Pollenz RS, Cooke PS, Peterson RE. School of Pharmacy, University of Wisconsin, Madison, Wisconsin, 53706, USA. Effects of stage of development and 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) exposure on aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (ARNT) protein concentrations in reproductive organs of male rats were determined. AhR protein levels in developing rat ventral and dorsolateral prostate decreased with age, declining approximately 70% between Postnatal Days (PND) 1 and 21. ARNT protein levels also decreased with age in dorsolateral, but not ventral prostate. The developmental decreases in prostatic AhR and ARNT protein were associated with decreases in AhR and ARNT mRNA. AhR and ARNT protein concentrations in fetal urogenital sinus on Gestation Days (GD) 16, 18, and 20 were similar to levels in ventral prostate on PND 7. TCDD exposure of adult male rats (0.2, 1, 5, or 25 micrograms/kg po, 24 h) decreased AhR but not ARNT protein in ventral and dorsolateral prostate, vas deferens, and epididymis. In utero and lactational TCDD exposure (1.0 micrograms/kg dam po, GD 15) did not alter ARNT levels but reduced prostatic AhR protein levels on PND 7 and delayed the developmental decrease in AhR protein in ventral and dorsolateral prostate. Finally, pretreatment of rat pups for 24 h with TCDD (5 micrograms/kg ip) down-regulated prostatic AhR protein on PND 7, but not on PND 1. Thus, prostatic AhR and ARNT protein and mRNA levels are regulated with age, whereas only AhR protein concentration is altered by TCDD exposure. Because in utero and lactational TCDD exposure only decreased prostatic AhR on PND 7, it is unlikely that down-regulation of AhR is the mechanism by which perinatal TCDD exposure impairs prostate development. Copyright 1999 Academic Press. PMID: 10053172 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 230: Mol Pharmacol. 1999 Mar;55(3):594-604. Regulation of cytochrome P-450 (CYP) 1B1 in mouse Hepa-1 variant cell lines: A possible role for aryl hydrocarbon receptor nuclear translocator (ARNT) as a suppressor of CYP1B1 gene expression. Eltom SE, Zhang L, Jefcoate CR. Center for Environmental Toxicology and Department of Pharmacology, University of Wisconsin Medical School, Madison 53706, USA. seeltom@facstaff.wisc.edu Cytochrome P-450 (CYP) 1B1 expression in mouse hepatoma (Hepa-1) wild-type (WT) cells was compared with responses in Hepa-1 variants LA1 and LA2, which, respectively, exhibit low aryl hydrocarbon receptor (AhR) level and defective AhR nuclear translocator (ARNT) protein. 10T1/2 mouse embryo fibroblasts express predominantly CYP1B1 and at a 100 times higher level than in Hepa-1 cells, whereas they express about 300-fold lower CYP1A1 than Hepa-1 cells. The expression of CYP1B1 in WT and LA1 variant, although at a much lower level, follows that of CYP1A1, reflecting their common regulation through the AhR. The LA2 (ARNT-defective) cells showed a major difference between CYP1B1 and CYP1A1 expression. Although CYP1A1 mRNA levels in LA2 were extremely low and unresponsive to 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD), basal CYP1B1 mRNA and protein were expressed at levels similar to those seen in TCDD-induced WT. The elevated basal CYP1B1 mRNA in LA2 cells decreased by 50% after transient transfection of ARNT cDNA, in parallel with substantial restoration of CYP1A1 induction. This implicates ARNT as a suppressor of CYP1B1 basal expression in Hepa cells. In transient CYP1B1-luciferase constructs in LA2 cells, ARNT shows stimulatory effects in the enhancer region but an inhibitory effect on the proximal promoter. Two CYP1B1 enhancer elements [xenobiotic-responsive element (XRE) 1/2 and XRE4] formed TCDD-unresponsive complexes of similar mobility to TCDD-stimulated AhR-ARNT complex with XRE5. However, because these two complexes were formed to the same extent in LA2 as in WT cells, they cannot be due to ARNT or contribute to ARNT-regulated suppression. PMID: 10051545 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 231: Toxicol Sci. 1999 Jan;47(1):76-85. RT-PCR quantification of AHR, ARNT, GR, and CYP1A1 mRNA in craniofacial tissues of embryonic mice exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin and hydrocortisone. Abbott BD, Schmid JE, Brown JG, Wood CR, White RD, Buckalew AR, Held GA. Reproductive Toxicology Division, National Health Effects and Environmental Research Laboratory, Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA. C57BL/6N mouse embryos exposed to hydrocortisone (HC) or 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) develop cleft palate. An interaction between these agents produces clefts at doses which alone are not teratogenic. The glucocorticoid receptor (GR) and dioxin receptor (AhR) mediated these responses and their gene expression was altered by TCDD and/or HC in palates examined on gestation day (GD) 14 by Northern blot analysis and in situ hybridization. The present study quantifies AhR, AhR nuclear translocator (ARNT), and GR mRNA at 4, 12, 24, and 48 h after exposure (time 0 = dose administration at 8 A.M. on gestation day 12) on GD12 to TCDD (24 micrograms/kg), HC (100 mg/kg) or HC (25 mg/kg) + TCDD (3 micrograms/kg). The induction of CYP1A1 mRNA was also quantified at 2, 4, 6, 12, 24, and 48 h for control and TCDD-exposed samples. Total RNA was prepared from midfacial tissue of 4-6 embryos/litter at each time and dose. An RNA internal standard (IS) for each gene was synthesized, which included the gene's primer sequences separated by a pUC19 plasmid sequence. Reverse transcription-polymerase chain reaction (RT-PCR) was performed on total RNA + IS using a range of 5-7 IS concentrations across a constant level of total RNA. PCR products were separated in gels (mRNA and IS-amplified sequences differed by 30-50 bases), ethidium bromide-stained, imaged (Hamamatsu Photonics Systems, Bridgewater, NJ), and quantified with NIH Image. CYP1A1 mRNA was significantly induced in the TCDD-exposed samples at all time points examined (p = 0.005 at 2 h and 0.001 after 2 h). During palatal shelf outgrowth on GD12, AhR mRNA levels increased significantly and this was not affected by treatment with TCDD or HC + TCDD. A significant increase in GR was detected at 24 h (p < 0.05) and this was unaffected by any of the exposures. Expression of ARNT increased at 12 h (p < 0.001); however, treatment with HC or HC + TCDD blocked this increase (p < 0.05). At 24 h, the TCDD-treated embryos had significantly lower ARNT mRNA compared with controls (p < 0.001). The relative overall expression level of the genes was AhR > ARNT > GR. Within individuals, expression of AhR and/or ARNT was highly correlated with GR level. In conclusion, CYP1A1 mRNA was expressed in developing craniofacial tissue and was highly induced by TCDD exposure. AhR, ARNT, and GR mRNA are upregulated in early palatogenesis, although not on the same schedule. The TCDD-induced decrease in ARNT at 24 h after dosing and the HC and HC + TCDD-induced delay in upregulation of ARNT may affect the dynamics of heterodimer formation between AhR and ARNT. The changes in ARNT mRNA level could also affect availability of this transcriptional regulator to interact with other potential partners, and these effects, separately or in combination, may be involved in disruption of normal embryonic development. PMID: 10048155 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 232: Toxicol Sci. 1999 Jan;47(1):62-75. AhR, ARNT, and CYP1A1 mRNA quantitation in cultured human embryonic palates exposed to TCDD and comparison with mouse palate in vivo and in culture. Abbott BD, Held GA, Wood CR, Buckalew AR, Brown JG, Schmid J. Reproductive Toxicology Division, National Health Effects and Environmental Research Laboratory, Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is developmentally toxic in many species and induces cleft palate in the C57BL/6N mouse embryo. Palatogenesis in mouse and human embryos involves homologous processes at the morphological, cellular, and molecular levels. In organ culture, mouse and human palates respond similarly to TCDD. The present study quantitates the expression of AhR, ARNT, and CYP1A1 mRNA in human embryonic palates in organ culture. Palatal tissues were exposed to 1 x 10(-10), 1 x 10(-9), or 1 x 10(-8) M TCDD or control medium and sampled at 0, 2, 4, and 6 hours for quantitative RT-PCR using a synthetic RNA internal standard. Similar measurements of CYP1A1 gene expression were collected for mouse palates cultured in this model. In human palates, AhR expression correlated with ARNT and CYP1A1 mRNA expression. TCDD induction of CYP1A1 was time- and concentration-dependent. The expression of these genes presented a uniform and continuous distribution across the group of embryos, with no subset of either high or low expressors/responders. The ratio of AhR to ARNT was approximately 4:1. AhR mRNA increased during the culture period in both treated and control subjects; however, ARNT expression was relatively constant. TCDD did not alter either AhR or ARNT expression in a consistent dose- or time-related manner. Comparison of human and mouse data showed a high correlation across species for the induction of CYP1A1. Human embryos expressed approximately 350 times less AhR mRNA than the mouse, and in earlier studies it was shown that human palates required 200 times more TCDD to produce the same effects. When the morphological, cellular, and molecular responses to TCDD between mouse and human are compared, it seems highly unlikely that human embryos could be exposed to sufficient TCDD to achieve changes in palatal differentiation that would lead to cleft palate. PMID: 10048154 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 233: Toxicol Appl Pharmacol. 1999 Feb 15;155(1):82-95. Physicochemical differences in the AH receptors of the most TCDD-susceptible and the most TCDD-resistant rat strains. Pohjanvirta R, Viluksela M, Tuomisto JT, Unkila M, Karasinska J, Franc MA, Holowenko M, Giannone JV, Harper PA, Tuomisto J, Okey AB. Department of Environmental Medicine, National Public Health Institute, Kuopio, FIN-70701, Finland. Long-Evans rats (strain Turku AB; L-E) are at least 1000-fold more sensitive (LD50 about 10 microg/kg) to the acute lethal effects of 2, 3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) than are Han/Wistar (Kuopio; H/W) rats (LD50 > 9600 microg/kg). The AH receptor (AHR) is believed to mediate the toxic effects of TCDD and related halogenated aromatic hydrocarbons. We compared the AHRs of L-E and H/W rats to determine if there were any structural or functional receptor differences that might be related to the dramatic difference in the sensitivity of these two strains to the lethal effects of TCDD. Cytosols from liver and lung of the sensitive L-E rats contained about twofold higher levels of specific binding sites for [3H]TCDD than occurred in H/W rats; the Kd for binding of [3H]TCDD to AHR in hepatic cytosols was similar between the two strains. Addition of the oxyanions, molybdate or tungstate (20 mM), had little effect upon ligand binding to AHR in hepatic cytosols from L-E rats whereas in cytosols from H/W rats these agents substantially diminished or totally abolished TCDD binding. The AHR in H/W cytosols also lost ligand-binding function when NaCl (20 to 400 mM) was added to the buffer whereas, in cytosols from L-E rats, the addition of 400 mM NaCl caused the receptor complex to shift from 9S to 6S during velocity sedimentation but did not destroy ligand binding function. AHR from hepatic cytosol of both the L-E and H/W rats could be transformed to the DNA-binding state in the presence of TCDD or other dioxin congeners as assessed by gel mobility shift assays. The most dramatic difference in AHR properties between L-E and H/W rats is molecular mass. Immunoblotting of cytosolic proteins revealed that the AHR in L-E rats has an apparent mass of approximately 106 kDa, similar to the mass of the receptor previously reported in several other common laboratory rat strains. In contrast, the mass of the AHR in H/W rats is approximately 98 kDa, significantly smaller than the mass of receptor reported in any other rat strains. F1 offspring of a cross between L-E and H/W rats expressed both the 106- and the 98-kDa protein. There was no apparent difference in the mass of the AHR nuclear translocator protein (ARNT) between the two strains, but the hepatic concentration of ARNT was about three times as high in L-E as in H/W rats. It will be interesting to find out how the altered structure of the AHR in H/W rats is related to their remarkable resistance to the lethal effects of TCDD. Copyright 1999 Academic Press. PMID: 10036221 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 234: Comp Biochem Physiol C Pharmacol Toxicol Endocrinol. 1998 Nov;121(1-3):23-53. The aryl hydrocarbon receptor: a comparative perspective. Hahn ME. Biology Department, Woods Hole Oceanographic Institution, MA 02543-1049, USA. mhahn@whoi.edu The aryl hydrocarbon receptor (Ah receptor or AHR) is a ligand-activated transcription factor involved in the regulation of several genes, including those for xenobiotic-metabolizing enzymes such as cytochrome P450 1A and 1B forms. Ligands for the AHR include a variety of aromatic hydrocarbons, including the chlorinated dioxins and related halogenated aromatic hydrocarbons whose toxicity occurs through activation of the AHR. The AHR and its dimerization partner ARNT are members of the emerging bHLH-PAS family of transcriptional regulatory proteins. In this review, our current understanding of the AHR signal transduction pathway in non-mammalian and other non-traditional species is summarized, with an emphasis on similarities and differences in comparison to the AHR pathway in rodents and humans. Evidence and prospects for the presence of a functional AHR in early vertebrates and invertebrates are also examined. An overview of the bHLH-PAS family is presented in relation to the diversity of bHLH-PAS proteins and the functional and evolutionary relationships of the AHR and ARNT to the other members of this family. Finally, some of the most promising directions for future research on the comparative biochemistry and molecular biology of the AHR and ARNT are discussed. Publication Types: Review PMID: 9972449 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 235: J Biol Chem. 1999 Jan 22;274(4):2060-71. Oxygen-regulated and transactivating domains in endothelial PAS protein 1: comparison with hypoxia-inducible factor-1alpha. O'Rourke JF, Tian YM, Ratcliffe PJ, Pugh CW. Erythropoietin Group, Room 425, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, OX3 9DS, United Kingdom. Endothelial PAS protein 1 (EPAS1) is a basic helix-loop-helix Per-AHR-ARNT-Sim transcription factor related to hypoxia-inducible factor-1alpha (HIF-1alpha). To analyze EPAS1 domains responsible for transactivation and oxygen-regulated function, we constructed chimeric fusions of EPAS1 with a GAL4 DNA binding domain, plus or minus the VP16 activation domain. Two transactivation domains were defined in EPAS1; a C-terminal domain (amino acids 828-870), and a larger internal domain (amino acids 517-682). These activation domains were interspersed by functionally repressive sequences, several of which independently conveyed oxygen-regulated activity. Two types of activity were defined. Sequences lying N-terminal to and overlapping the internal transactivation domain conferred regulated repression on the VP16 transactivator. Sequences lying C-terminal to this internal domain conveyed repression and oxygen-regulated activity on the native EPAS1 C-terminal activation domain, but not the Gal/VP16 fusion. Fusions containing internal but not C-terminal regulatory domains manifested regulation of fusion protein level. Comparison of EPAS1 with HIF-1alpha demonstrated a similar organization for both proteins, and for the C terminus defined a conserved RLL motif critical for inducibility. Overall, EPAS1 sequences were less inducible than those of HIF-1alpha, and inducibility was strikingly reduced as their expression level was increased. Despite these quantitative differences, EPAS1 regulation appeared similar to HIF-1alpha, conforming to a model involving the modulation of both protein level and activity, through distinct internal and C-terminal domains. PMID: 9890965 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 236: Genes Dev. 1999 Jan 1;13(1):20-5. Identification of a novel mechanism of regulation of Ah (dioxin) receptor function. Mimura J, Ema M, Sogawa K, Fujii-Kuriyama Y. Department of Chemistry, Graduate School of Science, Tohoku University, Aoba-ku, Sendai 980-8578, Japan. Ah receptor (AhR) is a ligand-activated transcription factor that mediates pleiotropic effects of environmental pollutants such as 2,3, 7,8-tetrachlorodibenzo-p-dioxin on host animals. In addition to induction of drug-metabolizing enzymes, the liganded AhR complex was found to activate gene expression of a factor designated AhR repressor (AhRR), which inhibits AhR function by competing with AhR for dimerizing with Arnt and binding to the XRE sequence. Thus, AhR and AhRR form a regulatory circuit in the xenobiotic signal transduction pathway and provide a novel mechanism of regulation of AhR function that may determine tissue-specific sensitivity to environmental pollutants. PMID: 9887096 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 237: Arch Biochem Biophys. 1999 Jan 15;361(2):223-30. The same xenobiotic response element is required for constitutive and inducible expression of the mammalian aldehyde dehydrogenase-3 gene. Boesch JS, Miskimins R, Miskimins WK, Lindahl R. Biochemistry and Molecular Biology Research Group, University of South Dakota School of Medicine, Vermillion, South Dakota, 57069, USA. The mammalian aldehyde dehydrogenase-3 gene (ALDH3) exhibits several aspects of tissue-specific expression. Certain normal tissues, such as the cornea, constitutively express ALDH3 at very high levels. Other tissues, such as normal liver, do not express ALDH3. In liver, ALDH3 is inducible by polycyclic aromatic hydrocarbon xenobiotics by an Ah-receptor (AhR)-mediated pathway in which a liganded AhR complexes with nuclear ARNT protein, and the complex binds to a xenobiotic response element (XRE) sequence located near -3.0 kb in the ALDH3 5' flanking region and initiates transcription. We used our recently developed rat corneal epithelium culture model (Boesch et al., J. Biol. Chem. 271, 5150-5157, 1996) to study the molecular basis of constitutive ALDH3 expression. Transient transfection assays of corneal epithelium using a battery of ALDH3 5' flanking region-CAT reporter gene constructs indicate that high constitutive ALDH3 expression involves the same cis-acting elements as xenobiotic-induced ALDH3 expression in liver. These elements include a strong basal promoter region and the XRE located near -3.0 kb. Western analysis confirms the presence of AhR and ARNT proteins in 3-methylcholanthrene-treated rat liver, as well as ARNT protein in rat corneal epithelium. No AhR protein is found in rat cornea. The -3.0-kb ALDH3 XRE region contains multiple overlapping transcription factor binding sequences, including consensus sites for AhR, ARNT, HNF1, HNF4, and C/ebp. Electrophoretic mobility shift assays (EMSAs) indicate that constitutive expression of ALDH3 in cornea involves binding of ARNT, HNF1, and HNF4 to the ALDH3-XRE in an Ah-receptor-independent, ARNT-requiring manner. Transient transfection of ALDH3-CAT reporter gene constructs possessing a mutation in either the ARNT- or HNF4-DNA binding sites of the XRE confirms the functional importance of these sequence motifs in constitutive ALDH3 expression. Copyright 1999 Academic Press. PMID: 9882450 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 238: Arch Biochem Biophys. 1999 Jan 1;361(1):156-63. Functional diversity of vertebrate ARNT proteins: identification of ARNT2 as the predominant form of ARNT in the marine teleost, Fundulus heteroclitus. Powell WH, Karchner SI, Bright R, Hahn ME. Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, 02543, USA. The aryl hydrocarbon receptor nuclear translocator (ARNT) is a member of the bHLH/PAS protein superfamily. ARNT dimerizes with several PAS superfamily members, including the ligand-activated aryl hydrocarbon receptor (AHR), forming a complex that alters transcription by binding specific elements within the promoters of target genes. Two genes encode different forms of the protein in rodents: ARNT1, which is widely expressed, and ARNT2, which is limited to the brain and kidneys of adults and specific neural and branchial tissues of embryos. In an effort to characterize aryl hydrocarbon signaling mechanisms in Fundulus heteroclitus, a marine teleost that can develop heritable xenobiotic resistance, we have isolated a liver cDNA encoding an ARNT homolog. The protein exhibits AHR-dependent DNA binding capability typical of other vertebrate ARNTs. Unexpectedly, phylogenetic analysis reveals that the cDNA encodes an ARNT2. This is the only detectable ARNT sequence in Fundulus liver, gill, ovary, and brain, suggesting that ARNT2 is the predominant form of ARNT in this species. Also surprising is the relative lack of sequence identity with another fish ARNT protein, rainbow trout ARNTb, which we show forms a distinct branch outside the ARNT1 and ARNT2 clades in phylogenetic analyses. Functional diversity of ARNT proteins in fish may have important implications for the assessment of aryl hydrocarbon effects on natural populations. The increasing use of fish models in developmental and toxicological studies underscores the importance of identifying taxon-specific roles of ARNT proteins and their potential dimeric partners in the PAS superfamily. Copyright 1999 Academic Press. PMID: 9882441 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 239: J Pharmacol Exp Ther. 1998 Dec;287(3):1113-8. Lipopolysaccharide activation of murine splenocytes and splenic B cells increased the expression of aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator. Marcus RS, Holsapple MP, Kaminski NE. Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA. These studies characterized the profile of AhR and ARNT expression in primary splenocytes and purified splenic B cells after cellular activation with lipopolysaccharide (LPS). LPS treatment of mouse splenocytes markedly increased the magnitude of both AhR and ARNT steady state mRNA expression. AhR mRNA expression peaked at 8 hr post-LPS activation and was increased by approximately 5-fold compared with freshly isolated splenocytes (i.e., time 0). ARNT mRNA expression began to increase at 8 hr postactivation, peaked at approximately 48 hr and was increased by approximately 4-fold when compared with nonactivated splenocytes at time 0. Western blotting also demonstrated an increase in the relative magnitude of both the AhR and ARNT proteins in LPS activated splenocytes. Likewise, the presence of the AhR, ARNT and cytochrome P450IA1 (CYP1A1) proteins were also detected in purified primary splenic B cells, and the magnitude of protein expression was enhanced in LPS activated splenic B cells at 12 and 24 hr relative to time matched controls for each of these proteins. In summary, these findings suggest that on LPS activation the magnitude of AhR and ARNT mRNA and protein increases in both splenocytes and purified primary splenic B cells. Moreover, because the increase in the relative magnitude of CYP1A1 protein in response to LPS occurred in the absence of exogenous AhR ligand, these results suggest that B-cell activation is sufficient to induce AhR nuclear translocation and binding to dioxin-responsive elements in the promoter region of AhR-responsive genes. PMID: 9864300 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 240: Drug Metab Dispos. 1998 Dec;26(12):1194-8. The aryl hydrocarbon receptor: studies using the AHR-null mice. Gonzalez FJ, Fernandez-Salguero P. Division of Basic Sciences, National Cancer Institute, Bethesda, MD 20892, USA. fjgonz@helix.nih.gov The aryl hydrocarbon receptor (AHR) is believed to mediate the toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), polychlorinated biphenyls, and polycyclic aromatic hydrocarbons. AHR is a member of the Per, ARNT, Sim/basic-helix-loop-helix superfamily of ligand-activated transcription factors that also harbors the transcription factors involved in the hypoxia response, development of the central nervous system, and day-night adaptations. To investigate the role of AHR in chemical toxicity and carcinogenesis and to determine any possible function in mammalian development and physiological homeostasis, AHR-null mice were developed. The AHR-null mice were resistant to the acute toxicity of TCDD and had an altered teratogenic response to this compound. These mice were found to have a number of abnormal phenotypes, thus confirming that AHR plays an important developmental and physiological role. Among the most consistent phenotypes was an altered liver pathology that was associated with accelerated rates of apoptosis. Evidence suggests that this may be related to an abnormal accumulation of levels of hepatic retinoic acid that cause an activation of transforming growth factor beta, resulting in stimulation of apoptosis. AHR may directly or indirectly control levels of a cytochrome P450 that is responsible for catabolizing retinoic acid. Publication Types: Review Review, Tutorial PMID: 9860927 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 241: Biochem Biophys Res Commun. 1998 Nov 9;252(1):225-31. Daily cycle of bHLH-PAS proteins, Ah receptor and Arnt, in multiple tissues of female Sprague-Dawley rats. Richardson VM, Santostefano MJ, Birnbaum LS. U.S. Environmental Protection Agency, National Health & Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, 27711, USA. The aryl hydrocarbon receptor (AhR) shares a common PAS domain with a number of genes that exhibit a pronounced circadian rhythm. Therefore, this study examined the daily cycle of AhR and AhR nuclear translocator (Arnt) protein expression in multiple tissues of female Sprague-Dawley rats. Rats were euthanized at 4, 7, and 11 am and 4, 7, and 11 pm after which whole tissue homogenates were made from multiple tissues. Western blot analysis showed that the daily cycle of relative AhR protein expression exhibits a similar oscillation pattern in the liver, lungs, and thymus. The daily cycle of relative Arnt protein expression exhibits a similar oscillation pattern in the liver and lungs. The apparent daily cycle of AhR and Arnt protein expression in multiple tissues was not observed within the spleen. This preliminary report is the first study to suggest that the PAS proteins, AhR and Arnt, exhibit a daily oscillation pattern within multiple target tissues which may give insight into the tissue-specific toxic and biochemical responses mediated through this dimerization pair, as well as the physiological function of these proteins. Copyright 1998 Academic Press. PMID: 9813174 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 242: Toxicol Pathol. 1998 Sep-Oct;26(5):657-71. Comment in: Toxicol Pathol. 1998 Sep-Oct;26(5):672-3. Mechanisms of ligand-induced aryl hydrocarbon receptor-mediated biochemical and toxic responses. Wilson CL, Safe S. Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466, USA. The ubiquitous environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) is a member of a broad group of halogenated aromatic hydrocarbons (HAHs) that is known to induce a wide range of toxic and biochemical responses in laboratory animals and humans. The effects of HAH exposure are mediated by binding to the cytosolic aryl hydrocarbon receptor (AhR), which is expressed in a tissue- and cell type-specific manner. The AhR is a ligand-activated transcription factor belonging to the basic helix-loop-helix/Per-AhR-Arnt-Sim (bHLH/PAS) superfamily of proteins. The mechanism of induction of gene transcription by TCDD involves ligand recognition and binding by the AhR, nuclear translocation, and dimerization with the AhR cofactor, AhR nuclear translocator (Arnt). The nuclear heterodimer interacts with cognate xenobiotic responsive elements (XREs) in promoter/enhancer regions of multiple Ah-responsive genes. Subsequent changes in chromatin structure and/or interaction of the AhR complex with the basal transcriptional machinery play a significant role in AhR-mediated gene expression. Although Arnt is a necessary component of a functional nuclear AhR complex, this protein also forms transcriptionally active heterodimers with other bHLH/PAS factors, including those involved in the transcriptional response to hypoxia. Arnt is ubiquitously expressed in mammalian systems, and results from transgenic mouse studies suggest that this protein plays a vital role in early mammalian embryonic development. Similar experiments suggest that the AhR may be involved in development of various organ systems. Thus, molecular mechanistic studies of TCDD action have contributed significantly to an improved understanding of the role of at least 2 bHLH/PAS proteins, as well as organ- and tissue-specific biochemical and toxic responses to this class of environmental toxins. Publication Types: Review Review, Tutorial PMID: 9789953 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 243: Teratology. 1998 Aug;58(2):30-43. AH receptor, ARNT, glucocorticoid receptor, EGF receptor, EGF, TGF alpha, TGF beta 1, TGF beta 2, and TGF beta 3 expression in human embryonic palate, and effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Abbott BD, Probst MR, Perdew GH, Buckalew AR. Division of Reproductive Toxicology, National Health Effects and Environmental Research Laboratory, Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA. Protein and mRNA for epidermal growth factor (EGF), transforming growth factor-alpha (TGF alpha), EGF receptor, transforming growth factor-beta 1 (TGF beta 1), TGF beta 2, TGF beta 3, glucocorticoid receptor (GR), the aryl hydrocarbon receptor (AhR), and the Ah receptor nuclear translocator (ARNT) were localized in gestational days (GD) 49-59 human embryonic secondary palates. The response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was determined for expression of these genes following palatal organ culture. Craniofacial tissues were shipped in medium from the Human Embryology Laboratory, University of Washington, Seattle, WA. Half of each specimen was cultured in control medium and half in medium containing TCDD at either 1 x 10(-8) or 1 x 10(-10) M. After fixation and paraffin-embedding, sections were examined either immunohistochemically or by in situ hybridization. Expression patterns were determined for each gene for the major stages of palatogenesis and in response to TCDD and compared to previously determined patterns of expression in the same developmental stages of palatogenesis for the mouse (GD49-59 in human palatogenesis corresponds to GD12-16 in the mouse). Human and mouse palates were dissimilar in particular spatiotemporal patterns of expression of these genes. Relative to patterns in mouse palatal development, human tissues demonstrated expression of EGF at early palatal stages, expression of EGF receptor and TGF alpha throughout fusion events, and uniform expression of TGF beta 3 in all epithelial regions without specifically higher levels in the medial cells. The responses to TCDD also differed in patterns of gene expression as well as in concentration required to induce hyperplasia of the medial epithelium. In summary, human palates expressed all of these regulatory genes, responses to TCDD were detected, and comparison between mouse and human palates revealed interspecies variation that may be a factor in each species' response to TCDD, as well as other teratogenic exposures. PMID: 9787404 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 244: Biochem Pharmacol. 1998 Sep 1;56(5):599-612. Regulation of cytochrome P450 enzymes by aryl hydrocarbon receptor in human cells: CYP1A2 expression in the LS180 colon carcinoma cell line after treatment with 2,3,7,8-tetrachlorodibenzo-p-dioxin or 3-methylcholanthrene. Li W, Harper PA, Tang BK, Okey AB. Department of Pharmacology, University of Toronto, Ontario, Canada. It has been difficult to study the regulation of cytochrome P4501A2 (CYP1A2) because expression of this enzyme is reported to be limited or absent in cell culture. We found that CYP1A2 can be induced significantly by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 3-methylcholanthrene (MC), or benz[a]anthracene in the human colon carcinoma cell line LS180. TCDD and MC each caused a dramatic elevation of CYP1A2 mRNA, as assessed by reverse transcription-polymerase chain reaction or by northern blot analysis. TCDD also increased immunoreactive CYP1A2 protein and the activity of phenacetin-O-deethylase, a diagnostic catalytic marker for CYP1A2. The induction of CYP1A2 at all levels (mRNA, protein, catalytic activity) was concentration- and time-dependent: the EC50 for mRNA induction by TCDD = 0.5 nM, and by MC = 1.4 microM. Inducible CYP1A2 mRNA also was detected at lower levels in two other human cell lines, the hepatoma cell line HepG2 and the breast carcinoma cell line MCF-7. CYP1A1 and CYP1B1, additional CYP1 enzymes regulated by the aryl hydrocarbon receptor (AHR), also were inducible by TCDD and MC in LS180 cells; their concentration-dependent induction was highly correlated with induction of CYP1A2 at mRNA, protein, and catalytic levels. CYP1B1 was constitutively expressed and inducible in the LS180, MCF-7, and HepG2 cell lines as well as in the human choriocarcinoma cell line JEG-3 and the squamous cell carcinoma line A431. CYP1A2 was neither constitutively expressed nor inducible in A431 or JEG-3 cells. The expression of mRNAs encoding the regulators of CYP1 enzymes-the AHR and its heterodimerization partner, the ARNT (AH receptor nuclear translocator) protein-was not altered by treatment with TCDD or MC. However, the cytosolic content of AHR protein and ARNT protein was depleted substantially following treatment with TCDD. The LS180 cell line should constitute a good model for further mechanistic studies on AHR-regulated CYP1A2 expression. PMID: 9783729 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 245: Toxicol Appl Pharmacol. 1998 Sep;152(1):200-10. Characterization of 2,3,7,8-tetrachlorodibenzofuran-dependent suppression and AH receptor pathway gene expression in the developing mouse mammary gland. Hushka LJ, Williams JS, Greenlee WF. Department of Pharmacology and Molecular Toxicology, University of Massachusetts Medical Center, Worcester, Massachusetts 01655, USA. The AH receptor (AHR) is a ligand-activated transcription factor and member of a growing family of homologous proteins implicated in development. In this study we have characterized the actions of 2,3, 7,8-tetrachlorodibenzofuran (TCDF), a well-studied AHR ligand, and the expression of AHR and selected AHR signal transduction pathway genes in the developing mouse mammary gland. High levels of AHR protein were observed in the mammary glands of C57Bl/6J (AHR +/+) mice during estrous-stimulated growth and branching of terminal end buds (TEBs). Comparative analysis of mammary gland development in AHR -/- and +/+ littermates revealed a 50% reduction in TEBs and an increase in blunt-ended terminal ducts in the AHR null animals. Treatment of mammary glands, removed from estrogen/progesterone-primed C57Bl/6J mice and maintained in organ culture, with TCDF suppressed lobule development (greater than twofold decreases in lobule number and size), with a concomitant suppression of DNA synthesis, as judged by a 35 to 45% decrease in [3H]thymidine incorporation in the TEBs. Immunohistochemical staining patterns for AHR, aryl hydrocarbon nuclear translocator (ARNT; the heterodimerization partner of AHR), and two AHR-regulated genes, Cyp1A1 and Cyp1B1, were similar and not altered by treatment of mammary glands in organ culture with TCDF. The observed differences in the development of mammary glands from AHR +/+ and -/- mice, associated expression of the AHR protein with hormone-dependent lobule development, and suppressive actions of TCDF support the position that, in C57Bl/6J mice, development of the mammary gland is at least in part AHR dependent. Development occurs in the absence of exogenous AHR ligand, suggesting that the unoccupied receptor may function to support the proliferative stages required for full lobule development. Copyright 1998 Academic Press. PMID: 9772216 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 246: Mol Pharmacol. 1998 Oct;54(4):671-7. The aryl hydrocarbon receptor interacts with transcription factor IIB. Swanson HI, Yang JH. Department of Pharmacology, University of Kentucky, Lexington, Kentucky 40536, USA. hswan@pop.uky.edu The aryl hydrocarbon receptor (AHR) and its DNA binding partner, the AHR nuclear translocator (ARNT), are basic helix-loop-helix transcription factors that mediate many of the toxic and carcinogenic effects of polyhalogenated aromatic hydrocarbons. The basic regions of the AHR and ARNT contact the GCGTG recognition site, whereas both their helix-loop-helix domains and periodicity-ARNT-single-minded domains participate in heterodimerization. To delineate the transcription factors that may facilitate DNA binding and transcriptional activation of the AHR/ARNT heterodimer, we questioned whether transcription factor IIB (TFIIB) may interact with either the AHR or ARNT and whether this interaction may affect the ability of the AHR/ARNT complex to bind DNA. Coaffinity precipitation assays demonstrated that both the AHR and ARNT were capable of interacting with TFIIB. Domain mapping experiments revealed that TFIIB interacts with the periodicity-ARNT-single-minded and carboxyl-terminal regions of the AHR. To determine whether the interaction between TFIIB and the AHR may affect DNA binding of the AHR and ARNT complex, we performed gel shift experiments in the absence and presence of TFIIB. The addition of TFIIB significantly increased the formation of the AHR/ARNT DNA binding complex, but only if TFIIB was first allowed to interact with the AHR before the addition of ARNT. These results indicate that TFIIB interacts with the AHR and may stabilize the DNA binding form of the AHR and thereby augment the ability of the AHR/ARNT complex to interact with its DNA recognition site. PMID: 9765510 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 247: Prostate. 1998 Oct 1;37(2):98-108. Expression of the aryl hydrocarbon receptor (AhR) and the aryl hydrocarbon receptor nuclear translocator (ARNT) in fetal, benign hyperplastic, and malignant prostate. Kashani M, Steiner G, Haitel A, Schaufler K, Thalhammer T, Amann G, Kramer G, Marberger M, Scholler A. Department of Urology, University of Vienna, Austria. BACKGROUND: Androgen-dependent tissue has been reported to be affected by chemical ligands of the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, which heterodimerizes with the aryl hydrocarbon receptor nuclear translocator protein (ARNT). METHODS: Fetal (n = 3), benign hyperplastic (BPH) (n = 10), and carcinomatous (CaP) (n = 19) prostate tissues were analyzed using immunohistochemistry. Western blot analysis was used to confirm the identity of the recognized proteins. RESULTS: Immunoblotting of enriched prostatic epithelial cells (EC) and stromal cells revealed constitutive expression of bands at around 110 kDa and 90 kDa, using anti-AhR and anti-ARNT, respectively. Immunohistology of the fetal specimens revealed heterogeneous cytoplasmic and nuclear AhR expression of immature EC and mesenchymal cells. Constitutive expression of AhR (primarily cytoplasmic) and ARNT (nuclear and cytoplasmic) by the majority of adult basal and secretory EC, CaP, and smooth muscle cells was confirmed in situ. The most intense anti-AhR/-ARNT reactivity was found on smooth muscle cells, followed by EC and fibrocytes. Secretory BPH-EC revealed significantly decreased AhR expression when compared to normal tissue segments. By contrast, anti-AhR reactivity was frequently increased in the more dedifferentiated tumor areas. CONCLUSIONS: These findings suggest that an undefined physiologic AhR ligand(s) as well as environmental factors may exert effects on EC and smooth muscle cells in the prostate through binding to these receptors. PMID: 9759704 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 248: J Biol Chem. 1998 Sep 18;273(38):24867-73. Structure and expression of the mouse AhR nuclear translocator (mArnt) gene. Wang F, Gao JX, Mimura J, Kobayashi A, Sogawa K, Fujii-Kuriyama Y. Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan. Aryl hydrocarbon receptor (AhR) nuclear translocator (Arnt) gene has been isolated and characterized from a mouse genomic DNA library. The gene is about 60 kilobases long and split into 22 exons. An unusual exon/intron junctional sequence was found in the 11th intron of the gene that begins with GC at its 5'-end. The exon/intron arrangement of mArnt gene differs greatly from those of the other members of the same basic-helix-loop-helix/PAS family. The gene is TATA-less and has several transcription start sites. The promoter region of the mArnt gene is GC-rich and contains a number of putative regulatory DNA sequences such as two GC-boxes, a cAMP-responsive element, E-box, AP-1 site, and CAAT-box. Deletion experiments revealed that all these DNA elements made substantial contributions to a high level of expression of the gene, except for the cAMP-responsive element. Of all, two GC-boxes displayed the most dominant enhancing effects. It was demonstrated that there exist specific factors binding to these DNA elements in the nuclear extracts of HeLa cells. Among them, Sp1 and Sp3, and CAAT-box binding factor-A were identified to bind the GC-boxes and CAAT-box, respectively. Expression of MyoD in HeLa cells stimulated the Arnt promoter activity by binding to the E-box. PMID: 9733792 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 249: Genomics. 1998 Jul 15;51(2):223-32. Physical localization of the mouse aryl hydrocarbon receptor nuclear translocator-2 (Arnt2) gene within the c112K deletion. Wines ME, Tiffany AM, Holdener BC. Program in Genetics, State University of New York at Stony Brook 11794-5215, USA. The albino deletions identify at least seven functional intervals essential for pre- and postnatal development in the 6- to 10-cM region surrounding the albino coat color (c = tyrosinase) locus on mouse chromosome 7. The c112K deletion identifies a putative thymus functional region not removed by the overlapping c3H deletion. Cloning the c3H proximal breakpoint provided a starting point for construction of an 840-kb BAC contig spanning the c112K and c3H (D7Ssb3Hp) proximal breakpoints. These breakpoints are separated by 320-350 kb. The aryl hydrocarbon receptor nuclear translocator-2 (Arnt2) is completely removed by the c112K deletion and spans 130-170 kb of the interval. Although Arnt2 is a candidate for the thymus defects in c112K homozygotes, the possibility that other as yet unidentified genes in the c112K deletion are responsible for the abnormalities has not been ruled out. Arnt2 is a member of the bHLH-PAS (Per, Ahr, Arnt, Sim) family of transcription factors and shares the highest similarity with Arnt. The survival of c112K homozygotes markedly contrasts the embryonic lethality observed in Arnt-deficient embryos and suggests distinct roles for these related transcription factors during embryogenesis. PMID: 9722945 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 250: Indian J Exp Biol. 1998 May;36(5):437-46. Gene regulation of cytochrome P450--an overview. Ramana KV, Kohli KK. Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India. Xenobiotics have played a role in elucidating the regulation of gene expression of hepatic cytochrome P450 in the eukaryotes. The major regulation of P450 genes in the eukaryotes is at the transcriptional and post transcriptional level. Polycyclic aromatic hydrocarbons regulate the gene expression by binding the cytosolic aryl hydrocarbon receptor and its translocation to the nucleus where it forms a ternary complex with aryl hydrocarbon nuclear translocator. The ternary complex PAH-AHR-ARNT acts as a transcription factor and binds aromatic hydrocarbon responsive element to increase the expression of CYP1A1 gene. Phenobarbitone and ethanol regulate the expression of respective P450s within CYP2 gene family by different mechanisms but without the involvement of a cytosolic receptor. PB uses phosphorylation as a switch to increase the affinity of the transcription factor(s) for the positive rather than negative PB regulatory element within CYP2B1/2. This is one of the novel ways that nature has designed for a protein to act as a negative as well as a positive acting transcription factor. Ethanol regulates the expression of CYP2E1 by posttranslational stabilization making it resistant to the proteolytic digestion. Steroids regulate expression of CYP3A genes through a receptor mediated mechanism. The binary complex of the steroid and its receptor increases the transcription of CYP3 genes by binding glucocorticoid responsive element which is already occupied by another protein. Peroxisome proliferators also follow a receptor mediated mechanism in which a binary complex of PP activated receptor and retinoid X receptor acts a transcription factor and increases the expression of CYP4A genes by binding peroxisome proliferator responsive element. These studies demonstrate that PAH, glucocorticoids and PP follow a receptor mediated whereas PB and ethanol follow a nonreceptor mediated mechanism for the regulation of respective P450 genes in the eukaryotes. Publication Types: Review Review, Tutorial PMID: 9717460 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 251: Biochem Biophys Res Commun. 1998 Aug 10;249(1):131-7. Depletion of arylhydrocarbon receptor during adipose differentiation in 3T3-L1 cells. Shimba S, Todoroki K, Aoyagi T, Tezuka M. Department of Hygienic Chemistry, College of Pharmacy, Nihon University, Chiba, Japan. Arylhydrocarbon receptor (AhR) is the receptor for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds. Although a physiological ligand for AhR has yet to be identified, several lines of evidence suggest that AhR may play an important role not only in the regulation of xenobiotic metabolism but also in the maintenance of homeostatic functions. When TCDD is administrated in vivo, this compound is primarily deposited in adipose tissue. Therefore, it is critical to know the states of AhR in adipose cells for assessing the expression of toxicities of TCDD and related compounds in vivo. In this report, we examined the levels of AhR protein and its associated protein (Arnt) during the adipose differentiation in 3T3-L1 cells. The level of AhR protein was found to decrease with ongoing adipose differentiation in 3T3-L1 cells. The binding activity to the xenobiotic response element and the cellular response to TCDD were also lowered as a result of adipose differentiation. These results indicate that the depletion of AhR is a novel event associated with adipose differentiation in 3T3-L1 cells and that the magnitude of the depletion of AhR is sufficient for 3T3-L1 cells to lose the functional response to xenobiotics. We also found a population of 3T3-L1 cells which have an adipose differentiation capability in the presence of high doses of TCDD. These cells lack nuclear AhR but not cytoplasmic AhR, suggesting a possible negative role of liganded nuclear AhR in adipose differentiation. The level of the Arnt protein also decreased as a result of the differentiation. However, the pattern of the depletion of the Arnt protein was distinct from that of the AhR protein. The data presented in this study will provide opportunities to carry out studies to better understand the roles of AhR in adipose cells which are the primary targets of TCDD. PMID: 9705844 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 252: Biochem Biophys Res Commun. 1998 Jul 30;248(3):554-8. Strain differences in cytochrome P4501A1 gene expression caused by 2,3,7,8-tetrachlorodibenzo-p-dioxin in the rat liver: role of the aryl hydrocarbon receptor and its nuclear translocator. Jana NR, Sarkar S, Yonemoto J, Tohyama C, Sone H. Chemical Exposure and Health Effects Research Team, National Institute for Environmental Studies, Ibaraki, Japan. Rat strain variation in hepatic cytochrome P4501A1 (CYP1A1) gene expression caused by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was investigated along with possible underlying mechanism. TCDD at a single oral dose of 13.5 ng/kg body weight significantly increased hepatic CYP1A1 mRNA expression in DRH, Long-Evans Cinamon (LEC), Long-Evans (LE), and Holtzman (HO) rats, but not in Sprague-Dawley (SD), Wistar-Imamichi (WI), Lewis (LEW), and Fisher-344 (F344) strains. All showed significant induction of CYP1A1 mRNA at a dose of 40 ng/kg, the relative levels decreasing in the order DRH, LEC, HO, LE, F344, WI, LEW, and SD. A more than 35-fold difference in the induction of CYP1A1 RNA was evident between the DRH and SD strains. Based on CYP1A1 induction, classification into two distinctly separate groups was possible, high responders (DRH, LEC, HO, and LE) and low responders (SD, LEW, WI, and F344). The expression levels closely correlated with the steady-state aryl hydrocarbon receptor (AhR) mRNA expression, this being approximately four-fold higher in the high than in the low responder group. Analysis of the aryl hydrocarbon receptor nuclear translocator (ARNT) showed the presence of a wild type as well as an alternately spliced variant in all strains, with a 45-bp deletion whose sequence corresponded to part of 5' end of the basic region of the basic helix-loop-helix domain. Expressed levels of both products were almost equal in all the strains except DRH, LEC and HO, where the wild form predominated. The results suggest that differential expression of both AhR and ARNT are responsible for rat strain-specific differences in TCDD induced CYP1A1 expression. PMID: 9703964 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 253: Nucleic Acids Res. 1998 Aug 1;26(15):3577-83. Assessment of aryl hydrocarbon receptor complex interactions using pBEVY plasmids: expressionvectors with bi-directional promoters for use in Saccharomyces cerevisiae. Miller CA 3rd, Martinat MA, Hyman LE. Environmental Health Sciences Department and Tulane-Xavier Center for Bioenvironmental Research,Tulane University School of Public Health and Tropical Medicine, 1430 Tulane Avenue, New Orleans,LA 70112, USA. The pBEVY (bi-directional expression vectors for yeast) plasmids were designed with constitutive and galactose-induced bi-directional promoters to direct the expression of multiple proteins in Saccharomyces cerevisiae . Using human estrogen receptor as a test gene, relatively balanced expression levels from each side of a bi-directional promoter were observed. Expression of a functional heterodimeric transcription factor composed of human aryl hydrocarbon receptor (Ahr) and aryl hydrocarbon receptor nuclear translocator (Arnt) proteins was accomplished using a single pBEVY plasmid. Previous studies suggest that inhibitory cross-talk between the estrogen receptor and the Ahr/Arnt complex may occur and that Hsp90-Ahr complex formation is important for Ahr-mediated signal transduction. Evidence for functional interaction among these proteins was investigated using pBEVY plasmids in a yeast system. No inhibitory cross-talk was observed in signaling assays performed with yeast that co-expressed Ahr, Arnt and estrogen receptor. In contrast, Ahr/Arnt-mediated signal transduction was reduced by 80% in a temperature-sensitive Hsp90 strain grown under non-permissive conditions. We conclude that pBEVY plasmids facilitate the examination of multiple protein interactions in yeast model systems. PMID: 9671822 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 254: Toxicol Appl Pharmacol. 1998 Jun;150(2):240-53. In utero and lactational exposure of the male rat to 2,3,7,8-tetrachlorodibenzo-p-dioxin impairs prostate development. 1. Effects on gene expression. Roman BL, Peterson RE. Environmental Toxicology Center, University of Wisconsin, Madison 53706, USA. In utero and lactational 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure decreases rat prostate weight without decreasing circulating androgen concentrations. Because one mechanism by which TCDD is thought to cause toxicity is by aryl hydrocarbon receptor (AhR)-mediated alterations in gene transcription, the goals of this study were to determine whether the developing prostate expresses the AhR and its dimerization partner, the AhR nuclear translocator (ARNT); to determine whether in utero and lactational TCDD exposure is capable of directly activating gene transcription in the developing prostate; and to identify prostatic mRNAs that exhibit altered abundance in response to in utero and lactational TCDD exposure. Pregnant Holtzman rats were administered TCDD (1.0 microgram/kg po) or vehicle on Gestation Day (GD) 15, and male offspring were euthanized between Postnatal Days (PNDs) 1 and 63. Using reverse transcriptase-polymerase chain reaction (RT-PCR), mRNAs encoding the AhR and ARNT were detected in both ventral and dorsolateral prostates from control animals throughout postnatal development. ARNT protein was expressed in the majority of stromal nuclei early in development, whereas ARNT expression in the prostate epithelium was initially cytoplasmic but became nuclear as development progressed. GD 15 TCDD exposure increased cytochrome P4501A1 (CYP1A1) mRNA and protein in whole prostates between PNDs 7 and 21. In these TCDD-exposed animals, CYP1A1 protein was localized to the epithelium. In order to define other genes in the developing prostate that might be regulated by TCDD at the level of mRNA, RNA samples from PND 21 whole prostates from control and TCDD-exposed animals were compared using mRNA differential display. Although no growth-regulatory candidates were identified using this screening technique, a ventral prostate-specific, androgen-regulated mRNA (20-kDa protein) was identified that seemed to be downregulated by TCDD exposure. Northern blot analysis confirmed this decrease at PND 21 and further showed that the downregulation was transient. Similar results were obtained for four additional androgen-regulated prostatic mRNAs (prostatic binding protein [PBP], Royal Winnipeg Ballet [RWB], probasin, and dorsal protein-1 [DP-1]), all of which are markers of a differentiated ductal epithelium. In contrast, TCDD exposure of adult male rats (25 micrograms TCDD/kg, 24 h) greatly induced CYP1A1 mRNA without affecting the abundance of prostate-specific, androgen-regulated mRNAs. These results suggest that the transient decreases in androgen-regulated prostatic mRNA abundance observed in response to in utero and lactational TCDD exposure were probably not the result of direct action of the activated AhR on these genes but instead were reflective of a TCDD-induced delay in prostate development. PMID: 9653055 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 255: Toxicol Appl Pharmacol. 1998 Jun;150(2):228-39. Responsiveness of the adult male rat reproductive tract to 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure: Ah receptor and ARNT expression, CYP1A1 induction, and Ah receptor down-regulation. Roman BL, Pollenz RS, Peterson RE. Environmental Toxicology Center, University of Wisconsin, Madison 53706, USA. Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) either in adulthood or during late fetal and early postnatal development causes a variety of adverse effects on the male rat reproductive system. It was therefore of interest to identify male rat reproductive organs and cell types within these organs that might be direct targets of TCDD exposure. Because TCDD toxicity could possibly be the result of alterations in gene transcription mediated by the TCDD/aryl hydrocarbon receptor (AhR)/AhR nuclear translocator (ARNT) complex, the presence of the AhR and ARNT in the various organs of the adult male reproductive tract was examined using Western blotting. Both proteins were detectable in all organs examined (testis, epididymis, vas deferens, ventral prostate, dorsolateral [combined dorsal and lateral] prostate, and seminal vesicle). Although technical difficulties precluded the immunohistochemical evaluation of AhR distribution in these organs, ARNT was localized in all organs in a variety of cell types, including germ cells, epithelial cells, fibroblasts, smooth muscle cells, and endothelial cells. Subcellular localization varied across organs and across cell types within these organs. In order to determine whether TCDD exposure could alter gene expression in these organs, animals were dosed with TCDD (25 micrograms/kg po) or vehicle and euthanized at 24 h, and cytochrome P4501A1 (CYP1A1) expression was evaluated. By Western blotting, only the ventral and dorsolateral prostates exhibited significant induction of CYP1A1. Immunohistochemistry confirmed this induction and localized CYP1A1 expression to epithelial cells of the ventral and lateral lobes of the prostate. Immunohistochemistry also revealed CYP1A1 induction in select epithelial cells in the epididymis and seminal vesicle, as well as endothelial cells in the vas deferens and seminal vesicle. No induction was observed in the testis. Finally, AhR and ARNT expression in TCDD-exposed and control animals was evaluated by Western blotting. Results revealed no effect of TCDD exposure on ARNT protein expression, while AhR expression was decreased to 5-51% of control in all organs examined. In summary, both AhR and ARNT were expressed in all organs of the adult male rat reproductive tract examined, and epithelial and/or endothelial cells within each of these organs (with the exception of the testis) were responsive to TCDD exposure in terms of CYP1A1 induction. In addition, all tissues exhibited marked reductions in AhR protein content after TCDD exposure that did not correlate with the magnitude of the CYP1A1 response. PMID: 9653054 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 256: Biochem Biophys Res Commun. 1998 Jun 18;247(2):383-6. Silencing of CYP1A1 expression in rabbits by DNA methylation. Takahashi Y, Suzuki C, Kamataki T. Faculty of Pharmaceutical Sciences, Hokkaido University, N12W6, Kita-ku, Hokkaido, Sapporo, 060-0812, Japan. Unlike most experimental animals, treatment of adult rabbits with 3-methylcholanthrene (MC) does not induce the expression of the CYP1A1 gene. In this study, we show that DNA methylation plays one of the key roles in the suppression of CYP1A1 gene expression. S1 nuclease protection assay showed that the induction of CYP1A1 mRNA by MC occurred in rabbit kidney RK13 cells but not in rabbit lung R9ab cells, while aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (Arnt) mRNAs were expressed in both cells at similar levels. Interestingly, the treatment of R9ab cells with a DNA demethylating agent, 5-aza-2'-deoxycitidine, resulted in the induction of the expression of the CYP1A1 gene by MC. The results indicate that DNA methylation is one of the factors involved in the loss of the MC-induced expression of the CYP1A1 gene. Thus, it seemed that the binding of the AhR/Arnt complex to the xenobiotic-responsive element (XRE) was inhibited by the hypermethylation of CpG dinucleotides within an XRE core sequence (5'-CGTG-3'). To explore this possibility, we compared the methylation status of XRE in R9ab cells with that in RK13 cells. A bisulfite sequence analysis using genomic DNAs from R9ab cells showed that the CpG site within XRE was highly methylated on both coding and non-coding strands. In contrast to this result, the hypomethylation of XRE was seen in RK13 cells. To examine whether or not the binding of the AhR/Arnt heterodimer to XRE is affected by the methylation status of XRE, a gel shift assay using a methylated XRE as a probe was carried out. As expected, the AhR/Arnt complex could not bind to the methylated XRE. From these results, we conclude that the cell type-specific transcription of the rabbit CYP1A1 gene is caused by DNA methylation. Copyright 1998 Academic Press. PMID: 9642136 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 257: J Biol Chem. 1998 Jun 19;273(25):15358-65. A novel response to dioxin. Induction of ecto-ATPase gene expression. Gao L, Dong L, Whitlock JP Jr. Department of Molecular Pharmacology, Stanford University School of Medicine, Stanford, California 94305-5332, USA. We used differential display to discover a new gene that the environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) regulates in mouse hepatoma cells. Its predicted amino acid sequence suggests that the gene encodes an ecto-ATPase that contains multiple glycosylation sites, conserved cysteine residues, and apyrase conserved regions. cDNA expression experiments in mouse hepatoma cells confirm that the new gene encodes an ecto-ATPase. Wild-type mouse hepatoma cells contain both constitutive and TCDD-inducible ecto-ATPase activity. Induction of ecto-ATPase gene expression by TCDD is direct and occurs at the transcriptional level. Studies in mutant hepatoma cells indicate that induction requires both the aromatic hydrocarbon receptor (AhR) and the AhR nuclear translocator (Arnt). Furthermore, induction requires AhR's transactivation domain, but not that of Arnt. Our findings reveal new aspects of dioxin's biological effects and TCDD-dependent gene regulation. PMID: 9624117 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 258: J Histochem Cytochem. 1998 Jul;46(7):825-32. Localization of xenobiotic-responsive element binding protein in rat hepatocyte nuclei after methylcholanthrene administration as revealed by in situ Southwestern hybridization. Asaka Y, Watanabe J, Kanamura S. Department of Anatomy, Kansai Medical University, Moriguchi, Osaka, Japan. Xenobiotic-responsive element binding protein (XRE-BP), a heterodimer of aryl hydrocarbon receptor (AhR) and its nuclear translocator (Arnt), regulates the transcription of cytochrome P-450 1A1 gene (CYP1A1) through XRE in response to xenobiotic inducers. For a better understanding of the regulatory mechanism of CYP1A1 through XRE, localization of XRE-BP was examined in liver sections or isolated hepatocyte nuclei from control and 3-methylcholanthrene (MC)-treated rats by in situ Southwestern hybridization, using synthetic XRE as a probe, and was observed by confocal laser scanning microscopy and electron microscopy. Gel mobility shift assay and competitive binding assay showed specificity of the synthetic XRE probe. XRE-BP was exclusively localized in hepatocyte nuclei in liver sections from animals 3 hr after MC injection, whereas the protein was absent in hepatocyte cytoplasm in MC-treated animals and in hepatocyte nuclei and cytoplasm in control animals. In isolated hepatocyte nuclei, XRE-BP began to accumulate in the central region between 0.5 and 3 hr, showed a peak between 3 and 6 hr, decreased gradually between 6 and 72 hr, and disappeared at 72 hr after MC injection. The protein was scarce in peripheral and nucleolar regions of the nucleus. Therefore, XRE-BP is formed in the nuclei of hepatocytes after MC stimulation. In addition, XRE-BP was found in isolated hepatocyte nuclei from control animals after preincubation with cytoplasmic lysate from MC-treated animals, although the protein was absent in the nuclei before the preincubation. These findings strongly suggest that AhR translocates from hepatocyte cytoplasm to the nucleus and forms XRE-BP in the nucleus after MC stimulation. PMID: 9632741 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 259: Nucleic Acids Res. 1998 Jun 15;26(12):3044-52. Functional and physical interactions between the estrogen receptor Sp1 and nuclear aryl hydrocarbon receptor complexes. Wang F, Hoivik D, Pollenz R, Safe S. Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA. 17beta-Estradiol (E2) induces cathepsin D gene expression in MCF-7 human breast cancer cells and previous analyses of the proximal promoter region of this gene identified two functional enhancer sequences; namely an Sp1(N)23estrogen-responsive element (ERE) half-site (-199 to -165) and an imperfect palindromic ERE (-119 to -107). A third region of the cathepsin D gene promoter (CD/L, -145 to -119) was also E2 responsive in transient transfection assays. A GC-rich sequence which contains two overlapping Sp1 binding sites (-145 to -135) was responsible for ER-mediated transactivation and required formation of an ER/Sp1 complex in which only the Sp1 protein bound DNA. E2 responsiveness of the CD/L sequence was also dependent on an adjacent overlapping GCGTG motif corresponding to the dioxin-responsive element (DRE) core binding sequence, which is the cognate response element for the heterodimeric aryl hydrocarbon receptor (AhR)/AhR nuclear translocator (ARNT) transcription factor complex. The results show that ER-mediated transactivation of CD/L was associated with the Sp1(N)2-4DRE (core) motif and involved formation of a multiprotein ER/Sp1-AhR/ARNT complex. These results illustrate a unique example of an endogenous role for AhR/ARNT in the absence of added AhR agonist and indicate that the cathepsin D gene proximal promoter region contains at least three different functional motifs associated with ER-mediated transactivation. PMID: 9611253 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 260: Mech Dev. 1998 Apr;73(1):117-23. Expression of ARNT, ARNT2, HIF1 alpha, HIF2 alpha and Ah receptor mRNAs in the developing mouse. Jain S, Maltepe E, Lu MM, Simon C, Bradfield CA. Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, IL, USA. The basic helix-loop-helix-PAS (bHLH-PAS) protein ARNT is a dimeric partner of the Ah receptor (AHR) and hypoxia inducible factor 1 alpha(HIF1 alpha). These dimers mediate biological responses to xenobiotic exposure and low oxygen tension. The recent cloning of ARNT and HIF1(homologues (ARNT2 and HIF2 alpha) indicates that at least six distinct bHLH-PAS heterodimeric combinations can occur in response to a number of environmental stimuli. In an effort to understand the biological relevance of this combinatorial complexity, we characterized their relative expression at a number of developmental time points by parallel in situ hybridization of adjacent tissue sections. Our results reveal that in general there is limited redundancy in the expression of these six transcription factors and that each of these bHLH-PAS members displays a unique pattern of developmental expression emerging as early as embryonic day 9.5. Copyright 1998 Elsevier Science Ireland Ltd. PMID: 9545558 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 261: Toxicol Sci. 1998 Apr;42(2):117-28. Female Sprague-Dawley rats exposed to a single oral dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin exhibit sustained depletion of aryl hydrocarbon receptor protein in liver, spleen, thymus, and lung. Pollenz RS, Santostefano MJ, Klett E, Richardson VM, Necela B, Birnbaum LS. Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston 29425, USA. There is currently little information concerning the time-dependent relationship between 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure and aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) protein concentration in vivo. Therefore, female Sprague-Dawley rats were given a single oral dose of TCDD (10 micrograms/kg), and the AHR and ARNT protein concentrations in liver, spleen, thymus, and lung determined by Western blotting. In liver, the concentration of AHR protein was significantly reduced 8 and 24 h postdosing as compared to time-matched controls. In spleen and lung, the concentration of AHR protein was reduced 3, 8, 24, and 168 h posttreatment compared to time-matched controls but returned to control levels by 336 h. In thymus, reductions in AHR protein concentration were observed 8, 24, 168, and 336 h postdosing as compared to time-matched controls. Significant reductions in the concentration of ARNT protein were not observed in any of the TCDD-exposed tissues. Functional studies in cell culture showed that exposure of a mouse hepatoma cell line (Hepa-1c1c7) and a rat smooth muscle cell line (A-7) to TCDD (1 nM) for 12 days resulted in a 50% reduction in TCDD-inducible reporter gene expression following subsequent challenge by an additional dose of TCDD (1 nM). Collectively, these results show that (i) TCDD-mediated depletion of AHR occurs in vivo, (ii) AHR protein does not rapidly recover to pretreatment levels even though the tissue concentration of TCDD has fallen, and (iii) reduction in AHR protein concentration correlates with reduction in TCDD-mediated reporter gene expression in mammalian culture cells. PMID: 9579024 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 262: Mol Pharmacol. 1998 Apr;53(4):623-9. Aryl hydrocarbon receptor-dependent suppression by 2,3,7, 8-tetrachlorodibenzo-p-dioxin of IgM secretion in activated B cells. Sulentic CE, Holsapple MP, Kaminski NE. Department of Pharmacology and Toxicology, Michigan State University, East Lansing 48824, USA. The immune system has been identified as a sensitive target for the toxic effects produced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Furthermore, the B cell has been identified as a sensitive cellular target of TCDD by previous cell-type fractionation studies from this laboratory. The mechanism responsible for the immunotoxic effects produced by TCDD is unclear; however, many of the biological effects of TCDD are thought to be mediated by the aryl hydrocarbon receptor (AhR). Here, we describe two B cell lines that differ considerably in their expression of the AhR and in their sensitivity to TCDD. Our results demonstrated a marked expression of the AhR protein in the CH12.LX B cell line but not in the BCL-1 B cell line. Transcripts for the AhR were not detected by reverse transcriptase-polymerase chain reaction in the BCL-1 cells. The AhR nuclear translocator (ARNT) protein was highly expressed in both cell lines. In addition, the AhR and ARNT are functional in CH12.LX cells as demonstrated by TCDD-induced CYP1A1 induction. TCDD did not induce CYP1A1 in BCL-1 cells. Furthermore, TCDD treatment resulted in suppression of lipopolysaccharide (LPS)-induced IgM secretion in CH12.LX cells. Conversely, TCDD-induced inhibition of IgM secretion was not demonstrated in LPS-stimulated BCL-1 cells, implicating a role for the AhR in the inhibition of B cell effector function. LPS-induced differentiation of the CH12.LX cells also resulted in a marked induction of Ahr expression which was not induced in LPS-stimulated BCL-1 cells. These studies have implicated the AhR as a critical factor in TCDD-induced inhibition of IgM secretion and have demonstrated an induction of AhR gene and protein expression after B cell activation. PMID: 9547351 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 263: Arch Biochem Biophys. 1998 Apr 1;352(1):9-18. Expression of functional aromatic hydrocarbon receptor and aromatic hydrocarbon nuclear translocator proteins in murine bone marrow stromal cells. Lavin AL, Hahn DJ, Gasiewicz TA. Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) acting through the aromatic hydrocarbon receptor (AhR) and its dimerization partner, the AhR nuclear translocator protein (arnt), elicits numerous toxicological effects including immunosuppression and thymic atrophy. Previous work has shown that TCDD alters bone marrow prothymocyte populations. These effects could be mediated at the lymphocyte level directly and/or through effects on bone marrow stromal cells, a population important in the support of lymphopoiesis. The purpose of this study was to characterize AhR and arnt expression in three murine bone marrow stromal cell lines (S17, M2-10B4, and BMS2) and in primary stromal cell cultures. Immunoblot analysis detected AhR protein in M2-10B4 and BMS2 cells. AhR protein was also detected in the primary cultures. Arnt protein could be detected in all cell cultures. Electrophoretic mobility shift assays detected TCDD-dependent dioxin-responsive element (DRE) binding in all three cell lines. DNA binding was sequence-specific and dependent on AhR, as demonstrated by the addition of unlabeled DRE DNA or of anti-AhR antibody. Results obtained with the primary cultures paralleled those seen with the stromal cell lines. The ED50 for induction of TCDD-dependent DRE binding in M2-10B4 cells was 0.21 nM. TCDD treatment did not induce stromal P4501A1 mRNA expression but did increase P4501B1 mRNA levels in all three cell lines and in the primary cultures. These results indicate that murine bone marrow stromal cells express AhR and arnt proteins. Furthermore, these proteins are functional in terms of their DRE-binding ability and potential to regulate mRNA levels in a gene-specific fashion. Copyright 1998 Academic Press. PMID: 9521805 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 264: Proc Natl Acad Sci U S A. 1998 Mar 17;95(6):2844-9. Caenorhabditis elegans orthologs of the aryl hydrocarbon receptor and its heterodimerization partner the aryl hydrocarbon receptor nuclear translocator. Powell-Coffman JA, Bradfield CA, Wood WB. Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309-0347, USA. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, until now described only in vertebrates, that mediates many of the carcinogenic and teratogenic effects of certain environmental pollutants. Here, we describe orthologs of AHR and its dimerization partner AHR nuclear translocator (ARNT) in the nematode Caenorhabditis elegans, encoded by the genes ahr-1 and aha-1, respectively. The corresponding proteins, AHR-1 and AHA-1, share biochemical properties with their mammalian cognates. Specifically, AHR-1 forms a tight association with HSP90, and AHR-1 and AHA-1 interact to bind DNA fragments containing the mammalian xenobiotic response element with sequence specificity. Yeast expression studies indicate that C. elegans AHR-1, like vertebrate AHR, requires some form of post-translational activation. Moreover, this requirement depends on the presence of the domains predicted to mediate binding of HSP90 and ligand. Preliminary experiments suggest that if AHR-1 is ligand-activated, its spectrum of ligands is different from that of the mammalian receptor: C. elegans AHR-1 is not photoaffinity labeled by a dioxin analog, and it is not activated by beta-naphthoflavone in the yeast system. The discovery of these genes in a simple, genetically tractable invertebrate should allow elucidation of AHR-1 function and identification of its endogenous regulators. PMID: 9501178 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 265: Biochem Pharmacol. 1998 Feb 15;55(4):489-97. Prolonged depletion of AH receptor without alteration of receptor mRNA levels after treatment of cells in culture with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Giannone JV, Li W, Probst M, Okey AB. Department of Pharmacology, University of Toronto, Ontario, Canada. Previous experiments have shown that the total cellular content of the AH receptor (AHR) drops rapidly after exposure of mouse hepatoma cells (Hepa-1) to the potent AHR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD); within 6 hr after treatment, less than 20% of the original cell content of AHR can be detected by radioligand binding or by immunoblotting. The goals of our current study were to determine the duration of receptor depletion following treatment with ligand and to determine if depletion is due to decreased expression of the Ahr gene that encodes the AHR. We found that depletion of AHR persisted for at least 72 hr after exposure to TCDD. Treatment with 3-methylcholanthrene caused a transient drop in total cell AHR, but the AHR levels returned to near pretreatment levels within 72 hr after the first exposure. TCDD treatment did not alter the levels of AHR mRNA as assessed by reverse transcription-polymerase chain reaction or slot blot assays. Thus, the decrease in AHR protein cannot be attributed to depression of transcription of the Ahr gene by TCDD. TCDD treatment did not alter the levels of the dimerization partner of the AHR, the AH receptor nuclear translocator protein (ARNT), or ARNT mRNA. In the presence of TCDD, both the AHR and the ARNT protein can be maintained at high levels in the nucleus if transcription is inhibited with actinomycin-D. In the absence of actinomycin-D, the AHR protein was lost rapidly, but the ARNT protein level in the cell was maintained. Together, these results suggest that the AHR protein is degraded through a selective mechanism that spares the ARNT protein and that the degradation pathway involves a protein that itself has a short half-life. PMID: 9514084 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 266: J Biochem (Tokyo). 1997 Dec;122(6):1075-9. Ah receptor, a novel ligand-activated transcription factor. Sogawa K, Fujii-Kuriyama Y. Department of Chemistry, Graduate School of Science, Tohoku University, Sendai. sogawa@mail.cc.tohoku.ac.jp The aryl hydrocarbon receptor (AhR) is widely distributed in vertebrates and is known to be involved in metabolism of xenobiotics including man-made chemicals, most of which act as a ligand for the receptor, although no endogenous ligand has yet been known. Upon binding a ligand, the receptor is activated to translocate to the nuclei, and during the nuclear translocation process, it is dissociated from the 90 kDa heat shock protein (Hsp90) to form a heterodimer with Arnt (Ah receptor nuclear translocator). The heterodimer complex binds a DNA response element termed xenobiotic responsive element (XRE) localized upstream of the target genes of many drug-metabolizing enzymes including cytochrome P4501A1 and glutathione S-transferase to activate their transcription. Recent cDNA cloning has revealed that the AhR, like Arnt, possesses characteristic structural motifs of basic helix-loop-helix and PAS domains responsible for DNA recognition, heterodimerization, and ligand binding, and functions as a novel receptor-type transcription factor. Publication Types: Review Review, Tutorial PMID: 9498548 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 267: J Biol Chem. 1997 Dec 12;272(50):31845-54. A mutation in the aryl hydrocarbon receptor (AHR) in a cultured mammalian cell line identifies a novel region of AHR that affects DNA binding. Sun W, Zhang J, Hankinson O. Department of Pathology and Laboratory Medicine, UCLA Jonsson Comprehensive Cancer Center, and Molecular Biology Institute, University of California, Los Angeles, California 90095-1732, USA. Introduction of a retroviral expression vector for the aryl hydrocarbon receptor (AHR) restores CYP1A1 inducibility to a mutant derivative of the Hepa-1 cell line that is defective in induction of CYP1A1 by ligands for the receptor. An AHR protein with normal ligand binding activity is expressed in the mutant but ligand treatment of mutant cell extract fails to induce binding of the AHR. ARNT (aryl hydrocarbon receptor nuclear translocator) dimer to the xenobiotic responsive element (XRE). AHR cDNAs derived from the mutant encode a protein that is unimpaired in ligand-dependent dimerization with ARNT, but the AHR.ARNT dimer so formed is severely impaired in XRE binding activity. The mutant cDNAs contain a C to G mutation at base 648, causing a cysteine to tryptophan alteration at amino acid 216, located between the PER-ARNT-SIM homology region (PAS) A and PAS B repeats. Introduction of the same mutation in the wild-type AHR sequence by site-directed mutagenesis similarity impaired XRE binding activity. Substitution with the conservative amino acid, serine, had no effect on XRE binding. The tryptophan mutation, but not the wild-type allele, was detectable in genomic DNA of the mutant. The implication that an amino acid within the PAS region may be involved in DNA binding indicates that the DNA binding behavior of AHR may be more anomalous than previously suspected. PMID: 9395531 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 268: J Biol Chem. 1997 Dec 12;272(50):31755-63. Signal transduction-mediated activation of the aryl hydrocarbon receptor in rat hepatoma H4IIE cells. Backlund M, Johansson I, Mkrtchian S, Ingelman-Sundberg M. Division of Molecular Toxicology, Institute of Environmental Medicine and Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 77 Stockholm, Sweden. We have investigated mechanisms of omeprazole (OME)-mediated induction of CYP1A1 and CYP3A, using the rat hepatoma H4IIE cell line, in comparison with mechanisms exerted by traditional aryl hydrocarbon receptor (AhR) ligands such as benso(a)pyrene (B(a)P) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). OME did not bind specifically to AhR, and it could not activate the AhR complex in rat cytosol to a xenobiotic-responsive element (XRE)-binding form in vitro. Genistein, a tyrosine kinase inhibitor, and daidzein, an inhibitor of casein kinase II, efficiently inhibited OME-mediated but not B(a)P- or TCDD-mediated induction of CYP1A1, as monitored at the transcriptional, mRNA, and protein levels as well as by analysis of activation of XRE-luciferase reporter constructs transfected into H4IIE cells. The protease inhibitor Nalpha-p-tosyl-L-lysine chloromethyl ketone (TLCK) and lavendustin A also had similar OME-specific effects. In addition, insulin pretreatment caused an almost complete inhibition of OME-dependent CYP1A1 induction but only partially affected TCDD and B(a)P-mediated induction of CYP1A1. Staurosporine, an inhibitor of protein kinase C, impaired the induction by both B(a)P and OME. OME caused an approximately 2-fold increase in the level of CYP3A expression, but all inhibitors used were ineffective in preventing this induction. Gel shift analysis with radiolabeled XRE and specific peptide antibodies toward AhR and aryl hydrocarbon receptor nuclear translocator protein (Arnt) revealed an OME-mediated translocation of the AhR.Arnt complex into the nuclei. Genistein inhibited the specific nuclear XRE binding caused by OME, but it potentiated the formation of the TCDD-induced XRE.AhR complex. Although daidzein was able to effectively inhibit the OME-stimulated CYP1A1 gene transcription, it did not influence the OME-dependent AhR.XRE complex formation. The data are consistent with a mechanism for OME-mediated induction of CYP1A1 that involves activation of the AhR complex via intracellular signal transduction systems and that is distinct from induction mediated by AhR ligands. PMID: 9395520 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 269: Proc Natl Acad Sci U S A. 1997 Dec 9;94(25):13743-8. Molecular evolution of two vertebrate aryl hydrocarbon (dioxin) receptors (AHR1 and AHR2) and the PAS family. Hahn ME, Karchner SI, Shapiro MA, Perera SA. Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA. mhahn@whoi.edu The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor through which halogenated aromatic hydrocarbons such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) cause altered gene expression and toxicity. The AHR belongs to the basic helix-loop-helix/Per-ARNT-Sim (bHLH-PAS) family of transcriptional regulatory proteins, whose members play key roles in development, circadian rhythmicity, and environmental homeostasis; however, the normal cellular function of the AHR is not yet known. As part of a phylogenetic approach to understanding the function and evolutionary origin of the AHR, we sequenced the PAS homology domain of AHRs from several species of early vertebrates and performed phylogenetic analyses of these AHR amino acid sequences in relation to mammalian AHRs and 24 other members of the PAS family. AHR sequences were identified in a teleost (the killifish Fundulus heteroclitus), two elasmobranch species (the skate Raja erinacea and the dogfish Mustelus canis), and a jawless fish (the lamprey Petromyzon marinus). Two putative AHR genes, designated AHR1 and AHR2, were found both in Fundulus and Mustelus. Phylogenetic analyses indicate that the AHR2 genes in these two species are orthologous, suggesting that an AHR gene duplication occurred early in vertebrate evolution and that multiple AHR genes may be present in other vertebrates. Database searches and phylogenetic analyses identified four putative PAS proteins in the nematode Caenorhabditis elegans, including possible AHR and ARNT homologs. Phylogenetic analysis of the PAS gene family reveals distinct clades containing both invertebrate and vertebrate PAS family members; the latter include paralogous sequences that we propose have arisen by gene duplication early in vertebrate evolution. Overall, our analyses indicate that the AHR is a phylogenetically ancient protein present in all living vertebrate groups (with a possible invertebrate homolog), thus providing an evolutionary perspective to the study of dioxin toxicity and AHR function. PMID: 9391097 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 270: J Biol Chem. 1997 Dec 26;272(52):32824-9. Expression of the human aryl hydrocarbon receptor complex in yeast. Activation of transcription by indole compounds. Miller CA 3rd. Department of Environmental Health Sciences and Center for Bioenvironmental Research, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana 70112, USA. rellim@mailhost.tcs.tulane.edu The human aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator protein (ARNT) were coexpressed in the yeast Saccharomyces cerevisiae to create a system for the study of this heterodimeric transcription factor. Specific transcriptional activation mediated by AHR/ARNT heterodimer, which is a functional indicator of receptor expression, was assessed by beta-galactosidase activity produced from a reporter plasmid. Yeast expressing AHR and ARNT displayed constitutive transcriptional activity that was not augmented by addition of AHR agonists in strains that required exogenous tryptophan for viability. In contrast, strains with an intact pathway for tryptophan biosynthesis responded to AHR agonists and had lower levels of background beta-galactosidase activity. Hexachlorobenzene, benzo(a)pyrene, and beta-naphthoflavone were effective AHR agonists in the yeast system, and had EC50 values of 200, 40, and 20 nM, respectively, for beta-galactosidase activity induction. Tryptophan, indole, indole acetic acid, and tryptamine activated transcription in yeast coexpressing AHR and ARNT (EC50 values approximately 300 microM). Indole-3-carbinol was an exceptionally potent AHR agonist (EC50 approximately 10 microM) in yeast. This yeast system is useful for the study of AHR/ARNT protein complexes, and may be generally applicable to the investigation of other multiprotein complexes. PMID: 9407059 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 271: Mol Cell Biol. 1998 Jan;18(1):525-35. Constitutive activation of the aromatic hydrocarbon receptor. Chang CY, Puga A. Department of Environmental Health, University of Cincinnati Medical Center, Ohio 45267-0056, USA. The ligand-activated aromatic hydrocarbon receptor (AHR) dimerizes with the AHR nuclear translocator (ARNT) to form a functional complex that transactivates expression of the cytochrome P-450 CYP1A1 gene and other genes in the dioxin-inducible [Ah] gene battery. Previous work from this laboratory has shown that the activity of the CYP1A1 enzyme negatively regulates this process. To study the relationship between CYP1A1 activity and Ah receptor activation we used CYP1A1-deficient mouse hepatoma c37 cells and CYP1A1- and AHR-deficient African green monkey kidney CV-1 cells. Using gel mobility shift and luciferase reporter gene expression assays, we found that c37 cells that had not been exposed to exogenous Ah receptor ligands already contained transcriptionally active AHR-ARNT complexes, a finding that we also observed in wild-type Hepa-1 cells treated with Ellipticine, a CYP1A1 inhibitor. In CV-1 cells, transient expression of AHR and ARNT leads to high levels of AHR-ARNT-dependent luciferase gene expression even in the absence of an agonist. Using a green fluorescent protein-tagged AHR, we showed that elevated reporter gene expression correlates with constitutive nuclear localization of the AHR. Transcriptional activation of the luciferase reporter gene observed in CV-1 cells is significantly decreased by (i) expression of a functional CYP1A1 enzyme, (ii) competition with chimeric or truncated AHR proteins containing the AHR ligand-binding domain, and (iii) treatment with the AHR antagonist alpha-naphthoflavone. These results suggest that a CYP1A1 substrate, which accumulates in cells lacking CYP1A1 enzymatic activity, is an AHR ligand responsible for endogenous activation of the Ah receptor. PMID: 9418899 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 272: Arch Biochem Biophys. 1997 Nov 15;347(2):163-73. Differentiation status of cultured murine keratinocytes modulates induction of genes responsive to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Jones CL, Reiners JJ Jr. Institute of Chemical Toxicology, Wayne State University, Detroit, Michigan 48201, USA. Primary murine keratinocytes were cultured in a chemically defined, serum-free medium which facilitated manipulation of their differentiation status. Exposure of basal cell and differentiating cultures to >/= 0.1 nM 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) preferentially elevated 7-ethoxyresorufin O-deethylase specific activities in differentiating cultures (28-fold versus 4-fold increases after 36 h of exposure). Semiquantitative reverse-transcription polymerase chain reaction (RT-PCR) analyses demonstrated the presence of constitutive mRNA transcripts corresponding to four known TCDD-inducible genes (e.g., Cyp1a1, Cyp1b1, Ahd4, and Nmo1) in both differentiating and proliferating cultures of murine keratinocytes. All four genes were induced in differentiating cultures following exposure to TCDD. No induction occurred in comparably treated basal cell cultures. Indirect immunofluorescence analyses demonstrated the presence of aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) proteins in both basal and differentiating keratinocytes. Both proteins appeared to be associated with the nucleus and their nuclear association was independent of prior exposure to TCDD. These studies suggest that AHR activation in murine skin is regulated as a function of the keratinocyte differentiation program. Copyright 1997 Academic Press. PMID: 9367521 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 273: J Biol Chem. 1997 Nov 28;272(48):30025-31. Inhibition of the transcription of CYP1A1 gene by the upstream stimulatory factor 1 in rabbits. Competitive binding of USF1 with AhR.Arnt complex. Takahashi Y, Nakayama K, Itoh S, Fujii-Kuriyama Y, Kamataki T. Division of Drug Metabolism, Faculty of Pharmaceutical Sciences, Hokkaido University, N12W6, Kita-ku, Sapporo, Hokkaido 060, Japan. kamataki@pharm.hokudai.ac.jp A xenobiotic-responsive element (XRE)-binding factor(s) other than the AhR.Arnt complex was found to inhibit the transcription of CYP1A1 gene in the liver from adult rabbits, known to be nonresponsive to CYP1A1 inducers. The constitutive factor(s) in liver nuclear extracts bound to the core sequence of XRE. The binding was eliminated by the presence of an excess amount of the AhR.Arnt complex synthesized in vitro. To identify the constitutive factor(s), a sequence similar to rabbit XRE was sought. It was found that the sequence of rabbit XRE overlapped with that of the upstream stimulatory factor 1 (USF1)-binding site in the mouse metallothionein I promoter. In fact, a super shift assay using a specific antibody against human USF1 indicated that USF1 was capable of binding to rabbit XRE. Additionally, the AhR.Arnt-mediated activation of XRE-TK/Luc reporter gene in RK13 cells was blocked by the transfection with a USF1 expression vector with the amounts of the expression vector transfected. These results indicate that the XRE of the rabbit CYP1A1 gene is recognized by the basic helix-loop-helix proteins to regulate the expression of CYP1A1 in both an agonistic (AhR.Arnt) and an antagonistic (USF1) manner. PMID: 9374477 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 274: J Biochem (Tokyo). 1997 Oct;122(4):703-10. CBP/p300 functions as a possible transcriptional coactivator of Ah receptor nuclear translocator (Arnt). Kobayashi A, Numayama-Tsuruta K, Sogawa K, Fujii-Kuriyama Y. Department of Chemistry, Graduate School of Science, Tohoku University, Sendai. A heterodimer of AhR (aryl hydrocarbon receptor) and Arnt (AhR nuclear translocator) conveys a transactivation signal of aromatic hydrocarbons such as 2,3,7,8-tetrachlorodibenzo-p-dioxin and 3-methylcholanthrene to the genes for a group of drug-metabolizing enzymes. This inducible expression of the genes is inhibited by adenovirus E1A, suggesting that CBP/p300 is somehow involved in the transactivation of the genes by the AhR and Arnt heterodimer. Yeast and mammalian two hybrid systems revealed that CBP/p300 interacted with the transactivation domain of Arnt, but not with that of AhR, via the CREB-binding domain. The pull down assay using GST-Arnt hybrid protein confirmed the interaction between Arnt and CBP/p300. Considering these results and that Arnt or Arnt2 functions as a common partner in the formation of transcriptional regulators with other bHLH/PAS proteins such as AhR, HLF, and HIF-1alpha, the possibility arises that CBP/p300 is extensively involved as a coactivator in the transactivation process by bHLH/PAS (a conserved sequence motif among Per, Arnt, and Sim) heterodimer transcription factors through interaction with Arnt or Arnt2. PMID: 9399571 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 275: Biochem Biophys Res Commun. 1997 Nov 17;240(2):293-7. Upstream stimulatory factor 1 (USF1) suppresses induction of CYP1A1 mRNA by 3-methylcholanthrene (MC) in HepG2 cells. Takahashi Y, Nakayama K, Itoh S, Kamataki T. Division of Drug Metabolism, Faculty of Pharmaceutical Sciences, Hokkaido University, Japan. In this study, an endogenous factor(s) involved in the suppression of the induction of CYP1A1 was studied. Analyzing the sequences, we found that the sequence of xenobiotic responsive element (XRE) in the upstream region of the human CYP1A1 gene was overlapped with that of the upstream stimulatory factor 1 (USF1)-binding site in mouse metallothionein I promoter. In fact, a gel shift assay using a specific competitor or mutant probes showed that the core sequence of human XRE was specifically recognized by USF1. The amount of USF1 in the nuclear extracts from HepG2 cells was smaller than that from rat and rabbit livers as assayed by the binding to XRE. To determine whether or not USF1 could inhibit the interaction of aryl hydrocarbon receptor (AhR)/AhR nuclear translocator (Arnt) complex with XRE, we transfected USF1-SR alpha expression vector into HepG2 cells. The results showed that no interaction of AhR/Arnt complex with XRE occurred even when the cells were treated with 2,3,7,8-tetrachlorodibenzofuran (TCDF). Furthermore, the S1 nuclease protection assay showed that the induction of CYP1A1 mRNA by 3-methylcholanthrene (MC) was depressed by the transfection of USF1-SR alpha into HepG2 cells. Thus, it is highly possible that USF1 negatively regulates the induction of CYP1A1 in humans. PMID: 9388470 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 276: Mol Pharmacol. 1997 Aug;52(2):202-11. 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 calculation of aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator protein in total cell lysates. Holmes JL, Pollenz RS. Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, 29425, USA. Western blot analysis was used to determine the concentration of the aryl hydrocarbon receptor nuclear translocator (ARNT) protein and aryl hydrocarbon receptor (AHR) in 11 mammalian cell culture lines derived from hepatic and nonhepatic tissues. The strategy was to first use Western blot analysis to determine the expression of ARNT or AHR in each cell line relative to its concentration in murine wild-type Hepa-1c1c7 (Hepa-1) cells. Actual ARNT and AHR concentrations in known amounts of total cell lysates were then determined by generating a standard curve with defined amounts of a highly purified ARNT or AHR protein and performing regression analysis. The results show that the level of ARNT expression in each of the cell lines is similar and represents approximately 0.001-0.002% of total cellular protein. The range of expression was only approximately 3-fold with wild-type Hepa-1 cells expressing the highest level of ARNT (33,000/cell) and canine kidney cells (MDCK line) expressing 14,000 ARNT molecules/cell. In contrast, the concentration of AHR varied by 65-fold over the different cell lines with the wild-type Hepa-1 expressing 323,000 AHR/cell and rat hepatoma cells (H4IIE) expressing 4700. The ratio of AHR to ARNT ranged from 0.3 in H4IIE cells to 10 in the Hepa-1 line with the majority of cells expressing 1-5 times more AHR than ARNT protein. Immunocytochemical staining of each cell line showed that ARNT was exclusively localized to the nuclear compartment and that a conserved nuclear localization signal mapped to the NH-terminal portion of the protein. PMID: 9271342 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 277: Biochemistry. 1997 Jul 22;36(29):9066-72. Ah receptor nuclear translocator protein heterogeneity is altered after heterodimerization with the Ah receptor. Tsai JC, Perdew GH. Department of Veterinary Science, The Pennsylvania State University, University Park, Pennsylvania 16802, USA. The Ah receptor (AhR) and the Ah receptor nuclear translocator (ARNT) are capable of forming a transcriptionally active heterodimeric complex. The biochemical events that are required for dimerization and transactivation are not fully understood. The purpose of this study was to determine whether covalent modifications of ARNT occur between ARNT existing in the monomeric form and after heterodimerization with the AhR and subsequent binding to DNA. Mouse hepatoma cell line 1c1c7 (Hepa 1) cytosol and ARNT immunoprecipitations were subjected to two-dimensional gel electrophoresis. ARNT was visualized with two antibodies, with distinct epitope specificity, and each detected a considerable level of charge heterogeneity. The pI range observed was 5.7-6.4, with the predominant form at a pI of 6.2. The AhR/ARNT heterodimer was immunoprecipitated from high-salt nuclear extract obtained from Hepa 1 cells treated with beta-naphthoflavone using an anti-AhR polyclonal antibody. This immunoprecipitate was subjected to two-dimensional gel electrophoresis, and coimmunoprecipitated ARNT was visualized. The results indicated that ARNT complexed with the AhR in the nucleus has an isoform pattern shifted toward the basic end, with the predominant isoform having a pI of 6.8. Thus, a significant shift in pI occurs during the dimerization and/or after binding to DNA. In vitro transformation of the AhR with 2,3,7,8-tetrachlorodibenzo-p-dioxin in cytosol leads to heterodimerization with ARNT. Two-dimensional gel electrophoresis of ARNT coimmunoprecipitated with the AhR revealed the same isoform pattern as seen in cytosol. This would indicate that each isoform of ARNT is capable of heterodimerizing with the AhRin vitro. ARNT is a phosphoprotein, and the more acidic isoforms appear to have a higher level of phosphorylation. PMID: 9220996 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 278: J Biol Chem. 1997 Jul 11;272(28):17640-7. A nuclear localization signal of human aryl hydrocarbon receptor nuclear translocator/hypoxia-inducible factor 1beta is a novel bipartite type recognized by the two components of nuclear pore-targeting complex. Eguchi H, Ikuta T, Tachibana T, Yoneda Y, Kawajiri K. Department of Biochemistry, Saitama Cancer Center Research Institute, 818 Komuro, Ina-machi, Kitaadachi-gun, Saitama 362, Japan. Aryl hydrocarbon receptor nuclear translocator (ARNT) is a component of the transcription factors, aryl hydrocarbon receptor (AhR) and hypoxia-inducible factor 1, which transactivate their target genes, such as CYP1A1 and erythropoietin, in response to xenobiotic aromatic hydrocarbons and to low O2 concentration, respectively. Since ARNT was isolated as a factor required for the nuclear translocation of AhR from the cytoplasm in response to xenobiotics, the subcellular localization of ARNT has been of great interest. In this investigation, we analyzed the subcellular distribution of ARNT using transient expression of a fusion gene with beta-galactosidase and microinjection of recombinant proteins containing various fragments of ARNT in the linker region of glutathione S-transferase/green fluorescent protein. We found a clear nuclear localization of ARNT in the absence of exogenous ligands to AhR, and identified the nuclear localization signal (NLS) of amino acid residues 39-61. The characterized NLS consists of 23 amino acids, and can be classified as a novel variant of the bipartite type on the basis of having two separate regions responsible for efficient nuclear translocation activity, but considerable deviation of the sequence from the consensus of the classical bipartite type NLSs. Like the well characterized NLS of the SV40 T-antigen, this variant bipartite type of ARNT NLS was also mediated by the two components of nuclear pore targeting complex, PTAC58 and PTAC97, to target to the nuclear rim in an in vitro nuclear transport assay. PMID: 9211913 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 279: Eur J Biochem. 1997 Jun 1;246(2):486-95. A point mutation responsible for defective function of the aryl-hydrocarbon-receptor nuclear translocator in mutant Hepa-1c1c7 cells. Numayama-Tsuruta K, Kobayashi A, Sogawa K, Fujii-Kuriyama Y. Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Japan. A 3,4-benzopyrene-resistant mutant clone (c4) of the mouse hepatoma Hepa-1c1c7 cell line was examined for the mutation that causes the defective function of aryl-hydrocarbon receptor (AHR) nuclear translocator (Arnt). Arnt dimerizes with AHR and mediates the induction signal of aryl-hydrocarbon hydroxylase activity. The Arnt cDNAs of c4 cells were cloned by reverse-transcription/PCR to compare the sequences with that of wild-type Arnt cDNA. The Arnt cDNA of c4 cells was found to have a single point mutation, leading to replacement of Gly326 with Asp between two internal repeats in the highly conserved Per-Arnt-Sim (PAS) domain, PAS A and PAS B. The inability of [Asp326]Arnt/AHR heterodimers to enhance reporter gene transcription under the control of the CYP1A1 gene promoter and enhancer confirmed that the G326-->D substitution was a causative mutation. While fluorescence microscopy and coimmunoprecipitation experiments showed that this mutant form of Arnt was not changed from wild-type Arnt in terms of nuclear localization or heterodimer formation with AHR, the binding activity of the [Asp326]Arnt x AHR heterodimer to the xenobiotic-responsive element was reduced markedly. Determination of the turnover rate in COS-7 cells transfected with expression plasmids for mutant Arnt or normal Arnt showed that the mutant protein turned over with an accelerated rate compared with that of the normal. Moreover, the mutant protein displayed increased proteolytic digestibility in vitro with various proteases. PMID: 9208942 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 280: Teratology. 1997 May;55(5):326-37. Effects of TCDD on Ah receptor, ARNT, EGF, and TGF-alpha expression in embryonic mouse urinary tract. Bryant PL, Clark GC, Probst MR, Abbott BD. Department of Biology, North Carolina Central University, Durham 27707, USA. Prenatal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces hydronephrosis in C57BL/6N mice. The etiology of this response involves TCDD-induced hyperplasia of ureteric epithelium, which occludes the ureteric lumen, blocking the flow of urine. The present study localizes and examines the effects of TCDD on the expression of the Ah receptor (AhR), the Ah receptor nuclear translocator (ARNT), epidermal growth factor (EGF), and transforming growth factor-alpha (TGF-alpha) in the epithelial cells of the developing urinary tract, particularly the ureteric bud derivatives (ureter and tubules). Pregnant C57BL/6N mice were dosed on gestation day (GD) 10 with either corn oil or TCDD at 12 micrograms/kg; a dose of 24 micrograms/kg is expected to induce 100% hydronephrosis. The metanephric urinary tract is morphologically detectable as early as GD 12; thus, embryos were removed on GD 12, 13, and 14, and the lower dorsal torso was prepared for immunohistochemistry or in situ hybridization. Regardless of treatment, the expression of both AhR and ARNT increased in epithelial cells of the ureter and AhR increased in the metanephric tubules from GD 12-14. In situ hybridization localized the expression of AhR and ARNT mRNAs to these derivatives of the ureteric bud and levels of mRNA increased throughout the developmental period examined. There were no significant effects of TCDD treatment on expression of AhR, while TCDD significantly decreased levels of ARNT in tubules on GD 14. The epithelial cells of the ureter and tubules expressed TGF-alpha and EGF. EGF increased from GD 12 to 13 in the tubules and ureter, but there was no difference from GD 13 to 14. Treatment with TCDD reduced TGF-alpha significantly only in tubules on GD 13. TCDD exposure significantly decreased EGF in ureter and tubule cells on both GD 13 and 14. In summary, the epithelial cells of the embryonic mouse urinary tract expressed AhR, ARNT, EGF, and TGF-alpha in developmentally dependent patterns. These proteins are involved in the regulation of embryonic cell proliferation during normal urinary tract development and are probably involved in the hyperplastic response to TCDD. PMID: 9261927 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 281: Mol Pharmacol. 1997 May;51(5):703-10. Effect of transforming growth factor-beta1 on expression of aryl hydrocarbon receptor and genes of Ah gene battery: clues for independent down-regulation in A549 cells. Dohr O, Sinning R, Vogel C, Munzel P, Abel J. Medical Institute of Environmental Hygiene, Heinrich-Heine-University of Dusseldorf, Department of Toxicology, Germany. An inhibitory effect on both constitutive and inducible expression of cytochrome P450 isoenzymes has been shown for different cytokines and growth factors. We previously described an inhibition of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1A1 mRNA and enzyme activity by transforming growth factor-beta1 (TGF-beta1) in human lung cancer A549 cells. In the present study, we report that not only TCDD-induced expression of CYP1A1 but also basal mRNA expression of CYP1A1, CYP1B1, and aryl hydrocarbon receptor (AHR) was down-regulated by TGF-beta1 in cells not treated with TCDD. In contrast, mRNA expression of the AHR partner protein Arnt (aryl hydrocarbon receptor nuclear translocator) was not influenced. Furthermore, TCDD-induced expression of CYP1B1 and NMO-1 was inhibited, and the IC50 values of 5-10 pM TGF-beta1 were in the same range as observed for inhibition of CYP1A1 and AHR mRNA expression. Transfection studies with a plasmid containing a luciferase reporter gene under control of two dioxin-responsive elements indicate an effect on AHR protein expression. Results of time-course studies revealed a parallel inhibition of AHR and CYP1 mRNA expression, indicating that TGF-beta1 is a direct negative regulator of transcription of these genes. The treatment of cells with cycloheximide led to a superinduction of TCDD-induced CYP1A1 and CYP1B1 mRNA expression and abolished the inhibitory effect of TGF-beta1 on basal as well as TCDD-induced CYP1 and AHR mRNA expression. TGF-beta1 seems not to influence the stability of AHR mRNA. The results suggest that TGF-beta1 induces rapid transcription and translation of an as-yet-unknown negative regulatory factor or factors that may directly regulate expression of AHR and genes of Ah gene battery. PMID: 9145908 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 282: Proc Natl Acad Sci U S A. 1997 Apr 29;94(9):4273-8. A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1alpha regulates the VEGF expression and is potentially involved in lung and vascular development. Ema M, Taya S, Yokotani N, Sogawa K, Matsuda Y, Fujii-Kuriyama Y. Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-77, Japan. We have isolated and characterized a cDNA for a novel Per-Arnt/AhR-Sim basic helix-loop-helix (bHLH-PAS) factor that interacts with the Ah receptor nuclear translocator (Arnt), and its predicted amino acid sequence exhibits significant similarity to the hypoxia-inducible factor 1alpha (HIF1alpha) and Drosophila trachealess (dTrh) gene product. The HIF1alpha-like factor (HLF) encoded by the isolated cDNA bound the hypoxia-response element (HRE) found in enhancers of genes for erythropoietin, vascular endothelial growth factor (VEGF), and various glycolytic enzymes, and activated transcription of a reporter gene harboring the HRE. Although transcription-activating properties of HLF were very similar to those reported for HIF1alpha, their expression patterns were quite different between the two factors; HLF mRNA was most abundantly expressed in lung, followed by heart, liver, and other various organs under normoxic conditions, whereas HIF1alpha mRNA was ubiquitously expressed at much lower levels. In lung development around parturition, HLF mRNA expression was markedly enhanced, whereas that of HIF1alpha mRNA remained apparently unchanged at a much lower level. Moreover, HLF mRNA expression was closely correlated with that of VEGF mRNA. Whole mount in situ hybridization experiments demonstrated that HLF mRNA was expressed in vascular endothelial cells at the middle stages (9.5 and 10.5 days postcoitus) of mouse embryo development, where HIF1alpha mRNA was almost undetectable. The high expression level of HLF mRNA in the O2 delivery system of developing embryos and adult organs suggests that in a normoxic state, HLF regulates gene expression of VEGF, various glycolytic enzymes, and others driven by the HRE sequence, and may be involved in development of blood vessels and the tubular system of lung. PMID: 9113979 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 283: J Biol Chem. 1997 Apr 25;272(17):11205-14. Activation of hypoxia-inducible factor-1; definition of regulatory domains within the alpha subunit. Pugh CW, O'Rourke JF, Nagao M, Gleadle JM, Ratcliffe PJ. Erythropoietin Group, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom. cwpugh@molbiol.ox.ac.uk Hypoxia-inducible factor-1 (HIF-1), a heterodimeric DNA binding complex composed of two basic-helix-loop-helix Per-AHR-ARNT-Sim proteins (HIF-1alpha and -1beta), is a key component of a widely operative transcriptional response activated by hypoxia, cobaltous ions, and iron chelation. To identify regions of HIF-1 subunits responsible for oxygen-regulated activity, we constructed chimeric genes in which portions of coding sequence from HIF-1 genes were either linked to a heterologous DNA binding domain or encoded between such a DNA binding domain and a constitutive activation domain. Sequences from HIF-1alpha but not HIF-1beta conferred oxygen-regulated activity. Two minimal domains within HIF-1alpha (amino acids 549-582 and amino acids 775-826) were defined by deletional analysis, each of which could act independently to convey inducible responses. Both these regions confer transcriptional activation, and in both cases adjacent sequences appeared functionally repressive in transactivation assays. The inducible operation of the first domain, but not the second, involved major changes in the level of the activator fusion protein in transfected cells, inclusion of this sequence being associated with a marked reduction of expressed protein level in normoxic cells, which was relieved by stimulation with hypoxia, cobaltous ions, or iron chelation. These results lead us to propose a dual mechanism of activation in which the operation of an inducible activation domain is amplified by regulation of transcription factor abundance, most likely occurring through changes in protein stability. PMID: 9111021 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 284: Toxicol Appl Pharmacol. 1997 Apr;143(2):407-19. Expression of the aryl hydrocarbon receptor (AhR) and AhR nuclear translocator during chick cardiogenesis is consistent with 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced heart defects. Walker MK, Pollenz RS, Smith SM. Department of Nutritional Sciences, University of Wisconsin, Madison 53706, USA. We examined cardiotoxicity induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the chick embryo and the cardiac expression of transcription factors, the aryl hydrocarbon receptor (AhR) which binds TCDD, and its dimer partner, the AhR nuclear translocator (Arnt). Chicken eggs were injected with control (triolein) or 1.0 pmol TCDD/g egg prior to incubation and collected on Day 10 when cardiomorphogenesis is complete. Relative to controls, TCDD increased heart wet weight (27.2 +/- 0.5 versus 36.6 +/- 1.3 mg, p < 0.001) and dry weight (2.7 +/- 0.1 versus 3.1 +/- 0.1 mg, p < 0.01), and tended to increase heart myosin content (3.5 +/- 0.6 versus 6.3 +/- 2.5 microg, p < 0.07), suggesting an increase in cardiac muscle mass and edema. Histologic and morphometric analyses revealed that 10/13 TCDD-exposed hearts exhibited enlarged right and left ventricles, thickened ventricular septum, and a thinner left ventricular wall with increased trabeculation, and 4/13 exhibited ventricular septal defects compared to controls (0/23). To evaluate AhR and Arnt expression, untreated chick embryos were collected on Days 2.2, 3, 4, 5, and 8 of incubation, preserved in Bouin's fixative, sectioned, and stained with AhR and Arnt antibodies. The AhR was expressed ubiquitously in cardiac myocytes, while Arnt expression was restricted to myocytes overlying developing septa: atrioventricular canal, outflow tract, and atrial and ventricular septa. Both proteins were absent from endocardium and endocardial-derived mesenchyme. In addition, cardiac expression of an AhR/Arnt target, cytochrome P4501A1, was restricted to myocardium coexpressing AhR and Arnt. Thus, the spatial and temporal expression of AhR and Arnt suggests that the developing myocardium and cardiac septa are potential targets of TCDD-induced teratogenicity, and such targets are also consistent with cardiac hypertrophy and septal defects observed following TCDD exposure. PMID: 9144457 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 285: J Biol Chem. 1997 Mar 28;272(13):8581-93. Characterization of a subset of the basic-helix-loop-helix-PAS superfamily that interacts with components of the dioxin signaling pathway. Hogenesch JB, Chan WK, Jackiw VH, Brown RC, Gu YZ, Pray-Grant M, Perdew GH, Bradfield CA. Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, Chicago, Illinois 60611, USA. In an effort to better understand the mechanism of toxicity of 2,3,7, 8-tetrachlorodibenzo-p-dioxin, we employed an iterative search of human expressed sequence tags to identify novel basic-helix-loop-helix-PAS (bHLH-PAS) proteins that interact with either the Ah receptor (AHR) or the Ah receptor nuclear translocator (ARNT). We characterized five new "members of the PAS superfamily," or MOPs 1-5, that are similar in size and structural organization to the AHR and ARNT. MOPs 1-4 have N-terminal bHLH and PAS domains and C-terminal variable regions. MOP5 contained the characteristic PAS domain and a variable C terminus; it is possible that the cDNA contains a bHLH domain, but the entire open reading frame has yet to be completed. Coimmunoprecipitation studies, yeast two-hybrid analysis, and transient transfection experiments demonstrated that MOP1 and MOP2 dimerize with ARNT and that these complexes are transcriptionally active at defined DNA enhancer sequences in vivo. MOP3 was found to associate with the AHR in vitro but not in vivo. This observation, coupled with the fact that MOP3 formed tighter associations with the 90-kDa heat shock protein than the human AHR, suggests that MOP3 may be a conditionally active bHLH-PAS protein that requires activation by an unknown ligand. The expression profiles of the AHR, MOP1, and MOP2 mRNAs, coupled with the observation that they all share ARNT as a common dimeric partner, suggests that the cellular pathways mediated by MOP1 and MOP2 may influence or respond to the dioxin signaling pathway. PMID: 9079689 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 286: Nature. 1997 Mar 27;386(6623):403-7. Abnormal angiogenesis and responses to glucose and oxygen deprivation in mice lacking the protein ARNT. Maltepe E, Schmidt JV, Baunoch D, Bradfield CA, Simon MC. Department of Pathology, Howard Hughes Medical Institute, University of Chicago, Illinois 60637, USA. The arylhydrocarbon-receptor nuclear translocator (ARNT) is a member of the basic-helix-loop-helix-PAS family of heterodimeric transcription factors which includes the arylhydrocarbon receptor (AHR), hypoxia-inducible factor-1alpha (HIF-1alpha) and the Drosophila single-minded protein (Sim). ARNT forms heterodimeric complexes with the arylhydrocarbon receptor, HIF-1alpha, Sim and the PAS protein Per. In response to environmental pollutants, AHR-ARNT heterodimers regulate genes involved in the metabolism of xenobiotics, whereas ARNT-HIF-1alpha heterodimers probably regulate those involved in the response to oxygen deprivation. By generating a targeted disruption of the Arnt locus in the mouse, we show here that Arnt-/- embryonic stem cells fail to activate genes that normally respond to low oxygen tension. Arnt-/- ES cells also failed to respond to a decrease in glucose concentration, indicating that ARNT is crucial in the response to hypoxia and to hypoglycaemia. Arnt-/- embryos were not viable past embryonic day 10.5 and showed defective angiogenesis of the yolk sac and branchial arches, stunted development and embryo wasting. The defect in blood vessel formation in Arnt-/- yolk sacs is similar to the angiogenic abnormalities reported for mice deficient in vascular endothelial growth factor or tissue factor. On the basis of these findings, we propose a model in which increasing tissue mass during organogenesis leads to the formation of hypoxic/nutrient-deprived cells, the subsequent activation of ARNT, and a concomitant increase in the expression of genes (including that encoding vascular endothelial growth factor) that promote vascularization of the developing yolk sac and solid tissues. PMID: 9121557 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 287: Arch Biochem Biophys. 1997 Mar 15;339(2):305-14. DNA binding and transcriptional enhancement by purified TCDD.Ah receptor complex. Henry EC, Kent TA, Gasiewicz TA. Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York, 14642, USA. henrye@envmed.rochester.edu The aryl hydrocarbon receptor (AhR) is a ligand-activated transcriptional enhancer which mediates the biochemical and toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and structurally related chemicals and which may have a role in the normal development of some tissues. We have previously reported the purification of the transformed TCDD.receptor complex from rat liver cytosol based on binding to its dioxin-responsive enhancer sequence (DRE) and that it comprises the AhR ligand-binding monomer and its dimerization partner, ARNT. The present studies were designed to compare the DRE-binding characteristics of the purified receptor with the cruder preparations that are commonly used and ultimately to determine whether the purified receptor complex itself (in the absence of additional cytosolic or nuclear factors) is capable of enhancing transcription in an in vitro system. The purified AhR retained in vitro DRE binding activity in the presence of carrier protein and dithiothreitol, and its affinity for the DRE oligonucleotide was equivalent to that of the other receptor preparations (crude and partially purified cytosolic and crude nuclear). When the ligand.receptor complex was bound to a DRE oligonucleotide containing BrdU and then UV-irradiated, two proteins in each of the receptor preparations were found to crosslink to BrdU-DRE, and we concluded that they are the AhR monomer and ARNT protein. All receptor preparations also gave a similar footprint of interaction with G-residues within the DRE consensus sequence, as assessed by methylation interference. Furthermore, purified and partially purified receptors were able to stimulate transcription from a DRE-containing template in a cell-free system in the presence of HeLa cell nuclear extract. Transcriptional enhancement was receptor dose-dependent, TCDD-dependent, and specific for the DRE sequence upstream of the promotor in our template construct. These data document for the first time that a purified TCDD.Ah receptor complex retains both specific DNA binding and transcriptional activities. This observation constitutes an important step toward understanding the mechanism of gene regulation by TCDD since it implies that the transformed receptor.ligand complex itself is competent as a transcriptional enhancer without a requirement for other factors. PMID: 9056263 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 288: Crit Rev Toxicol. 1997 Mar;27(2):109-34. Aryl hydrocarbon receptor-mediated signal transduction. Rowlands JC, Gustafsson JA. Department of Bioscience, Karolinska Institute, NOVUM, Huddinge, Sweden. The aryl hydrocarbon (or dioxin) receptor (AhR) is a ligand-activated basic helix-loop-helix (bHLH) protein that heterodimerizes with the bHLH protein ARNT (aryl hydrocarbon nuclear translocator) forming a complex that binds to xenobiotic regulatory elements in target gene enhancers. Genetic, biochemical, and molecular biology studies have revealed that the AhR mediates the toxic and biological effects of environmentally persistent dioxins and related compounds. Cloning of the receptor and its DNA-binding partner, ARNT, has facilitated detailed efforts to understand the mechanisms of AhR-mediated signal transduction. These studies have determined that this unique receptor consists of several functional domains and belongs to a subfamily of bHLH proteins that share a conserved motif termed the PAS domain. In addition, recent genetic studies have revealed that expression of the AhR is a requirement for proper embryonal development, which appears to be a common function shared by many other bHLH proteins. This review is a summary of recent molecular studies of AhR-mediated gene regulation. Publication Types: Review PMID: 9099515 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 289: J Biol Chem. 1997 Feb 14;272(7):4451-7. Two murine homologs of the Drosophila single-minded protein that interact with the mouse aryl hydrocarbon receptor nuclear translocator protein. Probst MR, Fan CM, Tessier-Lavigne M, Hankinson O. Jonsson Comprehensive Cancer Center, School of Medicine, UCLA, Los Angeles, California 90095, USA. Drosophila single-minded, which acts as a positive master gene regulator in central nervous system midline formation in Drosophila, its two mouse homologs SIM1 and SIM2, and the mammalian aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) proteins are members of the basic-helix-loop-helix.PAS family of transcription factors. In the yeast two-hybrid system, we demonstrate strong constitutive interaction of ARNT with SIM1 and SIM2 and fully ligand-dependent interaction of ARNT with AHR. Both the helix-loop-helix and the PAS regions of SIM1 and of ARNT are required for efficient heterodimerization. SIM1 and SIM2 do not form homodimers, and they do not interact with AHR. We also failed to detect homodimerization of ARNT. The interaction of ARNT with SIM1 was confirmed with in vitro synthesized proteins. Like AHR, in vitro synthesized SIM1 associates with the 90-kDa heat shock protein. SIM1 inhibits binding of the AHR.ARNT dimer to the xenobiotic response element in vitro. Introduction of SIM1 into hepatoma cells inhibits transcriptional transactivation by the endogenous AHR.ARNT dimer. The mouse SIM1. ARNT dimer binds only weakly to a proposed DNA target for the Drosophila SIM.ARNT dimer. In adult mice mRNA for SIM1 was expressed in lung, skeletal muscle, and kidney, whereas the mRNA for SIM2 was found in the latter two. ARNT is also expressed in these organs. Thus mouse SIM1 and SIM2 are novel heterodimerization partners for ARNT in vitro, and they may function both as positive and negative transcriptional regulators in vivo, during embryogenesis and in the adult organism. PMID: 9020169 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 290: Kidney Int. 1997 Feb;51(2):567-74. Oxygen- and dioxin-regulated gene expression in mouse hepatoma cells. Gassmann M, Kvietikova I, Rolfs A, Wenger RH. Institute of Physiology, University of Zurich-Irchel, Switzerland. The discovery that the oxygen-regulated transcription factor HIF-1 alpha and the dioxin receptor AhR share the common heterodimerization partner ARNT (HIF-1 beta) raised the question whether a cross-talk between oxygen and dioxin signal transduction pathways exists. To answer this question we investigated an ARNT-deficient mutant cell line (Hepa1C4), which has lost its capability of responding to dioxin. The results demonstrate that the presence of ARNT is indispensable for hypoxia-inducible HIF-1 DNA binding as well as for oxygen-regulated reporter gene activity mediated by the EPO 3' hypoxia response element (HRE). Hypoxic induction of the vascular endothelial growth factor (VEGF) gene, however, was only partially abrogated in Hepa1C4 cells, suggesting that HIF-1-independent oxygen signaling pathways might exist. We further studied HIF-1 and AhR/ARNT DNA binding activity as well as the regulation of oxygen- and xenobiotic-responsive genes by treating mouse Hepa1 hepatoma cells with hypoxia and/or the dioxin analogue ICZ. Hypoxia-inducible VEGF expression was found to be independent of ICZ-treatment, whereas ICZ-inducible cytochrome P-450IA1 expression was slightly reduced by hypoxic treatment of the cells. Interestingly, the enhancer function of a xenobiotic response element (XRE) linked to a reporter gene was induced by hypoxia, but expression of a HRE-containing reporter gene was not affected by ICZ treatment. PMID: 9027741 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 291: Eur J Biochem. 1996 Dec 15;242(3):512-8. Expression of aryl hydrocarbon receptor (AhR) and aryl hydrocarbon receptor nuclear translocator (Arnt) in adult rabbits known to be non-responsive to cytochrome P-450 1A1 (CYP1A1) inducers. Takahashi Y, Nakayama K, Shimojima T, Itoh S, Kamataki T. Division of Drug Metabolism, Faculty of Pharmaceutical Sciences, Hokkaido University, Japan. Induction of aryl hydrocarbon hydroxylase by aryl hydrocarbons occurs only in neonatal rabbits and not in adult rabbits [Kahl, G. F., Friederich, D. E., Bigelow, S. W., Okey, A. B. & Nebert, D. W. (1980) Dev. Pharmacol. Ther. 1,137-162]. In the present study, we isolated cDNA clones encoding aryl hydrocarbon receptor (AhR) and aryl hydrocarbon receptor nuclear translocator (Arnt) from adult rabbits. The deduced amino acid sequences of rabbit AhR and Arnt showed 80% and 94% identities with those of human AhR and Arnt, respectively. Rabbit AhR mRNA was predominantly expressed in the lung and liver. In contrast, rabbit Arnt mRNA was expressed at almost the same level in all tissues except for the heart, liver, and small intestine. Gel shift analysis showed that the AhR. Arnt complex could bind to the consensus xenobiotic-responsive element, which indicates that AhR expressed in adult rabbit liyers possessed binding activity to the consensus xenobiotic-responsive element in vitro, although aryl hydrocarbons did not induce the activity of AHH in adult rabbits. We propose that the incapability of adult rabbits to induce cytochrome P-450 1A1 (CYP1A1) is caused by factors other than AhR and Arnt. PMID: 9022676 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 292: Arch Biochem Biophys. 1996 Dec 15;336(2):297-308. Characterization of the aryl hydrocarbon receptor complex in human B lymphocytes: evidence for a distinct nuclear DNA-binding form. Masten SA, Shiverick KT. Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, 32610, USA. Kshiverick@qm.server.ufl.edu 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) suppresses B lymphocyte proliferation and immunoglobulin production. We previously reported that the aryl hydrocarbon receptor (AhR) complex, composed of the AhR ligand binding subunit and the Ah receptor nuclear translocator (ARNT), was constitutively present in nuclear extracts from two human B lymphocyte cell lines (Biochem. Biophys. Res. Commun. 212, 27-34, 1995). The present study compared the AhR complex in the IM-9 and PJS-91 human B lymphocyte and HepG2 human hepatoma cell lines. AhR mRNA levels in the two lymphocyte cell lines were substantially lower than those in HepG2 cells, as was immunoreactive AhR protein. In contrast, ARNT mRNA and protein were expressed at a high level in all three cell lines. TCDD induction of cytochrome P450 1A1 mRNA and protein was detected in only the PJS-91 lymphocyte cell line, and at a markedly lower level than that in HepG2 cells. In gel shift assays, the cytosolic DNA-binding AhR complex in IM-9 and PJS-91 cells was indistinguishable from that in HepG2 cells. In contrast, the nuclear DNA-binding AhR complex in IM-9 and PJS-91 cells consisted of several closely migrating species, one being recognized by an AhR antibody, while an ARNT antibody reacted with all species. Protein:DNA cross-linking analysis revealed the presence of a novel Mr 100,000 DNA-binding protein in nuclear extracts from IM-9 and PJS-91, but not HepG2, cells that was not recognized by either AhR or ARNT antibodies. These results show that IM-9 and PJS-91 human B cells constitutively express a distinct nuclear DNA-binding form of the AhR complex that may result from the presence of an additional protein or a structural variant of the AhR. PMID: 8954578 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 293: J Biol Chem. 1996 Dec 6;271(49):31657-65. Mapping the protein/DNA contact sites of the Ah receptor and Ah receptor nuclear translocator. Swanson HI, Yang J. Department of Pharmacology, University of Kentucky, University of Kentucky Medical Center, Lexington, Kentucky 40536, USA. hswan@pop.uky.edu The Ah receptor (AHR) and its DNA binding partner, the Ah receptor nuclear translocator (ARNT), are basic helix-loop-helix proteins distinguished by their PER, AHR, ARNT, and SIM (PAS) homology regions. To identify the amino acids of the AHR.ARNT heterodimer that contact the TNGCGTG recognition sequence, we have performed deletion mapping and amino acid substitutions within the N termini of both the AHR and ARNT. The ability of the variant AHR and ARNT proteins to bind DNA and activate gene transcription was determined by the gel shift analysis and transient transfection assays. We have found that the amino acids of ARNT that contact DNA are similar to those of other basic/helix-loop-helix proteins and include glutamic acid residue 83 and arginine residues 86 and 87. Although our initial experiments indicated that DNA binding of the AHR may involve two regions that are bordered by amino acids 9-17 and amino acids 34-42, further analysis demonstrated that only amino acids 34-39 are critical for the AHR.TNGC interaction. These experiments indicate that while the structural features of the ARNT.GTG complex may closely resemble that deduced for proteins such as Max, E47, and USF, the AHR.TNGC complex may represent a unique DNA binding form of basic/helix-loop-helix proteins. PMID: 8940186 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 294: J Biol Chem. 1996 Nov 29;271(48):30886-96. Isolation and expression of cDNAs from rainbow trout (Oncorhynchus mykiss) that encode two novel basic helix-loop-Helix/PER-ARNT-SIM (bHLH/PAS) proteins with distinct functions in the presence of the aryl hydrocarbon receptor. Evidence for alternative mRNA splicing and dominant negative activity in the bHLH/PAS family. Pollenz RS, Sullivan HR, Holmes J, Necela B, Peterson RE. Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA. pollenzr@musc.edu cDNAs encoding two distinct basic helix-loop-helix/PER-ARNT-SIM (bHLH/PAS) proteins with similarity to the mammalian aryl hydrocarbon nuclear translocator (ARNT) protein were isolated from RTG-2 rainbow trout gonad cells. The deduced proteins, termed rtARNTa and rtARNTb, are identical over the first 533 amino acids and contain a basic helix-loop-helix domain that is 100% identical to human ARNT. rtARNTa and rtARNTb differ in their COOH-terminal domains due to the presence of an additional 373 base pairs of sequence that have the characteristics of an alternatively spliced exon. The presence of the 373-base pair region causes a shift in the reading frame. rtARNTa lacks the sequence and has a COOH-terminal domain of 104 residues rich in proline, serine, and threonine. rtARNTb contains the sequence and has a COOH-terminal domain of 190 residues rich in glutamine and asparagine. mRNAs for both rtARNT splice variants were detected in RTG-2 gonad cells, trout liver, and gonad tissue. rtARNTa and rtARNb protein were identified in cell lysates from RTG-2 cells. Transfection of rtARNT expression vectors into murine Hepa-1 cells that are defective in ARNT function (type II) result in rtARNT protein expression localized to the nucleus. Treatment of these cells with 2,3,7,8-tetrachlorodibenzo-p-dioxin results in a 20-fold greater induction of endogenous P4501A1 protein in cells expressing rtARNTb when compared with rtARNTa, even though both proteins effectively dimerize with the aryl hydrocarbon receptor. The decreased function of rtARNTa appears to be due to inefficient binding of rtARNTa.AHR complexes to DNA. In addition, the presence of rtARNTa can reduce the aryl hydrocarbon receptor-dependent function of rtARNTb in vivo and in vitro. PMID: 8940073 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 295: Biochim Biophys Acta. 1996 Nov 15;1317(2):105-11. Retinoic acid affects the expression rate of the differentiation-related genes aryl hydrocarbon receptor, ARNT and keratin 4 in proliferative keratinocytes only. Wanner R, Panteleyev A, Henz BM, Rosenbach T. Department of Dermatology, Virchow-Clinic, Humboldt University, Berlin, Germany. rw@zedat.fu-berlin.de The environmental contaminant dioxin exerts most of its effects by activating the aryl hydrocarbon receptor (AhR). The AhR is considered to play not only a role in the regulation of xenobiotic metabolism, but also for development, growth, and differentiation. The transcript levels of the AhR and its associated translocator protein (ARNT) were found to increase with ongoing differentiation in the human keratinocyte cell line HaCaT. Correspondingly, in situ hybridization studies in normal human skin revealed an absence of AhR-expression in proliferating basal cells and increasing transcript levels in upper cell layers, in dependence of keratinocyte differentiation. AhR expression in differentiation-deficient hyperproliferative psoriatic skin was markedly decreased. When keratinocytes were continuously treated with 1 microM retinoic acid (RA), the upregulation of AhR- and ARNT-mRNA levels was inhibited as was keratin 4-expression, a marker of HaCaT-keratinocyte differentiation. In contrast, treatment of already differentiated cells with RA did not down-regulate these transcript levels. The mRNA levels of the prevalent retinoic acid receptors in keratinocytes, RAR gamma and RXR alpha, were not influenced by the process of differentiation or by addition of RA. Our data suggest that the regulation of AhR-, ARNT- and keratin 4-expression by RA is indirect and mediated by a yet to be identified factor. PMID: 8950195 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 296: Toxicol Appl Pharmacol. 1996 Nov;141(1):238-47. Dioxin induces transcription of fos and jun genes by Ah receptor-dependent and -independent pathways. Hoffer A, Chang CY, Puga A. Center for Environmental Genetics, University of Cincinnati Medical Center, Ohio 45267-0056, USA. Halogenated aromatic hydrocarbons, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; dioxin), and polycyclic aromatic hydrocarbons, such as benzo[a]pyrene, are environmental contaminants that cause many apparently unrelated toxic effects. In a previous study, we have shown that treatment of mouse hepatoma cells with TCDD or B(a)P results in an increase in mRNA levels of the immediate-early protooncogenes c-fos, c-jun, junB, and junD, and the concomitant increase of the DNA-binding activity of the transcription factor AP-1, a dimer of FOS and JUN proteins. To analyze the mechanism of fos/jun activation by TCDD we have used electrophoretic mobility shift and transient expression assays of reporter gene constructs containing response elements for 12-O-tetradecanoyl-phorbol-13-acetate (TRE), serum (SRE), cAMP (CRE), and aromatic hydrocarbons (AhRE) from the fos and jun genes fused to the firefly luciferase gene under the control of the SV40 minimal promoter. In mouse hepatoma Hepa-1 cells, which have Ah receptor (AHR) and Ah receptor nuclear translocator (ARNT) proteins, inclusion of TRE, SRE, and the AhRE motifs from c-jun and junD, but not CRE or the AhREs from c-fos, fosB, and junB, causes a large TCDD-dependent increase in luciferase expression. In agreement with these results, c-jun and junD, but not c-fos, fosB, and junB AhREs, competed with a canonical Cyp1A1 AhRE for binding to the AHR ARNT heterodimeric complex. In African Green Monkey CV-1 cells, which lack AHR, expression plasmids with AhRE motifs require coexpression of AHR and ARNT for TCDD to stimulate luciferase expression. In contrast, SRE-containing expression plasmids respond equally well to TCDD whether or not AHR and ARNT are coexpressed. These results suggest that TCDD induces expression of the immediate-early response genes fos and jun by activation of possibly three separate signal transduction pathways, at least one of which does not require a functional Ah receptor complex. PMID: 8917696 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 297: Toxicol Appl Pharmacol. 1996 Nov;141(1):76-83. Ah-receptor-dependent modulation of gene expression by aged and diluted sidestream cigarette smoke. Gebremichael A, Tullis K, Denison MS, Cheek JM, Pinkerton KE. Institute of Toxicology and Environmental Health, University of California, Davis 95616, USA. Cigarette smoke is known to induce cytochrome P4501A1 expression and activity in a variety of species. Although the elevation of this isozyme is assumed to be associated with the activation of the CYP1A1 gene through a ligand-mediated mechanism involving the Ah-receptor (AhR), this has not been determined. In this study we have examined the mechanism by which an ambient level of aged and diluted sidestream cigarette smoke (ADSS) induces cytochrome P4501A1. Effects of ADSS on C57BL/6N and DBA/ 2N mice were examined. Induction of P4501A1-associated ethoxyresorufin-O-dealkylase (EROD) activity was observed in the lungs of C57BL/6N mice, while there was no induction in DBA/ 2N mice. ADSS also induced EROD in wild-type mouse hepatoma (Hepa1c1c7) cells (hepa1), but not in variant hepa1 cells defective in the AhR nuclear translocator (ARNT) protein. ADSS exposure of recombinant hepa1 cells, stably transfected with a reporter plasmid containing the luciferase gene under control of several dioxin responsive enhancers (DREs), resulted in a time- and exposure-dependent induction of luciferase activity. ADSS-mediated induction of luciferase activity was inhibited by alpha-naphthoflavone (alpha NF), an Ah-receptor antagonist. Gel retardation analysis demonstrated that exposure to ADSS induced transformation and DNA binding of the AhR complex. In summary, our results not only indicate a role for the AhR in mediating the induction of P4501A1 by ADSS, but also demonstrate that environmentally relevant levels of ADSS must contain AhR ligands at sufficient concentrations to activate gene expression in an AhR-dependent manner. PMID: 8917678 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 298: J Biol Chem. 1996 Oct 18;271(42):26261-6. Protein kinase C modulates regulation of the CYP1A1 gene by the aryl hydrocarbon receptor. Chen YH, Tukey RH. Department of Pharmacology and UCSD Cancer Center, University of California, San Diego, La Jolla, California 92093-0636, USA. Transcriptional activation of the human CYP1A1 gene by halogenated and polycyclic aromatic hydrocarbons is mediated by the aryl hydrocarbon receptor (AhR) complex, a ligand-dependent transcription factor. A competent AhR comprises at least two components following nuclear translocation and DNA binding, the AhR and the AhR nuclear translocator (Arnt) protein, whose combined action on human CYP1A1 gene transcription is shown to be dependent upon functional protein kinase C (PKC). In the present study, we examined the effects of phorbol 12-myristate 13-acetate, a potent PKC activator, on the ligand-induced transcriptional activation of the CYP1A1 gene and cellular function of the AhR in human HepG2 101L cells. The 101L cells carry a stable transgene consisting of 1800 bases of 5'-flanking DNA and the promoter of the human CYP1A1 gene linked to the firefly luciferase structural gene (Postlind, H., Vu, T. P., Tukey, R. H. & Quattrochi, L. C. (1993) Toxicol. Appl. Pharmacol. 118, 255-262). Pretreatment of cells with 12-myristate 13-acetate enhanced ligand-induced CYP1A1 gene expression 2-3-fold. Inhibition of PKC activity blocked directly the transcriptional activation and the transactivation of the CYP1A1 gene, indicating a role for PKC in the AhR-mediated transcriptional activation process. However, the DNA binding activities of the in vitro activated and the induced nuclear AhR as measured by electrophoretic mobility shift analysis were not affected when CYP1A1 transcription was inhibited, indicating the actions of PKC to be a nuclear event that works in concert with or precedes AhR binding to the gene. These results illustrate that PKC is absolutely essential for the cellular and molecular events that control induction of CYP1A1 gene transcription. PMID: 8824276 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 299: Mol Cell Biol. 1996 Oct;16(10):5865-75. Two new members of the murine Sim gene family are transcriptional repressors and show different expression patterns during mouse embryogenesis. Ema M, Morita M, Ikawa S, Tanaka M, Matsuda Y, Gotoh O, Saijoh Y, Fujii H, Hamada H, Kikuchi Y, Fujii-Kuriyama Y. Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Japan. From a cDNA library of mouse skeletal muscle, we have isolated mouse Sim1 (mSim1) cDNA encoding a polypeptide of 765 amino acids with striking amino acid identify in basic helix-loop-helix (89% identify) and PAS (89 % identify) domains to previously identified mSim2, although the carboxy-terminal third of the molecule did not show any similarity to mSim2 or Drosophila Sim (dSim). Yeast two-hybrid analysis and coimmunoprecipitation experiments demonstrated that both of the mSim gene products interacted with Arnt even more efficiently than AhR, a natural partner of Arnt, suggesting a functional cooperativity with Arnt. In sharp contrast with dSim having transcriptional-enhancing activity in the carboxy-terminal region, the two mSims possessed a repressive activity toward Arnt in the heterodimer complex. This is the first example of bHLH-PAS proteins with transrepressor activity, although some genetic data suggest that dSim plays a repressive role in gene expression (Z. Chang, D. Price, S. Bockheim, M. J. Boedigheimer, R. Smith, and A. Laughon, Dev. Biol. 160:315-322, 1993; D. M. Mellerick and M. Nirenberg, Dev. Biol. 171:306-316, 1995). Whole-mount in situ hybridization showed restricted and characteristic expression patterns of the two mSim mRNAs in various tissues and organs during embryogenesis, such as those for the somite, the nephrogenic cord, and the mesencephalon (for mSim1) and those for the diencephalon, branchial arches, and limbs (for mSim2). From sequence similarity and chromosomal localization, it is concluded that mSim2 is an ortholog of hSim2, which is proposed to be a candidate gene responsible for Down's syndrome. The sites of mSim2 expression showed an overlap with the affected regions of the syndrome, further strengthening involvement of mSim2 in Down's syndrome. PMID: 8927054 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 300: Biochem Pharmacol. 1996 Sep 13;52(5):771-80. Identification of functional aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator in murine splenocytes. Williams CE, Crawford RB, Holsapple MP, Kaminski NE. Department of Pharmacology & Toxicology, Michigan State University, East Lansing 48824, USA. The objective of the present studies was to determine whether the aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (ARNT) protein are present and functional in B6C3F1 (C57BL/6 x C3H) mouse splenocytes. Northern analysis of poly(A) RNA isolated from splenocytes revealed transcripts of approximately 6.6 kb which hybridized to the AhR complementary DNA (cDNA) probe. Anti-AhR antibodies identified two major cytosolic forms of the AhR in splenocytes, approximately 95 and 104 kDa, corresponding to the codominately expressed Ahrb alleles in the B6C3F1 mice. Northern analysis utilizing an oligomer probe for ARNT identified three messenger RNA (mRNA) transcripts, approximately 5.6, 2.0, and 1.1 kb, in spleen which was consistent with the banding pattern observed in the B6C3F1 mouse liver. Western blotting confirmed the presence of the approximately 87 kDa ARNT protein in splenocytes. Protein quantitation by slot blot analysis demonstrated approximately 2.0-fold more AhR in liver than in splenocytes. Interestingly, ARNT was approximately 2.4-fold more abundant in splenocytes than in liver. Consistent with these results, comparison by quantitative reverse transcriptase-polymerase chain reaction analysis of AhR and ARNT transcripts in liver and splenocytes demonstrated approximately 2.3-fold more AhR transcripts in liver than in splenocytes and approximately 3.2-fold more ARNT transcripts in splenocytes than in liver. In addition, comparisons between AhR and ARNT transcripts isolated from the liver and splenocytes indicated a greater number of ARNT transcripts as compared with AhR in both preparations. TCDD treatment of splenocytes induced binding of the AhR nuclear complex to the dioxin-responsive enhancer (DRE) as detected by the electrophoretic mobility shift assay. These findings confirm that the AhR and ARNT are present in mouse splenocytes and are capable of binding to the DRE. PMID: 8765475 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 301: Mol Pharmacol. 1996 Sep;50(3):538-48. Trans-activation by the human aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator proteins: direct interactions with basal transcription factors. Rowlands JC, McEwan IJ, Gustafsson JA. Department of Biosciences, Karolinska Institute, NOVUM, Huddinge, Sweden. The aryl hydrocarbon (or dioxin) receptor (AhR) is a ligand-activated basic helix-loop-helix (bHLH) protein that heterodimerizes with the bHLH protein AhR nuclear translocator (ARNT) to form a complex that binds to xenobiotic regulatory elements in the enhancers of target genes. We used a series of fusion proteins, with a heterologous DNA-binding domain, to study independently the trans-activating function of the human AhR and ARNT proteins in yeast. The results confirm that both the human AhR and ARNT contain carboxyl-terminal trans-activation domains. The AhR has a complex trans-activation domain that is composed of multiple segments that function independently and exhibit varying levels of activation. Furthermore, these regions within the AhR cooperate when linked together, resulting in a synergistic activation of transcription. Fusion proteins of the AhR and ARNT trans-activation domains with the LexA DNA-binding domain, expressed in bacteria and purified to near-homogeneity, stimulated transcription of a minimal promoter in vitro in yeast nuclear extracts. Using this in vitro transcription assay, it was also possible to demonstrate that the AhR and ARNT trans-activation domains, in the absence of a DNA-binding domain, inhibited activated and basal transcription. Furthermore, in vitro the receptor bound selectively to the basal transcription factors, the TATA-binding protein and TFIIF, whereas ARNT bound preferentially to TFIIF. Taken together, these results suggest that AhR and ARNT activate target gene expression, at least in part, through direct interactions with basal transcription factors. PMID: 8794892 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 302: Exp Cell Res. 1996 Jul 10;226(1):154-63. Complementation of Ah receptor deficiency in hepatoma cells: negative feedback regulation and cell cycle control by the Ah receptor. Weiss C, Kolluri SK, Kiefer F, Gottlicher M. Research Center Karlsruhe, Institute of Genetics, Karlsruhe, D-76021, Germany. The Ah receptor (AhR) is a ligand-dependent transcription factor subunit that heterodimerizes with the AhR nuclear translocator (Arnt) and mediates the predominant biological effects of 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD). TCDD activates target genes in xenobiotica metabolism in many cell lines and, more specifically, delays G1-S progression of 5L hepatoma cells. Here we describe transient and stable AhR-expression analysis in AhR-deficient subclones of the TCDD-sensitive 5L cells. We tested the integrity of the AhR-signaling system beyond the lack of the receptor in the variant subclone and analyzed the role of AhR in cell cycle regulation. Transiently expressed AhR has a high basal activity on promoters containing AhR-binding sites, so-called XREs, when transfected into receptor-deficient variant cells compared to wild-type cells. Single- and double-hybrid analysis dissociates AhR ligand responsiveness, transactivation, and heterodimerization with Arnt from receptor binding to an XRE. Hybrid receptors also show the high basal activity in the absence of exogenous TCDD in AhR-deficient variant cells, indicating that the endogenous AhR-activating signal acts directly on the receptor rather than XRE-dependent promoters or DNA binding of the receptor. Stable expression of AhR in variant cell clones by retroviral infection fully reconstitutes TCDD responsiveness, including target-gene induction and delay of cell cycle progression. These AhR-reconstituted cells, like AhR-containing wild-type cells, show low basal activity of the transiently expressed AhR hybrid. Thus, the increased basal activity in AhR-deficient cells suggests a negative feedback control of AhR activity. In vitro ligand-binding assays are compatible with the idea that the increased basal activity is due to the accumulation of an AhR-binding endogenous ligand. In conclusion, AhR is causally responsible for TCDD-dependent cell cycle regulation and feedback control of AhR activity. PMID: 8660951 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 303: J Biol Chem. 1996 Jun 21;271(25):15117-23. The role of the aryl hydrocarbon receptor nuclear translocator (ARNT) in hypoxic induction of gene expression. Studies in ARNT-deficient cells. Wood SM, Gleadle JM, Pugh CW, Hankinson O, Ratcliffe PJ. Erythropoietin Group, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom. Hypoxia-inducible factor-1 (HIF-1), a DNA-binding complex implicated in the regulation of gene expression by oxygen, has been shown to consist of a heterodimer of two basic helix-loop-helix Per-AHR-ARNT-Sim (PAS) proteins, HIF-1alpha, and HIF-1beta. One partner, HIF-1beta, had been recognized previously as the aryl hydrocarbon receptor nuclear translocator (ARNT), an essential component of the xenobiotic response. In the present work, ARNT-deficient mutant cells, originally derived from the mouse hepatoma line Hepa1c1c7, have been used to analyze the role of ARNT/HIF-1beta in oxygen-regulated gene expression. Two stimuli were examined: hypoxia itself and desferrioxamine, an iron-chelating agent that also activates HIF-1. Induction of the DNA binding and transcriptional activity of HIF-1 was absent in the mutant cells, indicating an essential role for ARNT/HIF-1beta. Analysis of deleted ARNT/HIF-1beta genes indicated that the basic, helix-loop-helix, and PAS domains, but not the amino or carboxyl termini, were necessary for function in the response to hypoxia. Comparison of gene expression in wild type and mutant cells demonstrated the critical importance of ARNT/HIF-1beta in the hypoxic induction of a wide variety of genes. Nevertheless, for some genes a reduced response to hypoxia and desferrioxamine persisted in these mutant cells, clearly distinguishing ARNT/HIF-1beta-dependent and ARNT/HIF-1beta-independent mechanisms of gene activation by both these stimuli. PMID: 8662957 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 304: Biochem Mol Biol Int. 1996 Jun;39(3):589-93. Mapping the 90 kDa heat shock protein binding region of the Ah receptor. Perdew GH, Bradfield CA. Department of Veterinary Science, Pennsylvania State University, University Park 16802, USA. ghp2@psu.edu Expression of a series of Ah receptor (AhR) deletion mutants in an in vitro translation system has been previously used to map several functional domains of the murine AhR (Dolwick et al. (1993) Proc. Natl. Acad. Sci. USA 90, 8566-8570). In this report, quantitative immunoprecipitation of 90-kDa heat shock protein (hsp90) from reticulocyte lysate allowed us to measure the level of the AhR and AhR deletion mutants complexed with hsp90. After translation of a series of deletion mutants it was determined that there are two distinct domains important in forming a stable AhR/hsp90 complex, corresponding to amino acid sequences 1-166 and 289-347 of the AhR. Neither ARNT, nor Per were able to stably interact with hsp90. Thus, the AhR appears to be a unique member of the PAS domain family of proteins that binds a known ligand and stably interacts with hsp90. PMID: 8828811 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 305: J Biol Chem. 1996 May 24;271(21):12310-6. Cooperative interaction between AhR.Arnt and Sp1 for the drug-inducible expression of CYP1A1 gene. Kobayashi A, Sogawa K, Fujii-Kuriyama Y. Department of Chemistry, Faculty of Science, Tohoku University, Sendai, Japan. Expression of CYP1A1 gene is regulated in a substrate-inducible manner through at least two kinds of regulatory DNA elements in addition to the TATA sequence, XRE (xenobiotic responsive element), and BTE (basic transcription element), a GC box sequence. The trans-acting factor on the XRE is a heterodimer consisting of arylhydrocarbon receptor (AhR) and AhR nuclear translocator (Arnt), while Sp1 acts as a regulatory factor on the BTE. We have investigated how these factors interact with one another to induce expression of the CYP1A1 gene. Both in vivo transfection assays using Drosophila Schneider line 2 (SL2) cells, which is devoid of endogenous Sp1, AhR, and Arnt, and in vitro transcription assays using baculovirus-expressed AhR, Arnt, and Sp1 proteins revealed that these factors enhanced synergistically expression of the reporter genes driven by a model CYP1A1 promoter, consisting of four repeated XRE sequences and a BTE sequence, in agreement with previous observation (Yanagida, A., Sogawa, K., Yasumoto, K., and Fujii-Kuriyama, Y. (1990) Mol. Cell. Biol. 10, 1470-1475). We have proved by coimmunoprecipitation assays and DNase I footprinting that both AhR and Arnt interact with the zinc finger domain of Sp1 via their basic HLH/PAS domains. When either the AhR.Arnt heterodimer of Sp1 was bound to its cognate DNA element, DNA binding of the second factor was facilitated. Survey of DNA sequences in the promoter region shows that the XRE and GC box elements are commonly found in the genes whose expressions are induced by polycyclic aromatic hydrocarbons, suggesting that the two regulatory DNA elements and their cognate trans-acting factors constitute a common mechanism for induction of a group of drug-metabolizing enzymes. PMID: 8647831 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 306: Arch Biochem Biophys. 1996 May 1;329(1):47-55. Characterization of the activated form of the aryl hydrocarbon receptor in the nucleus of HeLa cells in the absence of exogenous ligand. Singh SS, Hord NG, Perdew GH. Department of Veterinary Science, Pennsylvania State University, University Park 16802, USA. The aryl hydrocarbon receptor (AhR) is known to mediate 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced toxic effects. Immunocytochemical studies revealed that AhR in HeLa cells is localized throughout the cell. Upon TCDD treatment most of the cytoplasmic receptor is translocated into the nucleus in a time-dependent manner. A significant amount of AhR was found to be tightly associated with the nuclear fraction of untreated HeLa cells. The level of receptor in the nuclear fraction was approximately 16% of the total cellular receptor pool. Further characterization of AhR heterocomplex from the HeLa nuclear fraction by sucrose density gradient analysis revealed that the AhR was present in the 6 S form, and that the nuclear AhR could be coimmunoprecipitated using anti-Arnt mAb. The ability of the AhR to specifically interact with dioxin-responsive elements (DRE) was demonstrated utilizing wild-type and two mutant DREs in gel shift assays. These results would suggest that, in HeLa cells, the AhR-Arnt heterodimer is associated with the nuclear fraction under normal culture conditions. Therefore, HeLa cells can be used as a model system to study the biochemical and molecular function of the Ah receptor and the process that leads to activation of the AhR in the absence of exogenous ligand. PMID: 8619634 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 307: J Biol Chem. 1996 Apr 12;271(15):8843-50. Functional characterization of DNA-binding domains of the subunits of the heterodimeric aryl hydrocarbon receptor complex imputing novel and canonical basic helix-loop-helix protein-DNA interactions. Bacsi SG, Hankinson O. Department of Pathology and Laboratory Medicine and the Jonsson Comprehensive Cancer Center, Medical School, University of California, Los Angeles California 90095-1781, USA. The aryl hydrocarbon receptor (AHR) and the aryl hydrocarbon receptor nuclear translocator (ARNT) belong to a novel subclass of basic helix-loop-helix transcription factors. The AHR.ARNT heterodimer binds to the xenobiotic responsive element (XRE). Substitution of each of four amino acids in the basic region of ARNT with alanine severely diminishes or abolishes XRE binding, intimating that these amino acids contact DNA bases. Three of these amino acids are conserved among basic helix-loop-helix proteins, and the corresponding amino acids of Max and USF are known to contact DNA bases. Alanine scanning mutagenesis of the basic domain of AHR and substitution with conservative amino acids at particular positions in this domain and in a more amino-proximal AHR segment previously shown to be required for XRE binding (Fukunaga, B. N., and Hankinson, O. (1996) J. Biol. Chem. 271, 3743-3749) demonstrate that the most carboxyl-proximal amino acid position of the basic domain and a position within the amino-proximal segment are intolerant to amino acid substitution with regard to XRE binding, suggesting that these two amino acids make base contacts. Amino acid positions in these AHR regions and in the ARNT basic region less adversely affected by substitution are also identified. The amino acids at these positions may contact the phosphodiester backbone. The apparent bipartite nature of the DNA binding region of AHR and the identity of those of its amino acids that apparently make DNA contacts impute a novel protein-DNA binding behavior for AHR. PMID: 8621524 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 308: J Biol Chem. 1996 Apr 5;271(14):7942-8. DNA binding by the heterodimeric Ah receptor. Relationship to dioxin-induced CYP1A1 transcription in vivo. Dong L, Ma Q, Whitlock JP Jr. Department of Molecular Pharmacology, Stanford University School of Medicine, Stanford, California 94305-5332, USA. The environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin induces the microsomal enzyme cytochrome P4501A1 by increasing the transcription rate of the CYP1A1 gene. Induction requires two basic helix-loop-helix proteins, the ligand-binding aromatic hydrocarbon receptor (AhR) and its heterodimerization partner, the AhR nuclear translocator (Arnt). The AhR/Arnt heterodimer induces transcription by binding to dioxin-responsive elements (DREs) within an enhancer upstream of the CYP1A1 gene. The basic regions of AhR and Arnt are crucial for DRE binding. We have mutated these regions in order to analyze the relationship between DRE binding (determined in vitro using an electrophoretic mobility shift assay) and induction of CYP1A1 transcription (determined in vivo by genetic complementation of AhR-defective and Arnt-defective mouse hepatoma cells, using an RNase protection assay to measure mRNA accumulation). Our findings reveal the amino acids in the basic regions of AhR/Arnt that are important for both DRE binding and induction of transcription. This information provides biological background for the interpretation of structural (e.g. crystallographic) studies of the interactions between AhR/Arnt and the DRE. Our findings also indicate that the in vitro behavior of the mutants does not consistently predict their functional activity in vivo. Thus, genetic complementation constitutes an important and stringent test for analyzing the effects of mutations on AhR/Arnt function. PMID: 8626473 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 309: Mol Cell Biol. 1996 Apr;16(4):1706-13. cDNA cloning and tissue-specific expression of a novel basic helix-loop-helix/PAS factor (Arnt2) with close sequence similarity to the aryl hydrocarbon receptor nuclear translocator (Arnt). Hirose K, Morita M, Ema M, Mimura J, Hamada H, Fujii H, Saijo Y, Gotoh O, Sogawa K, Fujii-Kuriyama Y. Department of Chemistry, Graduate School of Science, Tohoku University Sendai, Japan. We isolated mouse cDNA clones (Arnt2) that are highly similar to but distinct from the aryl hydrocarbon receptor (AhR) nuclear translocator (Arnt). The composite cDNA covered a 2,443-bp sequence consisting of a putative 2,136-bp open reading frame encoding a polypeptide of 712 amino acids. The predicted Arnt2 polypeptide carries a characteristic basic helix-loop-helix (bHLH)/PAS motif in its N-terminal region with close similarity (81% identity) to that of mouse Arnt and has an overall sequence identity of 57% with Arnt. Biochemical properties and interaction of Arnt2 with other bHLH/PAS proteins were investigated by coimmunoprecipitation assays, gel mobility shift assays, and the yeast two-hybrid system. Arnt2 interacted with AhR and mouse Sim as efficiently as Arnt, and the Arnt2-AhR complex recognized and bound specifically the xenobiotic responsive element (XRE) sequence. Expression of Arnt2 successfully rescued XRE-driven reporter gene activity in the Arnt-defective c4 mutant of Hepa-1 cells. RNA blot analysis revealed that expression of Arnt2 mRNA was restricted to the brains and kidneys of adult mice, while Arnt mRNA was expressed ubiquitously. In addition, whole-mount in situ hybridization of 9.5-day mouse embryos showed that Arnt2 mRNA was expressed in the dorsal neural tube and branchial arch 1, while Arnt transcripts were detected broadly in various tissues of mesodermal and endodermal origins. These results suggest that Arnt2 may play different roles from Arnt both in adult mice and in developing embryos. Finally, sequence comparison of the currently known bHLH/PAS proteins indicates a division into two phylogenetic groups: the Arnt group, containing Arnt, Arnt2, and Per, and the AhR group, consisting of AhR, Sim, and Hif-1alpha. PMID: 8657146 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 310: Mol Pharmacol. 1996 Mar;49(3):391-8. The aryl-hydrocarbon receptor, but not the aryl-hydrocarbon receptor nuclear translocator protein, is rapidly depleted in hepatic and nonhepatic culture cells exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Pollenz RS. Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, 29425, USA. Western blot analysis and indirect immunofluorescence microscopy were used to evaluate the fate of the aryl-hydrocarbon receptor (AhR) and aryl-hydrocarbon receptor nuclear translocator (Arnt) protein in culture cell models exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). In wild-type (WT) murine Hepa-1c1c7 cells, AhR protein was depleted by 85% after 4 hr of TCDD treatment as measured in total cell lysates. In contrast, the concentration of Arnt protein was unaffected by TCDD treatment in WT cells. Analysis of the AhR with immunofluorescence microscopy revealed that nuclear translocation of the liganded AhR preceded its depletion from cells. AhR protein was depleted from Hepa-1 type I variants (that contain a concentration of AhR that is 10% of WT) with a similar time course and to the same maximal level observed in WT cells (85%). The EC50 for AhR depletion in Hepa-1 cells was 39 pm TCDD and correspond to the EC50 for induction of P4501A1 protein. Murine embryonic fibroblasts (NIH-3T3), rat aortic smooth muscle cells (A7), and murine skeletal muscle cells (C2C12) all exhibited >90% depletion of the AhR after 2-4 hr of TCDD treatment. Arnt concentration was not affected by TCDD in these cell lines. These results indicate that the liganded AhR is rapidly depleted within the nuclear compartment of hepatic and nonhepatic cells in a manner independent of the Arnt protein. PMID: 8643077 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 311: Carcinogenesis. 1996 Mar;17(3):435-41. Autoregulation of human CYP1A1 gene promotor activity in HepG2 and MCF-7 cells. Jorgensen EC, Autrup H. Department of Environmental and Occupational Medicine, University of Aarhus, Denmark. Cytochrome CYP1A1 gene expression, induced by polycyclic aromatic hydrocarbons and dioxins, eg. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is regulated mainly at the level of transcription. Inducible activation of the CYP1A1 promotor is mediated by a ligand-dependent transcription factor dimer complex including the aryl hydrocarbon receptor (AHR) and the AHR nuclear translocator (ARNT) proteins. Additional factors seem to be involved in tissue- and cell-specific modification of the induction process. In the present study HepG2 and MCF-7 cell lines were used to examine a possible cell-specific autoregulation of CYP1A1 promotor function. Chimeric CYP1A1-CAT reporter constructs and a human CYP1A1 cDNA expression plasmid were used in transient co-expression experiments. In HepG2 cells co-expression of increasing amounts of CYP1A1 cDNA significantly down-regulated constitutive as well as the TCDD-induced CYP1A1 promotor driven CAT activity. In contrast, co-transfection of MCF-7 cells with a 3-fold molar excess of CYP1A1 cDNA relative to the CYP1A1-CAT reporter construct caused an approximately 2-fold increase in the TCDD-induced CAT activity, whereas no effect was observed on constitutive promotor activity. This autoregulatory mechanism(s) of the human CYP1A1 gene product was independent of specific 5' flanking promotor segments tested. RT-PCR analyses did not indicate any changes in mRNA level of AHR and ARNT in the co-transfection studies. Thus these studies show that the human CYP1A1 gene is exposed to cell-specific autoregulation, probably achieved via different functions of trans-acting factors. PMID: 8631128 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 312: J Biol Chem. 1996 Feb 16;271(7):3743-9. Identification of a novel domain in the aryl hydrocarbon receptor required for DNA binding. Fukunaga BN, Hankinson O. Department of Pathology and Laboratory Medicine, Medical School, University of California, Los Angeles, 90095, USA. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that binds DNA in the form of a heterodimer with the AHR nuclear translocator protein (ARNT). Both proteins possess basic helix-loop-helix motifs. ARNT binds to the side of the xenobiotic responsive element (XRE) that resembles an E-box (the sequence recognized by the majority of other basic helix-loop-helix proteins), whereas AHR binds to the side of the XRE that does not conform to the E-box sequence. The basic region of ARNT closely resembles those of other E-box-binding proteins, whereas the "nominal basic region" of AHR (amino acids 27 39), although required for XRE binding, deviates from this consensus. By extensive mutational analysis it is shown here that an additional block of amino acids of AHR (from tyrosine 9 to lysine 20) that contains a highly basic segment is required for XRE binding and transcriptional activation. Deletion of the first nine amino acids negates XRE binding. Substitution of either tyrosine 9 or arginine 14 with alanine eliminates XRE binding, whereas alanine substitutions at certain other sites within the block reduce but do not eliminate binding. The reported absence of the first nine amino acids in the purified protein may therefore be artifactual. These results suggest that the amino acids of AHR involved in binding to the XRE constitute a novel DNA-binding domain, comprising amino acids located within and amino-terminal to the nominal basic region. PMID: 8631989 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 313: Br J Cancer. 1996 Feb;73(3):316-22. Effect of transient expression of the oestrogen receptor on constitutive and inducible CYP1A1 in Hs578T human breast cancer cells. Wang WL, Thomsen JS, Porter W, Moore M, Safe S. Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466, USA. Hs578T human breast cancer cells are an oestrogen receptor (ER)-negative cell line. Treatment of these cells with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) resulted in formation of a 6.9 S nuclear aryl hydrocarbon (Ah) receptor complex, which bound to a [32P]dioxin-responsive element in a gel electrophoretic mobility shift assay. However, TCDD does not induce CYP1A1 gene expression or chloramphenicol acetyl transferase (CAT) activity in cells transiently transfected with pRNH11c or pMCAT5.12, which are Ah-responsive plasmids derived from the 5'-flanking region of the human and murine CYP1A1 genes respectively. Restoration of Ah responsiveness was investigated by co-transfecting Hs578T cells with pRNH11c or pMCAT5.12 and plasmids that express the ER (hER), Ah receptor (AhR) and AhR nuclear translocator (Arnt) proteins. ER expression resulted in significantly increased basal CAT activity; however, TCDD did not induce CAT activity in the transiently transfected cells. Expression of the AhR or Arnt proteins did not alter basal or inducible CAT activity. Expression of N- or C-terminal truncated ER in Hs578T resulted in differential regulation of Ah responsiveness. In Hs578T cells transiently expressing the ER, which contains C-terminal deletions (amino acids 282-595), basal CAT activity was also increased; however, Ah responsiveness was not restored. In contrast, transient expression of N-terminal-deleted (amino acids 1-178) ER resulted in a marked decrease in basal CAT activity but a restoration of Ah responsiveness. These results suggest that basal and inducible CAT activity in Hs578T cells transiently transfected with pRNH11c is modulated differentially by ER domains that are present in the N- and C-terminal regions of the ER. PMID: 8562336 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 314: Biochem Biophys Res Commun. 1996 Jan 17;218(2):588-94. cDNA cloning of a murine homologue of Drosophila single-minded, its mRNA expression in mouse development, and chromosome localization. Ema M, Suzuki M, Morita M, Hirose K, Sogawa K, Matsuda Y, Gotoh O, Saijoh Y, Fujii H, Hamada H, Fujii-Kuriyama Y. Department of Chemistry, Faculty of Science, Tohoku University, Sendai, Japan. A combination of the RT-PCR method and subsequent screening of the cDNA library of mouse skeletal muscle with the cDNA isolated by RT-PCR used as a probe led to isolation of cDNAs encoding a polypeptide (mSim) with bHLH and PAS domains which show high similarity to the corresponding regions of Drosophila Sim, a master regulator in neurogenesis. Experiments using a GST-fusion protein demonstrated that mSim heterodimerizes with Arnt (Ah receptor nuclear translocator), even more efficiently than AhR (Ah receptor) does with Arnt. RNA blot analysis using RNAs from various tissues of mice indicated that mSim transcript is expressed in several limited tissues such as muscle, kidney and lung of adult animals. Distribution of mSim mRNA was always accompanied with that of Arnt. All the results suggest a regulatory role of mSim in partnership with Arnt. Chromosomal location of the mSim gene was determined by fluorescent in situ hybridization to be localized on the C3.3-C4 band of mouse chromosome 16 which is syntenic with the human chromosome 21q22 carrying the Down syndrome critical region, where a gene highly homologous to the Drosophila sim was localized. Whole mount in situ hybridization using a unique part of mSim cDNA sequence showed that mSim mRNA was expressed in the ventral diencephalon, branchial arches and limbs. These findings will provide an approach to the cause of the Down syndrome as well as the elucidation of the functional roles of mSim in animal development. PMID: 8561800 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 315: Mol Cell Biol. 1996 Jan;16(1):430-6. Dioxin-induced CYP1A1 transcription in vivo: the aromatic hydrocarbon receptor mediates transactivation, enhancer-promoter communication, and changes in chromatin structure. Ko HP, Okino ST, Ma Q, Whitlock JP Jr. Department of Molecular Pharmacology, Stanford University School of Medicine, California 94305-5332, USA. We have analyzed the dioxin-inducible transcriptional control mechanism for the mouse CYP1A1 gene in its native chromosomal context. Our genetic and biochemical studies indicate that a C-terminal segment of the aromatic hydrocarbon receptor (AhR) contains latent transactivation capability and communicates the induction signal from enhancer to promoter. Thus, transactivation and enhancer-promoter communication may be congruent functions of AhR. Both functions require heterodimerization between AhR and the AhR nuclear translocator (Arnt). Our findings also indicate that heterodimerization activates AhR's latent transactivation function and silences that of Arnt. Furthermore, removal of Arnt's transactivation domain does not affect dioxin-induced CYP1A1 transcription in vivo. In addition, our studies demonstrate that dioxin-induced changes in chromatin structure occur by different mechanisms at the CYP1A1 enhancer and promoter and that events at an enhancer can be experimentally dissociated from events at the cognate promoter during mechanistic analyses of mammalian transcription in vivo. PMID: 8524325 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 316: Neuroreport. 1995 Dec 15;6(18):2557-60. The dioxin receptor and its nuclear translocator (Arnt) in the rat brain. Kainu T, Gustafsson JA, Pelto-Huikko M. Department of Anatomy, Tampere University Medical School, Finland. Dioxins are environmental pollutants, whose detrimental effects on health are the cause of wide public concern due to their accumulation in the food chain and resistance to metabolism. The most well known dioxin is 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Dioxins exert their effects through a ligand activated transcription factor termed the dioxin or aryl hydrocarbon receptor (Ahr), which acts in concert with another structurally related protein: the aryl hydrocarbon nuclear translocator (Arnt). In the present study, we have employed in situ hybridization to study the localization of the mRNAs for these two proteins in the rat brain. We found mRNAs for both Ahr and Arnt predominantly in the same neuronal populations: in the olfactory bulb, the hippocampus, and the cerebral and cerebellar cortices. Arnt, however, had a more widespread expression than Ahr in the brain. The present results demonstrate that dioxins may act directly in the brain and that the effects of dioxin may occur in discrete neuronal populations. However, in some parts of the brain, e.g. the hypothalamus, that are thought to be targets of the toxic effects of dioxins, we did not observe detectable levels of Ahr mRNA. Furthermore, it appears that Arnt may have additional functions in the brain, apart from being the heterodimerization partner of Ahr, possibly through heterodimerizing with other transcription factors. PMID: 8741762 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 317: J Biol Chem. 1995 Dec 8;270(49):29270-8. Identification of functional domains of the aryl hydrocarbon receptor. Fukunaga BN, Probst MR, Reisz-Porszasz S, Hankinson O. Department of Pathology and Laboratory Medicine, University of California, Los Angeles 90095-1786, USA. Functional domains of the mouse aryl hydrocarbon receptor (Ahr) were investigated by deletion analysis. Ligand binding was localized to a region encompassing the PAS B repeat. The ligand-mediated dissociation of Ahr from the 90-kDa heat shock protein (HSP90) does not require the aryl hydrocarbon receptor nuclear translocator (Arnt), but it is slightly enhanced by this protein. One HSP90 molecule appears to bind within the PAS region. The other molecule of HSP90 appears to require interaction at two sites: one over the basic helix-loop-helix region, and the other located within the PAS region. Each mutant was analyzed for dimerization with full-length mouse Arnt and subsequent binding of the dimer to the xenobiotic responsive element (XRE). In order to minimize any artificial steric hindrances to dimerization and XRE binding, each Ahr mutant was also tested with an equivalently deleted Arnt mutant. The basic region of Ahr is required for XRE binding but not for dimerization. Both the first and second helices of the basic helix-loop-helix motif and the PAS region are required for dimerization. These last results are analogous to those previously obtained for Arnt (Reisz-Porszasz, S., Probst, M.R., Fukunaga, B. N., and Hankinson, O. (1994) Mol. Cell. Biol. 14, 6075-6086) compatible with the notion that equivalent regions of Ahr and Arnt associate with each other. Deletion of the carboxyl-terminal half of Ahr does not affect dimerization or XRE binding but, in contrast to an equivalent deletion of Arnt, eliminates biological activity as assessed by an in vivo transcriptional activation assay, suggesting that this region of Ahr plays a more prominent role in transcriptional activation of the cyp1a1 gene than the corresponding region of Arnt. PMID: 7493958 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 318: J Biol Chem. 1995 Nov 3;270(44):26292-302. DNA binding specificities and pairing rules of the Ah receptor, ARNT, and SIM proteins. Swanson HI, Chan WK, Bradfield CA. Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, Chicago, Illinois 60611, USA. The Ah receptor (AHR), the Ah receptor nuclear translocator protein (ARNT), and single-minded protein (SIM) are members of the basic helix-loop-helix-PAS (bHLH-PAS) family of regulatory proteins. In this study, we examine the DNA half-site recognition and pairing rules for these proteins using oligonucleotide selection-amplification and coprecipitation protocols. Oligonucleotide selection-amplification revealed that a variety of bHLH-PAS protein combinations could interact, with each generating a unique DNA binding specificity. To validate the selection-amplification protocol, we demonstrated the preference of the AHR.ARNT complex for the sequence commonly found in dioxin-responsive enhancers in vivo (TNGCGTG). We then demonstrated that the ARNT protein is capable of forming a homodimer with a binding preference for the palindromic E-box sequence, CACGTG. Further examination indicated that ARNT may have a relaxed partner specificity, since it was also capable of forming a heterodimer with SIM and recognizing the sequence GT(G/A)CGTG. Coprecipitation experiments using various PAS proteins and ARNT were consistent with the idea that the ARNT protein has a broad range of interactions among the bHLH-PAS proteins, while the other members appear more restricted in their interactions. Comparison of this in vitro data with sites known to be bound in vivo suggests that the high affinity half-site recognition sequences for the AHR, SIM, and ARNT are T(C/T)GC, GT(G/A)C (5'-half-sites), and GTG (3'-half-sites), respectively. PMID: 7592839 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 319: Biochem Pharmacol. 1995 Oct 12;50(8):1295-302. Functional analysis of aryl hydrocarbon receptor nuclear translocator interactions with aryl hydrocarbon receptor in the yeast two-hybrid system. Yamaguchi Y, Kuo MT. Department of Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston 77030, USA. The aryl hydrocarbon receptor (AHR) mediates dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin)-induced transcriptional activation of a battery of genes by interaction with a cofactor, called aryl hydrocarbon receptor nuclear translocator (ARNT) protein. Both AHR and ARNT belong to a family of proteins that includes the Drosophila circadian-rhythm protein and "single-minded" protein. These proteins share a domain called the PAS domain. In addition to the PAS domain, both AHR and ARNT contain basic helix-loop-helix (bHLH) and glutamine (Q)-rich domains. The roles of these domains in the receptor-mediated transcriptional activation are not understood completely. By using the yeast two-hybrid system with the N-terminal half of AHR as a probe, which contains the bHLH and PAS regions, to screen cDNA libraries prepared from human lymphocytes and C57BL mouse liver for clones encoding proteins capable of binding to these regions, we isolated a partial ARNT cDNA clone. These results demonstrated that the N-terminal half of AHR is capable of interacting with ARNT in yeast (probably through the bHLH motif). A fusion protein containing the GAL4 DNA binding domain (DB) linked to the full-length AHR was not capable of activating expression of a reporter gene containing the GAL4 DNA binding site, suggesting that ligand-free AHR alone has no transactivating properties in yeast. However, the C-terminal portion (amino acid residues 580-797) of the AHR, including the Q-rich domain, could confer transactivation of the reporter gene expression in the same system, suggesting that the N-terminal portion of the AHR contains transcription repression properties. In contrast, GAL4(DB)-ARNT fusion protein was able to activate expression of the same reporter gene. Deletion analysis of ARNT revealed that the C-terminal 75 amino acids, including the Q-rich domain, exhibited full transactivation function in yeast and mammalian cells. These results revealed different structural organizations for the transactivation properties between AHR and ARNT, although both contained transactivation domains at the C-termini. PMID: 7488247 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 320: Dev Dyn. 1995 Oct;204(2):144-55. Developmental expression of two members of a new class of transcription factors: II. Expression of aryl hydrocarbon receptor nuclear translocator in the C57BL/6N mouse embryo. Abbott BD, Probst MR. Developmental Toxicology Division, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA. The aryl hydrocarbon receptor (AhR) and the AhR nuclear translocator protein (ARNT) are basic-helix-loop-helix (bHLH) proteins involved in transcriptional regulation. The AhR is a ligand-activated partner of the ARNT protein. Both proteins are required to transcriptionally regulate gene expression. ARNT must be complexed to AhR to permit binding to the regulatory DNA sequence. The AhR-ligand complex is known to mediate a range of biological responses, such as developmental toxicity, induction of cleft palate, and hydronephrosis. AhR and ARNT are expressed in human embryonic palatal cells and AhR was recently shown to have a specific developmental pattern of expression in the mouse embryo. In the present study, expression of ARNT is characterized in C57Bl/6N mouse embryos from gestation day (GD) 10-16 using immunohistochemistry and in situ hybridization. Af affinity purified antibody against human ARNT (1.1 micrograms/ml) was detected with an avidin-biotin-peroxidase complex. ARNT mRNA was localized with a 35S-RNA probe from pBM5/NEO-M1-1. Specific spatial and temporal patterns of ARNT expression emerged and mRNA and protein expression correlated. The GD 10-11 embryos showed highest levels of ARNT in neuroepithelial cells of the neural tube, visceral arches, otic and optic placodes, and preganglionic complexes. The heart also had significant expression of ARNT with strong nuclear localization. After GD11, expression in heart and brain declined. In GD 12-13 embryos expression was highest in the liver where expression increased from GD 12-16. At GD 15-16 the highest levels of ARNT occurred in adrenal gland and liver, although ARNT was also detected in submandibular gland, ectoderm, tongue, bone, and muscle. In all of these tissues ARNT was cytoplasmic as well as nuclear, except in some of the cortical adrenal cells in which ARNT was strongly cytoplasmic with little or no nuclear localization. These specific patterns of ARNT expression, which differ in certain tissues from the expression of AhR, suggest that ARNT may have additional roles in normal embryonic development. PMID: 8589438 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 321: Biochem Biophys Res Commun. 1995 Jul 6;212(1):144-50. Identification of a 120-kDa protein associated with aromatic hydrocarbon receptor nuclear translocator. Hossain A, Kikuchi H, Ikawa S, Sagami I, Watanabe M. Department of Molecular Genetics, Tohoku University, Sendai, Japan. The aromatic hydrocarbon receptor nuclear translocator (ARNT) is a basic helix-loop-helix-PAS protein which forms a heterodimer with aromatic hydrocarbon receptor (AHR), this heterodimer mediating the signal transduction in response to the various xenobiotics such as 2,3,7,8-tetrachlorodibenzo-p-dioxin and directly interacting with target genes by binding to xenobiotic responsive elements. An anti-ARNT antibody was raised in rabbits against the bacterially expressed ARNT of amino acids 21-328 from the N-terminal. Using this antibody, besides ARNT itself, we detected at least one protein, 120 kDa, in the immunoprecipitate of anti-ARNT antibodies in HepG2 cells as well as in Hepa-1 cells. However, this protein is not present in the immunoprecipitate of the anti-AHR antisera nor in that of the preimmune sera of the rabbits used for the immunization. PMID: 7611998 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 322: Mol Cell Biol. 1995 Jul;15(7):3714-21. Dioxin induces localized, graded changes in chromatin structure: implications for Cyp1A1 gene transcription. Okino ST, Whitlock JP Jr. Department of Molecular Pharmacology, Stanford University School of Medicine, California 94305-5332, USA. In mouse hepatoma cells, the environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, or dioxin) induces Cyp1A1 gene transcription, a process that requires two basic helix-loop-helix regulatory proteins, the aromatic hydrocarbon receptor (AhR) and the aromatic hydrocarbon receptor nuclear translocator (Arnt). We have used a ligation-mediated PCR technique to analyze dioxin-induced changes in protein-DNA interactions and chromatin structure of the Cyp1A1 enhancer-promoter in its native chromosomal setting. Dioxin-induced binding of the AhR/Arnt heteromer to enhancer chromatin is associated with a localized (about 200 bp) alteration in chromatin structure that is manifested by increased accessibility of the DNA; these changes probably reflect direct disruption of a nucleosome by AhR/Arnt. Dioxin induces analogous AhR/Arnt-dependent changes in chromatin structure and accessibility at the Cyp1A1 promoter. However, the changes at the promoter must occur by a different, more indirect mechanism, because they are induced from a distance and do not reflect a local effect of AhR/Arnt binding. Dose-response experiments indicate that the changes in chromatin structure at the enhancer and promoter are graded and mirror the graded induction of Cyp1A1 transcription by dioxin. We discuss these results in terms of a TCDD-induced shift in an equilibrium between nucleosomal and nonnucleosomal chromatin configurations. PMID: 7791778 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 323: Proc Natl Acad Sci U S A. 1995 Jun 6;92(12):5510-4. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Wang GL, Jiang BH, Rue EA, Semenza GL. Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287-3914, USA. Hypoxia-inducible factor 1 (HIF-1) is found in mammalian cells cultured under reduced O2 tension and is necessary for transcriptional activation mediated by the erythropoietin gene enhancer in hypoxic cells. We show that both HIF-1 subunits are basic-helix-loop-helix proteins containing a PAS domain, defined by its presence in the Drosophila Per and Sim proteins and in the mammalian ARNT and AHR proteins. HIF-1 alpha is most closely related to Sim. HIF-1 beta is a series of ARNT gene products, which can thus heterodimerize with either HIF-1 alpha or AHR. HIF-1 alpha and HIF-1 beta (ARNT) RNA and protein levels were induced in cells exposed to 1% O2 and decayed rapidly upon return of the cells to 20% O2, consistent with the role of HIF-1 as a mediator of transcriptional responses to hypoxia. PMID: 7539918 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 324: Carcinogenesis. 1995 Jun;16(6):1403-9. Expression of Ah receptor (TCDD receptor) during human monocytic differentiation. Hayashi S, Okabe-Kado J, Honma Y, Kawajiri K. Department of Biochemistry, Saitama Cancer Center Research Institute, Japan. We have previously found a high expression of human Ah receptor (TCDD receptor) mRNA in peripheral blood cells of individuals. In this paper, the expression of this gene in blood cells was first investigated in fractions of nucleated cells, revealing predominant expression of the Ah receptor gene in the monocyte fraction. Then the expression levels of AhR mRNA in various hematopoietic cell lines were examined together with those of Arnt and P450IA1. AhR was expressed at high levels in monocytoid U937, THP1, and HEL/S cells, and at moderate levels in promyelocytic HL60 cells and erythroblastic HEL cells. However, it was not detected in lymphoid cells MOLT4 (T cell) and BALL1 (B cell), nor in K562 erythroblasts. Furthermore, a specific induction of AhR during monocytic differentiation was investigated in HL60 and HEL cells. HL60 cells were induced to differentiate toward monocytes-macrophages by incubation with phorbol ester, showing a 5- to 2-fold increase of AhR mRNA. The incubation with transforming growth factor beta 1 and 1 alpha,25-dihydroxyvitamin D3 resulted in a 5- to 7-fold increase of AhR mRNA. The HEL cells also exhibited a similar elevation of AhR mRNA level, when they had differentiated toward monocyte-macrophage cells by these combined inducers, but little change in the mRNA level was observed when the cells were induced to differentiate into other cell types. Treatment of the differentiated HL60 cells with 3-methylcholanthrene, a ligand of AhR, induced the expression of the P450IA1 gene. These results indicated that expression of AhR mRNA was significantly induced during monocytic differentiation and that the differentiated cells were responsive to xenobiotics. Our results suggest that AhR may play an important role in the function of monocytes and also in the eventual activation of environmental carcinogens. PMID: 7788861 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 325: J Biol Chem. 1995 May 26;270(21):12697-703. Transcriptional activation by the mouse Ah receptor. Interplay between multiple stimulatory and inhibitory functions. Ma Q, Dong L, Whitlock JP Jr. Department of Molecular Pharmacology, Stanford University School of Medicine, California 94305-5332, USA. The aromatic hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that mediates cellular responses to the environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). We cloned AhR cDNA from C57BL/6 mouse liver and verified by transfection that it encodes a functional protein. Analyses of deletion mutants indicate that the carboxyl half of AhR contains several types of transactivation domain, which function independently of domains that mediate TCDD recognition, DNA binding, and heterodimerization with the Ah receptor nuclear translocator (Arnt) protein. The transactivation domains function independently of each other, display different levels of activity, and act synergistically when linked. In addition, AhR contains an 82-amino acid domain that inhibits transactivation. The inhibitory domain displays specificity, in that it blocks the transactivating functions of AhR and Arnt, but not that of the herpes simplex protein VP16. The inhibitory activity depends upon the cell type in which AhR is expressed, implying that a cell-specific protein mediates the effect. PMID: 7759522 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 326: Arch Biochem Biophys. 1995 Apr 1;318(1):166-74. Ah receptor phosphorylation: localization of phosphorylation sites to the C-terminal half of the protein. Mahon MJ, Gasiewicz TA. Department of Environmental Medicine, University of Rochester School of Medicine, New York 14642, USA. The aryl hydrocarbon receptor (AhR) is a transcriptional enhancer activated by the binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related xenobiotics. Ligand binding initiates a series of poorly understood molecular events which confers recognition of cis-acting elements located in regulatory regions of particular structural genes, such as CYP1A1. Several studies have suggested that AhR phosphorylation may be instrumental in activating the AhR to a DNA-binding state. In agreement with previous investigations, treatment of the AhR with acid phosphatase resulted in the loss of DNA-binding activity. To further evaluate the functional role of AhR phosphorylation we determined whether TCDD binding altered total AhR phosphorylation, and identified phosphorylated regions by the examination of chemical cleavage patterns. The AhR was isolated by immunoprecipitation from [32P]-orthophosphate-labeled Hepa 1 cells grown in the presence or absence of TCDD. Examination of the amount of 32P associated with the AhR indicated that the total level of AhR phosphorylation was not affected by ligand binding. Chemical cleavage with hydroxylamine and cyanogen bromide also revealed a similar pattern for liganded and unliganded AhR. The shortest regions of overlap determined by the chemical cleavage patterns localized phosphorylation sites to two regions in the C-terminal half of the AhR. One region is centrally located between amino acids 368 and 605 and within or adjacent to a DNA binding repressor domain. The other region is located at the glutamine-rich carboxyl terminus between amino acids 636 and 759. These data coupled with previous observations imply that total AhR phosphorylation is not altered by the ligand-elicited transformation to a DNA-binding form, but that phosphorylation nevertheless plays an important role in the ability of an active AhR-Arnt complex to associate with cis-acting regulatory elements. PMID: 7726558 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 327: Proc Natl Acad Sci U S A. 1995 Mar 14;92(6):1936-40. Possible function of Ah receptor nuclear translocator (Arnt) homodimer in transcriptional regulation. Sogawa K, Nakano R, Kobayashi A, Kikuchi Y, Ohe N, Matsushita N, Fujii-Kuriyama Y. Department of Chemistry, Faculty of Science, Tohoku University, Sendai, Japan. Arnt (Ah receptor nuclear translocator) is a member of a transcription factor family having characteristic motifs designated bHLH (basic helix-loop-helix) and PAS and was originally found as a factor forming a complex with Ah receptor (AhR) to bind the specific xenobiotic responsive element (XRE) sequence for induction of drug-metabolizing P4501A1. We have examined interaction of Arnt with other PAS proteins--Drosophila Per, Sim, and AhR--by the coimmunoprecipitation method. Arnt formed a homodimer with itself as well as heterodimers with the others by means of the PAS and HLH domains in a cooperative way. The Arnt homodimer binds the sequence of adenovirus major late promoter (MLP) with the E box core sequence CACGTG, suggesting that the CAC half of the XRE, CACGCN(A/T), recognized by the AhR-Arnt heterodimer is a target for Arnt. Cotransfection experiments using CV-1 cells with an Arnt expression plasmid and a MLP chloramphenicol acetyltransferase (CAT) reporter plasmid revealed that Arnt markedly activated CAT expression, indicative of a newly discovered regulatory role of Arnt. PMID: 7892203 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 328: Mol Pharmacol. 1995 Mar;47(3):432-8. Orientation of the heterodimeric aryl hydrocarbon (dioxin) receptor complex on its asymmetric DNA recognition sequence. Bacsi SG, Reisz-Porszasz S, Hankinson O. Laboratory of Structural Biology and Molecular Medicine, School of Medicine, University of California, Los Angeles 90024. The 2,3,7,8-tetrachlorodibenzo-p-dioxin-transformed aryl hydrocarbon receptor (AHR) complex binds to xenobiotic-responsive element (XRE) sequences in the 5' flanking region of the CYP1A1 gene, resulting in initiation of transcription. Both components of the transformed AHR complex [the ligand-binding AHR monomer and the AHR nuclear translocator (ARNT)] directly contact the XRE. These proteins belong to a novel subclass of basic helix-loop-helix transcription factors. The binding sites of AHR and ARNT on the asymmetric XRE were determined using nuclear extracts of 2,3,7,8-tetrachlorodibenzo-p-dioxin-treated Hepa-1c1c7 cells and a panel of double-stranded oligonucleotides containing XRE1 of the CYP1A1 gene (5'-TTGCGTGAGAA-3'), in which all combinations of three, two, or one of the thymines indicated were substituted by the photoreactive thymine analog 5-bromodeoxyuracil. Covalent cross-linking analysis and immunoprecipitation with antibodies specific for AHR or ARNT demonstrated that ARNT directly contacts the 3'-most thymine position, that AHR directly contacts the second thymine position, and that neither protein contacts the 5'-most thymine position. The thymine position contacted by ARNT lies within a three-nucleotide sequence (5'-GTG-3') identical to a half-site of an E-box element (5'-CACGTG-3') that is recognized by a number of other basic helix-loop-helix transcription factors. AHR binds to a portion of the XRE that does not resemble an E-box. Additional experiments demonstrated that neither protein loops over to contact residues located beyond the other's binding site. PMID: 7700240 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 329: Annu Rev Pharmacol Toxicol. 1995;35:307-40. The aryl hydrocarbon receptor complex. Hankinson O. Department of Pathology, University of California, Los Angeles 90024, USA. The heteromeric unliganded aryl hydrocarbon receptor complex (AHRC) contains the aryl hydrocarbon receptor monomer (AHR). Binding of polycyclic or halogenated aromatic hydrocarbon (PAH and HAH) ligand causes release of AHR, which then associates with the AHR nuclear translocator protein (ARNT) to generate the heterodimeric "transformed" AHRC. AHR and ARNT belong to a novel subclass of basic helix-loop-helix-containing transcription factors. The transformed AHRC binds xenobiotic responsive elements in responsive genes and turns on their transcription. Certain of these genes encode enzymes involved in the metabolic activation of PAHs to mutagenic derivatives. HAHs are not genotoxic: Their pathogenicity depends on the AHRC but not on their metabolism. Current research includes investigations directed towards delineating the pathways of HAH pathogenesis, ascertaining whether AHR can mediate signal transduction independently of DNA binding, understanding the mechanism of transcriptional activation, and investigating the potential roles of AHR and ARNT in development. Publication Types: Review PMID: 7598497 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 330: Pharmacogenetics. 1995;5 Spec No:S70-3. Genetic polymorphisms of drug-metabolizing enzymes and lung cancer susceptibility. Kawajiri K, Watanabe J, Eguchi H, Hayashi S. Department of Biochemistry, Saitama Cancer Center Research Institute, Japan. A close association of smoking-associated lung cancer incidence with the Msp 1 and 1le-Val polymorphisms of CYP1A1 gene was found in a Japanese population in terms of genotype frequency comparison and cigarette dose response. A synergistic increase in susceptibility to lung cancer was observed when the susceptible genotypes of CYP1A1 were combined with a deficient GSTM1 genotype. Individual difference in expression levels of Ahr and Arnt mRNAs was observed, and the expression levels of CYP1A1 appeared to associate with those of transcriptional factors. The Ahr protein has two different structures, ascribed to one amino acid replacement at codon 554 of Arg by Lys. However, this germ line polymorphism did not show a significant association with AHH inducibility nor lung cancer incidence. The p53 gene alterations in lung cancer tissues were more frequently observed among the patients with a susceptible allele of CYP1A1 gene. Publication Types: Review Review, Tutorial PMID: 7581493 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 331: J Cancer Res Clin Oncol. 1995;121(9-10):612-20. Transcriptional activation domains of the Ah receptor and Ah receptor nuclear translocator. Sogawa K, Iwabuchi K, Abe H, Fujii-Kuriyama Y. Department of Chemistry, Faculty of Science, Tohoku University, Sendai, Japan. The Ah receptor (AhR) and Ah receptor nuclear translocator (Arnt) heterodimer bind the xenobiotic-responsive element (XRE) sequence in the upstream region of the genes for some drug-metabolizing enzymes, such as P4501A1 and glutathione S-transferase Ya, to activate their transcription. This paper describes transcriptional activation domains of the AhR and Arnt as examined in vivo by DNA transfection experiments using GAL4-AhR or GAL4-Arnt chimeric plasmids and a reporter plasmid containing five GAL4 DNA binding sites. The major activation domain of Arnt was localized in a short segment of the C-terminal 34 amino acids, while the glutamine-rich domain of Arnt showed no transcriptional activity. This activation domain of Arnt could be further divided into two subdomains with some sequence similarity. Point mutation analysis of one of the subdomains revealed that bulky hydrophobic amino acids and neighboring acidic amino acids were necessary for the transcription-enhancing activity of Arnt. The C-terminal half of the AhR showed a strong transcription-stimulating activity, apparently five times as strong as that of Arnt. Further analysis of the activity revealed that the C-terminal transcriptional activity was distributed in several activation domains, one of which is rich in glutamine residues. These results indicate that the glutamine-rich domains of the AhR and Arnt function differently in the heterodimer regulatory complex. Previously, we showed that the enhancer activity of XRE was repressed by E1A proteins, especially the 12S form of E1A. Cotransfection experiments using an E1A12S expression plasmid and a GAL4-AhR or GAL4-Arnt expression plasmid demonstrated that E1A protein rather predominantly inhibited the transcriptional activity of Arnt. PMID: 7559746 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 332: J Biol Chem. 1994 Dec 16;269(50):31518-24. Potent transactivation domains of the Ah receptor and the Ah receptor nuclear translocator map to their carboxyl termini. Jain S, Dolwick KM, Schmidt JV, Bradfield CA. Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, Chicago, Illinois 60611. The Ah receptor (AHR) is a ligand-activated transcription factor that is structurally related to its dimerization partner, the Ah receptor nuclear translocator (ARNT), and two Drosophila proteins, SIM and PER. All four proteins contain a region of homology now referred to as a PAS homology domain. In addition, the AHR, ARNT, and SIM harbor a basic region helix-loop-helix motif in their N termini, whereas PER does not. Previous mapping studies of the AHR have demonstrated that the PAS domain contains sequences required for ligand recognition, dimerization, and interaction with the 90-kDa heat shock protein. They also have confirmed that the basic region helix-loop-helix domain plays a role in both dimerization and sequence-specific DNA binding. To identify domains involved in transactivation of target genes, we generated chimeras of AHR/ARNT deletion mutants with the DNA binding region of the yeast Gal4 protein, transiently expressed these in COS-1 cells, and monitored their capacity to activate the chloramphenicol acetyltransferase reporter gene under the control of a minimal promoter driven by enhancer elements recognized by Gal4. Extensive analysis of these fusions revealed that the AHR and ARNT harbor potent transactivation domains within their C termini. Importantly, the amino-terminal halves of both the AHR and ARNT were found to be devoid of transactivation activity. PMID: 7989319 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 333: J Biol Chem. 1994 Dec 2;269(48):30109-12. The 90-kDa heat shock protein is essential for Ah receptor signaling in a yeast expression system. Carver LA, Jackiw V, Bradfield CA. Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, Chicago, Illinois 60611. In an effort to provide a more powerful system to study the Ah receptor (AHR) signaling pathway, we expressed the AHR, its dimerization partner ARNT, and a beta-galactosidase (lacZ) reporter gene, driven by two dioxin-responsive enhancers, in the yeast Saccharomyces cerevisiae. In this system, the agonists beta-naphthoflavone and alpha-naphthoflavone induced transcription of the lacZ gene, with EC50 values of 7.9 x 10(-8) and 3.0 x 10(-7) M, respectively, while the nonagonist dexamethasone was without effect. As a first application of this system, we examined the relationship between the 90-kDa heat shock protein (hsp90) and AHR function. To accomplish this in a manner that was independent of the ARNT protein, we constructed a chimeric receptor in which the DNA binding and primary dimerization domains of the AHR were swapped with analogous domains from the LexA protein. Coexpression of this AHR-LexA chimera and a lacZ reporter gene driven by eight LexA operator sites in a yeast strain with regulatable levels of hsp90, yielded pharmacology that closely mirrored that of the AHR/ARNT/dioxin-responsive enhancer system described above, but only when hsp90 levels were held near their wild type levels. When hsp90 levels were reduced to approximately 5% of normal, AHR signaling in response to agonist was completely blocked despite normal cell growth. These results provide the first genetic evidence for the role of hsp90 in AHR signaling and provide the basis for a powerful new system in which to study this pathway. PMID: 7982913 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 334: Biochem Pharmacol. 1994 Nov 1;48(9):1767-78. Interaction of the regulatory domains of the murine Cyp1a1 gene with two DNA-binding proteins in addition to the Ah receptor and the Ah receptor nuclear translocator (ARNT). Carrier F, Chang CY, Duh JL, Nebert DW, Puga A. Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892. The aromatic hydrocarbon (Ah) receptor complex is a ligand-activated transcriptional activator consisting of at least two protein components. The ligand-binding component is the AhR protein, a cytosolic receptor encoded by the Ahr gene, which, upon ligand binding, translocates to the nucleus in a heterodimeric complex with the ARNT (Ah receptor nuclear translocator) component. The complex binds to several discrete DNA domains containing aromatic hydrocarbon responsive elements (AhRE) present in the regulatory region of the murine cytochrome P(1)450 Cyp1a1 gene and of the other genes in the [Ah] gene battery. As a consequence of binding, a transcriptional complex is formed that activates the expression of these genes by as yet unidentified mechanisms. We have analyzed DNA-protein interactions in four of these domains, specifically, the AhREs located between -1085 and -482 (sites A, C, E, and D) of the upstream regulatory region of the murine Cyp1a1 gene. We found that two DNA-binding proteins, present in cytosolic and nuclear extracts of mouse Hepa-1 cells, showed overlapping DNA-binding specificities to those of the Ah receptor. One of these proteins had an apparent molecular mass of 35-40 kDa, bound only to AhRE3 (site D), and has been identified tentatively as a member of the C/EBP family of transcription factors. The second protein, purified by DNA-affinity chromatography, had an apparent molecular mass of 95 kDa and bound to a larger DNA motif that included the AhRE sequence, in AhRE3 and AhRE5 (sites D and A), but not in AhRE1 or AhRE2 (sites C and E). This protein was not AhR nor was it ARNT, since it was found in receptorless (Ahr-) and in nuclear translocation-defective (Arnt-) cells, as well as in cells that had not been exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; dioxin), a potent inducer of Cyp1a1 expression. Evidence from in vivo methylation protection indicated that two G residues flanking AhRE3, one of which is required for binding of the 95-kDa protein, may be protected from methylation in uninduced cells and become exposed upon dioxin treatment, suggesting that the 95-kDa protein may be constitutively bound to AhRE3, and be displaced by binding of the Ah receptor complex. These results lend support to the concept that the transcriptional regulation of the [Ah] battery genes could be modulated by combinatorial interactions of the Ah receptor complex with other transcription factors. PMID: 7980646 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 335: Teratology. 1994 Nov;50(5):361-6. Immunohistochemical double-staining for Ah receptor and ARNT in human embryonic palatal shelves. Abbott BD, Probst MR, Perdew GH. Developmental Toxicology Division, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA. The aryl hydrocarbon receptor (AhR) and the AhR nuclear translocator protein (ARNT) are basic-helix-loop-helix-PAS (HLH) proteins involved in transcriptional regulation. Polycyclic aromatic halogenated chemicals, of which 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is the most potent, bind to the AhR. In the cellular cytoplasm, the AhR exists as a complex with the heat shock protein HSP90 and other small peptides. This complex dissociates following ligand binding and then the ligand-bound AhR binds ARNT. The ligand-AhR-ARNT complex interacts with a specific, nuclear DNA sequence, the dioxin response element (DRE), altering transcription of a regulated gene. Studies in hepatoma cell lines indicate that both proteins are required for regulation of transcription. In this study, AhR and ARNT were localized immunohistochemically in human embryonic palatal cells and specific patterns of expression were seen for each protein. A double-staining protocol revealed that epithelial cells expressed both AhR and ARNT, but in mesenchyme and nasal spine cartilage individual cells were identified which expressed either AhR or ARNT. This heterogeneous pattern may be a means of suppressing transcriptional regulation and also suggests the existence of other, unidentified basic-helix-loop-helix partner(s). The heterogeneous expression pattern may also reflect a complex role for these HLH proteins as transcriptional regulators of embryonic development. PMID: 7716743 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 336: J Biol Chem. 1994 Oct 21;269(42):26464-71. Baculovirus expression of the Ah receptor and Ah receptor nuclear translocater. Evidence for additional dioxin responsive element-binding species and factors required for signaling. Chan WK, Chu R, Jain S, Reddy JK, Bradfield CA. Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, Chicago, Illinois 60611. In an effort to facilitate the structural and biochemical analyses of the Ah receptor (AHR) and the Ah receptor nuclear translocator (ARNT), a baculovirus system was developed to express microgram-milligram quantities of the human version of these proteins. To simplify purification, a polyhistidine tag was cloned at their C termini so that the recombinant proteins could be specifically adsorbed to nickel-nitriloacetic acid-Sepharose. Expression studies revealed that approximately 23% of the overexpressed AHR was recovered in cell extracts with the remaining 77% forming insoluble aggregates. ARNT was found to be more soluble, with 90% recovery from cell extracts and only 10% aggregation. Photoaffinity labeling and gel shift assays demonstrated that the recombinant proteins bound ligand, heterodimerized, and recognized their cognate "dioxin response element" (DRE) in a manner similar to their native counterparts. Coexpression of the AHR and ARNT in Sf9 cells resulted in the in vivo generation of heterodimers that bound the DRE in the absence of ligand. Studies with the nickel-nitriloacetic acid-purified recombinant proteins demonstrated that the AHR and ARNT could bind DRE only when reconstituted with a heat-sensitive factor(s) present in soluble extracts from a variety of cell types. Use of these proteins also demonstrated the existence of at least three AHR-dependent DRE-binding species, suggesting that the AHR can bind to DRE in at least three distinct conformations. PMID: 7929368 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 337: J Biochem (Tokyo). 1994 Oct;116(4):845-51. Human arylhydrocarbon receptor: functional expression and chromosomal assignment to 7p21. Ema M, Matsushita N, Sogawa K, Ariyama T, Inazawa J, Nemoto T, Ota M, Oshimura M, Fujii-Kuriyama Y. Department of Chemistry, Faculty of Science, Tohoku University, Sendai. We isolated the human arylhydrocarbon receptor (AhR) cDNA from a human lung cDNA library, by using mouse AhR cDNA as a labeled probe. The nucleotide sequence of cloned human AhR cDNA is identical to the previously reported human AhR sequence [Dolwick et al. (1993), Mol. Pharmacol. 44, 911-917] from cell line HepG2. The overall amino acid identity with mouse AhR from cell line Hepa-1 is 72.5%. The human AhR expressed either in COS-7 cells or in a reticulocyte lysate in vitro translation system showed specific dioxin-binding activity and Arnt-dependent DNA-binding activity. Chromosomal localization of the AhR gene was determined to be chromosome 7p21 by fluorescent in situ hybridization and DNA blot hybridization using 23 human x mouse or Chinese hamster hybrid cell DNAs. PMID: 7883760 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 338: Mol Cell Biol. 1994 Sep;14(9):6075-86. Identification of functional domains of the aryl hydrocarbon receptor nuclear translocator protein (ARNT). Reisz-Porszasz S, Probst MR, Fukunaga BN, Hankinson O. Laboratory of Structural Biology and Molecular Medicine, University of California, Los Angeles 90024-1786. The activated aryl hydrocarbon receptor (AHR) and the AHR nuclear translocator (ARNT) bind DNA as a heterodimer. Both proteins represent a novel class of basic helix-loop-helix (bHLH)-containing transcription factors in that (i) activation of AHR requires the binding of ligand (e.g., 2,3,7,8-tetrachlorodibenzo-p-dioxin [TCDD]), (ii) the xenobiotic responsive element (XRE) recognized by the AHR/ARNT heterodimer differs from the recognition sequence for nearly all other bHLH proteins, and (iii) both proteins contain a PAS homology region, which in the Drosophila PER and SIM proteins functions as a dimerization domain. A cDNA for mouse ARNT has been cloned, and potential functional domains of ARNT were investigated by deletion analysis. A mutant lacking all regions of ARNT other than the bHLH and PAS regions is unimpaired in TCDD-dependent dimerization and subsequent XRE binding and only modestly reduced in ability to complement an ARNT-deficient mutant cell line, c4, in vivo. Both the first and second alpha helices of the bHLH region are required for dimerization. The basic region is required for XRE binding but not for dimerization. Deletion of either the A or B segments of the PAS region slightly affects TCDD-induced heterodimerization, while deletion of the complete PAS region severely affects (but does not eliminate) dimerization. Thus, ARNT possesses multiple domains required for maximal heterodimerization. Mutants deleted for PAS A, PAS B, and the complete PAS region all retain some degree of XRE binding, yet none can rescue the c4 mutant. Therefore, both the PAS A and PAS B segments, besides contributing to dimerization, apparently fulfill additional, unknown functions required for biological activity of ARNT. PMID: 8065341 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 339: Nucleic Acids Res. 1994 Aug 11;22(15):3038-44. Tissue specific expression of the rat Ah-receptor and ARNT mRNAs. Carver LA, Hogenesch JB, Bradfield CA. Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, Chicago, IL 60611. The Ah-receptor (AHR) is a ligand activated transcription factor that mediates the biological effects of agonists such as 2,3,7,8-tetrachlorodibenzo-p-dioxin. Upon binding agonists, the AHR dimerizes with a structurally related protein known as ARNT and this heterodimer then binds cognate enhancer elements and activates the expression of target genes. In this report we describe the cloning of the rat AHR cDNA and a fragment of the rat ARNT cDNA for use as probes in ribonuclease protection analysis. Ribonuclease protection analysis indicated that the rat AHR mRNA is expressed at the highest levels in the lung > thymus > kidney > liver while lower levels were expressed in heart and spleen. The rat AHR and ARNT mRNAs were expressed in a largely coordinate manner across the eight tissues examined with the exception of the placenta where AHR levels were relatively low compared to ARNT. In these experiments, a rare splice variant of the AHR was cloned that encoded a protein with a deletion in the ligand binding domain. In vitro expression studies demonstrated that in contrast to the full length AHR, the splice variant did not bind ligand nor did it bind to a cognate enhancer element in the presence of ARNT. PMID: 8065918 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 340: Cancer Res. 1994 May 15;54(10):2707-13. Antiestrogenic effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin on 17 beta-estradiol-induced pS2 expression. Zacharewski TR, Bondy KL, McDonell P, Wu ZF. Department of Pharmacology and Toxicology, University of Western Ontario, London, Canada. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) exhibits a broad spectrum of antiestrogenic activities in rodents and mammalian cells in culture. The effects of TCDD on 17 beta-estradiol (E2)-induction of pS2, a prognostic marker for breast cancer, were investigated in MCF-7, ZR-75, HeLa, and Hepa 1c1c7 wild-type and mutant cells. These effects were compared to the suppressive activities of the congener, 2,8-dichlorodibenzo-p-dioxin, and the established antiestrogens, ICI 164,384 and tamoxifen, in order to determine the relative potency of TCDD and to distinguish the mechanism of action of Ah receptor-mediated antiestrogens. Treatment of MCF-7 cells with 10 nM TCDD decreased E2-induced secreted pS2 protein levels by 50% and the induction of the transiently transfected -1100 to -86 pS2 promoter-regulated reporter gene (pS2-LUC) by 57%. Comparable effects on PS2-LUC activity were observed in HeLa and ZR-75 cells. In contrast, TCDD had minimal effects on pS2ERE(-405 to -393)-LUC induction, whereas treatment with 10 nM ICI 164,384 caused a 60% decrease in luciferase activity. In Hepa 1c1c7 wild-type and clone 1 (C1) mutant cells, TCDD also reduced E2 induction of pS2-LUC activity but had little effect in clone 4 (C4) or clone 12 (C12) mutant cells. However, suppression was reestablished following transfection of the human Ah receptor nuclear translocator (ARNT) complementary DNA expression vector into C4 cells and the mouse Ah receptor (AhR) complementary DNA expression vector into C12 cells. Induction of pS2-LUC activity by the ligand-dependent and -independent chimeric estrogen receptors (HE15, HE19, ERcVP16, and ERGR) were also used to examine the role of E2 metabolism and the mechanism of TCDD-mediated antiestrogenic activity. Induction by HE15 and ERcVP16 was suppressed by 57 and 74%, respectively, following treatment with TCDD, whereas ICI 164,384 was significantly less effective (38 and 20%, respectively). These results demonstrate a role for the Ah receptor in TCDD-mediated suppression of E2-induced pS2 expression. Data is presented demonstrating that the effect requires sequences within the pS2 promoter other than the estrogen response element and is independent of E2 oxidative metabolism. PMID: 8168101 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 341: Carcinogenesis. 1994 May;15(5):801-6. Interindividual difference in expression of human Ah receptor and related P450 genes. Hayashi S, Watanabe J, Nakachi K, Eguchi H, Gotoh O, Kawajiri K. Department of Biochemistry, Saitama Cancer Center Research Institute, Japan. The genomic clones of human aryl hydrocarbon receptor (Ahr) and Ahr nuclear translocator (Arnt) were isolated, and the structures of exon-intron junctions of these genes were partially determined. Based on the sequence information, a quantitative RT-PCR analysis was developed, and the expression of these genes was studied in various human tissues. mRNAs for Ahr and Arnt were widely expressed in human tissues and abundantly in lung. Individual difference in expression levels of Ahr and Arnt mRNA was observed in liver, lung and blood. In order to examine whether expression levels of Ahr and Arnt were associated with those of CYP1A1, we studied the expression of these mRNAs in blood among 20 healthy subjects, taking account of individuals' cigarette smoking habits. We found that the expression levels of CYP1A1 appeared to associate with those of Ahr and Arnt mRNAs (P < 0.06), and also that the expression of Ahr and Arnt was influenced by cigarette smoking. The expression of human Ahr and Arnt is reported here for the first time, providing a quantitative RT-PCR analysis as a useful tool for further studies. PMID: 7515333 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 342: Mol Pharmacol. 1994 Mar;45(3):428-38. The aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator protein show distinct subcellular localizations in Hepa 1c1c7 cells by immunofluorescence microscopy. Pollenz RS, Sattler CA, Poland A. McArdle Laboratory for Cancer Research, University of Wisconsin, Madison 53706. The aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (Arnt) protein were evaluated in the Hepa 1c1c7 (Hepa-1) cell line by indirect immunofluorescence microscopy and Western blot analysis. Wild-type (WT) Hepa-1 cells stained for AhR show intense cytoplasmic fluorescence with minimal nuclear reactivity. WT cells treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) show a time-dependent decrease in cytoplasmic AhR staining and a concomitant increase in nuclear fluorescence. WT cells stained for Arnt show nuclear fluorescence with minimal cytoplasmic reactivity, a pattern unchanged after TCDD treatment. Hepa-1 type II variants express normal levels of AhR but are defective in TCDD-mediated induction of cytochrome P4501A1. Type II variants stained for Arnt show reduced nuclear fluorescence, compared with WT cells, and express minimal levels of Arnt protein, as determined by Western blot analysis. Type II variants stained for the AhR show intense cytoplasmic fluorescence that becomes nuclear after TCDD treatment. Detailed evaluation by immunoelectron microscopy of the AhR and Arnt present in the nuclear compartment of WT cells shows that both proteins are uniformly distributed and do not appear to be associated with nuclear pores, membranes, or nucleoli. Western blot analysis of nuclei isolated from WT Hepa-1 cells fractionated with Nonidet P-40 shows that minimal levels of AhR or Arnt are retained in the nuclear fraction after TCDD treatment. Collectively, these results indicate that the unliganded AhR resides in the cytoplasm, Arnt is localized to the nucleus, and Hepa-1 cells defective in Arnt expression exhibit TCDD-mediated nuclear accumulation of the AhR. PMID: 8145729 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 343: Rev Physiol Biochem Pharmacol. 1994;125:1-42. Aryl hydrocarbon or dioxin receptor: biologic and toxic responses. Bock KW. Institute of Toxicology, University of Tubingen, Germany. 1. The AhR represents a ligand-activated transcription factor. Receptor agonists include planar aromatic compounds, a variety of heterocyclic plant constituents, and PCDD/PCDF. The latter lead to persistent activation of the receptor due to their strong binding affinity and long biologic half-life of over 10 years in human blood and fat. Practically every person on earth is exposed to these compounds via the diet (> 90%) and by high concentrations in mother's milk. PCDD/PCDF produced toxic responses in exposed people (primarily chloracne and immunosuppression) in the past. However, the present PCDD/PCDF levels (basal levels) in the general population are below those warranting toxicologic concern. 2. The AhR has been characterized as a helix-loop-helix transcription factor related to the Drosophila developmental genes sim and per. The cytosolic form of the receptor is present as an inactive complex with two subunits of HSP90. After ligand binding HSP90 is released and the receptor enters the nucleus as a heterodimer together with a related protein ARNT. It binds with high affinity to certain enhancer elements in the upstream region of several genes such as cytochrome P4501A1 (CYP1A1). The AhR transcriptionally activates several drug-metabolizing enzymes and proteins involved in growth/differentiation, such as the plasminogen activator inhibitor PAI-2 and IL-1 beta. In addition, it modulates the action of a number of other nuclear transcription factors such as receptors of the steroid hormone receptor superfamily and of cell surface receptors such as EGF. With the exception of CYP1A1 induction, little is known about the mechanism of transcriptional activation of the AhR-controlled genes. Many AhR-modulated biologic responses (such as modulation of the estrogen and EGF receptor) appear to be indirect. 3. Persistent activation of the AhR is probably responsible for toxic responses in experimental animals and humans. They are markedly tissue and species specific. In rodents a wasting syndrome, immunosuppression, teratogenicity, chloracne, and carcinogenicity/tumor promotion have been well studied. There is good evidence for an involvement for the AhR in these responses. However, the chain of events from receptor activation to the diverse toxic endpoints is largely unknown. Alteration of growth and differentiation of epithelial tissues may underlie most of the toxic responses. A lot has already been achieved, mostly by characterizing the AhR and transcriptional activation of CYP1A1. Still more work lies ahead of us, for example, elucidation of the physiologic roles of the AhR and of the chains of events from receptor activation to the various biologic and toxic endpoints. Publication Types: Review PMID: 7984872 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 344: Receptor. 1994 Winter;4(4):209-22. DNA sequence requirements for Ah receptor/Arnt recognition determined by in vitro transcription. McLane KE, Whitlock JP Jr. Scripps Research Institute, Department of Immunology, La Jolla, CA 92037. The enhancer of the mouse cytochrome P450 cyp1a1 gene, which contains six binding sites (A-F) for the Ah receptor (AhR), has been a useful model system for studying the mechanism of AhR-mediated activation of transcription. In the presence of ligand, AhR interacts with its dimerization partner, Arnt, and the heteromeric complex is able to bind DNA. In the present study, we test the effects of single base pair substitutions of site D on the ability of the AhR/Arnt heteromer to recognize this response element using an in vitro transcription system. Synthetic oligodeoxyribonucleotides corresponding to the wild-type sequence of site D, or single base pair mutations of that sequence, were used to compete for AhR/Arnt binding with the transcription template. Using this competition assay, the sequence of the core recognition motif 5'-GCGTG-3' was shown to be critical for AhR/Arnt binding, and the importance of the position and orientation of the G:C and A:T base pairs of this sequence was determined. PMID: 7894337 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 345: Exp Clin Immunogenet. 1994;11(2-3):65-74. Ah receptor: a novel ligand-activated transcription factor. Fujii-Kuriyama Y, Ema M, Mimura J, Sogawa K. Department of Chemistry, Faculty of Science, Tohoku University, Sendai, Japan. Two years have passed since the Ah receptor (AhR) and the AhR nuclear translocator (Arnt) were revealed to constitute a novel ligand-activated transcription factor by molecular biological studies on cloned cDNAs. Many interesting properties of the AhR/Arnt system have begun to be elucidated. We present here a brief summary of the molecular biology of AhR and Arnt. Publication Types: Review PMID: 7826667 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 346: Proc Natl Acad Sci U S A. 1993 Sep 15;90(18):8566-70. In vitro analysis of Ah receptor domains involved in ligand-activated DNA recognition. Dolwick KM, Swanson HI, Bradfield CA. Department of Pharmacology, Northwestern University Medical School, Chicago, IL 60611. The Ah receptor (AHR) is a basic helix-loop-helix protein that mediates the effects of 2,3,7,8-tetrachloro-dibenzo-p-dioxin. In this report, we describe a rabbit reticulocyte system that allows functional expression of both the AHR and its dimeric partner, the AHR nuclear translocator protein (ARNT). By using this in vitro system, we were able to reconstitute agonist binding to the AHR and agonist-induced AHR-ARNT recognition of a cognate DNA enhancer sequence. Expression of AHR deletion mutants revealed the location of N-terminal domains responsible for ligand and DNA recognition and C-terminal domains that play roles in agonist-induced DNA recognition. PMID: 8397410 [PubMed - indexed for MEDLINE] --------------------------------------------------------------- 347: Ann N Y Acad Sci. 1993 Jun 23;685:624-40. Role of the Ah receptor and the dioxin-inducible [Ah] gene battery in toxicity, cancer, and signal transduction. Nebert DW, Puga A, Vasiliou V. Department of Environmental Health, University of Cincinnati Medical Center, Ohio 45267-0056. 1. On the basis of our current knowledge about the evolution of drug-metabolizing enzymes, it appears to be extremely likely that these enzymes play a critical role in maintaining steady-state levels of the ligands involved in ligand-modulated transcription of genes effecting growth, differentiation, homeostasis, and neuroendocrine functions. 2. The original observations about genetic differences in CYP1A1 (cytochrome P1-450) induction by TCDD or benzo[a]pyrene in the mouse have led to an appreciation for a similar polymorphism in the human and the recent cloning of the murine Ah receptor (Ahr) and human Ah receptor nuclear translocator (ARNT) genes. It is most likely that the correlation between genetic differences in human or murine CYP1A1 inducibility by polycyclic hydrocarbons or TCDD and increased risk of cancer will be explained by differences in the AHR gene, leading to enhanced tumor promotion (rather than in the CYP1A1 structural gene). Perhaps the same will be found for birth defects, immunotoxicity, and other forms of toxic damage caused by these environmental chemicals. 3. In a manner similar to that of the phorbol ester tumor promoter, TCDD induces intracellular Ca2+ changes, accumulation of FOS and JUN mRNAs, and large increases in AP-1 transcription factor activity. Interestingly, these early effects of TCDD, and also of benzo[a]pyrene, appear not to require the Ah receptor. 4. Many genes are induced by TCDD, and many others are induced by electrophilic metabolites such as quinones and H2O2; using several mouse experimental systems, we have defined a subset of six of these genes as constituting the [Ah] battery by the sole criterion that a functional CYP1A1 or CYP1A2 enzyme is able to repress the expression of genes that are members of this gene battery. Publication Types: Review Review, Tutorial PMID: 8395783 [PubMed - indexed for MEDLINE] ---------------------------------------------------------------