1
|
Feng H, Cao H, Li J, Zhang H, Xue Q, Liu X, Zhang A, Fu J. Estrogenic activity of benzotriazole UV stabilizers evaluated through in vitro assays and computational studies. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 727:138549. [PMID: 32330715 DOI: 10.1016/j.scitotenv.2020.138549] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 06/11/2023]
Abstract
Benzotriazole UV stabilizers (BUVs) are used in a variety of products to prevent yellowing and degradation. However, knowledge of the estrogenic activity of BUVs is still lacking. In the present study, a strategy combining in vitro assays and computational studies was adopted to evaluate the estrogenic activity of BUVs. 2-(2-Hydroxy-5-methlphenyl) benzotriazole (UV-P), 2-(5-tert-butyl-2-hydroxyphenyl)benzotriazole (UV-PS), and 2-(3-Allyl-2-hydroxy-5-methylphenyl)-2H-benzotriazole (UV-9) induced partial estrogenic activity while 2-(2-hydroxy-5-tert-octyl-phenyl)benzotriazole (UV-329), 2-(3-s-butyl-5-tert-butyl-2-hydroxyphenyl)benzotriazole (UV-350), and 3-(2H-benzotriazolyl)-5- (1,1-di-methylethyl)-4-hydroxy-benzene-propanoic acid octyl esters (UV-384) showed no estrogenic activity in MVLN assays. The results of in vitro assays were in accord with the results of computational studies. Moreover, ICI 182,780 suppressed the estrogenic activity of BUVs both in the absence and presence of E2, demonstrating that the estrogen responsive element (ERE) transcription activities of BUVs are generated through an estrogen receptor (ER) mediated pathway. Our findings suggest that the endocrine disruption effects of BUVs are a cause for concern.
Collapse
Affiliation(s)
- Hongru Feng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Science, Beijing 100085, China; College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100190, China; Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Huiming Cao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Science, Beijing 100085, China; Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Juan Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Science, Beijing 100085, China
| | - Haiyan Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Qiao Xue
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Science, Beijing 100085, China
| | - Xian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Science, Beijing 100085, China
| | - Aiqian Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Science, Beijing 100085, China; College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100190, China.
| | - Jianjie Fu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Science, Beijing 100085, China.
| |
Collapse
|
2
|
Yoshida I, Ishida K, Yoshikawa H, Kitamura S, Hiromori Y, Nishioka Y, Ido A, Kimura T, Nishikawa JI, Hu J, Nagase H, Nakanishi T. In vivo profiling of 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced estrogenic/anti-estrogenic effects in female estrogen-responsive reporter transgenic mice. JOURNAL OF HAZARDOUS MATERIALS 2020; 385:121526. [PMID: 31732351 DOI: 10.1016/j.jhazmat.2019.121526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 06/10/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), commonly referred to simply as "dioxin", is a persistent environmental pollutant. Because of its high environmental persistence and biological accumulation, humans and animals are often exposed to TCDD. Therefore, the harmful effects on humans and animals is a major concern. Although studies have elucidated the adverse estrogenic and anti-estrogenic effects of TCDD, it is unclear in which tissues TCDD exerts these effects in vivo. To investigate the estrogen-related effects of TCDD in various tissues, we generated an improved estrogen-responsive reporter transgenic mouse in which the luciferase gene luc2 is expressed in response to estrogenic signals. Using these mice, we clarified that TCDD inhibits estrogenic signaling in liver and kidney but enhances estrogenic signaling in the pituitary gland in the same individual. Expression of aryl hydrocarbon receptor, aryl hydrocarbon receptor nuclear translocator, and estrogen receptor alpha mRNA was detected in liver, kidney, and pituitary gland, suggesting that the effects of TCDD on estrogenic signaling in these organs is independent of the expression pattern of these receptors. Thus, our results indicate that TCDD exerts both estrogenic and anti-estrogenic tissue-specific effects within the same individual.
Collapse
Affiliation(s)
- Ichiro Yoshida
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Keishi Ishida
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan; Research Fellow of the Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan
| | - Hiroshi Yoshikawa
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Sho Kitamura
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Youhei Hiromori
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan; Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie, 513-8670, Japan
| | - Yasushi Nishioka
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Akiko Ido
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Tomoki Kimura
- Faculty of Science and Engineering, Setsunan University, 17-8 Ikedanakamachi, Neyagawa, 572-8508, Japan
| | - Jun-Ichi Nishikawa
- Laboratory of Health Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Kyuban-cho, Koshien, Nishinomiya, Hyogo, 663-8179, Japan
| | - Jianying Hu
- MOE Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Hisamitsu Nagase
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan.
| |
Collapse
|
3
|
Warner GR, Mourikes VE, Neff AM, Brehm E, Flaws JA. Mechanisms of action of agrochemicals acting as endocrine disrupting chemicals. Mol Cell Endocrinol 2020; 502:110680. [PMID: 31838026 PMCID: PMC6942667 DOI: 10.1016/j.mce.2019.110680] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023]
Abstract
Agrochemicals represent a significant class of endocrine disrupting chemicals that humans and animals around the world are exposed to constantly. Agrochemicals can act as endocrine disrupting chemicals through a variety of mechanisms. Recent studies have shown that several mechanisms of action involve the ability of agrochemicals to mimic the interaction of endogenous hormones with nuclear receptors such as estrogen receptors, androgen receptors, peroxisome proliferator activated receptors, the aryl hydrocarbon receptor, and thyroid hormone receptors. Further, studies indicate that agrochemicals can exert toxicity through non-nuclear receptor-mediated mechanisms of action. Such non-genomic mechanisms of action include interference with peptide, steroid, or amino acid hormone response, synthesis and degradation as well as epigenetic changes (DNA methylation and histone modifications). This review summarizes the major mechanisms of action by which agrochemicals target the endocrine system.
Collapse
Affiliation(s)
- Genoa R Warner
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, 61802, IL, United States
| | - Vasiliki E Mourikes
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, 61802, IL, United States
| | - Alison M Neff
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, 61802, IL, United States
| | - Emily Brehm
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, 61802, IL, United States
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, 61802, IL, United States.
| |
Collapse
|
4
|
van den Brand AD, Rubinstein E, van den Berg M, van Duursen MBM. GH3 and RC-4BC cell lines are not suitable as in vitro models to study prolactin modulation and AHR responsiveness in rat pituitary. Mol Cell Endocrinol 2019; 496:110520. [PMID: 31352040 DOI: 10.1016/j.mce.2019.110520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/21/2019] [Accepted: 07/24/2019] [Indexed: 01/10/2023]
Abstract
Some environmental contaminants and pharmaceuticals increase the incidence of uterine tumors in toxicological studies with rats. These tumors can result from a hormonal imbalance due to rat-specific disrupted pituitary prolactin regulation, and are therefore of questionable relevance for humans. In this study we compared in vitro prolactin regulation in rat primary pituitary cells to that in pituitary cell lines, GH3 and RC-4BC. Moreover, we assessed the potential effects of aryl hydrocarbon receptor (AHR) activation on prolactin regulation by using two different AHR agonists, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and DELAQ, the N-deethylated minor metabolite of the pharmaceutical laquinimod. In rat primary pituitary cells, known prolactin stimulant thyrotropin-releasing hormone (TRH) marginally increased prolactin secretion (1.2-fold) and gene expression (1.3-fold). In contrast, synthetic dopamine receptor agonist quinpirole, a known inhibitor of prolactin release, significantly inhibited prolactin secretion (2.6-fold) and gene expression (3.6-fold). In GH3 cells, TRH strongly increased prolactin secretion (6.8-fold) and gene expression (30.8-fold), whereas quinpirole did not affect prolactin secretion nor gene expression. In RC-4BC cells, both TRH and quinpirole did not modulate prolactin secretion nor gene expression. Prolactin secretion and gene expression did not significantly change upon exposure to TCDD or DELAQ. However, DELAQ, but not TCDD, attenuated quinpirole-inhibited prolactin gene expression by 51% in primary pituitary cells. This study shows that pituitary prolactin regulation in rat primary pituitary cells in vitro is distinctly different from rat pituitary cell lines GH3 and RC-4BC. Therefore, effects on pituitary prolactin regulation in vitro should best be performed using rat primary pituitary cells. Additionally, AHR ligands may interact with rat pituitary prolactin regulation, but this appears to depend on the ligand and constitutive prolactin secretion. However, interpretation of the in vitro results with respect to occurrence of uterine tumors in rats should take the complex regulation of prolactin release in the pituitary into account as well as the in vivo hypothalamus-pituitary-gonadal (HPG) axis and its feedback loops.
Collapse
Affiliation(s)
- A D van den Brand
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM, the Netherlands.
| | - E Rubinstein
- TEVA Pharmaceutical Industries Ltd, Netanya, Israel
| | - M van den Berg
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM, the Netherlands
| | - M B M van Duursen
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3584 CM, the Netherlands; Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Wahlang B. Exposure to persistent organic pollutants: impact on women's health. REVIEWS ON ENVIRONMENTAL HEALTH 2018; 33:331-348. [PMID: 30110273 DOI: 10.1515/reveh-2018-0018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/18/2018] [Indexed: 05/23/2023]
Abstract
This literature review focuses on the causal relationship between persistent organic pollutants (POPs) exposure and women's health disorders, particularly cancer, cardio-metabolic events and reproductive health. Progressive industrialization has resulted in the production of a multitude of chemicals that are released into the environment on a daily basis. Environmental chemicals or pollutants are not only hazardous to our ecosystem but also lead to various health problems that affect the human population worldwide irrespective of gender, race or age. However, most environmental health studies that have been conducted, until recently, were exclusively biased with regard to sex and gender, beginning with exposure studies that were reported mostly in male, occupational workers and animal studies being carried out mostly in male rodent models. Health-related issues pertaining to women of all age groups have not been studied thoroughly and rather disregarded in most aspects of basic health science research and it is therefore pertinent that we address these limitations in environmental health. The review also addresses studies looking at the associations between health outcomes and exposures to POPs, particularly, polychlorinated biphenyls (PCBs), dioxins and pesticides, reported in cohort studies while accounting for gender differences. Considering that current levels of POPs in women can also impact future generations, informative guidelines related to dietary patterns and exposure history are needed for women of reproductive age. Additionally, occupational cohorts of highly exposed women worldwide, such as women working in manufacturing plants and female pesticide applicators are required to gather more information on population susceptibility and disease pathology.
Collapse
Affiliation(s)
- Banrida Wahlang
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, 505 S. Hancock Street, CTRB, Louisville, KY 40202-1617, USA
| |
Collapse
|
6
|
Cao H, Li X, Zhang W, Wang L, Pan Y, Zhou Z, Chen M, Zhang A, Liang Y, Song M. Anti-estrogenic activity of tris(2,3-dibromopropyl) isocyanurate through disruption of co-activator recruitment: experimental and computational studies. Arch Toxicol 2018; 92:1471-1482. [DOI: 10.1007/s00204-018-2159-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/17/2018] [Indexed: 12/21/2022]
|
7
|
Helle J, Keiler AM, Zierau O, Dörfelt P, Vollmer G, Lehmann L, Chittur SV, Tenniswood M, Welsh J, Kretzschmar G. Effects of the aryl hydrocarbon receptor agonist 3-methylcholanthrene on the 17β-estradiol regulated mRNA transcriptome of the rat uterus. J Steroid Biochem Mol Biol 2017; 171:133-143. [PMID: 28285017 DOI: 10.1016/j.jsbmb.2017.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 03/01/2017] [Accepted: 03/06/2017] [Indexed: 10/20/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are products of incomplete combustion of organic compounds, abundant in exhaust fumes and cigarette smoke. They act by binding to the aryl hydrocarbon receptor (AHR) which induces expression of phase 1 and phase 2 enzymes in the liver. PAH induced AHR activation may also lead to adverse effects by modulating other pathways, for example estrogen receptor (ER) signaling in the female reproductive tract. We have investigated the effects of the PAH 3-methylcholanthrene (3-MC) on 17β-estradiol (E2) dependent signaling in the uterus of ovariectomized rats to characterize the cross talk between AHR and ER on an mRNA transcriptome wide scale. A standard three day uterotrophic assay was performed in young adult Lewis rats. Treatment induced effects were analyzed using histology, immunohistochemistry and gene expression analysis by microarray and qPCR. 3-MC shows broad E2 antagonistic effects on uterine mRNA transcription of the vast majority of E2 regulated genes, significantly altering prostaglandin biosynthesis, complement activation, coagulation pathways and other inflammatory response pathways. The regulation of ER expression in the uterus, but not the regulation of E2 metabolism in the liver, was identified as a potentially important factor in mediating this general antiestrogenic effect. The regulation of prostaglandin biosynthesis by E2 is important for inflammation-like events during pregnancy including the initiation of birth. Our results suggest that adverse effects of PAHs on prostaglandin related pathways are likely caused by the interference with E2 signaling, specifically by inhibiting the E2 mediated downregulation of PGF2α. Characterization of the generalized antagonistic effect of 3-MC on E2 dependent signaling in the rat uterus thus contributes to a better understanding of molecular mechanisms of the toxicity of PAHs in female reproductive organs.
Collapse
Affiliation(s)
- Janina Helle
- Institute of Zoology, Molecular Cell Physiology and Endocrinology, Technische Universität Dresden, 01062, Dresden, Germany
| | - Annekathrin M Keiler
- Institute of Zoology, Molecular Cell Physiology and Endocrinology, Technische Universität Dresden, 01062, Dresden, Germany
| | - Oliver Zierau
- Institute of Zoology, Molecular Cell Physiology and Endocrinology, Technische Universität Dresden, 01062, Dresden, Germany
| | - Peggy Dörfelt
- Institute of Zoology, Molecular Cell Physiology and Endocrinology, Technische Universität Dresden, 01062, Dresden, Germany
| | - Günter Vollmer
- Institute of Zoology, Molecular Cell Physiology and Endocrinology, Technische Universität Dresden, 01062, Dresden, Germany
| | - Leane Lehmann
- Institute of Pharmacy and Food Chemistry, Universität Würzburg, 97070 Würzburg, Germany
| | - Sridar V Chittur
- Cancer Research Center and Department of Biomedical Sciences, University at Albany, NY 12144-2345, United States
| | - Martin Tenniswood
- Cancer Research Center and Department of Biomedical Sciences, University at Albany, NY 12144-2345, United States
| | - JoEllen Welsh
- Cancer Research Center and Department of Biomedical Sciences, University at Albany, NY 12144-2345, United States
| | - Georg Kretzschmar
- Institute of Zoology, Molecular Cell Physiology and Endocrinology, Technische Universität Dresden, 01062, Dresden, Germany.
| |
Collapse
|
8
|
Endocrine Disrupting Chemicals and Endometrial Cancer: An Overview of Recent Laboratory Evidence and Epidemiological Studies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14030334. [PMID: 28327540 PMCID: PMC5369169 DOI: 10.3390/ijerph14030334] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 02/13/2017] [Accepted: 02/22/2017] [Indexed: 01/10/2023]
Abstract
Background: Although exposure to endocrine disruptor compounds (EDCs) has been suggested as a contributing factor to a range of women's health disorders including infertility, polycystic ovaries and the early onset of puberty, considerable challenges remain in attributing cause and effect on gynaecological cancer. Until recently, there were relatively few epidemiological studies examining the relationship between EDCs and endometrial cancer, however, in the last years the number of these studies has increased. Methods: A systematic MEDLINE (PubMed) search was performed and relevant articles published in the last 23 years (from 1992 to 2016) were selected. Results: Human studies and animal experiments are confirming a carcinogenic effect due to the EDC exposure and its carcinogenesis process result to be complex, multifactorial and long standing, thus, it is extremely difficult to obtain the epidemiological proof of a carcinogenic effect of EDCs for the high number of confusing factors. Conclusions: The carcinogenic effects of endocrine disruptors are plausible, although additional studies are needed to clarify their mechanisms and responsible entities. Neverthless, to reduce endocrine disruptors (ED) exposure is mandatory to implement necessary measures to limit exposure, particularly during those periods of life most vulnerable to the impact of oncogenic environmental causes, such as embryonic period and puberty.
Collapse
|
9
|
Sechman A, Batoryna M, Antos PA, Hrabia A. Effects of PCB 126 and PCB 153 on secretion of steroid hormones and mRNA expression of steroidogenic genes (STAR, HSD3B, CYP19A1) and estrogen receptors (ERα, ERβ) in prehierarchical chicken ovarian follicles. Toxicol Lett 2016; 264:29-37. [PMID: 27832956 DOI: 10.1016/j.toxlet.2016.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/26/2016] [Accepted: 11/05/2016] [Indexed: 11/25/2022]
Abstract
The objective of this study was to assess the in vitro effects of dioxin-like PCB 126 and non-dioxin-like PCB 153 on basal and ovine LH (oLH)-stimulated testosterone (T) and estradiol (E2) secretion and expression of steroidogenic genes (STAR, HSD3B and CYP19A1) and estrogen receptors α (ERα) and β (ERβ) in white (WF) and yellowish (YF) prehierarchical follicles of the hen ovary. Steroid concentrations in a medium and gene expression in follicles following 6h of exposition were determined by RIA and real-time qPCR, respectively. Both PCBs increased basal and oLH-stimulated T secretion by the WF follicles. PCB 126 reduced basal E2 secretion by the WF follicles. PCB 153 elevated but PCB 126 reduced oLH-stimulated E2 secretion by the prehierarchical follicles. PCB 126 increased basal STAR and HSD3B and reduced CYP19A1 mRNA expression in these follicles. PCB 153 increased basal expression of STAR and HSD3B in YF follicles, but diminished HSD3B mRNA levels in the WF. The studied PCBs had an opposite effect on basal and oLH-stimulated CYP19A1 mRNA expression in prehierarchical follicles. Both PCBs modulated basal and inhibited oLH-stimulated ERα and ERβ gene expression in the prehierarchical follicles. In conclusion, data of the current study demonstrate the congener-specific effects of PCBs on sex steroid secretion by prehierarchical follicles of the chicken ovary, which are at least partly related to STAR, HSD3B and CYP19A1 gene expression. It is suggested that PCBs, by influencing follicular steroidogenesis and expression of estrogen receptors, may impair development and selection of yellowish follicles to the preovulatory hierarchy.
Collapse
Affiliation(s)
- Andrzej Sechman
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland.
| | - Marta Batoryna
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| | - Piotr A Antos
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| | - Anna Hrabia
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| |
Collapse
|
10
|
Barć J, Gregoraszczuk EL. Halowax 1051 affects steroidogenesis by down-regulation of aryl hydrocarbon and estrogen receptors and up-regulation of androgen receptor in porcine ovarian follicles. CHEMOSPHERE 2016; 144:467-474. [PMID: 26386772 DOI: 10.1016/j.chemosphere.2015.09.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/01/2015] [Accepted: 09/06/2015] [Indexed: 06/05/2023]
Abstract
Polychlorinated naphthalenes (PCNs) are thought to interact with the aryl hydrocarbon receptor (AHR) and to have enzyme-inducing properties comparable to polychlorinated dibenzo-p-dioxins, therefore activation of steroid hormone receptors in endocrine tissues is also possible. The aim of the present study was to examine the effects of PCNs mixture, Halowax 1051 on gene and protein expression of receptors: estradiol (ERα/β), androgen (AR) and AHRGene expression was evaluated by real-time PCR after 6 h of exposition and protein expression by Western blot after 24 h. Levels of sex steroids: androstenedione (A4), estradiol (E2) and testosterone (T) were measured by enzyme immunoassays. Results of the data show down-regulation of AHR gene expression after 6 h in parallel with an inhibition in AHR protein expression at doses 10 pg-10 ng/mL, down-regulation of ER at all doses used, and up-regulation of AR gene expression at doses 1 and 10 ng/mL without affecting their protein expression. To indicate the involvement of AHR, ERs and AR in the impact of PCNs on steroidogenesis, we used their specific blockers. Blocker of AHR reversed the inhibitory effect of Halowax 1051 on A4 secretion, and strengthened its effect on T secretion. Blockers of both ER and AR had no effect on Halowax 1051 action on steroids secretion. The results of this study suggest that AHR is involved in the effect of PCNs on steroidogenesis in the ovary. Additionally, we propose that cross-talk between AHR-ER and AHR-AR receptors mediates the effects of Halowax 1051 on ovarian follicles.
Collapse
Affiliation(s)
- Justyna Barć
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology, Jagiellonian University in Kraków, Kraków, Poland.
| | - Ewa Lucja Gregoraszczuk
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology, Jagiellonian University in Kraków, Kraków, Poland
| |
Collapse
|
11
|
Ghisari M, Long M, Tabbo A, Bonefeld-Jørgensen EC. Effects of currently used pesticides and their mixtures on the function of thyroid hormone and aryl hydrocarbon receptor in cell culture. Toxicol Appl Pharmacol 2015; 284:292-303. [PMID: 25684042 DOI: 10.1016/j.taap.2015.02.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/27/2015] [Accepted: 02/04/2015] [Indexed: 10/24/2022]
Abstract
Evidence suggest that exposure to pesticides can interfere with the endocrine system by multiple mechanisms. The endocrine disrupting potential of currently used pesticides in Denmark was analyzed as single compounds and in an equimolar mixture of 5 selected pesticides. The pesticides were previously analyzed for effects on the function of estrogen and androgen receptors, the aromatase enzyme and steroidogenesis in vitro. In this study, the effect on thyroid hormone (TH) function and aryl hydrocarbon receptor (AhR) transactivity was assessed using GH3 cell proliferation assay (T-screen) and AhR responsive luciferase reporter gene bioassay, respectively. Thirteen pesticides were analyzed as follows: 2-methyl-4-chlorophenoxyacetic acid, terbuthylazine, iodosulfuron-methyl-sodium, mesosulfuron-methyl, metsulfuron-methyl, chlormequat chloride, bitertanol, propiconazole, prothioconazole, mancozeb and its metabolite ethylene thiourea, cypermethrin, tau-fluvalinate, and malathion (currently banned in DK). In the T-screen, prothioconazole, malathion, tau-fluvalinate, cypermethrin, terbuthylazine and mancozeb significantly stimulated and bitertanol and propiconazole slightly reduced the GH3 cell proliferation. In the presence of triiodothyronine (T3), prothioconazole, tau-fluvalinate, propiconazole, cypermethrin and bitertanol significantly antagonized the T3-induced GH3 cell proliferation. Eleven of the tested pesticides agonized the AhR function, and bitertanol and prothioconazole inhibited the basal AhR activity. Bitertanol, propiconazole, prothioconazole and cypermethrin antagonized the TCDD-induced AhR transactivation at the highest tested concentration. The 5-component mixture had inducing effect but the combined effect could not be predicted due to the presence of bitertanol eliciting inhibitory effect. Upon removal of bitertanol from the mixture, the remaining four pesticides acted additively. In conclusion, our data suggest that pesticides currently used in Denmark can interfere with TH signaling and AhR function in vitro and might have the potential to cause endocrine disruption.
Collapse
Affiliation(s)
- Mandana Ghisari
- Centre for Arctic Health & Unit of Cellular and Molecular Toxicology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Manhai Long
- Centre for Arctic Health & Unit of Cellular and Molecular Toxicology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Agnese Tabbo
- Centre for Arctic Health & Unit of Cellular and Molecular Toxicology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Eva Cecilie Bonefeld-Jørgensen
- Centre for Arctic Health & Unit of Cellular and Molecular Toxicology, Department of Public Health, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
12
|
Abstract
Hormonal imprinting is an epigenetic process which is taking place perinatally at the first encounter between the developing hormone receptors and their target hormones. The hormonal imprinting influences the binding capacity of receptors, the hormone synthesis of the cells, and other hormonally regulated functions, as sexual behavior, aggressivity, empathy, etc. However, during the critical period, when the window for imprinting is open, molecules similar to the physiological imprinters as synthetic hormone analogs, other members of the hormone families, environmental pollutants, etc. can cause faulty imprinting with life-long consequences. The developing immune system, the cells of which also have receptors for hormones, is very sensitive to faulty imprinting, which causes alterations in the antibody and cytokine production, in the ratio of immune cells, in the defense against bacterial and viral infections as well as against malignant tumors. Immune cells (lymphocytes, monocytes, granulocytes and mast cells) are also producing hormones which are secreted into the blood circulation as well as are transported locally (packed transport). This process is also disturbed by faulty imprinting. As immune cells are differentiating during the whole life, faulty imprinting could develop any time, however, the most decisive is the perinatal imprinting. The faulty imprinting is inherited to the progenies in general and especially in the case of immune system. In our modern world the number and amount of artificial imprinters (e.g. endocrine disruptors and drugs) are enormously increasing. The effects of the faulty imprinters most dangerous to the immune system are shown in the paper. The present and future consequences of the flood of faulty imprintings are unpredictable however, it is discussed.
Collapse
Affiliation(s)
- György Csaba
- 1 Semmelweis University Department of Genetics, Cell and Immunobiology Budapest Hungary
| |
Collapse
|
13
|
Safe S, Lee SO, Jin UH. Role of the aryl hydrocarbon receptor in carcinogenesis and potential as a drug target. Toxicol Sci 2013; 135:1-16. [PMID: 23771949 PMCID: PMC3748760 DOI: 10.1093/toxsci/kft128] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/03/2013] [Indexed: 12/22/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is highly expressed in multiple organs and tissues, and there is increasing evidence that the AHR plays an important role in cellular homeostasis and disease. The AHR is expressed in multiple tumor types, in cancer cell lines, and in tumors from animal models, and the function of the AHR has been determined by RNA interference, overexpression, and inhibition studies. With few exceptions, knockdown of the AHR resulted in decreased proliferation and/or invasion and migration of cancer cell lines, and in vivo studies in mice overexpressing the constitutively active AHR exhibited enhanced stomach and liver cancers, suggesting a pro-oncogenic role for the AHR. In contrast, loss of the AHR in transgenic mice that spontaneously develop colonic tumors and in carcinogen-induced liver tumors resulted in increased carcinogenesis, suggesting that the receptor may exhibit antitumorigenic activity prior to tumor formation. AHR ligands also either enhanced or inhibited tumorigenesis, and these effects were highly tumor specific, demonstrating that selective AHR modulators that exhibit agonist or antagonist activities represent an important new class of anticancer agents that can be directed against multiple tumors.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843-4466, USA.
| | | | | |
Collapse
|
14
|
Nault R, Forgacs AL, Dere E, Zacharewski TR. Comparisons of differential gene expression elicited by TCDD, PCB126, βNF, or ICZ in mouse hepatoma Hepa1c1c7 cells and C57BL/6 mouse liver. Toxicol Lett 2013; 223:52-9. [PMID: 23994337 DOI: 10.1016/j.toxlet.2013.08.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/17/2013] [Accepted: 08/19/2013] [Indexed: 12/21/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a promiscuous receptor activated by structurally diverse synthetic and natural compounds. AhR activation may lead to ligand-specific changes in gene expression despite similarities in mode of action. Therefore, differential gene expression elicited by four structurally diverse, high affinity AhR ligands (2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 10nM, 30 μg/kg), 3,3',4,4',5-pentachlorobiphenyl (PCB126; 100nM, 300μg/kg), β-naphthoflavone (βNF; 10 μM, 90 mg/kg), and indolo[3,2-b]carbazole (ICZ; 1μM)) in mouse Hepa1c1c7 hepatoma cells and C57BL/6 mouse liver samples were compared. A total of 288, 183, 119, and 131 Hepa1c1c7 genes were differentially expressed (|fold-change|≥ 1.5, P1(t)≥ 0.9999) by TCDD, βNF, PCB126, and ICZ, respectively. Only ∼35% were differentially expressed by all 4 ligands in Hepa1c1c7 cells. In vivo, 661, 479, and 265 hepatic genes were differentially expressed following treatment with TCDD, βNF, and PCB126, respectively. Similar to Hepa1c1c7 cells, ≤ 34% of gene expression changes were common across all ligands. Principal components analysis identified time-dependent gene expression divergence. Comparisons of ligand-elicited expression between Hepa1c1c7 cells and mouse liver identified only 11 common gene expression changes across all ligands. Although metabolism may explain some ligand-specific gene expression changes, PCB126, βNF, and ICZ also elicited divergent expression compared to TCDD, suggestive of selective AhR modulation.
Collapse
Affiliation(s)
- Rance Nault
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | |
Collapse
|
15
|
Safe S. Endocrine disruptors and falling sperm counts: lessons learned or not! Asian J Androl 2012; 15:191-4. [PMID: 23001442 DOI: 10.1038/aja.2012.87] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA.
| |
Collapse
|
16
|
Brunnberg S, Andersson P, Poellinger L, Hanberg A. The constitutively active Ah receptor (CA-AhR) mouse as a model for dioxin exposure - effects in reproductive organs. CHEMOSPHERE 2011; 85:1701-1706. [PMID: 22014662 DOI: 10.1016/j.chemosphere.2011.09.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Revised: 08/25/2011] [Accepted: 09/22/2011] [Indexed: 05/31/2023]
Abstract
The dioxin/aryl hydrocarbon receptor (AhR) mediates most toxic effects of dioxins. In utero/lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) impairs fetal/neonatal development and the developing male reproductive tract are among the most sensitive tissues. TCDD causes antiestrogenic responses in rodent mammary gland and uterus and in human breast cancer cell lines in the presence of estrogen. Also, more recently an estrogen-like effect of TCDD/AhR has been suggested in the absence of estrogen. A transgenic mouse expressing a constitutively active AhR (CA-AhR) was developed as a model mimicking a situation of constant exposure to AhR agonists. Male and female reproductive tissues of CA-AhR mice were characterized for some of the effects commonly seen after dioxin exposure. Sexually mature CA-AhR female mice showed decreased uterus weight, while an uterotrophic assay in immature CA-AhR mice resulted in increased uterus weight. In immature mice, both TCDD-exposure and CA-AhR increased the expression of the estrogen receptor target gene Cathepsin D. When co-treated with 17β-estradiol no increase in Cathepsin D levels occurred in either TCDD-exposed or CA-AhR mice. In sexually mature male CA-AhR mice the weights of testis and ventral prostate were decreased and the epididymal sperm reserve was reduced. The results of the present study are in accordance with previous studies on dioxin-exposed rodents in that an activated AhR (here CA-AhR) leads to antiestrogenic effects in the presence of estrogen, but to estrogenic effects in the absence of estrogen. These results suggest the CA-AhR mouse model as a useful tool for studies of continuous low activity of the AhR from early development, resembling the human exposure situation.
Collapse
Affiliation(s)
- Sara Brunnberg
- Institute of Environmental Medicine, Karolinska Institutet, S-171 77 Stockholm, Sweden.
| | | | | | | |
Collapse
|
17
|
Beischlag TV, Luis Morales J, Hollingshead BD, Perdew GH. The aryl hydrocarbon receptor complex and the control of gene expression. Crit Rev Eukaryot Gene Expr 2008; 18:207-50. [PMID: 18540824 DOI: 10.1615/critreveukargeneexpr.v18.i3.20] [Citation(s) in RCA: 552] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that controls the expression of a diverse set of genes. The toxicity of the potent AhR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin is almost exclusively mediated through this receptor. However, the key alterations in gene expression that mediate toxicity are poorly understood. It has been established through characterization of AhR-null mice that the AhR has a required physiological function, yet how endogenous mediators regulate this orphan receptor remains to be established. A picture as to how the AhR/ARNT heterodimer actually mediates gene transcription is starting to emerge. The AhR/ARNT complex can alter transcription both by binding to its cognate response element and through tethering to other transcription factors. In addition, many of the coregulatory proteins necessary for AhR-mediated transcription have been identified. Cross talk between the estrogen receptor and the AhR at the promoter of target genes appears to be an important mode of regulation. Inflammatory signaling pathways and the AhR also appear to be another important site of cross talk at the level of transcription. A major focus of this review is to highlight experimental efforts to characterize nonclassical mechanisms of AhR-mediated modulation of gene transcription.
Collapse
Affiliation(s)
- Timothy V Beischlag
- Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | |
Collapse
|
18
|
Kortenkamp A. Ten years of mixing cocktails: a review of combination effects of endocrine-disrupting chemicals. ENVIRONMENTAL HEALTH PERSPECTIVES 2007; 115 Suppl 1:98-105. [PMID: 18174957 PMCID: PMC2174407 DOI: 10.1289/ehp.9357] [Citation(s) in RCA: 403] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Accepted: 10/04/2006] [Indexed: 05/17/2023]
Abstract
In the last 10 years, good evidence has become available to show that the combined effects of endocrine disruptors (EDs) belonging to the same category (e.g., estrogenic, antiandrogenic, or thyroid-disrupting agents) can be predicted by using dose addition. This is true for a variety of end points representing a wide range of organizational levels and biological complexity. Combinations of EDs are able to produce significant effect, even when each chemical is present at low doses that individually do not induce observable effects. However, comparatively little is known about mixtures composed of chemicals from different classes of EDs. Nevertheless, I argue that the accumulated evidence seriously undermines continuation with the customary chemical-by-chemical approach to risk assessment for EDs. Instead, we should seriously consider group-wise regulation of classes of EDs. Great care should be taken to define such classes by using suitable similarity criteria. Criteria should focus on common effects, rather than common mechanisms. In this review I also highlight research needs and identify the lack of information about exposure scenarios as a knowledge gap that seriously hampers progress with ED risk assessment. Future research should focus on investigating the effects of combinations of EDs from different categories, with considerable emphasis on elucidating mechanisms. This strategy may lead to better-defined criteria for grouping EDs for regulatory purposes. Also, steps should be taken to develop dedicated mixtures exposure assessment for EDs.
Collapse
|
19
|
Boverhof DR, Burgoon LD, Williams KJ, Zacharewski TR. Inhibition of estrogen-mediated uterine gene expression responses by dioxin. Mol Pharmacol 2007; 73:82-93. [PMID: 17942748 DOI: 10.1124/mol.107.040451] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) exhibits antiestrogenic properties, including the inhibition of estrogen-induced uterine growth and proliferation. The inhibition of estrogen-mediated gene expression through ER/AhR cross-talk has been proposed as a plausible mechanism; however, only a limited number of inhibited responses have been investigated that are unlikely to fully account for the antiuterotrophic effects of TCDD. Therefore, the effects of TCDD on ethynyl estradiol (EE)-mediated uterine gene expression were investigated using cDNA microarrays with complementary physiological and histological phenotypic anchoring. Mice were gavaged with vehicle, 3 daily doses of 10 mug/kg EE, a single dose of 30 mug/kg TCDD, or a combination of EE plus TCDD and sacrificed after 4, 12, 24, and 72 h. TCDD cotreatment inhibited EE-induced uterine wet weight by 37, 23, and 45% at 12, 24, and 72 h, respectively. TCDD cotreatment also reduced EE-mediated stromal edema, hypertrophy, and hyperplasia and induced marked luminal epithelial cell apoptosis. A 2 x 2 factorial microarray design was used to identify EE- and TCDD-specific differential gene expression responses as well as their interactive effects. Only 133 of the 2753 EE-mediated differentially expressed genes were significantly modulated by TCDD cotreatment, indicating a gene-specific inhibitory response. The EE-mediated induction of many genes, including trefoil factor 1 and keratin 14, were inhibited by greater than 90% by TCDD. Functional annotation of inhibited responses was associated with cell proliferation, water and ion transport, and maintenance of cellular structure and integrity. These inhibited responses correlate with the observed histological alterations and may contribute to the antiuterotrophic effects of TCDD.
Collapse
Affiliation(s)
- Darrell R Boverhof
- Michigan State University, Department of Biochemistry and Molecular Biology, 224 Biochemistry Building, Wilson Road, East Lansing, MI 48824-1319, USA
| | | | | | | |
Collapse
|
20
|
Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin administration and high-fat diet on the body weight and hepatic estrogen metabolism in female C3H/HeN mice. Toxicol Appl Pharmacol 2007; 226:107-18. [PMID: 17945325 DOI: 10.1016/j.taap.2007.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 08/23/2007] [Accepted: 08/23/2007] [Indexed: 12/23/2022]
Abstract
We studied the effect of administration of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) by i.p. injection once every 2 weeks in combination with a high-fat (HF) diet for 8 or 16 weeks on the body and organ weight changes as well as on the hepatic enzyme activity for estrogen metabolism in C3H/HeN female mice. Administration of TCDD at 100 microg/kg b.w. once every 2 weeks for 8 weeks increased the body weight by 46% in the HF diet-fed animals, but not in the regular diet-fed animals. This is the first observation suggesting that TCDD at a high dose (100 microg/kg b.w.), but not at lower doses (1 or 10 microg/kg b.w.), may have a strong obesity-inducing effect in C3H/HeN mice fed an HF diet. While TCDD increased liver weight and decreased thymus weight in animals, these effects were enhanced by feeding animals an HF diet. Metabolism studies showed that TCDD administration for 8 or 16 weeks increased the liver microsomal activity for the 2- and 4-hydroxylation of 17 beta-estradiol in animals fed a control diet, but surprisingly not in animals fed an HF diet. Treatment with TCDD dose-dependently increased the hepatic activity for the O-methylation of catechol estrogens in both control and HF diet-fed animals, and it also decreased the levels of liver microsomal sulfatase activity for hydrolysis of estrone-3-sulfate. TCDD did not significantly affect the hepatic enzyme activity for the glucuronidation or esterification of endogenous estrogens. It is suggested that enhanced metabolic inactivation of endogenous estrogens by hepatic estrogen-metabolizing enzymes in TCDD-treated, control diet-fed animals contributes importantly to the reduced incidence of estrogen-associated tumors in animals treated with TCDD.
Collapse
|
21
|
Ohtake F, Baba A, Takada I, Okada M, Iwasaki K, Miki H, Takahashi S, Kouzmenko A, Nohara K, Chiba T, Fujii-Kuriyama Y, Kato S. Dioxin receptor is a ligand-dependent E3 ubiquitin ligase. Nature 2007; 446:562-6. [PMID: 17392787 DOI: 10.1038/nature05683] [Citation(s) in RCA: 414] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Accepted: 02/16/2007] [Indexed: 12/12/2022]
Abstract
Fat-soluble ligands, including sex steroid hormones and environmental toxins, activate ligand-dependent DNA-sequence-specific transcriptional factors that transduce signals through target-gene-selective transcriptional regulation. However, the mechanisms of cellular perception of fat-soluble ligand signals through other target-selective systems remain unclear. The ubiquitin-proteasome system regulates selective protein degradation, in which the E3 ubiquitin ligases determine target specificity. Here we characterize a fat-soluble ligand-dependent ubiquitin ligase complex in human cell lines, in which dioxin receptor (AhR) is integrated as a component of a novel cullin 4B ubiquitin ligase complex, CUL4B(AhR). Complex assembly and ubiquitin ligase activity of CUL4B(AhR) in vitro and in vivo are dependent on the AhR ligand. In the CUL4B(AhR) complex, ligand-activated AhR acts as a substrate-specific adaptor component that targets sex steroid receptors for degradation. Thus, our findings uncover a function for AhR as an atypical component of the ubiquitin ligase complex and demonstrate a non-genomic signalling pathway in which fat-soluble ligands regulate target-protein-selective degradation through a ubiquitin ligase complex.
Collapse
Affiliation(s)
- Fumiaki Ohtake
- ERATO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Takahashi O, Oishi S, Yoneyama M, Ogata A, Kamimura H. Antiestrogenic effect of paradichlorobenzene in immature mice and rats. Arch Toxicol 2007; 81:505-17. [PMID: 17593412 DOI: 10.1007/s00204-007-0179-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Accepted: 01/09/2007] [Indexed: 10/23/2022]
Abstract
A significant increase/decrease in uterine and ovarian weights was occasionally seen in immature mice and rats subcutaneously administered paradichlorobenzene (PDCB) at doses of 22-67 mg/kg/day, but the results were not necessarily reproducible. PDCB at a dose of 800 mg/kg/day always reduced uterine and ovarian weights. Intraperitoneal PDCB at doses more than 400 mg/kg/day significantly inhibited the uterotrophic effect of beta-estradiol (E2) in CD-1 (ICR) mice. E2-induced uterotrophy was dose-dependently prevented by 204-400 mg PDCB/kg/day in C57BL/6N (Ah responsive) mice but not DBA/2N (Ah non-responsive) mice. While PDCB did not bind to estrogen receptor (ER(alpha)) up to 10(-3) M. Hepatic ethoxyresorufin-O-deethylase in adult female C57BL/6N mice was induced by i.p. administration of PDCB. Induction activity of PDCB may be 10(5)-10(6) times lower than that of 2,3,7,8-tetrachlorodibenzo-p-dioxin. These results suggest that PDCB is a weak antiestrogenic/antiuterotrophic compound possibly due to ER modulation through arylhydrocarbon receptor.
Collapse
Affiliation(s)
- Osamu Takahashi
- Department of Environmental Health and Toxicology, Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan.
| | | | | | | | | |
Collapse
|
23
|
Trewin AL, Woller MJ, Wimpee BAB, Conley LK, Baldridge MG, Hutz RJ. Short-Term Hormone Release from Adult Female Rat Hypothalamic and Pituitary Explants is not Altered by 2,3,7,8-Tetrachlorodibenzo-p-dioxin. J Reprod Dev 2007; 53:765-75. [PMID: 17325452 DOI: 10.1262/jrd.18101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
2, 3, 7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has adverse effects on reproduction, in part due to direct actions at the ovary. It is unclear whether effects are further mediated by glands that regulate ovarian function. We investigated whether effects of TCDD are mediated via the hypothalamic-pituitary axis. Hypothalamic and pituitary tissues were cultured in medium with and without TCDD. TCDD did not alter GnRH release from hypothalamic samples. It continued to be pulsatile with no differences in the average peak frequency, average peak amplitude, or baseline GnRH release. TCDD did not alter GnRH-induced release of gonadotropins from pituitary samples. There were no differences in average peak amplitude or baseline release. AhR, ARNT or ER alpha mRNA copy numbers in cultured pituitaries were not affected by TCDD. Our data suggest that TCDD effects on ovarian function are not mediated through the hypothalamic or pituitary release parameters tested in this study.
Collapse
Affiliation(s)
- Amanda Louise Trewin
- Department of Biology, University of Wisconsin-Platteville, Platteville, Wisconsin 53818, USA
| | | | | | | | | | | |
Collapse
|
24
|
Petersen SL, Krishnan S, Hudgens ED. The aryl hydrocarbon receptor pathway and sexual differentiation of neuroendocrine functions. Endocrinology 2006; 147:S33-42. [PMID: 16690800 DOI: 10.1210/en.2005-1157] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Historically, much of the research on health effects of environmental pollutants focused on ascertaining whether compounds were carcinogenic. More recent findings show that environmental contaminants also exert insidious effects by disrupting hormone action. Of particular concern are findings that developmental exposure to dioxins, chemicals that act through the aryl hydrocarbon receptor pathway, permanently alters sexually differentiated neural functions in animal models. In this review, we focus on mechanisms through which dioxins disrupt neuroendocrine development as exemplified by effects on a brain region critical for ovulation in rodents. We also provide evidence that dysregulation of GABAergic neural development may be a general mechanism underlying a broad spectrum of effects seen after perinatal dioxin exposure.
Collapse
Affiliation(s)
- Sandra L Petersen
- Department of Biology, University of Massachusetts-Amherst, Amherst, Massachusetts 01003 USA.
| | | | | |
Collapse
|
25
|
Hutz RJ, Carvan MJ, Baldridge MG, Conley LK, Heiden TK. Environmental toxicants and effects on female reproductive function. TRENDS IN REPRODUCTIVE BIOLOGY 2006; 2:1-11. [PMID: 18516253 PMCID: PMC2408384 DOI: 10.1901/jaba.2006.2-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the most toxic substances known to humans, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD or dioxin), is also highly pervasive in the environment. It is created naturally in volcanic eruptions and forest fires, and anthropogenically in waste incineration, chlorination processes and certain plastics manufacture. From reports of large industrial and other accidents, or from experimental studies, dioxin exposure has been correlated in animal models and/or humans with chloracne of the skin, organ cancers, hepatotoxicity, gonadal and immune changes, pulmonary and other diseases such as diabetes, skewing of the sex ratio, and infertility. We have demonstrated that the aromatic hydrocarbon receptor (AHR) that binds dioxin in tissues is localized in zebrafish, rat and rhesus monkey (Macaca mulatta) ovaries and in rat and human luteinizing granulosa cells (GC) (among other tissues), that labeled dioxin is specifically localized to granulosa cells of the ovarian follicle as observed by autoradiography, and that incubations of GC or ovarian fragments with environmentally relevant concentrations (fM to nM) of dioxin inhibit estradiol secretion significantly. Our experiments show that in human, non-human primate, rat, trout, and zebrafish ovarian tissues, dioxin inhibits estrogen synthesis at some level of the steroid biosynthetic pathway, most likely by inhibiting transcription of mRNAs for or activity of side-chain cleavage (Cyp11a1 gene) and/or aromatase (Cyp19a1 gene) enzymes, or conceivably other steroidogenic enzymes/factors. Such an untoward effect on estrogen synthesis in females exposed to dioxin environmentally may predispose them to defects in aspects of their fertility.
Collapse
Affiliation(s)
- R. J. Hutz
- Department of Biological Sciences and NIEHS Marine and Freshwater Biomedical Sciences Center and the Great Lakes Wisconsin Aquatic Technology and Environmental Research (WATER) Institute, University of Wisconsin-Milwaukee; the Medical College of Wisconsin; and the Wisconsin National Primate Research Center (WNPRC), USA
| | | | | | | | | |
Collapse
|
26
|
Safe S, Qin C, McDougal A. Development of selective aryl hydrocarbon receptor modulators for treatment of breast cancer. Expert Opin Investig Drugs 2005; 8:1385-96. [PMID: 15992156 DOI: 10.1517/13543784.8.9.1385] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
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.
Collapse
Affiliation(s)
- S Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466 USA.
| | | | | |
Collapse
|
27
|
Desaulniers D, Xiao GH, Leingartner K, Chu I, Musicki B, Tsang BK. Comparisons of brain, uterus, and liver mRNA expression for cytochrome p450s, DNA methyltransferase-1, and catechol-o-methyltransferase in prepubertal female Sprague-Dawley rats exposed to a mixture of aryl hydrocarbon receptor agonists. Toxicol Sci 2005; 86:175-84. [PMID: 15858227 DOI: 10.1093/toxsci/kfi178] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Non-ortho polychlorinated biphenyls (PCBs), polychlorinated dibenzodioxins (PCDDs), and polychlorinated dibenzofurans (PCDFs) are ubiquitous environmental contaminants that exert their toxicity mostly through activation of the aryl-hydrocarbon receptor (AhR), and are referred to as AhR agonists. The objective was to study, by real time reverse-transcriptase-polymerase chain reaction (RT-PCR), the effects of postnatal exposure to a reconstituted mixture of AhR agonists present in breast milk (3 non-ortho PCBs, 6 PCDDs, and 7 PCDFs, referred to here-in-after as AhRM) on mRNA expression of estrogen receptor (ERalpha), enzymes involved with the metabolism of estrogens [catechol-o-methyltransferase (Comt), cytochrome P450 (Cyp)1A1, 1B1 and 2B1], and DNA methyltransferase-1 (Dnmt1), in brain areas, liver and uterus of immature female rats. Neonates were exposed by gavage during postnatal day (PND) 1-20 with dosages equivalent to 1, 10, 100, and 1000 times the estimated average human exposure level, and were sacrificed at PND 21. None of the end points were affected in uterine cross-sections, or in samples of uterine tissue layers collected by laser capture microdissection. At 1000x, the AhRM reduced Dnmt1 mRNA abundance to 28% and 32% of control in the liver and hypothalamus, respectively. In the brain, Cyp1A1 was increased (409%) but ERalpha was reduced (66%). Similarly, mRNA abundance for Comt isoforms was reduced in the liver (45%) and brain areas (55-70%). AhRM at 100x, the lowest effective dose, exerted a 220% increase in brain cortex Comt [membrane bound (Mb)], a 219% increase in hepatic Cyp1B1, and a 63% decrease in hepatic Comt (soluble (S)+Mb). These results support the possibility that early exposure to environmental contaminants could lead to effects mediated by changes in DNA methylation and/or estrogen metabolism and signaling.
Collapse
Affiliation(s)
- D Desaulniers
- Health Canada, Healthy Environments and Consumer Safety Branch, Environmental & Occupational Toxicology Division, Ottawa, Ontario, Canada K1A 0L2.
| | | | | | | | | | | |
Collapse
|
28
|
Powers BE, Lin TM, Vanka A, Peterson RE, Juraska JM, Schantz SL. Tetrachlorodibenzo-p-dioxin exposure alters radial arm maze performance and hippocampal morphology in female AhR +/- mice. GENES BRAIN AND BEHAVIOR 2004; 4:51-9. [PMID: 15660668 DOI: 10.1111/j.1601-183x.2004.00098.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Perinatal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has been reported to alter spatial learning in rats tested on a radial arm maze (RAM). TCDD is believed to exert most of its effects through binding to the aryl hydrocarbon receptor (AhR). To determine whether the AhR mediates TCDD-induced alterations in spatial learning, we tested male and female AhR-knockout (AhR-/-), heterozygous (AhR+/-) and wild-type (AhR+/+) mice on the RAM. AhR+/- male and female mice were time mated, and treated dams were dosed with 5 microg TCDD/kg body weight on day 13 of gestation. When offspring reached adulthood, male and female AhR+/+, AhR+/- and AhR-/- mice from TCDD-exposed and unexposed litters were tested on the eight-arm RAM. After testing, we examined hippocampal morphology as visualized by the Timm's silver sulfide stain. TCDD-exposed female AhR+/- mice made more errors than their respective controls on the RAM and exhibited a decrease in the size of the intra- and infrapyramidal mossy fiber (IIP-MF) field of the hippocampus. None of the other TCDD-exposed groups differed from their respective control groups with regard to maze performance or hippocampal morphology. The reduction of IIP-MF field indicates a possible morphological basis for the learning deficit that was observed in the female AhR+/- mice. It is hypothesized that the effect of TCDD exposure is AhR dependent and that TCDD may alter GABAergic activity in the hippocampus of female mice during development.
Collapse
Affiliation(s)
- B E Powers
- Department of Veterinary Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | | | | | | | | | | |
Collapse
|
29
|
Thomsen JS, Kietz S, Ström A, Gustafsson JA. HES-1, a novel target gene for the aryl hydrocarbon receptor. Mol Pharmacol 2004; 65:165-71. [PMID: 14722248 DOI: 10.1124/mol.65.1.165] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Known mainly for its role as a toxin sensor, the aryl hydrocarbon receptor (AhR) complex is also involved in homeostasis regulation and differentiation processes and activated by xenobiotic compounds like 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Hairy and Enhancer of Split homolog-1 (HES-1) is a key regulator not only in differentiation, but also in the cell cycle, and we show here that HES-1 is a new target gene for AhR regulation. HES-1 is up-regulated by TCDD both at protein and mRNA levels in T47D human mammary carcinoma cells. Actinomycin D experiments have shown that the AhR-mediated up-regulation of HES-1 mRNA is caused by transcriptional activation of the HES-1 gene, and we have identified a functional AhR response element (XRE) at -48/-42 in the upstream regulatory region of human HES-1. The HES-1 protein down-regulates expression of its own gene, and the HES element overlaps the XRE. Our data indicate that HES-1 and the AhR complex compete for binding to the composite HES/XRE element. Also, we have previously shown that HES-1 is down-regulated by the estrogen receptor ligand 17beta-estradiol (E2). Up-regulation of HES-1 expression is correlated with suppression of cell proliferation, and the E2-mediated down-regulation of HES-1 therefore increases cell proliferation. It is known that TCDD exerts antiestrogenic action in breast tissue both in vivo and in vitro. Our observation that both the estrogen receptor and AhR signaling pathways regulate HES-1, but with opposing effects, suggests the existence of a new pathway by which AhR represses E2-signaling.
Collapse
Affiliation(s)
- Jane Sohn Thomsen
- Department of Biosciences at NOVUM, Karolinska Institutet, Huddinge, Sweden.
| | | | | | | |
Collapse
|
30
|
Abstract
In 1996, the US Congress directed the Environmental Protection Agency to produce screens and assays to detect estrogenic and other endocrine-disrupting chemicals in food and water. To date, there are none. Years have been wasted in attempts to utilize traditional toxicological approaches to solve the problem, when in retrospect, it is now apparent that the delay in part stems from the reluctance to attack the problem with entirely new approaches. To develop new testing protocols, it is necessary to set aside much of the dogma of toxicology and to begin again with open minds. A few pertinent examples are provided concerning what has been overlooked and what needs to be done. In particular, it is necessary to give close attention to the selection of animal strain and diet, factors that were only loosely controlled historically when one takes into consideration what has been learned in the last decade. Vast numbers of animals have been sacrificed, and more will be sacrificed, in futile attempts to validate assays and to develop safety standards unless knowledge gained over the past decade concerning the sensitivity and complexity of the endocrine system is taken into consideration.
Collapse
|
31
|
Foster WG, Agarwal SK. Environmental contaminants and dietary factors in endometriosis. Ann N Y Acad Sci 2002; 955:213-29; discussion 230-2, 396-406. [PMID: 11949949 DOI: 10.1111/j.1749-6632.2002.tb02782.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Endometriosis is an estrogen-dependent disease characterized by the presence of endometrial glands and stroma outside the uterine cavity. The etiology of this disease remains elusive, but is clearly influenced by genetic, immune, and endocrine factors. Exposure to environmental contaminants has recently been added to the list of potential factors that contribute to the pathogenesis of endometriosis. The objective of this paper is to review the weight of the evidence from hospital-based case-control studies and animal experiments for an association between exposure to environmental contaminants and endometriosis.
Collapse
Affiliation(s)
- Warren G Foster
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | | |
Collapse
|
32
|
Nilsson S, Mäkelä S, Treuter E, Tujague M, Thomsen J, Andersson G, Enmark E, Pettersson K, Warner M, Gustafsson JA. Mechanisms of estrogen action. Physiol Rev 2001; 81:1535-65. [PMID: 11581496 DOI: 10.1152/physrev.2001.81.4.1535] [Citation(s) in RCA: 1322] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Our appreciation of the physiological functions of estrogens and the mechanisms through which estrogens bring about these functions has changed during the past decade. Just as transgenic mice were produced in which estrogen receptors had been inactivated and we thought that we were about to understand the role of estrogen receptors in physiology and pathology, it was found that there was not one but two distinct and functional estrogen receptors, now called ER alpha and ER beta. Transgenic mice in which each of the receptors or both the receptors are inactive have revealed a much broader role for estrogens in the body than was previously thought. This decade also saw the description of a male patient who had no functional ER alpha and whose continued bone growth clearly revealed an important function of estrogen in men. The importance of estrogen in both males and females was also demonstrated in the laboratory in transgenic mice in which the aromatase gene was inactivated. Finally, crystal structures of the estrogen receptors with agonists and antagonists have revealed much about how ligand binding influences receptor conformation and how this conformation influences interaction of the receptor with coactivators or corepressors and hence determines cellular response to ligands.
Collapse
Affiliation(s)
- S Nilsson
- KaroBio AB and Department of Biosciences, Karolinska Institute, NOVUM, Huddinge, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
McDougal A, Gupta MS, Morrow D, Ramamoorthy K, Lee JE, Safe SH. Methyl-substituted diindolylmethanes as inhibitors of estrogen-induced growth of T47D cells and mammary tumors in rats. Breast Cancer Res Treat 2001; 66:147-57. [PMID: 11437101 DOI: 10.1023/a:1010608000074] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Diindolylmethane (DIM) is formed by acid catalyzed dimerization of the phytochemical indole-3-carbinol, and both compounds inhibit formation and/or growth of mammary tumors in rodents. In this study, we have investigated the aryl hydrocarbon receptor (AhR) agonist activity and inhibitory AhR-estrogen receptor crosstalk induced by the following methyl-substituted DIMs: 1,1'-dimethyl-, 2,2'-dimethyl-, 5,5'-dimethyl-, 6,6'-dimethyl-, and 7,7'-dimethylDIM and 1,1',2,2'-tetramethylDIM. The six compounds bound to the rat cytosolic AhR in a transformation assay but, at concentrations < or = 10 microM, exhibited minimal to non-detectable AhR agonist or antagonist activities associated with CYP1A1 induction. In contrast, the methyl-substituted DIMs inhibited estrogen-induced T47D human breast cancer cell growth and the four most active compounds (1,1'-, 2,2'-, 5,5'-dimethylDIM and 1,1',2,2'-tetramethylDIM) inhibited one or more estrogen-induced responses in the 21-day-old female B6C3F1 mice at a dose of 100 mg/kg/day (X3). Induction of hepatic CYP1A1-dependent activity was not observed at this high dose. The antitumorigenic activity of these compounds was examined in 7,12-dimethylbenz[a]anthracene-induced rat mammary tumor model in which the DIM analogs were orally administered (by gavage in corn oil) at a dose of 1 mg/kg/day (X10). 1,1'-DimethylDIM, 5,5'-dimethylDIM and 1,1',2,2'-tetramethylDIM significantly inhibited mammary tumor growth, and this was not accompanied by changes in organ/body weights or histopathology. These studies demonstrate that methyl-substituted DIMs are selective AhR modulators (SAhRMs) with potential for clinical treatment of breast cancer.
Collapse
Affiliation(s)
- A McDougal
- Department of Veterinary Physiology & Pharmacology, Texas A &M University, College Station 77843-4466, USA
| | | | | | | | | | | |
Collapse
|
34
|
Jana NR, Sarkar S, Ishizuka M, Yonemoto J, Tohyama C, Sone H. 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. MOLECULAR CELL BIOLOGY RESEARCH COMMUNICATIONS : MCBRC 2000; 4:174-80. [PMID: 11281733 DOI: 10.1006/mcbr.2001.0275] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
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.
Collapse
MESH Headings
- Azacitidine/pharmacology
- Cytochrome P-450 CYP1A1/genetics
- Cytochrome P-450 CYP1A1/metabolism
- Environmental Pollutants/pharmacology
- Enzyme Inhibitors/pharmacology
- Estradiol/pharmacology
- Estrogen Receptor alpha
- Estrogen Receptor beta
- Gene Expression Regulation
- Genes, Reporter
- Humans
- Plasminogen Activator Inhibitor 2/genetics
- Plasminogen Activator Inhibitor 2/metabolism
- Polychlorinated Dibenzodioxins/pharmacology
- Progesterone/pharmacology
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Recombinant Fusion Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Serine Proteinase Inhibitors/genetics
- Testosterone/pharmacology
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- N R Jana
- Chemical Exposure and Health Effects Research Team, National Institute for Environmental Studies, 16-2 Onogawa,Tsukuba, Ibaraki 305-0053, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Wormke M, Stoner M, Saville B, Safe S. Crosstalk between estrogen receptor alpha and the aryl hydrocarbon receptor in breast cancer cells involves unidirectional activation of proteasomes. FEBS Lett 2000; 478:109-12. [PMID: 10922479 DOI: 10.1016/s0014-5793(00)01830-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is an environmental toxin that activates the aryl hydrocarbon receptor (AhR) and disrupts multiple endocrine signaling pathways. T47D human breast cancer cells express a functional estrogen receptor alpha (ERalpha) and AhR, and treatment of these cells with 17beta-estradiol (E2) or TCDD resulted in a rapid proteasome-dependent decrease in immunoreactive ERalpha and AhR proteins (>60-80%), respectively. E2 did not affect the AhR, whereas TCDD induced proteasome-dependent degradation of both the AhR and ERalpha in T47D and MCF-7 human breast cancer cells, and these responses were specifically blocked by proteasome inhibitors. Thus, TCDD-induced degradation of ERalpha may contribute to the antiestrogenic activity of AhR agonists and this pathway may be involved in AhR-mediated disruption of other endocrine responses.
Collapse
Affiliation(s)
- M Wormke
- Department of Veterinary Physiology and Pharmacology, and Department of Biochemistry and Biophysics, Texas A&M University, 77843-4466, College Station, TX, USA
| | | | | | | |
Collapse
|
36
|
Klinge CM, Kaur K, Swanson HI. The aryl hydrocarbon receptor interacts with estrogen receptor alpha and orphan receptors COUP-TFI and ERRalpha1. Arch Biochem Biophys 2000; 373:163-74. [PMID: 10620335 DOI: 10.1006/abbi.1999.1552] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
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.
Collapse
Affiliation(s)
- C M Klinge
- Department of Biochemistry, University of Louisville School of Medicine, Louisville, Kentucky, 40292, USA.
| | | | | |
Collapse
|
37
|
Alvarez L, Randi A, Alvarez P, Piroli G, Chamson-Reig A, Lux-Lantos V, Kleiman de Pisarev D. Reproductive effects of hexachlorobenzene in female rats. J Appl Toxicol 2000; 20:81-7. [PMID: 10641019 DOI: 10.1002/(sici)1099-1263(200001/02)20:1<81::aid-jat629>3.0.co;2-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Hexachlorobenzene (HCB) is a polyhalogenated aromatic hydrocarbon widely distributed in the environment. In animal testing, HCB has been shown to be a reproductive toxin. Previous investigations of the effects of HCB on ovarian function have yielded equivocal results. Thus, the effects of chronic administration of HCB (1 g kg(-1) body wt.) on the ovary and pituitary hormone levels, hepatic and uterine oestradiol receptors, ovarian histopathological changes and oestrus cycle characteristics were investigated in spontaneously cycling rats. Our data demonstrate that HCB treatment, under the conditions of the present study, reduced circulating levels of oestradiol and prolactin without differences in serum concentrations of progesterone. Follicle-stimulating hormone serum levels were elevated. Hexachlorobenzene treatment resulted in irregularity of cycles, characterized mainly as prolonged periods of oestrus with a reduced number of ova recovered. In addition, HCB administration resulted in significantly decreased uterine nuclear oestrogen receptor levels. Histopathological examination revealed degenerative changes of the ovarian follicles and germinal epithelium and increased numbers of atresic follicles.
Collapse
Affiliation(s)
- L Alvarez
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
38
|
Klinge CM, Bowers JL, Kulakosky PC, Kamboj KK, Swanson HI. The aryl hydrocarbon receptor (AHR)/AHR nuclear translocator (ARNT) heterodimer interacts with naturally occurring estrogen response elements. Mol Cell Endocrinol 1999; 157:105-19. [PMID: 10619402 DOI: 10.1016/s0303-7207(99)00165-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
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.
Collapse
Affiliation(s)
- C M Klinge
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, KY 40292, USA.
| | | | | | | | | |
Collapse
|
39
|
Caruso JA, Laird DW, Batist G. Role of HSP90 in mediating cross-talk between the estrogen receptor and the Ah receptor signal transduction pathways. Biochem Pharmacol 1999; 58:1395-403. [PMID: 10513983 DOI: 10.1016/s0006-2952(99)00225-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tetrachlorodibenzo-p-dioxin (TCDD)-mediated gene transactivation via the Ah receptor (AhR) has been shown to be dependent upon estrogen receptor (ER) expression in human breast cancer cells. We have investigated the 90-kDa heat shock protein (HSP90) as a mediator of cross-talk between the AhR and the ER signal transduction pathways. The effect of HSP90 overexpression on receptor activity was determined by transient transfection assays using a HSP90 expression vector. Ligand-inducible gene expression was inhibited when the HSP90 expression vector was cotransfected with a TCDD-responsive reporter plasmid. However, overexpression of HSP90 did not block induction of an estrogen-responsive reporter plasmid. To determine whether ER facilitates AhR signaling through its ability to squelch HSP90, two vectors expressing protein products that bind HSP90 were transfected into MDA-MB-231 cells. Introduction of (i) He11, an ER deletion mutant that does not bind DNA, and (ii) the ligand-binding domain of human AhR, both led to increased basal and TCDD-inducible CYP1A1 expression. Finally, the subcellular distribution of HSP90 was investigated in human breast cancer cell lines. These studies showed HSP90 to be primarily cytoplasmic in ER-positive cell lines, whereas in matched ER-negative cell lines HSP90 was distributed equally between the cytoplasm and nucleus. Taken together, these results demonstrate that HSP90 can regulate AhR activity in vivo, and that Ah-responsiveness is dependent upon cellular ER content through a mechanism that involves HSP90.
Collapse
Affiliation(s)
- J A Caruso
- McGill Centre for Translational Research in Cancer, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
40
|
Kramer VJ, Giesy JP. Specific binding of hydroxylated polychlorinated biphenyl metabolites and other substances to bovine calf uterine estrogen receptor: structure-binding relationships. THE SCIENCE OF THE TOTAL ENVIRONMENT 1999; 233:141-61. [PMID: 10492903 DOI: 10.1016/s0048-9697(99)00221-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
The objectives of this research were: (1) to survey a wide variety of structurally diverse (and mostly chlorinated) aromatic chemicals for specific binding to the calf uterine estrogen receptor; (2) to develop a quantitative structure-binding relationship (QSBR) for hydroxylated polychlorinated biphenyls (OH-PCBs). This report specifically includes data on substances that did not exhibit specific binding to ER thereby exploring the structural requirements for specific binding to the estrogen receptor. Although several other QSBRs for OH-PCBs have been reported, this study presents data on a larger, environmentally relevant set of OH-PCBs than previously reported. Fifty three chemicals were tested for the ability to bind specifically to calf uterine estrogen receptor. All but three OH-PCBs bound specifically to calf uterine ER. For DDT compounds, receptor binding affinity followed the pattern: o,p'-DDT > o,p'-DDE > o,p'-DDD (Not active). Also exhibiting measurable affinity were 17 beta-estradiol (a positive control and the native ligand of the estrogen receptor), 2,4,6-trichlorobiphenyl and 4-chloro-2-isopropyl-5-methylphenol. Substances that did not bind to calf uterine estrogen receptor comprised several individual PCB congeners, chlorinated naphthalenes and naphthalenols, chlorinated bibenzyls, chlorinated phenols, and 9-chloro-retene. For 25 hydroxylated PCBs, a five parameter QSBR was developed using multiple linear regression and selection of the most parsimonius model from a total of seven molecular modeling parameters examined. The QSBR model predicted the ER binding log (IC50) to within one log unit.
Collapse
Affiliation(s)
- V J Kramer
- Fisheries and Wildlife Department, Michigan State University, East Lansing, USA.
| | | |
Collapse
|
41
|
Jana NR, Sarkar S, Ishizuka M, Yonemoto J, Tohyama C, Sone H. 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. Arch Biochem Biophys 1999; 368:31-9. [PMID: 10415108 DOI: 10.1006/abbi.1999.1288] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
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.
Collapse
MESH Headings
- Base Sequence
- Cytochrome P-450 CYP1A1/genetics
- DNA Primers/genetics
- Endometrial Neoplasms/genetics
- Endometrial Neoplasms/metabolism
- Estradiol/pharmacology
- Female
- Gene Expression/drug effects
- Humans
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Polychlorinated Dibenzodioxins/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Estradiol/genetics
- Receptors, Estradiol/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- N R Jana
- 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
| | | | | | | | | | | |
Collapse
|
42
|
Stephanou P, Pappas P, Vasiliou V, Marselos M. Prepubertal regulation of the rat dioxin-inducible aldehyde dehydrogenase (ALDH3). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1999; 463:143-50. [PMID: 10352679 DOI: 10.1007/978-1-4615-4735-8_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Affiliation(s)
- P Stephanou
- Department of Pharmacology, Medical School, University of Ioannina, Greece
| | | | | | | |
Collapse
|
43
|
Caruso JA, Batist G. Divergent mechanisms for loss of Ah-responsiveness in benzo[a]pyrene- and adriamycinR-resistant MCF-7 cells. Biochem Pharmacol 1999; 57:1253-63. [PMID: 10230769 DOI: 10.1016/s0006-2952(99)00041-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The intracellular aryl hydrocarbon receptor (AhR) mediates signal transduction by environmental pollutants such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and benzo[a]pyrene by functioning as a ligand-activated transcription factor. We have investigated AhR signaling in sublines of the human breast cancer cell line MCF-7 selected for resistance to AdriamycinR (AdrR) and benzo[a]pyrene (BP(R)). Previously we reported that AdrR cells have a loss of estrogen receptor (ER) expression and are Ah-nonresponsive. Here we show that AhR mRNA and protein are expressed at normal levels in AdrR cells, and the activated AhR complex is functionally capable of binding a xenobiotic responsive element. In MCF-7 cells AhR was depleted to 15% of normal levels after 4 hr TCDD treatment; however, 45% of AhR remained in AdrR cells during this time course. In BP(R) cells AhR mRNA levels were found to be decreased relative to wild-type cells, which led to decreased AhR protein levels and DNA-binding activity. Cellular ER content has been shown to correlate with Ah-responsiveness in human breast cancer cell lines. BP(R) cells were found to be ER-positive, although chronic (BP(R) cells) and acute (24 hr) exposure to benzo[a]pyrene led to significantly lower ER protein levels in MCF-7 cells. We conclude that loss of Ah-responsiveness occurs by different mechanisms in xenobiotic-resistant MCF-7 sublines: AhR mRNA is down-regulated in BP(R) cells, whereas AdrR cells are deficient in AhR signaling by a mechanism unrelated to AhR expression and activity.
Collapse
Affiliation(s)
- J A Caruso
- McGill Center for Translational Research in Cancer, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada
| | | |
Collapse
|
44
|
Jana NR, Sarkar S, Ishizuka M, Yonemoto J, Tohyama C, Sone H. Cross-talk between 2,3,7,8-tetrachlorodibenzo-p-dioxin and testosterone signal transduction pathways in LNCaP prostate cancer cells. Biochem Biophys Res Commun 1999; 256:462-8. [PMID: 10080920 DOI: 10.1006/bbrc.1999.0367] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
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.
Collapse
Affiliation(s)
- N R Jana
- Regional Environment Division, Environmental Health Sciences Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki, 305 0053, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Küchenhoff A, Seliger G, Klonisch T, Tscheudschilsuren G, Kaltwasser P, Seliger E, Buchmann J, Fischer B. Arylhydrocarbon receptor expression in the human endometrium. Fertil Steril 1999; 71:354-60. [PMID: 9988411 DOI: 10.1016/s0015-0282(98)00437-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To determine the expression and localization of the arylhydrocarbon (dioxin) receptor in human endometrium throughout the normal menstrual cycle. DESIGN Retrospective immunohistochemical and in situ hybridization study. SETTING Academic research unit. PATIENT(S) Premenopausal women (n = 86), aged 25 to 45 years, with histologically normal endometrium undergoing curettage or hysterectomy. INTERVENTION(S) Endometrial samples were collected from days 3 to 26 of the cycle by superficial scrapings of the uterine cavity or by hysterectomy. MAIN OUTCOME MEASURE(S) Expression of arylhydrocarbon receptor mRNA and protein. RESULT(S) Arylhydrocarbon receptor was expressed in 43% of the endometria studied and was correlated with the day of the cycle. The maximum of immunopositive endometria was found around the time of ovulation. Immunostaining decreased with increasing age of the patients. The receptor protein was localized exclusively in the apical part of the cytoplasm in the epithelial cells of the endometrial glands. In women positive for arylhydrocarbon receptor, arylhydrocarbon receptor mRNA was expressed in the cytoplasm of endometrial epithelial cells. CONCLUSION(S) Our results describe the expression of the arylhydrocarbon receptor in human endometrium and indicate a possible involvement of this transcription factor in endometrial function in women during the reproductive phase.
Collapse
Affiliation(s)
- A Küchenhoff
- Department of Anatomy and Cell Biology, Martin Luther University, Faculty of Medicine, Halle (Saale), Germany.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Reinhold J. HUTZ. Reproductive Endocrine Disruption by Environmental Xenobiotics that Modulate the Estrogen-Signaling Pathway, Particularly Tetrachlorodibenzo-p-dioxin (TCDD). J Reprod Dev 1999. [DOI: 10.1262/jrd.45.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- HUTZ Reinhold J.
- Department of Biological Sciences, NIEHS Marine and Freshwater Biomedical Sciences Center, University of Wisconsin-Milwaukee, Lapham Hall, Room 308, 3209 N. Maryland Avenue, Milwaukee, WI 53211-0413, USA
| |
Collapse
|
47
|
Ramamoorthy K, Gupta MS, Sun G, McDougal A, Safe SH. 3,3'4,4'-Tetrachlorobiphenyl exhibits antiestrogenic and antitumorigenic activity in the rodent uterus and mammary cells and in human breast cancer cells. Carcinogenesis 1999; 20:115-23. [PMID: 9934858 DOI: 10.1093/carcin/20.1.115] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
3,3',4,4'-Tetrachlorobiphenyl (tetraCB) binds to the aryl hydrocarbon receptor (AhR), and several reports have demonstrated that AhR agonists exhibit antiestrogenic and antitumorigenic activities in human breast cancer cells, the rodent uterus and breast. In contrast, a recent study showed that 3,3',4,4'-tetraCB bound the estrogen receptor (ER) and exhibited ER agonist activities, and we therefore have reinvestigated the estrogenic and antiestrogenic activities of 3,3',4,4'-tetraCB. Our results showed that 3,3',4,4'tetraCB and a structurally related analog, 3,3',4,4',5-pentaCB, did not bind the mouse uterine or human ER, did not induce proliferation of MCF-7 or T47D human breast cancer cells or induce reporter gene activity in cells transfected with E2-responsive constructs derived from the creatine kinase B (pCKB) or cathepsin D (pCD) gene promoters. Moreover, 3,3',4,4'-tetraCB and 3,3',4,4',5-pentaCB did not induce an increase in uterine wet weight, peroxidase activity or progesterone receptor binding in the 21-25-day-old female B6C3F1 mouse uterus. In contrast, both compounds inhibited 17beta-estradiol (E2)-induced cell proliferation and transactivation in MCF-7/T47D cells and uterine responses in B6C3F1 mice; surprisingly inhibition of E2-induced reporter gene activity was not observed in T47D cells transfected with pCKB, and this was observed as a cell-specific response with other AhR agonists. Additionally, 3,3',4,4'-tetraCB significantly inhibited mammary tumor growth in female Sprague-Dawley rats initiated with 7,12-dimethylbenzanthracene. Our results indicate that 3,3',4,4'-tetraCB does not exhibit ER agonist activity but exhibits a broad spectrum of antiestrogenic responses consistent with ligand-mediated AhR-ER crosstalk.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene
- Animals
- Anticarcinogenic Agents/pharmacology
- Anticarcinogenic Agents/therapeutic use
- Antineoplastic Agents, Hormonal/pharmacology
- Antineoplastic Agents, Hormonal/therapeutic use
- Binding, Competitive
- Breast Neoplasms/pathology
- Cell Division
- Estradiol/metabolism
- Estradiol/pharmacology
- Estrogen Antagonists/pharmacology
- Estrogen Antagonists/therapeutic use
- Estrogens
- Female
- Gene Expression Regulation/drug effects
- Genes, Reporter
- Humans
- Mammary Glands, Animal/drug effects
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Neoplasms, Hormone-Dependent/pathology
- Organ Size/drug effects
- Peroxidases/metabolism
- Polychlorinated Biphenyls/chemistry
- Polychlorinated Biphenyls/pharmacology
- Polychlorinated Biphenyls/therapeutic use
- Promegestone/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Estrogen/drug effects
- Receptors, Progesterone/drug effects
- Structure-Activity Relationship
- Transfection
- Tumor Cells, Cultured/drug effects
- Uterus/anatomy & histology
- Uterus/drug effects
- Uterus/enzymology
Collapse
Affiliation(s)
- K Ramamoorthy
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466, USA
| | | | | | | | | |
Collapse
|
48
|
Loeffler IK, Peterson RE. Interactive effects of TCDD and p,p'-DDE on male reproductive tract development in in utero and lactationally exposed rats. Toxicol Appl Pharmacol 1999; 154:28-39. [PMID: 9882589 DOI: 10.1006/taap.1998.8572] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The developing male rat reproductive system is highly sensitive to low doses of TCDD and p,p'-DDE (DDE), which exert antiandrogenic effects via different mechanisms. This study investigates the interactive effects of in utero and lactational exposure to a mixture of these compounds. Pregnant Holtzman rats received one of the following: vehicle on gestation day (GD) 14-18, 0.25 microgram/kg TCDD on GD15, 100 mg/kg DDE on GD 14-18, or 0.25 microgram/kg TCDD on GD15 and 100 mg/kg DDE on GD 14-18. Male offspring were euthanized on postnatal day (PND) 21 (weaning), PND 32 (prepuberty), PND 49 (puberty), and PND 63 (postpuberty). Coadministration of these doses of TCDD and DDE appeared to potentiate their individual actions on prostate weight on PND 21, while immunostaining for the prostatic androgen receptor exhibited patterns characteristic of the effects of both compounds individually. Cauda epididymal sperm number was reduced by each compound but was not further reduced by exposure to TCDD and DDE in combination. Anogenital distance, age at onset of puberty, daily sperm production, testicular and accessory sex organ weight (nonprostate), and levels of prostatic androgen-regulated gene transcripts are affected at higher doses of both compounds, but not at the doses used in the present study. Only DDE-treated animals retained nipples on PND 13. Serum androgen levels did not differ between treatment groups. In conclusion, the developing rat prostate is uniquely sensitive to the effects of TCDD and DDE, which may augment one another's effects in this organ.
Collapse
Affiliation(s)
- I K Loeffler
- School of Pharmacy and Environmental Toxicology Center, University of Wisconsin, Madison, Wisconsin, 53706, USA
| | | |
Collapse
|
49
|
Roman BL, Peterson RE. 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. Toxicol Appl Pharmacol 1998; 150:240-53. [PMID: 9653055 DOI: 10.1006/taap.1997.8362] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
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.
Collapse
Affiliation(s)
- B L Roman
- Environmental Toxicology Center, University of Wisconsin, Madison 53706, USA
| | | |
Collapse
|
50
|
Roman BL, Timms BG, Prins GS, Peterson RE. In utero and lactational exposure of the male rat to 2,3,7,8-tetrachlorodibenzo-p-dioxin impairs prostate development. 2. Effects on growth and cytodifferentiation. Toxicol Appl Pharmacol 1998; 150:254-70. [PMID: 9653056 DOI: 10.1006/taap.1998.8395] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the male Holtzman rat, in utero and lactational 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure decreases prostate weight without inhibiting testicular androgen production or decreasing circulating androgen concentrations. Therefore, the present study sought to characterize effects of TCDD exposure on prostate development, from very early outgrowth from the urogenital sinus (Gestation Day [GD] 20) until rapid growth and differentiation are essentially complete (Postnatal Day [PND] 32). Pregnant Holtzman rats were administered a single dose of TCDD (1.0 microgram/kg po) or vehicle on GD 15 and offspring were exposed via placental transfer (GD 20 euthanasia) or placental and subsequent lactational transfer until euthanasia (if before PND 21) or weaning. Results show that the prostatic epithelial budding process was impaired by in utero TCDD exposure, as evidence by significant decreases in the number of buds emerging from dorsal, lateral, and ventral aspects of the GD 20 urogenital sinus. Ventral prostate cell proliferation index was significantly decreased on PND 1 but was similar to or higher than control at later times, whereas apoptosis was an extremely rare event in ventral prostates from both control and TCDD-exposed animals. Delays were noted in the differentiation of pericordal smooth muscle cells and luminal epithelial cells. In addition, ventral prostates from approximately 40% of TCDD-exposed animals examined on PNDs 21 and 32 exhibited alterations in the histological arrangement of cell types that could not be explained by a developmental delay. Compared to controls, these ventral prostates exhibited a disorganized, hyperplastic epithelium containing fewer luminal epithelial cells and an increased density or continuous layer of basal epithelial cells, as well as thicker periductal smooth muscle sheaths. In addition, in ventral prostates from TCDD-exposed animals, the intensity of androgen receptor staining was relatively low in the central and distal epithelium, and the number of androgen receptor-positive cells was relatively high in the periductal stroma. These data suggest that in utero and lactational TCDD exposure interferes with prostate development by decreasing very early epithelial growth, delaying cytodifferentiation, and, in the most severely affected animals, producing alterations in epithelial and stromal cell histological arrangement and the spatial distribution of androgen receptor expression that may be of permanent consequence.
Collapse
Affiliation(s)
- B L Roman
- Environmental Toxicology Center, University of Wisconsin, Madison 53706, USA
| | | | | | | |
Collapse
|