1
|
Hokenson RE, Short AK, Chen Y, Pham AL, Adams ET, Bolton JL, Swarup V, Gall CM, Baram TZ. Unexpected Role of Physiological Estrogen in Acute Stress-Induced Memory Deficits. J Neurosci 2021; 41:648-662. [PMID: 33262247 PMCID: PMC7842761 DOI: 10.1523/jneurosci.2146-20.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 11/22/2022] Open
Abstract
Stress may promote emotional and cognitive disturbances, which differ by sex. Adverse outcomes, including memory disturbances, are typically observed following chronic stress, but are now being recognized also after short events, including mass shootings, assault, or natural disasters, events that consist of concurrent multiple acute stresses (MAS). Prior work has established profound and enduring effects of MAS on memory in males. Here we examined the effects of MAS on female mice and probed the role of hormonal fluctuations during the estrous cycle on MAS-induced memory problems and the underlying brain network and cellular mechanisms. Female mice were impacted by MAS in an estrous cycle-dependent manner: MAS impaired hippocampus-dependent spatial memory in early-proestrous mice, characterized by high levels of estradiol, whereas memory of mice stressed during estrus (low estradiol) was spared. As spatial memory requires an intact dorsal hippocampal CA1, we examined synaptic integrity in mice stressed at different cycle phases and found a congruence of dendritic spine density and spatial memory deficits, with reduced spine density only in mice stressed during high estradiol cycle phases. Assessing MAS-induced activation of brain networks interconnected with hippocampus, we identified differential estrous cycle-dependent activation of memory- and stress-related regions, including the amygdala. Network analyses of the cross-correlation of fos expression among these regions uncovered functional connectivity that differentiated impaired mice from those not impaired by MAS. In conclusion, the estrous cycle modulates the impact of MAS on spatial memory, and fluctuating physiological levels of sex hormones may contribute to this effect.SIGNIFICANCE STATEMENT: Effects of stress on brain functions, including memory, are profound and sex-dependent. Acute stressors occurring simultaneously result in spatial memory impairments in males, but effects on females are unknown. Here we identified estrous cycle-dependent effects of such stresses on memory in females. Surprisingly, females with higher physiological estradiol experienced stress-induced memory impairment and a loss of underlying synapses. Memory- and stress-responsive brain regions interconnected with hippocampus were differentially activated across high and low estradiol mice, and predicted memory impairment. Thus, at functional, network, and cellular levels, physiological estradiol influences the effects of stress on memory in females, providing insight into mechanisms of prominent sex differences in stress-related memory disorders, such as post-traumatic stress disorder.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christine M Gall
- Departments of Anatomy and Neurobiology
- Neurobiology and Behavior
| | - Tallie Z Baram
- Departments of Anatomy and Neurobiology
- Pediatrics
- Neurology, University of California-Irvine, Irvine, California 92697
| |
Collapse
|
2
|
Abstract
Sex, the states of being female or male, potentially interacts with all xenobiotic exposures, both inadvertent and deliberate, and influences their toxicokinetics (TK), toxicodynamics, and outcomes. Sex differences occur in behavior, exposure, anatomy, physiology, biochemistry, and genetics, accounting for female-male differences in responses to environmental chemicals, diet, and pharmaceuticals, including adverse drug reactions (ADRs). Often viewed as an annoying confounder, researchers have studied only one sex, adjusted for sex, or ignored it. Occupational epidemiology, the basis for understanding many toxic effects in humans, usually excluded women. Likewise, Food and Drug Administration rules excluded women of childbearing age from drug studies for many years. Aside from sex-specific organs, sex differences and sex × age interactions occur for a wide range of disease states as well as hormone-influenced conditions and drug distribution. Women have more ADRs than men; the classic sex hormone paradigm (gonadectomy and replacement) reveals significant interaction of sex and TK including absorption, distribution, metabolisms, and elimination. Studies should be designed to detect sex differences, describe the mechanisms, and interpret these in a broad social, clinical, and evolutionary context with phenomena that do not differ. Sex matters, but how much of a difference is needed to matter remains challenging.
Collapse
Affiliation(s)
- Michael Gochfeld
- Environmental and Occupational Health Sciences Institute and Consortium for Risk Evaluation with Stakeholder Participation at Rutgers—Robert Wood Johnson Medical School. Piscataway, New Jersey
| |
Collapse
|
3
|
Distinct roles for aryl hydrocarbon receptor nuclear translocator and ah receptor in estrogen-mediated signaling in human cancer cell lines. PLoS One 2012; 7:e29545. [PMID: 22235307 PMCID: PMC3250444 DOI: 10.1371/journal.pone.0029545] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 11/30/2011] [Indexed: 01/01/2023] Open
Abstract
The activated AHR/ARNT complex (AHRC) regulates the expression of target genes upon exposure to environmental contaminants such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Importantly, evidence has shown that TCDD represses estrogen receptor (ER) target gene activation through the AHRC. Our data indicates that AHR and ARNT act independently from each other at non-dioxin response element sites. Therefore, we sought to determine the specific functions of AHR and ARNT in estrogen-dependent signaling in human MCF7 breast cancer and human ECC-1 endometrial carcinoma cells. Knockdown of AHR with siRNA abrogates dioxin-inducible repression of estrogen-dependent gene transcription. Intriguingly, knockdown of ARNT does not effect TCDD-mediated repression of estrogen-regulated transcription, suggesting that AHR represses ER function independently of ARNT. This theory is supported by the ability of the selective AHR modulator 3′,4′-dimethoxy-α-naphthoflavone (DiMNF) to repress estrogen-inducible transcription. Furthermore, basal and estrogen-activated transcription of the genes encoding cathepsin-D and pS2 are down-regulated in MCF7 cells but up-regulated in ECC-1 cells in response to loss of ARNT. These responses are mirrored at the protein level with cathepsin-D. Furthermore, knock-down of ARNT led to opposite but corresponding changes in estrogen-stimulated proliferation in both MCF7 and ECC-1 cells. We have obtained experimental evidence demonstrating a dioxin-dependent repressor function for AHR and a dioxin-independent co-activator/co-repressor function for ARNT in estrogen signalling. These results provide us with further insight into the mechanisms of transcription factor crosstalk and putative therapeutic targets in estrogen-positive cancers.
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
Gochfeld M. Framework for gender differences in human and animal toxicology. ENVIRONMENTAL RESEARCH 2007; 104:4-21. [PMID: 16616135 DOI: 10.1016/j.envres.2005.12.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Accepted: 12/01/2005] [Indexed: 05/08/2023]
Abstract
Differences in exposure, anatomy, physiology, biochemistry, and behavior between males and females are a dominant theme in biology, transcending the plant and animal kingdoms. Yet differences due to sex and gender have not received adequate attention in human or animal toxicology nor always in epidemiology. Generalizations are often made about species' responses to xenobiotics, without data or consideration of female/male differences. Despite the leading role that pharmacology and drug development play in elucidating toxicokinetics, gender studies are relatively recent. Phenomenologic or clinical observations of sex differences often go unexplored, but pharmaceutical companies recognize the importance of enhanced understanding of toxicokinetics and toxicodynamics and emphasize the value of translational or integrational research--bringing laboratory findings to bedside applications and bedside questions to laboratory study. However, for many years Food and Drug Administration guidelines specifically precluded participation of females in many drug studies. Many occupational epidemiology studies, on which much of our understanding of toxic effects is based, begin by excluding women and minorities. Sex differentiation begins in the embryo under genetic and hormonal control. Changes affecting exposure, susceptibility, risk, and health continue throughout life. This paper provides a framework for analyzing the level(s) at which gender differences arise. The framework addresses exposure, toxicokinetics, toxicodynamics, and modulating influences. Men and women differ in many aspects of vulnerability to xenobiotics and other stressors, beginning with their opportunities for exposure. Toxicokinetic differences mainly involve metabolism, with few differences in absorption yet demonstrated. In addition, lifestyle, psychosocial, and hormonal factors modify the kinetics and responsiveness. Some phenomena fit the Classic Sex Hormone Paradigm in which castration (with and without hormone replacement) and administration of the opposite sex hormone demonstrate the primary regulatory role of sex hormones. Many phenomena, however, differ between males and females without showing a clear-cut relationship with the sex hormones. Since every cell both has a sex chromosome (X or Y) and is exposed to hormones, elegant techniques are just beginning to tease apart genetic from hormonal influences. Wherever possible, studies should use balanced gender and gender x age designs and should analyze data by sex and interactions, rather than simply adjusting for (discarding) gender. Power should be adequate, or lack of power (if inevitable) should be clearly stated.
Collapse
Affiliation(s)
- Michael Gochfeld
- Environmental and Occupational Health Sciences Institute, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
9
|
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
|
10
|
Dragin N, Dalton TP, Miller ML, Shertzer HG, Nebert DW. For dioxin-induced birth defects, mouse or human CYP1A2 in maternal liver protects whereas mouse CYP1A1 and CYP1B1 are inconsequential. J Biol Chem 2006; 281:18591-600. [PMID: 16636061 DOI: 10.1074/jbc.m601159200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin) induces cleft palate and hydronephrosis in mice, when exposed in utero; these effects are mediated by the aryl hydrocarbon receptor. The Cyp1a1, Cyp1a2, and Cyp1b1 genes are up-regulated by the aryl hydrocarbon receptor. To elucidate their roles in dioxin-induced teratogenesis, we compared Cyp1a1(-/-), Cyp1a2(-/-), and Cyp1b1(-/-) knock-out mice with Cyp1(+/+) wild-type mice. Dioxin was administered (25 microg/kg, gavage) on gestational day 10, and embryos were examined on gestational day 18. The incidence of cleft palate and hydronephrosis was not significantly different in fetuses from Cyp1a1(-/-), Cyp1b1(-/-), and Cyp1(+/+) wild-type mice. To fetuses carried by Cyp1a2(-/-) dams, however, this dose of dioxin was lethal; this effect was absolutely dependent on the maternal Cyp1a2 genotype and independent of the embryonic Cyp1a2 genotype. Dioxin levels were highest in adipose tissue, mammary gland, and circulating blood of Cyp1a2(-/-) mothers, compared with that in the Cyp1(+/+) mothers, who showed highest dioxin levels in liver. More dioxin reached the embryos from Cyp1a2(-/-) dams, compared with that from Cyp1(+/+) dams. Fetuses from Cyp1a2(-/-) dams exhibited a approximately 6-fold increased sensitivity to cleft palate, hydronephrosis, and lethality. Using the humanized hCYP1A1_1A2 transgenic mouse (expressing the human CYP1A1 and CYP1A2 genes in the absence of mouse Cyp1a2 gene), the teratogenic effects of dioxin reverted to the wild-type phenotype. These data indicate that maternal mouse hepatic CYP1A2, by sequestering dioxin and thus altering the pharmacokinetics, protects the embryos from toxicity and birth defects; substitution of the human CYP1A2 trans-gene provides the same protection. In contrast, neither CYP1A1 nor CYP1B1 appears to play a role in dioxin-mediated teratogenesis.
Collapse
Affiliation(s)
- Nadine Dragin
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, Ohio 45267-0056, USA
| | | | | | | | | |
Collapse
|
11
|
Vajda AM, Norris DO. Effects of steroids and dioxin (2,3,7,8-TCDD) on the developing wolffian ducts of the tiger salamander (Ambystoma tigrinum). Gen Comp Endocrinol 2005; 141:1-11. [PMID: 15707598 DOI: 10.1016/j.ygcen.2004.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Revised: 10/06/2004] [Accepted: 10/12/2004] [Indexed: 11/23/2022]
Abstract
This study was undertaken to investigate effects of the prototypical dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on steroid-dependent development of the wolffian ducts of an amphibian, the tiger salamander (Ambystoma tigrinum). Larvae with immature gonads and undeveloped mullerian ducts were injected with the steroid hormones estradiol (E2), dihydrotestosterone (DHT), or vehicle alone. Additionally, steroid-treated and vehicle-control larvae were immersed in sub-lethal solutions of technical grade TCDD (0, 0.0003, 0.003, 0.03, 0.3, and 3.0 microg TCDD/L). Both steroid treatments stimulated hypertrophy of the wolffian duct epithelium and an increase in mean epithelial cell size. Only DHT treatment stimulated epithelial cell proliferation. TCDD stimulated wolffian duct hypertrophy through an increase in mean epithelial cell size. TCDD acted as an androgen agonist on wolffian duct epithelial area and epithelial cell size. TCDD had no effect on wolffian duct epithelium among E2-injected animals. Stimulatory effects on cell size were observed at 0.0003 microg/L TCDD in saline-injected animals and at 0.003 microg/L TCDD in DHT-injected animals. Both E2 and DHT stimulated growth of the wolffian ducts early in development. Technical grade TCDD alone mimics E2 and DHT action but exhibits an androgen-agonistic action in the presence of exogenously administered DHT. Implications of possible interactions between TCDD and xenosteroids are discussed.
Collapse
Affiliation(s)
- Alan M Vajda
- Department of Integrative Physiology, Campus Box 354, University of Colorado, Boulder, CO 80309, USA.
| | | |
Collapse
|
12
|
Cunha GR, Cooke PS, Kurita T. Role of stromal-epithelial interactions in hormonal responses. ARCHIVES OF HISTOLOGY AND CYTOLOGY 2004; 67:417-34. [PMID: 15781983 DOI: 10.1679/aohc.67.417] [Citation(s) in RCA: 214] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Steroid sex hormones (17beta-estradiol, testosterone, dihydrotestosterone, and progesterone) and aryl hydrocarbons such as the dioxins regulate epithelial proliferation and secretory protein production and differentiation in their respective target organs in male and female urogenital tracts and mammary glands. Recent evidence has demonstrated that stromal-epithelial interactions are critical for mediating the effects of these molecules on epithelial cells. Our results have indicated that estradiol, testosterone, progesterone, and dioxin regulate epithelial proliferation (stimulation or inhibition) via paracrine mechanisms requiring the appropriate receptor in the stroma. The androgen receptor (AR), estrogen receptor alpha (ERalpha), progesterone receptor (PR), or aryl hydrocarbon receptor (AhR) in the epithelium are neither necessary nor sufficient for the regulation of epithelial proliferation. Moreover, during prostatic development, signaling through the stromal AR is required to induce prostatic epithelial identity, ductal morphogenesis and glandular differentiation. Epithelial functional differentiation is regulated in the prostate, uterus, and vagina via AR (prostate) and ERalpha(uterus and vagina). In these organs both epithelial and stromal steroid receptors are required for steroidal regulation of certain aspects of epithelial differentiation such as epithelial secretory protein production in the uterus and epithelial cornification in the vagina and prostate (squamous metaplasia). The mechanistic basis of these stromal-epithelial interactions is poorly understood, but growth factors appear to be mediators of these cell-cell interactions.
Collapse
Affiliation(s)
- Gerald R Cunha
- University of California, 3rd and Parnassus, Department of Anatomy, HSW 1323, San Francisco, USA.
| | | | | |
Collapse
|
13
|
Naruse M, Otsuka E, Naruse M, Ishihara Y, Miyagawa-Tomita S, Hagiwara H. Inhibition of osteoclast formation by 3-methylcholanthrene, a ligand for arylhydrocarbon receptor: suppression of osteoclast differentiation factor in osteogenic cells. Biochem Pharmacol 2004; 67:119-27. [PMID: 14667934 DOI: 10.1016/j.bcp.2003.08.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We investigated the effects of 3-methylcholanthrene (3MC), a ligand for arylhydrocarbon receptor (AhR), on osteoclastogenesis. Osteoclast-like cells, in cocultures with mouse spleen cells and clonal osteogenic stromal ST2 cells, are formed from spleen cells by a combination of the receptor activator of nuclear factor-kappaB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) produced by ST2 cells in response to 1alpha,25(OH)(2) Vitamin D(3). 3MC dose-dependently inhibited the formation of mono- and multinuclear osteoclast-like cells. However, 3MC did not inhibit the formation of osteoclast-like cells from mouse spleen cells which was supported by the exogenous soluble RANKL and M-CSF. 3MC did not affect the formation of an actin ring and pits on slices of dentine by osteoclast-like cells, both of which are typical indices of osteoclast activity. These results suggest that 3MC affects osteoclast-supporting cells such as ST2 cells but not osteoclast precursor cells and mature osteoclastic cells. When we measured the expression levels of RANKL mRNA in ST2 cells, 3MC dose-dependently decreased the level of this mRNA. However, 3MC did not affect levels of mRNAs for osteoprotegerin (OPG), M-CSF, and the receptor of 1alpha,25(OH)(2) Vitamin D(3) in ST2 cells. Furthermore, soluble RANKL was able to counteract the inhibitory effect of 3MC on the formation of osteoclast-like cells. Our findings indicate that 3MC inhibits osteoclastogenesis via the inhibition of RANKL expression in osteoblastic cells.
Collapse
Affiliation(s)
- M Naruse
- Department of Biological Sciences, Tokyo Institute of Technology, 226-8501, Yokohama, Japan
| | | | | | | | | | | |
Collapse
|
14
|
Naruse M, Ishihara Y, Miyagawa-Tomita S, Koyama A, Hagiwara H. 3-Methylcholanthrene, which binds to the arylhydrocarbon receptor, inhibits proliferation and differentiation of osteoblasts in vitro and ossification in vivo. Endocrinology 2002; 143:3575-81. [PMID: 12193573 DOI: 10.1210/en.2002-220003] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
3-Methylcholanthrene (3MC) is a ligand for arylhydrocarbon receptor (AhR), which binds dioxin. We examined the effects of 3MC on the proliferation and differentiation of osteoblasts using cultures of rat calvarial osteoblast-like cells (ROB cells) and mouse calvarial clonal preosteoblastic cells (MC3T3-E1 cells). Analysis by RT-PCR revealed that the mRNAs for AhR and AhR nuclear translocators were expressed in both ROB and MC3T3-E1 cells. Cell proliferation and the synthesis of DNA by ROB cells and MC3T3-E1 cells were markedly inhibited on exposure of cells to 3MC. Furthermore, 3MC reduced the activity of alkaline phosphatase and the rate of deposition of calcium by cells. The level of expression of mRNA for osteocalcin, which is a marker of osteoblastic differentiation, was also depressed by 3MC. Moreover, when 3MC (1 mg/kg body weight) was administered sc to pregnant mice at 10.5, 12.5, and 14.5 d post coitus, fetuses examined subsequently at 15.5 or 17.5 d post coitus revealed evidence of inhibition of appropriate calcification of bones. The treated metacarpals showed no subperiosteal bone matrix histologically. Our findings indicate that 3MC might have critical effects on the formation of bone both in vivo and in vitro.
Collapse
Affiliation(s)
- Masae Naruse
- Department of Biological Sciences, Tokyo Institute of Technology, Tokyo 226-8501, Japan
| | | | | | | | | |
Collapse
|
15
|
Abstract
To characterize the action of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) during both the follicular and luteal phases of the ovarian cycle, the direct effect of TCDD was investigated in vitro using a system of primary monolayer cell culture. Granulosa and theca cells were collected from the preovulatory follicles and cultured as a co-culture, thus resembling follicles in vivo. Luteal cells were isolated from the corpora lutea collected during the midluteal phase. In both cases cells were isolated from the ovaries of animals exhibiting natural estrus cycle. Results of these experiments suggest that TCDD decreases estradiol secretion by follicular cells and progesterone secretion by luteal cells in a dose-dependent manner. It was also shown that TCDD disrupts steroidogenesis through its influence on the activity of enzymes involved in the steroid biosynthesis cascade. In luteal cells, its action is mediated via the aryl hydrocarbon receptor (AhR) and is probably independent of estrogen receptor (ER) stimulation. Endocrine disruptors that interfere with estradiol production in the follicles can act as ovulatory disruptors, and while interfering with progesterone production by luteal cells they can act as abortifacients.
Collapse
Affiliation(s)
- Ewa L Gregoraszczuk
- Laboratory of Reproductive Physiology and Toxicology of Domestic Animals, Department of Animal Physiology, Institute of Zoology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Petroff BK, Gao X, Rozman KK, Terranova PF. The effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on weight gain and hepatic ethoxyresorufin-o-deethylase (EROD) induction vary with ovarian hormonal status in the immature gonadotropin-primed rat model. Reprod Toxicol 2001; 15:269-74. [PMID: 11390171 DOI: 10.1016/s0890-6238(01)00132-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Immature female rats received 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) during an induced proestrus or diestrus. The inhibitory effect of TCDD on acute weight gain and the induction of hepatic ethoxyresorufin-o-deethylase (EROD) activity by TCDD were greatest during proestrus. In a second experiment, ovariectomized rats received estradiol cypionate (ECP) or progesterone followed by TCDD. TCDD and estradiol each alone significantly inhibited weight gain. Progesterone potentiated the effects of TCDD on weight gain. The highest dose of ECP was associated with greater induction of hepatic EROD activity by TCDD than seen with TCDD alone. Estradiol modulates the induction of hepatic EROD activity by TCDD. Differential effects of TCDD on acute weight gain during proestrus vs. diestrus in this model do not mimic changes induced by estrogen alone. Hepatic responses to TCDD may vary according to phase of the female reproductive cycle.
Collapse
Affiliation(s)
- B K Petroff
- Center for Reproductive Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
19
|
Petroff BK, Roby KF, Gao X, Son D, Williams S, Johnson D, Rozman KK, Terranova PF. A review of mechanisms controlling ovulation with implications for the anovulatory effects of polychlorinated dibenzo-p-dioxins in rodents. Toxicology 2001; 158:91-107. [PMID: 11275352 DOI: 10.1016/s0300-483x(00)00367-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Polychlorinated dibenzo-p-dioxins (PCDDs) can impinge on female fertility by preventing ovulation. In this review, the aspects of normal ovulatory physiology most relevant to our current understanding of PCDD action on the ovary are briefly reviewed. This is followed by a comprehensive assessment of data relevant to the effects of PCDDs during ovulation in the rat. PCDDs interrupt ovulation through direct effects on the ovary in combination with dysfunction of the hypothalamo-hypophyseal axis.
Collapse
Affiliation(s)
- B K Petroff
- Center for Reproductive Sciences, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160-7417, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Petroff BK, Gao X, Rozman KK, Terranova PF. Interaction of estradiol and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in an ovulation model: evidence for systemic potentiation and local ovarian effects. Reprod Toxicol 2000; 14:247-55. [PMID: 10838126 DOI: 10.1016/s0890-6238(00)00075-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Immature rats were treated with estradiol cypionate, (ECP, 0, 0.1, 1, or 2 mg/kg s.c.) followed 24 h later by TCDD (0 or 10 microg/kg orally). Follicular development was induced with eCG [5 or 10 IU subcutaneously (s.c.)] followed by an ovulatory dose of hCG (10 IU s. c.). Inhibition of ovulation by TCDD was potentiated by ECP in hypophysectomized but not intact rats. Only hypophysectomized rats exposed systemically to TCDD and ECP exhibited weight loss. Pair feeding mimicked the combined effects of TCDD and ECP in hypophysectomized rats. In another experiment, intact rats received ECP s.c. (0 or 2 mg/kg) and TCDD into the ovarian bursa (0 or 250 ng). Another group of intact rats received TCDD orally (10 microg/kg) and ECP into the ovarian bursa (0 or 1.5 microg). Blockade of ovulation by systemic or local TCDD was alleviated by ECP pretreatment. Estrogen increased the systemic toxicity of TCDD in rats whereas antagonizing its direct ovarian effects.
Collapse
Affiliation(s)
- B K Petroff
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, G011 Lied Building, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
21
|
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
|
22
|
Ricci MS, Toscano DG, Mattingly CJ, Toscano WA. Estrogen receptor reduces CYP1A1 induction in cultured human endometrial cells. J Biol Chem 1999; 274:3430-8. [PMID: 9920887 DOI: 10.1074/jbc.274.6.3430] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) exerts its toxic action via the aryl hydrocarbon (Ah) receptor, which induces a battery of xenobiotic-metabolizing enzymes, including the cytochrome P450 isozyme, CYP1A1. TCDD-induced 7-ethoxycoumarin-O-deethylase activity was reduced 75% in cultured human endometrial ECC-1 cells exposed to various concentrations of 17beta-estradiol for up to 72 h, with a half-maximal effective concentration (EC50) of 0.9 nM. Reduced enzyme activity was correlated with decreased CYP1A1 mRNA levels, and transcription. Exposure to TCDD plus 17beta-estradiol also reduced CYP1A1 activity in MCF-7 breast cancer cells but not in Hep-3B human liver cells or HuE primary human keratinocytes, suggesting that the effect was specific to estrogen-regulated cells. Estrogen receptor antagonists 4-hydroxytamoxifen and 7alpha-[9-(4,4, 5,5,5-pentafluoro-pentylsulfinyl)nonyl]estra-1,3,5(10)-tr iene3, 17beta-diol restored TCDD-induced CYP1A1 transcription, steady-state mRNA levels, and enzymatic activity in ECC-1 cells. Gel mobility shift assay showed that 17beta-estradiol had little effect on Ah receptor binding to its DNA-responsive element. 17beta-Estradiol did not alter the induction of another Ah receptor-regulated gene, CYP1B1, suggesting that altered Ah receptor binding to DNA does not mediate reduced CYP1A1 transcription. Transfecting ECC-1 cells with a general transcription factor involved in CYP1A1 induction, nuclear factor-1, reversed 17beta-estradiol antagonism of dioxin induced-CYP1A1. The data suggest that 17beta-estradiol reduced CYP1A1 expression at the transcriptional level by squelching available nuclear factor-1, a transcription factor that interacts with both Ah and estrogen receptors.
Collapse
Affiliation(s)
- M S Ricci
- Department of Environmental Health Sciences and Center for Bioenvironmental Research, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana 70112-2699, USA
| | | | | | | |
Collapse
|
23
|
Wilker C. Are there redeeming qualities in one of nature's toxic orphan receptors? Toxicol Pathol 1998; 26:672-3. [PMID: 9789954 DOI: 10.1177/019262339802600511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- C Wilker
- Searle, Skokie, Illinois 60077, USA
| |
Collapse
|
24
|
Enan E, El-Sabeawy F, Moran F, Overstreet J, Lasley B. Interruption of estradiol signal transduction by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) through disruption of the protein phosphorylation pathway in adipose tissues from immature and mature female rats. Biochem Pharmacol 1998; 55:1077-90. [PMID: 9605431 DOI: 10.1016/s0006-2952(97)00683-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
At doses of 10-115 microg/kg, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) decreased body and adipose tissue weights of mature female rats. Doses below 10 microg TCDD/kg decreased body and adipose tissue weights of immature, but not mature females. Doses of 2 and 10 microg TCDD/kg decreased adipose tissue epidermal growth factor receptor (EGFR) binding activity 5 and 7 days later in immature and mature females, respectively. At these times, there was a decrease in the activities of tyrosine kinase (TK), mitogen-activated protein kinase (MAP2K), and protein kinase A (PKA). In mature females, estradiol (E2, 15 microg/kg) increased TK and PKA activities and decreased MAP2K activity. In immature females, E2 decreased TK and PKA activities but not MAP2K activity. TCDD abolished the stimulatory effect of E2 on TK and PKA in mature females, and in immature females TCDD potentiated the negative effect of E2 on all three kinases. TCDD decreased binding of [3H]E2 to cytosolic and nuclear estrogen receptors (ERs) of mature and immature females, and antagonized the stimulatory effect of E2 on ER binding activity. E2 increased DNA binding activity of the estrogen response element (ERE) and activator protein-1, and TCDD antagonized this effect. Geldanamycin, an inhibitor of Src tyrosine kinase, reduced the effects of TCDD on body and adipose tissue weights. Geldanamycin antagonized the effects of TCDD on EGFR binding activity and TK activity. In cell-free preparations, TCDD antagonized E2 action on TK activity in mature females, as well as E2 action on PKA activity in immature females. We hypothesize that TCDD antagonizes E2 action in female adipose tissues through disruption of common cytosolic signal transduction pathways.
Collapse
Affiliation(s)
- E Enan
- Department of Environmental Toxicology, and Institute of Toxicology and Environmental Health, University of California, Davis, USA.
| | | | | | | | | |
Collapse
|
25
|
Navas JM, Segner H. Antiestrogenic activity of anthropogenic and natural chemicals. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 1998; 5:75-82. [PMID: 19005814 DOI: 10.1007/bf02986390] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/1997] [Accepted: 10/05/1997] [Indexed: 05/27/2023]
Abstract
A number of natural and man-made chemicals possess antiestrogenic activity, i.e. they antagonize a broad spectrum of estrogen-induced responses in vertebrates. Examples of antiestrogens include dioxin, furan and PCB congeners, certain PAHs, pesticides and indol-3-carbinol derivatives. Major mechanisms of anti-estrogenicity are antagonistic action of chemicals at the estrogen receptor, or binding of chemicals to the arylhydrocarbon (Ah) receptor and subsequent interaction with estrogen-responsive genes. Toxicological consequences resulting from antiestrogenic activity have not been conclusively demonstrated to date, although antiestrogenic compounds could critically affect sensitive reproductive and developmental processes.
Collapse
Affiliation(s)
- J M Navas
- Department of Chemical Ecotoxicology, UFZ Centre for Environmental Research, Permoserstr. 15, D-04318, Leipzig, Germany
| | | |
Collapse
|
26
|
Drenth HJ, Bouwman CA, Seinen W, Van den Berg M. Effects of some persistent halogenated environmental contaminants on aromatase (CYP19) activity in the human choriocarcinoma cell line JEG-3. Toxicol Appl Pharmacol 1998; 148:50-5. [PMID: 9465263 DOI: 10.1006/taap.1997.8307] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 3,3',4,4',5-pentachlorobiphenyl (PCB126), a technical PCB mixture (Aroclor 1016), and a technical toxaphene mixture (Camphechlor) on aromatase (CYP19) activity were investigated in human choriocarcinoma JEG-3 cells. After 18 h incubation with TCDD, PCB126, Aroclor 1016 or toxaphene, ethoxyresorufin-O-deethylase (EROD), and aromatase activity were determined. To exclude serum effects, incubations were carried out with or without fetal calf serum in the medium. EROD activity was induced by both TCDD and PCB126 in the presence or absence of serum, which indicates that JEG-3 cells are responsive toward dioxin-like chemicals. Neither Aroclor 1016 nor toxaphene affected EROD activity in these cells. Calculated EC50 values for induction of EROD activity were 0.71 and 0.40 nM for TCDD, and 48 and 20 nM for PCB126 in presence or absence of serum, respectively. Incubation with TCDD or PCB126 with or without serum caused a concentration-dependent decrease in the aromatase activity of up to 4.9-fold. Calculated EC50 values for this effect were 52 pM and 13 nM for TCDD, and 75 and 48 nM for PCB126 in the presence and absence of serum, respectively. Aroclor 1016 and toxaphene had no effect on aromatase activity at concentrations up to 1.0 microM for Aroclor 1016 or 3.0 microM for toxaphene. These results show that aromatase activity can be decreased in a concentration dependent way within the same range where EROD activity is increased. In view of these results, possible effects of dioxin-like compounds on estrogen producing and androgen target cells should be studied in more detail.
Collapse
Affiliation(s)
- H J Drenth
- Research Institute of Toxicology, University of Utrecht, The Netherlands
| | | | | | | |
Collapse
|
27
|
Kharat I, Saatcioglu F. Antiestrogenic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin are mediated by direct transcriptional interference with the liganded estrogen receptor. Cross-talk between aryl hydrocarbon- and estrogen-mediated signaling. J Biol Chem 1996; 271:10533-7. [PMID: 8631852 DOI: 10.1074/jbc.271.18.10533] [Citation(s) in RCA: 173] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Aryl hydrocarbon receptor (AhR) ligands have diverse biological effects including striking antiestrogenic activity. We have investigated at the molecular level the antiestrogenic activity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). We show that the previously documented TCDD-mediated decrease in estradiol-inducible gene products such as cathepsin D (cat D) is due to a sharp decline in mRNA accumulation despite any change in estrogen receptor (ER) mRNA levels. The decline in cat D mRNA level is most likely due to a decrease in transcription of the cat D gene since TCDD blocks the ability of ER to transactivate from an estrogen response element. AhR is required for this activity as TCDD is no longer antiestrogenic in a mutant cell line that is deficient in functional AhR. We provide evidence that the loss of transactivation potential by ER in the presence of TCDD is due to a sharp decrease in its ability to bind to an estrogen response element. Reciprocally, estradiol treatment blocked TCDD-induced accumulation of CYP1A1 mRNA and AhR-mediated activation of the CYP1A1 promoter. This is due to the ability of liganded ER to interfere with the binding of AhR to the xenobiotic response element. These results provide a molecular mechanism for the antiestrogenic effects of TCDD and demonstrate the presence of a two-way crosstalk between the intracellular signaling pathways involving estrogens and aryl hydrocarbons.
Collapse
Affiliation(s)
- I Kharat
- Molecular Biology Laboratory, Maharishi International University, Fairfield, Iowa 52557, USA
| | | |
Collapse
|
28
|
Gierthy JF, Spink BC, Figge HL, Pentecost BT, Spink DC. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin, 12-O-tetradecanoylphorbol-13-acetate and 17 beta-estradiol on estrogen receptor regulation in MCF-7 human breast cancer cells. J Cell Biochem 1996; 60:173-84. [PMID: 8655628 DOI: 10.1002/(sici)1097-4644(19960201)60:2<173::aid-jcb2>3.0.co;2-u] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) exhibits remarkably potent antiestrogenic activity. To further elucidate the role of estrogen receptor (ER) regulation in this response, we examined the effects of exposure to TCDD in MCF-7 human breast cancer cells on ER mRNA levels by using an RNase protection assay, on ER accumulation by using an ER immunocytochemical essay (ER-ICA), and on ER function by competitive binding assays under conditions of saturating 17 beta-estradiol (E2). Comparative studies were conducted with E2 and 12-O-tetradecanoylphorbol-13-acetate (TPA), as both compounds are known to suppress ER expression. Our results indicate that 1 nM E2 and 100 nM TPA both suppress ER mRNA levels as early as 4 h after exposure and to 33.6% and 16.5% of control levels, respectively, after 72 h. In contrast, no significant effect on ER mRNA levels was attributed to exposure to 10 nM TCDD. A greater than 50% reduction in positive staining was observed by ER-ICA after 72 h exposure to 1 nM E2 and to 100 nM TPA, while only an 11% reduction in positive staining was observed with 10 nM TCDD. Specific binding of [3H]E2 under saturating conditions (10 nM E2) in whole cells was reduced by 50% in cultures exposed to 100 nM TPA, although no effect on binding was observed with exposure to 10 nM TCDD. In contrast, specific binding using subsaturating 1 nM [3H]E2 was depressed by 49% in MCF-7 cells exposed to 10 nM TCDD for 72 h. This depression was inhibited by a 1-h treatment with 5 microM alpha-naphthoflavone, which inhibits TCDD-induced, P450-mediated, E2 metabolism, and subsequent E2 depletion. In conclusion, while TPA and E2 effectively down-regulate ER expression, TCDD, under antiestrogenic conditions, has little if any effect on total ER levels in MCF-7 cells, and thus ER modulation is probably not necessary for the suppression of estrogenic activity in MCF-7 cells by TCDD.
Collapse
Affiliation(s)
- J F Gierthy
- Wadsworth Center, New York State Department of Health, Albany 12201-0509, USA
| | | | | | | | | |
Collapse
|
29
|
Krishnan V, Porter W, Santostefano M, Wang X, Safe S. Molecular mechanism of inhibition of estrogen-induced cathepsin D gene expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in MCF-7 cells. Mol Cell Biol 1995; 15:6710-9. [PMID: 8524236 PMCID: PMC230924 DOI: 10.1128/mcb.15.12.6710] [Citation(s) in RCA: 159] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
17 beta-Estradiol (E2) induces cathepsin D mRNA levels and intracellular levels of immunoreactive protein in MCF-7 human breast cancer cells. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) alone does not affect cathepsin D gene expression in this cell line; however, in cells cotreated with TCDD and E2, TCDD inhibited E2-induced cathepsin D mRNA levels, the rate of gene transcription, and levels of immunoreactive protein. The inhibitory responses were observed within 30 to 120 min after the cells were treated with TCDD. TCDD also inhibited E2-induced secreted alkaline phosphatase activity in aryl hydrocarbon (Ah)-responsive MCF-7 and wild-type mouse Hepa 1c1c7 cells cotransfected with the human estrogen receptor (hER) and the pBC12/S1/pac plasmid, which contains the 5' promoter region (-296/+57) of the cathepsin D gene and an alkaline phosphatase reporter gene. The E2-responsive ER/Sp1 sequence (-199 to -165) in the cathepsin D 5' region contains an imperfect GTGCGTG (-175/-181) xenobiotic responsive element (XRE); the role of this sequence in Ah responsiveness was investigated in gel electrophoretic mobility shift assays and with plasmid constructs containing a wild-type ER/Sp1 oligonucleotide or a mutant ER/Sp1-"XRE" oligonucleotide containing two C-->A mutations in the XRE sequence (antisense strand). In plasmid constructs which contained a chloramphenicol acetyltransferase reporter gene and the wild-type ER/Sp1 promoter sequence, E2-induced chloramphenicol acetyltransferase activity and mRNA levels were inhibited by TCDD whereas no inhibition was observed with the mutant ER/Sp1-"XRE" plasmids. Electrophoretic mobility shift assays showed that the nuclear or transformed cytosolic Ah receptor complex blocked formation of the ER-Sp1 complex with the wild-type but not the ER/Sp1 mutant oligonucleotide. Moreover, incubation of the wild-type bromodeoxyuridine-substituted ER/Sp1 oligonucleotide with the nuclear Ah receptor complex gave a specifically bound cross-linked 200-kDa band. These data demonstrate that Ah receptor-mediated inhibition of E2-induced cathepsin D gene expression is due to disruption of the ER-Sp1 complex by targeted interaction with an overlapping XRE.
Collapse
MESH Headings
- Alkaline Phosphatase/biosynthesis
- Animals
- Base Sequence
- Binding Sites
- Blotting, Northern
- Breast Neoplasms
- Cathepsin D/biosynthesis
- Cell Line
- Cell Nucleus/metabolism
- Chloramphenicol O-Acetyltransferase/biosynthesis
- Cloning, Molecular
- Estradiol/pharmacology
- Estrogen Antagonists/pharmacology
- Female
- Gene Expression Regulation, Enzymologic/drug effects
- Humans
- Mice
- Molecular Sequence Data
- Oligonucleotides, Antisense
- Polychlorinated Dibenzodioxins/pharmacology
- Promoter Regions, Genetic
- RNA, Messenger/analysis
- RNA, Messenger/biosynthesis
- Receptors, Aryl Hydrocarbon/biosynthesis
- Receptors, Aryl Hydrocarbon/physiology
- Receptors, Estrogen/biosynthesis
- Receptors, Estrogen/physiology
- Recombinant Proteins/biosynthesis
- Recombinant Proteins/metabolism
- Transfection
- Tumor Cells, Cultured
- Xenobiotics/pharmacology
Collapse
Affiliation(s)
- V Krishnan
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466, USA
| | | | | | | | | |
Collapse
|
30
|
Yang JH, Rhim JS. 2,3,7,8-Tetrachlorodibenzo-p-dioxin: molecular mechanism of carcinogenesis and its implication in human in vitro model. Crit Rev Oncol Hematol 1995; 18:111-27. [PMID: 7695826 DOI: 10.1016/1040-8428(94)00125-d] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- J H Yang
- Department of Preventive Medicine, Taegu Catholic University, School of Medicine, Republic of Korea
| | | |
Collapse
|
31
|
Safe SH. Modulation of gene expression and endocrine response pathways by 2,3,7,8-tetrachlorodibenzo-p-dioxin and related compounds. Pharmacol Ther 1995; 67:247-81. [PMID: 7494865 DOI: 10.1016/0163-7258(95)00017-b] [Citation(s) in RCA: 221] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The aryl hydrocarbon (Ah) receptor binds several different structural classes of chemicals, including halogenated aromatics, typified by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), polynuclear aromatic and heteropolynuclear aromatic hydrocarbons. TCDD induces expression of several genes including CYP1A1, and molecular biology studies show that the Ah receptor acts as a nuclear ligand-induced transcription factor that interacts with xenobiotic or dioxin responsive elements located in 5'-flanking regions of responsive genes. TCDD also elicits diverse toxic effects, modulates endocrine pathways and inhibits a broad spectrum of estrogen (17 beta-estradiol)-induced responses in rodents and human breast cancer cell lines. Molecular biology studies show that TCDD inhibited 17 beta-estradiol-induced cathepsin D gene expression by targeted interaction of the nuclear Ah receptor with imperfect dioxin responsive elements strategically located within the estrogen receptor-Sp1 enhancer sequence of this gene.
Collapse
Affiliation(s)
- S H Safe
- Texas A&M University, College Station 77843-4466, USA
| |
Collapse
|
32
|
Safe S, Krishnan V. Cellular and molecular biology of aryl hydrocarbon (Ah) receptor-mediated gene expression. ARCHIVES OF TOXICOLOGY. SUPPLEMENT. = ARCHIV FUR TOXIKOLOGIE. SUPPLEMENT 1995; 17:99-115. [PMID: 7786196 DOI: 10.1007/978-3-642-79451-3_8] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related compounds elicit diverse toxic and biochemical responses in laboratory animals and mammalian cells in culture. TCDD induces CYP1A1 gene expression and results of extensive research have delineated the molecular mechanism of this response. In target cells, TCDD initially binds to the aryl hydrocarbon (Ah) receptor which accumulates in the nucleus as an Ah-receptor:aryl hydrocarbon nuclear translocator (Arnt) protein heterodimeric complex. The nuclear Ah receptor complex acts as a ligand-induced transcription factor which binds to transacting genomic dioxin/xenobiotic responsive elements (DREs/XREs) located in the 5'-regulatory region upstream from the initiation start site and this interaction results in transactivation of gene transcription. DREs have been identified in several other genes which are induced by TCDD, including CYP1A2, aldehyde-3-dehydrogenase, NAD(P)H quinone oxidoreductase, and glutathione S transferase Ya and similar induction response pathways have been observed or proposed. However, TCDD and other Ah receptor agonists also inhibit expression of several genes and research in this laboratory has investigated inhibition of estrogen (E2)-induced genes including uterine epidermal growth factor, c-fos protooncogene, and the progesterone receptor, estrogen receptor (ER) and cathepsin D genes in human breast cancer cell lines. In MCF-7 human breast cancer cells, E2 induces cathepsin D gene expression and this is associated with formation of an ER/Sp1 complex at the sequence in the promoter region (-199/-165) of this gene. Within 30 min TCDD causes a rapid inhibition of E2-induced cathepsin D gene expression in MCF-7 cells. Moreover, using a series of synthetic oligonucleotides which include the wild-type ER/Sp1 and various mutants, it was shown by gel electromobility shift and transient transfection assays that the nuclear Ah receptor complex binds to an imperfect DRE located between the ER and Sp1 binding sequences. This interaction results in disruption of the ER/Sp1 complex and inhibition of E2-induced gene expression. These results illustrate that the nuclear Ah receptor complex also exhibits activity as a negative transcription factor via a mechanism which is similar to that reported for Ah receptor-mediated induction of gene expression.
Collapse
Affiliation(s)
- S Safe
- Texas A&M University, College Station 77843-4466, USA
| | | |
Collapse
|
33
|
Ahlborg UG, Lipworth L, Titus-Ernstoff L, Hsieh CC, Hanberg A, Baron J, Trichopoulos D, Adami HO. Organochlorine compounds in relation to breast cancer, endometrial cancer, and endometriosis: an assessment of the biological and epidemiological evidence. Crit Rev Toxicol 1995; 25:463-531. [PMID: 8611187 DOI: 10.3109/10408449509017924] [Citation(s) in RCA: 170] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
There is an increasing public and scientific concern that certain chlorinated compounds, recognized as environmental pollutants, may cause estrogen-related neoplastic disease in humans. The main hypothesis has been that certain organochlorines, through their estrogenic actions, might cause breast cancer. From experimental studies, both in vitro and in vivo, there is evidence that certain organochlorine compounds may cause estrogenic effects, whereas others may cause antiestrogenic effects. In limited studies, some of these compounds in high doses have also been shown to increase and reduce the frequency of estrogen-related tumors in animals. The epidemiological findings regarding the association between organochlorines and breast cancer are inconclusive. However, the largest and best designed study has been interpreted as negative with respect to DDT and polychlorinated biphenyls (PCB) in relation to breast cancer. Associations between organochlorine exposure and endometrial cancer or endometriosis have even more limited empirical basis. The hypothesis that human exposure to environmental levels or organochlorines would favor an estrogenic overactivity leading to an increase in estrogen-dependent formation of mammary or endometrial tumors is not supported by the existing in vitro, animal and epidemiological evidence. It can, however, not be conclusively rejected on the basis of available data.
Collapse
Affiliation(s)
- U G Ahlborg
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Harper N, Wang X, Liu H, Safe S. Inhibition of estrogen-induced progesterone receptor in MCF-7 human breast cancer cells by aryl hydrocarbon (Ah) receptor agonists. Mol Cell Endocrinol 1994; 104:47-55. [PMID: 7821706 DOI: 10.1016/0303-7207(94)90050-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
17 beta-Estradiol (E2) induces progesterone receptor (PR) binding, immunoreactive protein, nuclear PR formation and PR mRNA levels in MCF-7 human breast cancer cells. Gel mobility shift analysis of nuclear extracts from E2-treated cells also exhibited a higher intensity retarded band associated with formation of a PR complex with a consensus [32P]progesterone/glucocorticoid responsive element. In contrast, 1 nM 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) alone did not alter or decrease these same responses in MCF-7 cells; however, in cells co-treated with 1 nM TCDD plus 1 nM E2, TCDD significantly inhibited all the E2-induced responses. Scatchard analysis of PR binding demonstrated that TCDD decreased the number of E2-induced PR cellular binding sites but not the binding affinity of the PR for a radiolabeled promegestrone. In parallel studies, 3-methylcholanthrene, a prototypical polynuclear aromatic hydrocarbon, also inhibited E2-induced PR binding and immunoreactive protein. For a series of halogenated aromatics including 2,3,7,8- and 1,2,7,8-tetrachlorodibenzofuran, 1,3,7,8-TCDD and 6-methyl-1,3,8-trichlorodibenzofuran, their rank order potency for inhibiting E2-induced PR binding paralleled their rank order binding to the aryl hydrocarbon (Ah) receptor. These results support a role for the Ah receptor in mediating the antiestrogenic activity of polynuclear and halogenated aromatic hydrocarbons and illustrate cross-talk between the Ah and estrogen receptor signal transduction pathways.
Collapse
Affiliation(s)
- N Harper
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466
| | | | | | | |
Collapse
|
35
|
Holcomb M, Safe S. Inhibition of 7,12-dimethylbenzanthracene-induced rat mammary tumor growth by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Cancer Lett 1994; 82:43-7. [PMID: 8033067 DOI: 10.1016/0304-3835(94)90144-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related compounds inhibit diverse estrogen-induced responses in the rodent uterus and human breast cancer cells. The effects of a single non-toxic dose of TCDD (10 micrograms/kg) on the development of mammary tumors was investigated in female Sprague-Dawley rats treated with an oral dose of 7,12-dimethylbenzanthracene (DMBA) (20 mg). In rats which developed mammary tumors, subsequent treatment with corn oil (vehicle) resulted in a 3.9-fold increase in mammary tumor volume after 21 days. In contrast, a second group of rats with mammary tumors were treated with a non-toxic dose of TCDD (10 micrograms/kg) and after 21 days, the mean tumor volumes decreased from 89.7 +/- 53 mm3 to 24.9 +/- 28.5 mm3. Moreover, these results demonstrate the antitumorigenic activity of TCDD in female Sprague-Dawley rats.
Collapse
Affiliation(s)
- M Holcomb
- Texas A&M University, College Station 77843-4466
| | | |
Collapse
|
36
|
Frazier DE, Silverstone AE, Gasiewicz TA. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced thymic atrophy and lymphocyte stem cell alterations by mechanisms independent of the estrogen receptor. Biochem Pharmacol 1994; 47:2039-48. [PMID: 8010988 DOI: 10.1016/0006-2952(94)90079-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has both agonist and antagonist effects on estrogen-mediated activities and estrogen receptor (ER) levels in epithelial tissues following exposure. We previously demonstrated that TCDD alters bone marrow lymphocyte stem cells, including prothymocytes, as measured by functional assays and alterations in the lymphocyte stem cell-specific markers terminal deoxynucleotidyl transferase (TdT) and recombinase activating gene-1 (RAG-1). We have also shown that 17 beta-estradiol valerate (E2V) affects lymphocyte stem cells by reducing TdT and RAG-1 mRNA. It has been suggested that the effect of TCDD on these lymphocyte stem cells may be mediated directly or indirectly through estrogenic action and/or the ER. Studies were designed to evaluate whether endogenous estrogens or the ER mediate TCDD-elicited bone marrow alterations and thymic atrophy. Ovariectomy did not alter the sensitivity of mice to TCDD-induced thymic atrophy or to a reduction in TdT biosynthesis in bone marrow cells compared with either intact or sham-operated mice. The pure estrogen antagonist ICI 164,384 blocked E2V-induced uterine hypertrophy, thymic atrophy and reductions in lymphocyte stem cell markers. However, the antiestrogen failed to protect against TCDD-elicited thymic atrophy or bone marrow alterations in intact animals. The results are consistent with the hypothesis that the effects of TCDD on the thymus and/or bone marrow are mediated by mechanisms independent of estrogens or the ER.
Collapse
Affiliation(s)
- D E Frazier
- Department of Environmental Medicine, University of Rochester School of Medicine, NY 14642
| | | | | |
Collapse
|
37
|
Spink DC, Johnson JA, Connor SP, Aldous KM, Gierthy JF. Stimulation of 17 beta-estradiol metabolism in MCF-7 cells by bromochloro- and chloromethyl-substituted dibenzo-p-dioxins and dibenzofurans: correlations with antiestrogenic activity. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH 1994; 41:451-66. [PMID: 8145285 DOI: 10.1080/15287399409531856] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Mixed halo- and haloalkyl-substituted dibenzo-p-dioxins (DD) and dibenzofurans (DF) are known environmental contaminants, although there is limited information on the toxic effects of these compounds in human cells. In this study antiestrogenicity, a property of 2,3,7,8-Cl4-DD, was investigated with a series of bromochloro- and chloromethyl-substituted DDs and DFs. The effects of these compounds on the metabolism of 17 beta-estradiol (E2) and on the estrogen-dependent formation of multicellular foci in cultures of MCF-7 human breast cells were examined. Pretreatment of MCF-7 cells with 2,3,7,8-Cl4-DD induced pathways of E2 metabolism involving cytochrome P-450-catalyzed hydroxylation, methylation of the catechol estrogens, and conjugation. Several Br-Cl3-DD and Br2-Cl2-DD congeners with halogen substitution at the 2, 3, 7, and 8 positions also stimulated E2 metabolism with similar potency to that of 2,3,7,8-Cl4-DD; however, compounds with substitution of a methyl group for a halogen at any of these positions did not stimulate the metabolism of E2. For the series of compounds tested in MCF-7 cultures, a close correlation was observed between the antiestrogenicity as measured by the inhibition of estrogen-dependent postconfluent growth that results in focus formation and the efficacy with which the compounds stimulated the metabolism of E2. 2,3,7,8-TetrahaloDDs with one or two bromine atoms at these positions were highly antiestrogenic as determined by their inhibition of estrogen-dependent focus formation, whereas the methyl-substituted polychlorinated DDs and DFs investigated did not inhibit focus formation. These results indicate that the 2,3,7,8-substituted mixed halo-substituted DDs and DFs are of importance when the biologic effects of halogenated DD and DF congeners are considered, and provide additional evidence for the role of increased metabolism of E2 in the antiestrogenic effects of halogenated DDs and DFs.
Collapse
Affiliation(s)
- D C Spink
- Wadsworth Center for Laboratories and Research, New York State Department of Health, Albany 12201-0509
| | | | | | | | | |
Collapse
|
38
|
Fernandez P, Burghardt R, Smith R, Nodland K, Safe S. High passage T47D human breast cancer cells: altered endocrine and 2,3,7,8-tetrachlorodibenzo-p-dioxin responsiveness. Eur J Pharmacol 1994; 270:53-65. [PMID: 8157081 DOI: 10.1016/0926-6917(94)90080-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Low passage (L) T47D cells cultured for up to 12 months in media containing fetal bovine serum, fetal bovine serum plus 1 nM 17 beta-estradiol and estrogen-deficient media gave high passage cells denoted H, H(E+) and H(E-) cells, respectively, which exhibited differential responsiveness to 17 beta-estradiol, various growth factors, tamoxifen, 2,3,7,8-tetrachlorodibenzo-p-dioxin, and their combinations. Moreover, the altered mitogen/antimitogen responsiveness was paralleled by changes in hormone receptor levels, cellular architecture and ploidy. Estrogen receptor binding levels in the H, L and H(E+) cells varied from 36 to 155 fmol/mg protein; in contrast, the estrogen receptor binding in H(E-) cells exhibited a time-dependent increase from 81 to 1229 fmol/mg after culturing in estrogen-deficient media for approximately 12 months. Gel mobility shift assays of the nuclear estrogen receptor extracts from high and low passage cells with 32P-labeled estrogen responsive element showed that levels of the estrogen receptor-estrogen responsive element retarded band were lower in all the high passage cells compared to the low passage cells. These studies further illustrate the genetic instability of the T47D human breast cancer cell line and the resulting changes in mitogen and antimitogen responsiveness. In addition, the high passage H(E-) cells which express high estrogen receptor but low estrogen responsive element binding represent a unique model system for investigating the cellular and molecular biology of breast cancer cells which appear to be estrogen receptor-positive but are insensitive to antiestrogens.
Collapse
Affiliation(s)
- P Fernandez
- Department of Verterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466
| | | | | | | | | |
Collapse
|
39
|
Safe SH. Dietary and environmental estrogens and antiestrogens and their possible role in human disease. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 1994; 1:29-33. [PMID: 24234144 DOI: 10.1007/bf02986921] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Recent reports have suggested that dietary and environmental estrogens such as organochlorine pollutants may play a role in the increased incidence of breast cancer in women and disorders of the male reproductive tract. For example, elevated levels of DDE and polychlorinated biphenyls (PCB) have been measured in women with breast cancer. However, it should also be noted that numerous environmental and dietary compounds have also been characterized as antiestrogenic and as inhibitors of mammary cancer cell growthin vitro and/orin vivo. Some of these compounds include 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds, polynuclear aromatic hydrocarbons (PAH), other naturally-occurring TCDD receptor agonists, retinoids, phorbol esters, terpenes, fatty acids, and polysaccharides. Thus, it is possible that dietary and environmental estrogens and antiestrogens may be contra-active, and these interactions must be considered in the overall risk assessment of the potential adverse human and environmental health impacts of these chemicals.
Collapse
Affiliation(s)
- S H Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 77843-4466, College Station, TX, USA
| |
Collapse
|
40
|
Van den Berg M, De Jongh J, Poiger H, Olson JR. The toxicokinetics and metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs) and their relevance for toxicity. Crit Rev Toxicol 1994; 24:1-74. [PMID: 8172651 DOI: 10.3109/10408449409017919] [Citation(s) in RCA: 285] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
This article reviews the present state of the art regarding the toxicokinetics and metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs). The absorption, body distribution, and metabolism can vary greatly between species and also may depend on the congener and dose. In biota, the 2,3,7,8-substituted PCDDs and PCDFs are almost exclusively retained in all tissue types, preferably liver and fat. This selective tissue retention and bioaccumulation are caused by a reduced rate of biotransformation and subsequent elimination of congeners with chlorine substitution at the 2,3,7, and 8 positions. 2,3,7,8-Substituted PCDDs and PCDFs also have the greatest toxic and biological activity and affinity for the cytosolic arylhydrocarbon (Ah)-receptor protein. The parent compound is the causal agent for Ah-receptor-mediated toxic and biological effects, with metabolism and subsequent elimination of 2,3,7,8- substituted congeners representing a detoxification process. Congener-specific affinity of PCDDs and PCDFs for the Ah-receptor, the genetic events following receptor binding, and toxicokinetics are factors that contribute to the relative in vivo potency of an individual PCDD or PCDF in a given species. Limited human data indicate that marked species differences exist in the toxicokinetics of these compounds. Thus, human risk assessment for PCDDs and PCDFs needs to consider species-, congener-, and dose-specific toxicokinetic data. In addition, exposure to complex mixtures, including PCBs, has the potential to alter the toxicokinetics of individual compounds. These alterations in toxicokinetics may be involved in some of the nonadditive toxic or biological effects that are observed after exposure to mixtures of PCDDs or PCDFs with PCBs.
Collapse
Affiliation(s)
- M Van den Berg
- Research Institute of Toxicology, Utrecht University, The Netherlands
| | | | | | | |
Collapse
|
41
|
Abstract
Dioxin induces biological responses through interaction with a specific intracellular receptor, the Ah receptor, and the subsequent interaction of the Ah receptor with chromatin. We report the binding of the Ah receptor, partially purified from rabbit liver, to receptor binding factors in chromatin. Rabbit liver chromatin proteins (CP) were isolated by adsorption of chromatin to hydroxylapatite followed by sequential extraction with 1-8 M GdnHCl. To assay for receptor binding a portion of each CP fraction was reconstituted to rabbit double-stranded DNA using a reverse gradient dialysis of 7.5 to 0 M GdnHCl. These reconstituted nucleoacidic proteins were then examined for binding to [3H]-2,3,7,8-tetrachlorodibenzo-p-dioxin ([3H]TCDD)-receptor complexes by the streptomycin filter assay. Prior to the binding assay, [3H]TCDD-receptor complexes were partially purified by step elution from DEAE-cellulose columns. CP fractions 2, 5, and 7 were found to bind to the Ah receptor with high affinity. Scatchard analysis yielded Kd values in the nanomolar range. Competition with 2-fold excess unlabeled TCDD-receptor complexes was demonstrated, and binding was reduced markedly when the receptor was prepared in the presence of 10 mM molybdate. Such chromatin receptor binding factors (RBFs) may participate in the interaction of receptor with specific DNA sequences resulting in modulation of specific gene expression.
Collapse
Affiliation(s)
- R T Dunn
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, MO 63104
| | | | | |
Collapse
|
42
|
Moore M, Narasimhan TR, Wang X, Krishnan V, Safe S, Williams HJ, Scott AI. Interaction of 2,3,7,8-tetrachlorodibenzo-p-dioxin, 12-O-tetradecanoylphorbol-13-acetate (TPA) and 17 beta-estradiol in MCF-7 human breast cancer cells. J Steroid Biochem Mol Biol 1993; 44:251-61. [PMID: 8384872 DOI: 10.1016/0960-0760(93)90085-b] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and 12-O-tetradecanoylphorbol-13-acetate (TPA) are both tumor promoters which act through different mechanisms. In MCF-7 human breast cancer cells, both TCDD and TPA inhibited constitutive and 17 beta-estradiol-induced cell proliferation but showed no apparent interactive effects. TCDD also inhibited the 17 beta-estradiol-induced secretion of the 52-kDa protein (procathepsin D) and induced CYP1A1 gene expression whereas TPA alone was inactive for these responses. Moreover, TPA did not modulate the TCDD-mediated antiestrogenic or induction responses and did not decrease levels of the nuclear Ah receptor complex as determined in a gel mobility shift assay using a 32P-dioxin responsive element (DRE). The interactions of TPA and TCDD on the metabolism of [13C]glucose to [13C]lactate was also investigated using 13C-nuclear magnetic resonance spectroscopy. The rate of formation of [13C]lactate from [13C]glucose in MCF-7 cells treated with DMSO (control), 1 nM 17 beta-estradiol, 1 nM TCDD, 1 nM TCDD plus 1 nM 17 beta-estradiol, and 0.1 ng/ml TPA plus 1 nM 17 beta-estradiol was 28, 48, 20, 22 and 50 fmol lactate formed/cell/h, respectively. Thus, TCDD, but not TPA, inhibited this estrogen-induced response. However, a comparison of the rate of lactate formation in cells treated with TCDD plus 17 beta-estradiol (22 fmol/cell/h) or TCDD plus 17 beta-estradiol plus TPA (61 fmol/cell/h) showed that TPA significantly inhibited the TCDD-mediated antiestrogenic response. The results of these studies in MCF-7 cells demonstrate that the interactions of TCDD and TPA are highly response-specific and do not involve TPA-mediated downregulation of the nuclear Ah receptor complex.
Collapse
Affiliation(s)
- M Moore
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466
| | | | | | | | | | | | | |
Collapse
|
43
|
Peterson RE, Theobald HM, Kimmel GL. Developmental and reproductive toxicity of dioxins and related compounds: cross-species comparisons. Crit Rev Toxicol 1993; 23:283-335. [PMID: 8260069 DOI: 10.3109/10408449309105013] [Citation(s) in RCA: 390] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Developmental toxicity to TCDD-like congeners in fish, birds, and mammals, and reproductive toxicity in mammals are reviewed. In fish and bird species, the developmental lesions observed are species dependent, but any given species responds similarly to different TCDD-like congeners. Developmental toxicity in fish resembles "blue sac disease," whereas structural malformations can occur in at least one bird species. In mammals, developmental toxicity includes decreased growth, structural malformations, functional alterations, and prenatal mortality. At relatively low exposure levels, structural malformations are not common in mammalian species. In contrast, functional alterations are the most sensitive signs of developmental toxicity. These include effects on the male reproductive system and male reproductive behavior in rats, and neurobehavioral effects in monkeys. Human infants exposed during the Yusho and Yu-Cheng episodes, and monkeys and mice exposed perinatally to TCDD developed an ectodermal dysplasia syndrome that includes toxicity to the skin and teeth. Toxicity to the central nervous system in monkey and human infants is a potential part of the ectodermal dysplasia syndrome. Decreases in spermatogenesis and the ability to conceive and carry a pregnancy to term are the most sensitive signs of reproductive toxicity in male and female mammals, respectively.
Collapse
Affiliation(s)
- R E Peterson
- School of Pharmacy, University of Wisconsin, Madison 53706
| | | | | |
Collapse
|
44
|
Karageorgou M, Papadimitriou C, Marselos M. Sexual differentiation in the induction of the class 3 aldehyde dehydrogenase. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1993; 328:123-9. [PMID: 8493890 DOI: 10.1007/978-1-4615-2904-0_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- M Karageorgou
- Department of Pharmacology, Medical School, University of Ioannina, Greece
| | | | | |
Collapse
|
45
|
Liu H, Biegel L, Narasimhan TR, Rowlands C, Safe S. Inhibition of insulin-like growth factor-I responses in MCF-7 cells by 2,3,7,8-tetrachlorodibenzo-p-dioxin and related compounds. Mol Cell Endocrinol 1992; 87:19-28. [PMID: 1332906 DOI: 10.1016/0303-7207(92)90229-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Insulin-like growth factor-I (IGF-I) stimulated the growth and [3H]thymidine uptake in MCF-7 human breast cancer cells grown in serum- and growth factor-inactivated serum-containing media. Cotreatment of the cells with IGF-I plus 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) resulted in a significant decrease in mitogen-induced cell proliferation and [3H]thymidine uptake. Similar effects were observed for cells treated with 2,3,7,8-TCDD and IGF-I plus 17 beta-estradiol. The relative antimitogenic activities of 2,3,7,8-TCDD and related compounds followed the order 2,3,7,8-TCDD greater than 2,3,7,8-tetrachlorodibenzofuran (TCDF) greater than 1,2,7,8-TCDF greater than 1,3,7,8-TCDD which was similar to their aryl hydrocarbon (Ah) receptor binding affinities. The results showed that 2,3,7,8-TCDD did not alter the IGF-I receptor mRNA levels or the KD values for binding of [125I]IGF-I to the IGF-I receptor in MCF-7 cells. However, 2,3,7,8-TCDD significantly decreased the number of IGF-I-induced IGF-I receptor binding sites and this may play a role in the growth-inhibitory properties of 2,3,7,8-TCDD and related compounds and in the 'cross-talk' between the two endocrine-response pathways.
Collapse
Affiliation(s)
- H Liu
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station 77843-4466
| | | | | | | | | |
Collapse
|
46
|
Fernandez P, Safe S. Growth inhibitory and antimitogenic activity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in T47D human breast cancer cells. Toxicol Lett 1992; 61:185-97. [PMID: 1322575 DOI: 10.1016/0378-4274(92)90145-a] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The T47D human breast cancer cells used in this study express relative high levels of the progesterone receptor (637 +/- 118 fmol/mg protein) and lower levels of the estrogen and aryl hydrocarbon (Ah) receptors (81 +/- 3.4 and 55 +/- 8.2 fmol/mg protein, respectively). Treatment of these cells with 0.1, 1.0 and 10 nM concentrations of 17 beta-estradiol, transforming growth factor-alpha (TGF-alpha) and epidermal growth factor (EGF) resulted in concentration-dependent increase in cell proliferation and the ratios of mitogen-treated/control cell numbers were 2.46, 2.00 and 1.90, respectively. In contrast, insulin did not significantly stimulate T47D cell proliferation and insulin-like growth factor-I (IGF-I) was active only at a concentration of 10 nM. In parallel studies, the proliferative agents also stimulated the uptake of [3H]thymidine into cellular DNA. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) did not affect T47D cell growth at concentrations of 0.1 and 1.0 nM, whereas at a concentration of 10 nM a 44% decrease in cell numbers was observed. In cells cotreated with TCDD plus 100 ng/ml insulin or 10 nM 17 beta-estradiol, EGF, TGF-alpha and IGF-I, TCDD caused a concentration-dependent decrease in cell proliferation and [3H]thymidine uptake. For example, at a 10 nM concentration of TCDD there was a 32, 45, 29, 25 and 32% decrease in the 17 beta-estradiol, TGF-alpha, EGF, IGF-I and insulin-induced cell growth, respectively. These results confirm the antiproliferative activity of TCDD in T47D cells and this was similar to results previously reported in MCF-7 human breast cancer cells for the interaction of TCDD and 17 beta-estradiol. In addition, the data also show that TCDD inhibits the growth stimulatory effects of other polypeptide growth factors in T47D cells.
Collapse
Affiliation(s)
- P Fernandez
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466
| | | |
Collapse
|
47
|
DeVito MJ, Thomas T, Martin E, Umbreit TH, Gallo MA. Antiestrogenic action of 2,3,7,8-tetrachlorodibenzo-p-dioxin: tissue-specific regulation of estrogen receptor in CD1 mice. Toxicol Appl Pharmacol 1992; 113:284-92. [PMID: 1561637 DOI: 10.1016/0041-008x(92)90126-d] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a polychlorinated aromatic hydrocarbon with teratogenic and carcinogenic properties. Previous studies in our and other laboratories have demonstrated that TCDD has antiestrogenic properties. In order to elucidate the mechanism of action of TCDD on estrogen sensitive tissues, we studied its effects on serum estradiol and estrogen receptor (ER) levels in liver and uteri of CD1 mice. Treatment with TCDD did not result in alterations of serum estradiol levels at any of the doses tested (1.0-30 micrograms/kg). In contrast, TCDD treatment induced a dose-dependent decrease in hepatic and uterine ER protein as determined by an enzyme immunoassay and equilibrium binding assays. A decrease in cytosolic and nuclear ER levels in uteri occurred as early as 24 hr after initial treatment with 30 micrograms/kg TCDD and recovery occurred by 14 days. Hepatic cytosolic and nuclear ER also decreased at a dose of 30 micrograms/kg TCDD at 24 hr after treatment, but recovery occurred only by 21 days. Studies in ovariectomized mice indicate that the regulation of hepatic ER by TCDD is independent of ovarian factors, but ovariectomy inhibited the downregulation of uterine ER by TCDD. Furthermore, determination of TCDD-induced cytochrome P-450 levels indicates that the downregulation of uterine ER is uncoupled from induction of hepatic cytochrome P-450. This study indicates that the antiestrogenic effects of low doses of TCDD are mediated through its ability to decrease hepatic and uterine ER and are not due to alterations in serum estradiol levels. Our results on ovariectomized mice indicate that TCDD-induced downregulation of ER is tissue specific and may involve different mechanisms at transcriptional or posttranscriptional levels.
Collapse
Affiliation(s)
- M J DeVito
- Department of Environmental & Community Medicine, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway 08854
| | | | | | | | | |
Collapse
|
48
|
Zacharewski T, Harris M, Biegel L, Morrison V, Merchant M, Safe S. 6-Methyl-1,3,8-trichlorodibenzofuran (MCDF) as an antiestrogen in human and rodent cancer cell lines: evidence for the role of the Ah receptor. Toxicol Appl Pharmacol 1992; 113:311-8. [PMID: 1313996 DOI: 10.1016/0041-008x(92)90130-k] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
6-Methyl-1,3,8-trichlorodibenzofuran (MCDF) is a relatively nontoxic analog of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Treatment of aryl hydrocarbon (Ah)-responsive MCF-7 human breast cancer cells with 100 nM MCDF resulted in the inhibition of 17 beta-estradiol-induced proliferation and the secretion of the 34-, 52-, and 160-kDa proteins. After treatment of the cells with 17 beta-[3H]estradiol, 100 nM of MCDF caused a decrease in the accumulation of the radiolabeled nuclear estrogen receptor (ER) complex in these cells. In parallel experiments, the antiestrogenic effects of MCDF were also determined in Ah-responsive wild-type Hepa 1c1c7 cells and Ah-nonresponsive class 1 and class 2 mutant cells. Treatment of the wild-type cells with 17 beta-[3H]estradiol and 100 nM MCDF caused a decrease in the accumulation of radiolabeled nuclear ER complex in these cells whereas no significant effects were observed in the mutant cells as determined by velocity sedimentation analysis. Comparable results were obtained using ER antibodies to measure the decrease in immunoreactive nuclear ER. In addition, both actinomycin D and cycloheximide inhibited the MCDF-mediated decrease of nuclear ER levels in the Hepa 1c1c7 wild-type cells. Although 100 nM MCDF did not induce cytochrome P-450-dependent monooxygenases in the MCF-7 or Hepa 1c1c7 cell lines, incubation of nuclear extracts from the MCF-7 cells treated with 100 nM MCDF with a synthetic consensus dioxin responsive element (an oligonucleotide duplex of 26 bases) gave a retarded band in a gel-retardation assay. The data suggest that the antiestrogenic effects of MCDF does not require the induction of the CYP1A1 gene expression but may involve the induction of other genes.
Collapse
Affiliation(s)
- T Zacharewski
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466
| | | | | | | | | | | |
Collapse
|
49
|
Spink DC, Eugster HP, Lincoln DW, Schuetz JD, Schuetz EG, Johnson JA, Kaminsky LS, Gierthy JF. 17 beta-estradiol hydroxylation catalyzed by human cytochrome P450 1A1: a comparison of the activities induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin in MCF-7 cells with those from heterologous expression of the cDNA. Arch Biochem Biophys 1992; 293:342-8. [PMID: 1536570 DOI: 10.1016/0003-9861(92)90404-k] [Citation(s) in RCA: 160] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Exposure of MCF-7 breast cancer cells to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes an elevated cytochrome P450 content and a marked increase in the microsomal hydroxylation of 17 beta-estradiol (E2) at the C-2, C-4, C-15 alpha, and C-6 alpha positions. In this study we investigated the involvement of cytochromes P450 of the 1A gene subfamily in this metabolism of E2. Hydroxylation at each of these four positions of E2 was inhibited by P450 1A-subfamily inhibitors, alpha-naphthoflavone, benzo[a]pyrene, and 7-ethoxyresorufin. Northern blots showed that treatment of MCF-7 cells with TCDD resulted in production of the 2.6-kb CYP1A1 mRNA, but not the 3.0-kb CYP1A2 mRNA. Immunoblot analyses with anti-P450 1A antibodies confirmed the production of P450 1A1 protein in TCDD-treated MCF-7 cells. Anti-rat P450 1A IgG inhibited the hydroxylation of E2 at C-2, C-15 alpha, and C-6 alpha, but not hydroxylation at C-4. E2 hydroxylation by human cytochromes P450 1A1 and P450 1A2 was assessed in experiments with microsomes from Saccharomyces cerevisiae after transformation with cDNAs encoding the two cytochromes. The major hydroxylase activities of expressed human P450 1A1 were at the C-2, C-15 alpha, and C-6 alpha positions of E2; expressed human P450 1A2 catalyzed hydroxylation predominately at C-2. While both expressed P450s 1A1 and 1A2 had minor hydroxylase activities at the C-4 position, neither catalyzed a low-Km hydroxylation at C-4 similar to that observed with microsomes from TCDD-treated MCF-7 cells. These results provide strong evidence that P450 1A1 catalyzes the hydroxylations of E2 at the C-2, C-15 alpha, and C-6 alpha in incubations with microsomes from TCDD-treated MCF-7 cells, but suggest TCDD may also induce a cytochrome P450 E2 4-hydroxylase that is distinct from P450 1A1 or P450 1A2.
Collapse
Affiliation(s)
- D C Spink
- Wadsworth Center for Laboratories and Research, New York State Department of Health, Albany 12201-0509
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Dickerson R, Howie L, Safe S. The effect of 6-nitro-1,3,8-trichlorodibenzofuran as a partial estrogen in the female rat uterus. Toxicol Appl Pharmacol 1992; 113:55-63. [PMID: 1313194 DOI: 10.1016/0041-008x(92)90008-g] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Administration of 6-nitro-1,3,8-trichlorodibenzofuran (6-NCDF) caused a dose- and time-dependent increase in uterine wet weight and cytosolic and nuclear estrogen receptor (ER) and progesterone receptor (PR) levels in immature female Sprague-Dawley rats. These estrogenic effects persisted for up to 96 or 144 hr after initial administration of 6-NCDF and could be observed at a dose as low as 2 mumol/kg. In contrast, 6-NCDF (25 mumol/kg) did not increase rat uterine peroxidase activity or epidermal growth factor (EGF) receptor binding activity. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), which exhibits a broad spectrum of antiestrogenic effects in the female rat uterus, inhibited the 17 beta-estradiol-induced increase in uterine wet weights, cytosolic and nuclear ER and PR levels, peroxidase activity, and EGF receptor binding activity. In contrast, 2,3,7,8-TCDD inhibited the uterotropic effects caused by 6-NCDF but did not affect the 6-NCDF-induced uterine ER and PR levels. 6-NCDF is a weak inducer of hepatic microsomal ethoxyresorufin O-deethylase activity and competitively binds to the aryl hydrocarbon (Ah) receptor but not the PR or ER. Thus both 6-NCDF and 2,3,7,8-TCDD, two ligands which bind to the Ah receptor, exhibit both partial estrogenic and antiestrogenic properties and serve as useful models for delineating the complex biochemical interactions between the ER and Ah receptor signal transduction pathways.
Collapse
MESH Headings
- Animals
- Benzofurans/metabolism
- Benzofurans/pharmacology
- Binding, Competitive
- Cytochrome P-450 CYP1A1
- Cytochrome P-450 Enzyme System/metabolism
- Dose-Response Relationship, Drug
- Drug Interactions
- ErbB Receptors/drug effects
- ErbB Receptors/metabolism
- Estradiol/metabolism
- Female
- Microsomes, Liver/drug effects
- Microsomes, Liver/enzymology
- Microsomes, Liver/metabolism
- Nitro Compounds/metabolism
- Nitro Compounds/pharmacology
- Organ Size/drug effects
- Oxidoreductases/metabolism
- Peroxidase/metabolism
- Polychlorinated Dibenzodioxins/metabolism
- Polychlorinated Dibenzodioxins/pharmacology
- Rats
- Rats, Inbred Strains
- Receptors, Estrogen/drug effects
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/drug effects
- Receptors, Progesterone/metabolism
- Uterus/drug effects
- Uterus/enzymology
- Uterus/metabolism
Collapse
Affiliation(s)
- R Dickerson
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843
| | | | | |
Collapse
|