Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 174(1-3): 89-97, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17920789

ABSTRACT

The potential effect of receptor-mediated endocrine modulators across species is of increasing concern. In attempts to address these concerns, we are developing androgen and estrogen receptor binding assays using recombinant hormone receptors from a number of species across different vertebrate classes. The United States Environmental Protection Agency (USEPA) Office of Science Coordination and Policy (OSCP) requested that we develop a nonhuman mammalian receptor-binding assay for possible use in their Endocrine Disruptor Screening Program (EDSP). Since the chimpanzee androgen receptor is very similar to that of humans and thus possesses properties which could be exploited in future endocrine studies, we synthesized and expressed this gene in eukaryotic expression plasmids, baculovirus expression vectors and replication deficient adenovirus. In all ligand-binding and transcriptional activation assays tested, the chimpanzee receptor performed essentially identically to the human receptor. This suggests that the chimpanzee gene could substitute for the human gene in endocrine screening assays.


Subject(s)
Endocrine Disruptors/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Recombinant Proteins/metabolism , Adenoviridae/genetics , Androgens/metabolism , Animals , Baculoviridae/genetics , Binding, Competitive , Biological Assay , COS Cells , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Genetic Vectors , Humans , Metribolone/metabolism , Pan troglodytes , Plasmids , Transcriptional Activation , Transduction, Genetic
2.
Toxicol Appl Pharmacol ; 207(1): 78-88, 2005 Aug 22.
Article in English | MEDLINE | ID: mdl-16005038

ABSTRACT

PBDEs have been synthesized in large quantities as flame retardants for commercial products, such as electronic equipment and textiles. The rising in levels of PBDEs in tissues in wildlife species and in human milk and plasma samples over the past several years have raised concerns about possible health effects. Recently, we showed that the PBDE mixture, DE-71, delayed puberty and suppressed the growth of androgen-dependent tissues in male Wistar rat following a peri-pubertal exposure. These effects suggested that DE-71 may be either inducing steroid hormone metabolism or acting as an androgen receptor (AR) antagonist. To elucidate the potential anti-androgenic effects of this mixture, we evaluated DE-71 in several in vivo assays, which are responsive to alterations in androgen activity. In a pubertal exposure study designed to further evaluate the delay in preputial separation (PPS), we observed a dose-dependent delay in PPS with 60 and 120 mg/kg/day of DE-71 (4 and 5 days) and a corresponding suppression of ventral prostate (VP) and seminal vesicle growth at both doses. Adult males exposed to 60 mg/kg DE-71 for 3 days resulted in a significant increase in luteinizing hormone and a non-significant increase in testosterone, androstenedione and estrone. DE-71 also tested positive for anti-androgenic activity in an immature rat Hershberger assay, with decreases in mean VP and seminal vesicle weight following doses of 30-240 mg/kg. DE-71 and the individual BDE congeners which comprise the mixture (BDE-47, -99, -100, -153, -154) were also evaluated in vitro. First, AR binding was evaluated in a competitive binding assay using rat VP cytosol. In addition, we evaluated gene activation in a transcriptional activation assay using the MDA-kb2 cell line which contains an endogenous human AR and a transfected luciferase reporter. DE-71 and BDE-100 (2, 4, 6-pentaBDE) both inhibited AR binding, with IC50s of approximately 5 microM. In addition, DE-71 and two of the congeners (BDE-100 and BDE-47) inhibited DHT-induced transcriptional activation. The pattern of inhibition shown in the double-reciprocal plot for BDE-100 and the linear slope replot confirmed that the in vitro mechanism is pure competitive inhibition, with a inhibition constant (Ki) of 1 microM. The delay in puberty in the male rat and decreased growth of androgen-dependent tissues observed previously following exposure to DE-71 were likely due to this inhibition of AR binding by several of the congeners which make up this mixture.


Subject(s)
Androgen Antagonists/pharmacology , Androgen Receptor Antagonists , Phenyl Ethers/pharmacology , Polybrominated Biphenyls/pharmacology , Animals , Cell Line , Halogenated Diphenyl Ethers , Humans , Male , Rats , Rats, Wistar , Receptors, Androgen/metabolism , Transcriptional Activation/drug effects
3.
ILAR J ; 45(4): 425-37, 2004.
Article in English | MEDLINE | ID: mdl-15454681

ABSTRACT

The screening and testing program the US Environmental Protection Agency (EPA) is currently developing to detect endocrine-disrupting chemicals (EDCs) is described. EDCs have been shown to alter the following activities: hypothalamic-pituitary-gonadal (HPG) function; estrogen, androgen, and thyroid hormone synthesis; and androgen and estrogen receptor-mediated effects in mammals and other animals. The value and limitations of mammalian in vivo assays are described that involve the use of the laboratory rat, the EPA Endocrine Disruptor Screening and Testing Advisory Committee species of choice. The discussion includes the evaluation of high-priority chemicals positive in the Tier 1 Screening (T1S) battery, and of subsequent testing in the Tier 2 (T2) battery, with additional short-term screening assays proposed for use in T1.5 to eliminate any uncertainty about T1S results. Descriptions include the in vivo uterotropic assay, which detects estrogens and antiestrogens; the pubertal female assay, which assesses steroidogenesis, antithyroid activity, antiestrogenicity, and HPG function; and the Hershberger assay, which detects the weight of androgen-dependent tissues in castrate-immature-male rats (antiandrogens). Of the several alternative mammalian in vivo assays proposed, a short-term pubertal male rat assay appears most promising for inclusion in T1 or T1.5. An additional in utero-lactational screening protocol is being evaluated, but appears to be better suited for T1.5 or T2 due to the size, complexity, and duration of the assay. The adult intact male assay, also proposed as an alternative for T1, attempts to identify EDCs in a hormonal battery, but has limited value as a screen due to lack of sensitivity and specificity. For Tier 2 testing, the number of endocrine-sensitive endpoints and offspring (F1) examined in multigenerational tests must be thoughtfully expanded for EDCs on a mode-of-action-specific basis, with consideration given to tailoring T2 based on the results of T1S.


Subject(s)
Endocrine Glands/drug effects , Hormone Antagonists/toxicity , Models, Animal , Toxicity Tests/methods , Animals , Endocrine Glands/pathology , Endocrine Glands/physiopathology , Environmental Exposure/adverse effects , Female , Humans , Male , Rats , Species Specificity , United States , United States Environmental Protection Agency
4.
Biol Reprod ; 71(6): 1852-61, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15286035

ABSTRACT

Prenatal exposure to environmental chemicals that interfere with the androgen signaling pathway can cause permanent adverse effects on reproductive development in male rats. The objectives of this study were to 1) determine whether a documented antiandrogen butyl benzyl phthalate (BBP) and/or linuron (an androgen receptor antagonist) would decrease fetal testosterone (T) production, 2) describe reproductive developmental effects of linuron and BBP in the male, 3) examine the potential cumulative effects of linuron and BBP, and 4) investigate whether treatment-induced changes to neonatal anogenital distance (AGD) and juvenile areola number were predictive of adult reproductive alterations. Pregnant rats were treated with either corn oil, 75 mg/kg/day of linuron, 500 mg/kg/day of BBP, or a combination of 75 mg/kg/day linuron and 500 mg/kg/day BBP from gestational Day 14 to 18. A cohort of fetuses was removed to assess male testicular T and progesterone production, testicular T concentrations, and whole-body T concentrations. Male offspring from the remaining litters were assessed for AGD and number of areolae and then examined for alterations as young adults. Prenatal exposure to either linuron or BBP or BBP + linuron decreased T production and caused alterations to androgen-organized tissues in a dose-additive manner. Furthermore, treatment-related changes to neonatal AGD and infant areolae significantly correlated with adult AGD, nipple retention, reproductive malformations, and reproductive organ and tissue weights. In general, consideration of the dose-response curves for the antiandrogenic effects suggests that these responses were dose additive rather than synergistic responses. Taken together, these data provide additional evidence of cumulative effects of antiandrogen mixtures on male reproductive development.


Subject(s)
Androgen Antagonists/pharmacology , Linuron/pharmacology , Phthalic Acids/pharmacology , Sex Differentiation/drug effects , Anal Canal/anatomy & histology , Androgen Antagonists/administration & dosage , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Female , Fetus/metabolism , Genitalia, Male/anatomy & histology , Genitalia, Male/embryology , Linuron/administration & dosage , Male , Osmolar Concentration , Phthalic Acids/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects , Progesterone/metabolism , Rats , Rats, Sprague-Dawley , Testis/metabolism , Testosterone/metabolism
5.
Toxicology ; 181-182: 371-82, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12505339

ABSTRACT

The US Environmental Protection Agency (EPA) is developing a screening and testing program for endocrine disrupting chemicals (EDCs) to detect alterations of hypothalamic-pituitary-gonadal (HPG) function, estrogen (ER), androgen (AR) and thyroid hormone synthesis and AR and ER receptor-mediated effects in mammals and other animals. High priority chemicals would be evaluated in the Tier 1 Screening (T1S) battery and chemicals positive in T1S would then be tested (Tier 2). T1S includes in vitro ER and AR receptor binding and/or gene expression, an assessment of steroidogenesis and mammalian (rat) and nonmammalian in vivo assays (Table 1). In vivo, the uterotropic assay detects estrogens and antiestrogens, while steroidogenesis, antithyroid activity, (anti)estrogenicity and HPG function are assessed in a 'Pubertal Female Assay'. (Anti-) androgens are detected in the Hershberger Assay (weight of AR-dependent tissues in castrate-immature-male rats). Fish and amphibian assays also are being developed. The fathead minnow assay can identify EDCs displaying several mechanisms of concern, including AR and ER receptor agonists and antagonists and inhibitors of steroid hormone synthesis. An amphibian metamorphosis assay is being developed to detect thyroid-active substances. Several alternative mammalian in vivo assays have been proposed. Of these, a short-term pubertal male rat assay appears most promising. An in utero-lactational screening protocol also is being evaluated. For Tier 2, the numbers of endocrine sensitive endpoints and offspring (F1) examined in multigenerational tests need to be expanded for EDCs. Consideration should be given to tailoring T2, based on the results of T1S. Tier 1 and 2 also should examine relevant mixtures of EDCs. Toxicants that induce malformations in AR-dependent tissues produce cumulative effects even when two chemicals act via different mechanisms of action.


Subject(s)
Endocrine Glands/drug effects , Endocrine System Diseases/chemically induced , Xenobiotics/toxicity , Animals , Biological Assay , Endocrine System Diseases/pathology , Humans , Toxicology/methods , United States , United States Environmental Protection Agency
6.
Toxicol Sci ; 66(1): 82-90, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861975

ABSTRACT

The discovery of xenobiotics that interfere with androgen activity has highlighted the need to assess chemicals for their ability to modulate dihydrotestosterone (DHT)-receptor binding. Previous test systems have used cells transfected with plasmid containing a reporter gene. Here we report the use of transduction for gene delivery and assessment of the modulation of DHT-induced gene activation. Transduction, the ability of replication-defective viruses to deliver biologically competent genes, is a well understood biological process, which has been utilized to repair defective genes in humans as well as to express exogenous genes in rodent models. Human breast carcinoma cells (MDA-MB-453) containing endogenous copies of the androgen (hAR) and glucocorticoid (GR) receptors were transduced with replication-defective human adenovirus type 5 containing the luciferase (Luc) reporter gene driven by the AR- and GR-responsive glucocorticoid-inducible hormone response element found with the mammary tumor virus LTR (Ad/MLUC7). In a second set of experiments, CV-1 cells were transduced as above with MMTV-luc and also hAR. Cells were subcultured in 96-well plates, transduced with virus, exposed to chemicals, incubated for 48 h, lysed, and assayed for luciferase. Luc gene expression was induced in a dose-dependent manner by DHT, estradiol, and dexamethasone (MDA only) and inhibited by AR antagonist hydroxyflutamide (OHF), hydroxy-DDE, HPTE (2,2-bis(p-hydroxyphenyl)-1,1, 1-trichloroethane), a methoxychlor metabolite, and M1 and M2 (vinclozolin metabolites). The transduced cells responded to AR agonists and antagonists as predicted from our other studies, with a very robust and reproducible response. Over all replicates, 0.1 nM DHT induced luc expression by about 45-fold in CV-1 cells (intra-assay CV = 20%) and 1micromolar OHF inhibited DHT by about 80%. In the transduced MDA cells, 0.1 nM DHT induced luc by about 24-fold (intra-assay CV = 33%), which was inhibited by OHF by about 85%. DHT-induced luciferase activity peaked in both cell lines between 1 and 100 nM, displaying about 64- and 115-fold maximal induction in the CV-1 and MDA 453 cells, respectively. For agonists, a two-fold induction of luc over media control was statistically significant. For AR antagonists, a 25-30% inhibition of DHT-induced luc expression was typically statistically significant. Comparing the two assays, the transduced CV-1 cells were slightly more sensitive to AR-mediated responses, but the transduced MDA 453 cells were more responsive to GR agonists. In summary, these assays correctly identified the endocrine activity of all chemicals examined and displayed sensitivity with a relatively low variability and a high-fold induction over background. Adenovirus transduction for EDC screening has the potential to be employed in a high-throughput mode, and could easily be applied to other cell lines and utilized to deliver other receptors and reporter genes.


Subject(s)
Flutamide/analogs & derivatives , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/metabolism , Adenoviridae/genetics , Androgen Antagonists/pharmacology , Androgen Receptor Antagonists , Androgens/pharmacology , Animals , Cell Line , Dexamethasone/pharmacology , Dihydrotestosterone/pharmacology , Flutamide/pharmacology , Gene Expression/drug effects , Glucocorticoids/pharmacology , Humans , Luciferases/drug effects , Luciferases/genetics , Luciferases/metabolism , Mammary Tumor Virus, Mouse/genetics , Pesticides/pharmacology , Progesterone/pharmacology , Promoter Regions, Genetic/genetics , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Progesterone/agonists , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transduction, Genetic/methods , Transfection/methods , Tumor Cells, Cultured
7.
Toxicol Sci ; 62(2): 257-67, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11452138

ABSTRACT

Female mosquitofish (Gambusia affinis holbrooki) downstream from Kraft paper mills in Florida display masculinization of the anal fin, an androgen-dependent trait. The current investigation was designed to determine if water contaminated with pulp-mill effluent (PME) from the Fenholloway River in Florida displayed androgenic activity in vitro and to relate this activity to the reproductive status of female mosquitofish taken from this river. We tested water samples for androgenic activity from a reference site upstream of a Kraft pulp and paper mill on the Fenholloway River, from 3 sites downstream from the mill, and from another reference site on the Econfina River, also in Florida, where there is no paper mill. We also examined anal fin ray morphology in mosquitofish from these rivers for evidence of masculinization. Eighty percent of the female mosquitofish from the Fenholloway River were partially masculinized while another 10% were completely masculinized, based upon the numbers of segments in the longest anal fin ray (18.0 +/- 0.4 vs. 28.1 +/- 0.9 [p < 0.001]) in the Econfina River vs. the Fenholloway River, respectively). In a COS whole cell-binding assay, all 3 PME samples displayed affinity for human androgen receptor (hAR) (p < 0.001). In addition, PME induced androgen-dependent gene expression in CV-1 cells (cotransfected with pCMV hAR and MMTV luciferase reporter), which was inhibited by about 50% by coadministration of hydroxyflutamide (1 microM), an AR antagonist. Water samples collected upstream of the Kraft mill or from the Econfina River did not bind hAR or induce luciferase expression. When CV-1 cells were transfected with human glucocorticoid receptor (hGR) rather than hAR, PME failed to significantly induce MMTV-luciferase expression. Further evidence of the androgenicity was observed using a COS cell AR nuclear-translocalization assay. PME bound hAR and induced translocalization of AR into the nucleus. In contrast, AR remained perinuclear when treated with water from the control sites (indicating the absence of an AR ligand). Interestingly, PME also displayed "testosterone-like" immunoreactivity in a testosterone radioimmunoassay, whereas water from the reference sites did not. In summary, water collected downstream of the Kraft mill on the Fenholloway River contains unidentified androgenic substances whose presence is associated with masculinization of female mosquitofish.


Subject(s)
Androgens , Sex Characteristics , Water Pollutants, Chemical/toxicity , Animals , COS Cells , Female , Fishes , Humans , Immunohistochemistry , Industrial Waste , Male , Radioimmunoassay , Receptors, Androgen/metabolism , Testosterone/metabolism , Transcriptional Activation
8.
Hum Reprod Update ; 7(3): 248-64, 2001.
Article in English | MEDLINE | ID: mdl-11392371

ABSTRACT

Chemicals that act as androgen receptor (AR) agonists and antagonists or inhibit fetal steroidogenesis can induce reproductive malformations in humans and laboratory animals. Several environmental chemicals disrupt development in rats and/or rabbits at fetal concentrations at, or near, exposure levels seen in some segments of the human population. In rats, fetal tissues concentrations of 10-20 p.p.m. of the DDT metabolite, p,p'-DDE, are correlated with reproductive abnormalities in male offspring. These concentrations are similar to those measured in first-trimester human fetal tissues in the late 1960s. The pesticides vinclozolin, procymidone, linuron and DDT are AR antagonists. They reduce male rat anogenital distance, and induce areolas at relatively low dosages. Hypospadias, agenesis of the sex accessory tissues and retained nipples are seen in the middle dosages, while undescended testes and epididymal agenesis are seen in the highest doses. Phthalate esters (PE) inhibit testosterone synthesis during fetal life, but do not appear to be AR antagonists. Prenatal administration of a single low dose of dioxin (50-1,000 ng TCDD/kg) alters the differentiation of androgen-dependent tissues at p.p.t. concentrations, but the mechanism of action likely involves interaction with a hormone-like nuclear transcription factor, the hormone-like receptor AhR, rather than AR. p,p'-DDT and p,p'-DDE, vinclozolin and di-n-butyl phthalate affect reproductive function in rabbits when administered during prenatal and/or neonatal life. Cryptorchidism and carcinoma in situ-like (CIS) testicular lesions were seen in male rabbits treated during development with p,p'-DDT or p,p'-DDE. Extrapolation of effects from rodents to humans would be enhanced if future studies incorporate determination of tissue concentrations of the active metabolites. Knowledge of the tissue concentrations of the active toxicants also would provide an important link to in-vitro studies, which provide more useful mechanistic information when they are executed at relevant concentrations.


Subject(s)
Androgen Antagonists/pharmacology , Environmental Exposure , Genitalia, Male/drug effects , Genitalia, Male/growth & development , Animals , Animals, Laboratory/growth & development , Humans , Male
9.
Toxicol Sci ; 58(2): 339-49, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11099646

ABSTRACT

Phthalate esters (PE) such as DEHP are high production volume plasticizers used in vinyl floors, food wraps, cosmetics, medical products, and toys. In spite of their widespread and long-term use, most PE have not been adequately tested for transgenerational reproductive toxicity. This is cause for concern, because several recent investigations have shown that DEHP, BBP, DBP, and DINP disrupt reproductive tract development of the male rat in an antiandrogenic manner. The present study explored whether the antiandrogenic action of DEHP occurs by (1) inhibiting testosterone (T) production, or by (2) inhibiting androgen action by binding to the androgen receptor (AR). Maternal DEHP treatment at 750 mg/kg/day from gestational day (GD) 14 to postnatal day (PND) 3 caused a reduction in T production, and reduced testicular and whole-body T levels in fetal and neonatal male rats from GD 17 to PND 2. As a consequence, anogenital distance (AGD) on PND 2 was reduced by 36% in exposed male, but not female, offspring. By GD 20, DEHP treatment also reduced testis weight. Histopathological evaluations revealed that testes in the DEHP treatment group displayed enhanced 3ss-HSD staining and increased numbers of multifocal areas of Leydig cell hyperplasia as well as multinucleated gonocytes as compared to controls at GD 20 and PND 3. In contrast to the effects of DEHP on T levels in vivo, neither DEHP nor its metabolite MEHP displayed affinity for the human androgen receptor at concentrations up to 10 microM in vitro. These data indicate that DEHP disrupts male rat sexual differentiation by reducing T to female levels in the fetal male rat during a critical stage of reproductive tract differentiation.


Subject(s)
Abnormalities, Drug-Induced/etiology , Diethylhexyl Phthalate/toxicity , Genitalia, Male/abnormalities , Plasticizers/toxicity , Sex Differentiation/drug effects , Testosterone/biosynthesis , Animals , Body Weight/drug effects , Female , Leydig Cells/drug effects , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Testis/drug effects , Testis/metabolism , Testis/pathology
10.
Toxicol Sci ; 56(2): 389-99, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10910998

ABSTRACT

Antiandrogenic chemicals alter sex differentiation by several different mechanisms. Some, like flutamide, procymidone, or vinclozolin compete with androgens for the androgen receptor (AR), inhibit AR-DNA binding, and alter androgen-dependent gene expression in vivo and in vitro. Finasteride and some phthalate esters demasculinize male rats by inhibiting fetal androgen synthesis. Linuron, which is a weak competitive inhibitor of AR binding (reported Ki of 100 microM), alters sexual differentiation in an antiandrogenic manner. However, the pattern of malformations more closely resembles that produced by the phthalate esters than by vinclozolin treatment. The present study was designed to determine if linuron acted as an AR antagonist in vitro and in vivo. In vitro, we (1) confirmed the affinity of linuron for the rat AR, and found (2) that linuron binds human AR (hAR), and (3) acts as an hAR antagonist. Linuron competed with an androgen for rat prostatic AR (EC(50) = 100-300 microM) and human AR (hAR) in a COS cell-binding assay (EC(50) = 20 microM). Linuron inhibited dihydrotestosterone (DHT)-hAR induced gene expression in CV-1 and MDA-MB-453-KB2 cells (EC(50) = 10 microM) at concentrations that were not cytotoxic. In short-term in vivo studies, linuron treatment reduced testosterone- and DHT-dependent tissue weights in the Hershberger assay (oral 100 mg/kg/d for 7 days, using castrate-immature-testosterone propionate-treated male rats; an assay used for decades to screen for AR agonists and antagonists) and altered the expression of androgen-regulated ventral prostate genes (oral 100 mg/kg/d for 4 days). Histological effects of in utero exposure to linuron (100 mg/kg/d, day 14-18) or DBP (500 mg/kg/d, day 14 to postnatal day 3) on the testes and epididymides also are shown here. Taken together, these results support the hypothesis that linuron is an AR antagonist both in vivo and in vitro, but it remains to be determined if linuron alters sexual differentiation by additional mechanisms of action.


Subject(s)
Abnormalities, Drug-Induced , Androgen Antagonists/toxicity , Genitalia, Male/abnormalities , Herbicides/toxicity , Linuron/toxicity , Animals , COS Cells , Dibutyl Phthalate/toxicity , Dihydrotestosterone/pharmacology , Dose-Response Relationship, Drug , Female , Genitalia, Male/drug effects , Genitalia, Male/pathology , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Transcriptional Activation/drug effects
11.
Toxicol Ind Health ; 15(1-2): 94-118, 1999.
Article in English | MEDLINE | ID: mdl-10188194

ABSTRACT

Antiandrogenic chemicals alter sexual differentiation by a variety of mechanisms, and as a consequence, they induce different profiles of effects. For example, in utero treatment with the androgen receptor (AR) antagonist, flutamide, produces ventral prostate agenesis and testicular nondescent, while in contrast, finasteride, an inhibitor of 5 alpha-dihydrotestosterone (DHT) synthesis, rarely, if ever, induces such malformations. In this regard, it was recently proposed that dibutyl phthalate (DBP) alters reproductive development by a different mechanism of action than flutamide or vinclozolin (V), which are AR antagonists, because the male offsprings display an unusually high incidence of testicular and epididymal alterations--effects rarely seen after in utero flutamide or V treatment. In this study, we present original data describing the reproductive effects of 10 known or suspected anti-androgens, including a Leydig cell toxicant ethane dimethane sulphonate (EDS, 50 mg kg-1 day-1), linuron (L, 100 mg kg-1 day-1), p,p'-DDE (100 mg kg-1 day-1), ketoconazole (12-50 mg kg-1 day-1), procymidone (P, 100 mg kg-1 day-1), chlozolinate (100 mg kg-1 day-1), iprodione (100 mg kg-1 day-1), DBP (500 mg kg-1 day-1), diethylhexyl phthalate (DEHP, 750 mg kg-1 day-1), and polychlorinated biphenyl (PCB) congener no. 169 (single dose of 1.8 mg kg-1). Our analysis indicates that the chemicals discussed here can be clustered into three or four separate groups, based on the resulting profiles of reproductive effects. Vinclozolin, P, and DDE, known AR ligands, produce similar profiles of toxicity. However, p,p'-DDE is less potent in this regard. DBP and DEHP produce a profile distinct from the above AR ligands. Male offsprings display a higher incidence of epididymal and testicular lesions than generally seen with flutamide, P, or V even at high dosage levels. Linuron treatment induced a level of external effects consistent with its low affinity for AR [reduced anogenital distance (AGD), retained nipples, and a low incidence of hypospadias]. However, L treatment also induced an unanticipated degree of malformed epididymides and testis atrophy. In fact, the profile of effects induced by L was similar to that seen with DBP. These results suggest that L may display several mechanisms of endocrine toxicity, one of which involves AR binding. Chlozolinate and iprodione did not produce any signs of maternal or fetal endocrine toxicity at 100 mg kg-1 day-1. EDS produced severe maternal toxicity and a 45% reduction in size at birth, which resulted in the death of all neonates by 5 days of age. However, EDS only reduced AGD in male pups by 15%. Ketoconazole did not demasculinize or feminize males but rather displayed anti-hormonal activities, apparently by inhibiting ovarian hormone synthesis, which resulted in delayed delivery and whole litter loss. In summary, the above in vivo data suggest that the chemicals we studied alter male sexual differentiation via different mechanisms. The anti-androgens V, P, and p,p'-DDE produce flutamide-like profiles that are distinct from those seen with DBP, DEHP, and L. The effects of PCB 169 bear little resemblance to those of any known anti-androgen. Only in depth in vitro studies will reveal the degree to which one can rely upon in vivo studies, like those presented here, to predict the cellular and molecular mechanisms of developmental toxicity.


Subject(s)
Androgen Antagonists/toxicity , Genitalia, Male/abnormalities , Hydantoins , Pesticides/toxicity , Xenobiotics/toxicity , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/toxicity , Androgen Antagonists/pharmacology , Animals , Bridged Bicyclo Compounds/toxicity , Dibutyl Phthalate/toxicity , Dichlorodiphenyl Dichloroethylene/toxicity , Diethylhexyl Phthalate/toxicity , Female , Genitalia, Male/drug effects , Genitalia, Male/growth & development , Gonadal Steroid Hormones/metabolism , Ketoconazole/toxicity , Linuron/toxicity , Male , Oxazoles , Pesticides/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors, Androgen/drug effects , Receptors, Androgen/physiology , Sex Differentiation/drug effects , Xenobiotics/pharmacology
12.
Toxicol Ind Health ; 15(1-2): 65-79, 1999.
Article in English | MEDLINE | ID: mdl-10188192

ABSTRACT

Vinclozolin is a well-characterized antiandrogenic fungicide. It produces adverse effects when administered during sexual differentiation, and it alters reproductive function in adult male rats by acting as an androgen-antagonist. Two active metabolites of vinclozolin, M1 and M2, compete with natural androgens for the rat and human androgen receptors (ARs), an effect that blocks androgen-induced gene expression in vivo and in vitro. In addition to their effects during perinatal life, androgens play a key role in pubertal maturation in young males. In this regard, the present study was designed to examine the effects of peripubertal oral administration of vinclozolin (0, 10, 30, or 100 mg kg-1 day-1) on morphological landmarks of puberty, hormone levels, and sex accessory gland development in male rats. In addition, as binding of the M1 and M2 to AR alter the subcellular distribution of AR by inhibiting AR-DNA binding, we examined the effects of vinclozolin on AR distribution in the target cells after in vivo treatment. We also examined serum levels of vinclozolin, M1, and M2 in the treated males so that these could be related to the effects on the reproductive tract and AR distribution. Vinclozolin treatment delayed pubertal maturation (at 30 and 100 mg kg-1 day-1) and retarded sex accessory gland and epididymal growth. Serum luteinizing hormone (LH; significant at all dosage levels) and testosterone and 5 alpha-androstane, 3 alpha, 17 beta-diol (at 100 mg kg-1 day-1) levels were increased. Testis size and sperm production, however, were unaffected. It was apparent that these effects were concurrent with subtle alterations in the subcellular distribution of AR. In control animals, most AR were in the high salt cell fraction, apparently bound to the natural ligand and DNA. Vinclozolin treatment reduced the amount of AR in the high salt (bound to DNA) fraction and it increased AR levels in the low salt (inactive, not bound to DNA) fraction. M1 and M2 were found in the serum of animals from the two highest dosage groups, but they were present at levels well below their K1 values. In summary, these results suggest that when the vinclozolin metabolites occupy a small percentage of AR in the cell, this prevents maximal AR-DNA binding and alters in vivo androgen-dependent gene expression and protein synthesis, which in turn results in obvious alterations of morphological development and serum hormone levels. It is noteworthy that similar exposures during prenatal life result in a high incidence of malformations in male rats.


Subject(s)
Fungicides, Industrial/toxicity , Oxazoles/toxicity , Receptors, Androgen/physiology , Administration, Oral , Animals , DNA/drug effects , Dose-Response Relationship, Drug , Fungicides, Industrial/pharmacokinetics , Fungicides, Industrial/pharmacology , Gene Expression Regulation/drug effects , Male , Oxazoles/pharmacokinetics , Oxazoles/pharmacology , Rats , Rats, Long-Evans , Receptors, Androgen/drug effects , Testis/drug effects , Testis/growth & development , Testosterone/blood
13.
Toxicol Ind Health ; 15(1-2): 80-93, 1999.
Article in English | MEDLINE | ID: mdl-10188193

ABSTRACT

Procymidone is a dicarboximide fungicide structurally related to the well-characterized fungicide vinclozolin. Vinclozolin metabolites bind to mammalian androgen receptors (AR) and act as AR antagonists, inhibiting androgen-dependent gene expression in vivo and in vitro by inhibiting AR-binding to DNA. The current study was designed to determine if procymidone acted as an AR antagonist in vitro and to describe the dosage levels of procymidone that alter sexual differentiation in vivo. In vitro, procymidone inhibited androgen from binding the human AR (hAR) in COS (monkey kidney) cells transfected with hAR at 3.16 microM. In vitro, procymidone acted as an androgen antagonist, inhibiting dihydrotestosterone (DHT)-induced transcriptional activation at 0.2 microM in CV-1 cells (cotransfected with the hAR and a MMTV-luciferase reporter gene). In vivo, maternal procymidone exposure at 0, 25, 50, 100, or 200 mg kg-1 day-1 during gestation and early lactation (gestational day 14 to postnatal day 3) altered reproductive development of male offspring at all dosage levels tested. Male offspring exhibited shortened anogenital distance (at 25 mg kg-1 day-1 and above), permanent nipples, reduced weight of several androgen-dependent tissues (levator ani and bulbocavernosus muscles, prostate, seminal vesicles, Cowper's gland and glans penis), and malformations (hypospadias, cleft phallus, exposed os penis, vaginal pouch, hydronephrosis, occasional hydroureter, epididymal granulomas, and ectopic, undescended testes). In addition, perinatal procymidone treatment had a marked effect on the histology of the lateral and ventral prostatic and seminal vesicular tissues of the offspring (at 50 mg kg-1 day-1 and above). These effects consisted of fibrosis, cellular infiltration, and epithelial hyperplasia. This constellation of effects is similar to that produced by perinatal exposure to vinclozolin. However, procymidone appears to be slightly less potent in inducing malformations than vinclozolin by a factor of about two. In summary, the antiandrogenic activity of procymidone was demonstrated in vivo and in vitro in cell lines transfected with hAR. Since the role of androgens in mammalian sexual differentiation is highly conserved, it is likely that humans would be adversely affected by procymidone in a predictable manner if the human fetus was exposed to sufficient levels during critical stages of intrauterine and neonatal life.


Subject(s)
Bridged Bicyclo Compounds/toxicity , Fungicides, Industrial/toxicity , Gene Expression Regulation/drug effects , Genitalia, Male/growth & development , Receptors, Androgen/drug effects , Androgen Receptor Antagonists , Animals , Bridged Bicyclo Compounds/pharmacology , Cell Line/drug effects , Dose-Response Relationship, Drug , Female , Fungicides, Industrial/pharmacology , Genitalia, Male/abnormalities , Genitalia, Male/drug effects , Male , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Long-Evans , Receptors, Androgen/genetics
14.
Toxicol Appl Pharmacol ; 142(1): 192-200, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9007049

ABSTRACT

Vinclozolin and p,p'-DDE induce antiandrogenic developmental effects in vivo and are potent inhibitors of androgen receptor (AR) binding and AR-dependent gene expression in vitro. To determine whether this molecular mechanism is operative in vivo, the effects of these compounds on two androgen-regulated prostatic mRNAs were studied. Rats were sham operated or castrated and immediately implanted with one or two empty 2.5-cm silastic capsules or with one (1x) or two (2x) 2.5-cm capsules containing testosterone (T). T-implanted rats were treated by gavage for 4 days with vehicle (corn oil), vinclozolin (200 mg/kg/day), p,p'-DDE (200 mg/kg/day), or the antiandrogen flutamide (100 mg/kg/day) as a positive control. Vinclozolin, p,p'-DDE, and flutamide all induced a reciprocal decline in seminal vesicle (p < 0.01) and prostate (p < 0.01) weight as well as a reduction in immunohistochemical staining of AR in epididymal nuclei compared to vehicle-treated T-implanted controls. Specific AR antagonism was assessed by determining the ability of these chemicals to induce a testosterone-repressed prostatic message (i.e., TRPM-2) and/or repress a testosterone-induced prostatic message (i.e., prostatein subunit C3). Densitometry scans of Northern blots indicated that vinclozolin, p,p'-DDE, and flutamide each induced TRPM-2 mRNA and repressed C3 mRNA compared to vehicle-treated T-implanted controls. These antiandrogenic effects were competitively reduced in castrate rats implanted with two 2.5-cm T capsules (2x), where serum T levels were elevated more than twofold above physiological levels. Taken together, these data indicate that vinclozolin and p,p'-DDE act as antiandrogens in vivo by altering the expression of androgen-dependent genes.


Subject(s)
Androgen Antagonists/pharmacology , Dichlorodiphenyl Dichloroethylene/pharmacology , Gene Expression Regulation/drug effects , Molecular Chaperones , Oxazoles/pharmacology , Receptors, Androgen/drug effects , Androgen-Binding Protein/genetics , Animals , Clusterin , Drug Implants , Flutamide/pharmacology , Glycoproteins/genetics , Male , Orchiectomy , Organ Size/drug effects , Prostate/drug effects , Prostate/pathology , Prostatein , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Androgen/metabolism , Secretoglobins , Seminal Vesicles/drug effects , Seminal Vesicles/pathology , Testosterone/administration & dosage , Testosterone/pharmacology , Uteroglobin
SELECTION OF CITATIONS
SEARCH DETAIL
...