Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Toxicol Ind Health ; 15(1-2): 37-47, 1999.
Article in English | MEDLINE | ID: mdl-10188190

ABSTRACT

This study was designed to determine if long-term exposure to high doses of methoxychlor (M) would alter pituitary or testicular endocrine functions in either an estrogenic or antiandrogenic manner. Weanling male Long-Evans hooded rats were dosed daily with M (po) at 0, 200, 300, or 400 mg kg-1 day-1 for 10 months. Methoxychlor treatment delayed puberty by as much as 10 weeks and reduced fertility and copulatory plug formation in a dose-related manner at the initial mating. During mating, M-treated males exhibited shorter latencies to mount and ejaculate versus control males, but the number of intromissions prior to ejaculation was unaffected, indicating that M enhanced the arousal level in the males in an estrogen-dependent manner. Most treated males eventually mated but time-to-pregnancy was lengthened. Very low sperm counts were associated with infertility, while prolonged delays in puberty reduced fecundity. Methoxychlor treatment with 200 to 400 mg kg-1 day-1 failed to mimic the chronic effects of a sustained (8 months) low dose of estradiol-17 beta (3-mm silastic implants) on pituitary or testicular hormone levels. Estradiol administration increased pituitary weight 4-fold, serum levels of luteinizing hormone (LH) were reduced by almost 50%, and serum prolactin was increased 40-fold, while M did not affect any of these measures. These data demonstrate that M affects the CNS, epididymal sperm numbers, and the accessory sex glands and delays mating without significantly affecting the secretion of LH, prolactin, or testosterone. These data indicate that M did not alter pituitary endocrine function in either an estrogenic or antiandrogenic manner. To our knowledge, these data provide the first in vivo example of such a pronounced degree of target tissue selectivity to an environmental endocrine-disrupting chemical.


Subject(s)
Insecticides/toxicity , Luteinizing Hormone/metabolism , Methoxychlor/toxicity , Prolactin/metabolism , Sexual Behavior, Animal/drug effects , Testis/drug effects , Animals , Dose-Response Relationship, Drug , Insecticides/pharmacology , Luteinizing Hormone/drug effects , Male , Methoxychlor/pharmacology , Pituitary Gland/anatomy & histology , Pituitary Gland/drug effects , Pituitary Gland/physiology , Prolactin/drug effects , Rats , Rats, Inbred Strains , Sperm Count/drug effects , Testis/physiology , Xenobiotics/pharmacology , Xenobiotics/toxicity
2.
Toxicol Ind Health ; 15(1-2): 48-64, 1999.
Article in English | MEDLINE | ID: mdl-10188191

ABSTRACT

In humans and rodents, exposure to antiandrogenic chemicals during sexual differentiation can produce malformations of the reproductive tract. Perinatal administration of 100 or 200 mg vinclozolin (V) kg-1 day-1 during sexual differentiation in rats induces female-like anogenital distance (AGD), retained nipples, cleft phallus with hypospadias, suprainguinal ectopic scrota/testes, a vaginal pouch, epididymal granulomas, and small to absent sex accessory glands in male offspring. Vinclozolin is metabolized to at least two active forms, M1 and M2, that display antiandrogenic activity by binding the androgen receptor (AR). Here, we present information on the reproductive effects of oral treatment with low dosage levels of V during sexual differentiation of the male rat. Vinclozolin was administered to the dam at 0, 3.125, 6.25, 12.5, 25, 50, or 100 mg kg-1 day-1 from gestational day 14 to postnatal day 3 (the period of fetal/neonatal testicular testosterone synthesis and sexual differentiation). At doses of 3.125 mg V kg-1 and above, AGD was significantly reduced in newborn male offspring and the incidence of areolas was increased. These effects were associated with permanent alterations in other androgen-dependent tissues. Ventral prostate weight in one year old male offspring was reduced in all treatment groups (significant at 6.25, 25, 50, and 100 mg kg-1 day-1), and permanent nipples were detected in males at 3.125 (1.4%), 6.25 (3.6%), 12.5 (3.9%), 25 (8.5%), 50 (91%), and 100 (100%) mg V kg-1 day-1. To date, permanent nipples have not been observed in a control male from any study in our laboratory. Vinclozolin treatment at 50 and 100 mg kg-1 day-1 induced reproductive tract malformations and reduced ejaculated sperm numbers and fertility. Even though all of the effects of V likely result from the same initial event (AR binding), the different endpoints displayed a wide variety of dose-response curves and ED50's. The dose-response data for several of the functional endpoints failed to display an obvious threshold. These data demonstrate that V produces subtle alterations in sexual differentiation of the external genitalia, ventral prostate, and nipple tissue in male rat offspring at dosage levels below the previously described no-observed-effect-level (NOEL). These effects occur at a dosage level an order of magnitude below that required to induce malformations and reduce fertility. Hence, multigenerational reproduction studies of antiandrogenic chemicals that were not conducted under the Environmental Protection Agency's new Harmonized Multigenerational Test Guidelines, which include endpoints sensitive to antiandrogens at low dosage levels, could yield a NOEL that is at least an order of magnitude too high.


Subject(s)
Fungicides, Industrial/toxicity , Genitalia, Male/growth & development , Oxazoles/toxicity , Prostate/growth & development , Administration, Oral , Animals , Dose-Response Relationship, Drug , Female , Fungicides, Industrial/pharmacology , Genitalia, Male/drug effects , Male , Nipples/drug effects , Nipples/growth & development , Oxazoles/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects , Prostate/drug effects , Rats , Rats, Long-Evans , Reproduction/drug effects , Risk Assessment
3.
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
4.
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
5.
Mol Endocrinol ; 13(3): 440-54, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10077001

ABSTRACT

Natural and pharmacological androgen receptor (AR) ligands were tested for their ability to induce the AR NH2-terminal and carboxyl-terminal (N/C) interaction in a two-hybrid protein assay to determine whether N/C complex formation distinguishes in vivo AR agonists from antagonists. High-affinity agonists such as dihydrotestosterone, mibolerone, testosterone, and methyltrienolone at concentrations between 0.1 and 1 nM induce the N/C interaction more than 40-fold. The lower affinity anabolic steroids, oxandrolone and fluoxymesterone, require concentrations of 10-100 nM for up to 23-fold induction of the N/C interaction. However no N/C interaction was detected in the presence of the antagonists, hydroxyflutamide, cyproterone acetate, or RU56187, at concentrations up to 1 microM, or with 1 microM estradiol, progesterone, or medroxyprogesterone acetate; each of these steroids at 1-500 nM inhibited the dihydrotestosterone-induced N/C interaction, with medroxyprogesterone acetate being the most effective. In transient and stable cotransfection assays using the mouse mammary tumor virus reporter vector, all ligands displayed concentration-dependent AR agonist activity that paralleled induction of the N/C interaction, with antagonists and weaker agonists failing to induce the N/C interaction. AR dimerization and DNA binding in mobility shift assays and AR stabilization reflected, but were not dependent on, the N/C interaction. The results indicate that the N/C interaction facilitates agonist potency at low physiological ligand concentrations as detected in transcription, dimerization/DNA binding, and stabilization assays. However the N/C interaction is not required for agonist activity at sufficiently high ligand concentrations, nor does its inhibition imply antagonist activity.


Subject(s)
Androgen Antagonists/pharmacology , Androgen Receptor Antagonists , Androgens , Dihydrotestosterone/pharmacology , Medroxyprogesterone Acetate/pharmacology , Animals , COS Cells/drug effects , COS Cells/metabolism , DNA/metabolism , Dimerization , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Luciferases/drug effects , Luciferases/genetics , Luciferases/metabolism , Mammary Tumor Virus, Mouse/genetics , Metribolone/pharmacology , Mice , Nandrolone/analogs & derivatives , Nandrolone/pharmacology , Nitriles/pharmacology , Peptide Fragments/metabolism , Progesterone Congeners/pharmacology , Receptors, Androgen/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Testosterone Congeners/pharmacology , Transcription, Genetic , Transfection
6.
Reprod Fertil Dev ; 10(1): 105-11, 1998.
Article in English | MEDLINE | ID: mdl-9727600

ABSTRACT

Steroid hormone receptors control fundamental events in embryonic development and sex differentiation through their function as ligand-inducible transcription factors. The consequences of disrupting these processes can be especially profound during development due to the crucial role hormones play in controlling transient and irreversible developmental processes. Several environmental chemicals, including metabolites of the fungicide vinclozolin and the pesticide DDT, disrupt male reproductive development and function by inhibiting androgen receptor mediated events. A variety of in vitro and in vivo approaches have been used to determine the molecular basis of environmental antiandrogen toxicity. These chemicals commonly bind androgen receptor with moderate affinity and act as antagonists by inhibiting transcription of androgen dependent genes.


Subject(s)
Androgen Antagonists , Androgen Receptor Antagonists , Environmental Pollutants/adverse effects , Androgens/physiology , Animals , Humans , Male , Receptors, Androgen/physiology , Sex Differentiation/physiology
8.
Toxicol Appl Pharmacol ; 146(1): 11-20, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9299592

ABSTRACT

Male rats exposed in utero to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) display reduced fertility as a consequence of the direct action of TCDD on the epididymides, as well as delayed puberty and altered reproductive organ weights. The current study provides dose-response data for the reproductive effects of TCDD, administered during pregnancy, with an emphasis on the effects of TCDD on testicular, epididymal, and ejaculated sperm numbers. Long Evans Hooded rats were dosed by gavage with 0, 0.05, 0.20, or 0.80 microg TCDD/kg on Day 15 of gestation. After birth, growth, viability, and developmental landmarks were monitored in both male and female offspring. Shortly after puberty (49 and 63 days of age) and at 15 months of age, male offspring were necropsied. Growth and viability of the pups were reduced only at 0.80 microg TCDD/kg, eye opening was accelerated (all dosage groups), and puberty was delayed (at 0.20 and 0.80 microg TCDD/kg). Treated progeny displayed transient reductions in ventral prostate and seminal vesicle weights, while epididymal sperm reserves and glans penis size were permanently reduced. Ejaculated sperm numbers were reduced (45% in the 0.8 and by 25% in the 0.05 and 0.2 microg TCDD/kg dosage groups) to a greater degree than were cauda or caput/corpus epididymal or testicular (unaffected) sperm numbers. In conclusion, administration of TCDD on Day 15 of pregnancy at 0.05 microg/kg altered eye opening and reduced ejaculated sperm counts, while higher dosage levels also delayed puberty and permanently reduced cauda epididymal sperm reserves.


Subject(s)
Fetus/drug effects , Genitalia, Male/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , Dose-Response Relationship, Drug , Female , Male , Organ Size/drug effects , Pregnancy , Rats , Sperm Count/drug effects , Spermatogenesis/drug effects , Testosterone/blood
9.
J Mol Med (Berl) ; 75(3): 198-207, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9106076

ABSTRACT

Industrial chemicals and environmental pollutants can disrupt reproductive development in wildlife and humans by mimicking or inhibiting the action of the gonadal steroid hormones, estradiol and testosterone. The toxicity of these so-called environmental endocrine disruptors is especially insidious during sex differentiation and development due to the crucial role of gonadal steroid hormones in regulating these processes. This review describes the mechanism of toxicity and clinical implications of a new class of environmental chemicals that inhibit androgen-mediated sex development. For several of these chemicals, including the agricultural fungicide vinclozolin and the ubiquitous and persistent 1,1,1-trichloro-2,2-bis (p-chlorophenyl)ethane metabolite, 1,1-dichloro-2,2-bis(p-chlorophenyl) ethylene, the molecular mechanism of action and the adverse developmental effects on male sex differentiation have been elucidated and are used as examples. Environmental chemicals with antiandrogenic activity offer profound implications with regard to recent clinical observations that suggest an increasing incidence of human male genital tract malformations, male infertility, and female breast cancer. Finally, in light of increasing concern over the potential endocrine disrupting effects of environmental pollutants, an in vitro/in vivo investigational strategy is presented which has proved useful in identifying chemicals with antiandrogen activity and their mechanism of action.


Subject(s)
Androgen Antagonists/toxicity , Environmental Pollutants/toxicity , Sex Differentiation/drug effects , Androgen Receptor Antagonists , Androgens , Dichlorodiphenyl Dichloroethylene/toxicity , Gene Expression Regulation , Humans , Hypospadias/etiology , Male , Oxazoles/toxicity , Plant Extracts/toxicity , Serenoa
10.
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
11.
Chem Res Toxicol ; 9(8): 1240-8, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8951225

ABSTRACT

Comparative molecular field analysis (CoMFA), a three-dimensional quantitative structure-activity relationship (3D-QSAR) paradigm, was used to examine the estrogen receptor (ER) binding affinities of a series of structurally diverse natural, synthetic, and environmental chemicals of interest. The CoMFA/3D-QSAR model is statistically robust and internally consistent, and successfully illustrates that the overall steric and electrostatic properties of structurally diverse ligands for the estrogen receptor are both necessary and sufficient to describe the binding affinity. The ability of the model to accurately predict the ER binding affinity of an external test set of molecules suggests that structure-based 3D-QSAR models may be used to supplement the process of endocrine disruptor identification through prioritization of novel compounds for bioassay. The general application of this 3D-QSAR model within a toxicological framework is, at present, limited only by the quantity and quality of biological data for relevant biomarkers of toxicity and hormonal responsiveness.


Subject(s)
Estrogens/chemistry , Animals , Chemical Phenomena , Chemistry, Physical , Cytosol/metabolism , Estrogens/metabolism , Female , In Vitro Techniques , Ligands , Mice , Models, Molecular , Molecular Conformation , Receptors, Estrogen/metabolism , Structure-Activity Relationship , Uterus/metabolism
12.
Toxicology ; 112(3): 173-82, 1996 Sep 02.
Article in English | MEDLINE | ID: mdl-8845038

ABSTRACT

Vinclozolin, a dicarboximide fungicide, alters morphological sex differentiation in male rats following perinatal exposure. The occurrence of these abnormalities correlates with the in vivo formation of two antiandrogenic metabolites of vinclozolin, (i.e. 2-[[(3,5-dichlorophenyl)-carbamoyl]oxy]-2-methyl-3-butenoic acid (Mt) and 3',5'-dichloro-2-hydroxy-2-methylbut-3-enanilide (M2)), which are potent inhibitors of rat androgen receptor binding. As steroid hormone receptors exhibit promiscuity in their ability to bind different ligands, the present study evaluated the ability of these vinclozolin metabolites to bind to the estrogen (ER) and progesterone (PR) receptors in vitro, and to alter ER and PR function following in vivo exposure. To this end, in vitro ligand binding assays demonstrated that both M1 and M2 can compete with endogenous ligand for binding to the PR (Ki = 400 and 60 microM, respectively). In contrast, neither metabolite exhibited the ability to bind ER. Subsequent in vivo studies to evaluate the potential of vinclozolin to alter ER or PR function demonstrate that, (1) the estrogen-dependent increases in uterine weight and PR induction were not altered by vinclozolin; (2) the distribution of nuclear and cytosolic PR was not altered following short-term vinclozolin exposure; and (3) vinclozolin did not disrupt ovulation in cycling female rats. These studies indicate that although vinclozolin metabolites can compete for binding to the PR in vitro, concentrations of these metabolites do not reach sufficient levels to disrupt female reproductive function following short-term in vivo exposure to vinclozolin. In addition, these studies demonstrate the importance of correlating in vitro receptor binding data with in vivo studies in order to understand the physiological consequences of exposure to environmental toxicants.


Subject(s)
Fungicides, Industrial/toxicity , Oxazoles/toxicity , Receptors, Estrogen/antagonists & inhibitors , Receptors, Progesterone/antagonists & inhibitors , Anilides/metabolism , Anilides/toxicity , Animals , Binding, Competitive , Carbamates/metabolism , Carbamates/toxicity , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Female , Fungicides, Industrial/metabolism , In Vitro Techniques , Lethal Dose 50 , Ligands , Male , Organ Size/drug effects , Ovariectomy , Ovulation/drug effects , Oxazoles/metabolism , Rats , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Sex Differentiation/drug effects , Software , Uterus/drug effects , Uterus/metabolism
13.
Drug Metab Dispos ; 24(7): 725-33, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8818568

ABSTRACT

Specific cell types of the mammalian testes demonstrate varying susceptibility to toxic insult by chemical agents. The mammalian testis is divided into two major compartments: seminiferous tubules, the site of spermatogenesis, and interstitium, which contains the Leydig cells. Glutathione S-transferase (GST) expression was examined in isolated compartments of the rat testis and in segments of the epididymis. Western blot analysis revealed the presence of GST class alpha, mu, and pi bands in each of the isolated compartments of the testis, and HPLC analysis of monomeric isoforms provided evidence for differential expression of multiple GST isoforms in testicular compartments. All major isoforms (e.g., forms 1, 2, 3, 4, 6, 7, 8, 9, and 11) were detected in the cytosol of whole testis. Isoform subunit 4 was the major form in the tubule, whereas isoform subunit 11 is the dominant form in the Leydig cells. Isoform subunits 3, 4, and 6 were enriched in the tubules as compared to interstitial or Leydig cells. The preferential action of reproductive toxicants at specific stages of aging may be due to an age-dependent expression of the activating or detoxifying enzymes in the reproductive tract. Therefore, the age-dependent expression of testicular GST isoforms was also examined. Expression of isoform subunits 2 and 4 displayed an age dependence, with the largest increase in these subunits occurring between ages 4 and 15 weeks. Isoform expression did not correlate with serum testosterone levels. HPLC analysis of the GST isoforms in the longitudinal segments of the epididymis and vas deferens revealed differential expression within these segments. Total GST protein and catalytic activity was highest in the caput epididymis and progressively decreased toward the vas deferens. Isoform subunit 2 was the major form expressed in the epididymis. The results of this study indicate that the GSTs are differentially expressed in testicular compartments and epididymal segments, and that this may contribute to susceptibility of different cell types to xenobiotic damage.


Subject(s)
Epididymis/enzymology , Glutathione Transferase/metabolism , Testis/enzymology , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Dinitrochlorobenzene/chemistry , Leydig Cells/enzymology , Liver/enzymology , Male , Rats , Rats, Sprague-Dawley , Seminiferous Tubules/enzymology , Testis/cytology , Vas Deferens/enzymology , Xenobiotics/toxicity
14.
Toxicol Ind Health ; 12(3-4): 515-31, 1996.
Article in English | MEDLINE | ID: mdl-8843568

ABSTRACT

In humans and rodents, exposure to hormonally active chemicals during sex differentiation can produce morphological pseudohermaphrodism (Schardein, 1993; Gray, 1992). For example, hormonally active drugs like DES (estrogenic), Danazol (androgenic), and progestins cause urogenital malformations in the reproductive tracts of humans and rodents. The current discussion will present new information on the effects of toxic chemicals and pesticides that act on reproductive development via novel mechanisms, including germ cell toxicity, antiandrogenicity, and Ah-receptor binding. Information will be presented that describes how exposure during critical stages of life to synthetic chemicals present in our environment, such as benzidine-based dyes, antiandrogenic fungicides, 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD), and PCB congener 169, result in abnormal rodent sex differentiation. In rodents, perinatal exposure to fetal germ cell toxicants reduced the reproductive potential of female, and permanently reduced sperm production in male progeny. Phenotypic sex differentiation, however, was unaffected by these germ cell toxicants. In contrast, antiandrogenic drugs and fungicides induced profound alterations in phenotypic sex differentiation. Effects such as hypospadias, ectopic testes, vaginal pouches, agenesis of the ventral prostate, and nipple retention in male rats were observed commonly. Although these antiandrogens induced no permanent effects in female progeny, another class of chemicals, the Ah-receptor mediated toxicants, did reduce fertility in both male and female rat offspring. Cauda epididymal sperm numbers were reduced permanently in TCDD-exposed male rat and hamster progeny, and female progeny displayed malformations of the external genitalia. Other toxicants produced dramatic alterations of sex differentiation (uterus unicornis, agenesis of the vas and epididymis, and undescended testes), via mechanisms that have not been characterized yet. Since these adult/pubertal alterations resulted from gestational and/or neonatal exposures, future studies should include a comprehensive assessment of reproductive function after perinatal exposure because the developing animal is extremely sensitive to toxicants during sex differentiation, and many of the effects are difficult to detect until late in life.


Subject(s)
Androgen Antagonists/toxicity , Germ Cells/drug effects , Polychlorinated Dibenzodioxins/toxicity , Sex Differentiation/drug effects , Abnormalities, Drug-Induced , Animals , Cricetinae , Embryonic and Fetal Development/drug effects , Female , Germ Cells/cytology , Gestational Age , Male , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Reproduction/drug effects , Sperm Count/drug effects
15.
Toxicol Appl Pharmacol ; 137(2): 219-27, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8661347

ABSTRACT

Comparative molecular field analysis (CoMFA), a three-dimensional quantitative structure-activity relationship (3D-QSAR) paradigm, was used to examine androgen receptor-binding affinities of a series of structurally diverse natural, synthetic, and environmental chemicals of interest. The CoMFA/3D-QSAR model successfully illustrates that the overall steric and electrostatic properties of structurally diverse ligands for the androgen receptor are necessary and sufficient to describe the binding affinity. The practical utility of models of this type is demonstrated using parent compounds in the training set and known as well as putative biological metabolites as test set molecules. The ability of the model to accurately predict binding affinity of test set molecules suggests that structure-based 3D-QSAR models may be used to supplement the process of hazard identification. The application of 3D-QSAR models within a toxicological framework is, at present, limited by the quantity and quality of biological data for relevant biomarkers of toxicity.


Subject(s)
Ligands , Receptors, Androgen/metabolism , Hazardous Substances/analysis , Hazardous Substances/metabolism , Models, Molecular , Receptors, Androgen/chemistry , Steroids/chemistry , Steroids/metabolism , Structure-Activity Relationship
16.
Fundam Appl Toxicol ; 29(1): 1-17, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8838635

ABSTRACT

The ability of foreign compounds to affect the functioning of various endocrine systems is currently thought responsible for a wide variety of effects. The presentations in this Symposium reviewed the evidence for and against the involvement of endocrine systems in several different aspects of reproduction. The mechanism behind the ability of a triazine herbicide to cause enhanced appearance of mammary tumors in one strain of female rats is reviewed by Stevens. The data suggest that enhanced aging, not direct mammary modulation, is responsible. Dietary phytoestrogens, the mediators of their actions, their effects in various biological systems, and the relationships between phytoestrogen producers and consumers are all provocatively and succinctly reviewed by Hughes. Kelce presents the strategy used to dissect the mode and mechanisms of action of a fungicide that opened a new awareness in reproductive toxicology: the possibility of xenobiotics being antiandrogens. Finally, to heighten our understanding of the interplay among hormonal systems in vivo, Hess reviews the data that show that androgens are not the only hormones important in the development of the male reproductive system: the pituitary is shown to play a critical role at specific stages of development. The breadth of these presentations, and the implications of their findings, should make us pause and realize how much there is still to discover about the interaction between the reproductive system and anthropogenic compounds.


Subject(s)
Endocrine Glands/physiology , Hormones/physiology , Reproduction/physiology , Aging/drug effects , Aging/physiology , Androgen Antagonists/toxicity , Animals , Animals, Newborn , Atrazine/toxicity , Estrogens, Non-Steroidal/pharmacology , Female , Fungicides, Industrial/toxicity , Herbicides/toxicity , Humans , Male , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/physiopathology , Pituitary Gland/physiology , Plant Growth Regulators/pharmacology , Rats , Rats, Sprague-Dawley , Reproduction/drug effects , Testis/drug effects , Testis/physiology
17.
J Biol Chem ; 270(34): 19998-20003, 1995 Aug 25.
Article in English | MEDLINE | ID: mdl-7650017

ABSTRACT

The mechanism of antiandrogenic activity of vinclozolin (3-(3,5-dichlorophenyl)-5-methyl-5-vinyloxazolidine-2,4-dione), a dicarboximide fungicide under investigation for its potential adverse effects on human male reproduction, was investigated using recombinant human androgen receptor (AR). The two primary metabolites of vinclozolin in plants and mammals are M1 (2-[[3,5-dichlorophenyl)-carbamoyl]oxy]-2-methyl-3-butenoic acid) and M2 (3',5'-dichloro-2-hydroxy-2-methylbut-3-enanilide). Both metabolites, in a dose-dependent manner, target AR to the nucleus and inhibit androgen-induced transactivation mediated by the mouse mammary tumor virus promoter. M2 is a 50-fold more potent inhibitor than M1 and only 2-fold less than hydroxyflutamide. In the presence of dihydrotestosterone (50 nM), M2 (0.2-10 microM) inhibits androgen-induced AR binding to androgen response element DNA. In the absence of dihydrotestosterone, concentrations of 10 microM M2 or hydroxyflutamide promote AR binding to androgen response element DNA and activation of transcription. Agonist activities of M2 and hydroxyflutamide occur at 10-fold lower concentrations with the mutant AR (Thr877 to Ala) endogenous to LNCaP human prostate cancer cells. The results indicate that androgen antagonists can act as agonists, depending on ligand binding affinity, concentration, and the presence of competing natural ligands.


Subject(s)
Androgen Receptor Antagonists , Androgens , Flutamide/analogs & derivatives , Fungicides, Industrial/toxicity , Oxazoles/toxicity , Animals , Binding, Competitive , Cell Line , DNA/genetics , DNA/metabolism , Dihydrotestosterone/pharmacology , Flutamide/pharmacology , Fungicides, Industrial/metabolism , Humans , Kinetics , Male , Oxazoles/metabolism , Receptors, Androgen/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reproduction/drug effects , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured
18.
Nature ; 375(6532): 581-5, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7791873

ABSTRACT

The increase in the number of reports of abnormalities in male sex development in wildlife and humans coincided with the introduction of 'oestrogenic' chemicals such as DDT (1,1,1-trichloro-2,2-bis(p-chlorophenyl)ethane) into the environment. Although these phenotypic alterations are thought to be mediated by the oestrogen receptor, they are also consistent with inhibition of androgen receptor-mediated events. Here we report that the major and persistent DDT metabolite, p,p'-DDE (1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene), has little ability to bind the oestrogen receptor, but inhibits androgen binding to the androgen receptor, androgen-induced transcriptional activity, and androgen action in developing, pubertal and adult male rats. The results suggest that abnormalities in male sex development induced by p,p'-DDE and related environmental chemicals may be mediated at the level of the androgen receptor.


Subject(s)
Androgen Antagonists/toxicity , Androgen Receptor Antagonists , Dichlorodiphenyl Dichloroethylene/toxicity , Animals , Binding, Competitive , Cell Line , DDT/metabolism , Dichlorodiphenyl Dichloroethylene/chemistry , Dichlorodiphenyl Dichloroethylene/metabolism , Genes, Reporter , Haplorhini , Humans , Male , Molecular Structure , Rats , Receptors, Estrogen/metabolism , Sexual Maturation/drug effects , Transcription, Genetic/drug effects
19.
Toxicol Appl Pharmacol ; 131(1): 108-18, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7878665

ABSTRACT

Prenatal administration of relatively low doses of TCDD alters reproductive development and fertility of the progeny. Fertility was reduced in the progeny of Wistar rats exposed to 0.5 micrograms TCDD/kg/day from Gestational Day (GD) 6 to GD 15. In a three-generation reproduction study, TCDD reduced fertility of Sprague-Dawley rats in the F1 and F2 but not the F0 (no developmental exposure) generation at 0.01 microgram/kg/day in the diet. Furthermore, administration of TCDD on GD 15 (at 0.064 to 1 microgram/kg) both demasculinized and feminized morphology and behavior of Holtzman male rat offspring. Our objectives were to expand the observations of Mably et al. (1992, Toxicol, Appl. Pharmacol. 114, 97-107, 108-117, 118-126) on the effects of gestational administration of a single dose of TCDD to another strain of rat and another species, the hamster. In the first study, Long Evans (LE) hooded rats were dosed by gavage with 1 microgram TCDD/kg on GD 8 (during the period of major organogenesis) or GD 15 (the gestational day used by Mably et al.). In the second study, pregnant Syrian hamsters, a species relatively insensitive to the lethal effects of TCDD, were dosed on GD 11, equivalent to GD 15 in the rat, with TCDD at 2 micrograms/kg. When LE rats were dosed on GD 15, or when hamsters were dosed on GD 11, puberty (preputial separation) was delayed by about 3 days, ejaculated sperm counts were reduced by at least 58%, and epididymal sperm storage was reduced by 38%. Testicular sperm production was less affected. The sex accessory glands were also reduced in size in LE rat offspring treated on GD 15 despite the fact that serum testosterone (T), T production by the testis in vitro, and androgen receptor (AR) levels were not reduced. Some reproductive measures, such as anogenital distance and male sex behavior, were altered by TCDD treatment in rat but not hamster offspring. Since T and AR levels appeared normal in the sex accessory glands and the epididymis following perinatal TCDD exposure, the alterations in these tissues are not likely to have resulted from an alteration of the androgenic status of the male offspring.


Subject(s)
Androgens/physiology , Ejaculation/drug effects , Epididymis/drug effects , Epididymis/physiology , Genitalia, Male/drug effects , Genitalia, Male/growth & development , Polychlorinated Dibenzodioxins/toxicity , Prenatal Exposure Delayed Effects , Receptors, Androgen/physiology , Sperm Count/drug effects , Aging/physiology , Animals , Cricetinae , Female , Fertility/drug effects , Genitalia, Male/anatomy & histology , Luteinizing Hormone/pharmacology , Male , Mesocricetus , Organ Size/drug effects , Pregnancy , Prostate/metabolism , Prostate/ultrastructure , Rats , Rats, Inbred Strains , Receptors, Androgen/metabolism , Seminal Vesicles/metabolism , Seminal Vesicles/ultrastructure , Sexual Behavior, Animal/drug effects , Sexual Maturation/drug effects , Testis/drug effects , Testis/metabolism , Testosterone/biosynthesis , Testosterone/blood
20.
Toxicol Appl Pharmacol ; 130(2): 248-56, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7871538

ABSTRACT

It has been reported that ethane dimethanesulfonate (EDS) is a Leydig cell toxicant that affects rats and hamsters (Kerr et al., 1987), while, in contrast, the Leydig cells of mice are relatively insensitive to the toxicant. In the rat, there is a rapid decline in levels of testosterone (T) within hours after EDS administration. However, T production, spermiogenesis, and fertility are restored within a few weeks as new Leydig cells are formed from undifferentiated cells in the interstitium of the testis. In an earlier study, we found, as expected, that ejaculated sperm counts (ESCs) reached a nadir 10 days after adult rats were dosed with EDS at 65 mg/kg ip along with serum and testicular T, testis and seminal vesicle weights, and in vitro T production, while, in contrast, EDS at 65 mg/kg had no effect on these endpoints in the Syrian hamster (Gray et al., 1992). In the current study, when EDS was administered to 6, 12, and 18 month old hamsters at 100 mg/kg, it produced subtle effects on serum T and sex accessory gland weights, while dramatic effects were seen in similarly exposed rats. In addition, when testes were examined by light microscopy all treated rats displayed severely reduced Leydig cell numbers, while, in contrast, only one-third of the EDS-treated hamsters were affected, having moderately reduced Leydig cell numbers. In support of the histological data, 3 beta-HSD enzyme activity was reduced by 99% of control in EDS-treated rats, but it was reduced by only 35% of control in EDS-treated hamsters. An in vitro analysis of the effects of EDS on LH-stimulated T production by quartered testes demonstrated that the hamster testis was less sensitive to the direct effects of EDS than the rat testis. The IC50 after 3 hr in culture was greater than 1800 micrograms EDS/ml for the hamster quarter testes, while the IC50 for the rat quarter testes was 320 micrograms EDS/ml. In summary, these results demonstrate in vivo and in vitro that Leydig cells of hamsters are less sensitive to EDS than those of the adult rat.


Subject(s)
Leydig Cells/drug effects , Mesylates/toxicity , Aging/metabolism , Animals , Cricetinae , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Mesocricetus , Organ Size/drug effects , Rats , Species Specificity , Testis/drug effects , Testis/pathology , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...