Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-24230975

ABSTRACT

The growth, development, and ERα and Vtg-I gene expressions of Japanese ricefish (Oryzias latipes; medaka) exposed to different concentrations of estriol (E3), including one environmentally relevant concentration, during embryo-adult life stages were evaluated. At the early life stage, fertilized eggs were exposed to 5, 50, 500, 5000ng/L E3 for 15days, and the hatched fry were exposed continuously to the same concentrations for an additional 15days. Exposure to 500 and 5000ng/L E3 resulted in adverse effects on hatchability and time to hatching. At 5000ng/L, the gross abnormality rate was increased and the number of females that hatched was twice that of males. When the fish were exposed to 5-5000ng/L E3 for further 60days, the male hepatosomatic index (HSI) was increased at 5000ng/L. The female gonadosomatic index (GSI) was decreased at 500 and 5000ng/L E3, while the male GSI at 5000ng/L E3 was increased and sex reversal was also found at this concentration. Quantitative RT-PCR showed that the hepatic vitellogenin-I (Vtg-I) genes were up-regulated in females at 500 and 5000ng/L E3 and in males at all E3 concentrations, whereas E3 did not affect estrogen receptor α (ERα) mRNA transcription. These results showed that E3 at environmental concentration of 5ng/L has no adverse effects on growth and development of the Japanese medaka. However, in this study, if we only focused on Vtg gene change in males, E3 had strong estrogenic effects on male medaka under the conditions of these experiments.


Subject(s)
Estriol/administration & dosage , Estriol/toxicity , Estrogens/metabolism , Sex Characteristics , Animals , Female , Male , Oryzias/growth & development , Oryzias/metabolism , Time Factors
2.
Environ Toxicol Chem ; 31(6): 1407-15, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22488655

ABSTRACT

In an effort to assess the combined risk estrone (E1), 17ß-estradiol (E2), 17α-ethinyl estradiol (EE2), and estriol (E3) pose to aquatic wildlife across United States watersheds, two sets of predicted-no-effect concentrations (PNECs) for significant reproductive effects in fish were compared to predicted environmental concentrations (PECs). One set of PNECs was developed for evaluation of effects following long-term exposures. A second set was derived for short-term exposures. Both sets of PNECs are expressed as a 17ß-estradiol equivalent (E2-eq), with 2 and 5 ng/L being considered the most likely levels above which fish reproduction may be harmed following long-term and short-term exposures, respectively. A geographic information system-based water quality model, Pharmaceutical Assessment and Transport Evaluation (PhATE™), was used to compare these PNECs to mean and low flow concentrations of the steroid estrogens across 12 U.S. watersheds. These watersheds represent approximately 19% of the surface area of the 48 North American states, contain 40 million people, and include over 44,000 kilometers of rivers. This analysis determined that only 0.8% of the segments (less than 1.1% of kilometers) of these watersheds would have a mean flow E2-eq concentration exceeding the long-term PNEC of 2.0 ng/L; only 0.5% of the segments (less than 0.8% of kilometers) would have a critical low flow E2-eq exceeding the short-term PNEC of 5 ng/L. Those few river segments where the PNECs were exceeded were effluent dominated, being either headwater streams with a publicly owned treatment works (POTW), or flowing through a highly urbanized environment with one or several POTWs. These results suggest that aquatic species in most U.S. surface waters are not at risk from steroid estrogens that may be present as a result of human releases.


Subject(s)
Endocrine Disruptors/toxicity , Estrogens/toxicity , Water Pollution, Chemical/statistics & numerical data , Water Supply/statistics & numerical data , Animals , Endocrine Disruptors/analysis , Estradiol/analysis , Estradiol/toxicity , Estriol/analysis , Estriol/toxicity , Estrogens/analysis , Estrone/analysis , Estrone/toxicity , Ethinyl Estradiol/analysis , Ethinyl Estradiol/toxicity , Fishes , Humans , Risk Assessment , Rivers/chemistry , United States , Urbanization , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/toxicity
3.
Environ Toxicol Chem ; 31(6): 1396-406, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22488680

ABSTRACT

The authors derive predicted-no-effect concentrations (PNECs) for the steroid estrogens (estrone [E1], 17ß-estradiol [E2], estriol [E3], and 17α-ethinylestradiol [EE2]) appropriate for use in risk assessment of aquatic organisms. In a previous study, they developed a PNEC of 0.35 ng/L for EE2 from a species sensitivity distribution (SSD) based on all available chronic aquatic toxicity data. The present study updates that PNEC using recently published data to derive a PNEC of 0.1 ng/L for EE2. For E2, fish were the most sensitive taxa, and chronic reproductive effects were the most sensitive endpoint. Using the SSD methodology, we derived a PNEC of 2 ng/L for E2. Insufficient data were available to construct an SSD for E1 or E3. Therefore, the authors used in vivo vitellogenin (VTG) induction studies to determine the relative potency of the steroid estrogens to induce VTG. Based on the relative differences between in vivo VTG induction, they derive PNECs of 6 and 60 ng/L for E1 and E3, respectively. Thus, for long-term exposures to steroid estrogens in surface water (i.e., >60 d), the PNECs are 6, 2, 60, and 0.1 ng/L for E1, E2, E3, and EE2, respectively. Higher PNECs are recommended for short-term (i.e., a few days or weeks) exposures.


Subject(s)
Estradiol/toxicity , Estriol/toxicity , Estrone/toxicity , Ethinyl Estradiol/toxicity , Water Pollutants, Chemical/toxicity , Animals , Aquatic Organisms/drug effects , Environmental Exposure/adverse effects , Environmental Exposure/statistics & numerical data , Fishes , No-Observed-Adverse-Effect Level , Risk Assessment , Vitellogenins
4.
Environ Health Perspect ; 119(1): 63-70, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20826375

ABSTRACT

BACKGROUND: Interest and concern regarding potentially estrogenic substances have resulted in development of model systems to evaluate mechanisms of such chemicals. Microarray studies have indicated that estradiol (E2)-stimulated uterine responses can be divided into early and late phases. Comparison of E2 uterine transcript profiles and those of other estrogenic chemicals of interest in vivo indicates mechanisms and activities of test compounds. OBJECTIVES: We compared transcript responses and mechanisms of response using mouse reproductive tracts after treatment with E2, estriol (E3), bisphenol A (BPA), and 2,2-bis(p-hydroxyphenyl)-1,1,1-trichloroethane (HPTE). METHODS: Uterine RNA from ovariectomized wild-type mice, estrogen receptor α (ERα) knockout (αERKO) mice, and mice expressing a DNA-binding-deficient ERα (KIKO) treated with E2, E3, BPA, or HPTE for 2 or 24 hr was analyzed by microarray. Resulting regulated transcripts were compared by hierarchical clustering and correlation analysis, and response patterns were verified by reverse-transcription real-time polymerase chain reaction (RT-PCR). RESULTS: Both xenoestrogens, BPA and HPTE, showed profiles highly correlated to that of E2 in the early response phase (2 hr), but the correlation diminished in the later response phase (24 hr), similar to the known weak estrogen E3. Both xenoestrogens also mimicked E2 in samples from KIKO mice, indicating that they are able to utilize the indirect tethering mode of ERα signaling. No response was detected in ERα-null uteri, indicating that ERα mediates the responses. CONCLUSION: Our study forms a basis on which patterns of response and molecular mechanisms of potentially estrogenic chemicals can be assessed.


Subject(s)
Estrogens/toxicity , Phenols/toxicity , Uterus/drug effects , Animals , Benzhydryl Compounds , Estriol/toxicity , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression/drug effects , Gene Expression Profiling , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Uterus/metabolism
5.
J Cataract Refract Surg ; 35(7): 1260-5, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19545818

ABSTRACT

PURPOSE: To compare the efficacy and safety of estriol and triamcinolone acetonide suspensions in visualizing the prolapsed vitreous body in the anterior chamber after posterior capsule rupture in animal models. SETTING: Tsukuba University Institute of Clinical Medicine, Ibaraki, Japan. METHODS: To evaluate efficacy, triamcinolone acetonide or estriol suspension was injected into the anterior chambers of porcine eyes after intentional posterior capsule rupture. To evaluate safety, triamcinolone acetonide 5.0 mg or estriol in 0.1 mL suspension was injected into the anterior chamber of New Zealand white rabbits. Slitlamp examinations, intraocular pressure (IOP), corneal endothelial cell density (ECD) measurements, and histologic examinations were performed up to 28 days after the injection. RESULTS: Triamcinolone acetonide and estriol were equally effective in allowing visualization of the prolapsed vitreous body in the anterior chamber. The granules of triamcinolone acetonide or estriol disappeared 1 day after the injection and did not affect the IOP or corneal ECD. No statistically significant histological changes were observed in the eyes 28 days after the injection of triamcinolone acetonide or estriol. CONCLUSIONS: Estriol was effective for the visualization of the prolapsed vitreous body in the anterior chamber after posterior capsule rupture. In experimental models, no significant side effects were observed after the injection of estriol in the anterior chamber. Results suggest that estriol is an alternative reagent for visualizing the vitreous body, especially in steroid responders, because it has no glucocorticoid or mineralocorticoid activity.


Subject(s)
Anterior Chamber/pathology , Estriol , Eye Diseases/diagnosis , Lens Capsule, Crystalline/injuries , Vitreous Body/pathology , Animals , Anterior Chamber/drug effects , Cell Count , Endothelium, Corneal/pathology , Estriol/toxicity , Eye Diseases/etiology , Intraocular Pressure , Male , Models, Animal , Prolapse , Rabbits , Rupture , Staining and Labeling/methods , Swine , Triamcinolone Acetonide/toxicity , Vitrectomy
6.
Climacteric ; 10 Suppl 2: 62-5, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17882675

ABSTRACT

Certain estrogen metabolites can act as carcinogens in in vitro and animal experiments. The clinical relevance remains unclear. However, in the presence of factors that could influence estradiol metabolism, such as smoking or genetic polymorphisms, it seems prudent to prefer transdermal therapy to minimize the production of possible toxic metabolites. In addition, various defense mechanisms operate in the physiologic human body that prevent the formation of possible toxic intermediate products of estradiol metabolism, especially during oxidative stress. Only under rare special conditions is it conceivable that the human body cannot react sufficiently.


Subject(s)
Breast Neoplasms/chemically induced , Carcinogens/toxicity , Estradiol/analogs & derivatives , Estriol/analogs & derivatives , Estrogen Replacement Therapy/adverse effects , Steroid 16-alpha-Hydroxylase/toxicity , Administration, Cutaneous , Animals , Breast Neoplasms/metabolism , Estradiol/toxicity , Estriol/toxicity , Estrogens, Catechol , Female , Humans
7.
Article in English | MEDLINE | ID: mdl-16860000

ABSTRACT

Managed by the Organisation for Economic Co-operation and Development (OECD), a comprehensive work is carried out in numerous laboratories to develop test guidelines for the detection of endocrine disrupting chemicals in humans, and various animal species. Development of tests to detect chemicals with endocrine disrupting properties in fish is a part of that work. A Fish Sexual Development Test (FSDT) (an extension of the existing OECD TG 210, fish early life stage toxicity test), proposed as an international test guideline for the detection of endocrine disrupting chemicals, was evaluated by water exposure of juvenile zebrafish to the three natural estrogens: estrone, 17beta-estradiol, and estriol and the synthetic androgen trenbolone (trenbolone acetate). As endpoints, vitellogenin induction and histological changes including changes in sex ratios were investigated. The sex ratio was significantly altered towards females from 49 ng/l estrone, 54 ng/l 17beta-estradiol and 22 microg/l estriol, respectively. An all male population was observed from exposure to 9.7 ng/l trenbolone and above. Significant vitellogenin induction in whole body homogenate was measured after exposure to 14 ng/l estrone, 54 ng/l 17beta-estradiol and 0.6 mug/l estriol, respectively. Significant vitellogenin reduction was measured after exposure to 193 ng/l trenbolone or higher. The present results provide strong evidence that the FSDT is a sensitive test toward estrogenic and especially androgenic exposure and the validation of the FSDT as an OECD test guideline should continue.


Subject(s)
Endocrine Disruptors/toxicity , Sex Ratio , Vitellogenins/analysis , Zebrafish/physiology , Animals , Biomarkers/analysis , Disorders of Sex Development , Estradiol/toxicity , Estriol/toxicity , Estrone/toxicity , Female , Male , Ovary/drug effects , Ovary/physiology , Sexual Development/drug effects , Testis/drug effects , Testis/physiology , Trenbolone Acetate/analogs & derivatives , Trenbolone Acetate/toxicity , Zygote/drug effects
8.
J Endocrinol ; 183(1): 91-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15525577

ABSTRACT

Several investigators have suggested that certain hydroxylated metabolites of 17beta-estradiol (E2) are the proximate carcinogens that induce mammary carcinomas in estrogen-sensitive rodent models. The studies reported here were designed to examine the carcinogenic potential of different levels of E2 and the effects of genotoxic metabolites of E2 in an in vivo model sensitive to E2-induced mammary cancer. The potential induction of mammary tumors was determined in female ACI rats subcutaneously implanted with cholesterol pellets containing E2 (1, 2, or 3 mg), or 2-hydroxyestradiol (2-OH E2), 4-hydroxyestradiol (4-OH E2), 16alpha-hydroxyestradiol (16alpha-OH E2), or 4-hydoxyestrone (4-OH E1) (equimolar to 2 mg E2). Treatment with 1, 2, or 3 mg E2 resulted in the first appearance of a mammary tumor between 12 and 17 weeks, and a 50% incidence of mammary tumors was observed at 36, 19, and 18 weeks respectively. The final cumulative mammary tumor incidence in rats treated with 1, 2, or 3 mg E2 for 36 weeks was 50%, 73%, and 100% respectively. Treatment of rats with pellets containing 2-OH E2, 4-OH E2, 16alpha-OH E2, or 4-OH E1 did not induce any detectable mammary tumors. The serum levels of E2 in rats treated with a 1 or 3 mg E2 pellet for 12 weeks was increased 2- to 6-fold above control values (approximately 30 pg/ml). Treatment of rats with E2 enhanced the hepatic microsomal metabolism of E2 to E1, but did not influence the 2- or 4-hydroxylation of E2). In summary, we observed a dose-dependent induction of mammary tumors in female ACI rats treated continuously with E2; however, under these conditions 2-OH E2, 4-OH E2, 16alpha-OH E2, and 4-OH E1 were inactive in inducing mammary tumors.


Subject(s)
Carcinoma in Situ/chemically induced , Carcinoma, Ductal, Breast/chemically induced , Estradiol/analogs & derivatives , Estrogens/toxicity , Mammary Neoplasms, Experimental/chemically induced , Animals , Dose-Response Relationship, Drug , Drug Implants , Estradiol/toxicity , Estriol/toxicity , Estrogens, Catechol , Female , Hydroxyestrones/toxicity , Rats , Rats, Inbred ACI
9.
Proc Natl Acad Sci U S A ; 100(7): 3913-8, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12655060

ABSTRACT

Mechanisms of estrogen-induced tumorigenesis in the target organ are not well understood. It has been suggested that oxidative stress resulting from metabolic activation of carcinogenic estrogens plays a critical role in estrogen-induced carcinogenesis. We tested this hypothesis by using an estrogen-induced hamster renal tumor model, a well established animal model of hormonal carcinogenesis. Hamsters were implanted with 17beta-estradiol (betaE2), 17alpha-estradiol (alphaE2), 17alpha-ethinylestradiol (alphaEE), menadione, a combination of alphaE2 and alphaEE, or a combination of alphaEE and menadione for 7 months. The group treated with betaE2 developed target organ specific kidney tumors. The kidneys of hamsters treated with alphaE2, alphaEE, or menadione alone did not show any gross evidence of tumor. Kidneys of hamsters treated with a combination of alphaE2 and alphaEE showed early signs of proliferation in the interstitial cells. Kidneys of hamsters treated with a combination of menadione and alphaEE showed foci of tumor with congested tubules and atrophic glomeruli. betaE2-treated tumor-bearing kidneys showed >2-fold increase in 8-iso-prostaglandin F(2alpha) (8-iso-PGF(2alpha)) levels compared with untreated controls. Kidneys of hamsters treated with a combination of menadione and alphaEE showed increased 8-iso-PGF(2alpha) levels compared with untreated controls, whereas no increase in 8-iso-PGF(2alpha) was detected in kidneys of alphaEE-treated group. A chemical known to produce oxidative stress or a potent estrogen with poor ability to produce oxidative stress, were nontumorigenic in hamsters, when given as single agents, but induced renal tumors, when given together. Thus, these data provide evidence that oxidant stress plays a crucial role in estrogen-induced carcinogenesis.


Subject(s)
Estrogens/toxicity , Kidney Neoplasms/chemically induced , Kidney/pathology , Oxidative Stress/physiology , Animals , Cricetinae , Estradiol/toxicity , Estriol/toxicity , Ethinyl Estradiol/toxicity , Immunohistochemistry , Kidney/drug effects , Kidney Neoplasms/pathology , Male , Mesocricetus , Prostaglandins/metabolism , Vitamin K 3/pharmacology
10.
Mutagenesis ; 17(4): 345-52, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12110632

ABSTRACT

Cumulative exposure to oestrogen has been linked to increased risk of breast cancer. Whilst oestrogens induce cancers in rodent bioassays it is unclear whether the mechanisms involved are genotoxic and/or epigenetic. The cytokinesis block micronucleus (CBMN) and the alkaline single cell-gel electrophoresis 'Comet' assays were used to examine MCF-7 cells for chromosomal damage and DNA single-strand breaks (SSBs), respectively. The comet-forming activities of oestrogens were also tested in a 72 h primary culture of cells isolated from freshly expressed breast milk. Micronuclei (MN) were scored in 500 binucleate cells per treatment and SSBs were quantified by comet tail length (CTL) (microm). Effects on mitotic rate (per cent binucleate cells) and cell viability (per cent plating efficiency) were also assessed. beta-Oestradiol, oestrone and oestriol were tested for genotoxicity in the 10(-10)-10(-4) M and 10(-10)-10(-2) M concentration ranges in the CBMN and Comet assays, respectively. Beta-Oestradiol, following 24 h treatment but not 120 h treatment, induced increases (up to 3-fold) in MN at a concentration of 10(-9) M. Oestrone induced dose-related increases in MN (up to 5-fold) following both 24 and 120 h treatment, whereas oestriol appeared not to induce MN. All three oestrogens induced dose-related increases in per cent binucleate cells suggesting that they enhance mitotic rate. In the Comet assay both beta-oestradiol and oestrone induced dose-related increases in SSBs (up to 7-fold over control CTL) and were significantly comet-forming (P < 0.0001) at concentrations as low as 10(-9) and 10(-8) M, respectively, whereas oestriol was less genotoxic. All three oestrogens were significantly comet-forming (P < 0.0001) in a primary culture of breast milk cells, suggesting that they can damage the target cells from which breast cancers may eventually arise.


Subject(s)
Breast/cytology , Estrogens/toxicity , Adult , Breast/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinoma/drug therapy , Carcinoma/genetics , Cell Survival/drug effects , Cells, Cultured , Comet Assay , Dose-Response Relationship, Drug , Estradiol/toxicity , Estriol/toxicity , Estrone/toxicity , Female , Humans , Micronucleus Tests , Milk, Human/cytology , Milk, Human/drug effects , Mitosis/drug effects , Mutagens/toxicity
12.
Cell Tissue Res ; 249(3): 533-40, 1987 Sep.
Article in English | MEDLINE | ID: mdl-3664603

ABSTRACT

The luminal epithelium of adult ovariectomized mice responds to estradiol-17 beta with a synchronised wave of DNA synthesis and mitosis. Estriol, however, although producing a similar DNA-synthetic and mitotic response fails to cause an increase in cell number owing to a wave of cell death occurring at mitosis. In the present study it was shown that cells died by two different routes. The majority died by apoptosis but, unusually, a minority also died by necrosis. In the apoptotic cells the cytoplasm became dense, the endoplasmic reticulum and nuclear cisternae dilated; chromatin became marginated the nucleus shrank and became deeply infolded and contorted. Apoptosis, however, was uncharacteristic in that the nucleus failed to fragment, form caps or show disruption before the cells died by membrane rupture. Furthermore, the cells were frequently lost in sheets from the epithelium into the lumen. Part of the biochemical explanation for this onset of cell death comes from the accelerated loss from the tissue of estriol when compared to estradiol-17 beta. This resulted in a decline in protein and rRNA biosynthesis and a failure to complete ribosomal maturation. Evidence in favour of this explanation came from experiments that showed a return to the estradiol-17 beta level of response and an inhibition of cell death when the occupancy of the estriol receptor was maintained.


Subject(s)
Estradiol/toxicity , Estriol/toxicity , Uterus/pathology , Animals , Cell Survival , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/ultrastructure , Epithelium/drug effects , Epithelium/ultrastructure , Female , Mice , Ovariectomy , Ribosomes/drug effects , Ribosomes/ultrastructure , Uterus/drug effects , Uterus/ultrastructure
13.
Cancer Res ; 43(9): 4114-8, 1983 Sep.
Article in English | MEDLINE | ID: mdl-6871853

ABSTRACT

It had been found previously that exposure of human lymphocytes in vitro to diethylstilbestrol (DES), a synthetic estrogen and known human carcinogen, led to the induction of sister chromatid exchanges. More sister chromatid exchanges were induced in cells from pregnant women than from men. To see if the effects of DES could be induced by other estrogens, lymphocytes from a man and a pregnant woman were treated in vitro with the natural estrogens estradiol and estriol. These did not induce sister chromatid exchanges. To see if the presence of exogenous female hormones might be responsible for the increase in DES-induced sister chromatid exchanges seen in cells from pregnant women, lymphocytes from a man and a pregnant woman were also treated in vitro simultaneously with DES, estradiol, estriol, and progesterone. Treatments with these exogenous hormones did not alter the number of DES-induced sister chromatid exchanges. Our previous studies showed that DES also inhibits in vitro proliferation of lymphocytes. The results reported here show that estradiol also strongly inhibits this proliferation but that estriol is only a weak inhibitor. The cause of delayed cell proliferation induced by DES and estradiol was 2-fold: some of the cells were delayed in phytohemagglutinin-mediated blast cell transformation but, additionally, most cells had a prolonged cell cycle because of an extended G2 phase. These studies also showed that DES, but not estradiol or estriol, induced a low level of polyploidy in human lymphocytes.


Subject(s)
Cell Cycle/drug effects , Crossing Over, Genetic/drug effects , Diethylstilbestrol/toxicity , Estradiol/toxicity , Estriol/toxicity , Lymphocytes/physiology , Sister Chromatid Exchange/drug effects , Cells, Cultured , DNA Replication/drug effects , Female , Humans , Interphase/drug effects , Kinetics , Lymphocytes/drug effects , Male , Metaphase/drug effects , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...