Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
SAR QSAR Environ Res ; 13(1): 69-88, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12074393

RESUMO

Recent legislation mandates the US Environmental Protection Agency (EPA) to develop a screening and testing program for potential endocrine disrupting chemicals (EDCs), of which xenoestrogens figure prominently. Under the legislation, a large number of chemicals will undergo various in vitro and in vivo assays for their potential estrogenicity, as well as other hormonal activities. There is a crucial need for priority setting before this strategy can be effectively implemented. Here we report an integrated computational approach to priority setting using estrogen receptor (ER) binding as an example. This approach rationally integrates different predictive computational models into a "Four-Phase" scheme so that it can effectively identify potential estrogenic EDCs based on their predicted ER relative binding affinity (RBA). The system has been validated using an in-house ER binding assay dataset for 232 chemicals that was designed to have both broad structural diversity and a wide range of binding affinities. When applied to 58,000 chemicals identified by Walker et al. as candidates for endocrine disruption screening, some 9100 chemicals were predicted to bind to ER. Of these, only 3600 were expected to bind to ER at RBA values up to 100,000-fold less than that of 17beta-estradiol. The method ruled out 83% of the chemicals as non-binders with a very low rate of false negatives. We believe that the same integrated scheme will be equally applicable to endpoints of other endocrine disrupting mechanisms, e.g. androgen receptor binding.


Assuntos
Estrogênios não Esteroides/efeitos adversos , Modelos Químicos , Receptores de Estrogênio/efeitos dos fármacos , Bioensaio , Sistema Endócrino/efeitos dos fármacos , Sistema Endócrino/fisiologia , Estrogênios não Esteroides/farmacologia , Previsões , Humanos , Receptores de Estrogênio/fisiologia , Relação Estrutura-Atividade , Xenobióticos/efeitos adversos , Xenobióticos/farmacologia
2.
J Chem Inf Comput Sci ; 41(1): 186-95, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11206373

RESUMO

Endocrine disruptors (EDs) have a variety of adverse effects in humans and animals. About 58,000 chemicals, most having little safety data, must be tested in a group of tiered assays. As assays will take years, it is important to develop rapid methods to help in priority setting. For application to large data sets, we have developed an integrated system that contains sequential four phases to predict the ability of chemicals to bind to the estrogen receptor (ER), a prevalent mechanism for estrogenic EDs. Here we report the results of evaluating two types of QSAR models for inclusion in phase III to quantitatively predict chemical binding to the ER. Our data set for the relative binding affinities (RBAs) to the ER consists of 130 chemicals covering a wide range of structural diversity and a 6 orders of magnitude spread of RBAs. CoMFA and HQSAR models were constructed and compared for performance. The CoMFA model had a r2 = 0.91 and a q2LOO = 0.66. HQSAR showed reduced performance compared to CoMFA with r2 = 0.76 and q2LOO = 0.59. A number of parameters were examined to improve the CoMFA model. Of these, a phenol indicator increased the q2LOO to 0.71. When up to 50% of the chemicals were left out in the leave-N-out cross-validation, the q2 remained significant. Finally, the models were tested by using two test sets; the q2pred for these were 0.71 and 0.62, a significant result which demonstrates the utility of the CoMFA model for predicting the RBAs of chemicals not included in the training set. If used in conjunction with phases I and II, which reduced the size of the data set dramatically by eliminating most inactive chemicals, the current CoMFA model (phase III) can be used to predict the RBA of chemicals with sufficient accuracy and to provide quantitative information for priority setting.


Assuntos
Estrogênios/química , Estrogênios/farmacologia , Modelos Moleculares , Relação Quantitativa Estrutura-Atividade , Animais , Ligação Competitiva , Estrogênios/metabolismo , Ratos , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo
3.
Toxicol Sci ; 54(1): 138-53, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10746941

RESUMO

We have utilized a validated (standardized) estrogen receptor (ER) competitive-binding assay to determine the ER affinity for a large, structurally diverse group of chemicals. Uteri from ovariectomized Sprague-Dawley rats were the ER source for the competitive-binding assay. Initially, test chemicals were screened at high concentrations to determine whether a chemical competed with [3H]-estradiol for the ER. Test chemicals that exhibited affinity for the ER in the first tier were subsequently assayed using a wide range of concentrations to characterize the binding curve and to determine each chemical's IC50 and relative binding affinity (RBA) values. Overall, we assayed 188 chemicals, covering a 1 x 10(6)-fold range of RBAs from several different chemical or use categories, including steroidal estrogens, synthetic estrogens, antiestrogens, other miscellaneous steroids, alkylphenols, diphenyl derivatives, organochlorines, pesticides, alkylhydroxybenzoate preservatives (parabens), phthalates, benzophenone compounds, and a number of other miscellaneous chemicals. Of the 188 chemicals tested, 100 bound to the ER while 88 were non-binders. Included in the 100 chemicals that bound to the ER were 4-benzyloxyphenol, 2,4-dihydroxybenzophenone, and 2,2'-methylenebis(4-chlorophenol), compounds that have not been shown previously to bind the ER. It was also evident that certain structural features, such as an overall ring structure, were important for ER binding. The current study provides the most structurally diverse ER RBA data set with the widest range of RBA values published to date.


Assuntos
Receptores de Estrogênio/metabolismo , Xenobióticos/metabolismo , Animais , Benzofenonas/metabolismo , Benzofenonas/farmacologia , Ligação Competitiva/efeitos dos fármacos , Compostos de Bifenilo/metabolismo , Compostos de Bifenilo/farmacologia , Feminino , Inseticidas/metabolismo , Inseticidas/farmacologia , Ligantes , Fenóis/metabolismo , Fenóis/farmacologia , Ácidos Ftálicos/metabolismo , Ácidos Ftálicos/farmacologia , Ratos , Ratos Sprague-Dawley , Esteroides/metabolismo , Útero/metabolismo
4.
Biol Reprod ; 61(1): 91-100, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10377036

RESUMO

The synthetic estrogen diethylstilbestrol (DES) is a potent neonatal endocrine disruptor in the hamster. To test the specificity of this phenomenon, newborn animals were treated with 100 microgram of either DES or the natural estrogen, estradiol-17beta (E2). Of the two, neonatal DES exposure caused greater morphological disruption throughout the female reproductive tract in prepubertal animals and in adults that either retained their ovaries or were ovariectomized and then given the same levels of chronic E2 stimulation. In the uterus, a characteristic histopathological profile, including enhancement of both hyperplastic and apoptotic activity, was initiated prepubertally and exclusively in the endometrial epithelial cell compartment from the neonatally DES-treated animals and then was promoted by E2 stimulation during adulthood. Interestingly, apoptotic activity was not detected in an area of endometrial epithelium that progressed to the neoplastic state in a DES-exposed animal. Lastly, chronic estrogen induction of lactoferrin was also restricted to the DES-exposed endometrium. We conclude that 1) DES is more active than E2 as a perinatal endocrine disruptor in the hamster and 2) this experimental system should be generally useful as a means to screen compounds for such activity and then probe their mechanism of action.


Assuntos
Dietilestilbestrol/farmacologia , Estradiol/farmacologia , Estrogênios não Esteroides/farmacologia , Genitália Feminina/efeitos dos fármacos , Envelhecimento , Animais , Animais Recém-Nascidos , Cricetinae , Endométrio/anatomia & histologia , Endométrio/efeitos dos fármacos , Feminino , Genitália Feminina/anatomia & histologia , Genitália Feminina/crescimento & desenvolvimento , Imuno-Histoquímica , Mesocricetus , Ovariectomia , Útero/anatomia & histologia , Útero/efeitos dos fármacos
5.
Biol Reprod ; 60(5): 1172-82, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10208980

RESUMO

Insulin-like growth factor (IGF)-I and IGF-II play a number of important roles in growth and differentiation, and IGF-binding proteins (IGFBPs) modulate IGF biological activity. IGF-I has been shown previously to be essential for normal uterine development. Therefore, we used in situ hybridization assays to characterize the unique tissue- and developmental stage-specific pattern of expression for each IGF and IGFBP gene in the rat uterus during perinatal development (gestational day [GD]-20 to postnatal day [PND]-24). IGF-I and IGFBP-1 mRNAs were expressed in all uterine tissues throughout this period. IGFBP-3 mRNA was not detectable at GD-20 but became detectable beginning at PND-5, and the signal intensity appeared to increase during stromal and muscle development. IGFBP-4 mRNA was abundant throughout perinatal development in the myometrium and in the stroma, particularly near the luminal epithelium. IGFBP-5 mRNA was abundantly expressed in myometrium throughout perinatal development. IGFBP-6 mRNA was detected throughout perinatal development in both the stroma and myometrium in a diffuse expression pattern. IGF-II and IGFBP-2 mRNAs were not detected in perinatal uteri. Our results suggest that coordinated temporal and spatial expression of IGF-I and its binding proteins (IGFBP-1,-3,-4,-5, and -6) could play important roles in perinatal rodent uterine development.


Assuntos
Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/biossíntese , RNA Mensageiro/biossíntese , Somatomedinas/biossíntese , Útero/embriologia , Útero/metabolismo , Animais , Feminino , Hibridização In Situ , Gravidez , Sondas RNA , Ratos , Ratos Sprague-Dawley
6.
J Toxicol Environ Health ; 52(5): 385-401, 1997 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-9388532

RESUMO

Physiologically based pharmacokinetic (PBPK) models for pregnancy are inherently more complex than conventional PBPK models due to the growth of the maternal and embryo/fetal tissues. Physiological parameters such as compartmental volumes or flow rates are relatively constant at any particular time during gestation when an acute experiment might be conducted, but vary greatly throughout the course of gestation (e.g., contrast relative fetal weight during the first month of gestation with the ninth month). Maternal physiological parameters change during gestation, depending upon the particular system; for example, cardiac output increases by approximately 50% during human gestation; plasma protein concentration decreases during pregnancy; overall metabolism remains fairly constant. Maternal compartmental volumes may change by 10-30%; embryo/fetal volume increases over a billionfold from conception to birth. Data describing these physiological changes in the human are available from the literature. Human embryo/fetal growth can be well described using the Gompertz equation. By contrast, very little of these same types of data is available for the laboratory animal. In the rodent there is a dearth of information during organogenesis as to embryo weights, and even less organ or tissue weight or volume data during embryonic or fetal periods. Allometric modeling offers a reasonable choice to extrapolate (approximately) from humans to animals; validation, however, is confined to comparisons with limited data during the late embryonic and fetal periods of development (after gestation d 11 in the rat and mouse). Embryonic weight measurements are limited by the small size of the embryo and the current state of technology. However, the application of the laser scanning confocal microscope (LSCM) to optically section intact embryos offers the capability of precise structural measurements and computer-generated three-dimensional reconstruction of early embryos. Application of these PBPK models of pregnancy in laboratory animal models at teratogenically sensitive periods of development provides exposure values at specific target tissues. These exposures provide fundamentally important data to help design and interpret molecular probe investigations into mechanisms of teratogenesis.


Assuntos
Embrião de Mamíferos/fisiologia , Feto/fisiologia , Modelos Biológicos , Farmacocinética , Gravidez/fisiologia , Animais , Embrião de Mamíferos/metabolismo , Feminino , Feto/metabolismo , Idade Gestacional , Humanos , Gravidez/metabolismo , Roedores , Especificidade da Espécie
7.
Environ Health Perspect ; 105(10): 1116-24, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9353176

RESUMO

The recognition of adverse effects due to environmental endocrine disruptors in humans and wildlife has focused attention on the need for predictive tools to select the most likely estrogenic chemicals from a very large number of chemicals for subsequent screening and/or testing for potential environmental toxicity. A three-dimensional quantitative structure-activity relationship (QSAR) model using comparative molecular field analysis (CoMFA) was constructed based on relative binding affinity (RBA) data from an estrogen receptor (ER) binding assay using calf uterine cytosol. The model demonstrated significant correlation of the calculated steric and electrostatic fields with RBA and yielded predictions that agreed well with experimental values over the entire range of RBA values. Analysis of the CoMFA three-dimensional contour plots revealed a consistent picture of the structural features that are largely responsible for the observed variations in RBA. Importantly, we established a correlation between the predicted RBA values for calf ER and their actual RBA values for human ER. These findings suggest a means to begin to construct a more comprehensive estrogen knowledge base by combining RBA assay data from multiple species in 3D-QSAR based predictive models, which could then be used to screen untested chemicals for their potential to bind to the ER. Another QSAR model was developed based on classical physicochemical descriptors generated using the CODESSA (Comprehensive Descriptors for Structural and Statistical Analysis) program. The predictive ability of the CoMFA model was superior to the corresponding CODESSA model.


Assuntos
Exposição Ambiental , Estrogênios/metabolismo , Receptores de Estrogênio/metabolismo , Humanos , Modelos Lineares , Estrutura Molecular , Especificidade da Espécie , Relação Estrutura-Atividade
8.
Cancer Res ; 57(10): 1903-8, 1997 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9157983

RESUMO

Prenatal exposure to the synthetic estrogen diethylstilbestrol (DES) causes morphogenetic alterations and neoplasia in the human reproductive tract. In the hamster, neonatal DES exposure alters early uterine morphogenesis and induces endometrial adenocarcinomas in adults. We now demonstrate that the preneoplastic stages of this phenomenon in the hamster reflect an abnormal uterotropic response to estrogen that is characterized by hyperplastic lesions in the endometrial epithelium and includes an immune and/or inflammatory component. Interestingly, biochemical and in situ analysis revealed that the hyperplastic epithelium is also an active site of cell death by apoptosis. To further probe the mechanism of this phenomenon, uteri from 7-day-old control or DES-exposed donors were transplanted into the cheek pouches of control or neonatally DES-exposed adult hosts, and both host groups were treated to provide high circulating levels of estradiol. Among the four ectopic scenarios, histopathological lesions (epithelial hyperplasia, dysplasia, and apoptosis), segregated almost exclusively to the two that consisted of neonatally DES-exposed uteri. The virtual absence of lesions in control uteri transplanted to DES hosts eliminated host systemic factors as causative agents. Therefore, we conclude that DES or its metabolites alter the cellular physiology and/or composition of the developing uterus (initiating event) in such a way that it thereafter responds abnormally to estrogenic stimulation (promoting event). These observations serve to further define a unique experimental system for probing: (a) various aspects of the clinical "DES Syndrome"; (b) how estrogen regulates normal uterine growth and morphogenesis; and (c) how this process can degenerate to the unregulated neoplastic state.


Assuntos
Apoptose/efeitos dos fármacos , Dietilestilbestrol/toxicidade , Hiperplasia Endometrial/induzido quimicamente , Estradiol/farmacologia , Lesões Pré-Cancerosas/induzido quimicamente , Neoplasias Uterinas/induzido quimicamente , Útero/efeitos dos fármacos , Útero/patologia , Animais , Animais Recém-Nascidos , Bochecha , Cricetinae , Hiperplasia Endometrial/patologia , Estradiol/sangue , Feminino , Masculino , Mesocricetus , Lesões Pré-Cancerosas/patologia , Gravidez , Estimulação Química , Neoplasias Uterinas/patologia , Útero/transplante
9.
Biol Reprod ; 56(5): 1239-44, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9160724

RESUMO

In the developing rodent uterus, the estrogen agonist activity of triphenylethylene antiestrogens such as tamoxifen alters uterine luminal epithelium morphology and inhibits uterine gland genesis. We examined uterine growth and differentiation in female offspring from date-mated Sprague-Dawley rats given the structurally related antiestrogen, toremifene, by s.c. injection in 10 microl of sesame oil on postnatal days (PND) 1-5, 10-14, or 20-24. Toremifene given on PND 10-14, a period of rapid uterine gland differentiation, caused a dose-related increase in uterine weight, tripled luminal epithelium cell height, and completely inhibited uterine gland development on PND 14 at doses of 10 microg or higher. Based on this dose-response analysis, a 10-microg dose of toremifene was chosen to assess uterine development after neonatal exposure (PND 1-5). Uterine weights and luminal epithelium cell heights were significantly increased by toremifene on PND 5 but returned to control levels by PND 26. Uterine gland numbers were reduced to 50% those of controls on PND 26. Dose-related uterine weight and luminal epithelium cell height increases were also observed in rats given toremifene on PND 20-24. This estrogen agonist activity of toremifene, revealed primarily in the uterine luminal epithelium, indicates that toremifene is developmentally toxic.


Assuntos
Antagonistas de Estrogênios/farmacologia , Toremifeno/farmacologia , Útero/efeitos dos fármacos , Útero/crescimento & desenvolvimento , Fatores Etários , Animais , Animais Recém-Nascidos , Epitélio/efeitos dos fármacos , Epitélio/crescimento & desenvolvimento , Antagonistas de Estrogênios/administração & dosagem , Feminino , Ratos , Ratos Sprague-Dawley , Toremifeno/administração & dosagem
10.
Biol Reprod ; 55(6): 1221-30, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8949877

RESUMO

During the first 2 wk of postnatal life, the rodent uterus undergoes a period of marked growth and differentiation. To further examine the role of the estrogen receptor (ER) in the mediation of uterine development, we analyzed the ontogeny of ER mRNA expression in the postnatal rat uterus using in situ hybridization. ER mRNA was present in the uterine stroma on the day of birth and progressively increased in abundance during the first 2 wk of postnatal life. In contrast, ER mRNA was not detectable in the luminal epithelium at birth and did not become abundant in this region until postnatal day (P) 7. ER mRNA abundance increased in the luminal epithelium and in the invaginating and fully formed glandular epithelium during the second week of life. At P21 ER mRNA was more abundant in the glandular epithelium than in any other uterine cell type. These results are consistent with, and extend the findings of, previous studies using uterine homogenate binding assays and immunohistochemistry to define ER ontogeny in rodents. Delineation of the temporal and cell-type specific pattern of ER mRNA ontogeny in the postnatal rat uterus furthers our understanding of the molecular basis of both endogenous and exogenous estrogen effects on uterine growth and development.


Assuntos
Expressão Gênica , Receptores de Estrogênio/genética , Útero/crescimento & desenvolvimento , Envelhecimento , Animais , Autorradiografia , Epitélio/metabolismo , Feminino , Hibridização In Situ , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Útero/metabolismo
11.
Biol Reprod ; 54(1): 160-7, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8838013

RESUMO

To assess the effects of the steroidal antiestrogen ICI 182,780 on postnatal uterine development, female Sprague-Dawley rats were given s.c. injections of ICI 182,780 (0.1-100 micrograms/rat) on each of postnatal days (PND) 10-14. ICI 182,780 inhibited uterine growth, as measured by uterine weight, in a dose-dependent manner but had no effect on either uterine luminal epithelium hypertrophy or gland genesis. Immunohistochemical analysis revealed that ICI 182,780 (10 micrograms) markedly reduced uterine estrogen receptor (ER) immunoreactivity in all uterine cell types while tamoxifen (10 micrograms) increased ER immunoreactivity, most notably in the luminal epithelium. In addition, tamoxifen increased uterine weight and induced luminal epithelium hypertrophy but inhibited uterine gland genesis--outcomes also seen with synthetic estrogens such as diethylstilbestrol. To test the hypothesis that these effects are a consequence of the estrogen agonist activity of tamoxifen, rats were cotreated with ICI 182,780 (10 micrograms, PND 8-14) and tamoxifen (10 micrograms, PND 10-14). ICI 182,780 greatly reduced or completely blocked tamoxifen-induced uterine weight gain, luminal epithelium hypertrophy, tamoxifen-induced ER immunoreactivity, and the inhibition of uterine gland genesis. ICI 182,780 given daily on PND 1-5 did not alter PND 5 uterine weight or uterine differentiation on PND 26. We conclude that postnatal exposure to ICI 182,780 does not affect uterine growth or differentiation at an age when the uterus is not dependent on estrogen for growth, i.e., PND 1-5, but does inhibit later endogenous estrogen-dependent uterine growth. The blockade of tamoxifen-induced uterine developmental alterations by ICI 182,780 demonstrates that these tamoxifen effects result from its estrogen agonist activity.


Assuntos
Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Estrogênios/fisiologia , Tamoxifeno/farmacologia , Útero/efeitos dos fármacos , Útero/crescimento & desenvolvimento , Animais , Epitélio/metabolismo , Estradiol/farmacologia , Feminino , Fulvestranto , Imuno-Histoquímica , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/metabolismo , Útero/metabolismo
12.
Biol Reprod ; 53(4): 863-72, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8547482

RESUMO

We tested the hypothesis that ovarian and/or adrenal factors contribute to uterine growth, differentiation, and acquisition of estrogen responsiveness in the postnatal rat. In untreated rats, normalized uterine weight (5.2 mg/10 g BW on postnatal days [PND] 1-10) increased by about 35% on PND 11-19; PND 6 ovariectomy (OVX) eliminated this increase. Adrenalectomy (ADX) on PND 6 lowered normalized uterine weight only when combined with OVX and only on PND 16 and 19, demonstrating the presence of uterotropic adrenal products. OVX +/- ADX on PND 6 delayed uterine gland genesis by about 2 days but did not alter final gland numbers. There was no change in the normal pattern of luminal epithelium morphology. A uterotropic response to 17 beta-estradiol (E2) occurred on PND 10 in OVX rats and on PND 12 in OVX + ADX rats, but not until PND 14 in controls. We conclude that normal uterine growth is independent of the ovaries and adrenals prior to PND 10, partially dependent during PND 10-15, and completely dependent during PND 16-26. Additionally, a uterotropic response to exogenous E2 occurs concomitantly with, but independently of, the endogenous estrogen surge. Finally, while uterine gland genesis is slightly retarded by OVS +/- ADX, estrogens from these organs do not induce uterine differentiation.


Assuntos
Glândulas Suprarrenais/fisiologia , Diferenciação Celular , Ovário/fisiologia , Útero/crescimento & desenvolvimento , Adrenalectomia , Animais , Desoxicorticosterona/farmacologia , Estradiol/farmacologia , Feminino , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , Ratos , Ratos Sprague-Dawley , Útero/citologia
13.
Proc Soc Exp Biol Med ; 208(3): 307-13, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7878071

RESUMO

Phytoestrogens found in clover, alfalfa, and soybeans have caused reproductive toxicity in several mammalian species. Other estrogens, such as diethylstilbestrol (DES), are developmental toxicants, reducing uterine estrogen receptor (ER) concentration, altering uterine growth, and eliciting reproductive tract abnormalities in the rat. The present study examines the effects of the phytoestrogens coumestrol and equol on the developing rat uterus. Various doses of these compounds were injected sc on postnatal days (PND) 1-5 or 1-10 to ascertain their effects on uterine weight and ER levels, and on PND 10-14 to determine their effects on uterine weight and gland genesis. Coumestrol (PND 1-5) was about 10(-3) as potent as DES in increasing uterine weight (wet or dry) while equol increased dry weight only, with a potency of 10(-5) that of DES. Although the 10 and 100 micrograms doses of coumestrol (PND 1-5 or 1-10) initially increased uterine wet weight, by PND 20 uterine weights either equaled or fell significantly below controls. The 100-micrograms dose of coumestrol (PND 1-5 or 1-10) reduced ER levels at all ages, while the 10-micrograms dose was not as effective. Equol (PND 1-5 or 1-10) did not affect ER levels. Premature uterine gland genesis occurred by PND 9 for the PND 1-5 100-micrograms coumestrol dose. When given on PND 10-14 (the critical period of gland genesis), 10 micrograms and 100 micrograms of coumestrol and 10 micrograms DES greatly increased uterine weight, while no effect was elicited by equol. Although coumestrol and equol inhibited uterine gland genesis in a dose-dependent manner, neither abolished gland genesis as did 10 micrograms of DES or tamoxifen. These data demonstrate that coumestrol elicits uterine biochemical and morphological toxicity much like DES. Equol decreased uterine gland number without increasing uterine wet weight or luminal epithelial hypertrophy, which is inconsistent with either an estrogenic or antiestrogenic action in the uterus.


Assuntos
Cromanos/toxicidade , Cumestrol/toxicidade , Isoflavonas , Útero/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Dietilestilbestrol/toxicidade , Relação Dose-Resposta a Droga , Equol , Feminino , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/análise , Útero/crescimento & desenvolvimento
14.
Proc Soc Exp Biol Med ; 208(1): 67-71, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7892298

RESUMO

The phytoestrogens, coumestrol and equol, are weakly estrogenic. Here, we have examined their ability to induce responses in the neonatal rat uterus. Potent estrogens such as diethylstilbestrol (DES) and 17 beta-estradiol which initially double uterine weight on postnatal Day (PND) 5 when given on PND 1-5 subsequently reduce both uterine growth and gland development at later ages. In this study, Sprague-Dawley pups were treated neonatally (PND 1-5) with various doses of coumestrol and equol, and sacrificed at different ages to determine alterations in biochemical and morphological endpoints. Other rats were injected with the same compounds during the critical period of gland genesis (PND 10-14) to examine their effects on gland development. At the 100 micrograms coumestrol dose, on PND 1-5, premature gland development and increased uterine weight were observed. However, at later ages, uterine weight was significantly lowered and there was a severe suppression in the estrogen receptor (ER) levels. Equol lowered uterine weight at the later ages but did not affect ER levels. When given on PND 10-14, both coumestrol and equol caused a dose-dependent inhibition of gland genesis though not as severe as either DES or tamoxifen. Coumestrol was about 10(3) more potent than equol as an estrogen and behaved much like DES with respect to its effects on uterine weight, glands, and ER levels. At the doses used in this study, equol failed to demonstrate either estrogenic or antiestrogenic activity.


Assuntos
Cromanos/farmacologia , Cumestrol/farmacologia , Estrogênios não Esteroides/farmacologia , Isoflavonas , Útero/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Equol , Feminino , Hipertrofia , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/biossíntese , Útero/crescimento & desenvolvimento , Útero/patologia
15.
Proc Soc Exp Biol Med ; 203(3): 297-303, 1993 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8516342

RESUMO

Triphenylethylene antiestrogens are considered weak estrogen agonists based on their limited ability to induce estrogen responses, in particular uterine growth. We compared the uterotrophic activity of naturally occurring and synthetic estrogens with that of antiestrogens by quantitating uterine wet weight and hypertrophy in the uterine luminal and glandular epithelium. Immature rats received five daily injections of either an estrogen (17 beta-estradiol [E2], diethylstilbestrol [DES], or ethynyl estradiol [EE]) or an antiestrogen (tamoxifen [TAM], monohydroxytamoxifen [OH-TAM], or clomiphene citrate [CC]) (0.001-100 micrograms/rat/day) subcutaneously in sesame oil and were sacrificed approximately 2 hr after the last injection. Both DES and EE increased uterine weight at doses between 0.01-100 micrograms/rat/day; E2 was about 10-fold less potent. The antiestrogens increased uterine weight only slightly. DES, EE, and the three antiestrogens each increased luminal epithelium hypertrophy to over 3-fold above that in controls. While the potencies of these synthetic compounds differed (DES = EE > OH-TAM > TAM = CC), each hypertrophic response occurred over two log doses, and the response curves displayed identical slopes. E2, however, required a range of four log doses to achieve the same degree of luminal epithelium hypertrophy. The three antiestrogens elicited glandular epithelium hypertrophy up to 2-fold above controls at the same doses that induced luminal epithelium hypertrophy; the order of potency was OH-TAM > TAM = CC. However, the three estrogens increased glandular epithelium hypertrophy only marginally. Thus, under dosing conditions commonly used to assess uterotrophic activity, these "antiestrogens" are complete, albeit less potent, estrogen agonists in the luminal epithelium and, unlike estrogens, induce hypertrophy in the glandular epithelium.


Assuntos
Antagonistas de Estrogênios/farmacologia , Estrogênios/farmacologia , Útero/patologia , Animais , Clomifeno/farmacologia , Dietilestilbestrol/farmacologia , Relação Dose-Resposta a Droga , Epitélio/patologia , Estradiol/farmacologia , Antagonistas de Estrogênios/administração & dosagem , Estrogênios/administração & dosagem , Etinilestradiol/farmacologia , Feminino , Hipertrofia , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tamoxifeno/farmacologia , Útero/efeitos dos fármacos
16.
In Vitro Cell Dev Biol Anim ; 29A(5): 408-14, 1993 May.
Artigo em Inglês | MEDLINE | ID: mdl-8314735

RESUMO

We have developed a system for serum-free culture of separated uterine epithelium and stroma from 11-day-old rats recombined on extracellular matrix extracted from Englebreth-Holm-Swarm tumors. Epithelium grew and, after 2 days in culture, developed into luminal epithelial spheres (LES) surrounding a fluid-filled lumen. Individual LES cells maintained epithelial cell characteristics such as basally located nuclei, apical microvilli (oriented toward the lumen), lateral membranes with interdigitations and desmosomes, secretory Golgi complexes, and abundant mitochondria and rough endoplasmic reticulum. Secretory vesicles were ubiquitous throughout the luminal fluid. Addition of 17 beta-estradiol to the growth medium increased the number and longevity of the LES. Prior exposure of uteri to tamoxifen via s.c. injection in vivo on postnatal Days 1 to 5 reduced or completely inhibited formation of LES in vitro. These effects occurred regardless of whether the stromal or epithelial component of the recombinant tissue was exposed to tamoxifen. These data suggest a directive property of neonatal stroma in culture resulting in the formation of highly secretory spherical epithelial structures completely enclosing a lumen. LES formation is responsive to both estrogen (positive response) and antiestrogen (negative response).


Assuntos
Matriz Extracelular/fisiologia , Tamoxifeno/farmacologia , Útero/citologia , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Divisão Celular , Separação Celular , Células Cultivadas , Células Epiteliais , Epitélio/efeitos dos fármacos , Epitélio/ultraestrutura , Feminino , Microscopia Eletrônica , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo , Útero/efeitos dos fármacos , Útero/ultraestrutura
17.
Differentiation ; 51(1): 49-54, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1451961

RESUMO

The Syrian hamster cheek pouch was evaluated as a convenient transplantation site for studies of estrogen-dependent uterine growth and morphogenesis. At one month of age, hosts were either ovariectomized (Ovex) or ovariectomized and estrogen implanted (Ovex+E2), and at the same time the uterus from a 7-day old untreated donor was transplanted into the host's right cheek pouch. Periodic inspection (by simple eversion of the pouch) revealed viable transplants in the majority of hosts for both groups, and clear evidence of estrogen-dependent transplant growth that continued for at least 9 months. At that time, weight of the transplanted uterus was comparable to that of a given host's own in situ uterus, but uteri at both sites weighed six to eightfold more in Ovex+E2 hosts than in Ovex hosts. Histological analysis also revealed similar degrees of endometrial atrophy in Ovex hosts and hypertrophy/hyperplasia in Ovex+E2 hosts for both in situ and transplanted uteri. Furthermore, while only scant and rudimentary endometrial glands developed in both in situ and transplanted uteri within Ovex hosts, uteri at both sites within the Ovex+E2 hosts were riddled with cystic glandular structures and exhibited marked leukocytic infiltration. These data demonstrate that neonatal uteri transplanted to the hamster cheek pouch will grow, differentiate and follow an endocrine-responsive morphogenetic program that is quantitatively and qualitatively consistent with that of the host's in situ uterus. Lastly, we were able to cleanly separate epithelium from the stroma of 5-day old hamster uteri, reassociate the two tissues in vitro, transplant the recombinants into cheek pouches of adult female hamsters and subsequently observe growth and maintenance of a generally normal uterine morphology and differentiated function.


Assuntos
Estradiol/farmacologia , Útero/citologia , Animais , Animais Recém-Nascidos , Bochecha , Cricetinae , Células Epiteliais , Epitélio/efeitos dos fármacos , Feminino , Mesocricetus , Morfogênese/efeitos dos fármacos , Ovariectomia , Transplante Autólogo , Transplante Heterotópico , Útero/efeitos dos fármacos , Útero/transplante
18.
Proc Soc Exp Biol Med ; 199(4): 501-8, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1549630

RESUMO

It has been proposed that the anticonvulsant drug phenytoin (PHT) and glucocorticoids induce orofacial clefting by the same mechanism. Previous work had demonstrated that PHT treatment significantly increased endogenous maternal corticosterone concentrations for approximately 48 hr after dosing in A/J mice. The purpose of the present investigation was to determine whether PHT is embryotoxic in the absence of endogenous maternal glucocorticoids. Maternal adrenal glands were removed on Day 7 of gestation, and the incidence of clefting after PHT treatment was determined. There was a high level of maternal toxicity following adrenalectomy (ADX) and PHT treatment at either 60 or 75 mg/kg. This increased toxicity did not appear to be due to altered maternal drug levels in ADX mice. There was a significant increase in the clefting incidence among offspring of ADX dams treated with PHT at 60 mg/kg. This dose of PHT did not elevate maternal corticosterone levels in ADX dams. These data suggest that PHT is capable of producing clefts in the absence of endogenous maternal corticosterone.


Assuntos
Anormalidades Induzidas por Medicamentos/etiologia , Adrenalectomia , Fissura Palatina/induzido quimicamente , Fenitoína/toxicidade , Animais , Corticosterona/sangue , Feminino , Reabsorção do Feto/induzido quimicamente , Camundongos , Fenitoína/sangue , Gravidez
19.
J Steroid Biochem Mol Biol ; 42(1): 23-8, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1558818

RESUMO

Diethylstilbestrol (DES) treatment of female rats on postnatal days (PND) 1-5 reduces uterine growth, estrogen receptor (ER) level and gland number by PND 25, while daily DES treatment on PND 1-25 increases uterine growth 4-fold, further reduces ER level and completely suppresses gland formation. We now report the persistence of these effects in adults. By PND 60, uterine weight was 70% of controls in rats injected with DES on PND 1-5 but only 10% of controls in rats injected PND 1-10 or longer. In fact, uterine weights were the same on PND 10 and 60. Uterine gland numbers were reduced to 30% of controls in all DES-treated rats regardless of exposure length; however, luminal and glandular epithelial cell heights were reduced to less than 50 and 70%, respectively, of controls when DES was given on PND 1-25 but not when given on PND 1-5. Ovariectomy 7 days prior to sacrifice on PND 60 reduced uterine weight in controls by 67% and in rats injected with DES on PND 1-5 by 53%, but had no effect in rats injected with DES on PND 1-10. DES exposure at either PND 1-5 or 1-10 lowered ER levels by 35-50% at both 60 and 90 days. Treatment with a high dose of estradiol (E2) 1 week before sacrifice significantly down-regulated ER to the same concentration in all treatment groups at PND 60 and 90. Following E2 treatment, all groups also showed increased uterine weight at PND 60 and 90. These data show there is a short period of development (PND 5-10) in which further DES exposure indirectly inhibits uterine growth.


Assuntos
Dietilestilbestrol/farmacologia , Receptores de Estrogênio/metabolismo , Útero/efeitos dos fármacos , Envelhecimento , Animais , Dietilestilbestrol/administração & dosagem , Regulação para Baixo/efeitos dos fármacos , Feminino , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , Ratos , Ratos Endogâmicos , Útero/crescimento & desenvolvimento , Útero/metabolismo
20.
In Vitro Cell Dev Biol ; 27A(6): 442-6, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1869485

RESUMO

We have developed a tissue culture system using an extract of basement membrane (extracellular matrix) which promotes the in vitro growth and development of uterine luminal epithelium from the 5-day-old rat. Uterine luminal epithelium, free of stroma, was obtained as short tubes by trypsinization of uterine segments followed by mechanical separation. Epithelial segments were grown in a serum-free medium on culture dishes coated with an extracellular matrix. After 2 days, rapid cell growth resulted in monolayer cultures, which subsequently formed organoid structures similar to differentiated uterine glands present in uterine tissue taken from older rats. Electron microscopy of cultures revealed columnar cells with basally located nuclei, apical microvilli, lateral membranes with interdigitations, desmosomes, and secretory Golgi complexes, all features found in functioning uterine epithelium in vivo. This model will allow the in vitro investigation of the development of uterine epithelium-specific functions free of the influence of stromal cell factors.


Assuntos
Extratos Celulares/farmacologia , Matriz Extracelular/fisiologia , Útero/citologia , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais , Epitélio/efeitos dos fármacos , Epitélio/ultraestrutura , Matriz Extracelular/química , Feminino , Microscopia Eletrônica , Ratos , Ratos Endogâmicos F344 , Útero/efeitos dos fármacos , Útero/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...