Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Environ Health Perspect ; 128(3): 37001, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32186404

RESUMO

BACKGROUND: Embryo implantation relies on precise hormonal regulation, associated gene expression changes, and appropriate female reproductive tract tissue architecture. Female mice exposed neonatally to the phytoestrogen genistein (GEN) at doses similar to those in infants consuming soy-based infant formulas are infertile due in part to uterine implantation defects. OBJECTIVES: Our goal was to determine the mechanisms by which neonatal GEN exposure causes implantation defects. METHODS: Female mice were exposed to GEN on postnatal days (PND)1-5 and uterine tissues collected on PND5, PND22-26, and during pregnancy. Analysis of tissue weights, morphology, and gene expression was performed using standard histology, confocal imaging with three-dimensional analysis, real-time reverse transcription polymerase chain reaction (real-time RT-PCR), and microarrays. The response of ovariectomized adults to 17ß-estradiol (E2) and artificial decidualization were measured. Leukemia inhibitory factor (LIF) injections were given intraperitoneally and implantation sites visualized. Gene expression patterns were compared with curated data sets to identify upstream regulators. RESULTS: GEN-exposed mice exhibited reduced uterine weight gain in response to E2 treatment or artificial decidualization compared with controls; however, expression of select hormone responsive genes remained similar between the two groups. Uteri from pregnant GEN-exposed mice were posteriorized and had reduced glandular epithelium. Implantation failure was not rescued by LIF administration. Microarray analysis of GEN-exposed uteri during early pregnancy revealed significant overlap with several conditional uterine knockout mouse models, including Foxa2, Wnt4, and Sox17. These models exhibit reduced endometrial glands, features of posteriorization and implantation failure. Expression of Foxa2, Wnt4, and Sox17, as well as genes important for neonatal uterine differentiation (Wnt7a, Hoxa10, and Msx2), were severely disrupted on PND5 in GEN-exposed mice. DISCUSSION: Our findings suggest that neonatal GEN exposure in mice disrupts expression of genes important for uterine development, causing posteriorization and diminished gland function during pregnancy that contribute to implantation failure. These findings could have implications for women who consumed soy-based formulas as infants. https://doi.org/10.1289/EHP6336.


Assuntos
Implantação do Embrião/efeitos dos fármacos , Genisteína/efeitos adversos , Fitoestrógenos/efeitos adversos , Útero/efeitos dos fármacos , Animais , Feminino , Camundongos , Gravidez , Útero/crescimento & desenvolvimento , Útero/fisiopatologia
2.
EMBO J ; 36(21): 3175-3193, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29021282

RESUMO

Methionine metabolism is critical for epigenetic maintenance, redox homeostasis, and animal development. However, the regulation of methionine metabolism remains unclear. Here, we provide evidence that SIRT1, the most conserved mammalian NAD+-dependent protein deacetylase, is critically involved in modulating methionine metabolism, thereby impacting maintenance of mouse embryonic stem cells (mESCs) and subsequent embryogenesis. We demonstrate that SIRT1-deficient mESCs are hypersensitive to methionine restriction/depletion-induced differentiation and apoptosis, primarily due to a reduced conversion of methionine to S-adenosylmethionine. This reduction markedly decreases methylation levels of histones, resulting in dramatic alterations in gene expression profiles. Mechanistically, we discover that the enzyme converting methionine to S-adenosylmethionine in mESCs, methionine adenosyltransferase 2a (MAT2a), is under control of Myc and SIRT1. Consistently, SIRT1 KO embryos display reduced Mat2a expression and histone methylation and are sensitive to maternal methionine restriction-induced lethality, whereas maternal methionine supplementation increases the survival of SIRT1 KO newborn mice. Our findings uncover a novel regulatory mechanism for methionine metabolism and highlight the importance of methionine metabolism in SIRT1-mediated mESC maintenance and embryonic development.


Assuntos
Desenvolvimento Embrionário/genética , Epigênese Genética , Metionina Adenosiltransferase/genética , Metionina/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Sirtuína 1/genética , Acetilação , Animais , Apoptose , Diferenciação Celular , Embrião de Mamíferos , Histonas/genética , Histonas/metabolismo , Metabolômica , Metionina/administração & dosagem , Metionina Adenosiltransferase/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Células-Tronco Embrionárias Murinas/citologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , S-Adenosilmetionina/metabolismo , Sirtuína 1/deficiência
3.
J Am Assoc Lab Anim Sci ; 52(2): 130-41, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23562095

RESUMO

The lowest observed adverse effect level for bisphenol A (BPA) in mice and rats is currently poorly defined due to inconsistent study designs and results in published studies. The objectives of the current study were to (1) compare the estrogenic content of rodent diets, bedding, cages, and water bottles to evaluate their impact on the estrogenic activity of BPA and (2) review the literature on BPA to determine the most frequently reported diets, beddings, cages, and water bottles used in animal studies. Our literature review indicated that low-dose BPA animal studies have inconsistent results and that factors contributing to this inconsistency are the uses of high-phytoestrogen diets and the different routes of exposure. In 44% (76 of 172) of all reports, rodents were exposed to BPA via the subcutaneous route. Our literature review further indicated that the type of diet, bedding, caging, and water bottles used in BPA studies were not always reported. Only 37% (64 of 172) of the reports described the diet used. In light of these findings, we recommend the use of a diet containing low levels of phytoestrogen (less than 20 µg/g diet) and metabolizable energy (approximately 3.1 kcal/g diet) and estrogen-free bedding, cages, and water bottles for studies evaluating the estrogenic activity of endocrine-disrupting compounds such as BPA. The oral route of BPA exposure should be used when results are to be extrapolated to humans.


Assuntos
Compostos Benzidrílicos/efeitos adversos , Compostos Benzidrílicos/análise , Disruptores Endócrinos/efeitos adversos , Fenóis/efeitos adversos , Fenóis/análise , Ração Animal/análise , Animais , Disruptores Endócrinos/análise , Metabolismo Energético , Feminino , Abrigo para Animais , Masculino , Camundongos , Fitoestrógenos/efeitos adversos , Fitoestrógenos/análise , Ratos
4.
Biol Reprod ; 87(1): 10, 1-10, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22553218

RESUMO

Treatment of neonatal mice with the phytoestrogen genistein (50 mg/kg/day) results in complete female infertility caused in part by preimplantation embryo loss in the oviduct between Days 2 and 3 of pregnancy. We previously demonstrated that oviducts of genistein-treated mice are "posteriorized" as compared to control mouse oviducts because they express numerous genes normally restricted to posterior regions of the female reproductive tract (FRT), the cervix and vagina. We report here that neonatal genistein treatment resulted in substantial changes in oviduct expression of genes important for the FRT mucosal immune response, including immunoglobulins, antimicrobials, and chemokines. Some of the altered immune response genes were chronically altered beginning at the time of neonatal genistein treatment, indicating that these alterations were a result of the posteriorization phenotype. Other alterations in oviduct gene expression were observed only in early pregnancy, immediately after the FRT was exposed to inflammatory or antigenic stimuli from ovulation and mating. The oviduct changes affected development of the surviving embryos by increasing the rate of cleavage and decreasing the trophectoderm-to-inner cell mass cell ratio at the blastocyst stage. We conclude that both altered immune responses to pregnancy and deficits in oviduct support for preimplantation embryo development in the neonatal genistein model are likely to contribute to infertility phenotype.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Genisteína/toxicidade , Imunidade nas Mucosas/efeitos dos fármacos , Oviductos/efeitos dos fármacos , Oviductos/imunologia , Fitoestrógenos/toxicidade , Animais , Animais Recém-Nascidos , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/imunologia , Desenvolvimento Embrionário/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes MHC da Classe II/efeitos dos fármacos , Genisteína/administração & dosagem , Imunidade nas Mucosas/genética , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/genética , Infertilidade Feminina/imunologia , Infertilidade Feminina/metabolismo , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Oviductos/metabolismo , Oviductos/patologia , Fitoestrógenos/administração & dosagem , Gravidez
6.
Environ Health Perspect ; 119(11): 1575-82, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21810550

RESUMO

BACKGROUND: Preimplantation embryo loss during oviduct transit has been observed in adult mice after a 5-day neonatal exposure to the phytoestrogen genistein (Gen; 50 mg/kg/day). OBJECTIVE: We investigated the mechanisms underlying the contribution of the oviduct to infertility. METHODS: Female mice were treated on postnatal days 1-5 with corn oil or Gen (50 mg/kg/day). We compared morphology, gene expression, and protein expression in different regions of the reproductive tracts of Gen-treated mice with those of control littermates at several time points. RESULTS: Neonatal Gen treatment resulted in substantial changes in expression of genes that modulate neonatal oviduct morphogenesis, including Hoxa (homeobox A cluster), Wnt (wingless-related MMTV integration site), and hedgehog signaling genes. An estrogen receptor antagonist blocked these effects, indicating that they were induced by the estrogenic activity of Gen. Oviducts of adults treated neonatally with Gen had abnormal morphology and were stably "posteriorized," as indicated by altered Hoxa gene patterning during the time of treatment and dramatic, permanent up-regulation of homeobox genes (e.g., Pitx1, Six1) normally expressed only in the cervix and vagina. CONCLUSIONS: Neonatal exposure to estrogenic environmental chemicals permanently disrupts oviduct morphogenesis and adult gene expression patterns, and these changes likely contribute to the infertility phenotype.


Assuntos
Genisteína/toxicidade , Infertilidade/induzido quimicamente , Oviductos/efeitos dos fármacos , Fitoestrógenos/toxicidade , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Genisteína/farmacologia , Immunoblotting , Infertilidade/genética , Camundongos , Análise em Microsséries , Morfogênese , Oviductos/crescimento & desenvolvimento , Oviductos/patologia , Fitoestrógenos/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo
7.
Biol Reprod ; 80(3): 425-31, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19005167

RESUMO

Female mice treated neonatally with the phytoestrogen genistein (50 mg/kg/day) have multioocyte follicles, lack regular estrous cyclicity, and are infertile even after superovulation. To determine the cause of their infertility, we examined oocyte developmental competence and timing of embryo loss. Eggs obtained by superovulation of genistein-treated or control females were equally capable of being fertilized in vitro and cultured to the blastocyst stage. However, if eggs were fertilized in vivo, retrieved at the pronucleus stage, and cultured, there was a significant reduction in the percentage of embryos from genistein-treated females reaching the blastocyst stage. When these blastocysts were transferred to pseudopregnant recipients, the number of live pups produced was similar to that in controls. Preimplantation embryo development in vivo was examined by flushing embryos from the oviduct and/or uterus. Similar numbers of one-cell and two-cell embryos were obtained from genistein-treated and control females. However, significantly fewer embryos (<50%) were obtained from genistein-treated females on postcoital Days 3 and 4. To determine if neonatal genistein treatment altered the ability of the uterus to support implantation, blastocysts from control donors were transferred to control and genistein-treated pseudopregnant recipients. These experiments demonstrated that genistein-treated females are not capable of supporting normal implantation of control embryos. Taken together, these results suggest that oocytes from mice treated neonatally with genistein are developmentally competent; however, the oviductal environment and the uterus have abnormalities that contribute to the observed reproductive failure.


Assuntos
Implantação do Embrião/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Genisteína/toxicidade , Infertilidade Feminina/induzido quimicamente , Fitoestrógenos/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Células Cultivadas , Modelos Animais de Doenças , Implantação do Embrião/fisiologia , Transferência Embrionária , Desenvolvimento Embrionário/fisiologia , Feminino , Fertilização/efeitos dos fármacos , Fertilização/fisiologia , Genisteína/farmacologia , Infertilidade Feminina/fisiopatologia , Camundongos , Camundongos Endogâmicos ICR , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Ovário/efeitos dos fármacos , Ovário/fisiopatologia , Oviductos/efeitos dos fármacos , Oviductos/fisiopatologia , Fitoestrógenos/farmacologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Útero/efeitos dos fármacos , Útero/fisiopatologia
8.
Environ Health Perspect ; 115(12): 1717-26, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18087589

RESUMO

BACKGROUND: The optimum test diet and rodent species/strain for evaluating endocrine-disrupting compounds (EDCs) are critical. OBJECTIVES: We conducted studies to evaluate rodent species sensitivity and the effects of diets varying in phytoestrogen content on the time of vaginal opening (VO) in CD-1 mice, Fischer 344 (F344) rats, and CD Sprague-Dawley (S-D) rats. METHODS: Mice were weaned on postnatal day (PND) 15 and rats on PND19 and randomly assigned to control or test diets. Body weights, food consumption, and time of VO were recorded. RESULTS: The time of VO was significantly advanced in F344 rats fed diets containing daidzein and genistein, whereas these same diets did not advance VO in S-D rats. When animals were fed the AIN-76A diet spiked with genistein, time of VO was significantly advanced at all doses in CD-1 mice, at the two highest doses in F344 rats, and at the highest dose in S-D rats. The time of VO in F344 rats was more highly correlated with the phytoestrogen content than with the total metabolizable energy (ME) of 12 diets. CONCLUSIONS: The S-D rat is less sensitive to dietary phytoestrogens compared with the F344 rat or the CD-1 mouse, suggesting that the S-D rat is not the ideal model for evaluating estrogenic activity of EDCs. The profound effects of dietary phytoestrogens on the time of VO, an estrogen-sensitive marker, indicate that a standardized open-formula phytoestrogen-free diet containing a low ME level should be used to optimize the sensitivity of estrogenic bioassays.


Assuntos
Dieta , Fitoestrógenos/análise , Fitoestrógenos/farmacologia , Vagina/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Feminino , Genisteína/análise , Genisteína/farmacologia , Isoflavonas/análise , Isoflavonas/farmacologia , Camundongos , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley
9.
Mol Nutr Food Res ; 51(7): 832-44, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17604387

RESUMO

Studies in our laboratory have shown that exposure to genistein causes deleterious effects on the developing female reproductive system. Mice treated neonatally on days 1-5 by subcutaneous injection of genistein (0.5-50 mg/kg) exhibited altered ovarian differentiation leading to multioocyte follicles (MOFs) at 2 months of age. Ovarian function and estrous cyclicity were also disrupted by neonatal exposure to genistein with increasing severity observed over time. Reduced fertility was observed in mice treated with genistein (0.5, 5, or 25 mg/kg) and infertility was observed at 50 mg/kg. Mammary gland and behavioral endpoints were also affected by neonatal genistein treatment. Further, transgenerational effects were observed; female offspring obtained from breeding genistein treated females (25 mg/kg) to control males had increased MOFs. Thus, neonatal treatment with genistein at environmentally relevant doses caused adverse consequences on female development which is manifested in adulthood. Whether adverse effects occur in human infants exposed to soy-based products such as soy infant formulas is unknown but the neonatal murine model may help address some of the current uncertainties since we have shown that many effects obtained from feeding genistin, the glycosolated form of genistein found in soy formula, are similar to those obtained from injecting genistein.


Assuntos
Genisteína/farmacologia , Fitoestrógenos/farmacologia , Reprodução/efeitos dos fármacos , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Dieta , Ciclo Estral/efeitos dos fármacos , Feminino , Genisteína/administração & dosagem , Genisteína/efeitos adversos , Humanos , Recém-Nascido , Masculino , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Comportamento Materno/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/crescimento & desenvolvimento , Ovário/fisiologia , Fitoestrógenos/administração & dosagem , Fitoestrógenos/efeitos adversos , Reprodução/fisiologia , Neoplasias Uterinas/induzido quimicamente
10.
Reprod Toxicol ; 23(3): 308-16, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17250991

RESUMO

Studies in our laboratory have shown that developmental exposure to genistein causes deleterious effects on the reproductive system. Oral exposure to genistin (25mg/kg) increases uterine weight at 5 days of age similar to subcutaneous injection of genistein (20mg/kg) suggesting that subcutaneous injection of genistein is a suitable model for oral exposure to genistin. Mice treated neonatally by subcutaneous injection of genistein (0.5-50mg/kg) exhibit altered ovarian differentiation leading to multi-oocyte follicles (MOFs). Ovarian function and estrous cyclicity were disrupted in genistein treated mice with increasing severity over time. Reduced fertility was observed in mice treated with genistein (0.5, 5, or 25mg/kg) and infertility was observed at 50mg/kg. Females generated from genistein 25mg/kg females bred to control males have increased MOFs suggesting these effects can be transmitted to subsequent generations. Thus, neonatal treatment with genistein at environmentally relevant doses caused adverse consequences on reproduction in adulthood.


Assuntos
Genisteína/administração & dosagem , Genitália Feminina/efeitos dos fármacos , Fitoestrógenos/administração & dosagem , Reprodução/efeitos dos fármacos , Animais , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/química , Feminino , Genisteína/química , Genitália Feminina/fisiopatologia , Humanos , Estrutura Molecular , Fitoestrógenos/química , Estilbenos/administração & dosagem
11.
J AOAC Int ; 89(4): 1189-96, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16918037

RESUMO

Studies have shown that developmental exposure to genistein alters murine reproductive differentiation, resulting in abnormal ovarian development (multioocyte follicles) and uterine neoplasia later in life. Further, reproductive function was altered. Prolonged estrous cyclicity was observed following neonatal genistein treatment (0.5-50 mg/kg) on Days 1-5 with dose- and age-related increase in severity. Fertility, determined at 2, 4, and 6 months, showed decreased numbers of genistein-treated females (0.5 or 5 mg/kg) delivering live pups and reduced numbers of pups. At 6 months, 60% of 0.5 mg/kg and 40% of 5 mg/kg groups delivered live pups compared to 100% of controls. At 2 months, half the mice treated with 25 mg/kg of genistein and none treated with 50 mg/kg delivered live pups, although half of the latter group showed signs of pregnancy with few small implantation sites. Ovarian function was disrupted in the low genistein-dosed mice with increased numbers of corpora lutea (CLs) compared to controls and increased ovulated oocytes following exogenous gonadotropins treatment. In contrast, mice treated with high genistein doses had decreased numbers of CLs; ovulation could be restored with exogenous gonadotropins. Thus, neonatal treatment with genistein at environmentally relevant doses caused adverse consequences on ovarian development and reproductive function.


Assuntos
Animais Recém-Nascidos , Genisteína/efeitos adversos , Reprodução/efeitos dos fármacos , Animais , Corpo Lúteo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ciclo Estral/efeitos dos fármacos , Feminino , Genisteína/farmacologia , Gonadotropinas/metabolismo , Camundongos , Ovário/efeitos dos fármacos , Fitoestrógenos/metabolismo , Fatores de Tempo
12.
Endocrinology ; 147(10): 4871-82, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16857750

RESUMO

Developmental effects of genistein (Gen) on the mammary gland were investigated using outbred female CD-1 mice treated neonatally on d 1-5 by sc injections at doses of 0.5, 5, or 50 mg/kg.d. Examination of mammary gland whole mounts (no. 4) before puberty (4 wk) revealed no morphological differences in development after Gen treatment. However, mice treated with Gen-50 had stunted development characterized by less branching at 5 wk and decreased numbers of terminal end buds at 5 and 6 wk. Conversely, at 6 wk, Gen-0.5-treated mice exhibited advanced development with increased ductal elongation compared with controls. Measurements of hormone receptor levels showed increased levels of progesterone receptor protein and estrogen receptor-beta mRNA in Gen-0.5-treated mice compared with controls; ERalpha expression was decreased after all doses of Gen treatment. Lactation ability, measured by pup weight gain and survival, was not affected after neonatal Gen-0.5 and Gen-5. Mice treated with Gen-50 did not deliver live pups; therefore, lactation ability could not be determined. Evaluation of mammary glands in aged mice (9 months) showed no differences between Gen-0.5-treated mice and controls but mice treated with Gen-5 and Gen-50 exhibited altered morphology including reduced lobular alveolar development, dilated ducts, and focal areas of "beaded" ducts lined with hyperplastic ductal epithelium. In summary, neonatal Gen exposure altered mammary gland growth and development as well as hormone receptor levels at all doses examined; higher doses of Gen led to permanent long-lasting morphological changes.


Assuntos
Animais Recém-Nascidos/fisiologia , Genisteína/farmacologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Fitoestrógenos/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Hormônios/sangue , Imuno-Histoquímica , Lactação/efeitos dos fármacos , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Gravidez , Receptores da Prolactina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sobrevida
13.
Biol Reprod ; 73(4): 798-806, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15930323

RESUMO

Outbred female CD-1 mice were treated with genistein (Gen), the primary phytoestrogen in soy, by s.c. injections on Neonatal Days 1-5 at doses of 0.5, 5, or 50 mg/kg per day (Gen-0.5, Gen-5, and Gen-50). The day of vaginal opening was observed in mice treated with Gen and compared with controls, and although there were some differences, they were not statistically significant. Gen-treated mice had prolonged estrous cycles with a dose- and age-related increase in severity of abnormal cycles. Females treated with Gen-0.5 or Gen-5 bred to control males at 2, 4, and 6 mo showed statistically significant decreases in the number of live pups over time with increasing dose; at 6 mo, 60% of the females in the Gen-0.5 group and 40% in the Gen-5 group delivered live pups compared with 100% of controls. Mice treated with Gen-50 did not deliver live pups. At 2 mo, >60% of the mice treated with Gen-50 were fertile as determined by uterine implantation sites, but pregnancy was not maintained; pregnancy loss was characterized by fewer, smaller implantation sites and increased reabsorptions. Mice treated with lower doses of Gen had increased numbers of corpora lutea compared with controls, while mice treated with the highest dose had decreased numbers; however, superovulation with eCG/hCG yielded similar numbers of oocytes as controls. Serum levels of progesterone, estradiol, and testosterone were similar between Gen-treated and control mice when measured before puberty and during pregnancy. In summary, neonatal treatment with Gen caused abnormal estrous cycles, altered ovarian function, early reproductive senescence, and subfertility/infertility at environmentally relevant doses.


Assuntos
Ciclo Estral/efeitos dos fármacos , Genisteína/efeitos adversos , Ovário/crescimento & desenvolvimento , Reprodução/efeitos dos fármacos , Aborto Espontâneo , Animais , Animais Recém-Nascidos , Animais não Endogâmicos , Relação Dose-Resposta a Droga , Implantação do Embrião , Meio Ambiente , Feminino , Tamanho da Ninhada de Vivíparos , Masculino , Camundongos , Ovário/efeitos dos fármacos , Ovário/fisiologia , Fitoestrógenos/efeitos adversos , Gravidez , Resultado da Gravidez
14.
J Chromatogr B Analyt Technol Biomed Life Sci ; 777(1-2): 179-89, 2002 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-12270211

RESUMO

The estrogenic responses of several phytoestrogens including genistein, daidzein, coumestrol, alpha-zearalanol, zearalenone, naringenin, taxifolin and biochanin A were compared over a wide dose range using an in vitro assay that measures transcriptional activation of the estrogen receptor (ER) and an in vivo immature mouse uterotrophic assay consisting of measuring uterine wet weight increase plus sensitive morphological and biochemical endpoints in the uterus. The transcriptional activation assay showed activation of the ER by all compounds tested except taxifolin with varying magnitudes of response as compared to estradiol or diethylstilbestrol. Results from the uterotropic bioassay showed that genistein, coumestrol, zearalanol, and zearalenone caused an increase in uterine wet weight, while naringenin, taxifolin, daidzein and biochanin A failed to do so over the dose range tested. However, sensitive morphological and biochemical parameters such as uterine epithelial cell height increase, uterine gland number increase, and induction of the estrogen-responsive protein lactoferrin demonstrated that all compounds tested in this study gave some measure of estrogenicity although a wide range of estrogenic responses across compounds was shown. Use of multiple in vitro and in vivo estrogenic endpoints as described in this paper will be useful in developing estrogenic profiles for individual compounds and ultimately mixtures of compounds. Furthermore, having an estrogenic "fingerprint" for each phytochemical is an essential first step in determining potential adverse effects of exposure to phytoestrogens.


Assuntos
Estrogênios não Esteroides/farmacologia , Isoflavonas , Ativação Transcricional/efeitos dos fármacos , Útero/efeitos dos fármacos , Animais , Feminino , Imuno-Histoquímica , Lactoferrina/metabolismo , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Fitoestrógenos , Preparações de Plantas , Receptores de Estrogênio/genética , Útero/citologia , Útero/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA