Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 21(10): e3002334, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37856394

RESUMEN

Tissue development entails genetically programmed differentiation of immature cell types to mature, fully differentiated cells. Exposure during development to non-mutagenic environmental factors can contribute to cancer risk, but the underlying mechanisms are not understood. We used a mouse model of endometrial adenocarcinoma that results from brief developmental exposure to an estrogenic chemical, diethylstilbestrol (DES), to determine causative factors. Single-cell RNA sequencing (scRNAseq) and spatial transcriptomics of adult control uteri revealed novel markers of uterine epithelial stem cells (EpSCs), identified distinct luminal and glandular progenitor cell (PC) populations, and defined glandular and luminal epithelium (LE) cell differentiation trajectories. Neonatal DES exposure disrupted uterine epithelial cell differentiation, resulting in a failure to generate an EpSC population or distinguishable glandular and luminal progenitors or mature cells. Instead, the DES-exposed epithelial cells were characterized by a single proliferating PC population and widespread activation of Wnt/ß-catenin signaling. The underlying endometrial stromal cells had dramatic increases in inflammatory signaling pathways and oxidative stress. Together, these changes activated phosphoinositide 3-kinase/AKT serine-threonine kinase signaling and malignant transformation of cells that were marked by phospho-AKT and the cancer-associated protein olfactomedin 4. Here, we defined a mechanistic pathway from developmental exposure to an endocrine disrupting chemical to the development of adult-onset cancer. These findings provide an explanation for how human cancers, which are often associated with abnormal activation of PI3K/AKT signaling, could result from exposure to environmental insults during development.


Asunto(s)
Adenocarcinoma , Fosfatidilinositol 3-Quinasas , Animales , Femenino , Ratones , Adenocarcinoma/inducido químicamente , beta Catenina/genética , beta Catenina/metabolismo , Diferenciación Celular , Estrógenos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Útero
2.
Proc Natl Acad Sci U S A ; 115(18): E4189-E4198, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29666266

RESUMEN

Early transient developmental exposure to an endocrine active compound, diethylstilbestrol (DES), a synthetic estrogen, causes late-stage effects in the reproductive tract of adult mice. Estrogen receptor alpha (ERα) plays a role in mediating these developmental effects. However, the developmental mechanism is not well known in male tissues. Here, we present genome-wide transcriptome and DNA methylation profiling of the seminal vesicles (SVs) during normal development and after DES exposure. ERα mediates aberrations of the mRNA transcriptome in SVs of adult mice following neonatal DES exposure. This developmental exposure impacts differential diseases between male (SVs) and female (uterus) tissues when mice reach adulthood due to most DES-altered genes that appear to be tissue specific during mouse development. Certain estrogen-responsive gene changes in SVs are cell-type specific. DNA methylation dynamically changes during development in the SVs of wild-type (WT) and ERα-knockout (αERKO) mice, which increases both the loss and gain of differentially methylated regions (DMRs). There are more gains of DMRs in αERKO compared with WT. Interestingly, the methylation changes between the two genotypes are in different genomic loci. Additionally, the expression levels of a subset of DES-altered genes are associated with their DNA methylation status following developmental DES exposure. Taken together, these findings provide an important basis for understanding the molecular and cellular mechanism of endocrine-disrupting chemicals (EDCs), such as DES, during development in the male mouse tissues. This unique evidence contributes to our understanding of developmental actions of EDCs in human health.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Dietilestilbestrol/efectos adversos , Receptor alfa de Estrógeno/metabolismo , Estrógenos no Esteroides/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Vesículas Seminales/metabolismo , Transcriptoma/efectos de los fármacos , Animales , Metilación de ADN/genética , Dietilestilbestrol/farmacología , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/genética , Estrógenos no Esteroides/farmacología , Sitios Genéticos , Masculino , Ratones , Ratones Noqueados
3.
Nucleic Acids Res ; 46(11): 5487-5503, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29648668

RESUMEN

Little is known regarding how steroid hormone exposures impact the epigenetic landscape in a living organism. Here, we took a global approach to understanding how exposure to the estrogenic chemical, diethylstilbestrol (DES), affects the neonatal mouse uterine epigenome. Integration of RNA- and ChIP-sequencing data demonstrated that ∼80% of DES-altered genes had higher H3K4me1/H3K27ac signal in close proximity. Active enhancers, of which ∼3% were super-enhancers, had a high density of estrogen receptor alpha (ERα) binding sites and were correlated with alterations in nearby gene expression. Conditional uterine deletion of ERα, but not the pioneer transcription factors FOXA2 or FOXO1, prevented the majority of DES-mediated changes in gene expression and H3K27ac signal at target enhancers. An ERα dependent super-enhancer was located at the Padi gene locus and a topological connection to the Padi1 TSS was documented using 3C-PCR. Chromosome looping at this site was independent of ERα and DES exposure, indicating that the interaction is established prior to ligand signaling. However, enrichment of H3K27ac and transcriptional activation at this locus was both DES and ERα-dependent. These data suggest that DES alters uterine development and consequently adult reproductive function by modifying the enhancer landscape at ERα binding sites near estrogen-regulated genes.


Asunto(s)
Dietilestilbestrol/farmacología , Receptor alfa de Estrógeno/metabolismo , Estrógenos no Esteroides/farmacología , Regulación de la Expresión Génica/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Útero/embriología , Animales , Sitios de Unión/genética , Receptor alfa de Estrógeno/genética , Estrógenos no Esteroides/metabolismo , Femenino , Proteína Forkhead Box O1/genética , Factor Nuclear 3-beta del Hepatocito/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Metilación/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Regiones Promotoras Genéticas/genética
4.
Biol Reprod ; 101(2): 306-317, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31201420

RESUMEN

Enhancer of zeste homolog 2 (EZH2) is a rate-limiting catalytic subunit of a histone methyltransferase, polycomb repressive complex, which silences gene activity through the repressive histone mark H3K27me3. EZH2 is critical for epigenetic effects of early estrogen treatment, and may be involved in uterine development and pathologies. We investigated EZH2 expression, regulation, and its role in uterine development/function. Uterine epithelial EZH2 expression was associated with proliferation and was high neonatally then declined by weaning. Pre-weaning uterine EZH2 expression was comparable in wild-type and estrogen receptor 1 knockout mice, showing neonatal EZH2 expression is ESR1 independent. Epithelial EZH2 was upregulated by 17ß-estradiol (E2) and inhibited by progesterone in adult uteri from ovariectomized mice. To investigate the uterine role of EZH2, we developed a EZH2 conditional knockout (Ezh2cKO) mouse using a cre recombinase driven by the progesterone receptor (Pgr) promoter that produced Ezh2cKO mice lacking EZH2 in Pgr-expressing tissues (e.g. uterus, mammary glands). In Ezh2cKO uteri, EZH2 was deleted neonatally. These uteri had reduced H3K27me3, were larger than WT, and showed adult cystic endometrial hyperplasia. Ovary-independent uterine epithelial proliferation and increased numbers of highly proliferative uterine glands were seen in adult Ezh2cKO mice. Female Ezh2cKO mice were initially subfertile, and then became infertile by 9 months. Mammary gland development in Ezh2cKO mice was inhibited. In summary, uterine EZH2 expression is developmentally and hormonally regulated, and its loss causes aberrant uterine epithelial proliferation, uterine hypertrophy, and cystic endometrial hyperplasia, indicating a critical role in uterine development and function.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Útero/enzimología , Útero/crecimiento & desarrollo , Animales , Proteína Potenciadora del Homólogo Zeste 2/genética , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Femenino , Histonas/metabolismo , Glándulas Mamarias Animales/enzimología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Embarazo , Progesterona/metabolismo
5.
Toxicol Pathol ; 47(8): 1049-1071, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31833458

RESUMEN

During the past 20 years, investigations involving endocrine active substances (EAS) and reproductive toxicity have dominated the landscape of ecotoxicological research. This has occurred in concert with heightened awareness in the scientific community, general public, and governmental entities of the potential consequences of chemical perturbation in humans and wildlife. The exponential growth of experimentation in this field is fueled by our expanding knowledge into the complex nature of endocrine systems and the intricacy of their interactions with xenobiotic agents. Complicating factors include the ever-increasing number of novel receptors and alternate mechanistic pathways that have come to light, effects of chemical mixtures in the environment versus those of single EAS laboratory exposures, the challenge of differentiating endocrine disruption from direct cytotoxicity, and the potential for transgenerational effects. Although initially concerned with EAS effects chiefly in the thyroid glands and reproductive organs, it is now recognized that anthropomorphic substances may also adversely affect the nervous and immune systems via hormonal mechanisms and play substantial roles in metabolic diseases, such as type 2 diabetes and obesity.


Asunto(s)
Disruptores Endocrinos/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/patología , Reproducción/efectos de los fármacos , Animales , Congresos como Asunto , Femenino , Desarrollo Fetal/efectos de los fármacos , Corazón/efectos de los fármacos , Corazón/embriología , Humanos , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Especificidad de la Especie , Testículo/efectos de los fármacos , Testículo/embriología , Testículo/patología , Útero/efectos de los fármacos , Útero/embriología , Útero/patología
6.
Biol Reprod ; 98(4): 449-464, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29325037

RESUMEN

Understanding factors that regulate zygotic genome activation (ZGA) is critical for determining how cells are reprogrammed to become totipotent or pluripotent. There is limited information regarding how this process occurs physiologically in early mammalian embryos. Here, we identify a mediator complex subunit, MED13, as translated during mouse oocyte maturation and transcribed early from the zygotic genome. Knockdown and conditional knockout approaches demonstrate that MED13 is essential for ZGA in the mouse, in part by regulating expression of the embryo-specific chromatin remodeling complex, esBAF. The role of MED13 in ZGA is mediated in part by interactions with E2F transcription factors. In addition to MED13, its paralog, MED13L, is required for successful preimplantation embryo development. MED13L partially compensates for loss of MED13 function in preimplantation knockout embryos, but postimplantation development is not rescued by MED13L. Our data demonstrate an essential role for MED13 in supporting chromatin reprogramming and directed transcription of essential genes during ZGA.


Asunto(s)
Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica , Complejo Mediador/metabolismo , Oocitos/metabolismo , Animales , Cromatina/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Genoma , Complejo Mediador/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cigoto/metabolismo
7.
Toxicol Pathol ; 46(5): 574-596, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29895210

RESUMEN

Developmental exposure to estrogenic chemicals is an established risk factor for cancer of the female reproductive tract. This increase in risk has been associated with disruption of normal patterns of cellular differentiation during critical stages of morphogenesis. The goal of this study was to document uterine epithelial phenotypes over time following neonatal treatment with the synthetic estrogen diethylstilbestrol (DES) or the soy phytoestrogen genistein (GEN) in female CD-1 mice. Both DES and GEN induced three distinct populations of abnormal endometrial epithelial cells: luminal (SIX1+/P63-/CK14-/CK18+), basal (SIX1+/P63+/CK14+/CK18-), and mixed/bipotential (SIX1+/P63-/CK14+/CK18+), which were all established by early adulthood. In older animals, DES and GEN resulted in uterine carcinomas with mixed glandular, basal, and squamous cell elements. All carcinomas were composed largely of the three abnormal cell types. These findings identify novel epithelial differentiation patterns in the uterus and support the idea that disruption of cellular programming in early development can influence cancer risk later in life.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Neoplasias Endometriales/inducido químicamente , Endometrio/efectos de los fármacos , Estrógenos/toxicidad , Morfogénesis/efectos de los fármacos , Lesiones Precancerosas/inducido químicamente , Animales , Animales Recién Nacidos , Dietilestilbestrol/toxicidad , Neoplasias Endometriales/patología , Endometrio/crecimiento & desarrollo , Endometrio/patología , Femenino , Genisteína/toxicidad , Inmunohistoquímica , Ratones Endogámicos , Lesiones Precancerosas/patología
8.
J Pathol ; 242(2): 246-259, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28295343

RESUMEN

Epithelial membrane protein-2 (EMP2) is a tetraspan protein predicted to regulate placental development. Highly expressed in secretory endometrium and trophectoderm cells, previous studies suggest that it may regulate implantation by orchestrating the surface expression of integrins and other membrane proteins. In order to test the role of EMP2 in pregnancy, mice lacking EMP2 (Emp2-/- ) were generated. Emp2-/- females are fertile but have reduced litter sizes when carrying Emp2-/- but not Emp2+/- fetuses. Placentas of Emp2-/- fetuses exhibit dysregulation in pathways related to neoangiogenesis, coagulation, and oxidative stress, and have increased fibrin deposition and altered vasculature. Given that these findings often occur due to placental insufficiency resulting in an oxygen-poor environment, the expression of hypoxia-inducible factor-1 alpha (HIF-1α) was examined. Placentas from Emp2-/- fetuses had increased total HIF-1α expression in large part through an increase in uterine NK (uNK) cells, demonstrating a unique interplay between uNK cells and trophoblasts modulated through EMP2. To determine if these results translated to human pregnancy, placentas from normal, term deliveries or those complicated by placental insufficiency resulting in intrauterine growth restriction (IUGR) were stained for EMP2. EMP2 was significantly reduced in both villous and extravillous trophoblast populations in IUGR placentas. Experiments in vitro using human trophoblast cells lines indicate that EMP2 modulates angiogenesis by altering HIF-1α expression. Our results reveal a novel role for EMP2 in regulating trophoblast function and vascular development in mice and humans, and suggest that it may be a new biomarker for placental insufficiency. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Glicoproteínas de Membrana/genética , Oxígeno/metabolismo , Insuficiencia Placentaria/genética , Animales , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Fibrina/genética , Fibrina/metabolismo , Técnicas de Inactivación de Genes , Recombinación Homóloga , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica , Placenta/irrigación sanguínea , Placenta/metabolismo , Placenta/patología , Insuficiencia Placentaria/metabolismo , Insuficiencia Placentaria/patología , Placentación , Embarazo , Trofoblastos/metabolismo , Trofoblastos/patología , Útero/irrigación sanguínea , Útero/metabolismo , Útero/patología
9.
Proc Natl Acad Sci U S A ; 109(11): 4169-74, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22371584

RESUMEN

Mammalian fertilization is accompanied by oscillations in egg cytoplasmic calcium (Ca(2+)) concentrations that are critical for completion of egg activation. These oscillations are initiated by Ca(2+) release from inositol 1,4,5-trisphosphate (IP(3))-sensitive intracellular stores. We tested the hypothesis that Ca(2+) influx across the plasma membrane was a requisite component of egg activation signaling, and not simply a Ca(2+) source for store repletion. Using intracytoplasmic sperm injection (ICSI) and standard in vitro fertilization (IVF), we found that Ca(2+) influx was not required to initiate resumption of meiosis II. However, even if multiple oscillations in intracellular Ca(2+) occurred, in the absence of Ca(2+) influx, the fertilized eggs failed to emit the second polar body, resulting in formation of three pronuclei. Additional experiments using the Ca(2+) chelator, BAPTA/AM, demonstrated that Ca(2+) influx is sufficient to support polar body emission and pronucleus formation after only a single sperm-induced Ca(2+) transient, whereas BAPTA/AM-treated ICSI or fertilized eggs cultured in Ca(2+)-free medium remained arrested in metaphase II. Inhibition of store-operated Ca(2+) entry had no effect on ICSI-induced egg activation, so Ca(2+) influx through alternative channels must participate in egg activation signaling. Ca(2+) influx appears to be upstream of CaMKIIγ activity because eggs can be parthenogenetically activated with a constitutively active form of CaMKIIγ in the absence of extracellular Ca(2+). These results suggest that Ca(2+) influx at fertilization not only maintains Ca(2+) oscillations by replenishing Ca(2+) stores, but also activates critical signaling pathways upstream of CaMKIIγ that are required for second polar body emission.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Óvulo/citología , Óvulo/metabolismo , Animales , Tampones (Química) , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ciclo Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cromatina/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Fertilización In Vitro , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Ratones , Modelos Biológicos , Óvulo/efectos de los fármacos , Inyecciones de Esperma Intracitoplasmáticas
10.
Biol Reprod ; 90(3): 63, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24501176

RESUMEN

Fully grown oocytes in the ovary are arrested at prophase of meiosis I because of high levels of intraoocyte cAMP that maintain increased levels of cAMP-dependent protein kinase (PKA) activity. Following the luteinizing hormone surge at the time of ovulation, cAMP levels drop, resulting in a reduction in PKA activity that triggers meiotic resumption. Although much is known about the molecular mechanisms of how PKA activity fluctuations initiate the oocyte's reentry into meiosis, significantly less is known about the requirement for PKA activity in the oocyte after exit from the prophase I arrest. Here we show that although PKA activity decreases in the oocyte upon meiotic resumption, it increases throughout meiotic progression from the time of germinal vesicle breakdown (GVBD) until the metaphase II (MII) arrest. Blocking this meiotic maturation-associated increase in PKA activity using the pharmacological inhibitor H89 resulted in altered kinetics of GVBD, defects in chromatin and spindle dynamics, and decreased ability of oocytes to reach MII. These effects appear to be largely PKA specific because inhibitors targeting other kinases did not have the same outcomes. To determine potential proteins that may require PKA phosphorylation during meiosis, we separated oocyte protein extracts on an SDS-PAGE gel, extracted regions of the gel that had corresponding immune reactivity towards an anti-PKA substrate antibody, and performed mass spectrometry and microsequencing. Using this approach, we identified transducin-like enhancer of split-6 (TLE6)-a maternal effect gene that is part of the subcortical maternal complex-as a putative PKA substrate. TLE6 localized to the oocyte cortex throughout meiosis in a manner that is spatially and temporally consistent with the localization of critical PKA subunits. Moreover, we demonstrated that TLE6 becomes phosphorylated in a narrow window following meiotic resumption, and H89 treatment can completely block this phosphorylation when added prior to GVBD but not after. Taken together, these results highlight the importance of oocyte-intrinsic PKA in regulating meiotic progression after the prophase I arrest and offer new insights into downstream targets of its activity.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Oocitos/fisiología , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Proteínas Co-Represoras , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Electroforesis en Gel de Poliacrilamida , Femenino , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Isoquinolinas/metabolismo , Isoquinolinas/farmacología , Masculino , Espectrometría de Masas , Meiosis/fisiología , Profase Meiótica I/efectos de los fármacos , Metafase/fisiología , Ratones , Datos de Secuencia Molecular , Oligopéptidos/metabolismo , Oocitos/enzimología , Oocitos/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Especificidad por Sustrato , Sulfonamidas/metabolismo , Sulfonamidas/farmacología
11.
Epigenetics ; 18(1): 2139986, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36328762

RESUMEN

Clinically, developmental exposure to the endocrine disrupting chemical, diethylstilboestrol (DES), results in long-term male and female infertility. Experimentally, developmental exposure to DES results in abnormal reproductive tract phenotypes in male and female mice. Previously, we reported that neonatal DES exposure causes ERα-mediated aberrations in the transcriptome and in DNA methylation in seminal vesicles (SVs) of adult mice. However, only a subset of DES-altered genes could be explained by changes in DNA methylation. We hypothesized that alterations in histone modification may also contribute to the altered transcriptome during SV development. To test this idea, we performed a series of genome-wide analyses of mouse SVs at pubertal and adult developmental stages in control and DES-exposed wild-type and ERα knockout mice. Neonatal DES exposure altered ERα-mediated mRNA and lncRNA expression in adult SV, including genes encoding chromatin-modifying proteins that can impact histone H3K27ac modification. H3K27ac patterns, particularly at enhancers, and DNA methylation were reprogrammed over time during normal SV development and after DES exposure. Some of these reprogramming changes were ERα-dependent, but others were ERα-independent. A substantial number of DES-altered genes had differential H3K27ac peaks at nearby enhancers. Comparison of gene expression changes, H3K27ac marks and DNA methylation marks between adult SV and adult uterine tissue from ovariectomized mice neonatally exposed to DES revealed that most of the epigenetic changes and altered genes were distinct in the two tissues. These findings indicate that the effects of developmental DES exposure cause reprogramming of reproductive tract tissue differentiation through multiple epigenetic mechanisms.


Asunto(s)
Dietilestilbestrol , Receptor alfa de Estrógeno , Animales , Ratones , Masculino , Femenino , Dietilestilbestrol/farmacología , Receptor alfa de Estrógeno/genética , Metilación de ADN , Estudio de Asociación del Genoma Completo , Epigénesis Genética , Expresión Génica
12.
Biol Reprod ; 87(1): 10, 1-10, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22553218

RESUMEN

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.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Genisteína/toxicidad , Inmunidad Mucosa/efectos de los fármacos , Oviductos/efectos de los fármacos , Oviductos/inmunología , Fitoestrógenos/toxicidad , Animales , Animales Recién Nacidos , Desarrollo Embrionario/genética , Desarrollo Embrionario/inmunología , Desarrollo Embrionario/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes MHC Clase II/efectos de los fármacos , Genisteína/administración & dosificación , Inmunidad Mucosa/genética , Infertilidad Femenina/inducido químicamente , Infertilidad Femenina/genética , Infertilidad Femenina/inmunología , Infertilidad Femenina/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Oviductos/metabolismo , Oviductos/patología , Fitoestrógenos/administración & dosificación , Embarazo
13.
Biol Reprod ; 86(3): 1-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22075476

RESUMEN

Metastasis-associated protein 3 (MTA3) is a constituent of the Mi-2/nucleosome remodeling and deacetylase (NuRD) protein complex that regulates gene expression by altering chromatin structure and can facilitate cohesin loading onto DNA. The biological function of MTA3 within the NuRD complex is unknown. Herein, we show that MTA3 was expressed highly in granulosa cell nuclei of all ovarian follicle stages and at lower levels in corpora lutea. We tested the hypothesis that MTA3-NuRD complex function is required for granulosa cell proliferation. In the ovary, MTA3 interacted with NuRD proteins CHD4 and HDAC1 and the core cohesin complex protein RAD21. In cultured mouse primary granulosa cells, depletion of endogenous MTA3 using RNA interference slowed cell proliferation; this effect was rescued by coexpression of exogenous MTA3. Slowing of cell proliferation correlated with a significant decrease in cyclin B1 and cyclin B2 expression. Granulosa cell populations lacking MTA3 contained a significantly higher percentage of cells in G2/M phase and a lower percentage in S phase compared with control cells. Furthermore, MTA3 depletion slowed entry into M phase as indicated by reduced phosphorylation of histone H3 at serine 10. These findings provide the first evidence to date that MTA3 interacts with NuRD and cohesin complex proteins in the ovary in vivo and regulates G2/M progression in proliferating granulosa cells.


Asunto(s)
Ciclo Celular/fisiología , División Celular/fisiología , Proliferación Celular , Fase G2/fisiología , Células de la Granulosa/citología , Proteínas de Neoplasias/fisiología , Animales , Proteínas de Ciclo Celular/fisiología , Células Cultivadas , Proteínas Cromosómicas no Histona/fisiología , Ciclina B1/fisiología , Ciclina B2/fisiología , Femenino , Células de la Granulosa/fisiología , Técnicas In Vitro , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/fisiología , Ratones , Ratones Endogámicos , Modelos Animales , Cohesinas
14.
Reproduction ; 143(3): 247-60, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22223686

RESUMEN

Phytoestrogens, estrogenic compounds derived from plants, are ubiquitous in human and animal diets. These chemicals are generally much less potent than estradiol but act via similar mechanisms. The most common source of phytoestrogen exposure to humans is soybean-derived foods that are rich in the isoflavones genistein and daidzein. These isoflavones are also found at relatively high levels in soy-based infant formulas. Phytoestrogens have been promoted as healthy alternatives to synthetic estrogens and are found in many dietary supplements. The aim of this review is to examine the evidence that phytoestrogen exposure, particularly in the developmentally sensitive periods of life, has consequences for future reproductive health.


Asunto(s)
Desarrollo Infantil/efectos de los fármacos , Fitoestrógenos/farmacología , Reproducción/efectos de los fármacos , Animales , Dieta , Humanos , Lactante , Alimentos Infantiles/efectos adversos , Fórmulas Infantiles/química , Recién Nacido , Fitoestrógenos/efectos adversos , Reproducción/fisiología
15.
J Nutr ; 140(12): 2322S-2325S, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20980642

RESUMEN

Ovarian function in adults is controlled by hormones circulating in the body. The primary hormone responsible for cyclicity in animals and humans is estrogen. Estrogen is mostly produced in the ovary and enters the circulation where it then signals the brain for a response. The parts of the brain that controls reproductive hormones are the hypothalamus and anterior pituitary. Estrogen stimulates the hypothalamus to produce gonadotropin releasing hormone, which in turn signals the anterior pituitary to produce follicle stimulating hormone and luteinizing hormone. These hormones enter the circulation and signal the ovary to ovulate. Substances with estrogenic activity can potentially interfere with this signaling if levels of activity are sufficient to cause a response. Soy foods contain estrogenic substances called phytoestrogens. The predominant phytoestrogens found in soy are genistein and daidzein. The female reproductive system is dependent on hormones for proper function and phytoestrogens at very high levels can interfere with this process. This paper summarizes the literature on adult soy consumption and its effect on ovarian function.


Asunto(s)
Proteínas de Soja , Estrógenos/biosíntesis , Femenino , Humanos
16.
Environ Health Perspect ; 128(3): 37001, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32186404

RESUMEN

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.


Asunto(s)
Implantación del Embrión/efectos de los fármacos , Genisteína/efectos adversos , Fitoestrógenos/efectos adversos , Útero/efectos de los fármacos , Animales , Femenino , Ratones , Embarazo , Útero/crecimiento & desarrollo , Útero/fisiopatología
17.
Endocrinology ; 161(2)2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31883000

RESUMEN

Estrogen receptor α (ESR1; encoded by Esr1) is a crucial nuclear transcription factor for female reproduction and is expressed throughout the female reproductive tract. To assess the function of ESR1 in reproductive tissues without confounding effects from a potential developmental defect arising from global deletion of ESR1, we generated a mouse model in which Esr1 was specifically ablated during postnatal development. To accomplish this, a progesterone receptor Cre line (PgrCre) was bred with Esr1f/f mice to create conditional knockout of Esr1 in reproductive tissues (called PgrCreEsr1KO mice) beginning around 6 days after birth. In the PgrCreEsr1KO oviduct, ESR1 was most efficiently ablated in the isthmic region. We found that at 3.5 days post coitus (dpc), embryos were retrieved from the uterus in control littermates while all embryos were retained in the PgrCreEsr1KO oviduct. Additionally, serum progesterone (P4) levels were significantly lower in PgrCreEsr1KO compared to controls at 3.5 dpc. This finding suggests that expression of ESR1 in the isthmus and normal P4 levels allow for successful embryo transport from the oviduct to the uterus. Therefore, alterations in oviductal isthmus ESR1 signaling and circulating P4 levels could be related to female infertility conditions such as tubal pregnancy.


Asunto(s)
Desarrollo Embrionario , Receptor alfa de Estrógeno/fisiología , Trompas Uterinas/fisiología , Útero/metabolismo , Animales , Estradiol/sangre , Femenino , Fertilidad , Hormona Luteinizante/sangre , Masculino , Ratones , Ratones Noqueados , Hipófisis/metabolismo , Embarazo , Embarazo Tubario/metabolismo , Progesterona/sangre
18.
Biol Reprod ; 81(4): 690-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19535786

RESUMEN

Developmental exposure to endocrine-disrupting compounds is hypothesized to adversely affect female reproductive physiology by interfering with the organization of the hypothalamic-pituitary-gonadal axis. Here, we compared the effects of neonatal exposure to two environmentally relevant doses of the plastics component bisphenol-A (BPA; 50 microg/kg and 50 mg/kg) with the ESR1 (formerly known as ERalpha)-selective agonist 4,4',4''-(4-propyl-[(1)H]pyrazole-1,3,5-triyl)trisphenol (PPT; 1 mg/kg) on the development of the female rat hypothalamus and ovary. An oil vehicle and estradiol benzoate (EB; 25 microg) were used as negative and positive controls. Exposure to EB, PPT, or the low dose of BPA advanced pubertal onset. A total of 67% of females exposed to the high BPA dose were acyclic by 15 wk after vaginal opening compared with 14% of those exposed to the low BPA dose, all of the EB- and PPT-treated females, and none of the control animals. Ovaries from the EB-treated females were undersized and showed no evidence of folliculogenesis, whereas ovaries from the PPT-treated females were characterized by large antral-like follicles, which did not appear to support ovulation. Severity of deficits within the BPA-treated groups increased with dose and included large antral-like follicles and lower numbers of corpora lutea. Sexual receptivity, examined after ovariectomy and hormone replacement, was normal in all groups except those neonatally exposed to EB. FOS induction in hypothalamic gonadotropic (GnRH) neurons after hormone priming was impaired in the EB- and PPT-treated groups but neither of the BPA-treated groups. Our data suggest that BPA disrupts ovarian development but not the ability of GnRH neurons to respond to steroid-positive feedback.


Asunto(s)
Estrógenos no Esteroides/toxicidad , Hipotálamo/efectos de los fármacos , Ovario/efectos de los fármacos , Fenoles/toxicidad , Conducta Sexual Animal/efectos de los fármacos , Animales , Animales Recién Nacidos , Compuestos de Bencidrilo , Relación Dosis-Respuesta a Droga , Estrógenos no Esteroides/administración & dosificación , Ciclo Estral/efectos de los fármacos , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Masculino , Células Neuroendocrinas/efectos de los fármacos , Ovario/patología , Fenoles/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Long-Evans , Maduración Sexual/efectos de los fármacos
19.
Mol Cancer Res ; 17(12): 2369-2382, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597742

RESUMEN

Early-life exposure to estrogenic chemicals can increase cancer risk, likely by disrupting normal patterns of cellular differentiation. Female mice exposed neonatally to the synthetic estrogen diethylstilbestrol (DES) develop metaplastic and neoplastic uterine changes as adults. Abnormal endometrial glands express the oncofetal protein sine oculis homeobox 1 (SIX1) and contain cells with basal [cytokeratin (CK)14+/18-] and poorly differentiated features (CK14+/18+), strongly associating SIX1 with aberrant differentiation and cancer. Here, we tested whether SIX1 expression is necessary for abnormal endometrial differentiation and DES-induced carcinogenesis by using Pgr-cre to generate conditional knockout mice lacking uterine Six1 (Six1 d/d). Interestingly, corn oil (CO) vehicle-treated Six1 d/d mice develop focal endometrial glandular dysplasia and features of carcinoma in situ as compared with CO wild-type Six1 (Six1 +/+) mice. Furthermore, Six1 d/d mice neonatally exposed to DES had a 42% higher incidence of endometrial cancer relative to DES Six1 +/+ mice. Although DES Six1 d/d mice had >10-fold fewer CK14+/18- basal cells within the uterine horns as compared with DES Six1 +/+ mice, the appearance of CK14+/18+ cells remained a feature of neoplastic lesions. These findings suggest that SIX1 is required for normal endometrial epithelial differentiation, CK14+/18+ cells act as a cancer progenitor population, and SIX1 delays DES-induced endometrial carcinogenesis by promoting basal differentiation of CK14+/18+ cells. In human endometrial biopsies, 35% of malignancies showed CK14+/18+ expression, which positively correlated with tumor stage and grade and was not present in normal endometrium. IMPLICATIONS: Aberrant epithelial differentiation is a key feature in both the DES mouse model of endometrial cancer and human endometrial cancer. The association of CK14+/18+ cells with human endometrial cancer provides a novel cancer biomarker and could lead to new therapeutic strategies.


Asunto(s)
Dietilestilbestrol/toxicidad , Hiperplasia Endometrial/genética , Neoplasias Endometriales/genética , Estrógenos/toxicidad , Proteínas de Homeodominio/genética , Animales , Animales Recién Nacidos , Carcinogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Aceite de Maíz/farmacología , Dietilestilbestrol/farmacología , Modelos Animales de Enfermedad , Hiperplasia Endometrial/inducido químicamente , Hiperplasia Endometrial/patología , Neoplasias Endometriales/inducido químicamente , Neoplasias Endometriales/patología , Endometrio/efectos de los fármacos , Endometrio/patología , Células Epiteliales/efectos de los fármacos , Estrógenos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Queratina-14/genética , Ratones
20.
Int J Androl ; 31(2): 201-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18315718

RESUMEN

Environmental chemicals with hormone-like activity can disrupt the programming of endocrine signalling pathways that are established during perinatal life and result in adverse consequences that may not be apparent until much later in life. Increasing evidence implicates developmental exposure to environmental hormone mimics with a growing list of adverse health consequences in both males and females. Most recently, obesity has been proposed to be yet another adverse health effect of exposure to endocrine disrupting chemicals (EDCs) during critical stages of development. Obesity is quickly becoming a significant human health crisis because it is reaching epidemic proportions worldwide, and is associated with chronic illnesses such as diabetes and cardiovascular disease. In this review, we summarize the literature reporting an association of EDCs and the development of obesity, and further describe an animal model of exposure to diethylstilbestrol that has proven useful in studying mechanisms involved in abnormal programming of various oestrogen target tissues during differentiation. Together, these data suggest new targets (i.e. adipocyte differentiation and mechanisms involved in weight homeostasis) of abnormal programming by EDCs, and provide evidence that support the scientific term 'the developmental origins of adult disease'. The emerging idea of an association of EDCs and obesity expands the focus on obesity from intervention and treatment to include prevention and avoidance of these chemical modifiers.


Asunto(s)
Disruptores Endocrinos/toxicidad , Obesidad/inducido químicamente , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Feto/fisiopatología , Humanos , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA