Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
PLoS Genet ; 14(11): e1007787, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30452456

RESUMEN

Successful embryo implantation requires a receptive endometrium. Poor uterine receptivity can account for implantation failure in women who experience recurrent pregnancy loss or multiple rounds of unsuccessful in vitro fertilization cycles. Here, we demonstrate that the transcription factor Forkhead Box O1 (FOXO1) is a critical regulator of endometrial receptivity in vivo. Uterine ablation of Foxo1 using the progesterone receptor Cre (PgrCre) mouse model resulted in infertility due to altered epithelial cell polarity and apoptosis, preventing the embryo from penetrating the luminal epithelium. Analysis of the uterine transcriptome after Foxo1 ablation identified alterations in gene expression for transcripts involved in the activation of cell invasion, molecular transport, apoptosis, ß-catenin (CTNNB1) signaling pathway, and an increase in PGR signaling. The increase of PGR signaling was due to PGR expression being retained in the uterine epithelium during the window of receptivity. Constitutive expression of epithelial PGR during this receptive period inhibited expression of FOXO1 in the nucleus of the uterine epithelium. The reciprocal expression of PGR and FOXO1 was conserved in human endometrial samples during the proliferative and secretory phase. This demonstrates that expression of FOXO1 and the loss of PGR during the window of receptivity are interrelated and critical for embryo implantation.


Asunto(s)
Implantación del Embrión/genética , Implantación del Embrión/fisiología , Endometrio/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Animales , Núcleo Celular/metabolismo , Polaridad Celular/genética , Polaridad Celular/fisiología , Decidua/fisiología , Endometrio/citología , Femenino , Proteína Forkhead Box O1/deficiencia , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Embarazo , Receptores de Progesterona/deficiencia , Transducción de Señal
2.
FASEB J ; 26(3): 1218-27, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22155565

RESUMEN

The ovarian steroid progesterone, acting through the progesterone receptor (PR), coordinates endometrial epithelial-stromal cell communication, which is critical for its development and function. PR expression in these cellular compartments is under tight temporal and endocrine control. Although ex vivo studies demonstrated the importance of stromal PR expression, they failed to show a role for epithelial PR in uterine function. Here, the in vivo role of PR in the uterine epithelium is defined using floxed PR (PR(f/f)) mice crossed to Wnt7a-Cre mice. Progesterone was unable to stimulate the expression of its epithelial target genes, including Ihh, in the Wnt7a-Cre(+)PR(f/-) mice. Analysis was conducted on Ihh to determine whether PR directly regulates epithelial gene transcription. ChIP-on-chip analysis identified PR binding sites in the 5'-flanking region of Ihh. Cotransfection of the proximal Ihh promoter with PR demonstrated that PR directly regulates Ihh transcription. Female Wnt7a-Cre(+)PR(f/-) mice are infertile due to defects in embryo attachment, stromal cell decidualization, and the inability to cease estrogen-induced epithelial cell proliferation. Finally, progesterone was unable to inhibit neonatal endometrial glandular development in Wnt7a-Cre(+)PR(f/-) mice. Thus, epithelial PR is necessary for the regulation of progesterone epithelial target gene expression, as well as uterine function and development.


Asunto(s)
Células Epiteliales/metabolismo , Receptores de Progesterona/fisiología , Útero/fisiología , Proteínas Wnt/fisiología , Animales , Sitios de Unión/genética , Proliferación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina/métodos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Estrógenos/farmacología , Femenino , Fertilidad/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Ratones Transgénicos , Embarazo , Progesterona/farmacología , Regiones Promotoras Genéticas/genética , Unión Proteica , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Útero/efectos de los fármacos , Útero/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
3.
Chromosome Res ; 20(1): 247-58, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22105695

RESUMEN

The chromosome status of the mammalian embryo initiates a multistage process of sexual development in which the bipotential reproductive system establishes itself as either male or female. These events are governed by intricate cell-cell and interorgan communication that is regulated by multiple signaling pathways. The hedgehog signaling pathway was originally identified for its key role in the development of Drosophila, but is now recognized as a critical developmental regulator in many species, including humans. In addition to its developmental roles, the hedgehog signaling pathway also modulates adult organ function, and misregulation of this pathway often leads to diseases, such as cancer. The hedgehog signaling pathway acts through its morphogenetic ligands that signal from ligand-producing cells to target cells over a specified distance. The target cells then respond in a graded manner based on the concentration of the ligands that they are exposed to. Through this unique mechanism of action, the hedgehog signaling pathway elicits cell fate determination, epithelial-mesenchymal interactions, and cellular homeostasis. Here, we review current findings on the roles of hedgehog signaling in the sexually dimorphic development of the reproductive organs with an emphasis on mammals and comparative evidence in other species.


Asunto(s)
Proteínas Hedgehog/genética , Diferenciación Sexual , Sexo , Transducción de Señal , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Desarrollo Fetal , Regulación del Desarrollo de la Expresión Génica , Genes , Células Germinativas/citología , Células Germinativas/crecimiento & desarrollo , Células Germinativas/metabolismo , Proteínas Hedgehog/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Mamíferos , Reproducción , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo
4.
Biol Reprod ; 87(5): 104, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22954793

RESUMEN

WNT4 is required for normal ovarian follicle development and female fertility in mice, but how its signal is transduced remains unknown. Fzd1 encodes a WNT receptor whose expression is markedly induced in both mural granulosa cells and cumulus cells during the preovulatory period, in a manner similar to Wnt4. To study the physiological roles of FZD1 in ovarian physiology and to determine whether it serves as receptor for WNT4, Fzd1-null mice were created by gene targeting. Whereas rare Fzd1(-/-) females were sterile because of uterine fibrosis and ovarian tubulostromal hyperplasia, most were subfertile, producing ≈1 fewer pup per litter on average relative to controls. Unlike WNT4-deficient mice, ovaries from Fzd1(-/-) mice had normal weights, numbers of follicles, steroid hormone production, and WNT4 target gene expression levels. Microarray analyses of granulosa cells from periovulatory follicles revealed few genes whose expression was altered in Fzd1(-/-) mice. However, gene expression analyses of cumulus-oocyte complexes (COCs) revealed a blunted response of both oocyte (Zp3, Dppa3, Nlrp5, and Bmp15) and cumulus (Btc, Ptgs2, Sema3a, Ptx3, Il6, Nts, Alcam, and Cspg2) genes to the ovulatory signal, whereas the expression of these genes was not altered in WNT4-deficient COCs from Wnt4(tm1.1Boer/tm1.1Boer);Tg (CYP19A1-cre)1Jri mice. Despite altered gene expression, cumulus expansion appeared normal in Fzd1(-/-) COCs both in vitro and in vivo. Together, these results indicate that Fzd1 is required for normal female fertility and may act in part to regulate oocyte maturation and cumulus cell function, but it is unlikely to function as the sole ovarian WNT4 receptor.


Asunto(s)
Células del Cúmulo/fisiología , Fertilidad/fisiología , Receptores Frizzled/fisiología , Regulación de la Expresión Génica/fisiología , Animales , Femenino , Receptores Frizzled/deficiencia , Receptores Frizzled/genética , Expresión Génica , Células de la Granulosa/metabolismo , Ratones , Ratones Noqueados , Análisis por Micromatrices , Folículo Ovárico/crecimiento & desarrollo , Ovario/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Vía de Señalización Wnt/fisiología
5.
FASEB J ; 25(4): 1176-87, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21163860

RESUMEN

WNT4, a member of the Wnt family of ligands, is critical for the development of the female reproductive tract. Analysis of Wnt4 expression in the adult uterus during pregnancy indicates that it may play a role in the regulation of endometrial stromal cell proliferation, survival, and differentiation, which is required to support the developing embryo. To investigate the role of Wnt4 in adult uterine physiology, conditional ablation of Wnt4 using the PR(cre) mouse model was accomplished. Ablation of Wnt4 rendered female mice subfertile due to a defect in embryo implantation and subsequent defects in endometrial stromal cell survival, differentiation, and responsiveness to progesterone signaling. In addition to altered stromal cell function, the uteri of PR(cre/+)Wnt4(f/f) (Wnt4(d/d)) mice displayed altered epithelial differentiation characterized by a reduction in the number of uterine glands and the emergence of a p63-positive basal cell layer beneath the columnar luminal epithelial cells. The altered epithelial cell phenotype was further escalated by chronic estrogen treatment, which caused squamous cell metaplasia of the uterine epithelium in the Wnt4(d/d) mice. Thus, WNT4 is a critical regulator not only of proper postnatal uterine development, but also embryo implantation and decidualization.


Asunto(s)
Decidua/fisiología , Útero/fisiología , Proteínas Wnt/fisiología , Animales , Apoptosis/efectos de los fármacos , Implantación del Embrión/fisiología , Femenino , Ratones , Embarazo , Progesterona/fisiología , Transducción de Señal/fisiología , Útero/crecimiento & desarrollo , Proteína Wnt4
6.
Biol Reprod ; 82(4): 706-13, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20018910

RESUMEN

Normal endometrial function requires a balance of progesterone (P4) and estrogen (E2) effects. E2 acts to stimulate the proliferation of uterine epithelial cells, while P4 action inhibits E2-mediated proliferation of the epithelium. P4 through its cognate receptor, the P4 receptor (Pgr), has important roles in the establishment and maintenance of pregnancy. In previous studies, we have identified ERBB receptor feedback inhibitor 1 (Errfi1) as a downstream target of Pgr action in the uterus. Herein, we show that Errfi1 mRNA expression was significantly increased in the uterus after Day 2.5 of gestation. Its expression is also induced in the uterus by acute E2 treatment, and this induction is synergistically induced by chronic E2 and P4 treatment. Although it is known that conditional ablation of Errfi1 in the Pgr-positive cells (Errfi1(d/d)) results in infertility, the function of Errfi1 in reproductive biology has remained elusive. Using Errfi1(d/d) mice, we have identified Errfi1 as an important mediator of uterine implantation. Epithelial ESR1 and target genes were significantly increased in the uteri of Errfi1(d/d) mice. Our results identify a new signaling paradigm of steroid hormone regulation in female reproductive biology that adds insight into the underlying dysregulation of hormonal signaling in human reproductive disorders such as endometriosis and endometrial cancer.


Asunto(s)
Implantación del Embrión/genética , Receptor alfa de Estrógeno/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Animales , Implantación del Embrión/efectos de los fármacos , Implantación del Embrión/fisiología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Edad Gestacional , Hormonas Esteroides Gonadales/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Útero/efectos de los fármacos , Útero/metabolismo , Útero/fisiología
7.
Biol Reprod ; 82(5): 991-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20130264

RESUMEN

Previous work has identified Indian hedgehog (Ihh) as a major mediator of progesterone signaling during embryo implantation. Ihh acts through its downstream effector smoothened (Smo) to activate the GLI family of transcription factors. In order to gain a better understanding of Ihh action during embryo implantation, we expressed a Cre-recombinase-dependent constitutively activated SMO in the murine uterus using the Pgr(tm2(cre)Lyd) (PR(cre)) mouse model [Pgr(tm2(cre)Lyd+)Gt(ROSA)26Sor(tm1(Smo/EYFP)Amc)(+) (PR(cre/+)SmoM2(+))]. Female PR(cre/+)SmoM2(+) mice were infertile. They exhibited normal serum progesterone levels and normal ovulation, but their ova failed to be fertilized in vivo and their uterus failed to undergo the artificially induced decidual response. Examination of the PR(cre/+)SmoM2(+) uteri revealed numerous features such as uterine hypertrophy, the presence of a stratified luminal epithelial cell layer, a reduced number of uterine glands, and an endometrial stroma that had lost its normal morphologic characteristics. Microarray analysis of 3-mo-old PR(cre/+)SmoM2(+) uteri demonstrated a chondrocytic signature and confirmed that constitutive activation of PR(cre/+)SmoM2(+) increased extracellular matrix production. Thus, constitutive activation of Smo in the mouse uterus alters postnatal uterine differentiation which interferes with early pregnancy. These results provide new insight into the role of Hedgehog signaling during embryo implantation.


Asunto(s)
Implantación del Embrión/fisiología , Infertilidad Femenina/metabolismo , Progesterona/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Útero/crecimiento & desarrollo , Animales , Diferenciación Celular/fisiología , Decidua/crecimiento & desarrollo , Femenino , Regulación de la Expresión Génica/fisiología , Proteínas Hedgehog/metabolismo , Metaboloma , Ratones , Ratones Mutantes , Embarazo , Análisis por Matrices de Proteínas , Receptores de Progesterona/metabolismo , Transducción de Señal/fisiología , Receptor Smoothened , Útero/citología , Útero/metabolismo
8.
Biol Reprod ; 82(4): 783-90, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20056671

RESUMEN

Conditional ablation of Indian hedgehog (Ihh) in the murine uterus results in mice that are sterile because of defects in embryo implantation. We performed microarray analysis on these mice at the time point at which the Ihh target genes are induced by the administration of exogenous hormone to mimic Day 3.5 of pregnancy. This analysis identified 863 genes altered by the conditional ablation of Ihh. Of these, genes that regulated the cell cycle were overrepresented. In addition, genes involved in epidermal growth factor (EGF) and estrogen (E2) signaling were found to be deregulated upon Ihh ablation. Furthermore, upon conditional ablation of Ihh, 15-mo-old mice exhibited hallmarks of estrogenized uteri, such as cystically dilated glands and hyalinized stroma. Thus, Ihh regulates embryo implantation by having an impact on the cell cycle, EGF signaling, and E2 signaling.


Asunto(s)
Ciclo Celular/genética , Factor de Crecimiento Epidérmico/metabolismo , Estradiol/metabolismo , Eliminación de Gen , Proteínas Hedgehog/genética , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Implantación del Embrión/genética , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Epidérmico/fisiología , Estradiol/farmacología , Estradiol/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genes cdc/efectos de los fármacos , Genes cdc/fisiología , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/fisiología , Ratones , Ratones Transgénicos , Modelos Biológicos , Embarazo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Útero/efectos de los fármacos , Útero/metabolismo , Útero/fisiología
9.
Mol Endocrinol ; 22(10): 2336-52, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18687735

RESUMEN

Dicer is an evolutionarily conserved ribonuclease III that is necessary for microRNA (miRNA) processing and the synthesis of small interfering RNAs from long double-stranded RNA. Although it has been shown that Dicer plays important roles in the mammalian germline and early embryogenesis, the functions of Dicer-dependent pathways in the somatic cells of the female reproductive tract are unknown. Using a transgenic line in which Cre recombinase is driven by the anti-Müllerian hormone receptor type 2 promoter, we conditionally inactivated Dicer1 in the mesenchyme of the developing Müllerian ducts and postnatally in ovarian granulosa cells and mesenchyme-derived cells of the oviducts and uterus. Deletion of Dicer in these cell types results in female sterility and multiple reproductive defects including decreased ovulation rates, compromised oocyte and embryo integrity, prominent bilateral paratubal (oviductal) cysts, and shorter uterine horns. The paratubal cysts act as a reservoir for spermatozoa and oocytes and prevent embryos from transiting the oviductal isthmus and passing the uterotubal junction to enter the uterus for implantation. Deep sequencing of small RNAs in oviduct revealed down-regulation of specific miRNAs in Dicer conditional knockout females compared with wild type. The majority of these differentially expressed miRNAs are predicted to regulate genes important for Müllerian duct differentiation and mesenchyme-derived structures, and several of these putative target genes were significantly up-regulated upon conditional deletion of Dicer1. Thus, our findings reveal diverse and critical roles for Dicer and its miRNA products in the development and function of the female reproductive tract.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Endorribonucleasas/metabolismo , Genitales Femeninos/fisiología , Infertilidad/genética , Animales , ARN Helicasas DEAD-box/genética , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/anatomía & histología , Endorribonucleasas/genética , Estradiol/sangre , Ciclo Estral/fisiología , Femenino , Hormona Folículo Estimulante/sangre , Genitales Femeninos/anatomía & histología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , MicroARNs/metabolismo , Oocitos/patología , Oviductos/anatomía & histología , Oviductos/patología , Ovulación/fisiología , Embarazo , Regiones Promotoras Genéticas , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Ribonucleasa III , Espermatozoides/citología
10.
Science ; 357(6352): 717-720, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28818950

RESUMEN

The sexual differentiation paradigm contends that the female pattern of the reproductive system is established by default because the male reproductive tracts (Wolffian ducts) in the female degenerate owing to a lack of androgen. Here, we discovered that female mouse embryos lacking Coup-tfII (chicken ovalbumin upstream promoter transcription factor II) in the Wolffian duct mesenchyme became intersex-possessing both female and male reproductive tracts. Retention of Wolffian ducts was not caused by ectopic androgen production or action. Instead, enhanced phosphorylated extracellular signal-regulated kinase signaling in Wolffian duct epithelium was responsible for the retention of male structures in an androgen-independent manner. We thus suggest that elimination of Wolffian ducts in female embryos is actively promoted by COUP-TFII, which suppresses a mesenchyme-epithelium cross-talk responsible for Wolffian duct maintenance.


Asunto(s)
Factor de Transcripción COUP II/fisiología , Genitales Masculinos/embriología , Diferenciación Sexual/fisiología , Conductos Mesonéfricos/embriología , Andrógenos/metabolismo , Andrógenos/farmacología , Animales , Factor de Transcripción COUP II/genética , Embrión de Mamíferos , Femenino , Masculino , Ratones , Ratones Noqueados , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Diferenciación Sexual/genética , Transducción de Señal
11.
Cell Rep ; 17(5): 1414-1425, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27783953

RESUMEN

Altered progesterone responsiveness leads to female infertility and cancer, but underlying mechanisms remain unclear. Mice with uterine-specific ablation of GATA binding protein 2 (Gata2) are infertile, showing failures in embryo implantation, endometrial decidualization, and uninhibited estrogen signaling. Gata2 deficiency results in reduced progesterone receptor (PGR) expression and attenuated progesterone signaling, as evidenced by genome-wide expression profiling and chromatin immunoprecipitation. GATA2 not only occupies at and promotes expression of the Pgr gene but also regulates downstream progesterone responsive genes in conjunction with the PGR. Additionally, Gata2 knockout uteri exhibit abnormal luminal epithelia with ectopic TRP63 expressing squamous cells and a cancer-related molecular profile in a progesterone-independent manner. Lastly, we found a conserved GATA2-PGR regulatory network in both human and mice based on gene signature and path analyses using gene expression profiles of human endometrial tissues. In conclusion, uterine Gata2 regulates a key regulatory network of gene expression for progesterone signaling at the early pregnancy stage.


Asunto(s)
Endometrio/metabolismo , Redes Reguladoras de Genes/genética , Progesterona/metabolismo , Animales , Secuencia de Bases , Secuencia Conservada/genética , Implantación del Embrión , Femenino , Factor de Transcripción GATA2/metabolismo , Humanos , Ratones , Fosfoproteínas/metabolismo , Embarazo , Progesterona/sangre , Unión Proteica/genética , Receptores de Progesterona/metabolismo , Transducción de Señal/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo
12.
Mol Endocrinol ; 28(12): 2025-37, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25333515

RESUMEN

Progesterone receptors (PRs) are phosphorylated on multiple sites, and a variety of roles for phosphorylation have been suggested by cell-based studies. Previous studies using PR-null mice have shown that PR plays an important role in female fertility, regulation of uterine growth, the uterine decidualization response, and proliferation as well as ductal side-branching and alveologenesis in the mammary gland. To study the role of PR phosphorylation in vivo, a mouse was engineered with homozygous replacement of PR with a PR serine-to-alanine mutation at amino acid 191. No overt phenotypes were observed in the mammary glands or uteri of PR S191A treated with progesterone (P4). In contrast, although PR S191A mice were fertile, litters were 19% smaller than wild type and the estrous cycle was lengthened slightly. Moreover, P4-dependent gene regulation in primary mammary epithelial cells (MECs) was altered in a gene-selective manner. MECs derived from wild type and PR S191A mice were grown in a three-dimensional culture. Both formed acinar structures that were morphologically similar, and proliferation was stimulated equally by P4. However, P4 induction of receptor activator of nuclear factor-κB ligand and calcitonin was selectively reduced in S191A cultures. These differences were confirmed in freshly isolated MECs. Chromatin immunoprecipitation analysis showed that the binding of S191A PR to some of the receptor activator of nuclear factor-κB ligand enhancers and a calcitonin enhancer was substantially reduced. Thus, the elimination of a single phosphorylation site is sufficient to modulate PR activity in vivo. PR contains many phosphorylation sites, and the coordinate regulation of multiple sites is a potential mechanism for selective modulation of PR function.


Asunto(s)
Receptores de Progesterona/química , Receptores de Progesterona/metabolismo , Serina/metabolismo , Animales , Femenino , Inmunohistoquímica , Ratones , Fosforilación , Receptores de Progesterona/genética , Serina/química
13.
Mol Endocrinol ; 27(3): 548-54, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23340253

RESUMEN

In order to understand the biology of the endometrium and potentially develop new diagnostic tools and treatments for endometrial diseases, the highly orchestrated gene expression/regulation that occurs within the uterus must first be understood. Even though a wealth of information on endometrial gene expression/regulation is available, this information is scattered across several different resources in formats that can be difficult for the average bench scientist to query, integrate, and utilize. The Endometrium Database Resource (EDR) was created as a single evolving resource for protein- and micro-RNA-encoding genes that have been shown by gene expression microarray, Northern blot, or other experiments in the literature to have their expression regulated in the uterus of humans, mice, rats, cows, domestic pigs, guinea pigs, and sheep. Genes are annotated in EDR with basic gene information (eg, gene symbol and chromosome), gene orthologs, and gene ontologies. Links are also provided to external resources for publication/s, nucleic and amino acid sequence, gene product function, and Gene Expression Omnibus (GEO) phase expression graph information. The resource also allows for direct comparison of relative gene expression in different microarray experiments for genes shown in the literature to be differentially expressed in the uterus. It is available via a user-friendly, web-based interface and is available without charge or restriction to the entire scientific community. The EDR can be accessed at http://edr.research.bcm.edu.


Asunto(s)
Bases de Datos Genéticas , Endometrio/metabolismo , Investigación , Animales , Femenino , Regulación de la Expresión Génica , Humanos , Anotación de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos
14.
PLoS One ; 7(10): e47359, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133512

RESUMEN

Fetal testis steroidogenesis plays an important role in the reproductive development of the male fetus. While regulators of certain aspects of steroidogenesis are known, the initial driver of steroidogenesis in the human and rodent fetal testis is unclear. Through comparative analysis of rodent fetal testis microarray datasets, 54 candidate fetal Leydig cell-specific genes were identified. Fetal mouse testis interstitial expression of a subset of these genes with unknown expression (Crhr1, Gramd1b, Itih5, Vgll3, and Vsnl1) was verified by whole-mount in situ hybridization. Among the candidate fetal Leydig cell-specific factors, three receptors (CRHR1, PRLR, and PROKR2) were tested for a steroidogenic function using ex vivo fetal testes treated with receptor agonists (CRH, PRL, and PROK2). While PRL and PROK2 had no effect, CRH, at low (approximately 1 to 10) nM concentration, increased expression of the steroidogenic genes Cyp11a1, Cyp17a1, Scarb1, and Star in GD15 mouse and GD17 rat testes, and in conjunction, testosterone production was increased. Exposure of GD15 fetal mouse testis to a specific CRHR1 antagonist blunted the CRH-induced steroidogenic gene expression and testosterone responses. Similar to ex vivo rodent fetal testes, ≥ 10 nM CRH exposure of MA-10 Leydig cells increased steroidogenic pathway mRNA and progesterone levels, showing CRH can enhance steroidogenesis by directly targeting Leydig cells. Crh mRNA expression was observed in rodent fetal hypothalamus, and CRH peptide was detected in rodent amniotic fluid. Together, these data provide a resource for discovering factors controlling fetal Leydig cell biology and suggest that CRHR1 activation by CRH stimulates rat and mouse fetal Leydig cell steroidogenesis in vivo.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Intersticiales del Testículo/metabolismo , Receptores de Hormona Liberadora de Corticotropina/agonistas , Receptores de Hormona Liberadora de Corticotropina/fisiología , Testículo/embriología , Líquido Amniótico/metabolismo , Animales , Proteína C-Reactiva/metabolismo , Hormonas Gastrointestinales/metabolismo , Genómica , Inmunohistoquímica/métodos , Masculino , Ratones , Neuropéptidos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Prolactina/biosíntesis , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Esteroides/metabolismo , Testosterona/metabolismo , Factores de Tiempo , Urocortinas/metabolismo
15.
Gene Expr Patterns ; 12(5-6): 196-203, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22476030

RESUMEN

In mammals, such as mouse and human, timely production of the progesterone receptor (PR) in the proper uterine compartments is critical for preparing the uterus for the initiation and maintenance of pregnancy. Developmentally, the expression of GATA2, a member of the six member zinc-finger family of transcription factors, has been shown to be necessary for multiple non-related tissues, such as the hematopoietic system, adipose maturation and the urogential system. We recently identified Gata2 as a potential progesterone target gene in the mouse uterus; however, the expression of the GATA genes in the mouse uterus during pregnancy has not been demonstrated. In the present study, we examined the expression of GATA2 protein during the phases of pregnancy, including early pregnancy where progesterone (P4) signaling is critical in order to facilitate the window of receptivity for embryo implantation and during the decidualization of the uterine stroma, a process of cellular proliferation and differentiation which is necessary for maintenance of the invading embryo until placentation occurs. Here, we report that GATA2 protein is expressed in the uterine luminal and glandular epithelium pre-implantation, spatio-temporally co-localizing with that of the PR. Additionally, GATA2 continues to be expressed in the decidualized stroma throughout early pregnancy indicating a role in the maintenance of decidual cells. Based on these findings, we conclude that GATA2 is expressed during critical phases of early pregnancy, similar to that of the PR, and that it may play a major role in mediating P4 signaling in the mouse uterus.


Asunto(s)
Factor de Transcripción GATA2/metabolismo , Embarazo/metabolismo , Útero/metabolismo , Animales , Línea Celular , Decidua/metabolismo , Implantación del Embrión , Endometrio/metabolismo , Femenino , Humanos , Ratones , Promegestona/metabolismo , Seudoembarazo/metabolismo , Receptores de Progesterona/agonistas , Receptores de Progesterona/análisis
16.
Mol Endocrinol ; 26(8): 1428-42, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22638070

RESUMEN

Progesterone (P4) signaling through its nuclear transcription factor, the progesterone receptor (PR), is essential for normal uterine function. Although deregulation of PR-mediated signaling is known to underscore uterine dysfunction and a number of endometrial pathologies, the early molecular mechanisms of this deregulation are unclear. To address this issue, we have defined the genome-wide PR cistrome in the murine uterus using chromatin immunoprecipitation (ChIP) followed by massively parallel sequencing (ChIP-seq). In uteri of ovariectomized mice, we identified 6367 PR-binding sites in the absence of P4 ligand; however, this number increased at nearly 3-fold (18,432) after acute P4 exposure. Sequence analysis revealed that approximately 73% of these binding sites contain a progesterone response element or a half-site motif recognized by the PR. Many previously identified P4 target genes known to regulate uterine function were found to contain PR-binding sites, confirming the validity of our methodology. Interestingly, when the ChIP-seq data were coupled with our microarray expression data, we identified a novel regulatory role for uterine P4 in circadian rhythm gene expression, thereby uncovering a hitherto unexpected new circadian biology for P4 in this tissue. Further mining of the ChIP-seq data revealed Sox17 as a direct transcriptional PR target gene in the uterus. As a member of the Sox transcription factor family, Sox17 represents a potentially novel mediator of PR action in the murine uterus. Collectively, our first line of analysis of the uterine PR cistrome provides the first insights into the early molecular mechanisms that underpin normal uterine responsiveness to acute P4 exposure. Future analysis promises to reveal the PR interactome and, in turn, potential therapeutic targets for the diagnosis and/or treatment of endometrial dysfunction.


Asunto(s)
Proteínas HMGB/metabolismo , Receptores de Progesterona/metabolismo , Factores de Transcripción SOXF/metabolismo , Útero/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Secuencia de Consenso , Femenino , Genoma , Humanos , Ratones , Ratones Endogámicos C57BL , Anotación de Secuencia Molecular , Progesterona/fisiología , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Elementos de Respuesta , Análisis de Secuencia de ADN , Transducción de Señal , Transcriptoma
17.
J Oncol ; 2010: 139087, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19884980

RESUMEN

Endometrial cancer is the most common gynecological cancer. Estrogen-dependent endometrioid carcinoma is the most common type of endometrial cancer, and alterations in the expression of PTEN and K-ras have been associated with this disease. To study the roles of Pten and K-ras in endometrial cancer, we generated Pten ablation and oncogenic K-ras mutation in progesterone receptor positive cells (PR(cre/+)Pten(f/f)K-ras(G12D)). Double mutant mice dramatically accelerated the development of endometrial cancer compared to a single mutation of either gene. Histological analysis showed that all of the 1-month old double mutant female mice developed endometrial cancer with myometrial invasion. The expression of PR was downregulated in double mutant mice compared to a single mutation of either gene which resulted in decreased suppression of estrogen signaling. Therefore, these results suggest a synergistic effect of dysregulation of the Pten and K-ras signaling pathways during endometrial tumorigenesis.

18.
Mol Endocrinol ; 24(6): 1251-66, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20363875

RESUMEN

Bone morphogenetic proteins (BMPs) have diverse roles in development and reproduction. Although several BMPs are produced by oocytes, thecal cells, and granulosa cells of developing follicles, the in vivo functions of most of these ligands are unknown. BMP signals are transduced by multiple type I and type II TGFbeta family receptors, and of the type I receptors, BMP receptor 1A (BMPR1A) and BMP receptor 1B (BMPR1B) are known to be expressed in rodent granulosa cells. Female mice homozygous null for Bmpr1b are sterile due to compromised cumulus expansion, but the function of BMPR1A in the ovary is unknown. To further decipher a role for BMP signaling in mouse granulosa cells, we deleted Bmpr1a in the granulosa cells of the ovary and found Bmpr1a conditional knockout females to be subfertile with reduced spontaneous ovulation. To explore the redundant functions of BMP receptor signaling in the ovary, we generated Bmpr1a Bmpr1b double-mutant mice, which developed granulosa cell tumors that have evidence of increased TGFbeta and hedgehog signaling. Thus, similar to SMAD1 and SMAD5, which have redundant roles in suppressing granulosa cell tumor development in mice, two type I BMP receptors, BMPR1A and BMPR1B, function together to prevent ovarian tumorigenesis. These studies support a role for a functional BMP signaling axis as a tumor suppressor pathway in the ovary, with BMPR1A and BMPR1B acting downstream of BMP ligands and upstream of BMP receptor SMADs.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Fertilidad/fisiología , Células de la Granulosa/metabolismo , Neoplasias Ováricas/metabolismo , Lesiones Precancerosas/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Células del Cúmulo/metabolismo , Células del Cúmulo/patología , Ciclo Estral/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Células de la Granulosa/patología , Proteínas Hedgehog/metabolismo , Hormonas/sangre , Ratones , Ratones Noqueados , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/fisiopatología , Lesiones Precancerosas/patología , Lesiones Precancerosas/fisiopatología , Transducción de Señal
19.
Semin Cell Dev Biol ; 19(2): 178-86, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18280760

RESUMEN

In order for a successful pregnancy to occur, the embryo must attach to the luminal epithelial cells and invade into the stroma. Then, the surrounding stromal cells need to undergo decidualization in order to establish the vasculature necessary for survival of the embryo. These events in early pregnancy are tightly regulated by the steroid hormones, estrogen (E2) and progesterone (P4), through their cognate receptors, the estrogen receptor (ER) and the progesterone receptor (PR), respectively. Using a mouse model in which the PR has been ablated, it was demonstrated that the PR is necessary for embryo implantation and decidualization. Therefore, understanding the mechanism of PR action in the adult uterus is necessary in order to understand the events of early pregnancy. Insights from both mouse models and human samples have been integral in elucidating uterine PR action. These studies have shown that not only PR target genes, but also mediators of PR action are important for correct PR action in early pregnancy. Many of the genes involved in PR action in early pregnancy have also been shown to have roles in uterine diseases such as endometriosis and endometrial cancer. Therefore, the integration of mouse and human studies on PR action in the uterus will be important for the future understanding of uterine diseases and in the development of treatment for these diseases.


Asunto(s)
Implantación del Embrión/fisiología , Receptores de Progesterona/fisiología , Útero/metabolismo , Animales , Femenino , Humanos , Ratones , Ratones Noqueados , Modelos Animales , Modelos Biológicos , Embarazo , Receptores de Progesterona/análisis , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transducción de Señal/genética , Enfermedades Uterinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA