Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Endocrinol ; 28(4): 499-511, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24552588

RESUMO

Ovarian follicles form through a process in which somatic pregranulosa cells encapsulate individual germ cells from germ cell syncytia. Complementary expression of the Notch ligand, Jagged1, in germ cells and the Notch receptor, Notch2, in pregranulosa cells suggests a role for Notch signaling in mediating cellular interactions during follicle assembly. Using a Notch reporter mouse, we demonstrate that Notch signaling is active within somatic cells of the embryonic ovary, and these cells undergo dramatic reorganization during follicle histogenesis. This coincides with a significant increase in the expression of the ligands, Jagged1 and Jagged2; the receptor, Notch2; and the target genes, Hes1 and Hey2. Histological examination of ovaries from mice with conditional deletion of Jagged1 within germ cells (J1 knockout [J1KO]) or Notch2 within granulosa cells (N2 knockout [N2KO]) reveals changes in follicle dynamics, including perturbations in the primordial follicle pool and antral follicle development. J1KO and N2KO ovaries also contain multi-oocytic follicles, which represent a failure to resolve germ cell syncytia, and follicles with enlarged oocytes but lacking somatic cell growth, signifying a potential role of Notch signaling in follicle activation and the coordination of follicle development. We also observed decreased cell proliferation and increased apoptosis in the somatic cells of both conditional knockout lines. As a consequence of these defects, J1KO female mice are subfertile; however, N2KO female mice remain fertile. This study demonstrates important functions for Jagged1 and Notch2 in the resolution of germ cell syncytia and the coordination of somatic and germ cell growth within follicles of the mouse ovary.


Assuntos
Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Receptor Notch2/metabolismo , Transdução de Sinais , Animais , Apoptose , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Células Germinativas/metabolismo , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/metabolismo , Especificidade de Órgãos , Folículo Ovariano/citologia , Receptor Notch2/deficiência , Proteínas Serrate-Jagged
2.
Endocrinology ; 152(1): 303-12, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21084447

RESUMO

Activin, a member of the TGF-ß superfamily, is an important modulator of FSH synthesis and secretion and is involved in reproductive dysfunctions and cancers. It also regulates ovarian follicle development. To understand the mechanisms and pathways by which activin regulates follicle function, we performed a microarray study and identified 240 activin regulated genes in mouse granulosa cells. The gene most strongly inhibited by activin was Cyp26b1, which encodes a P450 cytochrome enzyme that degrades retinoic acid (RA). Cyp26b1 has been shown to play an important role in male germ cell meiosis, but its expression is largely lost in the ovary around embryonic d 12.5. This study demonstrated that Cyp26b1 mRNA was expressed in granulosa cells of follicles at all postnatal developmental stages. A striking inverse spatial and temporal correlation between Cyp26b1 and activin-ßA mRNA expression was observed. Cyp26b1 expression was also elevated in a transgenic mouse model that has decreased activin expression. The Cyp26 inhibitor R115866 stimulated the proliferation of primary cultured mouse granulosa cells, and a similar effect was observed with RA and activin. A pan-RA receptor inhibitor, AGN194310, abolished the stimulatory effect of either RA or activin on granulosa cell proliferation, indicating an involvement of RA receptor-mediated signaling. Overall, this study provides new insights into the mechanisms of activin action in the ovary. We conclude that Cyp26b1 is expressed in the postnatal mouse ovary, regulated by activin, and involved in the control of granulosa cell proliferation.


Assuntos
Ativinas/metabolismo , Proliferação de Células , Sistema Enzimático do Citocromo P-450/metabolismo , Regulação da Expressão Gênica/fisiologia , Células da Granulosa/citologia , Células da Granulosa/fisiologia , Ativinas/genética , Animais , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/genética , Feminino , Masculino , Metalotioneína/genética , Metalotioneína/metabolismo , Camundongos , Camundongos Transgênicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ácido Retinoico 4 Hidroxilase , Tretinoína/metabolismo
3.
J Biol Chem ; 282(50): 36755-65, 2007 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-17951260

RESUMO

Activin, a member of the transforming growth factor-beta superfamily, is an important modulator of follicle-stimulating hormone synthesis and secretion in the pituitary and plays autocrine/paracrine roles in the regulation of ovarian follicle development. From a microarray study on mouse ovarian granulosa cells, we discovered that the estrogen receptor beta (ERbeta) is inducible by activin. We previously demonstrated that estrogen suppresses activin gene expression, suggesting a feedback relationship between these two follicle-regulating hormones. The purpose of this study was to investigate fully activin A regulation of ER expression. Real time reverse transcription-PCR assays on cultured granulosa cells showed that both ERalpha and ERbeta mRNAs were induced by activin A at 4, 12, and 24 h in a dose-responsive manner. Western blots confirmed an increase in their protein levels. Consistent with increased ERalpha and ERbeta expression, activin A stimulated estradiol-induced estrogen response element promoter activity. Activin A stimulation of ER expression was a direct effect at the level of gene transcription, as it was not abolished by cycloheximide but was abolished by actinomycin D, and in transfected granulosa cells activin A stimulated ERalpha promoter activity. To investigate the effect of activin in vivo and, thus, its biological significance, we examined ER expression in inhibin transgenic mice that have decreased activin expression and discovered that these mice had decreased ERalpha and ERbeta expression in the ovary. We also found that ER mRNA levels were decreased in Müllerian inhibiting substance promoter (MIS)-Smad2 dominant negative mice that have impaired activin signaling through Smad2, and small interfering RNAs targeting Smad2 or Smad3 suppressed ERalpha promoter activation, suggesting that Smad2 and Smad3 are involved in regulating ER levels. Therefore, this study reveals an important role for activin in inducing the expression of ERs in the mouse ovary and suggests important interplay between activin and estrogen signaling.


Assuntos
Ativinas/biossíntese , Receptor alfa de Estrogênio/biossíntese , Receptor beta de Estrogênio/biossíntese , Regulação da Expressão Gênica/fisiologia , Células da Granulosa/metabolismo , Transdução de Sinais/fisiologia , Ativinas/genética , Ativinas/farmacologia , Animais , Hormônio Antimülleriano/genética , Hormônio Antimülleriano/metabolismo , Comunicação Autócrina/fisiologia , Células Cultivadas , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Relação Dose-Resposta a Droga , Estradiol/genética , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/citologia , Camundongos , Camundongos Transgênicos , Comunicação Parácrina/fisiologia , Hipófise/metabolismo , Regiões Promotoras Genéticas/fisiologia , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
4.
Mol Endocrinol ; 21(8): 1969-83, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17536003

RESUMO

The GHRH receptor is expressed in the somatotroph cell of the anterior pituitary, where it functions to mediate GHRH-stimulated GH release. To study pituitary and somatotroph cell-specific expression of this gene, a transgenic mouse model and complementary cell culture experiments were developed. The activity of the 1.6-kb proximal rat GHRH receptor promoter was examined in vivo by generating transgenic mice with the promoter directing expression of a luciferase reporter. The promoter directs tissue-specific expression; luciferase is highly expressed in the pituitary but absent from 14 other tissues. Immunocytochemistry experiments show that transgene expression is targeted to GH-expressing somatotroph cells. The transgene is 5-fold more highly expressed in males than females, and there is an increase in transgene expression leading up to the onset of puberty. The 1.6-kb promoter was further examined in cell culture experiments, which revealed that the promoter is selectively activated in pituitary cells and that promoter-reporter expression in nonpituitary cells can be enhanced by the pituitary-specific transcription factor Pit-1. EMSAs identified 10 short regions that specifically bind Pit-1 with highly variable relative affinities. The highest affinity site was previously identified and is required for Pit-1 activation of the promoter. Four additional sites contribute to Pit-1 regulation of the promoter and are important to achieving full activation of the gene. The results show that the 1.6-kb promoter is sufficient to direct tissue- and cell-specific expression in vivo and is regulated by Pit-1.


Assuntos
Regulação da Expressão Gênica/fisiologia , Adeno-Hipófise/metabolismo , Receptores de Neuropeptídeos/biossíntese , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/biossíntese , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Fator de Transcrição Pit-1/fisiologia , Animais , Animais Geneticamente Modificados , Genes Reporter , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Ratos , Transgenes
5.
Endocrinology ; 148(8): 3595-604, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17431007

RESUMO

Chronic ovulation as a contributing factor for the development of epithelial ovarian cancer in women has long been an outstanding hypothesis. To test the incessant ovulation hypothesis, mice were superovulated using weekly ip injections of pregnant mare serum gonadotropin (5 IU/animal), followed 48 h later by human chorionic gonadotropin (5 IU/animal). Wild-type CD1 mice were used along with CD1 mice expressing a Smad2 dominant-negative (Smad2DN) transgene under the control of the Müllerian inhibiting substance promoter that targets expression to the ovary and enhances cyst formation. After chronic injections, ovaries were analyzed from animals 6 months of age for the total adjusted number of cysts, cyst area, cyst location, and key signaling pathways. All observed cysts were confirmed to be of epithelial origin. The number of cysts was not significantly different between superovulated and control mice in either the wild-type or Smad2DN groups. However, the combination of the Smad2DN transgene and superovulation resulted in an increase in cyst formation compared with normal littermates that were unstimulated. Rapid proliferation of the cells lining the cysts was detected using bromodeoxyuridine and phospho-histone 3 immunohistochemistry but was not different in the ovarian surface epithelium or in the cyst lining between groups. These data suggest that chronic superovulation in Smad2DN mice results in a higher incidence of cyst formation compared with unstimulated controls, but the epithelial lined cysts did not progress to cancer over the course of this study.


Assuntos
Cistos Ovarianos/fisiopatologia , Ovulação/fisiologia , Proteína Smad2/genética , Proteína Smad2/metabolismo , Superovulação/fisiologia , Ativinas/metabolismo , Animais , Divisão Celular/fisiologia , Doença Crônica , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Feminino , Gonadotropinas Equinas/farmacologia , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Cistos Ovarianos/complicações , Cistos Ovarianos/patologia , Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/fisiopatologia , Fosforilação , Transdução de Sinais/fisiologia , Superovulação/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
6.
Endocrinology ; 148(5): 1968-76, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17255206

RESUMO

In the ovary, the steroid hormone estrogen and the TGF-beta superfamily member activin are both produced by granulosa cells and they both have intraovarian functions. Emerging evidence has indicated an interaction of these two signaling pathways. Based on the fact that estrogen and activin can impact early follicle formation and development, we hypothesize that estrogen treatment may alter activin signaling in the neonatal ovary. Therefore, this study was designed to examine the effect of neonatal diethylstilbestrol (DES) and estradiol (E(2)) exposure on the mRNA and protein levels of the key factors involved in activin signaling in the mouse ovary. CD-1 mouse pups were given daily injections of DES, E(2), or oil on postnatal d 1-5, and ovaries and sera were collected on d 19. Neonatal DES or E(2) exposure decreased the number of small antral follicles, induced multioocytic follicle formation, and decreased activin beta-subunit mRNA and protein levels. Consistent with local loss of beta-subunit expression, the phosphorylation of Smad 2, a marker of activin-dependent signaling, was decreased in the estrogen-treated ovaries. The decreased beta-subunit expression resulted in a decrease in serum inhibin levels, with a corresponding increase in FSH. Estrogen also suppressed activin subunit gene promoter activities, suggesting a direct transcriptional effect. Overall, this study demonstrates that activin subunits are targets of estrogen action in the early mouse ovary.


Assuntos
Receptores de Ativinas/genética , Ativinas/genética , Estradiol/farmacologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Transdução de Sinais/efeitos dos fármacos , Receptores de Ativinas/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Ativinas/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Dietilestilbestrol/farmacologia , Estrogênios não Esteroides/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Imuno-Histoquímica , Inibinas/genética , Camundongos , Camundongos Endogâmicos , Tamanho do Órgão , Folículo Ovariano/citologia , Gravidez , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia
7.
Dev Biol ; 298(1): 149-54, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16925987

RESUMO

The importance of the initial follicle pool in fertility in female adult mammals has recently been debated. Utilizing a mathematical model of the dynamics of follicle progression (primordial to primary to secondary), we examined whether the initial follicle pool is sufficient for adult fertility through reproductive senescence in CD1 mice. Follicles in each stage were counted from postnatal day 6 through 12 months and data were fit to a series of first-order differential equations representing two mechanisms: an initial pool of primordial follicles as the only follicle source (fixed pool model), or an initial primordial follicle pool supplemented by germline stem cells (stem cell model). The fixed pool model fit the experimental data, accurately representing the maximum observed primary follicle number reached by 4-6 months of age. Although no germline stem cells could be identified by SSEA-1 immunostaining, the stem cell model was tested using a range of de novo primordial follicle production rates. The stem cell model failed to describe the observed decreases in follicles over time and did not parallel the accumulation and subsequent reduction in primary follicles during the early fertile lifespan of the mouse. Our results agree with established dogma that the initial endowment of ovarian follicles is not supplemented by an appreciable number of stem cells; rather, it is sufficient to ensure the fertility needs of the adult mouse.


Assuntos
Fertilidade , Antígenos CD15/metabolismo , Modelos Teóricos , Folículo Ovariano/fisiologia , Ovário/metabolismo , Animais , Animais Recém-Nascidos , Proliferação de Células , Feminino , Células Germinativas/fisiologia , Camundongos , Modelos Biológicos , Oócitos , Transdução de Sinais , Células-Tronco/fisiologia , Fatores de Tempo
8.
Dev Biol ; 298(1): 132-48, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16930587

RESUMO

Mammalian females enter puberty with follicular reserves that exceed the number needed for ovulation during a single lifetime. Follicular depletion occurs throughout reproductive life and ends in menopause, or reproductive senescence, when the follicle pool is exhausted. The mechanisms regulating the production of a species-specific initial follicle pool are not well understood. However, the establishment of a follicular reserve is critical to defining the length of reproductive cyclicity. Here we show that activin A (rh-ActA), a known regulator of follicle formation and growth in vitro, increased the number of postnatal mouse primordial follicles by 30% when administered to neonatal animals during the time of germline cyst breakdown and follicle assembly. This expansion in the initial follicle pool was characterized by a significant increase in both germ cell and granulosa cell proliferation. However, the excess follicles formed shortly after birth did not persist into puberty and both adult rh-ActA- and vehicle-treated animals demonstrated normal fertility. A follicle atresia kinetic constant (k(A)) was modeled for the two groups of animals, and consistent with the empirical data, the k(A) for rh-ActA-treated was twice that of vehicle-treated animals. Kinetic constants for follicle formation, follicle loss and follicle expansion from birth to postnatal day 19 were also derived for vehicle and rh-ActA treatment conditions. Importantly, introduction of exogenous rh-ActA revealed an intrinsic ovarian quorum sensing mechanism that controls the number of follicles available at puberty. We propose that there is an optimal number of oocytes present at puberty, and when the follicle number is exceeded, it occurs at the expense of oocyte quality. The proposed mechanism provides a means by which the ovary eliminates excess follicles containing oocytes of poor quality prior to puberty, thus maintaining fertility in the face of abnormal hormonal stimuli in the prepubertal period.


Assuntos
Ativinas/metabolismo , Ativinas/fisiologia , Fertilidade , Folículo Ovariano/fisiologia , Ovário/metabolismo , Receptores de Ativinas/metabolismo , Ativinas/farmacocinética , Ativinas/farmacologia , Animais , Animais Recém-Nascidos , Proliferação de Células , Feminino , Hormônio Foliculoestimulante/sangue , Células Germinativas/fisiologia , Camundongos , Modelos Biológicos , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Folículo Ovariano/efeitos dos fármacos , Ovário/citologia , Transdução de Sinais , Células-Tronco/fisiologia
9.
Endocrinology ; 147(5): 2338-45, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16484319

RESUMO

The ovarian surface epithelium (OSE) is a monolayer of cells that surround the ovary and accommodate repeated tear and repair in response to ovulation. OSE cells are thought to be the progenitors of 90% of ovarian cancers. Currently, the total amount of proliferation of the OSE has not been reported in response to one ovulatory event. In this study, proliferation of the OSE was quantified in response to superovulation induced by ip injection of pregnant mare serum gonadotropin (PMSG) and human chorionic gonadotropin (hCG) in immature 27-d-old CD1 mice using bromodeoxyuridine (BrdU). BrdU incorporation into the OSE cells was measured from the time of hCG injection for a total cumulative label of 12 h. BrdU incorporation was also measured from the time of PMSG injection for a total label of 60 h to correlate proliferation with specific gonadotropin stimulation. The OSE proliferation was significantly higher in superovulated animals compared with control mice at all time points. Proliferation was also analyzed in discrete anatomical sections and indicated that OSE covering antral follicles and corpora lutea proliferated more rapidly than OSE distal to follicular growth. Finally, apoptosis was assessed in response to ovulation, and virtually no cell death within the OSE was detected. These data demonstrate that the OSE, especially near antral follicles and corpora lutea, proliferates significantly in response to the gonadotropins PMSG and hCG. Therefore, ovarian surface cell division in response to ovulation could contribute to ovarian cancer by proliferation-induced DNA mutations and transformed cell progression.


Assuntos
Células Epiteliais/citologia , Gonadotropinas/metabolismo , Ovário/metabolismo , Animais , Apoptose , Bromodesoxiuridina/farmacologia , Proliferação de Células , Corpo Lúteo/metabolismo , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Modelos Biológicos , Modelos Estatísticos , Mutação , Folículo Ovariano/metabolismo , Ovulação , Superovulação , Fatores de Tempo
10.
Endocrinology ; 146(12): 5228-36, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16141389

RESUMO

Pelvic pain is a common presenting ailment in women often linked to ovulation, endometriosis, early pregnancy, ovarian cancer, and cysts. Clear differential diagnosis for each condition caused by these varied etiologies is difficult and may slow the delivery of therapy that, in the case of ovarian cancer, could be fatal. Ovarian endosalpingiosis, a pelvic condition typified by the presence of cystic glandular structures lined by benign tubal/salpingeal epithelium, is also associated with pelvic pain in women. The exact cellular antecedents of these epithelial lined cystic structures are not known, nor is there a known link to ovarian cancer. A mouse model of ovarian endosalpingiosis has been developed by directing a dominant-negative version of the TGF-beta transcription factor, Smad2, to the ovary using the Müllerian-inhibiting substance promoter (MIS-Smad2-dn). Female mice develop an ovarian endosalpingeal phenotype as early as 3 months of age. Importantly, cysts continuous with the ovarian surface epithelial have been identified, indicating that these cyst cells may be derived from the highly plastic ovarian surface epithelial cell layer. A second transgenic mouse model that causes loss of activin action (inhibin alpha-subunit transgenic mice) develops similar cystic structures, supporting a TGF-beta/activin/Smad2 dependence in the onset of this disease.


Assuntos
Coristoma/patologia , Cistos/patologia , Modelos Animais de Doenças , Tubas Uterinas , Doenças Ovarianas/patologia , Ativinas/antagonistas & inibidores , Animais , Hormônio Antimülleriano , Feminino , Fertilidade , Hormônio Foliculoestimulante/sangue , Expressão Gênica , Marcação de Genes/métodos , Genes Dominantes , Glicoproteínas/genética , Inibinas/genética , Camundongos , Camundongos Transgênicos , Doenças Ovarianas/genética , Ovário/patologia , Fenótipo , Regiões Promotoras Genéticas , Proteína Smad2/genética , Hormônios Testiculares/genética , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...