Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Transl Med ; 19(1): 90, 2021 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-33639972

RESUMO

BACKGROUND: Adult granulosa cell tumor (aGCT) is a rare type of stromal cell malignant cancer of the ovary characterized by elevated estrogen levels. aGCTs ubiquitously harbor a somatic mutation in FOXL2 gene, Cys134Trp (c.402C < G); however, the general molecular effect of this mutation and its putative pathogenic role in aGCT tumorigenesis is not completely understood. We previously studied the role of FOXL2C134W, its partner SMAD3 and its antagonist FOXO1 in cellular models of aGCT. METHODS: In this work, seeking more comprehensive profiling of FOXL2C134W transcriptomic effects, we performed an RNA-seq analysis comparing the effect of FOXL2WT/SMAD3 and FOXL2C134W/SMAD3 overexpression in an established human GC line (HGrC1), which is not luteinized, and bears normal alleles of FOXL2. RESULTS: Our data shows that FOXL2C134W/SMAD3 overexpression alters the expression of 717 genes. These genes include known and novel FOXL2 targets (TGFB2, SMARCA4, HSPG2, MKI67, NFKBIA) and are enriched for neoplastic pathways (Proteoglycans in Cancer, Chromatin remodeling, Apoptosis, Tissue Morphogenesis, Tyrosine Kinase Receptors). We additionally expressed the FOXL2 antagonistic Forkhead protein, FOXO1. Surprisingly, overexpression of FOXO1 mitigated 40% of the altered genome-wide effects specifically related to FOXL2C134W, suggesting it can be a new target for aGCT treatment. CONCLUSIONS: Our transcriptomic data provide novel insights into potential genes (FOXO1 regulated) that could be used as biomarkers of efficacy in aGCT patients.


Assuntos
Tumor de Células da Granulosa , Neoplasias Ovarianas , Adulto , Linhagem Celular Tumoral , DNA Helicases , Feminino , Proteína Forkhead Box L2 , Proteína Forkhead Box O1/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Tumor de Células da Granulosa/genética , Humanos , Mutação , Proteínas Nucleares , Neoplasias Ovarianas/genética , Proteína Smad3/genética , Fatores de Transcrição , Transcriptoma/genética
2.
J Transl Med ; 19(1): 428, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34654452

RESUMO

BACKGROUND: In the ovarian follicle, the Theca Cells (TCs) have two main functions: preserving morphological integrity and, importantly, secreting steroid androgen hormones. TCs express the essential enzyme 17α-hydroxylase/17,20-desmolase (CYP17), which permits the conversion of pregnenolone and progesterone into androgens. Dysregulation of CYP17 enzyme activity due to an intrinsic ovarian defect is hypothesized to be a cause of hyperandrogenism in women. Androgen excess is observed in women with polycystic ovary syndrome (PCOS) resulting from excess endogenous androgen production, and in transgender males undergoing exogenous testosterone therapy after female sex assignment at birth. However, the molecular and morphological effects of Cyp17 overexpression and androgen excess on folliculogenesis is unknown. METHODS: In this work, seeking a comprehensive profiling of the local outcomes of the androgen excess in the ovary, we generated a transgenic mouse model (TC17) with doxycycline (Dox)-induced Cyp17 overexpression in a local and temporal manner. TC17 mice were obtained by a combination of the Tet-dependent expression system and the Cre/LoxP gene control system. RESULTS: Ovaries of Dox-treated TC17 mice overexpressed Cyp17 specifically in TCs, inducing high testosterone levels. Surprisingly, TC17 ovarian morphology resembled the human ovarian features of testosterone-treated transgender men (partially impaired folliculogenesis, hypertrophic or luteinized stromal cells, atretic follicles, and collapsed clusters). We additionally assessed TC17 fertility denoting a perturbation of the normal reproductive functions (e.g., low pregnancy rate and numbers of pups per litter). Finally, RNAseq analysis permitted us to identify dysregulated genes (Lhcgr, Fshr, Runx1) and pathways (Extra Cellular Matrix and Steroid Synthesis). CONCLUSIONS: Our novel mouse model is a versatile tool to provide innovative insights into study the effects of Cyp17 overexpression and hyperandrogenism in the ovary.


Assuntos
Síndrome do Ovário Policístico , Células Tecais , Androgênios/farmacologia , Animais , Família 17 do Citocromo P450 , Feminino , Humanos , Masculino , Camundongos , Fenótipo , Esteroide 17-alfa-Hidroxilase/genética
3.
J Assist Reprod Genet ; 33(8): 1067-77, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27155601

RESUMO

PURPOSE: Factors that differentially regulate oocyte and granulosa cell growth within the early preantral follicle and how these factors differ at each stage of follicle growth remain poorly understood. The aim of this study was to isolate and evaluate the effect of recombinant growth and differentiation factor 9 (GDF9) on oocyte and granulosa cell growth at the primary and early secondary stages of preantral follicle growth during in vitro culture. METHODS: Primary stage follicles (diameters of 50-89 µm) and early secondary stage follicles (diameters of 90-120 µm) were isolated from immature mice, and individual, intact follicles were cultured in vitro in the presence and absence of recombinant GDF9. The effects of GDF9 on follicle growth were determined by the assessment of changes in the follicle volume during culture. The growth of the granulosa cell and oocyte compartments of the follicles was evaluated separately at each stage. RESULTS: GDF9 significantly increased the growth of isolated follicles at both the primary and early secondary follicle stages. Independent evaluation of the granulosa cell and oocyte compartments revealed that, while GDF9 promoted granulosa cell growth at both stages of folliculogenesis, oocyte growth was stage specific. GDF9 promoted growth of the oocyte at the primary, but not the early secondary, follicle stage. CONCLUSIONS: These findings demonstrate a stage-specific role for GDF9 in the regulation of oocyte and granulosa cell growth at the primary and early secondary stages of preantral follicle development.


Assuntos
Células da Granulosa/metabolismo , Fator 9 de Diferenciação de Crescimento/farmacologia , Oócitos/crescimento & desenvolvimento , Oogênese/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , Animais , Feminino , Células da Granulosa/citologia , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/citologia , Oogênese/fisiologia , Técnicas de Cultura de Órgãos , Folículo Ovariano/citologia
4.
Biol Reprod ; 93(3): 69, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26203175

RESUMO

Polycystic ovary syndrome (PCOS) pathophysiology is poorly understood, due partly to lack of PCOS animal models fully recapitulating this complex disorder. Recently, a PCOS rat model using letrozole (LET), a nonsteroidal aromatase inhibitor, mimicked multiple PCOS phenotypes, including metabolic features absent in other models. Given the advantages of using genetic and transgenic mouse models, we investigated whether LET produces a similar PCOS phenotype in mice. Pubertal female C57BL/6N mice were treated for 5 wk with LET, which resulted in increased serum testosterone and normal diestrus levels of estradiol, similar to the hyperandrogenemia and follicular phase estrogen levels of PCOS women. As in PCOS, ovaries from LET mice were larger, polycystic, and lacked corpora lutea versus controls. Most LET females were acyclic, and all were infertile. LET females displayed elevated serum LH levels and higher Lhb mRNA in the pituitary. In contrast, serum FSH and Fshb were significantly reduced in LET females, demonstrating differential effects on gonadotropins, as in PCOS. Within the ovary, LET females had higher Cyp17, Cyp19, and Fsh receptor mRNA expression. In the hypothalamus, LET females had higher kisspeptin receptor mRNA expression but lower progesterone receptor mRNA levels. LET females also gained more weight than controls, had increased abdominal adiposity and adipocyte size, elevated adipose inflammatory mRNA levels, and impaired glucose tolerance, mirroring the metabolic phenotype in PCOS women. This is the first report of a LET paradigm in mice that recapitulates both reproductive and metabolic PCOS phenotypes and will be useful to genetically probe the PCOS condition.


Assuntos
Inibidores Enzimáticos/toxicidade , Nitrilas/toxicidade , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/patologia , Reprodução/efeitos dos fármacos , Triazóis/toxicidade , Animais , Corpo Lúteo/metabolismo , Diestro/metabolismo , Ciclo Estral/efeitos dos fármacos , Feminino , Hiperandrogenismo/sangue , Hiperandrogenismo/induzido quimicamente , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Kisspeptinas/biossíntese , Kisspeptinas/genética , Letrozol , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Síndrome do Ovário Policístico/metabolismo , Gravidez , Testosterona/sangue
5.
Biochem Biophys Res Commun ; 434(2): 401-6, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23583200

RESUMO

Estrogen is known to play a pivotal role in granulosa cell responses to follicle-stimulating hormone (FSH) that is critical for the establishment of dominant follicles and subsequent ovulation in mammals. Thus, elucidating the cellular and molecular mechanisms that regulate FSH activity is important to understand female fertility. We previously discovered that the oocyte is required for estrogen to exert its positive effects on FSH activity in rat granulosa cells. This finding supports the new concept that estrogen action in granulosa cells is mediated by the oocyte. In the current study, we explored the underlying mechanism. In the presence of oocytes, estrogens enhanced FSH-induced increases in aromatase, steroidogenic acute regulatory protein and FSH receptor mRNA expression as well as cAMP production. However, as forskolin did not mimic FSH activity this indicated that coexistence of estrogen/oocytes increases FSH activity at a site upstream of adenylate cyclase in granulosa cells. We therefore sought a possible involvement of the autoregulatory molecules for FSH receptor, G protein-coupled receptor kinases (GRKs) and ß-arrestins in enhancing FSH activity in response to the estrogen/oocyte co-treatment in granulosa cells. Among the seven known GRK and two ß-arrestin molecules, we found that estrogens with oocytes suppressed FSH-induced GRK-6 mRNA expression. Consistent with this finding, transfecting granulosa cells with small interfering RNA of GRK-6 significantly increased FSH induction of aromatase mRNA, suggesting that endogenous GRK-6 plays an inhibitory role in FSH-induced aromatase mRNA expression. Consequently, these findings strongly suggest that GRK-6 is involved in the mechanism by which estrogen and oocytes synergistically augment FSH activity in granulosa cells.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Quinases de Receptores Acoplados a Proteína G/metabolismo , Células da Granulosa/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Animais , Aromatase/genética , Aromatase/metabolismo , Células Cultivadas , Técnicas de Cocultura , AMP Cíclico/biossíntese , Dietilestilbestrol/farmacologia , Sinergismo Farmacológico , Estrogênios/farmacologia , Feminino , Hormônio Foliculoestimulante/metabolismo , Quinases de Receptores Acoplados a Proteína G/genética , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Oócitos/citologia , Oócitos/metabolismo , Cultura Primária de Células , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do FSH/genética , Receptores do FSH/metabolismo , Transfecção
6.
Mol Reprod Dev ; 78(1): 9-21, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21226076

RESUMO

The oocyte plays an important role in regulating and promoting follicle growth, and thereby its own development, by the production of oocyte growth factors that predominantly act on supporting granulosa cells via paracrine signaling. Genetic studies in mice demonstrated critical roles of two key oocyte-derived growth factors belonging to the transforming growth factor-ß (TGF-ß) superfamily, growth and differentiation factor-9 (GDF-9) and bone morphogenetic protein-15 (BMP-15), in ovarian function. The identification of Bmp15 and Gdf9 gene mutations as the causal mechanism underlying the highly prolific or infertile nature of several sheep strains in a dosage-sensitive manner also highlighted the crucial role these two genes play in ovarian function. Similarly, large numbers of mutations in the GDF9 and BMP15 genes have been identified in women with premature ovarian failure and in mothers of dizygotic twins. The purpose of this article is to review the genetic studies of GDF-9 and BMP-15 mutations identified in women and sheep, as well as describing the various knockout and overexpressing mouse models, and to summarize the molecular and biological functions that underlie the crucial role of these two oocyte factors in female fertility.


Assuntos
Proteína Morfogenética Óssea 15/metabolismo , Fertilidade , Fator 9 de Diferenciação de Crescimento/metabolismo , Insuficiência Ovariana Primária/metabolismo , Animais , Proteína Morfogenética Óssea 15/genética , Modelos Animais de Doenças , Feminino , Células da Granulosa/metabolismo , Fator 9 de Diferenciação de Crescimento/genética , Humanos , Camundongos , Mutação , Oócitos/metabolismo , Insuficiência Ovariana Primária/genética , Ovinos
7.
J Endocr Soc ; 3(11): 2064-2081, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31701078

RESUMO

Adult granulosa cell tumor (aGCT) is a rare type of ovarian cancer characterized by estrogen excess. Interestingly, only the single somatic mutation FOXL2 C134W was found across virtually all aGCTs. We previously reported that FOXL2C134W stimulates CYP19 transcription synergistically with SMAD3, leading to elevated estradiol synthesis in a human granulosa cell line (HGrC1). This finding suggested a key role for FOXL2C134W in causing the typical estrogen overload in patients with aGCTs. We have now investigated the effect of FOXO1, a tumor suppressor, on CYP19 activation by FOXL2C134W in the presence of SMAD3. Intriguingly, FOXO1 antagonized the positive, synergistic effect of FOXL2C134W and SMAD3 on CYP19 transcription. Similar to FOXL2C134W, FOXO1 binds SMAD3 but not the proximal FOXL2C134W binding site (-199 bp) of the CYP19 promoter identified in our earlier studies. The results of a competitive binding assay suggested a possible underlying mechanism in which FOXO1 sequesters SMAD3 away from FOXL2C134W, thereby negating the cooperative action of FOXL2C134W and SMAD3 in inducing CYP19 expression. To our knowledge, this study is the first to demonstrate the ability of FOXO1 to restore an altered CYP19 expression by FOXL2C134W and SMAD3 and provides insight as to why FOXO1 deficiency promotes GCT development in mice.

8.
Protein Sci ; 17(2): 362-70, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18227435

RESUMO

Bone morphogenetic protein-15 (BMP-15) is an oocyte-secreted factor critical for the regulation of ovarian physiology. When recombinant human BMP-15 (rhBMP-15) produced in human embryonic kidney 293 cells was subjected to SDS-PAGE analysis, two mature protein forms corresponding to 16 kDa (P16) and 17 kDa (P17) were observed. Despite the physiological relevance and critical function of BMP-15 in female reproduction, little is known about the structure of rhBMP-15. Here, we have analyzed the structure of the rhBMP-15 mature proteins (P16 and P17) using state-of-the-art proteomics technology. Our findings are as follows: (1) the N-terminal amino acid of P16 and P17 is pyroglutamic acid; (2) the Ser residue at the sixth position of P16 is phosphorylated; (3) P17 is O-glycosylated at Thr10; and (4) the C-terminal amino acid of P16 and P17 is truncated. These findings are the first knowledge of the structure of rhBMP-15 mature protein toward understanding the molecular basis of BMP-15 function and could provide an important contribution to the rapidly progressing research area involving oocyte-specific growth factors in modulation of female fertility.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Sequência de Aminoácidos , Animais , Proteína Morfogenética Óssea 15 , Glicosilação , Fator 9 de Diferenciação de Crescimento , Humanos , Dados de Sequência Molecular , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
9.
Endocrinology ; 149(2): 812-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18006624

RESUMO

Two highly homologous oocyte-secreted growth factors, bone morphogenetic protein (BMP)-15 and growth and differentiation factor (GDF)-9, are known to control folliculogenesis and ovulation through direct effects on granulosa cells in the developing follicles. Although much is known about the expression and biology of these proteins, the impact of posttranslational modifications of BMP-15 and GDF-9 is unknown. Here, we report that: 1) recombinant human (rh) BMP-15 and rhGDF-9 are phosphorylated; 2) the phosphorylation is essential for bioactivity; and 3) the dephosphorylated forms of rhBMP-15 and rhGDF-9 can abolish the bioactivity of rhBMP-15, rhGDF-9, and rhBMP-7, but not rh activin A. These results indicate that the phosphorylation state of rhBMP-15 and rhGDF-9 is a determinant of their agonistic and antagonistic activities.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Fertilidade/fisiologia , Células da Granulosa/citologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteína Morfogenética Óssea 15 , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/farmacologia , Células Cultivadas , Feminino , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/fisiologia , Fator 9 de Diferenciação de Crescimento , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Rim/citologia , Folículo Ovariano/citologia , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/fisiologia , Fosforilação , Ratos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Smad/metabolismo , Timidina/farmacocinética , Fator de Crescimento Transformador beta/farmacologia , Trítio
10.
Endocrinology ; 149(6): 2807-15, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18308851

RESUMO

Whereas mutations in the bmp15 gene cause infertility in ewes and women due to defects in folliculogenesis, most defects in female mice lacking bone morphogenetic protein (BMP)-15 are confined to the ovulation process, supportive of the observation that functional mouse BMP-15 is barely detected in oocytes in vivo until after the LH surge. In addition, the mouse BMP-15 proprotein is not processed into the functional mature protein in transfected cells. However, a chimeric protein consisting of the human proregion, human cleavage site, and mouse mature region (termed hhmBMP-15) is processed and the mature protein secreted. To study the role of BMP-15 in folliculogenesis, we generated transgenic mice overexpressing hhmBMP-15, exclusively in oocytes during folliculogenesis and confirmed the overexpression of mouse BMP-15 mature protein. Immature transgenic mice exhibited accelerated follicle growth with decreased primary follicles and an increase in secondary follicles. Granulosa cells of immature mice displayed an increased mitotic index and decreased FSH receptor mRNA expression. Adult mice had normal litter sizes but an increased number of atretic antral follicles. Interestingly, aging mice exhibited an early onset of acyclicity marked by increased diestrus length and early occurrence of constant diestrus. These findings indicate the role of BMP-15 in vivo in promoting follicle growth and preventing follicle maturation, resulting in an early decline in the ovarian reserve of transgenic mice. Therefore, the lack of mouse BMP-15 during early folliculogenesis in the wild-type mice may be relevant to their polyovulatory nature as well as the preservation of ovarian function as the mice age.


Assuntos
Estro , Fertilidade/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Oócitos/fisiologia , Folículo Ovariano/fisiologia , Ovulação/fisiologia , Animais , Proteína Morfogenética Óssea 15 , Feminino , Regulação da Expressão Gênica , Fator 9 de Diferenciação de Crescimento , Humanos , Camundongos , Camundongos Transgênicos , Ovulação/genética , Reação em Cadeia da Polimerase
11.
J Clin Endocrinol Metab ; 93(5): 1827-33, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18285408

RESUMO

CONTEXT: In women with polycystic ovary syndrome (PCOS), excess ovarian androgen production is driven by increased LH secretion. Studies conducted in animals suggest that the granulosa cell may influence LH-stimulated theca cell androgen production. OBJECTIVE: The objective of this study was to determine whether FSH enhances androgen production in women with PCOS compared with that of normal women. DESIGN: A prospective study was conducted to compare androgen production in response to FSH in two groups of women. SETTING: The study was conducted in a General Clinical Research Center in a tertiary academic medical center. PATIENTS: Women with PCOS, 18-35 yr (n = 20), and normal ovulatory controls, 18-35 yr (n = 10), were recruited for study. INTERVENTIONS: Serial blood samples were obtained over a 24-h period after an iv injection of recombinant human FSH (150 IU). MAIN OUTCOME MEASURES: The main outcome measures were serum 17-hydroxyprogesterone (17-OHP), androstenedione (A), dehydroepiandrosterone (DHEA), testosterone (T), and inhibin B (Inh B) responses after FSH administration. RESULTS: Basal serum 17-OHP, A, and T levels were markedly increased in women with PCOS compared with that observed in normal women. Basal DHEA and Inh B levels were similar to those of normal controls. After FSH injection, PCOS women demonstrated enhanced production of 17-OHP, A, DHEA, and Inh B, whereas in normal women no increases were observed. T levels declined slightly in both groups. CONCLUSIONS: These findings provide evidence that, in PCOS women, theca cell androgen production is enhanced by FSH administration and suggest a granulosa-theca cell paracrine mechanism.


Assuntos
Androgênios/biossíntese , Hormônio Foliculoestimulante/farmacologia , Síndrome do Ovário Policístico/sangue , 17-alfa-Hidroxiprogesterona/sangue , Adolescente , Adulto , Androstenodiona/sangue , Desidroepiandrosterona/sangue , Estradiol/sangue , Feminino , Humanos , Hidrocortisona/sangue , Inibinas/sangue , Hormônio Luteinizante/sangue , Estudos Prospectivos , Testosterona/sangue
12.
Endocr Rev ; 25(1): 72-101, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14769828

RESUMO

Using molecular, cellular, and genetic approaches, recent studies examining the role of the bone morphogenetic protein (BMP) family of growth factors in the reproductive system have led to significant breakthroughs in our understanding of mammalian reproduction and fertility. Gene expression studies have revealed that key components of the BMP system (ligands, receptors, signaling molecules, and binding proteins) exhibit coordinated spatial and temporal expression patterns in fundamental cell types throughout the reproductive system. Availability of recombinant BMPs has enabled functional studies that have demonstrated important biological activities of BMPs in controlling cellular proliferation, differentiation, and apoptosis in reproductive tissues. The physiological importance of the BMP system for mammalian reproduction has been further highlighted by the elucidation of the aberrant reproductive phenotypes of animals with naturally occurring mutations or targeted deletions of certain BMP family genes. Collectively, these studies have established the concept that the BMP system plays a crucial role in fertility in female and male mammals. The purpose of this article is to review the evidence underpinning the importance of the BMP system in mammalian reproduction.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Reprodução/fisiologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular , Divisão Celular , Feminino , Fertilidade/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Transgênicos
13.
Vitam Horm ; 107: 317-348, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29544636

RESUMO

Growth and differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are oocyte-secreted factors with a leading role in the control of ovarian function in female reproduction, modulating both the cell fate of the somatic granulosa cells and the quality and developmental competence of the egg. This short review aims to consolidate the molecular aspects of GDF9 and BMP15 and their integral actions in female fertility to understand particularly their effects on oocyte quality and fetal growth. The significant consequences of mutations in the GDF9 and BMP15 genes in women with dizygotic twins as well as the clinical relevance of these oocyte factors in the pathogenesis of primary ovarian insufficiency and polycystic ovary syndrome are also addressed.


Assuntos
Proteína Morfogenética Óssea 15/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fator 9 de Diferenciação de Crescimento/metabolismo , Ciclo Menstrual/metabolismo , Oócitos/metabolismo , Oogênese , Ovário/fisiologia , Animais , Proteína Morfogenética Óssea 15/química , Feminino , Predisposição Genética para Doença , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Fator 9 de Diferenciação de Crescimento/química , Humanos , Mutação , Oócitos/citologia , Oócitos/patologia , Ovário/citologia , Ovário/patologia , Ovário/fisiopatologia , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Síndrome do Ovário Policístico/fisiopatologia , Gravidez , Gravidez de Gêmeos/genética , Gravidez de Gêmeos/metabolismo , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/fisiopatologia , Conformação Proteica , Especificidade da Espécie , Gêmeos Dizigóticos
14.
Endocrinology ; 159(4): 1690-1703, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29471425

RESUMO

Germline knockout studies in female mice demonstrated an essential role for forkhead box L2 (FOXL2) in early follicle development, whereas an inducible granulosa cell (GC)-specific deletion of Foxl2 in adults has shown ovary-to-testis somatic sex reprogramming. In women, over 120 different germline mutations in the FOXL2 gene have been shown to cause blepharophimosis/ptosis/epicantus inversus syndrome associated with or without primary ovarian insufficiency. By contrast, a single somatic mutation (FOXL2C134W) accounts for almost all adult-type GC tumors (aGCTs). To test the hypothesis that FOXL2C134W differentially regulates the expression of aGCT markers, we investigated the effect of FOXL2C134W on inhibin B and P450 aromatase expression using a recently established human GC line (HGrC1), which we now show to bear two normal alleles of FOXL2. Neither FOXL2wt nor FOXL2C134W regulate INHBB messenger RNA (mRNA) expression. However, FOXL2C134W selectively displays a 50-fold induction of CYP19 mRNA expression dependent upon activin A. Mechanistically, the CYP19 promoter is activated in a similar way by FOXL2C134W interaction with SMAD3, but not by FOXL2wt. SMAD2 had no effect. Moreover, FOXL2C134W interactions with SMAD3 and with the FOX binding element located at -199 bp upstream of the ATG initiation codon of CYP19 are more sustainable than FOXL2wt. Thus, FOXL2C134W potentiates CYP19 expression in HGrC1 cells via enhanced recruitment of SMAD3 to a proximal FOX binding element. These findings may explain the pathophysiology of estrogen excess in patients with aGCT.


Assuntos
Aromatase/metabolismo , Proteína Forkhead Box L2/metabolismo , Regulação da Expressão Gênica , Inibinas/metabolismo , Proteína Smad3/metabolismo , Aromatase/genética , Linhagem Celular , Feminino , Proteína Forkhead Box L2/genética , Células da Granulosa/metabolismo , Humanos , Inibinas/genética , Regiões Promotoras Genéticas , Proteína Smad3/genética , Transcrição Gênica
15.
Am J Reprod Immunol ; 78(1)2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28337819

RESUMO

PROBLEM: Plasminogen activator inhibitor-1 (PAI-1) is elevated in women with polycystic ovary syndrome (PCOS), but the regulation in granulosa cells (GCs) is unclear. METHOD OF STUDY: PAI-1 expression in PCOS ovaries was investigated immunohistologically. PAI-1 expressions in HGrC1, a human GC cell line, were investigated at mRNA and activity levels. The expressions of TGF-ß and TNF-α in peritoneal fluid mononuclear cells (PFMCs) were measured with quantitative PCR. RESULTS: Little PAI-1 expression is observed in healthy GCs, whereas GCs of PCOS and atretic follicle exhibit distinct expression in vivo. In vitro study using HGrC1 shows that TGF-ß and TNF-α increase PAI-1 mRNA and its activity, and both together exhibit a synergistic effect. The expression of PAI-1 mRNA is suppressed by simvastatin. Moreover, insulin-sensitizing drugs (metformin, pioglitazone, and rosiglitazone) suppress LPS-induced TGF-ß and TNF-α mRNA expression in PFMC. CONCLUSION: Statin and insulin-sensitizing drugs may provide a potential therapy for PCOS via down-regulation of PAI-1 expression in GCs and down-regulation of TGF-ß and TNF-α expression in PFMC, respectively.


Assuntos
Células da Granulosa/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hipoglicemiantes/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Necrose Tumoral alfa/genética , Líquido Ascítico/citologia , Linhagem Celular , Feminino , Células da Granulosa/metabolismo , Humanos , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos , Folículo Ovariano/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Síndrome do Ovário Policístico/metabolismo , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Sinvastatina/farmacologia
16.
Sci Rep ; 7(1): 10824, 2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28883502

RESUMO

Recent studies report the involvement of intra-ovarian factors, such as inflammation and oxidative stress, in the pathophysiology of polycystic ovary syndrome (PCOS), the most common endocrine disorder of reproductive age women. Endoplasmic reticulum (ER) stress is a local factor that affects various cellular events during a broad spectrum of physiological and pathological conditions. It may also be an important determinant of pro-fibrotic remodeling during tissue fibrosis. In the present study, we showed that ER stress was activated in granulosa cells of PCOS patients as well as in a well-established PCOS mouse model. Pharmacological inducers of ER stress, tunicamycin and thapsigargin, were found to increase the expression of pro-fibrotic growth factors, including transforming growth factor (TGF)-ß1, in human granulosa cells, and their expression also increased in granulosa cells of PCOS patients. By contrast, treatment of PCOS mice with an ER stress inhibitor, tauroursodeoxycholic acid or BGP-15, decreased interstitial fibrosis and collagen deposition in ovaries, accompanied by a reduction in TGF-ß1 expression in granulosa cells. These findings suggest that ER stress in granulosa cells of women with PCOS contributes to the induction of pro-fibrotic growth factors during ovarian fibrosis, and that ER stress may serve as a therapeutic target in PCOS.


Assuntos
Estresse do Retículo Endoplasmático , Fibrose/fisiopatologia , Células da Granulosa/patologia , Células da Granulosa/fisiologia , Síndrome do Ovário Policístico/patologia , Animais , Células Cultivadas , Colágeno/análise , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Ovário/patologia , Síndrome do Ovário Policístico/complicações , Fator de Crescimento Transformador beta1/metabolismo
17.
Endocrinology ; 147(1): 201-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16210364

RESUMO

Bone morphogenetic protein-15 (BMP-15) is a member of the TGFbeta family known to regulate ovarian functions in mammals. The structure and function of BMP-15 in lower vertebrates are less known. In this study, we cloned the zebrafish BMP-15 (zfBMP-15) cDNA and depicted its genomic organization. The zfBMP-15 cDNA encodes a protein of 384 amino acids. The mature protein has 46-51% sequence identities to fugu, chicken, and mammalian BMP-15. It also shares 38-46% homology with growth and differentiation factor-9 in fishes, chicken, and mammals. Phylogenetic analysis further confirms that the zfBMP-15 is most closely related to BMP-15 from other species, whereas the growth and differentiation factor-9 peptides from fish to mammals form a distinct branch. Comparison of zfBMP-15 cDNA with zebrafish genome database revealed that zfBMP-15 is encoded by a gene with two exons and one intron, located on chromosome 6. BMP-15 mRNA is expressed in the ovary and testis and, to a lesser extent, brain, liver, gut, heart, and muscle. Real-time PCR revealed that BMP-15 is expressed in follicles at all stages of development with no significant changes over the course of folliculogenesis. Using in situ hybridization and immunocytochemistry, we detected BMP-15 in both oocytes and follicular cells. Incubation of follicles with antiserum against zfBMP15 increased oocyte maturation, whereas incubation with recombinant human BMP-15 suppressed human chorionic gonadotropin-induced oocyte maturation. These findings suggest that BMP-15 plays a role in regulating gonadal functions in fish, in particular oocyte maturation.


Assuntos
DNA Complementar/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Oócitos/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteína Morfogenética Óssea 15 , Galinhas , Clonagem Molecular , Primers do DNA , Feminino , Fator 9 de Diferenciação de Crescimento , Humanos , Mamíferos , Dados de Sequência Molecular , Especificidade de Órgãos , Filogenia , Reação em Cadeia da Polimerase , Peixe-Zebra/classificação , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
18.
Reprod Med Biol ; 5(4): 245-248, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29699253

RESUMO

Naturally occurring mutations in the oocyte-specific factor, bone morphogenetic protein-15 (BMP-15), cause infertility in women and in ewes. In contrast to these monoovulatory mammals, the targeted deletion of BMP-15 in polyovulatory mice results in subfertility with only minimal defects in the ovulation process. Given the established role of BMP-15 in governing the progression of folliculogenesis, it is hypothesized that species-specific differences in the BMP-15 system are involved in species-specific determination of ovulation quota and litter size. Recent data using in vitro cell transfection methodology indicate that, in contrast to human BMP-15 which is successfully processed and secreted, the mouse BMP-15 proprotein is resistant to proteolytic cleavage. Thus, no functional mature BMP-15 is secreted in vitro. Further studies have shown that the functional mature form of BMP-15 is barely detectable in mouse oocytes in vivo until just before ovulation, when it is markedly increased. The general hypothesis to emerge from these observations is that the species-specific differences in the defects caused by mutations in the bmp15 gene between monoovulatory ewes and women and polyovulatory mice might be attributed to the timing of the production of BMP-15 mature protein. (Reprod Med Biol 2006; 5: 245-248).

19.
Endocrinology ; 146(2): 941-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15539559

RESUMO

Oocyte-granulosa cell communication is essential for oocyte development. The aims of this study were: 1) to determine the effect of FSH on expression of Kit ligand (KL), growth/differentiation factor-9, bone morphogenetic protein (BMP)-15, and Kit during growth of oocyte-granulosa cell complexes (OGCs) in vitro; 2) to investigate the role of BMP-15 in regulation of KL expression; and 3) to correlate mRNA expression with oocyte growth. OGCs from 12-d-old mice were cultured for up to 7 d in the presence of FSH [0.05 ng/ml (low), 5 ng/ml (high)] or BMP-15 (10 or 100 ng/ml). Transcripts were quantified using real-time RT-PCR, and oocyte and OGC diameters were measured. FSH regulated KL expression in a biphasic manner, with low FSH decreasing the KL-1/KL-2 ratio, and high FSH increasing the KL-1/KL-2 ratio, compared with controls (P < 0.05). The decrease in KL-1/KL-2 ratio with low FSH was due to increased KL-2 mRNA expression. Both FSH concentrations increased OGC diameter (P < 0.05), but only low FSH promoted oocyte growth (P < 0.05). High FSH also decreased BMP-15 expression (P < 0.05). FSH-stimulated oocyte growth was inhibited by Gleevec, an inhibitor of Kit activity. BMP-15 increased both KL-1 and KL-2 mRNA levels in a dose-dependent manner (P < 0.05) but did not alter the KL-1/KL-2 ratio or promote oocyte growth. When the KL-1/KL-2 ratio was increased by exogenous KL-1, FSH-stimulated oocyte growth was suppressed (P < 0.05), suggesting that lowered KL-1/KL-2 ratio is important for oocyte growth. In summary, the correct concentration of FSH is crucial for appropriate modulation of KL and BMP-15 to promote oocyte growth.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Oócitos/citologia , Oócitos/fisiologia , Animais , Proteína Morfogenética Óssea 15 , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Feminino , Expressão Gênica/efeitos dos fármacos , Fator 9 de Diferenciação de Crescimento , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Mutantes , Proteínas Oncogênicas/genética , Oócitos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit , RNA Mensageiro/análise , Fator de Células-Tronco/genética
20.
Endocrinology ; 146(8): 3362-7, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15878960

RESUMO

The establishment of dominant ovarian follicles that are capable of ovulating fertilizable oocytes is a fundamental determinant of female fertility. This process is governed by pituitary gonadotropins as well as local ovarian factors. Within the follicle, estrogen acts in an autocrine/paracrine manner to enhance FSH action in the granulosa cells. These effects include the augmentation of P450aromatase expression and estradiol production. This feed-forward effect of estrogen is believed to play a key role in follicle dominance. Here we found the essential role of the oocyte in this physiological process using primary cultures of rat granulosa cells. In the presence, but not absence, of oocytes, estrogen amplified FSH-stimulated increases in mRNA expression of P450aromatase, FSH receptor, LH receptor, and inhibin alpha-, betaA-, and betaB-subunits as well as cAMP production. Thus, oocytes mediate the estrogen enhancement of FSH action in the granulosa cells. In comparison with FSH, cotreatment with estrogen and oocytes failed to amplify the stimulatory effects of forskolin or 8-bromoadenosine-cAMP on granulosa cell responses including P450aromatase mRNA expression and cAMP production, indicating that estrogen/oocytes amplify FSH action at a site upstream of adenylate cyclase. These findings support the novel conclusion that communication between the oocyte and granulosa cells plays a crucial role in mediating estrogen action during FSH-dependent folliculogenesis.


Assuntos
Dietilestilbestrol/farmacologia , Estrogênios/farmacologia , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/fisiologia , Oócitos/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , AMP Cíclico/metabolismo , Feminino , Células da Granulosa/efeitos dos fármacos , Cinética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA