Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.021
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Cell Physiol ; 236(9): 6619-6629, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33512728

RESUMO

Estradiol (E2), one of the main steroid hormones secreted by the ovaries, plays an important role in maintaining normal female reproductive function. Ovarian granulosa cells are the main source of E2 production because these cells express aromatase, which is encoded by the CYP19A1 gene and catalyzes the final step in E2 biosynthesis from androgens. Transforming growth factor-beta 1 (TGF-ß1) and its receptors are expressed in human granulosa cells, and TGF-ß1 expression can be detected in human follicular fluid. To date, TGF-ß1 has been shown to regulate various ovarian functions. However, whether aromatase can be regulated by TGF-ß1 in human granulosa cells has not been determined. In the present study, we demonstrate that TGF-ß1 stimulates aromatase expression in primary human granulosa-lutein cells and in the human ovarian granulose-like tumor cell line, KGN. We used pharmacological inhibitors and small interfering RNA-mediated knockdown approaches to reveal that the SMAD2 and ERK1/2 signaling pathways are involved in TGF-ß1-induced aromatase expression and E2 production. These results not only provide important insights into the molecular mechanisms that mediate TGF-ß1-induced aromatase expression and E2 production in human granulosa cells but also increase the understanding of the normal physiological roles of TGF-ß1 in the ovary.


Assuntos
Aromatase/metabolismo , Estradiol/biossíntese , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Feminino , Humanos , Modelos Biológicos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo
2.
Cell Biol Int ; 45(1): 127-139, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32997425

RESUMO

Follicular fluid (FF) is essential for developing ovarian follicles. Besides the oocytes, FF has abundant undifferentiated somatic cells containing stem cell properties, which are discarded in daily medical procedures. Earlier studies have shown that FF cells could differentiate into primordial germ cells via forming embryoid bodies, which produced oocyte-like cells (OLC). This study aimed at isolating mesenchymal stem cells (MSC) from FF and evaluating the impacts of bone morphogenetic protein 15 (BMP15) on the differentiation of these cells into OLCs. Human FF-derived cells were collected from 78 women in the assisted fertilization program and cultured in human recombinant BMP15 medium for 21 days. Real-time polymerase chain reaction and immunocytochemistry staining characterized MSCs and OLCs. MSCs expressed germline stem cell (GSC) markers, such as OCT4 and Nanog. In the control group, after 15 days, OLCs were formed and expressed zona pellucida markers (ZP2 and ZP3), and reached 20-30 µm in diameter. Ten days after induction with BMP15, round cells developed, and the size of OLCs reached 115 µm. A decrease ranged from 0.04 to 4.5 in the expression of pluripotency and oocyte-specific markers observed in the cells cultured in a BMP15-supplemented medium. FF-derived MSCs have an innate potency to differentiate into OLCs, and BMP15 is effective in promoting the differentiation of these cells, which may give an in vitro model to examine germ cell development.


Assuntos
Proteína Morfogenética Óssea 15/farmacologia , Diferenciação Celular/efeitos dos fármacos , Líquido Folicular/citologia , Células-Tronco Mesenquimais/citologia , Oócitos/citologia , Adipogenia/efeitos dos fármacos , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 15/metabolismo , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Estradiol/biossíntese , Feminino , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Oócitos/metabolismo
3.
Reprod Fertil Dev ; 33(5): 363-371, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33641714

RESUMO

MicroRNAs (miRNAs) have been determined to participate in the process of oestradiol production. Generally, there are two pathways by which oestradiol levels change, one being the state of cells (i.e. the status of enzymes involved in the synthesis of hormones such as oestradiol) and the other being the number of cells that secrete oestradiol. It is known that oestrogens are the main steroids produced by granulosa cells (GCs) of mature ovarian follicles. In this study we explored the function of miR-18b in rabbit GCs by overexpressing or inhibiting its activity. We found that miR-18b silencing promoted the secretion of oestradiol by significantly affecting the expression of steroidogenesis-related genes. Thus, miR-18b may act as a negative regulator of the production of enzymes related to oestradiol synthesis and affect oestradiol production. Furthermore, the effects of miR-18b on the proliferation, cell cycle and apoptosis of GCs were investigated using a cell counting kit (CCK-8) proliferation assay, detection of annexin V-fluorescein isothiocyanate apoptosis, flow cytometry and quantitative polymerase chain reaction. The results showed that miR-18b upregulated GC apoptosis (miR-18b overexpression decreases cell growth and stimulates apoptosis). These findings suggest that miR-18b and the oestrogen receptor 1 (ESR1) gene may be attractive targets to further explore the molecular regulation of GCs. The miR-18b may also explain, in part, the abnormal folliculogenesis in mammals caused by conditions such as polycystic ovary syndrome, primary ovarian insufficiency, and others.


Assuntos
Células da Granulosa/fisiologia , MicroRNAs/fisiologia , Animais , Apoptose , Ciclo Celular , Proliferação de Células , Células Cultivadas , Epigênese Genética/genética , Estradiol/biossíntese , Estradiol/genética , Receptor alfa de Estrogênio/genética , Feminino , Expressão Gênica , Inativação Gênica , MicroRNAs/genética , Coelhos , Transfecção
4.
Addict Biol ; 26(2): e12918, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32476209

RESUMO

Despite advances in prevention and treatment, cigarette smoking remains a leading cause of preventable death in the United States. Although men and women are equally likely to attempt to quit smoking cigarettes, women are far less likely to achieve abstinence both during and following cessation treatment. Recent evidence suggests that ovarian hormone levels may play a role in successful abstinence attempts in women smokers. The primary goal of this exploratory prospective observational study was to estimate the association between within-participant levels of progesterone and estradiol with associated cigarettes smoked per day in adult women smokers (n = 104). The primary study outcome was self-reported cigarettes smoked per day (CPD) during a 2-week observational period collected using a daily smoking diary. Additionally, participants collected saliva daily, from which hormone levels (progesterone and estradiol) were derived. Higher within-participant progesterone levels were associated with a significant decrease in CPD (p = .008) whereas within-participant estradiol levels were unrelated to CPD (p = .25). Regression models indicated a single change in the trajectory of smoking behavior for both within-participant progesterone and estradiol. When progesterone values were below the change point, there was a significant inverse relationship between within-participant progesterone levels and smoking behavior (p = .025) whereas the relationship was attenuated for higher within-participant progesterone levels (p = .59). The effect of estradiol on smoking behavior was not significant when it was either below (p = .92) or above (p = .16) the change point. Higher within-participant levels of progesterone but not estradiol are associated with reduced CPD in nontreatment seeking women smokers.


Assuntos
Estradiol/análise , Progesterona/análise , Saliva/química , Fumar Tabaco/epidemiologia , Adolescente , Adulto , Estradiol/biossíntese , Feminino , Humanos , Pessoa de Meia-Idade , Progesterona/biossíntese , Estudos Prospectivos , Estados Unidos/epidemiologia , Adulto Jovem
5.
Environ Toxicol ; 36(4): 665-674, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33258555

RESUMO

Numerous studies have shown that endocrine-disrupting chemicals are one of the important pathogenic factors in women with polycystic ovary syndrome. Our previous study has revealed that bisphenol A (BPA) can cause steroid hormone imbalance, polycystic ovary, and estrus cycle disorder. In this study, we aimed to explore the effect of BPA, a typical environmental estrogen, on the synthesis of steroid hormones in human ovarian granulosa KGN cells. Exposure of KGN cells to BPA (0.5, 5, 50, and 500 µg/L) resulted in the decrease of progesterone (P), estradiol (E2), and the ratio of estradiol to testosterone (E2/T). BPA affected the expression of genes related to steroid hormone synthesis in KGN cells, including the decreased expression of the steroidogenic acute regulatory protein, ferredoxin, and ferredoxin reductase genes during progesterone synthesis; upregulating the expression of cytochrome p450 oxidoreductase gene associated with E2 and T synthesis; and the downregulated cytochrome P450 family 1 subfamily A member 1 and cytochrome P450 family 1 subfamily B member 1 in E2 degradation. BPA also reduced the expression of stimulatory G proteins (GS) in follicle-stimulating hormone receptor (FSHR)/GS/adenylate cyclase (AC) signaling pathway. In summary, our research has demonstrated that environment-relevant level of BPA exposure leads to steroid hormone synthesis disorder in human ovarian granulosa cells, which might cause the reduction of gene expression in hormone synthesis and the suppression of the FSHR/GS/AC signaling pathway.


Assuntos
Compostos Benzidrílicos/toxicidade , Disruptores Endócrinos/toxicidade , Estradiol/biossíntese , Células da Granulosa/efeitos dos fármacos , Fenóis/toxicidade , Progesterona/biossíntese , Testosterona/biossíntese , Animais , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Relação Dose-Resposta a Droga , Regulação para Baixo , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Regulação para Cima
6.
J Neuroinflammation ; 17(1): 37, 2020 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-31992325

RESUMO

BACKGROUND: Tibolone is a synthetic steroid used in clinical practice for the treatment of climacteric symptoms and osteoporosis. Active metabolites of tibolone, generated in target tissues, have an affinity for estrogen and androgen receptors. Astrocytes are direct targets for estrogenic compounds and previous studies have shown that tibolone protects brain cortical neurons in association with a reduction in reactive astrogliosis in a mouse model of traumatic brain injury. Since phagocytosis is a crucial component of the neuroprotective function exerted by astrocytes, in the present study, we have assessed whether tibolone regulates phagocytosis in primary astrocytes incubated with brain-derived cellular debris. METHODS: Male and female astrocyte cell cultures were obtained from newborn (P0-P2) female and male Wistar rats. Astrocytic phagocytosis was first characterized using carboxylate beads, Escherichia coli particles, or brain-derived cellular debris. Then, the effect of tibolone on the phagocytosis of Cy3-conjugated cellular debris was quantified by measuring the intensity of Cy3 dye-emitted fluorescence in a given GFAP immunoreactive area. Before the phagocytosis assays, astrocytes were incubated with tibolone in the presence or absence of estrogen or androgen receptor antagonists or an inhibitor of the enzyme that synthesizes estradiol. The effect of tibolone on phagocytosis was analyzed under basal conditions and after inflammatory stimulation with lipopolysaccharide. RESULTS: Tibolone stimulated phagocytosis of brain-derived cellular debris by male and female astrocytes, with the effect being more pronounced in females. The effect of tibolone in female astrocytes was blocked by a selective estrogen receptor ß antagonist and by an androgen receptor antagonist. None of these antagonists affected tibolone-induced phagocytosis in male astrocytes. In addition, the inhibition of estradiol synthesis in the cultures enhanced the stimulatory effect of tibolone on phagocytosis in male astrocytes but blocked the effect of the steroid in female cells under basal conditions. However, after inflammatory stimulation, the inhibition of estradiol synthesis highly potentiated the stimulation of phagocytosis by tibolone, particularly in female astrocytes. CONCLUSIONS: Tibolone exerts sex-specific regulation of phagocytosis in astrocytes of both sexes, both under basal conditions and after inflammatory stimulation.


Assuntos
Astrócitos/efeitos dos fármacos , Inflamação/patologia , Norpregnenos/farmacologia , Fagocitose/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Antagonistas de Receptores de Andrógenos/farmacologia , Animais , Estradiol/biossíntese , Antagonistas de Estrogênios/farmacologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Inflamação/induzido quimicamente , Lipopolissacarídeos , Masculino , Microglia/efeitos dos fármacos , Ratos , Ratos Wistar
7.
Neurobiol Learn Mem ; 175: 107300, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32882397

RESUMO

BACKGROUND: Hippocampal aromatase is responsible for local synthesis of 17ß-estradiol (E2) that has much higher concentrations than serum levels in males and females. Letrozole, an aromatase inhibitor, passes through the brain barriers, distributes to the brain, and affects local E2 synthesis. Here, the effects of intra-cerebroventricular (ICV) letrozole administration in the presence and absence of gonads were examined on the cognitive abilities of male and female rats. METHOD: Animals received intra-ICV injection of letrozole or vehicle for 14 consecutive days. Spatial working memory, novel object recognition memory, and anxiety-related behavior, were evaluated using Y-maze, object recognition test, and elevated plus maze, respectively. The E2 levels in the serum and hippocampal tissue were measured by the ELISA technique. RT-PCR was performed to assess the hippocampal estrogen receptors (ER) expression. Moreover, letrozole effect on neuronal activity of CA1 pyramidal neurons was studied by in vivo single-unit recording. RESULTS: Letrozole (0.2, 0.4, and 0.8 µg) significantly decreased the hippocampal E2 levels compared to the vehicle group. Letrozole caused cognitive impairments in a dose-dependent manner in male and female rats in the presence or absence of gonads. Dose-response analysis revealed that the minimum effective dose of letrozole on the behavioral measures was 0.4 µg. Letrozole also caused an up-regulation of ERα and ERß and a down-regulation of GPR30 gene expression. The firing rate of pyramidal neurons was reduced by letrozole in gonadal-intact animals. CONCLUSION: The detrimental effects of letrozole treatment on cognitive abilities in the presence and absence of gonads indicate that local E2 synthesis in the hippocampus is a crucial factor in normal cognitive performance. The suppressive effect of letrozole on hippocampal neuronal firing might alter synaptic plasticity that is critical for memory formation. These data potentially suggest that memory deficits following letrozole administration should be monitored.


Assuntos
Inibidores da Aromatase/farmacologia , Cognição/efeitos dos fármacos , Estradiol/biossíntese , Hipocampo/efeitos dos fármacos , Letrozol/farmacologia , Células Piramidais/efeitos dos fármacos , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Disfunção Cognitiva , Teste de Labirinto em Cruz Elevado , Estradiol/sangue , Estradiol/metabolismo , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/genética , Feminino , Hipocampo/metabolismo , Injeções Intraventriculares , Masculino , Teste de Campo Aberto , Orquiectomia , Ovariectomia , Células Piramidais/metabolismo , Ratos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Análise de Célula Única
8.
Reproduction ; 160(6): 955-967, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33112769

RESUMO

Luteinization is the event of corpus luteum formation, a way of follicle cells transformation and a process of steroidogenesis alteration. As the core clock gene, Bmal1 was involved in the regulation of ovulation process and luteal function afterwards. Till now, the underlying roles of luteinization played by Bmal1 remain unknown. To explore the unique role of Bmal1 in luteal steroidogenesis and its underlying pathway, we investigated the luteal hormone synthesis profile in Bmal1 knockout female mice. We found that luteal hormone synthesis was notably impaired, and phosphorylation of PI3K/NfκB pathway was significantly activated. Then, the results were verified in in vitro cultured cells, including isolated Bmal1 interference granulosa cells (GCs) and theca cells (TCs), respectively. Hormones levels of supernatant culture media and mRNA expressions of steroidogenesis-associated genes (star, Hsd3ß2, cyp19a1 in GCs, Lhcgr, star, Hsd3ß2, cyp17a1 in TCs) were mutually decreased, while the phosphorylation of PI3K/NfκB was promoted during in vitro luteinization. After PI3K specific-inhibitor LY294002 intervention, mRNA expressions of Lhcgr and Hsd3ß2 were partially rescued in Bmal1 interference TCs, together with significantly increased androstenedione and T synthesis. Further exploration in TCs demonstrated BMAL1 interacted directly but negatively with NfκB p65 (RelA), a subunit which was supposed as a mediator in Bmal1-governed PI3K signaling regulation. Taken together, we verified the novel role of Bmal1 in luteal steroidogenesis, achieving by negative interplay with RelA-mediated PI3K/NfκB pathway.


Assuntos
Fatores de Transcrição ARNTL/fisiologia , Hormônios Esteroides Gonadais/biossíntese , Células da Granulosa/metabolismo , Luteinização , Folículo Ovariano/metabolismo , Células Tecais/metabolismo , Androstenodiona/biossíntese , Animais , Estradiol/biossíntese , Feminino , Células da Granulosa/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Folículo Ovariano/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Progesterona/biossíntese , Testosterona/biossíntese , Células Tecais/patologia , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
9.
RNA Biol ; 17(12): 1798-1810, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32559120

RESUMO

Polycystic ovary syndrome (PCOS) causes anovulatory infertility in women of reproductive age, but etiopathogenesis of PCOS remains undetermined. Taurine up-regulated 1 (TUG1), an evolutionarily conserved long non-coding RNA, performs various biological functions; however, the role of TUG1 in PCOS remains unclear. Herein, TUG1 expression was assayed in granulosa cells (GCs) of 100 patients with PCOS and 100 control participants. Receiver operating characteristic (ROC) curve analysis was conducted to determine the diagnostic value of TUG1 in PCOS. TUG1 expression was also silenced in KGN cells to explore the role of TUG1 in cellular proliferation, apoptosis, cell-cycle progression, autophagy, and steroidogenesis. We found that TUG1 levels were dramatically increased in the PCOS group compared with those of the control group; this increased expression was related to a rising antral follicle count (R = 0.209, P < 0.001 versus control). The ROC curve indicated a significant separation between PCOS group and the control group (AUC: 0.702; 95% CI: 0.630-0.773; P < 0.001). TUG1 showed a predominantly nuclear localization in human GCs. TUG1 knockdown reduced cellular proliferation, and promoted MAPKs pathway-dependent apoptosis and P21-dependent autophagy, but may not affect cell-cycle progression. TUG1 knockdown increased aromatase expression and oestradiol biosynthesis. Our results indicate that increased TUG1 expression in PCOS GCs may contribute to excessive follicular activation and growth, and may disrupt the selection of dominant follicle. Our study shows that TUG1 can be used as a diagnostic biomarker for PCOS.


Assuntos
Regulação da Expressão Gênica , Síndrome do Ovário Policístico/genética , RNA Longo não Codificante/genética , Adulto , Apoptose/genética , Aromatase/metabolismo , Autofagia/genética , Biomarcadores , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Suscetibilidade a Doenças , Estradiol/biossíntese , Feminino , Hormônio Foliculoestimulante/sangue , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células da Granulosa/metabolismo , Humanos , Insulina/administração & dosagem , Insulina/uso terapêutico , Sistema de Sinalização das MAP Quinases , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Síndrome do Ovário Policístico/diagnóstico , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo , Curva ROC , Adulto Jovem
10.
Toxicol Ind Health ; 36(12): 1010-1018, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33190593

RESUMO

Exposure to an electromagnetic field (EMF) can have adverse effects on many organs and tissues, including the reproductive system. This study aimed to investigate the effects of EMF exposure during prenatal and postnatal periods on ovarian development in rat offspring. In this study, rat pups born from eight pregnant rats were used. EMF exposure was initiated on the first day of pregnancy and continued until the 42nd postnatal day. The blood and ovarian tissue samples of female offspring in sham and EMF groups were collected when they reached the age of 42 days. Follicle-stimulating hormone levels were significantly higher in the EMF group than in the sham group. Estradiol levels were significantly lower in the EMF group than in the sham group. Tissue-inducible nitric oxide synthase (iNOS) levels and expression were significantly greater in the EMF group than in the sham group. In the EMF group, congestion, bleeding areas, and degeneration of follicle structures were observed in ovarian tissue. The findings suggest that exposure to 50-Hz, 3-mT EMF used in this study during prenatal and postnatal periods may lead to impaired ovarian structure and function in female offspring. EMF may affect ovarian physiology by increasing iNOS levels and may lead to fertility disorders.


Assuntos
Campos Eletromagnéticos/efeitos adversos , Ovário/efeitos da radiação , Efeitos Tardios da Exposição Pré-Natal/veterinária , Animais , Animais Recém-Nascidos , Estradiol/biossíntese , Feminino , Hormônio Foliculoestimulante/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Gravidez , Ratos , Ratos Sprague-Dawley
11.
Am J Physiol Endocrinol Metab ; 316(6): E1071-E1080, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30939050

RESUMO

The transition from granulosa cell (GC) to luteal cell involves a change from estrogen production to predominantly progesterone production. We analyzed the role of Krüppel-like factor 4 (Klf4), a transcriptional repressor used to generate pluripotent cells, in that transition. After luteinizing hormone (LH)/human chorionic gonadotropin treatment of preovulatory follicles, a major but transient increase in Klf4 transcript levels was detected. Therefore, we enquired whether Klf4 is involved in the rapid decline of aromatase, the key estrogen-producing enzyme, using preovulatory GCs obtained from pregnant mare serum gonadotropin-primed immature rat ovaries. Cyp19A1 expression in GCs transfected with FLAG-Klf4 or Klf4-specific siRNA was analyzed by real-time PCR and immunofluorescence staining. Cyp19A1 decreased when Klf4 was overexpressed, and Cyp19A1 and estradiol biosynthesis increased when Klf4 was knocked down. The mechanism by which Klf4 regulates Cyp19A1 expression was investigated using Cyp19A1 promoter-luciferase reporter assays and chromatin immunoprecipitation assays. The results revealed that the steroidogenic factor-1 (SF1)-binding motif, but not the specificity protein 1 (Sp1) binding element or the CACCC motif, was required for Klf4-mediated repression of Cyp19A1 promoter activity. Here we showed that Klf4 suppressed endogenous Cyp19A1 transcript and protein production, and this resulted from direct binding of Klf4 to the SF1 recognition motif in the Cyp19A1 promoter. These findings suggest that Klf4 is a physiologic regulator of Cyp19A1 expression in response to the LH surge in preovulatory GCs and that it has an essential role in the luteal transition in steroidogenesis.


Assuntos
Aromatase/genética , Estradiol/biossíntese , Células da Granulosa/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Hormônio Luteinizante/metabolismo , Progesterona/biossíntese , Animais , Aromatase/metabolismo , Diferenciação Celular , Feminino , Fase Folicular , Técnicas de Silenciamento de Genes , Gonadotropinas Equinas , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Regiões Promotoras Genéticas , Ratos
12.
Cell Tissue Res ; 375(3): 743-754, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30377784

RESUMO

We previously demonstrated that female Runx3 knockout (Runx3-/-) mice were anovulatory and their uteri were atrophic and that Runx3 mRNA was expressed in granulosa cells. To clarify how Runx3 regulates folliculogenesis and ovulation, we examine the effects of Runx3 knockout on the gene expression of growth factors associated with folliculogenesis and enzymes associated with steroidogenesis. In Runx3-/- mouse ovaries, the numbers of primary and antral follicles were lower than those in wild-type (wt) mice at 3 weeks of age, indicating that the loss of Runx3 affects folliculogenesis. The expression of genes encoding activin and inhibin subunits (Inha, Inhba and Inhbb) was also decreased in ovaries from the Runx3-/- mice compared with that in wt mice. Moreover, the expression of the genes Cyp11a1 and Cyp19a1 encoding steroidogenic enzymes was also decreased. In cultured granulosa cells from 3-week-old mouse ovaries, Cyp19a1 mRNA levels were lower in Runx3-/- mice than those in wt mice. Follicle-stimulating hormone (FSH) treatment increased Cyp19a1 mRNA levels in both wt and Runx3-/- granulosa cells in culture but the mRNA level in Runx3-/- granulosa cells was lower than that in wt ones, indicating that granulosa cells could not fully function in the absence of Runx3. At 3 weeks of age, gonadotropin α subunit, FSHß subunit and luteinizing hormone (LH) ß subunit mRNA levels were decreased in Runx3-/- mice. These findings suggest that Runx3 plays a key role in female reproduction by regulating folliculogenesis and steroidogenesis in granulosa cells.


Assuntos
Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Células da Granulosa/metabolismo , Organogênese , Esteroides/biossíntese , Animais , Subunidade alfa 3 de Fator de Ligação ao Core/deficiência , Estradiol/biossíntese , Feminino , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Organogênese/efeitos dos fármacos , Progesterona/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores da Gonadotropina/genética , Receptores da Gonadotropina/metabolismo
13.
Mol Reprod Dev ; 86(8): 1023-1032, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31179605

RESUMO

Long noncoding RNAs (lncRNAs) have been demonstrated to play vital roles in mammalian reproduction. Our previous research revealed that lncRNA Gm2044 is highly expressed in mouse spermatocytes and regulates male germ cell function. The gene annotation database BioGPS shows that Gm2044 is not only highly expressed in testicular tissue but also in ovarian tissue, which suggests that Gm2044 may be involved in female reproductive development. In this study, we confirmed that lncRNA Gm2044 promotes 17ß-estradiol synthesis in mouse pre-antral follicular granulosa cells (mpGCs). Furthermore, bioinformatics methods, western blot, and the luciferase assay proved that Gm2044 functions as a miR-138-5p sponge to inhibit the direct target of miR-138-5p, Nr5a1, which enhances 17ß-estradiol synthesis through cyp19a1 activation. Taken together, our results provide an insight into the mechanistic roles of lncRNA Gm2044 for 17ß-estradiol synthesis by acting as competing-endogenous RNAs to modulate the function of mpGCs. Studying the potential lncRNAs, which regulate estradiol release, will be beneficial for the diagnosis and treatment of steroid hormone-related disease.


Assuntos
Estradiol/biossíntese , Células da Granulosa/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Aromatase/biossíntese , Feminino , Camundongos , Camundongos Endogâmicos ICR , Fator Esteroidogênico 1/biossíntese
14.
Mol Reprod Dev ; 86(2): 166-174, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30625262

RESUMO

In cattle and other species, the fetal ovary is steroidogenically active before follicular development commences, and there is evidence that estradiol and progesterone inhibit follicle formation and activation. Estradiol levels decline sharply around the time of follicle formation. In the present study, we hypothesized that FGF10 and FGF18, which inhibit estradiol secretion from granulosa cells of antral follicles, also regulate fetal ovarian steroid production. Fetuses were collected at local abattoirs, and age determined by crown-rump length measurements. Real-time polymerase chain reaction assays with RNA extracted from whole ovaries revealed that the abundance of CYP19A1 messenger RNA (mRNA) decreased from 60 to 90 days of gestation, which is consistent with the decline in estradiol secretion previously observed. Immunohistochemistry revealed the presence of FGF18 in ovigerous cords in early gestation and in oocytes later in fetal age (≥150 days). The abundance of FGF18 mRNA increased after Day 90 gestation. Addition of recombinant FGF18 to fetal ovarian pieces inhibited estradiol and progesterone secretion in vitro, whereas FGF10 was without effect. Consistent with these results, FGF18 decreased levels of mRNA for CYP19A1 and CYP11A1 in ovarian pieces in vitro. These data suggest that FGF18 may be an intraovarian factor that regulates steroidogenesis in fetal ovaries.


Assuntos
Estradiol/biossíntese , Feto/metabolismo , Fatores de Crescimento de Fibroblastos/biossíntese , Células da Granulosa/metabolismo , Progesterona/biossíntese , Animais , Bovinos , Feminino , Feto/citologia , Idade Gestacional , Células da Granulosa/citologia
15.
Mol Reprod Dev ; 86(11): 1582-1591, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31353672

RESUMO

In the bovine oviduct, estradiol (E2) stimulates secretion and cell proliferation, whereas progesterone (P4) suppresses them. In this study, we have evaluated the effect of two superstimulatory protocols (follicle-stimulating hormone [FSH] or FSH combined with equine chorionic gonadotropin [eCG]) on the oviductal levels of E2 and P4 and its outcome on oviductal cells. Compared with the control group (a single pre-ovulatory follicle), we have observed that the cows submitted to FSH/eCG treatment showed a higher concentration of E2 in the oviduct tissue, together with a higher abundance of messenger RNA encoding steroid receptors (ESR1 and progesterone receptor), and genes linked to gamete interactions and regulation of polyspermy (oviduct-specific glycoprotein 1, heat-shock protein family A member 5, α-l-fucosidase 1 [FUCA1], and FUCA2) in the infundibulum and ampulla segments of the oviduct. However, we did not observe any modulation of gene expression in the isthmus segment. Even though the FSH protocol upregulated some of the genes analyzed, we may infer that the steady effect of FSH combined with eCG on oviduct regulation might benefit fertilization and may potentially increase pregnancy rates.


Assuntos
Gonadotropina Coriônica/farmacologia , Estradiol/biossíntese , Tubas Uterinas/metabolismo , Fertilização , Transcrição Gênica/efeitos dos fármacos , Animais , Bovinos , Tubas Uterinas/citologia , Feminino , Cavalos
16.
Mol Reprod Dev ; 86(1): 63-74, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30372551

RESUMO

Cumulus cells secreting steroid hormones have important functions in oocyte development. Several members of the short-chain dehydrogenase/reductase (SDR) family are critical to the biosynthesis of steroid hormones. NADPH-dependent retinol dehydrogenase/reductase ( NRDR), a member of the SDR superfamily, is overexpressed in pig breeds that also show high levels of androstenone. However, the potential functions and regulatory mechanisms of NRDR in pig ovaries have not been reported to date. The present study demonstrated that NRDR is highly expressed in pig ovaries and is specifically located in cumulus granulosa cells. Functional studies showed that NRDR inhibition increased estradiol synthesis. Both pregnant mare serum gonadotropin and human chorionic gonadotropin downregulated the expression of NRDR in pig cumulus granulosa cells. When the relationship between reproductive traits and single-nucleotide polymorphisms (SNPs) of the NRDR gene was examined, we found that two SNPs affected reproductive traits. SNP rs701332503 was significantly associated with a decrease in the total number of piglets born during multiparity, and rs326982309 was significantly associated with an increase in the average birth weight during primiparity. Thus, NRDR has an important role in steroid hormone biosynthesis in cumulus granulosa cells, and NRDR SNPs are associated with changes in porcine reproduction traits.


Assuntos
Oxirredutases do Álcool , Estradiol/biossíntese , Ovário/enzimologia , Paridade/genética , Polimorfismo de Nucleotídeo Único , Característica Quantitativa Herdável , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Animais , Gonadotropina Coriônica/farmacologia , Estradiol/genética , Feminino , Gonadotropinas Equinas/farmacologia , Humanos , Gravidez , Suínos
17.
Neurochem Res ; 44(8): 1781-1795, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31254250

RESUMO

The hippocampus-derived neuroestradiol plays a major role in neuroplasticity, independent of circulating estradiol that originates from gonads. The response of hypothalamus-pituitary regions towards the synthesis of neuroestradiol in the hippocampus is an emerging scientific concept in cognitive neuroscience. Hippocampal plasticity has been proposed to be regulated via neuroblasts, a major cellular determinant of functional neurogenesis in the adult brain. Defects in differentiation, integration and survival of neuroblasts in the hippocampus appear to be an underlying cause of neurocognitive disorders. Gonadotropin receptors and steroidogenic enzymes have been found to be expressed in neuroblasts in the hippocampus of the brain. However, the reciprocal relationship between hippocampal-specific neuroestradiol synthesis along neuroblastosis and response of pituitary based feedback regulation towards regulation of estradiol level in the hippocampus have not completely been ascertained. Therefore, this conceptual article revisits (1) the cellular basis of neuroestradiol synthesis (2) a potential relationship between neuroestradiol synthesis and neuroblastosis in the hippocampus (3) the possible involvement of aberrant neuroestradiol production with mitochondrial dysfunctions and dyslipidemia in menopause and adult-onset neurodegenerative disorders and (4) provides a hypothesis for the possible existence of the hypothalamic-pituitary-hippocampal (HPH) axis in the adult brain. Eventually, understanding the regulation of hippocampal neurogenesis by abnormal levels of neuroestradiol concentration in association with the feedback regulation of HPH axis might provide additional cues to establish a neuroregenerative therapeutic management for mood swings, depression and cognitive decline in menopause and neurocognitive disorders.


Assuntos
Estradiol/metabolismo , Hipocampo/fisiologia , Menopausa/fisiologia , Doenças Neurodegenerativas/fisiopatologia , Neurogênese/fisiologia , Hipófise/fisiologia , Envelhecimento/fisiologia , Animais , Estradiol/biossíntese , Feminino , Hipocampo/fisiopatologia , Humanos , Doenças Mitocondriais/fisiopatologia , Plasticidade Neuronal/fisiologia , Hipófise/fisiopatologia
18.
J Pineal Res ; 66(2): e12546, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30586196

RESUMO

The interaction of granulosa cells (GCs) with oocytes is important to regulate follicle development. The exogenous melatonin promoting the maturation of oocytes by GCs has been approved in pig, however, the transcriptome profile and the functions of the genes regulated by melatonin in GCs have not yet to be fully characterized. In this study, we found melatonin could stimulate the synthesis of estradiol in pig GCs. The RNA-seq was used to explore the effects of melatonin on gene expression, a total of 89 differentially expressed genes (DEGs) were identified. Gene ontology analysis showed DEGs which associated with regulation of cell proliferation, cell cycle, and anti-apoptosis were significantly enriched. The functions of two DEGs, NOTCH2 and FILIP1L, were studied in pig GCs. The results showed that NOTCH2 inhibited the synthesis of estradiol, but FILIP1L promoted the synthesis of estradiol. Furthermore, inhibiting NOTCH2 in granulosa cells cocultured with cumulus-oocyte-complexes had no obvious effect on the maturation of pig oocyte, but could upregulate the cleavage rate of oocyte. We proved that FILIP1L had no effect on the maturation and cleavage of pig oocytes. Our work deepens the understanding of melatonin's effects on GCs and oocyte. The DEGs we found will be beneficial to reveal mechanisms of melatonin acting on GCs and oocytes and design the pharmacological interventions.


Assuntos
Estradiol/biossíntese , Expressão Gênica/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Melatonina/farmacologia , Oócitos/efeitos dos fármacos , Animais , Feminino , Células da Granulosa/metabolismo , Suínos
19.
J Appl Toxicol ; 39(7): 945-954, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30834569

RESUMO

Fluorinated diiodine alkanes (FDIAs), important industrial intermediates in the synthesis of various perfluorinated compounds, which are distributed widely in wildlife and humans. Recent studies showed that FDIAs had in vitro estrogenic effects. However, to date, little information is available regarding the in vivo estrogenic effects of FDIAs and the mechanisms are unclear. In this study, a combination of in vitro and in vivo assays was used to investigate the estrogenic effects of FDIAs. We tested the in vitro estrogenic effects and estrogen receptor-related gene expression via MCF-7 cell assay. The hormone level of estradiol and the expression of estrogenic synthesis genes were measured in the H295R cell assay. Finally, the in vivo effects of FDIAs on development and estrogen-related gene expression were assessed in the zebrafish embryos assay. The results demonstrated that FDIAs could exhibit estrogenic activity through inducing cell proliferation (1.6-6.7-fold of the control) and estrogen receptor alpha gene expression (1.07-1.39-fold of the control), altering estradiol production (1.14-1.22-fold of the control) and the major estrogenic synthesis gene expression of CYP19 (1.22-1.31-fold of the control), disrupting the estrogen-related genes (esr1 and cyp19b) levels in zebrafish (1.52-2.99-fold and 2.95-5.00-fold of the control for esr1 and cyp19b, respectively). The current findings indicated the potential estrogenic effects of FDIAs and provided novel information for human risk assessment.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Estradiol/metabolismo , Estrogênios/toxicidade , Hidrocarbonetos Fluorados/toxicidade , Hidrocarbonetos Iodados/toxicidade , Peixe-Zebra , Alcanos/toxicidade , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Estradiol/biossíntese , Receptor alfa de Estrogênio/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7
20.
Reprod Domest Anim ; 54(11): 1459-1469, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31424586

RESUMO

Female fertility potential is based on the development and growth of ovarian follicles. Our previous study showed that miR-17-5p was significantly differently expressed in pre-ovulatory ovarian follicles of Large White (LW) and Chinese Taihu (CT) sows. In the present study, we investigated the role of miR-17-5p in ovarian follicle development. We demonstrated that miR-17-5p overexpression significantly decreased the luciferase reporter activity containing the E2F transcription factor 1 (E2F1) 3'untranslated region (3'UTR) and suppressed the E2F1 expression, whereas the miR-17-5p inhibition increased the E2F1 expression in porcine granulosa cells (pGCs). Meanwhile, miR-17-5p overexpression or E2F1 knockdown promoted cell growth, follicular development marker genes (LHR, CYP19A1 and AREG) expression and oestradiol synthesis, and miR-17-5p inhibition suppressed cell growth, follicular development marker genes (LHR, CYP19A1 and AREG) expression and oestradiol synthesis in pGCs. Furthermore, E2F1 knockdown increased CYP19A1 promoter activity. This study suggests that miR-17-5p regulates pGC growth and oestradiol synthesis by targeting E2F1 gene.


Assuntos
Proliferação de Células , Fator de Transcrição E2F1/genética , Células da Granulosa/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Animais , Estradiol/biossíntese , Feminino , Técnicas de Silenciamento de Genes , MicroRNAs/genética , Folículo Ovariano/metabolismo , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA