Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Biol Reprod ; 107(3): 800-812, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-35639639

RESUMO

Estradiol-17ß has been shown to promote primordial follicle formation and to involve bone morphogenetic protein 2 (BMP2) as a downstream effector to promote primordial follicle in hamsters. However, the molecular mechanism whereby these factors regulate ovarian somatic cells to pre-granulosa cells transition leading to primordial follicle formation remains unclear. The objective of this study was to determine whether BMP2 and/or estradiol-17ß would regulate the expression of specific ovarian transcriptome during pre-granulosa cells transition and primordial follicle formation in the mouse ovary. BMP2 mRNA level increased during the period of primordial follicle formation with the concurrent presence of BMP2 protein in ovarian somatic cells. Estradiol-17ß but not BMP2 exposure led to increased expression of ovarian BMP2 messenger RNA (mRNA), and the effect of estradiol-17ß could not be suppressed by 4-[6-[4-(1-Piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline dihydrochloride (LDN) 193189. BMP2 or estradiol-17ß stimulated primordial follicle formation without inducing apoptosis. Ribonucleic acid-sequence analysis (RNA-seq) of ovaries exposed to exogenous BMP2 or estradiol-17ß revealed differential expression of several thousand genes. Most of the differentially expressed genes, which were common between BMP2 or estradiol-17ß treatment demonstrated concordant changes, suggesting that estradiol-17ß and BMP2 affected the same set of genes during primordial follicle formation. Further, we have identified that estradiol-17ß, in cooperation with BMP2, could affect the expression of three major transcription factors, GATA binding protein 2, GATA binding protein 4 and Early growth response 2, and one serine protease, hepsin, in pre-granulosa cells during primordial follicle formation. Taken together, results of this study suggest that estradiol-17ß and BMP2 may regulate ovarian gene expression that promote somatic cells to pre-granulosa cells transition and primordial follicle formation in the mouse ovary.


Assuntos
Estradiol , Ovário , Transcriptoma , Animais , Proteína Morfogenética Óssea 2/farmacologia , Cricetinae , Estradiol/farmacologia , Feminino , Camundongos , Ovário/metabolismo , RNA Mensageiro/metabolismo
2.
FASEB J ; 23(11): 3752-65, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19608628

RESUMO

The objective of this study was to determine whether surface-modified nanoparticles enhance permeability across nasal mucosa, while retaining the effectiveness of the payload. The uptake and permeability of polystyrene nanoparticles (PS-NPs; FluoSpheres) was evaluated across the various regions of the bovine nasal epithelia following conjugation with deslorelin and transferrin. Uptake and transport of PS-NPs, deslorelin-PS-NPs, and transferrin-PS-NPs exhibited regional differences in the order: inferior turbinate posterior (ITP) > medium turbinate posterior (MTP) > medium turbinate anterior (MTA). Uptake and transport also exhibited directionality and temperature dependence in these tissues. Further, uptake as well as transport of functionalized nanoparticles could be inhibited by excess free functionalizing ligand. Confocal microscopy indicated the presence of functionalized nanoparticles in respiratory epithelial cells, as well as other cell types of the nasal tissue. We chose the ITP region for further studies with deslorelin or transferrin-conjugated poly-l-lactide-co-glycolide nanoparticles (PLGA-NPs) encapsulating an anti-VEGF intraceptor (Flt23k) plasmid. Transport of the nanoparticles, as well as the plasmid from the nanoparticles, exhibited the following order: transferrin-PLGA-NPs > deslorelin-PLGA-NPs > PLGA-NPs >> plasmid. The ability of the nanoparticles transported across the nasal tissue to retain the effectiveness of the Flt23k plasmid was evaluated by measuring transfection efficiency (percentage of cells expressing GFP) and VEGF inhibition in LNCaP and PC-3 prostate cancer cells. Transfection efficiencies and VEGF inhibition in LNCaP and PC-3 cells exhibited the following trend: transferrin-PLGA-NPs >or= deslorelin-PLGA-NPs > PLGA-NPs >> plasmid. Further, functionalized nanoparticles exhibited transfection efficiencies and VEGF inhibition significantly superior compared with the routinely used transfecting agent, lipofectamine. Formulating plasmids into nanoparticulate delivery systems enhances the transnasal delivery and gene therapy at remote target cancer cells, which can be further enhanced by nanoparticle functionalization with deslorelin or transferrin.


Assuntos
Mucosa Nasal/metabolismo , Administração Intranasal , Animais , Bovinos , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos , Ácido Láctico/administração & dosagem , Nanopartículas , Ácido Poliglicólico/administração & dosagem , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Transferrina/administração & dosagem , Pamoato de Triptorrelina/administração & dosagem , Pamoato de Triptorrelina/análogos & derivados
3.
Circ Res ; 103(2): 186-93, 2008 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-18566341

RESUMO

We have previously reported that the expression of Angiotensin II (Ang II) type 1 receptors (AT1R) was increased in the rostral ventrolateral medulla (RVLM) of rabbits with chronic heart failure (CHF) and in the RVLM of normal rabbits infused with intracerebroventricular (ICV) Ang II. The present study investigated whether oxidant stress plays a role in Ang II-induced AT1R upregulation and its relationship to the transcription factor activator protein 1 (AP1) in CHF rabbits and in the CATHa neuronal cell line. In CATHa cells, Ang II significantly increased AT1R mRNA by 123+/-11%, P<0.01; c-Jun mRNA by 90+/-20%, P<0.01; c-fos mRNA by 148+/-49%, P<0.01; NADPH oxidase activity by 126+/-43%, P<0.01 versus untreated cells. Tempol and Apocynin reversed the increased expression of AT1R mRNA, c-Jun mRNA, c-fos mRNA, and superoxide production induced by Ang II. We also examined the effect of ICV Tempol on the RVLM of CHF rabbits. Compared to vehicle treated CHF rabbits, Tempol significantly decreased AT1R protein expression (1.6+/-0.29 versus 0.88+/-0.16, P<0.05), phosphorylated Jnk protein (0.4+/-0.05 versus 0.2+/-0.04, P<0.05), cytosolic phosphorylated c-Jun (0.56+/-0.1 versus 0.36+/-0.05, P<0.05), and nuclear phosphorylated c-Jun (0.67+/-0.1 versus 0.3+/-0.08, P<0.01). Tempol also significantly decreased the AP-1-DNA binding activity in the RVLM of CHF rabbits compared to the vehicle group (9.14 x 10(3) versus 41.95 x 10(3) gray level P<0.01). These data suggest that Ang II induces AT1R upregulation at the transcriptional level by induction of oxidant stress and activation of AP1 in both cultured neuronal cells and in intact brain of rabbits. Antioxidant agents may be beneficial in CHF and other states where brain Ang II is elevated by decreasing AT1R expression through the Jnk and AP1 pathway.


Assuntos
Insuficiência Cardíaca/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Receptor Tipo 1 de Angiotensina/metabolismo , Angiotensina II/metabolismo , Animais , Antioxidantes/farmacologia , Células Cultivadas , Doença Crônica , Óxidos N-Cíclicos/farmacologia , Modelos Animais de Doenças , Insuficiência Cardíaca/patologia , MAP Quinase Quinase 4/metabolismo , Masculino , NADPH Oxidases/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/metabolismo , Coelhos , Espécies Reativas de Oxigênio/metabolismo , Marcadores de Spin , Superóxidos/metabolismo , Fator de Transcrição AP-1/metabolismo
4.
Carcinogenesis ; 30(9): 1581-90, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19520794

RESUMO

Estrogen receptors (ERs) [ERalpha (Esr1) and ERbeta (Esr2)] are expressed in the human colon, but during the multistep process of colorectal carcinogenesis, expression of both ERalpha and ERbeta is lost, suggesting that loss of ER function might promote colorectal carcinogenesis. Through crosses between an ERalpha knockout and Apc(Min) mouse strains, we demonstrate that ERalpha deficiency is associated with a significant increase in intestinal tumor multiplicity, size and burden in Apc(Min/+) mice. Within the normal intestinal epithelium of Apc(Min/+) mice, ERalpha deficiency is associated with an accumulation of nuclear beta-catenin, an indicator of activation of the Wnt-beta-catenin-signaling pathway, which is known to play a critical role in intestinal cancers. Consistent with the hypothesis that ERalpha deficiency is associated with activation of Wnt-beta-catenin signaling, ERalpha deficiency in the intestinal epithelium of Apc(Min/+) mice also correlated with increased expression of Wnt-beta-catenin target genes. Through crosses between an ERbeta knockout and Apc(Min) mouse strains, we observed some evidence that ERbeta deficiency is associated with an increased incidence of colon tumors in Apc(Min/+) mice. This effect of ERbeta deficiency does not involve modulation of Wnt-beta-catenin signaling. Our studies suggest that ERalpha and ERbeta signaling modulate colorectal carcinogenesis, and ERalpha does so, at least in part, by regulating the activity of the Wnt-beta-catenin pathway.


Assuntos
Receptor alfa de Estrogênio/deficiência , Receptor beta de Estrogênio/deficiência , Genes APC , Neoplasias Intestinais/etiologia , Transdução de Sinais/fisiologia , Animais , Caderinas/análise , Colo/química , Ciclina D1/análise , Estradiol/sangue , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Feminino , Neoplasias Intestinais/genética , Masculino , Camundongos , Ovário/patologia , Proteínas Wnt/fisiologia , beta Catenina/análise , beta Catenina/fisiologia
5.
Peptides ; 30(2): 351-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18992782

RESUMO

Deslorelin, a luteinizing hormone releasing hormone (LHRH) agonist, is transported via the LHRH-receptor (LHRH-R) and exhibits regional variation as follows: inferior turbinate posterior (ITP)>medium turbinate posterior (MTP)>medium turbinate anterior (MTA) of the bovine nasal mucosa. Differential LHRH-R expression in various regions of the nose is a potential explanation for regional variation in deslorelin transport. Thus, the objective was to determine whether LHRH-R expression exhibits regional variation in bovine nasal mucosa. LHRH-R density (B(max)) and affinity constant (K(d)) were determined by saturation experiments using 0.5mg tissue in the presence of increasing amounts of I(125)-deslorelin (100-100,000 cpm) at 4 degrees C for 4h. The 50% inhibitory concentration (IC(50)) was determined by competition experiments using various amounts of unlabelled deslorelin (0.01-3000 ng) at 4 degrees C for 4h. LHRH-R mRNA and protein expressions were determined using real-time PCR and Western blot analysis, respectively. LHRH-R B(max) and K(d) varied between the regions of excised bovine nasal mucosa: ITP>MTP>MTA. The inhibition experiments yielded two IC(50) concentrations which exhibited trends similar to B(max) and K(d). Real-time PCR and Western blot analysis indicated that LHRH-R expression exhibits similar trends: ITP>MTP>MTA. We identified two deslorelin binding sites in the nasal tissues, with high affinity sites representing approximately 60-70% of the total sites available. In summary, regional differences in nasal deslorelin transport correlate with regional differences in LHRH-R expression, with LHRH-R expression, peptide binding, and transport being the highest in the inferior turbinate posterior region of the nose.


Assuntos
Mucosa Nasal/metabolismo , Receptores LHRH/metabolismo , Pamoato de Triptorrelina/análogos & derivados , Animais , Transporte Biológico , Bovinos , Expressão Gênica , RNA Mensageiro/metabolismo , Receptores LHRH/genética , Pamoato de Triptorrelina/metabolismo , Conchas Nasais/metabolismo
6.
Mol Endocrinol ; 22(2): 403-14, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17916653

RESUMO

In most mammals, prostaglandin F2alpha (PGF2alpha) is believed to be a trigger that induces the regression of the corpus luteum (CL), whereby progesterone synthesis is inhibited, the luteal structure involutes, and the reproductive cycle resumes. Studies have shown that the early growth response 1 (EGR1) protein can induce the expression of proapoptotic proteins, suggesting that EGR1 may play a role in luteal regression. Our hypothesis is that EGR1 mediates the actions of PGF2alpha by inducing the expression of TGF beta1 (TGFB1), a key tissue remodeling protein. The levels of EGR1 mRNA and protein were up-regulated in the bovine CL during PGF2alpha-induced luteolysis in vivo and in PGF2alpha-treated luteal cells in vitro. Using chemical and genetic approaches, the RAF/MAPK kinase (MEK) 1/ERK pathway was identified as a proximal signaling event required for the induction of EGR1 in PGF2alpha-treated cells. Treatment with PGF2alpha increased the expression of TGFB1 mRNA and protein as well as the binding of EGR1 protein to TGFB1 promoter in bovine luteal cells. The effect of PGF2alpha on TGFB1 expression was mimicked by a protein kinase C (PKC)/RAF/MEK1/ERK activator or adenoviral-mediated expression of EGR1. The stimulatory effect of PGF2alpha on TGFB1 mRNA and TGFB1 protein secretion was inhibited by blockade of MEK1/ERK signaling and by adenoviral-mediated expression of NAB2, an EGR1 binding protein that inhibits EGR1 transcriptional activity. Treatment of luteal cells with TGFB1 reduced progesterone secretion, implicating TGFB1 in luteal regression. These studies demonstrate that PGF2alpha stimulates the expression of EGR1 and TGFB1 in the CL. We suggest that EGR1 plays a role in the expression of genes whose cognate proteins coordinate luteal regression.


Assuntos
Corpo Lúteo/efeitos dos fármacos , Dinoprosta/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/genética , Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta1/genética , Animais , Northern Blotting , Western Blotting , Bovinos , Núcleo Celular/metabolismo , Imunoprecipitação da Cromatina , Corpo Lúteo/metabolismo , AMP Cíclico/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Imuno-Histoquímica , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Hormônio Luteinizante/farmacologia , MAP Quinase Quinase 1/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
7.
Endocrinology ; 149(9): 4452-61, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18499747

RESUMO

Estradiol-17beta (E2) plays an important role in the formation and development of primordial follicles, but the mechanisms remain unclear. G protein-coupled receptor 30 (GPR30) can mediate a rapid and transcription-independent E2 signaling in various cells. The objectives of this study were to examine whether GPR30 was expressed in the neonatal hamster ovary and whether it could mediate estrogen action during the formation of primordial follicles. GPR30 mRNA levels decreased from the 13th day of gestation (E13) through the second day of postnatal (P2) life, followed by steady increases from P3 through P6. Consistent with the changes in mRNA levels, GPR30 protein expression decreased from E13 to P2 followed by a significant increase by P7, the day before the first appearance of primordial follicles in the hamster ovary. GPR30 was expressed both in the oocytes and somatic cells, although the expression in the oocytes was low. GPR30 protein was located primarily in the perinuclear endoplasmic reticulum, which was also the site of E2-BSA-FITC (E2-BSA-fluorescein isothiocyanate) binding. E2 or E2-BSA increased intracellular calcium in neonatal hamster ovary cells in vitro. Exposure to GPR30 small interfering RNA in vitro significantly reduced GPR30 mRNA and protein levels in cultured hamster ovaries, attenuated E-BSA binding to cultured P6 ovarian cells, and markedly suppressed estrogen-stimulated primordial follicle formation. These results suggest that a membrane estrogen receptor, GPR30, is expressed in the ovary during perinatal development and mediates E2 action on primordial follicle formation.


Assuntos
Estradiol/farmacologia , Folículo Ovariano/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Animais , Animais Recém-Nascidos , Células Cultivadas , Cricetinae , Embrião de Mamíferos , Estradiol/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Ovário/efeitos dos fármacos , Ovário/metabolismo , Ovário/fisiologia , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Distribuição Tecidual
8.
Circ Res ; 99(9): 1004-11, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-17008603

RESUMO

Chronic heart failure (CHF) is a leading cause of mortality in developed countries. Angiotensin II (Ang II) plays an important role in the development and progression of CHF. Many of the important functions of Ang II are mediated by the Ang II type 1 receptor (AT(1)R), including the increase in sympathetic nerve activity in CHF. However, the central regulation of the AT(1)R in the setting of CHF is not well understood. This study investigated the AT(1)R in the rostral ventrolateral medulla (RVLM) of rabbits with CHF, its downstream pathway, and its gene regulation by the transcription factor activator protein 1 (AP-1). Studies were performed in 5 groups of rabbits: sham (n=5), pacing-induced (3 to 4 weeks) CHF (n=5), CHF with intracerebroventricular (ICV) losartan treatment (n=5), normal with ICV Ang II treatment (n=5), and normal with ICV Ang II plus losartan treatment (n=5). AT(1)R mRNA and protein expressions, plasma Ang II, and AP-1-DNA binding activity were significantly higher in RVLM of CHF compared with Sham rabbits (240.4+/-30.2%, P<0.01; 206.6+/-25.8%, P<0.01; 280+/-36.5%, P<0.05; 207+/-16.4%, P<0.01, respectively). Analysis of the stress-activated protein kinase/Jun N-terminal kinase (SAPK/JNK) pathway showed that phosphorylated c-Jun proteins, phosphorylated JNK proteins, and JNK activity increased significantly in RVLM of CHF compared with sham (262.9+/-48.1%, 213.8+/-27.7%, 148.2+/-10.1% of control, respectively). Importantly, ICV losartan in CHF rabbits attenuated these increases. ICV Ang II in normal rabbits simulated the molecular changes seen in CHF. This effect was blocked by concomitant ICV losartan. In addition, Ang II-induced AT(1)R expression was blocked by losartan and a JNK inhibitor, but not by extracellular signal-regulated kinase or p38 MAP kinase inhibitors in a neuronal cell culture. These data suggest that central Ang II activates the AT(1)R, SAPK/JNK pathway. AP-1 may further regulate gene expression in RVLM in the CHF state.


Assuntos
Baixo Débito Cardíaco/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Receptor Tipo 1 de Angiotensina/biossíntese , Fator de Transcrição AP-1/metabolismo , Regulação para Cima , Angiotensina II/administração & dosagem , Angiotensina II/sangue , Angiotensina II/farmacologia , Animais , Peso Corporal , Baixo Débito Cardíaco/enzimologia , Baixo Débito Cardíaco/genética , Linhagem Celular , Ecocardiografia , Coração/anatomia & histologia , Coração/fisiologia , Injeções Intraventriculares , Sistema de Sinalização das MAP Quinases , Masculino , Tamanho do Órgão , Proteínas Proto-Oncogênicas c-jun/biossíntese , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/metabolismo , Coelhos , Receptor Tipo 1 de Angiotensina/genética
9.
J Immunotoxicol ; 15(1): 73-81, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29648480

RESUMO

Skeletal health consequences associated with inflammatory diseases of the airways significantly contribute to morbidity. Sex differences have been described independently for lung and bone diseases. Repetitive inhalant exposure to lipopolysaccharide (LPS) induces bone loss and deterioration in male mice, but comparison effects in females are unknown. Using an intranasal inhalation exposure model, 8-week-old C57BL/6 male and female mice were treated daily with LPS (100 ng) or saline for 3 weeks. Bronchoalveolar lavage fluids, lung tissues, tibias, bone marrow cells, and blood were collected. LPS-induced airway neutrophil influx, interleukin (IL)-6 and neutrophil chemoattractant levels, and bronchiolar inflammation were exaggerated in male animals as compared to female mice. Trabecular bone micro-CT imaging and analysis of the proximal tibia were conducted. Inhalant LPS exposures lead to deterioration of bone quality only in male mice (not females) marked by decreased bone mineral density, bone volume/tissue volume ratio, trabecular thickness and number, and increased bone surface-to-bone volume ratio. Serum pentraxin-2 levels were modulated by sex differences and LPS exposure. In proof-of-concept studies, ovarectomized female mice demonstrated LPS-induced bone deterioration, and estradiol supplementation of ovarectomized female mice and control male mice protected against LPS-induced bone deterioration findings. Collectively, sex-specific differences exist in LPS-induced airway inflammatory consequences with significant differences found in bone quantity and quality parameters. Male mice demonstrated susceptibility to bone loss and female animals were protected, which was modulated by estrogen. Therefore, sex differences influence the biologic response in the lung-bone inflammatory axis in response to inhalant LPS exposures.


Assuntos
Reabsorção Óssea/imunologia , Osso e Ossos/imunologia , Terapia de Reposição Hormonal , Inflamação/imunologia , Pulmão/imunologia , Animais , Reabsorção Óssea/tratamento farmacológico , Estradiol/uso terapêutico , Feminino , Inflamação/tratamento farmacológico , Exposição por Inalação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Sexo , Tomografia Computadorizada por Raios X
10.
Endocrinology ; 148(4): 1707-16, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17194746

RESUMO

The role of E2 on primordial follicle formation was examined by treating neonatal hamsters with 1 or 2 microg estradiol cypionate (ECP) at age postnatal d 1 (P1) and P4 or by in vitro culture of embryonic d 15 (E15) ovaries with 1, 5, or 10 ng/ml estradiol-17beta (E2). The specificity of E2 action was examined by ICI 182,780. One microgram of ECP maintained serum levels of E2 within the physiological range, significantly reduced apoptosis, and stimulated the formation and development of primordial follicles. In contrast, 2 microg ECP increased serum E2 levels to 400 pg/ml and had significantly less influence on primordial follicle formation. In vivo, ICI 182,780 significantly increased apoptosis and caused a modest reduction in primordial follicle formation. The formation and development of primordial follicles in vitro increased markedly with 1 ng/ml E2, and the effect was blocked by ICI 182,780. Higher doses of E2 had no effect on primordial follicle formation but significantly up-regulated apoptosis, which was blocked by ICI 182,780. CYP19A1 mRNA expression occurred by E13 and increased with the formation of primordial follicles. P4 ovaries synthesized E2 from testosterone, which increased further by FSH. Both testosterone and FSH maintained ovarian CYP19A1 mRNA, but FSH up-regulated the expression. These results suggest that neonatal hamster ovaries produce E2 under FSH control and that E2 action is essential for the survival and differentiation of somatic cells and the oocytes leading to the formation and development of primordial follicles. This supportive action of E2 is lost when hormone levels increase above a threshold.


Assuntos
Estradiol/farmacologia , Folículo Ovariano/embriologia , Animais , Animais Recém-Nascidos , Aromatase/genética , Aromatase/metabolismo , Células Cultivadas , Cricetinae , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Morfogênese/efeitos dos fármacos , Morfogênese/genética , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Gravidez , RNA Mensageiro/metabolismo
11.
Endocrinology ; 148(10): 4853-64, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17640985

RESUMO

The nongenomic actions of estradiol-17beta are mediated by transmembrane estrogen receptors. Recently, G protein-coupled receptor 30 (GPR30) has been suggested to be a transmembrane estrogen receptor that can mediate rapid and transcription-independent estradiol-17beta signaling in different cell types. However, the expression, regulation, or biological relevance of GPR30 in the ovary remains unknown. We examined the expression and hormonal regulation of GPR30 mRNA and protein in hamster ovarian cells during the estrous cycle and after hypophysectomy and hormone replacement. GPR30 protein expression was high in the theca, appreciable in the granulosa, but low in luteal cells. GPR30 protein levels in granulosa and theca cells increased steadily with the development of preantral and antral follicles, respectively. GPR30 mRNA and protein levels increased significantly on diestrous (d 3 of the estrous cycle), but decreased on d 4 at 1600 h after the LH surge. GPR30 mRNA levels increased significantly after hypophysectomy. Although steroid treatment failed to alter ovarian GPR30 mRNA levels, either FSH or LH effectively reduced the levels. Interestingly, the decrease in GPR30 mRNA corresponded to a marked increase in the receptor protein levels. FSH treatment, either alone or together with LH, resulted in a marked increase in GPR30 immunostaining in granulosa cells. LH alone significantly increased immunostaining in theca cells. These results suggest that GPR30 is expressed in the membrane of hamster granulosa and theca cells, and the expression is regulated by gonadotropins. The unique pattern of GPR30 expression suggests that gonadotropin-regulated follicular cell functions may involve GPR30 activity.


Assuntos
Gonadotropinas/fisiologia , Hormônios/fisiologia , Ovário/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sequência de Aminoácidos , Animais , Clonagem Molecular , Cricetinae , DNA Complementar , Sinergismo Farmacológico , Estro/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Gonadotropinas/farmacologia , Hormônios/farmacologia , Hipofisectomia , Hormônio Luteinizante/metabolismo , Hormônio Luteinizante/farmacologia , Mesocricetus , Dados de Sequência Molecular , Ovário/fisiologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Distribuição Tecidual
13.
Sci Rep ; 7(1): 15581, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29138526

RESUMO

Primordial follicle (PF) pool determines the availability of follicles for ovulation in all mammals. Premature depletion of the PF reserve leads to subfertility or infertility. Bone morphogenetic protein 2 (BMP2) promotes PF formation by facilitating oocyte and granulosa cell development. Estradiol-17ß (E2) upregulates PF formation in developing hamster ovaries. However, if BMP2 mediates E2 effect is not known. We hypothesize that E2 facilitates the effect of BMP2 on somatic to granulosa cell transition. BMP2 and E2 together significantly upregulated the percentage of PFs in hamster fetal ovaries in vitro compared with either of the treatments alone. E2 also promoted BMP2 expression in vivo. Inhibition of BMP2 receptors suppressed E2-stimulation of PF formation while knockdown of BMP2 in vitro significantly suppressed the E2 effect. In contrast, estrogen receptor blocker did not affect BMP2 action. Inhibition of the activity of E2 or BMP2 receptors, either alone or combined during the last two days of the culture (C6-C8) resulted in a significant decrease in PF formation by C8, suggesting that both BMP2 and E2 action is essential for somatic cell differentiation for PF formation. Together, the results suggest that E2 activates BMP2-BMPR system leading to the formation of primordial follicles.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular/genética , Estradiol/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Animais , Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/genética , Diferenciação Celular/efeitos dos fármacos , Cricetinae , Estradiol/biossíntese , Antagonistas de Estrogênios/administração & dosagem , Antagonistas do Receptor de Estrogênio/administração & dosagem , Feminino , Feto , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células da Granulosa/metabolismo , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Ovulação/genética , Ovulação/metabolismo
14.
Endocrinology ; 147(4): 1725-34, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16384866

RESUMO

Postnatal growth differentiation factor 9 (GDF-9) expression in the hamster oocytes precedes the formation of primordial follicles. We examined the functional significance of GDF-9 in primordial folliculogenesis in the hamster ovary using RNA interference knockdown of GDF-9 mRNA and protein expression. Fifteen-day-old fetal ovaries were cultured for 9 d with or without 1 ng FSH, 1 microl Metafectane, 100 nM control nontargeting small interfering RNA (siRNA), GDF-9 siRNA, or GDF-9 siRNA + FSH, and the development of primordial follicles examined. The efficiency of siRNA transfecting ovarian cells in the organ culture was tested by culturing ovaries with siGlo, a nontargeting control siRNA labeled with Cy3. More than 90% of cells in the ovary were siGlo positive, and neither the Metafectane nor the siRNA-induced cellular apoptosis. Control siRNA did not affect the basal levels of GDF-9 mRNA, but GDF-9 siRNA slightly but significantly reduced the level. FSH markedly up-regulated the levels of GDF-9 mRNA and protein, and the effect was completely suppressed by GDF-9 siRNA. However, GDF-9 siRNA did not affect the levels of bone morphogenetic protein receptor IA or beta-actin mRNA. GDF-9 siRNA alone also reduced GDF-9 protein expression. Concurrent with GDF-9 expression, FSH significantly augmented primordial follicle formation, but the effect was abolished by GDF-9 siRNA. These results suggest that endogenous GDF-9 plays an important role in somatic cell differentiation and the formation of primordial follicles. Furthermore, FSH, by virtue of regulating GDF-9 expression, modulates oocyte regulation of primordial follicles formation.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Oócitos/metabolismo , Folículo Ovariano/embriologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Cricetinae , Feminino , Hormônio Foliculoestimulante/farmacologia , Fator 9 de Diferenciação de Crescimento , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Mesocricetus , Dados de Sequência Molecular , Folículo Ovariano/fisiologia , RNA Interferente Pequeno/farmacologia
15.
Endocrinology ; 146(7): 3185-93, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15802500

RESUMO

Exposure to testosterone (T) during d 30-90 of fetal life results in low-birth-weight offspring, hypergonadotropism, multifollicular ovaries, and early cessation of cyclicity. The multifollicular phenotype may result from failure of follicles to regress and consequent follicular persistence or, alternatively, increased follicular recruitment. We tested the hypothesis that prenatal exposure to excess T causes intrauterine growth retardation and increases ovarian follicular recruitment. Time-mated pregnant ewes were treated with 100 mg T propionate in cottonseed oil or vehicle twice weekly from d 30-90 of gestation. Ewes were euthanized near term, from d 139-141 of gestation (term is 147 d). After determining fetal measures and organ weights, ovaries were removed from fetuses of control and T-treated dams, and follicular distribution in each ovary was determined by morphometric quantification. Total number and percentage distribution of the various classes of follicles (primordial, primary, preantral, and antral follicles) were compared between treatment groups. Prenatally T-treated female fetuses were smaller in size, had an increased head circumference to fetal weight ratio (P < 0.01), increased adrenal to fetal weight ratio (P < 0.05), decreased number of follicles (P < 0.05), a decrease in percentage of primordial follicles (P < 0.001), and a corresponding increase in the remaining classes of follicles (P < 0.05). Ovarian findings support decreased ovarian reserve and enhanced follicular recruitment, potential contributors of early reproductive failure. The extent to which metabolic changes associated with intrauterine growth retardation contribute toward altered trajectory of ovarian folliculogenesis remains to be determined.


Assuntos
Androgênios/farmacologia , Retardo do Crescimento Fetal/induzido quimicamente , Folículo Ovariano/efeitos dos fármacos , Ovário/efeitos dos fármacos , Testosterona/farmacologia , Animais , Feminino , Peso Fetal/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Folículo Ovariano/patologia , Folículo Ovariano/fisiologia , Ovário/patologia , Ovário/fisiologia , Gravidez , Ovinos
16.
Mol Cell Endocrinol ; 400: 1-9, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25462584

RESUMO

The usefulness of azaline B, a GnRH antagonist, in suppressing gonadotropin secretion in the golden hamster was examined by examining follicular development, steroidogenesis and expression of steroidogenic enzymes. Serum levels of P and E declined significantly, while FSH or LH was undetectable in azaline B-treated hamsters. FSH significantly increased serum E levels, whereas LH upregulated serum P levels. The formation of antral follicles ceased in azaline-treated hamsters, but was reversed by FSH with or without LH supplement. FSH also activated the primordial follicle pool resulting in increased formation of primary and preantral follicles. Further, an increasing trend in the formation of preantral follicles in response to E or E + P, and the formation of antral follicles in response to E + P treatment was evident. The level of Cyp11a1 mRNA increased markedly in LH- or LH + FSH-treated hamsters, whereas FSH with or without LH upregulated Cyp17a1, Cyp19a1 and Fshr mRNA expression. E without or with P also upregulated ovarian Cyp19a1 mRNA expression. The expression of enzyme protein corroborated the mRNA data. In summary, azaline B is an efficient GnRH antagonist in the hamster, and will be useful in studying the selective effect of gonadotropins on ovarian functions without disrupting the physiological functions of other hormones in ovarian cells.


Assuntos
Ciclo Estral/efeitos dos fármacos , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Luteinizante/metabolismo , Folículo Ovariano/efeitos dos fármacos , Animais , Aromatase/genética , Aromatase/metabolismo , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Estradiol/sangue , Estradiol/farmacologia , Ciclo Estral/fisiologia , Feminino , Hormônio Foliculoestimulante/genética , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Injeções Subcutâneas , Hormônio Luteinizante/genética , Hormônio Luteinizante/farmacologia , Mesocricetus , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Progesterona/sangue , Progesterona/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do FSH/genética , Receptores do FSH/metabolismo , Transdução de Sinais
17.
Mol Cell Endocrinol ; 400: 41-7, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25462586

RESUMO

FSH plays an important role in ovarian follicular development, and it functions via the G-protein coupled FSH receptor. The objectives of the present study were to determine if full-length FSHR mRNA and corresponding protein were expressed in fetal through postnatal hamster ovaries to explain the FSH-induced primordial follicle formation, and if FSH or estrogen (E) would affect the expression. A full-length and two alternately spliced FSHR transcripts were expressed from E14 through P20. The level of the full-length FSHR mRNA increased markedly through P7 before stabilizing at a lower level with the formation and activation of primordial follicles. A predicted 87 kDa FSHR protein band was detected in fetal through P4 ovaries, but additional bands appeared as ovary developed. FSHR immunosignal was present in undifferentiated somatic cells and oocytes in early postnatal ovaries, but was granulosa cells specific after follicles formed. Both eCG and E significantly up-regulated full-length FSHR mRNA levels. Therefore, FSHR is expressed in the hamster ovary from the fetal life to account for FSH-induced primordial follicle formation and cAMP production. Further, FSH or E regulates the receptor expression.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Folículo Ovariano/metabolismo , RNA Mensageiro/genética , Receptores do FSH/genética , Processamento Alternativo , Animais , Animais Recém-Nascidos , Gonadotropina Coriônica/farmacologia , AMP Cíclico/biossíntese , Embrião de Mamíferos , Estradiol/farmacologia , Ciclo Estral/fisiologia , Feminino , Feto , Hormônio Foliculoestimulante/farmacologia , Injeções Subcutâneas , Mesocricetus , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Receptores do FSH/metabolismo , Transdução de Sinais
18.
Sci Rep ; 5: 12664, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26219655

RESUMO

Primordial follicles (PF) are formed when somatic cells differentiate into flattened pregranulosa cells, invaginate into the oocyte nests and encircle individual oocytes. We hypothesize that BMP2 regulates PF formation by promoting the transition of germ cells into oocytes and somatic cells into pregranulosa cells. E15 hamster ovaries were cultured for 8 days corresponding to postnatal day 8 (P8) in vivo, with or without BMP2, and the formation of PF was examined. BMP2 was expressed in the oocytes as well as ovarian somatic cells during development. BMP2 exposure for the first two days or the last two days or the entire 8 days of culture led to increase in PF formation suggesting that BMP2 affected both germ cell transition and somatic cell differentiation. Whereas an ALK2/3 inhibitor completely blocked BMP2-induced PF formation, an ALK2-specific inhibitor was partially effective, suggesting that BMP2 affected PF formation via both ALK2 and ALK3. BMP2 also reduced apoptosis in vitro. Further, more meiotic oocytes were present in BMP2 exposed ovaries. In summary, the results provide the first evidence that BMP2 regulates primordial follicle formation by promoting germ cell to oocyte transition and somatic cell to pre-granulosa cells formation and it acts via both ALK2 and ALK3.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Ovário/efeitos dos fármacos , Receptores de Ativinas Tipo I/antagonistas & inibidores , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Animais , Animais Recém-Nascidos , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Cricetinae , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Masculino , Mesocricetus , Microscopia Confocal , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Folículo Ovariano/citologia , Folículo Ovariano/embriologia , Ovário/citologia , Ovário/embriologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Quinolinas/farmacologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
19.
Endocrinology ; 143(6): 2385-98, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021204

RESUMO

Spatiotemporal expression patterns of ER-alpha and ER-beta protein and mRNA in hamster ovarian cells during the estrous cycle and following hypophysectomy and selective hormone replacement were evaluated by immunofluorescence, immunoblotting and in situ hybridization analyses. Whereas ER-beta mRNA and protein expression predominated in granulosa cells and ER-alpha expression was in interstitial and thecal cells, overlap in receptor subtype expression across cell types was evident. Both ER subtypes were present from primordial follicle stage onward. ER-alpha mRNA levels and immunoreactivity started increasing from D3:0900 h in interstitial and granulosa cells and peaked on the proestrous (D4:0900 h). Regionalized higher expression of ER-alpha in granulosa cells in and around the forming antrum was evident. Surface epithelial cells were also positive. ER-beta mRNA and protein expression increased markedly in granulosa and interstitial cells on D2:0900 h, reached a peak on D3:0900 h, and then declined sharply on D4:0900 h. No change in ER expression occurred following the preovulatory gonadotropin surge. Whereas FSH or human CG stimulated ER-alpha mRNA and protein expression in hypophysectomized hamsters, only FSH could stimulate ER-beta mRNA and protein, and the effect was significantly attenuated by human CG. ER expression was stimulated by estrogen, but progesterone strongly inhibited estrogen action. These results indicate that ER expression is cell type specific to the larger extent and is critically regulated by reproductive hormones.


Assuntos
Hormônios Esteroides Gonadais/farmacologia , Gonadotropinas/farmacologia , Ovário/fisiologia , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética , Esteroides/farmacologia , Animais , Western Blotting , Clonagem Molecular , Cricetinae , DNA Complementar/biossíntese , DNA Complementar/genética , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Hipofisectomia , Hibridização In Situ , Mesocricetus , Ovário/citologia , Ovário/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Endocrinology ; 145(12): 5757-66, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15345677

RESUMO

Perinatal expression of estrogen receptor (ER) protein and mRNA and the influence of FSH on this process were examined by immunofluorescence and RT-PCR using ovaries from fetal (d 13-15 of gestation) and postnatal [postnatal d 1-15 (P1-P15)] hamsters and from 8-d-old hamsters exposed in utero to an anti-FSH serum on d 12 of gestation and saline or equine chorionic gonadotropin (eCG) on P1. A few somatic cells expressing ERalpha immunoreactivity appeared first on d 14 of gestation and increased markedly by P8-P15 in the interstitial cells and granulosa cells of primordial follicles. In contrast, appreciable ERbeta immunoreactivity was localized on d 13 of gestation, and more cells expressed ERbeta immunoreactivity by P1-P8. By P7, ERbeta immunoreactivity was present in cells adjacent to the oocytes, and by P8, ERbeta was preferentially localized in the granulosa cells. Receptor immunoreactivities decreased markedly in P8 ovaries exposed in utero to the FSH antiserum but were reversed with postnatal eCG replacement. Oocytes and somatic cells expressed ERalpha and ERbeta mRNA, and levels of ER mRNA in the ovary increased by P7-P8, corresponding to the appearance of primordial follicles. Thereafter, only ERbeta mRNA levels increased progressively with postnatal ovary development. Similar to ER protein, mRNA levels decreased significantly in FSH antiserum-treated ovaries but were restored by eCG. These results indicate that both ER subtypes are expressed in undifferentiated somatic cells and the oocytes during perinatal ovary development in the hamster; however, ERbeta expression segregates with the differentiation of granulosa cells. Furthermore, ER expression and differentiation of somatic cells to granulosa cells depend on perinatal FSH action.


Assuntos
Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Hormônio Foliculoestimulante/fisiologia , Ovário/fisiologia , Animais , Cricetinae , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Imunofluorescência , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Oócitos/fisiologia , Ovário/embriologia , Ovário/crescimento & desenvolvimento , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA