Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cell Endocrinol ; 526: 111211, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33582214

RESUMO

Follicle stimulating hormone (FSH), composed of FSHα and FSHß subunits, is essential for female follicle development and male spermatogenesis. The recombinant human FSH (rhFSH) products on the market are mainly generated from mammalian cells and are expensive. Large animal mammary gland bioreactors are urgently needed to produce large amounts of rhFSH. However, there are currently no effective methods to prepare rhFSH by large animals mainly due to the fact that excessive accumulation of FSH might cause many adverse effects in animals. We herein report the development and characterization of functional self-assembled rhFSH produced in goat mammary epithelial cells (GMECs). FSHα and FSHß stably expressed in Chinese hamster ovary (CHO) cell lines were secreted into culture medium and well glycosylated. Importantly, FSHα and FSHß expressed apart were able to assemble into functional FSH. We next inserted human FSHα or FSHß gene separately into goat ß-Lactoglobulin locus in GMECs by CRISPR/Cas9. Inactive FSHα and FSHß subunits expressed from GMECs assembled into rhFSH as analyzed by His-tag pull down assay. Functional assessment of rhFSH by cAMP induction assay, mouse ovulation induction and rat ovarian weight gain experiments showed that the bioactivity of self-assembled rhFSH expressed by GMECs was comparable to that of Gonal-F both in vitro and in vivo. Our study demonstrated that FSHα and FSHß can be separately expressed and assembled into functional rhFSH, and provided the basis for future preparing FSH by goat mammary gland bioreactor with less health problems on the producing animals.


Assuntos
Células Epiteliais/metabolismo , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade alfa de Hormônios Glicoproteicos/biossíntese , Cabras/fisiologia , Glândulas Mamárias Animais/citologia , Proteínas Recombinantes/biossíntese , Animais , Aromatase/genética , Aromatase/metabolismo , Sequência de Bases , Células CHO , Sistemas CRISPR-Cas/genética , Cricetulus , AMP Cíclico/metabolismo , Endocitose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Estradiol/sangue , Feminino , Subunidade beta do Hormônio Folículoestimulante/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Subunidade alfa de Hormônios Glicoproteicos/farmacologia , Glicosilação , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Lactoglobulinas/genética , Ligantes , Camundongos , Ácido N-Acetilneuramínico/metabolismo , Ovulação/efeitos dos fármacos , Subunidades Proteicas/farmacologia , RNA Guia de Cinetoplastídeos/metabolismo , Ratos , Proteínas Recombinantes/farmacologia , Aumento de Peso/efeitos dos fármacos
2.
Biomed Pharmacother ; 102: 494-501, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29579710

RESUMO

Luteal phase defects (LPD) are an important etiology of infertility which has increased in recent years. Studies have shown that bu-shen-zhu-yun decoction (BSZY-D) can lower the expression of estrogen receptor and progesterone receptor, in rats endometrium of embryonic implantation period, which upregulated by mifepristone, and improve uterine receptivity. The aim of present study was to determine the effect of BSZY-D on the synthesis and secretion of gonadotropic hormones in the anterior pituitary cells of rats. Rats were treated with saline (control) or BSZY-D two times/day for three estrous cycles by gavage. The cerebrospinal fluid (CSF) were collected for further cell treatment. The components in BSZY-D, serum and CSF were analysed by High Performance Liquid Chromatography (HPLC). Cells were either pretreated with normal CSF or BSZY-D/CSF before being stimulated with or without cetrorelix. The mRNA and proteins levels of receptors, hormones, and transcription factors were detected by RT-PCR, western blot analysis and immunostaining. We show that non-toxic concentrations of cetrorelix, a GnRH antagonist, can reduce the mRNA and protein levels of GnRHR, LH, and FSH. This effect could be reversed by the addition of BSZY-D/CSF. We also show decreased mRNA and protein expression of transcription factors, such as CREB, and Egr-1 and secretory vescicles, including SNAP-25 and Munc-18 upon treatment with cetrorelix could be reversed post co-treatment with BSZY-D/CSF. These results indicate that BSZY-D/CSF treatment led to increased levels of GnRHR, transcription factors, and secretory vesicles leading to increased secretion of FSH and LH. Thus, BSZY-D presents a promising candidate to treat luteal phase defects and infertility.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hormônio Luteinizante Subunidade beta/biossíntese , Hormônio Luteinizante Subunidade beta/metabolismo , Adeno-Hipófise/citologia , Animais , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cromatografia Líquida de Alta Pressão , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Proteínas Munc18/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores LHRH/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos
3.
J Biol Chem ; 292(23): 9815-9829, 2017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-28385888

RESUMO

Neuroendocrine control of reproduction by brain-secreted pulses of gonadotropin-releasing hormone (GnRH) represents a longstanding puzzle about extracellular signal decoding mechanisms. GnRH regulates the pituitary gonadotropin's follicle-stimulating hormone (FSH) and luteinizing hormone (LH), both of which are heterodimers specified by unique ß subunits (FSHß/LHß). Contrary to Lhb, Fshb gene induction has a preference for low-frequency GnRH pulses. To clarify the underlying regulatory mechanisms, we developed three biologically anchored mathematical models: 1) parallel activation of Fshb inhibitory factors (e.g. inhibin α and VGF nerve growth factor-inducible), 2) activation of a signaling component with a refractory period (e.g. G protein), and 3) inactivation of a factor needed for Fshb induction (e.g. growth differentiation factor 9). Simulations with all three models recapitulated the Fshb expression levels obtained in pituitary gonadotrope cells perifused with varying GnRH pulse frequencies. Notably, simulations altering average concentration, pulse duration, and pulse frequency revealed that the apparent frequency-dependent pattern of Fshb expression in model 1 actually resulted from variations in average GnRH concentration. In contrast, models 2 and 3 showed "true" pulse frequency sensing. To resolve which components of this GnRH signal induce Fshb, we developed a high-throughput parallel experimental system. We analyzed over 4,000 samples in experiments with varying near-physiological GnRH concentrations and pulse patterns. Whereas Egr1 and Fos genes responded only to variations in average GnRH concentration, Fshb levels were sensitive to both average concentration and true pulse frequency. These results provide a foundation for understanding the role of multiple regulatory factors in modulating Fshb gene activity.


Assuntos
Simulação por Computador , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/biossíntese , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Humanos , Hormônio Luteinizante Subunidade beta/biossíntese , Modelos Biológicos , Proteínas Proto-Oncogênicas c-fos/metabolismo
4.
J Endocrinol ; 229(3): 331-41, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27029473

RESUMO

Pituitary follicle-stimulating hormone (FSH) synthesis is regulated by transforming growth factorßsuperfamily ligands, most notably the activins and inhibins. Bone morphogenetic proteins (BMPs) also regulate FSHß subunit (Fshb) expression in immortalized murine gonadotrope-like LßT2 cells and in primary murine or ovine primary pituitary cultures. BMP2 signals preferentially via the BMP type I receptor, BMPR1A, to stimulate murine Fshb transcription in vitro Here, we used a Cre-lox approach to assess BMPR1A's role in FSH synthesis in mice in vivo Gonadotrope-specific Bmpr1a knockout animals developed normally and had reproductive organ weights comparable with those of controls. Knockouts were fertile, with normal serum gonadotropins and pituitary gonadotropin subunit mRNA expression. Cre-mediated recombination of the floxed Bmpr1a allele was efficient and specific, as indicated by PCR analysis of diverse tissues and isolated gonadotrope cells. Furthermore, BMP2 stimulation of inhibitor of DNA binding 3 expression was impaired in gonadotropes isolated from Bmpr1a knockout mice, confirming the loss of functional receptor protein in these cells. Treatment of purified gonadotropes with small-molecule inhibitors of BMPR1A (and the related receptors BMPR1B and ACVR1) suppressed Fshb mRNA expression, suggesting that an autocrine BMP-like molecule might regulate FSH synthesis. However, deletion of Bmpr1a and Acvr1 in cultured pituitary cells did not alter Fshb expression, indicating that the inhibitors had off-target effects. In sum, BMPs or related ligands acting via BMPR1A or ACVR1 are unlikely to play direct physiological roles in FSH synthesis by murine gonadotrope cells.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Fertilidade/fisiologia , Gonadotrofos/fisiologia , Gonadotropinas Hipofisárias/biossíntese , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/fisiologia , Animais , 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/fisiologia , Células Cultivadas , Feminino , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/genética , Gonadotrofos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
J Mol Endocrinol ; 55(2): 95-106, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26162607

RESUMO

Neuromedin U (NMU) and neuromedin S (NMS) play inhibitory roles in the regulation of food intake and energy homeostasis in mammals. However, their functions are not clearly established in teleost fish. In the present study, nmu and nms homologs were identified in several fish species. Subsequently, their cDNA sequences were cloned from the orange-spotted grouper (Epinephelus coioides). Sequence analysis showed that the orange-spotted grouper Nmu proprotein contains a 21-amino acid mature Nmu peptide (Nmu-21). The Nms proprotein lost the typical mature Nms peptide, but it retains a putative 34-amino acid peptide (Nmsrp). In situ hybridization revealed that nmu- and nms-expressing cells are mainly localized in the hypothalamic regions associated with appetite regulation. Food deprivation decreased the hypothalamic nmu mRNA levels but induced an increase of nms mRNA levels. Periprandial expression analysis showed that hypothalamic expression of nmu increased significantly at 3 h post-feeding, while nms expression was elevated at the normal feeding time. I.p. injection of synthetic Nmu-21 peptide suppressed the hypothalamic neuropeptide y (npy) expression, while Nmsrp administration significantly increased the expression of npy and orexin in orange-spotted grouper. Furthermore, the mRNA levels of LH beta subunit (lhß) and gh in the pituitary were significantly down-regulated after Nmu-21 peptide administration, while Nmsrp was able to significantly stimulate the expression of FSH beta subunit (fshß), prolactin (prl), and somatolaction (sl). Our results indicate that nmu and nms possess distinct neuroendocrine functions and pituitary functions in the orange spotted grouper.


Assuntos
Bass/genética , Metabolismo Energético/genética , Proteínas de Peixes/genética , Neuropeptídeo Y/genética , Neuropeptídeos/genética , Sequência de Aminoácidos , Animais , Apetite/genética , Sequência de Bases , Clonagem Molecular , Ingestão de Alimentos/genética , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Hipotálamo/citologia , Hipotálamo/metabolismo , Hibridização In Situ , Hormônio Luteinizante Subunidade beta/genética , Dados de Sequência Molecular , Neuropeptídeo Y/biossíntese , Neuropeptídeos/biossíntese , Orexinas/biossíntese , Hipófise/metabolismo , Prolactina/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Análise de Sequência de DNA , Inanição/genética
6.
J Biol Chem ; 289(23): 16164-75, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24778184

RESUMO

Gonadotropin-releasing hormone (GnRH) is secreted in brief pulses from the hypothalamus and regulates follicle-stimulating hormone ß-subunit (FSHß) gene expression in pituitary gonadotropes in a frequency-sensitive manner. The mechanisms underlying its preferential and paradoxical induction of FSHß by low frequency GnRH pulses are incompletely understood. Here, we identify growth differentiation factor 9 (GDF9) as a GnRH-suppressed autocrine inducer of FSHß gene expression. GDF9 gene transcription and expression were preferentially decreased by high frequency GnRH pulses. GnRH regulation of GDF9 was concentration-dependent and involved ERK and PKA. GDF9 knockdown or immunoneutralization reduced FSHß mRNA expression. Conversely, exogenous GDF9 induced FSHß expression in immortalized gonadotropes and in mouse primary pituitary cells. GDF9 exposure increased FSH secretion in rat primary pituitary cells. GDF9 induced Smad2/3 phosphorylation, which was impeded by ALK5 knockdown and by activin receptor-like kinase (ALK) receptor inhibitor SB-505124, which also suppressed FSHß expression. Smad2/3 knockdown indicated that FSHß induction by GDF9 involved Smad2 and Smad3. FSHß mRNA induction by GDF9 and GnRH was synergistic. We hypothesized that GDF9 contributes to a regulatory loop that tunes the GnRH frequency-response characteristics of the FSHß gene. To test this, we determined the effects of GDF9 knockdown on FSHß induction at different GnRH pulse frequencies using a parallel perifusion system. Reduction of GDF9 shifted the characteristic pattern of GnRH pulse frequency sensitivity. These results identify GDF9 as contributing to an incoherent feed-forward loop, comprising both intracellular and secreted components, that regulates FSHß expression in response to activation of cell surface GnRH receptors.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/genética , Regulação da Expressão Gênica/fisiologia , Fator 9 de Diferenciação de Crescimento/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Primers do DNA , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Hormônio Liberador de Gonadotropina/fisiologia , Fator 9 de Diferenciação de Crescimento/genética , Masculino , Camundongos , Hipófise/citologia , Hipófise/metabolismo , RNA Interferente Pequeno , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Proteínas Smad/genética , Proteínas Smad/metabolismo , Transcrição Gênica
7.
Endocrinology ; 155(5): 1970-81, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24601881

RESUMO

FSH is an essential regulator of mammalian reproduction. Its synthesis by pituitary gonadotrope cells is regulated by multiple endocrine and paracrine factors, including TGFß superfamily ligands, such as the activins and inhibins. Activins stimulate FSH synthesis via transcriptional regulation of its ß-subunit gene (Fshb). More recently, bone morphogenetic proteins (BMPs) were shown to stimulate murine Fshb transcription alone and in synergy with activins. BMP2 signals via its canonical type I receptor, BMPR1A (or activin receptor-like kinase 3 [ALK3]), and SMAD1 and SMAD5 to stimulate transcription of inhibitor of DNA binding proteins. Inhibitor of DNA binding proteins then potentiate the actions of activin-stimulated SMAD3 to regulate the Fshb gene in the gonadotrope-like LßT2 cell line. Here, we report the unexpected observation that BMP2 also stimulates the SMAD2/3 pathway in these cells and that it does so directly via ALK3. Indeed, this novel, noncanonical ALK3 activity is completely independent of ALK4, ALK5, and ALK7, the type I receptors most often associated with SMAD2/3 pathway activation. Induction of the SMAD2/3 pathway by ALK3 is dependent upon its own previous activation by associated type II receptors, which phosphorylate conserved serine and threonine residues in the ALK3 juxtamembrane glycine-serine-rich domain. ALK3 signaling via SMAD3 is necessary for the receptor to stimulate Fshb transcription, whereas its activation of the SMAD1/5/8 pathway alone is insufficient. These data challenge current dogma that ALK3 and other BMP type I receptors signal via SMAD1, SMAD5, and SMAD8 and not SMAD2 or SMAD3. Moreover, they suggest that BMPs and activins may use similar intracellular signaling mechanisms to activate the murine Fshb promoter in immortalized gonadotrope-like cells.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Gonadotrofos/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Ativinas/antagonistas & inibidores , Ativinas/metabolismo , Animais , Proteína Morfogenética Óssea 2/agonistas , Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/agonistas , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Linhagem Celular , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Inativação Gênica , Genes Reporter , Humanos , Camundongos , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Interferente Pequeno , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteína Smad2/antagonistas & inibidores , Proteína Smad2/genética , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/genética , Transcrição Gênica
8.
Endocrinology ; 155(5): 1874-86, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24484170

RESUMO

Kisspeptin regulates reproductive events, including puberty and ovulation, primarily via GnRH neurons. Prolonged treatment of prepubertal striped bass females with kisspeptin (Kiss) 1 or Kiss2 peptides failed to enhance puberty but suggested a gnrh-independent pituitary control pathway. Kiss2 inhibited, but Kiss1 stimulated, FShß expression and gonadal development, although hypophysiotropic gnrh1 and gnrh receptor expression remained unchanged. In situ hybridization and immunohistochemistry on brains and pituitaries revealed a differential plasticity between the 2 kisspeptin neurons. The differences were most pronounced at the prespawning phase in 2 regions along the path of gnrh1 axons: the nucleus lateralis tuberis (NLT) and the neurohypophysis. Kiss1 neurons appeared in the NLT and innervated the neurohypophysis of prespawning males and females, reaching Lh gonadotropes in the proximal pars distalis. Males, at all reproductive stages, had Kiss2 innervations in the NLT and the neurohypophysis, forming large axonal bundles in the former and intermingling with gnrh1 axons. Unlike in males, only preovulatory females had massive NLT-neurohypophysis staining of kiss2. Kiss2 neurons showed a distinct appearance in the NLT pars ventralis-equivalent region only in spawning zebrafish, indicating that this phenomenon is widespread. These results underscore the NLT as important nuclei for kisspeptin action in 2 facets: 1) kisspeptin-gnrh interaction, both kisspeptins are involved in the regulation of gnrh release, in a stage- and sex-dependent manner, especially at the prespawning phase; and 2) gnrh-independent effect of Kiss peptides on the pituitary, which together with the plastic nature of their neuronal projections to the pituitary implies that a direct gonadotropic regulation is plausible.


Assuntos
Bass/fisiologia , Proteínas de Peixes/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Kisspeptinas/metabolismo , Maturidade Sexual , Proteínas de Xenopus/metabolismo , Animais , Aquicultura , Axônios/efeitos dos fármacos , Axônios/metabolismo , Relação Dose-Resposta a Droga , Implantes de Medicamento , Feminino , Fármacos para a Fertilidade Feminina/farmacologia , Proteínas de Peixes/biossíntese , Proteínas de Peixes/genética , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/genética , Sistema Hipotálamo-Hipofisário/citologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Hipotálamo Médio/citologia , Hipotálamo Médio/efeitos dos fármacos , Hipotálamo Médio/crescimento & desenvolvimento , Hipotálamo Médio/metabolismo , Kisspeptinas/administração & dosagem , Kisspeptinas/farmacologia , Maryland , Neuro-Hipófise/citologia , Neuro-Hipófise/efeitos dos fármacos , Neuro-Hipófise/crescimento & desenvolvimento , Neuro-Hipófise/metabolismo , Maturidade Sexual/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas de Xenopus/administração & dosagem , Proteínas de Xenopus/farmacologia
9.
Mol Endocrinol ; 27(8): 1283-94, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23770611

RESUMO

Kisspeptin signaling through its receptor, Kiss1R, is crucial for many reproductive functions including puberty, sex steroid feedback, and overall fertility. Although the importance of Kiss1R in the brain is firmly established, its role in regulating reproduction at the level of the pituitary is not well understood. This study presents molecular analysis of the role of kisspeptin and Kiss1R signaling in the transcriptional regulation of the gonadotropin gene ß-subunits, LHß and FSHß, using LßT2 gonadotrope cells and murine primary pituitary cells. We show that kisspeptin induces LHß and FSHß gene expression, and this induction is protein kinase C dependent and mediated by the immediate early genes, early growth response factor 1 and cFos, respectively. Additionally, kisspeptin induces transcription of the early growth response factor 1 and cFos promoters in LßT2 cells. Kisspeptin also increases gonadotropin gene expression in mouse primary pituitary cells in culture. Furthermore, we find that Kiss1r expression is enhanced in the pituitary of female mice during the estradiol-induced LH surge, a critical component of the reproductive cycle. Overall, our findings indicate that kisspeptin regulates gonadotropin gene expression through the activation of Kiss1R signaling through protein kinase C, inducing immediate early genes in vitro, and responds to physiologically relevant cues in vivo, suggesting that kisspeptin affects pituitary gene expression to regulate reproductive function.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/metabolismo , Gonadotrofos/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante Subunidade beta/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células Cultivadas , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Feminino , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/genética , Expressão Gênica , Regulação da Expressão Gênica , Genes Precoces/genética , Gonadotrofos/citologia , Hormônio Luteinizante Subunidade beta/biossíntese , Hormônio Luteinizante Subunidade beta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Receptores de Kisspeptina-1 , Reprodução/genética , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica
10.
Biol Reprod ; 88(3): 78, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23426431

RESUMO

Activins were discovered and, in fact, named more than a quarter century ago based on their abilities to stimulate pituitary follicle-stimulating hormone (FSH) synthesis and secretion. However, it is only in the last decade that we have finally come to understand their underlying mechanisms of action in gonadotroph cells. In this minireview, we chronicle the research that led to the recent discovery of forkhead box L2 (FOXL2) as an essential mediator of activin-regulated FSH beta subunit (Fshb) transcription in vitro and in vivo.


Assuntos
Ativinas/metabolismo , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Fatores de Transcrição Forkhead/metabolismo , Animais , Proteína Forkhead Box L2 , Regulação da Expressão Gênica , Humanos
11.
Mol Endocrinol ; 27(3): 407-21, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23340250

RESUMO

Impairments in pituitary FSH synthesis or action cause infertility. However, causes of FSH dysregulation are poorly described, in part because of our incomplete understanding of mechanisms controlling FSH synthesis. Previously, we discovered a critical role for forkhead protein L2 (FOXL2) in activin-stimulated FSH ß-subunit (Fshb) transcription in immortalized cells in vitro. Here, we tested the hypothesis that FOXL2 is required for FSH synthesis in vivo. Using a Cre/lox approach, we selectively ablated Foxl2 in murine anterior pituitary gonadotrope cells. Conditional knockout (cKO) mice developed overtly normally but were subfertile in adulthood. Testis size and spermatogenesis were significantly impaired in cKO males. cKO females exhibited reduced ovarian weight and ovulated fewer oocytes in natural estrous cycles compared with controls. In contrast, ovaries of juvenile cKO females showed normal responses to exogenous gonadotropin stimulation. Both male and female cKO mice were FSH deficient, secondary to diminished pituitary Fshb mRNA production. Basal and activin-stimulated Fshb expression was similarly impaired in Foxl2 depleted primary pituitary cultures. Collectively, these data definitively establish FOXL2 as the first identified gonadotrope-restricted transcription factor required for selective FSH synthesis in vivo.


Assuntos
Fertilidade , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Fatores de Transcrição Forkhead/deficiência , Gonadotrofos/metabolismo , Ativinas/farmacologia , Animais , Feminino , Subunidade beta do Hormônio Folículoestimulante/deficiência , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Loci Gênicos/genética , Gonadotrofos/efeitos dos fármacos , Gonadotropinas/sangue , Cavalos , Humanos , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Especificidade de Órgãos/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Ovulação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recombinação Genética/genética , Células de Sertoli/citologia , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Espermatogênese/efeitos dos fármacos , Testículo/citologia , Testículo/efeitos dos fármacos , Testículo/metabolismo
12.
Mol Endocrinol ; 27(2): 253-65, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23275456

RESUMO

GnRH is a potent hypothalamic regulator of gonadotropin hormones, LH and FSH, which are both expressed within the pituitary gonadotrope and are necessary for the stimulation of gametogenesis and steroidogenesis in the gonads. Differential regulation of LH and FSH, which is essential for reproductive fitness, is achieved, in part, through the varying of GnRH pulse frequency. However, the mechanism controlling the increase in FSH during the periods of low GnRH has not been elucidated. Here, we uncover another level of regulation by GnRH that contributes to differential expression of the gonadotropins and may play an important role for the generation of the secondary rise of FSH that stimulates folliculogenesis. GnRH stimulates LHß and FSHß subunit transcription via induction of the immediate early genes, Egr1 and c-Fos, respectively. Here, we determined that GnRH induces rapidly both Egr1 and c-Fos, but specifically decreases the rate of c-Fos degradation. In particular, GnRH modulates the rate of c-Fos protein turnover by inducing c-Fos phosphorylation through the ERK1/2 pathway. This extends the half-life of c-Fos, which is normally rapidly degraded. Confirming the role of phosphorylation in promoting increased protein activity, we show that a c-Fos mutant that cannot be phosphorylated by GnRH induces lower expression of the FHSß promoter than wild-type c-Fos. Our studies expand upon the role of GnRH in the regulation of gonadotropin gene expression by highlighting the role of c-Fos posttranslational modification that may cause higher levels of FSH during the time of low GnRH pulse frequency to stimulate follicular growth.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/biossíntese , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Linhagem Celular , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Hormônio Luteinizante Subunidade beta/biossíntese , Hormônio Luteinizante Subunidade beta/genética , Camundongos , Mutação , Folículo Ovariano/metabolismo , Fosforilação , Hipófise/metabolismo , Processamento de Proteína Pós-Traducional , Transcrição Gênica
13.
Mol Endocrinol ; 27(2): 224-37, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23211523

RESUMO

The regulation of gonadotropin synthesis by GnRH plays an essential role in the neuroendocrine control of reproduction. The known signaling mechanisms involved in gonadotropin synthesis have been expanding. For example, involvement of ß-catenin in LHß induction by GnRH has been discovered. We examined the role of ß-catenin in FSHß gene expression in LßT2 gonadotrope cells. GnRH caused a sustained increase in nuclear ß-catenin levels, which was significantly reduced by c-Jun N-terminal kinase (JNK) inhibition. Small interfering RNA-mediated knockdown of ß-catenin mRNA demonstrated that induction of FSHß mRNA by GnRH depended on ß-catenin and that regulation of FSHß by ß-catenin occurred independently of the JNK-c-jun pathway. ß-Catenin depletion had no impact on FSHß mRNA stability. In LßT2 cells transfected with FSHß promoter luciferase fusion constructs, GnRH responsiveness was conferred by the proximal promoter (-944/-1) and was markedly decreased by ß-catenin knockdown. However, none of the T-cell factor/lymphoid enhancer factor binding sites in that region were required for promoter activation by GnRH. Chromatin immunoprecipitation further corroborated the absence of direct interaction between ß-catenin and the 1.8-kb FSHß promoter. To elucidate the mechanism for the ß-catenin effect, we analyzed approximately 1 billion reads of next-generation RNA sequencing ß-catenin knockdown assays and selected the nuclear cofactor breast cancer metastasis-suppressor 1-like (Brms1L) as one candidate for further study. Subsequent experiments confirmed that Brms1L mRNA expression was decreased by ß-catenin knockdown as well as by JNK inhibition. Furthermore, knockdown of Brms1L significantly attenuated GnRH-induced FSHß expression. Thus, our findings indicate that the expression of Brms1L depends on ß-catenin activity and contributes to FSHß induction by GnRH.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/genética , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas Repressoras/biossíntese , beta Catenina/metabolismo , Animais , Antracenos/farmacologia , Sítios de Ligação , Linhagem Celular , Imunoprecipitação da Cromatina , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Expressão Gênica , Gonadotropinas/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Camundongos , Regiões Promotoras Genéticas , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição TCF/metabolismo , beta Catenina/genética
14.
Biol Reprod ; 86(5): 144, 1-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22357547

RESUMO

The hypothalamic-pituitary-gonadal axis is central to normal reproductive function. This pathway begins with the release of gonadotropin-releasing hormone in systematic pulses by the hypothalamus. Gonadotropin-releasing hormone is bound by receptors on gonadotroph cells in the anterior pituitary gland and stimulates the synthesis and secretion of luteinizing hormone and, to some extent, follicle-stimulating hormone. Once stimulated by these glycoprotein hormones, the gonads begin gametogenesis and the synthesis of sex hormones. In humans, mutations of the forkhead transcription factor, FOXP3, lead to an autoimmune disorder known as immunodysregulation, polyendocrinopathy, and enteropathy, X-linked syndrome. Mice with a mutation in the Foxp3 gene have a similar autoimmune syndrome and are infertile. To understand why FOXP3 is required for reproductive function, we are investigating the reproductive phenotype of Foxp3 mutant mice (Foxp3(sf/Y)). Although the gonadotroph cells appear to be intact in Foxp3(sf/Y) mice, luteinizing hormone beta (Lhb) and follicle-stimulating hormone beta (Fshb) expression are significantly decreased, demonstrating that these mice exhibit a hypogonadotropic hypogonadism. Hypothalamic expression of gonadotropin-releasing hormone is not significantly decreased in Foxp3(sf/Y) males. Treatment of Foxp3(sf/Y) males with a gonadotropin-releasing hormone receptor agonist does not rescue expression of Lhb or Fshb. Interestingly, we do not detect Foxp3 expression in the pituitary or hypothalamus, suggesting that the infertility seen in Foxp3(sf/Y) males is a secondary effect, possibly due to loss of FOXP3 in immune cells. Pituitary expression of glycoprotein hormone alpha (Cga) and prolactin (Prl) are significantly reduced in Foxp3(sf/Y) males, whereas the precursor for adrenocorticotropic hormone, pro-opiomelanocortin (Pomc), is increased. Human patients diagnosed with IPEX often exhibit thyroiditis due to destruction of the thyroid gland by autoimmune cells. We find that Foxp3(sf/Y) mice have elevated expression of thyroid-stimulating hormone beta (Tshb), suggesting that they may suffer from thyroiditis as well. Expression of the pituitary transcription factors, Pitx1, Pitx2, Lhx3, and Egr1, is normal; however, expression of Foxl2 and Gata2 is elevated. These data are the first to demonstrate a defect at the pituitary level in the absence of FOXP3, which contributes to the infertility observed in mice with Foxp3 loss of function mutations.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/biossíntese , Fatores de Transcrição Forkhead/metabolismo , Hormônio Luteinizante Subunidade beta/biossíntese , Hipófise/metabolismo , Animais , Fatores de Transcrição Forkhead/genética , Hormônio Liberador de Gonadotropina/biossíntese , Hipogonadismo/tratamento farmacológico , Hipogonadismo/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Hipófise/efeitos dos fármacos , Pró-Opiomelanocortina/biossíntese , Prolactina/biossíntese , Receptores LHRH/agonistas , Tireotropina Subunidade beta/biossíntese , Fatores de Transcrição/biossíntese
15.
J Reprod Dev ; 58(1): 147-55, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22134063

RESUMO

We cloned the LIM-homeodomain protein LHX2 as a transcription factor for the porcine follicle-stimulating hormone ß subunit gene (Fshß) by the Yeast One-Hybrid Cloning System using the upstream region of -852/-746 bases (b) from the transcription start site, called Fd2, as a bait sequence. The reporter assay in LßT2 and CHO cells revealed the presence of an LHX2-responsive region other than Fd2. A potential LHX2 binding sequence was confirmed as AATTAAT containing a consensus homeodomain binding core sequence AATT by Systematic Evolution of Ligands by Exponential Enrichment analysis. DNase I footprinting demonstrated three AATTAAT sequences located at regions -835/-829, -818/-812 and -806/-800 b in the Fd2 region and 12 binding sites in the distal and proximal regions mostly containing an AATT-core sequence. RT-PCR analysis of Lhx2 expression during porcine fetal and postnatal pituitary development showed a gradual increase from fetal day (f) 40 to postnatal day (p) 8 followed by a slight decrease to p230, suggesting that LHX2 may play its role largely in the late fetal and postnatal periods. The analyses of Lhx2 expression in pituitary tumor-derived cell lines showed their expressions in cell lines including αT31, LßT2 and others. Since LHX2 was previously identified as a transcription factor for Cga and the in vitro experiments in the present study suggested that LHX2 regulated the expression of Fshß, it is possible that LHX2 controls the synthesis of FSH at the transcription level.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/genética , Regulação da Expressão Gênica , Proteínas com Homeodomínio LIM/metabolismo , Suínos/genética , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Células CHO , Linhagem Celular Tumoral , Clonagem Molecular , Cricetinae , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Proteínas com Homeodomínio LIM/genética , Dados de Sequência Molecular , Hipófise/crescimento & desenvolvimento , Hipófise/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Transcrição Gênica
16.
Fertil Steril ; 93(8): 2465-85, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20403589

RESUMO

The pituitary gonadotropin hormones, FSH and LH, are key regulators of reproductive physiology. Though the two hormones are produced by the same cell type, often in response to the same endocrine and paracrine regulators, they sub-serve different biological functions and their synthesis and secretion are differentially regulated. This stems largely from differences in transcriptional, post-transcriptional, and post-translational regulation of their unique beta subunits. That is, both hormones are dimeric glycoproteins and share a common alpha subunit. Their unique beta subunits, however, derive from different genes encoding distinct proteins. Past and recent research indicates synthesis and release of the two hormones are subject to extensive and independent regulation. LH appears to be secreted predominantly via the regulated secretory pathway, whereas FSH release is largely constitutive. As such, investigations of FSH-beta subunit synthesis may lend direct insight into mechanisms underlying patterns of secreted FSH, more so than investigations of the LHbeta subunit. Here, we review recent investigations of transcriptional regulation of the FSH-beta subunit gene from different mammalian species, including humans. The results reveal both conserved and species-specific regulatory mechanisms that might contribute to inter-species variation in FSH release.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/biossíntese , Ativinas/fisiologia , Androgênios/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Estrogênios/fisiologia , Feminino , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Folistatina/fisiologia , Fator de Transcrição GATA2/fisiologia , Regulação da Expressão Gênica , Subunidade alfa de Hormônios Glicoproteicos , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Humanos , Inibinas/fisiologia , Proteínas com Homeodomínio LIM , Masculino , Dados de Sequência Molecular , Fatores de Transcrição Box Pareados/fisiologia , Regiões Promotoras Genéticas/fisiologia , Alinhamento de Sequência , Especificidade da Espécie , Fator Esteroidogênico 1/fisiologia , Fatores de Transcrição
17.
Protein Expr Purif ; 68(2): 183-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19635568

RESUMO

Bovine follicle-stimulating hormone (bFSH), a pituitary gonadotropin, is a heterodimer hormone that consists of a common alpha-subunit non-covalently associated with the hormone-specific beta-subunit. Unfortunately, expression levels of recombinant bFSH or its subunits are invariably low. We report here the secretory expression of biologically active bFSHalpha and bFSHbeta subunit in the methylotrophic yeast Hansenula polymorpha. A slightly higher level of expression of recombinant bFSH subunits was achieved by using the Saccharomyces cerevisiae-derived calnexin (ScCne1) as a chaperone in engineered H. polymorpha strains. The preliminary data also suggested that bFSH subunits expressed in H. polymorpha appeared to be less-glycosylated. This isoform had been shown to be 80% increase in in vivo bioactivity compared with the hyperglycosylated Pichia pastoris-derived recombinant bFSHalpha/beta. More sophisticated applications of bFSH would profit from the assembled less-glycosylated heterodimer.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Subunidade alfa de Hormônios Glicoproteicos/biossíntese , Pichia/genética , Proteínas Recombinantes de Fusão/biossíntese , Animais , Calnexina/genética , Calnexina/metabolismo , Bovinos , Cromatografia de Afinidade , Códon/metabolismo , Subunidade beta do Hormônio Folículoestimulante/química , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidade alfa de Hormônios Glicoproteicos/química , Subunidade alfa de Hormônios Glicoproteicos/genética , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Glicosilação , Histidina/química , Histidina/genética , Oligopeptídeos/química , Oligopeptídeos/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
18.
Reprod Biol Endocrinol ; 7: 66, 2009 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-19552818

RESUMO

BACKGROUND: Activin A increases production of follicle stimulating hormone (FSH) by inducing transcription of its beta subunit (FSHB). This induction has been studied here in LbetaT2 gonadotropes using transient expression of ovine FSHBLuc (-4741 bp of ovine FSHB promoter plus exon/intron 1 linked to Luc). Several sequences between -169/-58 bp of the ovine FSHB proximal promoter are necessary for induction by activin A in LbetaT2 cells, but deletions between -4741/-752 bp decrease induction > 70% suggesting the existence of other important 5' sequences. Induction disappears if a minimal T81 thymidine kinase promoter replaces the ovine FSHB TATA box and 3' exon/intron. The study reported here was designed to determine if sequences outside -169/-58 bp are important for induction of ovine FSHB by activin A. METHODS: Progressively longer deletions of ovine FSHBLuc were created between -4741/-195 bp. Deletions internal to this region were created also, but replaced with substitute DNA. The ovine FSHB TATA box region (-40/+3 bp) was replaced by thymidine kinase and rat prolactin minimal promoters, and substitutions were made in 3' intron/exon sequences. All constructs were tested for basal and activin A-induced expression in LbetaT2 cells. RESULTS: Successive 5' deletions progressively lowered fold-induction by activin A from 9.5 to zero, but progressively increased basal expression. Replacing deletions with substitute DNA showed no changes in basal expression or fold-induction. Induction by activin A was supported by the minimal rat prolactin promoter (TATA box) but not the thymidine kinase promoter (no TATA box). Replacement mutations in the 3' region did not decrease induction by activin A. CONCLUSION: The data show that specific ovine FSHB sequences 5' to -175 bp or 3' of the transcription start site are not required for induction by activin A. A minimal TATA box promoter supports induction by activin A, but the sequence between the TATA box and transcription start site seems unimportant.


Assuntos
Ativinas/farmacologia , Subunidade beta do Hormônio Folículoestimulante/genética , Regiões Promotoras Genéticas/genética , TATA Box , Animais , Sequência de Bases , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Gonadotrofos/metabolismo , Camundongos , Ratos , Ovinos
20.
Gen Comp Endocrinol ; 158(2): 178-82, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18664367

RESUMO

In many animals, including the three-spine stickleback (Gasterosteus aculeatus), photoperiod strongly influences reproduction. The aim of this study was to investigate if feedback mechanisms on the brain-pituitary-gonadal axis play a role in mediating the photoperiodic response in the stickleback. To that end, stickleback males, exposed to either non-stimulatory short photoperiod (light/dark 8:16) or under stimulatory long photoperiod (LD 16:8), were subjected to either sham-operation, castration, castration combined with treatment with the androgens 11-ketoandrostenedione (11KA) and testosterone (T), and the effects on levels of luteinizing hormone (LH) and follicle-stimulating hormone (FSH)-beta mRNA were analyzed. During breeding season the kidney of the stickleback male hypertrophies and produces a glue used for building nests. Kidney weight and expression of both LH-beta and FSH-beta were higher in sham-operated fish kept under long than under short photoperiod. Under both photoperiods, LH-beta mRNA levels were lower in castrated males compared to sham-operated males and treatment with 11KA and T increased expression, indicating a positive feedback. A positive feedback was also found on FSH-beta expression under long photoperiod, where castration decreased, and androgen replacement restored FSH-beta mRNA expression. On the contrary, castration under short photoperiod instead increased FSH-beta levels whereas treatment with 11KA and T decreased FSH-beta expression, indicating a negative feedback on FSH-beta under these conditions. The positive feedback on FSH-beta expression under stimulatory photoperiod may accelerate maturation, whereas the negative feedback under inhibitory photoperiod may suppress maturation. This could be part of the mechanisms by which photoperiod controls maturation.


Assuntos
Androstenos/farmacologia , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Hormônio Luteinizante Subunidade beta/biossíntese , Smegmamorpha/fisiologia , Testosterona/farmacologia , Animais , Retroalimentação/fisiologia , Subunidade beta do Hormônio Folículoestimulante/genética , Hormônio Luteinizante Subunidade beta/genética , Masculino , Hibridização de Ácido Nucleico , Orquiectomia/veterinária , Fotoperíodo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Maturidade Sexual/efeitos dos fármacos , Maturidade Sexual/fisiologia , Smegmamorpha/genética , Smegmamorpha/metabolismo , Testículo/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...