Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34967900

RESUMO

For billions of years before electric light was invented, life on Earth evolved under the pattern of light during the day and darkness during the night. Through evolution, nearly all organisms internalized the temporal rhythm of Earth's 24-hour rotation and evolved self-sustaining biological clocks with a ~24-hour rhythm. These internal rhythms are called circadian rhythms, and the molecular constituents that generate them are called molecular circadian clocks. Alignment of molecular clocks with the environmental light-dark rhythms optimizes physiology and behavior. This phenomenon is particularly true for reproductive function, in which seasonal breeders use day length information to time yearly changes in fertility. However, it is becoming increasingly clear that light-induced disruption of circadian rhythms can negatively impact fertility in nonseasonal breeders as well. In particular, the luteinizing hormone surge promoting ovulation is sensitive to circadian disruption. In this review, we will summarize our current understanding of the neuronal networks that underlie circadian rhythms and the luteinizing hormone surge.


Assuntos
Ritmo Circadiano/fisiologia , Hormônio Luteinizante/metabolismo , Rede Nervosa/fisiologia , Neurônios/fisiologia , Animais , Relógios Circadianos , Ritmo Circadiano/genética , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano , Feminino , Hormônio Liberador de Gonadotropina , Hipotálamo , Kisspeptinas , Luz , Masculino , Neuropeptídeos/fisiologia , Roedores , Núcleo Supraquiasmático/fisiologia , Fatores de Transcrição/fisiologia
2.
Mol Neurobiol ; 57(2): 1217-1232, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31705443

RESUMO

The brain's primary circadian pacemaker, the suprachiasmatic nucleus (SCN), is required to translate day-length and circadian rhythms into neuronal, hormonal, and behavioral rhythms. Here, we identify the homeodomain transcription factor ventral anterior homeobox 1 (Vax1) as required for SCN development, vasoactive intestinal peptide expression, and SCN output. Previous work has shown that VAX1 is required for gonadotropin-releasing hormone (GnRH/LHRH) neuron development, a neuronal population controlling reproductive status. Surprisingly, the ectopic expression of a Gnrh-Cre allele (Gnrhcre) in the SCN confirmed the requirement of both VAX1 (Vax1flox/flox:Gnrhcre, Vax1Gnrh-cre) and sine oculis homeobox protein 6 (Six6flox/flox:Gnrhcre, Six6Gnrh-cre) in SCN function in adulthood. To dissociate the role of Vax1 and Six6 in GnRH neuron and SCN function, we used another Gnrh-cre allele that targets GnRH neurons, but not the SCN (Lhrhcre). Both Six6Lhrh-cre and Vax1Lhrh-cre were infertile, and in contrast to Vax1Gnrh-cre and Six6Gnrh-cre mice, Six6Lhrh-cre and Vax1Lhrh-cre had normal circadian behavior. Unexpectedly, ~ 1/4 of the Six6Gnrh-cre mice were unable to entrain to light, showing that ectopic expression of Gnrhcre impaired function of the retino-hypothalamic tract that relays light information to the brain. This study identifies VAX1, and confirms SIX6, as transcription factors required for SCN development and function and demonstrates the importance of understanding how ectopic CRE expression can impact the results.


Assuntos
Proteínas de Homeodomínio/metabolismo , Neuropeptídeos/metabolismo , Núcleo Supraquiasmático/crescimento & desenvolvimento , Núcleo Supraquiasmático/fisiologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Ritmo Circadiano/fisiologia , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo
3.
Endocrinology ; 157(12): 4914-4929, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27704948

RESUMO

Circadian rhythms synchronize physiological processes with the light-dark cycle and are regulated by a hierarchical system initiated in the suprachiasmatic nucleus, a hypothalamic region that receives direct photic input. The suprachiasmatic nucleus then entrains additional oscillators in the periphery. Circadian rhythms are maintained by a molecular transcriptional feedback loop, of which brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (BMAL1) is a key member. Disruption of circadian rhythms by deletion of the BMAL1 gene (Bmal1 knockout [KO]) induces a variety of disease states, including infertility in males, due to unidentified mechanisms. We find that, despite normal sperm function, Bmal1 KO males fail to mate with receptive females, indicating a behavioral defect. Mating is dependent on pheromone detection, as are several other behaviors. We determined that Bmal1 KO males also fail to display aggression and avoidance of predator scent, despite intact main olfactory function. Moreover, the vomeronasal organ, a specialized pheromone-responsive organ, was also functionally intact, as determined by calcium imaging in response to urine pheromone stimulus. However, neural circuit tracing using c-FOS activation revealed that, although Bmal1 KO males displayed appropriate activation in the olfactory bulb and accessory olfactory bulb, the bed nucleus of the stria terminalis and the medial preoptic area (areas responsible for integration of copulatory behaviors) failed to activate highly in response to the female scent. This indicates that neural signaling in select behavioral centers is impaired in the absence of BMAL1, likely underlying Bmal1 KO male copulatory defects, demonstrating the importance of the BMAL1 protein in the maintenance of neural circuits that drive pheromone-mediated mating behaviors.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Hipotálamo/metabolismo , Rede Nervosa/metabolismo , Neurônios/metabolismo , Reprodução/fisiologia , Comportamento Sexual Animal/fisiologia , Órgão Vomeronasal/metabolismo , Fatores de Transcrição ARNTL/genética , Animais , Masculino , Camundongos , Área Pré-Óptica/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
4.
PLoS One ; 11(7): e0158597, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27389022

RESUMO

Gonadotropin-releasing hormone (GnRH), a neuropeptide released from a small population of neurons in the hypothalamus, is the central mediator of the hypothalamic-pituitary-gonadal axis, and is required for normal reproductive development and function. Evolutionarily conserved regulatory elements in the mouse, rat, and human Gnrh1 gene include three enhancers and the proximal promoter, which confer Gnrh1 gene expression specifically in GnRH neurons. In immortalized mouse hypothalamic GnRH (GT1-7) neurons, which show pulsatile GnRH release in culture, RNA sequencing and RT-qPCR revealed that expression of a novel long noncoding RNA at Gnrh1 enhancer 1 correlates with high levels of GnRH mRNA expression. In GT1-7 neurons, which contain a transgene carrying 3 kb of the rat Gnrh1 regulatory region, both the mouse and rat Gnrh1 enhancer-derived noncoding RNAs (GnRH-E1 RNAs) are expressed. We investigated the characteristics and function of the endogenous mouse GnRH-E1 RNA. Strand-specific RT-PCR analysis of GnRH-E1 RNA in GT1-7 cells revealed GnRH-E1 RNAs that are transcribed in the sense and antisense directions from distinct 5' start sites, are 3' polyadenylated, and are over 2 kb in length. These RNAs are localized in the nucleus and have a half-life of over 8 hours. In GT1-7 neurons, siRNA knockdown of mouse GnRH-E1 RNA resulted in a significant decrease in the expression of the Gnrh1 primary transcript and Gnrh1 mRNA. Over-expression of either the sense or antisense mouse GnRH-E1 RNA in immature, migratory GnRH (GN11) neurons, which do not express either GnRH-E1 RNA or GnRH mRNA, induced the transcriptional activity of co-transfected rat Gnrh1 gene regulatory elements, where the induction requires the presence of the rat Gnrh1 promoter. Together, these data indicate that GnRH-E1 RNA is an inducer of Gnrh1 gene expression. GnRH-E1 RNA may play an important role in the development and maturation of GnRH neurons.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Neurônios/metabolismo , Precursores de Proteínas/genética , RNA não Traduzido/genética , Animais , Dactinomicina/química , Fertilidade , Humanos , Hipotálamo/metabolismo , Camundongos , Células NIH 3T3 , Neuropeptídeos/metabolismo , Poliadenilação , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Ratos , Análise de Sequência de RNA
5.
J Neurosci ; 36(12): 3506-18, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27013679

RESUMO

Hypothalamic gonadotropin-releasing hormone (GnRH) neurons are at the apex of the hypothalamic-pituitary-gonadal axis that regulates mammalian fertility. Herein we demonstrate a critical role for the homeodomain transcription factor ventral anterior homeobox 1 (VAX1) in GnRH neuron maturation and show that Vax1 deletion from GnRH neurons leads to complete infertility in males and females. Specifically, global Vax1 knock-out embryos had normal numbers of GnRH neurons at 13 d of gestation, but no GnRH staining was detected by embryonic day 17. To identify the role of VAX1 specifically in GnRH neuron development,Vax1(flox)mice were generated and lineage tracing performed in Vax1(flox/flox):GnRH(cre):RosaLacZ mice. This identified VAX1 as essential for maintaining expression of Gnrh1 The absence of GnRH staining in adult Vax1(flox/flox):GnRH(cre)mice led to delayed puberty, hypogonadism, and infertility. To address the mechanism by which VAX1 maintains Gnrh1 transcription, the capacity of VAX1 to regulate Gnrh1 transcription was evaluated in the GnRH cell lines GN11 and GT1-7. As determined by luciferase and electrophoretic mobility shift assays, we found VAX1 to be a direct activator of the GnRH promoter through binding to four ATTA sites in the GnRH enhancer (E1) and proximal promoter (P), and able to compete with the homeoprotein SIX6 for occupation of the identified ATTA sites in the GnRH promoter. We conclude that VAX1 is expressed in GnRH neurons where it is required for GnRH neuron expression of GnRH and maintenance of fertility in mice. SIGNIFICANCE STATEMENT: Infertility classified as idiopathic hypogonadotropic hypogonadism (IHH) is characterized by delayed or absent sexual maturation and low sex steroid levels due to alterations in neuroendocrine control of the hypothalamic-pituitary-gonadal axis. The incidence of IHH is 1-10 cases per 100,000 births. Although extensive efforts have been invested in identifying genes giving rise to IHH, >50% of cases have unknown genetic origins. We recently showed that haploinsufficiency of ventral anterior homeobox 1 (Vax1) leads to subfertility, making it a candidate in polygenic IHH. In this study, we investigate the mechanism by which VAX1 controls fertility finding that VAX1 is required for maintenance of Gnrh1 gene expression and deletion of Vax1 from GnRH neurons leads to complete infertility.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Homeodomínio/metabolismo , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Infertilidade/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Animais , Feminino , Fertilidade , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Camundongos Knockout , Neuropeptídeos/genética
6.
Biol Reprod ; 93(3): 69, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26203175

RESUMO

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


Assuntos
Inibidores Enzimáticos/toxicidade , Nitrilas/toxicidade , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/patologia , Reprodução/efeitos dos fármacos , Triazóis/toxicidade , Animais , Corpo Lúteo/metabolismo , Diestro/metabolismo , Ciclo Estral/efeitos dos fármacos , Feminino , Hiperandrogenismo/sangue , Hiperandrogenismo/induzido quimicamente , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Kisspeptinas/biossíntese , Kisspeptinas/genética , Letrozol , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Síndrome do Ovário Policístico/metabolismo , Gravidez , Testosterona/sangue
7.
J Neurosci ; 31(2): 426-38, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21228153

RESUMO

The hypothalamus, pituitary, and gonads coordinate to direct the development and regulation of reproductive function in mammals. Control of the hypothalamic-pituitary-gonadal axis is dependent on correct migration of gonadotropin-releasing hormone (GnRH) neurons from the nasal placode to the hypothalamus, followed by proper synthesis and pulsatile secretion of GnRH, functions absent in patients with hypogonadal hypogonadism. In this study, we identify sine oculis-related homeobox 6 (Six6) as a novel factor necessary for proper targeting of GnRH expression to the limited population of GnRH neurons within the adult mouse hypothalamus and demonstrate that it is required for proper reproductive function in both male and female mice. Female Six6-null mice exhibit a striking decrease in fertility, failing to progress through the estrous cycle normally, show any signs of successful ovulation, or produce litters. Although basal gonadotropin production in these mice is relatively normal, analysis of GnRH expression reveals a dramatic decrease in total GnRH neuron numbers. We show that expression of Six6 is dramatically increased during GnRH neuronal maturation and that overexpression of Six6 induces GnRH transcription in neuronal cells. Finally, we demonstrate that this induction in GnRH expression is mediated via binding of Six6 to evolutionarily conserved ATTA sites located within the GnRH proximal promoter. Together, these data indicate that Six6 plays an important role in the regulation of GnRH expression and hypothalamic control of fertility.


Assuntos
Fertilidade/fisiologia , Hormônio Liberador de Gonadotropina/biossíntese , Proteínas de Homeodomínio/fisiologia , Hipotálamo/metabolismo , Reprodução/fisiologia , Transativadores/fisiologia , Animais , Linhagem Celular , Ciclo Estral/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/genética , Proteínas de Homeodomínio/genética , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Ovulação/fisiologia , Gravidez , Regiões Promotoras Genéticas , Fatores Sexuais , Transativadores/genética , Transcrição Gênica
8.
Mol Endocrinol ; 24(10): 1949-64, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20667983

RESUMO

GnRH, the central regulator of reproductive function, is produced by only approximately 800 highly specialized hypothalamic neurons. Previous studies identified a minimal promoter [GnRH minimal promoter (GnRH-P)] (-173/+1) and a neuron-specific enhancer [GnRH-enhancer (E)1] (-1863/-1571) as regulatory regions in the rat gene that confer this stringent specificity of GnRH expression to differentiated GnRH neurons. In transgenic mice, these two elements target only GnRH neurons but fail to drive expression in the entire population, suggesting the existence of additional regulatory regions. Here, we define two novel, highly conserved, upstream enhancers in the GnRH gene termed GnRH-E2 (-3135/-2631) and GnRH-E3 (-4199/-3895) that increase neuron-specific GnRH expression through interactions with GnRH-E1 and GnRH-P. GnRH-E2 and GnRH-E3 regulate GnRH expression through similar mechanisms via Oct-1, Msx1, and Dlx2, which bind both GnRH-E2 and the GnRH-E3 critical region at -3952/-3895. Overexpression of Dlx2 increases transcription through GnRH-E2 and GnRH-E3. Remarkably, these novel elements are contained within the 3' untranslated region of the neighboring upstream gene, yet are marked endogenously by histone modification signatures consistent with those of enhancers. Thus, GnRH-E2 and GnRH-E3 are novel regulatory elements that, together with GnRH-E1 and GnRH-P, confer the specificity of GnRH expression to differentiated and mature GnRH neurons.


Assuntos
Elementos Facilitadores Genéticos , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Homeodomínio/metabolismo , Hipotálamo/metabolismo , Transcrição Gênica , Regiões 3' não Traduzidas , Animais , Sequência de Bases , Linhagem Celular , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Canais de Potássio/genética , Regiões Promotoras Genéticas , Ratos , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Neuroendocrinology ; 84(6): 353-63, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17192702

RESUMO

Orexin A, a recently discovered hypothalamic peptide, has been shown to have a stimulatory effect on release of gonadotropin-releasing hormone (GnRH) from rat hypothalamic explants in vitro. However, it is presently unclear whether in vivo this effect is mediated directly at the level of the GnRH neuron, or via multiple afferent neuronal connections. Therefore, in the present study, we investigated the direct action of orexin A on GnRH neurons using the immortalized GnRH-secreting GT1-7 hypothalamic cells. Orexin-1 receptor (OX1R) expression was detected in GT1-7 cells by RT-PCR and Western blot. Results showed that 0.1-1 nM orexin A, when administered in culture media for 4 h, can significantly stimulate GnRH mRNA expression in GT1-7 cells (p < 0.05). Administration of 1 microM OX1R antagonist, SB-334867, completely blocked the observed orexin A responses in these cells, indicating that orexin A stimulation of GnRH neurons is specifically through OX1R. Moreover, 0.1 nM orexin A stimulated GnRH release after 30-45 min. To examine possible signal transduction pathways involved in mediating these effects, a MEK inhibitor (UO-126), PKC inhibitor (calphostin C), and PKA inhibitor (H-89), were used, with each blocking orexin A-induced GnRH transcription and release from immortalized cells. Collectively, our results show that orexin A is capable of directly stimulating GnRH transcription and neuropeptide release from these immortalized hypothalamic neurons, and that the effects of orexin A appear to be mediated via the OX1R, coupled with activation of the PKC-, MAPK- and PKA-signaling pathways. It is suggested that the stimulatory effect of orexin A on GnRH transcription and release may also occur directly at the level of GnRH neurons in vivo.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Neuropeptídeos/farmacologia , Neurotransmissores/farmacologia , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Hormônio Liberador de Gonadotropina/genética , Humanos , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Neurônios/metabolismo , Receptores de Orexina , Orexinas , Proteína Quinase C/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia
10.
Mol Endocrinol ; 19(11): 2769-79, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15994198

RESUMO

The GnRH gene uses two well-defined regions to target expression to a small population of hypothalamic GnRH neurons: a 173-bp proximal promoter and a 300-bp enhancer localized at approximately -1800 to -1500 bp from the start site. Interaction of multiple factors with the GnRH enhancer and promoter is required to confer neuron-specific expression in vivo and in cells in culture. In addition, the expression of the GnRH gene is regulated by numerous neurotransmitters and hormones. Several of these effectors act through membrane receptors to trigger the protein kinase C pathway, and 12-O-tetradecanoyl phorbol-13-acetate (TPA), a modulator of this pathway, has been shown to suppress GnRH gene expression through the promoter. We find that TPA suppresses expression through the GnRH enhancer as well as the promoter. In the enhancer, an Oct-1 binding site, a Pbx/Prep binding site, Msx/Dlx binding sites, and a previously unidentified protein-binding element at -1793, all contribute to TPA suppression. TPA treatment leads to decreased binding of Oct-1 and Pbx1a/Prep to their sites. However, a complex formed by GT1-7 nuclear extracts on the -1793 site is not affected by TPA treatment. It is known that cooperative interaction among multiple factors is necessary for GnRH gene expression; thus, one mechanism by which TPA suppresses GnRH gene expression is to disengage some of these factors from their cis-regulatory elements.


Assuntos
Regulação para Baixo , Elementos Facilitadores Genéticos/genética , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Proteína Quinase C/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Elementos Facilitadores Genéticos/efeitos dos fármacos , Regulação da Expressão Gênica , Hipotálamo/citologia , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Ratos , Proteínas Repressoras/metabolismo , Deleção de Sequência , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica/efeitos dos fármacos
11.
J Biol Chem ; 280(19): 19156-65, 2005 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15743757

RESUMO

Gonadotropin-releasing hormone (GnRH) is the central regulator of the hypothalamic-pituitary-gonadal axis, controlling sexual maturation and fertility in diverse species from fish to humans. GnRH gene expression is limited to a discrete population of neurons that migrate through the nasal region into the hypothalamus during embryonic development. The GnRH regulatory region contains four conserved homeodomain binding sites (ATTA) that are essential for basal promoter activity and cell-specific expression of the GnRH gene. MSX and DLX are members of the Antennapedia class of non-Hox homeodomain transcription factors that regulate gene expression and influence development of the craniofacial structures and anterior forebrain. Here, we report that expression patterns of the Msx and Dlx families of homeodomain transcription factors largely coincide with the migratory route of GnRH neurons and co-express with GnRH in neurons during embryonic development. In addition, MSX and DLX family members bind directly to the ATTA consensus sequences and regulate transcriptional activity of the GnRH promoter. Finally, mice lacking MSX1 or DLX1 and 2 show altered numbers of GnRH-expressing cells in regions where these factors likely function. These findings strongly support a role for MSX and DLX in contributing to spatiotemporal regulation of GnRH transcription during development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Hormônio Liberador de Gonadotropina/biossíntese , Hormônio Liberador de Gonadotropina/genética , Proteínas de Homeodomínio/química , Fatores de Transcrição/química , Animais , Sequência de Bases , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Núcleo Celular/metabolismo , Embrião de Mamíferos/metabolismo , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Fator de Transcrição MSX1 , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Células NIH 3T3 , Neurônios/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Regiões Promotoras Genéticas , Prosencéfalo/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transcrição Gênica , Transfecção
12.
J Biol Chem ; 279(29): 30287-97, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15138251

RESUMO

Gonadotropin-releasing hormone (GnRH) is the central regulator of reproductive function. Expression of the GnRH gene is confined to a rare population of neurons scattered throughout the hypothalamus. Restricted expression of the rat GnRH gene is driven by a multicomponent enhancer and an evolutionarily conserved promoter. Oct-1, a ubiquitous POU homeodomain transcription factor, was identified as an essential factor regulating GnRH transcription in the GT1-7 hypothalamic neuronal cell line. In this study, we conducted a two-hybrid interaction screen in yeast using a GT1-7 cDNA library to search for specific Oct-1 cofactors. Using this approach, we isolated Pbx1b, a TALE homeodomain transcription factor that specifically associates with Oct-1. We show that heterodimers containing Pbx/Prep1 or Pbx/Meis1 TALE homeodomain proteins bind to four functional elements within the GnRH regulatory region, each in close proximity to an Oct-1-binding site. Cotransfection experiments indicate that TALE proteins are essential for GnRH promoter activity in the GT1-7 cells. Moreover, Pbx1 and Oct-1, as well as Prep1 and Oct-1, form functional complexes that enhance GnRH gene expression. Finally, Pbx1 is expressed in GnRH neurons in embryonic as well as mature mice, suggesting that the associations between TALE homeodomain proteins and Oct-1 regulate neuron-specific expression of the GnRH gene in vivo.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , Cromatina/metabolismo , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/química , Dimerização , Relação Dose-Resposta a Droga , Biblioteca Gênica , Glutationa Transferase/metabolismo , Proteínas de Homeodomínio , Fator C1 de Célula Hospedeira , Hipotálamo/metabolismo , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Neurônios/metabolismo , Fator 1 de Transcrição de Octâmero , Oligonucleotídeos/química , Testes de Precipitina , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Proteínas Repressoras , Fatores de Transcrição/química , Transcrição Gênica , Transfecção , Técnicas do Sistema de Duplo-Híbrido
13.
J Neurosci ; 23(35): 11202-13, 2003 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-14657179

RESUMO

Although it has long been established that episodic secretion of gonadotropin-releasing hormone (GnRH) from the hypothalamus is required for normal gonadotropin release, the molecular and cellular mechanisms underlying the synchronous release of GnRH are primarily unknown. We used the GT1-7 mouse hypothalamic cell line as a model for GnRH secretion, because these cells release GnRH in a pulsatile pattern similar to that observed in vivo. To explore possible molecular mechanisms governing secretory timing, we investigated the role of the molecular circadian clock in regulation of GnRH secretion. GT1-7 cells express many known core circadian clock genes, and we demonstrate that oscillations of these components can be induced by stimuli such as serum and the adenylyl cyclase activator forskolin, similar to effects observed in fibroblasts. Strikingly, perturbation of circadian clock function in GT1-7 cells by transient expression of the dominant-negative Clock-Delta19 gene disrupts normal ultradian patterns of GnRH secretion, significantly decreasing mean pulse frequency. Additionally, overexpression of the negative limb clock gene mCry1 in GT1-7 cells substantially increases GnRH pulse amplitude without a commensurate change in pulse frequency, demonstrating that an endogenous biological clock is coupled to the mechanism of neurosecretion in these cells and can regulate multiple secretory parameters. Finally, mice harboring a somatic mutation in the Clock gene are subfertile and exhibit a substantial increase in estrous cycle duration as revealed by examination of vaginal cytology. This effect persists in normal light/dark (LD) cycles, suggesting that a suprachiasmatic nucleus-independent endogenous clock in GnRH neurons is required for eliciting normal pulsatile patterns of GnRH secretion.


Assuntos
Ritmo Circadiano/fisiologia , Proteínas de Drosophila , Proteínas do Olho , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/biossíntese , Hipotálamo/metabolismo , Células Fotorreceptoras de Invertebrados , Fatores de Transcrição ARNTL , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Relógios Biológicos/genética , Relógios Biológicos/fisiologia , Proteínas CLOCK , Proteínas de Ciclo Celular , Linhagem Celular , Ritmo Circadiano/genética , Criptocromos , Estro/genética , Estro/fisiologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Flavoproteínas/genética , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Fluorescência Verde , Hipotálamo/citologia , Proteínas Luminescentes/genética , Camundongos , Camundongos Mutantes , Células NIH 3T3 , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Circadianas Period , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Mol Endocrinol ; 16(11): 2413-25, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12403831

RESUMO

GnRH gene expression is restricted to a tiny population of neurons scattered throughout the mediobasal hypothalamus. The combination of a 300-bp enhancer and the 173-bp promoter from the rat GnRH gene can confer this narrow specificity in transgenic mice and in transfections of hypothalamic GT1-7 cells. In the present study, we identify repeated CAATT elements in the 3' region of the rat GnRH enhancer that bind a tissue-restricted protein complex and play a significant role in cell-restricted expression of the GnRH gene. Deletions of multiple repeats demonstrate their importance in transcriptional activity. In fact, even mutation of a single repeat reduces expression. This reduction can be compensated by the conserved GnRH promoter, which also contains such elements and binds this protein complex. In Southwestern analysis, three proteins from GT1-7 nuclear extract bind to the CAATT element, and these proteins are not found in NIH3T3 cells. This cell-specific protein complex has properties of the Q50 homeodomain family of transcription factors and binds to as many as seven binding sites in the enhancer and promoter to play a key role in GnRH gene expression in the hypothalamus.


Assuntos
Núcleo Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Hormônio Liberador de Gonadotropina/genética , Neurônios/fisiologia , Células 3T3 , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular , Clonagem Molecular , Hipotálamo/fisiologia , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Plasmídeos , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Transcrição Gênica , Transfecção
15.
Menopause ; 9(2): 145-50, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11875334

RESUMO

OBJECTIVE: Women are increasingly turning to herbal therapies in an effort to manage their menopausal symptoms. In this study, we investigate the estrogenic activity of four selected herbs commonly used in menopause, namely dong quai, ginseng, black cohosh, and licorice root. DESIGN: We investigated the effect of these selected herbs on cell proliferation of MCF-7 cells, a human breast cancer cell line. We also assessed their estrogenic activity in a transient gene expression assay system using HeLa cells co-transfected with an estrogen-dependent reporter plasmid in the presence of human estrogen receptor ER alpha or ER beta cDNA. Finally, we investigated the estrogenic activity of these herbs using a bioassay in mice. RESULTS: Dong quai and ginseng both significantly induced the growth of MCF-7 cells by 16- and 27-fold, respectively, over that of untreated control cells, while black cohosh and licorice root did not. The herbs tested failed to show transactivation of either hER alpha or hER beta and had no effect on uterine weight in vivo when administered orally to mice for a period of 4 days. CONCLUSIONS: Our studies show that dong quai and ginseng stimulate the growth of MCF-7 cells independent of estrogenic activity. Because of the lack of efficacy and the potential for adverse effects, use of these herbs in humans warrants caution pending further study.


Assuntos
Fogachos/tratamento farmacológico , Fitoterapia , Preparações de Plantas/farmacologia , Preparações de Plantas/uso terapêutico , Receptores de Estrogênio/efeitos dos fármacos , Angelica sinensis , Animais , Caulophyllum , Divisão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Glycyrrhiza , Humanos , Menopausa , Camundongos , Camundongos Endogâmicos , Ovariectomia , Panax , Células Tumorais Cultivadas/efeitos dos fármacos
16.
Endocrinology ; 143(4): 1404-12, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11897697

RESUMO

The GnRH-expressing neurons are the ultimate regulator of reproductive function. GnRH gene expression is limited to this small population of neurons in the hypothalamus. Transfections using 3 kb of the rat or mouse 5'-regulatory region provide specific gene expression in the hypothalamic cell line GT1-7. The combination of two elements, a 300-bp enhancer and a 173-bp promoter, recapitulates specificity in GT1-7 cells. It was not known whether these elements could specifically target gene expression throughout development in the whole animal. We demonstrate that the 3-kb rat GnRH regulatory region provides a higher degree of specificity than the equivalent mouse sequence in a mouse hypothalamic cell line. Moreover, combination of the enhancer and the promoter of the rat gene targets expression to GnRH neurons in transgenic mice in a developmentally appropriate manner. Transgene expression is regulated by activin A, a known activator of GnRH gene expression. In contrast, the enhancer on a heterologous promoter produces inappropriate expression in vivo. We conclude that the enhancer and promoter regions of the rat GnRH gene are necessary for targeted expression to hypothalamic neurons and are sufficient to confer regulated, cell type-specific expression to a reporter gene in vivo.


Assuntos
Regulação da Expressão Gênica/genética , Hormônio Liberador de Gonadotropina/genética , Neurônios/metabolismo , Ativinas/fisiologia , Animais , Vírus do Sarcoma Aviário/genética , Linhagem Celular , Movimento Celular/genética , Feminino , Genes Reporter/genética , Hormônio Liberador de Gonadotropina/biossíntese , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Imuno-Histoquímica , Subunidades beta de Inibinas/fisiologia , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Plasmídeos/genética , Gravidez , Ratos , Especificidade da Espécie , Transfecção , Transgenes/genética , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA