Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nat Neurosci ; 24(12): 1660-1672, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34795451

RESUMO

Neurons that produce gonadotropin-releasing hormone (GnRH), which control fertility, complete their nose-to-brain migration by birth. However, their function depends on integration within a complex neuroglial network during postnatal development. Here, we show that rodent GnRH neurons use a prostaglandin D2 receptor DP1 signaling mechanism during infancy to recruit newborn astrocytes that 'escort' them into adulthood, and that the impairment of postnatal hypothalamic gliogenesis markedly alters sexual maturation by preventing this recruitment, a process mimicked by the endocrine disruptor bisphenol A. Inhibition of DP1 signaling in the infantile preoptic region, where GnRH cell bodies reside, disrupts the correct wiring and firing of GnRH neurons, alters minipuberty or the first activation of the hypothalamic-pituitary-gonadal axis during infancy, and delays the timely acquisition of reproductive capacity. These findings uncover a previously unknown neuron-to-neural-progenitor communication pathway and demonstrate that postnatal astrogenesis is a basic component of a complex set of mechanisms used by the neuroendocrine brain to control sexual maturation.


Assuntos
Hormônio Liberador de Gonadotropina , Maturidade Sexual , Astrócitos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/fisiologia , Neurônios/fisiologia , Maturidade Sexual/fisiologia
2.
Sci Rep ; 11(1): 1996, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33479437

RESUMO

Female puberty is subject to Polycomb Group (PcG)-dependent transcriptional repression. Kiss1, a puberty-activating gene, is a key target of this silencing mechanism. Using a gain-of-function approach and a systems biology strategy we now show that EED, an essential PcG component, acts in the arcuate nucleus of the hypothalamus to alter the functional organization of a gene network involved in the stimulatory control of puberty. A central node of this network is Kdm6b, which encodes an enzyme that erases the PcG-dependent histone modification H3K27me3. Kiss1 is a first neighbor in the network; genes encoding glutamatergic receptors and potassium channels are second neighbors. By repressing Kdm6b expression, EED increases H3K27me3 abundance at these gene promoters, reducing gene expression throughout a gene network controlling puberty activation. These results indicate that Kdm6b repression is a basic mechanism used by PcG to modulate the biological output of puberty-activating gene networks.


Assuntos
Histona Desmetilases com o Domínio Jumonji/genética , Kisspeptinas/genética , Complexo Repressor Polycomb 2/genética , Puberdade/genética , Animais , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Neurônios/metabolismo , Sistemas Neurossecretores/crescimento & desenvolvimento , Sistemas Neurossecretores/metabolismo , Proteínas do Grupo Polycomb/genética , Regiões Promotoras Genéticas/genética , Puberdade/fisiologia , Ratos , Biologia de Sistemas
3.
Hum Mol Genet ; 28(8): 1357-1368, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30608578

RESUMO

The initiation of puberty is orchestrated by an augmentation of gonadotropin-releasing hormone (GnRH) secretion from a few thousand hypothalamic neurons. Recent findings have indicated that the neuroendocrine control of puberty may be regulated by a hierarchically organized network of transcriptional factors acting upstream of GnRH. These include enhanced at puberty 1 (EAP1), which contributes to the initiation of female puberty through transactivation of the GnRH promoter. However, no EAP1 mutations have been found in humans with disorders of pubertal timing. We performed whole-exome sequencing in 67 probands and 93 relatives from a large cohort of familial self-limited delayed puberty (DP). Variants were analyzed for rare, potentially pathogenic variants enriched in case versus controls and relevant to the biological control of puberty. We identified one in-frame deletion (Ala221del) and one rare missense variant (Asn770His) in EAP1 in two unrelated families; these variants were highly conserved and potentially pathogenic. Expression studies revealed Eap1 mRNA abundance in peri-pubertal mouse hypothalamus. EAP1 binding to the GnRH1 promoter increased in monkey hypothalamus at the onset of puberty as determined by chromatin immunoprecipitation. Using a luciferase reporter assay, EAP1 mutants showed a reduced ability to trans-activate the GnRH promoter compared to wild-type EAP1, due to reduced protein levels caused by the Ala221del mutation and subcellular mislocation caused by the Asn770His mutation, as revealed by western blot and immunofluorescence, respectively. In conclusion, we have identified the first EAP1 mutations leading to reduced GnRH transcriptional activity resulting in a phenotype of self-limited DP.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Puberdade Tardia/genética , Securina/genética , Adolescente , Adulto , Animais , Criança , Feminino , Regulação da Expressão Gênica/genética , Hormônio Liberador de Gonadotropina/genética , Humanos , Hipotálamo/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Neurônios/metabolismo , Regiões Promotoras Genéticas/genética , Puberdade/genética , Puberdade/fisiologia , RNA Mensageiro/genética , Securina/fisiologia , Maturidade Sexual/genética , Transativadores/genética , Fatores de Transcrição/genética , Sequenciamento do Exoma , Adulto Jovem
4.
Nat Commun ; 9(1): 1977, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773799

RESUMO

Population studies elucidating the genetic architecture of reproductive ageing have been largely limited to European ancestries, restricting the generalizability of the findings and overlooking possible key genes poorly captured by common European genetic variation. Here, we report 26 loci (all P < 5 × 10-8) for reproductive ageing, i.e. puberty timing or age at menopause, in a non-European population (up to 67,029 women of Japanese ancestry). Highlighted genes for menopause include GNRH1, which supports a primary, rather than passive, role for hypothalamic-pituitary GnRH signalling in the timing of menopause. For puberty timing, we demonstrate an aetiological role for receptor-like protein tyrosine phosphatases by combining evidence across population genetics and pre- and peri-pubertal changes in hypothalamic gene expression in rodent and primate models. Furthermore, our findings demonstrate widespread differences in allele frequencies and effect estimates between Japanese and European associated variants, highlighting the benefits and challenges of large-scale trans-ethnic approaches.


Assuntos
Envelhecimento/genética , Povo Asiático/genética , Loci Gênicos/fisiologia , Menarca/genética , Menopausa/genética , Adolescente , Adulto , Fatores Etários , Animais , Criança , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Frequência do Gene/fisiologia , Variação Genética/fisiologia , Humanos , Hipotálamo/metabolismo , Japão , Macaca mulatta , Metanálise como Assunto , Pessoa de Meia-Idade , Modelos Animais , Ratos Sprague-Dawley , População Branca/genética
5.
Nat Commun ; 9(1): 57, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29302059

RESUMO

Polycomb group (PcG) proteins control the timing of puberty by repressing the Kiss1 gene in hypothalamic arcuate nucleus (ARC) neurons. Here we identify two members of the Trithorax group (TrxG) of modifiers, mixed-lineage leukemia 1 (MLL1), and 3 (MLL3), as central components of an activating epigenetic machinery that dynamically counteracts PcG repression. Preceding puberty, MLL1 changes the chromatin configuration at the promoters of Kiss1 and Tac3, two genes required for puberty to occur, from repressive to permissive. Concomitantly, MLL3 institutes a chromatin structure that changes the functional status of a Kiss1 enhancer from poised to active. RNAi-mediated, ARC-specific Mll1 knockdown reduced Kiss1 and Tac3 expression, whereas CRISPR-Cas9-directed epigenome silencing of the Kiss1 enhancer selectively reduced Kiss1 activity. Both interventions delay puberty and disrupt reproductive cyclicity. Our results demonstrate that an epigenetic switch from transcriptional repression to activation is crucial to the regulatory mechanism controlling the timing of mammalian puberty.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Hipotálamo/metabolismo , Proteína de Leucina Linfoide-Mieloide/genética , Puberdade/genética , Animais , Sistemas CRISPR-Cas , Cromatina , Epigênese Genética , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Kisspeptinas/genética , Macaca mulatta , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Taquicininas/genética
6.
Nat Commun ; 6: 10195, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26671628

RESUMO

In primates, puberty is unleashed by increased GnRH release from the hypothalamus following an interval of juvenile quiescence. GWAS implicates Zinc finger (ZNF) genes in timing human puberty. Here we show that hypothalamic expression of several ZNFs decreased in agonadal male monkeys in association with the pubertal reactivation of gonadotropin secretion. Expression of two of these ZNFs, GATAD1 and ZNF573, also decreases in peripubertal female monkeys. However, only GATAD1 abundance increases when gonadotropin secretion is suppressed during late infancy. Targeted delivery of GATAD1 or ZNF573 to the rat hypothalamus delays puberty by impairing the transition of a transcriptional network from an immature repressive epigenetic configuration to one of activation. GATAD1 represses transcription of two key puberty-related genes, KISS1 and TAC3, directly, and reduces the activating histone mark H3K4me2 at each promoter via recruitment of histone demethylase KDM1A. We conclude that GATAD1 epitomizes a subset of ZNFs involved in epigenetic repression of primate puberty.


Assuntos
Epigênese Genética , Fatores de Transcrição GATA/genética , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/metabolismo , Puberdade/genética , RNA Mensageiro/metabolismo , Animais , Western Blotting , Imunoprecipitação da Cromatina , Feminino , Imunofluorescência , Hormônio Foliculoestimulante/metabolismo , Fatores de Transcrição GATA/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Histona Desmetilases/metabolismo , Hibridização in Situ Fluorescente , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Macaca mulatta , Masculino , Neurocinina B/genética , Neurocinina B/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Dedos de Zinco/genética
7.
Front Neuroendocrinol ; 36: 90-107, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25171849

RESUMO

Substantial progress has been made in recent years toward deciphering the molecular and genetic underpinnings of the pubertal process. The availability of powerful new methods to interrogate the human genome has led to the identification of genes that are essential for puberty to occur. Evidence has also emerged suggesting that the initiation of puberty requires the coordinated activity of gene sets organized into functional networks. At a cellular level, it is currently thought that loss of transsynaptic inhibition, accompanied by an increase in excitatory inputs, results in the pubertal activation of GnRH release. This concept notwithstanding, a mechanism of epigenetic repression targeting genes required for the pubertal activation of GnRH neurons was recently identified as a core component of the molecular machinery underlying the central restraint of puberty. In this chapter we will discuss the potential contribution of various mechanisms of epigenetic regulation to the hypothalamic control of female puberty.


Assuntos
Epigênese Genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/fisiologia , Neurônios/metabolismo , Puberdade/fisiologia , Maturidade Sexual/fisiologia , Animais , Feminino , Humanos
8.
Neuroendocrinology ; 99(2): 94-107, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24686008

RESUMO

The importance of the Kiss1 gene in the control of reproductive development is well documented. However, much less is known about the transcriptional regulation of Kiss1 expression in the hypothalamus. Critical for these studies is an accurate identification of the site(s) where Kiss1 transcription is initiated. Employing 5'-RACE PCR, we detected a transcription start site (TSS1) used by the hypothalamus of rats, mice, nonhuman primates and humans to initiate Kiss1 transcription. In rodents, an exon 1 encoding 5'-untranslated sequences is followed by an alternatively spliced second exon, which encodes 5'-untranslated regions of two different lengths and contains the translation initiation codon (ATG). In nonhuman primates and humans, exon 2 is not alternatively spliced. Surprisingly, in rat mediobasal hypothalamus (MBH), but not preoptic area (POA), an additional TSS (TSS2) located upstream from TSS1 generates an exon 1 longer (377 bp) than the TSS1-derived exon 1 (98 bp). The content of TSS1-derived transcripts increased at puberty in the POA and MBH of female rats. It also increased in the MBH after ovariectomy, and this change was prevented by estrogen. In contrast, no such changes in TSS2-derived transcript abundance were detected. Promoter assays showed that the proximal TSS1 promoter is much more active than the putative TSS2 promoter, and that only the TSS1 promoter is regulated by estrogen. These differences appear to be related to the presence of a TATA box and binding sites for transcription factors activating transcription and interacting with estrogen receptor-α in the TSS1, but not TSS2, promoter.


Assuntos
Estrogênios/farmacologia , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , RNA Mensageiro/metabolismo , Maturidade Sexual , Sítio de Iniciação de Transcrição , Transcrição Gênica/efeitos dos fármacos , Animais , Receptor alfa de Estrogênio/efeitos dos fármacos , Terapia de Reposição de Estrogênios , Éxons/genética , Feminino , Humanos , Macaca mulatta , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Ovariectomia , Regiões Promotoras Genéticas/genética , Ratos , Ratos Sprague-Dawley , Transcrição Gênica/genética
9.
Nat Neurosci ; 16(3): 281-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23354331

RESUMO

The timing of puberty is controlled by many genes. The elements coordinating this process have not, however, been identified. Here we show that an epigenetic mechanism of transcriptional repression times the initiation of female puberty in rats. We identify silencers of the Polycomb group (PcG) as principal contributors to this mechanism and show that PcG proteins repress Kiss1, a puberty-activating gene. Hypothalamic expression of two key PcG genes, Eed and Cbx7, decreased and methylation of their promoters increased before puberty. Inhibiting DNA methylation blocked both events and resulted in pubertal failure. The pubertal increase in Kiss1 expression was accompanied by EED loss from the Kiss1 promoter and enrichment of histone H3 modifications associated with gene activation. Preventing the eviction of EED from the Kiss1 promoter disrupted pulsatile gonadotropin-releasing hormone release, delayed puberty and compromised fecundity. Our results identify epigenetic silencing as a mechanism underlying the neuroendocrine control of female puberty.


Assuntos
Epigênese Genética , Hipotálamo/fisiologia , Maturidade Sexual/fisiologia , Animais , Metilação de DNA , Estradiol/sangue , Feminino , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Histonas/genética , Histonas/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo , Ratos , Ratos Sprague-Dawley
10.
Mol Cell Endocrinol ; 351(2): 184-98, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22209758

RESUMO

Mammalian puberty is initiated by an increased pulsatile release of gonadotropin-releasing hormone (GnRH) from specialized neurons located in the hypothalamus. GnRH secretion is controlled by neuronal and glial networks, whose activity appears to be coordinated via transcriptional regulation. One of the transcription factors involved in this process is thought to be the recently described gene Enhanced at Puberty 1 (EAP1), which encodes a protein with dual transcriptional activity. In this study we used gene reporter and chromatin immunoprecipitation (ChIP) assays to examine the hypothesis that EAP1 expression is controlled by transcriptional regulators earlier postulated to serve as central nodes of a gene network involved in the neuroendocrine control of puberty. These regulators include Thyroid Transcription Factor 1 (TTF1), Yin Yang 1 (YY1), and CUX1, in addition to EAP1 itself. While TTF1 has been shown to facilitate the advent of puberty, YY1 (a zinc finger protein component of the Polycomb silencing complex) may play a repressive role. The precise role of CUX1 in this context is not known, but like EAP1, CUX1 can either activate or repress gene transcription. We observed that DNA segments of two different lengths (998 and 2744bp) derived from the 5'-flanking region of the human EAP1 gene display similar transcriptional activity. TTF1 stimulates transcription from both DNA segments with equal potency, whereas YY1, CUX1, and EAP1 itself, behave as transcriptional repressors. All four proteins are recruited in vivo to the EAP1 5'-flanking region. These observations suggest that EAP1 gene expression is under dual transcriptional regulation imposed by a trans-activator (TTF1) and two repressors (YY1 and CUX1) previously postulated to be upstream components of a puberty-controlling gene network. In addition, EAP1 itself appears to control its own expression via a negative auto-feedback loop mechanism. Further studies are needed to determine if the occupancy of the EAP1 promoter by these regulatory factors changes at the time of puberty.


Assuntos
Redes Reguladoras de Genes , Genes Reguladores/genética , Genes Supressores de Tumor , Proteínas de Neoplasias/genética , Animais , Sítios de Ligação , Linhagem Celular , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Proteínas de Neoplasias/biossíntese , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Puberdade/genética , Ratos , Ratos Sprague-Dawley , Sequências Reguladoras de Ácido Nucleico , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Securina , Fatores de Transcrição , Transcrição Gênica , Fator de Transcrição YY1/genética , Fator de Transcrição YY1/metabolismo
11.
Endocrinology ; 153(1): 350-61, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22128022

RESUMO

Mammalian reproductive cyclicity requires the periodic discharge of GnRH from hypothalamic neurons into the portal vessels connecting the neuroendocrine brain to the pituitary gland. GnRH secretion is, in turn, controlled by changes in neuronal and glial inputs to GnRH-producing neurons. The transcriptional control of this process is not well understood, but it appears to involve several genes. One of them, termed enhanced at puberty 1 (EAP1), has been postulated to function in the female hypothalamus as an upstream regulator of neuroendocrine reproductive function. RNA interference-mediated inhibition of EAP1 expression, targeted to the preoptic region, delays puberty and disrupts estrous cyclicity in rodents, suggesting that EAP1 is required for the normalcy of these events. Here, we show that knocking down EAP1 expression in a region of the medial basal hypothalamus that includes the arcuate nucleus, via lentiviral-mediated delivery of RNA interference, results in cessation of menstrual cyclicity in female rhesus monkeys undergoing regular menstrual cycles. Neither lentiviruses encoding an unrelated small interfering RNA nor the placement of viral particles carrying EAP1 small interfering RNA outside the medial basal hypothalamus-arcuate nucleus region affected menstrual cycles, indicating that region-specific expression of EAP1 in the hypothalamus is required for menstrual cyclicity in higher primates. The cellular mechanism by which EAP1 exerts this function is unknown, but the recent finding that EAP1 is an integral component of a powerful transcriptional-repressive complex suggests that EAP1 may control reproductive cyclicity by inhibiting downstream repressor genes involved in the neuroendocrine control of reproductive function.


Assuntos
Hipotálamo/fisiologia , Macaca mulatta/genética , Macaca mulatta/fisiologia , Ciclo Menstrual/genética , Ciclo Menstrual/fisiologia , Animais , Sequência de Bases , Primers do DNA/genética , Feminino , Expressão Gênica , Hipotálamo/anatomia & histologia , Técnicas In Vitro , Macaca mulatta/anatomia & histologia , Sistemas Neurossecretores/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética
12.
Mol Ther ; 19(12): 2152-62, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22031240

RESUMO

To date, a therapy for Huntington's disease (HD), a genetic, neurodegenerative disorder, remains elusive. HD is characterized by cell loss in the basal ganglia, with particular damage to the putamen, an area of the brain responsible for initiating and refining motor movements. Consequently, patients exhibit a hyperkinetic movement disorder. RNA interference (RNAi) offers therapeutic potential for this disorder by reducing the expression of HTT, the disease-causing gene. We have previously demonstrated that partial suppression of both wild-type and mutant HTT in the striatum prevents behavioral and neuropathological abnormalities in rodent models of HD. However, given the role of HTT in various cellular processes, it remains unknown whether a partial suppression of both alleles will be safe in mammals whose neurophysiology, basal ganglia anatomy, and behavioral repertoire more closely resembles that of a human. Here, we investigate whether a partial reduction of HTT in the normal non-human primate putamen is safe. We demonstrate that a 45% reduction of rhesus HTT expression in the mid- and caudal putamen does not induce motor deficits, neuronal degeneration, astrogliosis, or an immune response. Together, these data suggest that partial suppression of wild-type HTT expression is well tolerated in the primate putamen and further supports RNAi as a therapy for HD.


Assuntos
Doença de Huntington/genética , Doença de Huntington/terapia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Animais , Comportamento Animal , Western Blotting , Dependovirus/genética , Avaliação Pré-Clínica de Medicamentos , Gliose/metabolismo , Gliose/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteína Huntingtina , Imunidade Ativa , Técnicas Imunoenzimáticas , Inflamação/metabolismo , Inflamação/patologia , Macaca mulatta , Imageamento por Ressonância Magnética , Masculino , MicroRNAs/administração & dosagem , MicroRNAs/genética , Atividade Motora , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Proteínas Nucleares/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real
13.
Mol Cell Endocrinol ; 342(1-2): 8-19, 2011 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-21672609

RESUMO

Kisspeptin, the product of the KiSS1 gene, has emerged as a key component of the mechanism by which the hypothalamus controls puberty and reproductive development. It does so by stimulating the secretion of gonadotropin releasing hormone (GnRH). Little is known about the transcriptional control of the KiSS1 gene. Here we show that a set of proteins postulated to be upstream components of a hypothalamic network involved in controlling female puberty regulates KiSS1 transcriptional activity. Using RACE-PCR we determined that transcription of KiSS1 mRNA is initiated at a single transcription start site (TSS) located 153-156bp upstream of the ATG translation initiation codon. Promoter assays performed using 293 MSR cells showed that the KiSS1 promoter is activated by TTF1 and CUX1-p200, and repressed by EAP1, YY1, and CUX1-p110. EAP1 and CUX-110 were also repressive in GT1-7 cells. All four TFs are recruited in vivo to the KiSS1 promoter and are expressed in kisspeptin neurons. These results suggest that expression of the KiSS1 gene is regulated by trans-activators and repressors involved in the system-wide control of mammalian puberty.


Assuntos
Regulação da Expressão Gênica , Kisspeptinas/genética , Transcrição Gênica , Imunoprecipitação da Cromatina , Feminino , Células HeLa , Humanos , Hipotálamo/metabolismo , Regiões Promotoras Genéticas/genética , Puberdade/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transativadores/metabolismo , Sítio de Iniciação de Transcrição
14.
Endocrinology ; 152(6): 2353-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21486931

RESUMO

We previously identified synaptic cell adhesion molecule 1 (SynCAM1) as a component of a genetic network involved in the hypothalamic control of female puberty. Although it is well established that SynCAM1 is a synaptic adhesion molecule, its contribution to hypothalamic function is unknown. Here we show that, in addition to the expected neuronal localization illustrated by its presence in GnRH neurons, SynCAM1 is expressed in hypothalamic astrocytes. Cell adhesion assays indicated that SynCAM is recognized by both GnRH neurons and astrocytes as an adhesive partner and promotes cell-cell adhesiveness via homophilic, extracellular domain-mediated interactions. Alternative splicing of the SynCAM1 primary mRNA transcript yields four mRNAs encoding membrane-spanning SynCAM1 isoforms. Variants 1 and 4 are predicted to be both N and O glycosylated. Hypothalamic astrocytes and GnRH-producing GT1-7 cells express mainly isoform 4 mRNA, and sequential N- and O-deglycosylation of proteins extracted from these cells yields progressively smaller SynCAM1 species, indicating that isoform 4 is the predominant SynCAM1 variant expressed in astrocytes and GT1-7 cells. Neither cell type expresses the products of two other SynCAM genes (SynCAM2 and SynCAM3), suggesting that SynCAM-mediated astrocyte-astrocyte and astrocyte-GnRH neuron adhesiveness is mostly mediated by SynCAM1 homophilic interactions. When erbB4 receptor function is disrupted in astrocytes, via transgenic expression of a dominant-negative erbB4 receptor form, SynCAM1-mediated adhesiveness is severely compromised. Conversely, SynCAM1 adhesive behavior is rapidly, but transiently, enhanced in astrocytes by ligand-dependent activation of erbB4 receptors, suggesting that erbB4-mediated events affecting SynCAM1 function contribute to regulate astrocyte adhesive communication.


Assuntos
Astrócitos/citologia , Moléculas de Adesão Celular/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Hipotálamo/metabolismo , Imunoglobulinas/metabolismo , Neurônios/citologia , Sequência de Aminoácidos , Animais , Astrócitos/metabolismo , Adesão Celular , Molécula 1 de Adesão Celular , Moléculas de Adesão Celular/genética , Comunicação Celular , Linhagem Celular , Feminino , Imunoglobulinas/genética , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Neurônios/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais
15.
Endocr Dev ; 17: 44-51, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19955755

RESUMO

The initiation of mammalian puberty requires an increased pulsatile release of gonadotropin-releasing hormone (GnRH) from the hypothalamus. This increase is brought about by changes in transsynaptic and glial-neuronal communication. Coordination of these cellular interactions likely requires the participation of sets of genes hierarchically arranged within functionally connected networks. Using high throughput, genetic, molecular and bioinformatics strategies, in combination with a systems biology approach, three transcriptional regulators of the pubertal process have been identified, and the structure of at least one hypothalamic gene network has been proposed. A genomewide analysis of hypothalamic DNA methylation revealed profound changes in methylation patterns associated with the onset of female puberty. Pharmacological disruption of two epigenetic marks associated with gene silencing (DNA methylation and histone deacetylation) resulted in pubertal failure, instead of advancing the onset of puberty, suggesting that disruption of these two silencing mechanisms leads to activation of repressor genes whose expression would normally decrease at puberty. These observations suggest that the genetic underpinnings of puberty are polygenic rather than specified by a single gene, and that epigenetic mechanisms may provide coordination and transcriptional plasticity to this genetic network.


Assuntos
Puberdade/fisiologia , Epigenômica , Feminino , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Humanos , Hipotálamo/fisiologia , Masculino , Puberdade/genética
16.
Glia ; 57(4): 362-79, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18803307

RESUMO

Studies in rodents have shown that astroglial erbB tyrosine kinase receptors are key regulatory elements in neuron-glia communication. Although both astrocytes and deregulation of erbB functions have been implicated in the pathogenesis of many common human brain disorders, erbB signaling in native human brain astrocytes has never been explored. Taking advantage of our ability to perform primary cultures from the cortex and the hypothalamus of human fetuses, we conducted a thorough analysis of erbB signaling in human astrocytes. We showed that human cortical astrocytes express erbB1, erbB2, and erbB3, whereas human hypothalamic astrocytes express erbB1, erbB2, and erbB4 receptors. Ligand-dependent activation of different erbB receptor heterodimeric complexes in these two populations of astrocytes translated into different morphological and proliferative responses. Although morphological plasticity was more pronounced in hypothalamic astrocytes than in cortical astrocytes, the former showed a lower mitogenic potential. Decreasing erbB4 expression via siRNA-mediated gene knockdown revealed that erbB4 constitutively restrains basal proliferative activity in hypothalamic astrocytes. We further show that treatment of human astrocytes with a protein kinase C activator results in rapid tyrosine phosphorylation of erbB receptors that involves cleavage of endogenous membrane bound erbB ligands by metalloproteinases. Together, these results indicate that erbB signaling in primary human brain astrocytes is functional, region-specific, and can be activated in a paracrine and/or autocrine manner. In addition, by revealing that some aspects of astroglial erbB signaling are different between human and rodents, our results provide a molecular framework to explore the potential involvement of astroglial erbB signaling deregulation in human brain disorders.


Assuntos
Astrócitos/fisiologia , Córtex Cerebral/citologia , Receptores ErbB/metabolismo , Hipotálamo/citologia , Transdução de Sinais/fisiologia , Análise de Variância , Bromodesoxiuridina , Proliferação de Células , Células Cultivadas , Receptores ErbB/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Feto , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imunoprecipitação/métodos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuregulina-1/farmacologia , RNA Interferente Pequeno/farmacologia , Receptor ErbB-4 , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador alfa/farmacologia , Tirosina/metabolismo , Vimentina/metabolismo
17.
Endocrinology ; 150(4): 1870-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19022886

RESUMO

A gene termed EAP1 (enhanced at puberty 1) was recently identified as a transcriptional regulator of female neuroendocrine reproductive function. We have now used in vivo and in vitro assays, and the female rat as an animal model, to determine whether Eap1 gene expression is regulated by ovarian steroids. Eap1 mRNA abundance decreases in both the hypothalamus and cerebral cortex during the infantile-juvenile phases of development, but it increases selectively in the hypothalamus at puberty, suggesting that in contrast to the general decline in expression observed in immature animals, the region-specific increase in Eap1 mRNA levels that occurs at puberty might be elicited by ovarian steroids. This is, however, not the case, because hypothalamic Eap1 mRNA levels increase at the expected time of puberty in rats ovariectomized at the beginning of the juvenile period. Although a subpopulation of EAP1-containing cells in the medial basal hypothalamus (MBH) and preoptic area express estrogen receptor-alpha (ERalpha), the 5'-flanking region of the rat Eap1 (rEap1) gene does not contain a complete estrogen-responsive element, and no such estrogen-responsive element is detected within 100 kb of the rEap1 locus. Functional promoter assays showed that neither estradiol (E(2)) alone nor a combination of E(2) plus progesterone increases rEap1 gene transcription. Likewise, E(2) administered to ovariectomized immature rats elicited a robust surge of LH but increased neither preoptic area nor MBH Eap1 mRNA levels. E(2)/progesterone-treated rats showed a massive elevation in plasma LH but only a modest increase in Eap1 mRNA levels, limited to the MBH. These results indicate that hypothalamic Eap1 expression is not directly controlled by ovarian steroids and suggest that Eap1 expression increases at puberty driven by ovary-independent, centrally initiated events.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hipotálamo/metabolismo , Ovário/metabolismo , Esteroides/farmacologia , Transativadores/genética , Transativadores/metabolismo , Animais , Células Cultivadas , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Ovariectomia , Gravidez , Progesterona/farmacologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro , Ratos , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
18.
Neuroendocrinology ; 88(3): 199-211, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18547942

RESUMO

NELL2, a protein containing EGF-like repeats, is almost exclusively expressed in the nervous system. In the mammalian brain, NELL2 expression is mostly neuronal. NELL2 was previously found to be a secreted protein that functions during embryonic development as a neuronal differentiation and survival factor. We now show that the Nell2 gene is selectively expressed in the two major subtypes of glutamatergic neurons described in the postnatal brain: those containing the vesicular glutamate transporter 1 and those expressing vesicular glutamate transporter 2. No Nell2 mRNA is detected in GABAergic neurons. Likewise, GnRH neurons are devoid of NELL2. During prepubertal development of the female rat, Nell2 mRNA abundance increases selectively in the medial basal hypothalamus, reaching maximal values at the end of the juvenile period, to decline at the time of puberty to intermediate levels. Similar, but less pronounced changes are observed in the preoptic area, but they are absent in the cerebral cortex. A well-established glutamatergic function in the neuroendocrine brain is to enhance release of GnRH, the neurohormone controlling sexual development and the time of puberty. In vivo disruption of NELL2 synthesis via intraventricular administration of antisense oligodeoxynucleotides reduced GnRH release from the medial basal hypothalamus and delayed the initiation of female puberty. These results identify NELL2 as a new component of glutamatergic neurons and provide evidence for its physiological involvement in a major, glutamate-dependent, process of neuroendocrine regulation.


Assuntos
Ácido Glutâmico/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Maturidade Sexual/fisiologia , Animais , Feminino , Expressão Gênica , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/genética , Sistemas Neurossecretores/metabolismo , Sistemas Neurossecretores/fisiologia , Especificidade de Órgãos/genética , RNA Mensageiro/metabolismo , Ratos , Maturidade Sexual/genética
19.
Endocrinology ; 148(11): 5147-61, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17615149

RESUMO

Much has been learned in recent years about the central mechanisms controlling the initiation of mammalian puberty. It is now clear that this process requires the interactive participation of several genes. Using a combination of high throughput, molecular, and bioinformatics strategies, in combination with a system biology approach, we singled out from the hypothalamus of nonhuman primates and rats a group of related genes whose expression increases at the time of female puberty. Although these genes [henceforth termed tumor-related genes (TRGs)] have diverse cellular functions, they share the common feature of having been earlier identified as involved in tumor suppression/tumor formation. A prominent member of this group is KiSS1, a gene recently shown to be essential for the occurrence of puberty. Cis-regulatory analysis revealed the presence of a hierarchically arranged gene set containing five major hubs (CDP/CUTL1, MAF, p53, YY1, and USF2) controlling the network at the transcriptional level. In turn, these hubs are heavily connected to non-TRGs involved in the transcriptional regulation of the pubertal process. TRGs may be expressed in the mammalian hypothalamus as components of a regulatory gene network that facilitates and integrates cellular and cell-cell communication programs required for the acquisition of female reproductive competence.


Assuntos
Redes Reguladoras de Genes , Genes Neoplásicos , Hipotálamo/metabolismo , Maturidade Sexual/genética , Regulação para Cima , Região 5'-Flanqueadora , Animais , Sítios de Ligação , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Macaca mulatta , Modelos Biológicos , Sistemas Neurossecretores/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo
20.
J Neurosci ; 26(51): 13167-79, 2006 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-17182767

RESUMO

Thyroid transcription factor 1 (TTF1) [also known as Nkx2.1 (related to the NK-2 class of homeobox genes) and T/ebp (thyroid-specific enhancer-binding protein)], a homeodomain gene required for basal forebrain morphogenesis, remains expressed in the hypothalamus after birth, suggesting a role in neuroendocrine function. Here, we show an involvement of TTF1 in the control of mammalian puberty and adult reproductive function. Gene expression profiling of the nonhuman primate hypothalamus revealed that TTF1 expression increases at puberty. Mice in which the Ttf1 gene was ablated from differentiated neurons grew normally and had normal basal ganglia/hypothalamic morphology but exhibited delayed puberty, reduced reproductive capacity, and a short reproductive span. These defects were associated with reduced hypothalamic expression of genes required for sexual development and deregulation of a gene involved in restraining puberty. No extrapyramidal impairments associated with basal ganglia dysfunction were apparent. Thus, although TTF1 appears to fulfill only a morphogenic function in the ventral telencephalon, once this function is satisfied in the hypothalamus, TTF1 remains active as part of the transcriptional machinery controlling female sexual development.


Assuntos
Gânglios da Base/fisiologia , Diferenciação Celular/genética , Deleção de Genes , Neurônios/citologia , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Reprodução/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Animais , Gânglios da Base/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Hipotálamo/citologia , Hipotálamo/fisiologia , Macaca mulatta , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neurônios/fisiologia , Proteínas Nucleares/deficiência , Comportamento Sexual Animal/fisiologia , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA