Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
Proc Natl Acad Sci U S A ; 108(38): 16104-9, 2011 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-21896757

RESUMO

Astrocytes in the hypothalamus release prostaglandin E(2) (PGE(2)) in response to cell-cell signaling initiated by neurons and glial cells. Upon release, PGE(2) stimulates the secretion of gonadotropin-releasing hormone (GnRH), the neuropeptide that controls reproduction, from hypothalamic neuroendocrine neurons. Whether this effect on GnRH secretion is accompanied by changes in the firing behavior of these neurons is unknown. Using patch-clamp recording we demonstrate that PGE(2) exerts a dose-dependent postsynaptic excitatory effect on GnRH neurons. These effects are mimicked by an EP2 receptor agonist and attenuated by protein kinase A (PKA) inhibitors. The acute blockade of prostaglandin synthesis by indomethacin (INDO) or the selective inhibition of astrocyte metabolism by fluoroacetate (FA) suppresses the spontaneous firing activity of GnRH neurons in brain slices. Similarly, GnRH neuronal activity is reduced in mice with impaired astrocytic PGE(2) release due to defective erbB signaling in astrocytes. These results indicate that astrocyte-to-neuron communication in the hypothalamus is essential for the activity of GnRH neurons and suggest that PGE(2) acts as a gliotransmitter within the GnRH neurosecretory system.


Assuntos
Astrócitos/metabolismo , Dinoprostona/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Alprostadil/análogos & derivados , Alprostadil/farmacologia , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Hormônio Liberador de Gonadotropina/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Indometacina/farmacologia , Isoquinolinas/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Inibidores de Proteínas Quinases/farmacologia , Receptores de Prostaglandina E Subtipo EP2/agonistas , Sulfonamidas/farmacologia
5.
Horm Behav ; 64(2): 175-86, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23998662

RESUMO

This article is part of a Special Issue "Puberty and Adolescence". Puberty is a major developmental milestone controlled by the interaction of genetic factors and environmental cues of mostly metabolic and circadian nature. An increased pulsatile release of the decapeptide gonadotropin releasing hormone (GnRH) from hypothalamic neurosecretory neurons is required for both the initiation and progression of the pubertal process. This increase is brought about by coordinated changes that occur in neuronal and glial networks associated with GnRH neurons. These changes ultimately result in increased neuronal and glial stimulatory inputs to the GnRH neuronal network and a reduction of transsynaptic inhibitory influences. While some of the major players controlling pubertal GnRH secretion have been identified using gene-centric approaches, much less is known about the system-wide control of the overall process. Because the pubertal activation of GnRH release involves a diversity of cellular phenotypes, and a myriad of intracellular and cell-to-cell signaling molecules, it appears that the overall process is controlled by a highly coordinated and interactive regulatory system involving hundreds, if not thousands, of gene products. In this article we will discuss emerging evidence suggesting that these genes are arranged as functionally connected networks organized, both internally and across sub-networks, in a hierarchical fashion. According to this concept, the core of these networks is composed of transcriptional regulators that, by directing expression of downstream subordinate genes, provide both stability and coordination to the cellular networks involved in initiating the pubertal process. The integrative response of these gene networks to external inputs is postulated to be coordinated by epigenetic mechanisms.


Assuntos
Redes Reguladoras de Genes , Sistemas Neurossecretores/fisiologia , Primatas/fisiologia , Maturidade Sexual/genética , Biologia de Sistemas/métodos , Animais , Epigênese Genética/fisiologia , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Ratos
6.
Mol Ther ; 19(6): 1058-69, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21487395

RESUMO

Other labs have previously reported the ability of adeno-associated virus serotype 9 (AAV9) to cross the blood-brain barrier (BBB). In this report, we carefully characterized variables that might affect AAV9's efficiency for central nervous system (CNS) transduction in adult mice, including dose, vehicle composition, mannitol coadministration, and use of single-stranded versus self-complementary AAV. We report that AAV9 is able to transduce approximately twice as many neurons as astrocytes across the entire extent of the adult rodent CNS at doses of 1.25 × 10¹², 1 × 10¹³, and 8 × 10¹³ vg/kg. Vehicle composition or mannitol coadministration had only modest effects on CNS transduction, suggesting AAV9 crosses the BBB by an active transport mechanism. Self-complementary vectors were greater than tenfold more efficient than single-stranded vectors. When this approach was applied to juvenile nonhuman primates (NHPs) at the middle dose (9-9.5 × 10¹² vg/kg) tested in mice, a reduction in peripheral organ and brain transduction was observed compared to mice, along with a clear shift toward mostly glial transduction. Moreover, the presence of low levels of pre-existing neutralizing antibodies (NAbs) mostly occluded CNS and peripheral transduction using this delivery approach. Our results indicate that high peripheral tropism, limited neuronal transduction in NHPs, and pre-existing NAbs represent significant barriers to human translation of intravascular AAV9 delivery.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Animais , Feminino , Células HEK293 , Células HeLa , Humanos , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase , Primatas
7.
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
8.
Mol Cells ; 45(8): 537-549, 2022 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-35950455

RESUMO

Preproenkephalin (PPE) is a precursor molecule for multiple endogenous opioid peptides Leu-enkephalin (ENK) and Met-ENK, which are involved in a wide variety of modulatory functions in the nervous system. Despite the functional importance of ENK in the brain, the effect of brain-derived factor(s) on PPE expression is unknown. We report the dual effect of neural epidermal growth factor (EGF)-likelike 2 (NELL2) on PPE gene expression. In cultured NIH3T3 cells, transfection of NELL2 expression vectors induced an inhibition of PPE transcription intracellularly, in parallel with downregulation of protein kinase C signaling pathways and extracellular signal-regulated kinase. Interestingly, these phenomena were reversed when synthetic NELL2 was administered extracellularly. The in vivo disruption of NELL2 synthesis resulted in an increase in PPE mRNA level in the rat brain, suggesting that the inhibitory action of intracellular NELL2 predominates the activation effect of extracellular NELL2 on PPE gene expression in the brain. Biochemical and molecular studies with mutant NELL2 structures further demonstrated the critical role of EGF-like repeat domains in NELL2 for regulation of PPE transcription. These are the first results to reveal the spatio-specific role of NELL2 in the homeostatic regulation of PPE gene expression.


Assuntos
Fator de Crescimento Epidérmico , Proteínas do Tecido Nervoso , Animais , Encefalinas , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/farmacologia , Expressão Gênica , Camundongos , Células NIH 3T3 , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Precursores de Proteínas , Ratos
9.
J Biol Chem ; 285(32): 25074-84, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20538601

RESUMO

NELL2 (neural tissue-specific epidermal growth factor-like repeat domain-containing protein) is a secreted glycoprotein that is predominantly expressed in neural tissues. We reported previously that NELL2 mRNA abundance in brain is increased by estrogen (E2) treatment and that NELL2 is involved in the E2-dependent organization of a sexually dimorphic nucleus in the preoptic area. In this study we cloned the mouse NELL2 promoter and found it to contain two half-E2 response elements. Electrophoretic mobility shift assays and promoter assays showed that E2 and its receptors (ERalpha and ERbeta) stimulated NELL2 transcription by binding to the two half-E2 response elements. Hippocampal neuroprogenitor HiB5 cells expressing recombinant NELL2 showed increased cell survival under cell death-inducing conditions. Blockade of endogenous synthesis of NELL2 in HiB5 cells abolished the cell survival effect of E2 and resulted in a decrease in phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2). These data suggest that the NELL2 gene is trans-activated by E2 and contributes to mediating the survival promoting effects of E2 via intracellular signaling pathway of ERK.


Assuntos
Estrogênios/metabolismo , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/metabolismo , Animais , Apoptose , Morte Celular , Primers do DNA/genética , Hipocampo/metabolismo , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Modelos Biológicos , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Elementos de Resposta , Transdução de Sinais
10.
J Neurosci Res ; 89(6): 840-51, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21394759

RESUMO

Fxyd1 encodes a trans-membrane protein that modulates Na(+) ,K(+) -ATPase activity and is a substrate for multiple protein kinases. Fxyd1 expression is repressed by methyl CpG-binding protein 2 (Mecp2) in the frontal cortex (FC) but not in the cerebellum (CB) of the mouse brain. Consistently with these observations, FXYD1 mRNA abundance is increased in the FC of Rett syndrome (RTT) patients with MECP2 mutations. Because Fxyd1 is implicated in the regulation of neuronal excitability, understanding how Fxyd1 expression is controlled is important. Here we report that basal expression of Fxyd1a and Fxyd1b, the two main alternatively spliced forms of Fxyd1 mRNA, is lower in the FC than in the CB. This difference is accompanied by increased Mecp2 recruitment to the promoter region of these two Fxyd1 mRNA forms. DNA methylation of both promoters is more frequent in the FC than in the CB, and in both cases the most frequently methylated CpG dinucleotides are adjacent to [A/T](4) sequences required for high-affinity Mecp2 binding. Consistently with these features of epigenetic silencing, histone 3 acetylated at lysines 9 and 14 (H3K9/14ac) and histone 3 methylated at lysine 4 (H3K4me3), both activating histone marks, were associated with the Fxyd1 promoter to a lesser degree in the FC than in the CB. These results indicate that differential Fxyd1 expression in these two brain regions is, at least in part, regulated by an epigenetic mechanism involving increased DNA methylation of the two alternative Fxyd1 promoters, enhanced Mecp2 recruitment, and reduced association of activating histones.


Assuntos
Encéfalo/metabolismo , Epigênese Genética/genética , Regulação da Expressão Gênica/genética , Proteínas de Membrana/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Fosfoproteínas/metabolismo , Animais , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Metilação de DNA , Feminino , Histonas/metabolismo , Proteínas de Membrana/genética , Proteína 2 de Ligação a Metil-CpG/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas/genética , RNA Mensageiro/metabolismo
11.
Reproduction ; 142(2): 319-31, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21646391

RESUMO

Excessive nerve growth factor (NGF) production by the ovary, achieved via a transgenic approach, results in arrested antral follicle growth, reduced ovulatory capacity, and a predisposition to cyst formation in response to mildly elevated LH levels. Two salient features in these mutant mice (termed 17NF) are an elevated production of 17α-hydroxyprogesterone (17-OHP(4)), testosterone, and estradiol (E(2)) in response to gonadotropins, and an increased frequency of granulosa cell (GC) apoptosis. In this study, we show that the increase in steroidal response is associated with enhanced expression of Cyp17a1, Hsd17b, and Cyp19a1, which encode the enzymes catalyzing the synthesis of 17-OHP(4), testosterone, and E(2) respectively. Using a proteomic approach, we identified stathmin (STMN1), as a protein that is overproduced in 17NF ovaries. In its phosphorylated state, STMN1 mediates a cell death signal initiated by tumor necrosis factor α (TNF). STMN1 is expressed in GCs and excessive NGF increases its abundance as well as that of its forms phosphorylated at serine (Ser) 16, 25, and 38. TNF synthesis is also increased in 17NF ovaries, and this change is abolished by blocking neurotrophic tyrosine kinase receptors. Inhibiting TNF actions in vivo by administering a soluble TNF receptor prevented the increase in total and phosphorylated STMN1 production, as well as GC apoptosis in NGF-overproducing ovaries. These results indicate that an excess of NGF in the ovary promotes steroidogenesis by enhancing the expression of enzyme genes involved in 17-OHP(4), testosterone, and E(2) synthesis, and causes GC apoptosis by activating a TNF/ STMN1-mediated cell death pathway.


Assuntos
Apoptose , Células da Granulosa/metabolismo , Fator de Crescimento Neural/metabolismo , Ovário/metabolismo , Transdução de Sinais , Estatmina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , 17-Hidroxiesteroide Desidrogenases/genética , 17-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Aromatase/genética , Aromatase/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônios Esteroides Gonadais/metabolismo , Gonadotropinas/farmacologia , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Fator de Crescimento Neural/genética , Ovário/efeitos dos fármacos , Ovário/enzimologia , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estatmina/genética , Esteroide 17-alfa-Hidroxilase/genética , Esteroide 17-alfa-Hidroxilase/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores
12.
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
13.
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
14.
Eur J Neurosci ; 32(12): 2003-10, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21143655

RESUMO

Mammalian puberty is initiated by an increased pulsatile release of the neuropeptide gonadotropin-releasing hormone (GnRH) from hypothalamic neuroendocrine neurons. Although this increase is primarily set in motion by neuronal networks synaptically connected to GnRH neurons, glial cells contribute to the process via at least two mechanisms. One involves production of growth factors acting via receptors endowed with either serine-threonine kinase or tyrosine kinase activity. The other involves plastic rearrangements of glia-GnRH neuron adhesiveness. Growth factors of the epidermal growth factor family acting via erbB receptors play a major role in glia-to-GnRH neuron communication. In turn, neurons facilitate astrocytic erbB signaling via glutamate-dependent cleavage of erbB ligand precursors. The genetic disruption of erbB receptors delays female sexual development due to impaired erbB ligand-induced glial prostaglandin E(2) release. The adhesiveness of glial cells to GnRH neurons involves at least two different cell-cell communication systems endowed with both adhesive and intracellular signaling capabilities. One is provided by synaptic cell adhesion molecule (SynCAM1), which establishes astrocyte-GnRH neuron adhesiveness via homophilic interactions and the other involves the heterophilic interaction of neuronal contactin with glial receptor-like protein tyrosine phosphatase-ß. These findings indicate that the interaction of glial cells with GnRH neurons involves not only secreted bioactive molecules, but also cell-surface adhesive proteins able to set in motion intracellular signaling cascades.


Assuntos
Neuroglia/metabolismo , Neurônios/metabolismo , Sistemas Neurossecretores/fisiologia , Puberdade/fisiologia , Animais , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Feminino , Ácido Glutâmico/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Sistemas Neurossecretores/citologia , Receptor ErbB-2/metabolismo , Receptor ErbB-4 , Maturidade Sexual/fisiologia , Transdução de Sinais/fisiologia
15.
J Clin Invest ; 117(8): 2145-54, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17627301

RESUMO

The initiation of mammalian puberty and the maintenance of female reproductive cycles are events controlled by hypothalamic neurons that secrete the decapeptide gonadotropin-releasing hormone (GnRH). GnRH secretion is, in turn, controlled by changes in neuronal and glial inputs to GnRH-producing neurons. The hierarchical control of the process is unknown, but it requires coordinated regulation of these cell-cell interactions. Here we report the functional characterization of a gene (termed enhanced at puberty 1 [EAP1]) that appears to act as an upstream transcriptional regulator of neuronal networks controlling female reproductive function. EAP1 expression increased selectively at puberty in both the nonhuman primate and rodent hypothalamus. EAP1 encoded a nuclear protein expressed in neurons involved in the inhibitory and facilitatory control of reproduction. EAP1 transactivated genes required for reproductive function, such as GNRH1, and repressed inhibitory genes, such as preproenkephalin. It contained a RING finger domain of the C3HC4 subclass required for this dual transcriptional activity. Inhibition of EAP1 expression, targeted to the rodent hypothalamus via lentivirus-mediated delivery of EAP1 siRNAs, delayed puberty, disrupted estrous cyclicity, and resulted in ovarian abnormalities. These results suggest that EAP1 is a transcriptional regulator that, acting within the neuroendocrine brain, contributes to controlling female reproductive function.


Assuntos
Ciclo Estral/metabolismo , Hormônio Liberador de Gonadotropina/biossíntese , Sistema Hipotálamo-Hipofisário/metabolismo , Proteínas de Neoplasias/biossíntese , Neurônios/metabolismo , Precursores de Proteínas/biossíntese , Maturidade Sexual , Fatores de Transcrição/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Regulação para Baixo/genética , Ciclo Estral/genética , Feminino , Hormônio Liberador de Gonadotropina/genética , Humanos , Sistema Hipotálamo-Hipofisário/citologia , Lentivirus , Macaca mulatta , Proteínas de Neoplasias/genética , Neuroglia/citologia , Neuroglia/patologia , Neurônios/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ovário/citologia , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Precursores de Proteínas/genética , Estrutura Terciária de Proteína/genética , Ratos , Ratos Sprague-Dawley , Securina , Maturidade Sexual/genética , Fatores de Transcrição/genética , Transdução Genética
16.
Methods ; 49(1): 70-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19559089

RESUMO

Non-human primates (NHPs) are an invaluable resource for the study of genetic regulation of disease mechanisms. The main disadvantage of using NHPs as a preclinical model of human disease is the difficulty of manipulating the monkey genome using conventional gene modifying strategies. Lentiviruses offer the possibility of circumventing this difficulty because they can infect and transduce either dividing or nondividing cells, without producing an immune response. In addition, lentiviruses can permanently integrate into the genome of host cells, and are able to maintain long-term expression. In this article we describe the lentiviral vectors that we use to both express transgenes and suppress expression of endogenous genes via RNA interference (RNAi) in NHPs. We also discuss the safety features of currently available vectors that are especially important when lentiviral vectors are used in a species as closely related to humans as NHPs. Finally, we describe in detail the lentiviral vector production protocol we use and provide examples of how the vector can be employed to target peripheral tissues and the brain.


Assuntos
Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Lentivirus/genética , Primatas/genética , Primatas/metabolismo , Animais , Humanos
17.
Sci Rep ; 10(1): 10073, 2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32572045

RESUMO

Cats are a critical pre-clinical model for studying adeno-associated virus (AAV) vector-mediated gene therapies. A recent study has described the high prevalence of anti-AAV neutralizing antibodies among domestic cats in Switzerland. However, our knowledge of pre-existing humoral immunity against various AAV serotypes in cats is still limited. Here, we show that, although antibodies binding known AAV serotypes (AAV1 to AAV11) are prevalent in cats living in the Northeastern United States, these antibodies do not necessarily neutralize AAV infectivity. We analyzed sera from 35 client-owned, 20 feral, and 30 specific pathogen-free (SPF) cats for pre-existing AAV-binding antibodies against the 11 serotypes. Antibody prevalence was 7 to 90% with an overall median of 50%. The AAV-binding antibodies showed broad reactivities with other serotypes. Of 44 selected antibodies binding AAV2, AAV6 or AAV9, none exhibited appreciable neutralizing activities. Instead, AAV6 or AAV9-binding antibodies showed a transduction-enhancing effect. AAV6-binding antibodies were highly prevalent in SPF cats (83%), but this was primarily due to cross-reactivity with preventive vaccine-induced anti-feline panleukopenia virus antibodies. These results indicate that prevalent pre-existing immunity in cats is not necessarily inhibitory to AAV and highlight a substantial difference in the nature of AAV-binding antibodies in cats living in geographically different regions.


Assuntos
Anticorpos Antivirais/metabolismo , Dependovirus/imunologia , Soro/imunologia , Animais , Anticorpos Neutralizantes/metabolismo , Gatos , Dependovirus/classificação , Imunidade Humoral , New England , Sorogrupo , Suíça , Transdução Genética
18.
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
19.
Reproduction ; 138(1): 131-40, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19357131

RESUMO

Recent studies have demonstrated that neurotrophins (NTs) and their NTRK tyrosine kinase receptors, thought to be exclusively required for the development of the nervous system, are also involved in controlling ovarian development. Here, we show that primordial follicle formation is decreased in the absence of nerve growth factor (NGF) or its receptor NTRK1, and in the absence of NTRK2, the receptor for neurotrophin-4 (NTF4) and brain-derived neurotrophic factor (BDNF). This deficiency is not due to premature oocyte loss, because the ovaries of Ntrk1(-/-) and Ntrk2(-/-) mice do not show an increased rate of oocyte death antedating the initiation of folliculogenesis. Moreover, exposure of NGF-deficient ovaries to NGF rescues the defect in follicular assembly, if NTRK1 receptors are present, suggesting that the absence of NTs causes a delay, and not an irretrievable loss, of follicle formation. Both the number of secondary follicles and FSH receptor (FSHR) expression are diminished in Ntrk1- and Ntrk2-null ovaries, but not in ovaries lacking the common NT receptor NGFR. Transient exposure of wild-type ovaries to NTF4 increases Fshr gene expression and enhances the ability of the ovary to respond to FSH with formation of cyclin D2, a cell cycle protein mediating the proliferative actions of FSH in the ovary. These results indicate that both NTRK1 and NTRK2 receptors are necessary for the timely assembly of primordial follicles and for sustaining early follicular development. They also suggest that a mechanism by which NTRK2 receptors facilitate subsequent follicle development is by inducing the formation of functional FSHR.


Assuntos
Glicoproteínas de Membrana/metabolismo , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Ovário/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptor trkA/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Ciclina D2/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Crescimento Neural/deficiência , Fator de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Oócitos/patologia , Folículo Ovariano/patologia , Ovário/patologia , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Receptor trkA/deficiência , Receptor trkA/genética , Receptores do FSH/metabolismo , Transdução de Sinais , Fatores de Tempo , Técnicas de Cultura de Tecidos
20.
Endocrinology ; 149(1): 50-6, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17947351

RESUMO

A substantial fraction of the noradrenergic innervation targeting the mammalian ovary is provided by neurons of the celiac ganglion. Although studies in the rat have shown that noradrenergic nerves reach the ovary near the time of birth, it is unknown how the functional capacity of this innervation unfolds during postnatal ovarian development. To address this issue, we assessed the ability of the developing ovary to incorporate and release (3)H-norepinephrine. Incorporation of (3)H-norepinephrine was low during the first 3 wk of postnatal life, but pharmacological inhibition of norepinephrine (NE) neuronal uptake with cocaine showed that an intact transport mechanism for NE into nerve terminals is already in place by the first week after birth. Consistent with this functional assessment, the mRNA encoding the NE transporter was also expressed in the celiac ganglion at this time. During neonatal-infantile development [postnatal (PN) d 5-20], the spontaneous, vesicle-independent outflow of recently taken up NE was high, but the NE output in response to K(+)-induced depolarization was low. After PN d 20, spontaneous outflow decreased and the response to K(+) increased markedly, reaching maximal values by the time of puberty. Tyramine-mediated displacement of NE stored in vesicles, which displace vesicular NE, showed that vesicle-dependent NE storage becomes functional by PN d 12 and that vesicular release increases during the juvenile-peripubertal phases of sexual development. These results indicate that vesicular release of NE from ovarian noradrenergic nerves begins to operate by the third week of postnatal life, becoming fully functional near the time of puberty.


Assuntos
Fibras Adrenérgicas/fisiologia , Norepinefrina/metabolismo , Ovário/crescimento & desenvolvimento , Ovário/inervação , Sistema Nervoso Simpático/crescimento & desenvolvimento , Fibras Adrenérgicas/metabolismo , Animais , Animais Recém-Nascidos , Cálcio/farmacologia , Feminino , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Ovário/fisiologia , Ratos , Ratos Sprague-Dawley , Maturidade Sexual/fisiologia , Vesículas Transportadoras/metabolismo , Trítio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA