Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Medicinas Complementares
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.
Glia ; 67(2): 309-320, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30485552

RESUMO

Gonadotropin releasing hormone (GnRH)-secretion is not only regulated by neuronal factors but also by astroglia cells via growth factors and ErbB receptors of the epidermal growth factor family. Studies in transgenic mice carrying mutations in the ErbB receptor system experience impaired reproductive capacity. In addition, some of these animals show a typical skin phenotype with wavy hair and curly whiskers. The rat strain SPRD-CU3 (CU3), examined in this study, displays a similar skin phenotype and a significant impairment of the timing of puberty onset and reproductive performance, suggesting a disruption in the astrocytic to GnRH neuronal communication. To address this issue, we analyzed astrocytic prostaglandin E2 (PGE2 ) release from primary hypothalamic astrocytic cell cultures after stimulation with transforming growth factor α (TGFα), ligand for ErbB1/ErbB2, or Neuregulin 1 beta 2 (NRG1ß2 ), ligand for ErbB4/ErbB2 signaling pathway. Compared to cultures from wild type animals, astrocytic cultures from CU3 rats were unable to respond to NRG stimulation, suggesting a disruption of the ErbB4/ErbB2 signaling pathway. This is confirmed by mutational analysis of ErbB4 that revealed a single point mutation at 3125 bp resulting in an amino acid change from proline to glutamine located at the carboxy-terminal region. As a consequence, substantial conformational changes occur in the transmembrane and intracellular domain of the protein, affecting the ability to form a receptor dimer with a partner and the ability to function as a transcriptional regulator. Thus, astroglia to GnRH neuronal signaling via ErbB4 is essential of timely onset of puberty and reproductive function.


Assuntos
Astrócitos/efeitos dos fármacos , Dinoprostona/metabolismo , Transtornos do Desenvolvimento Sexual/patologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurregulinas/farmacologia , Neurônios/metabolismo , Receptor ErbB-4/genética , Animais , Astrócitos/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Transtornos do Desenvolvimento Sexual/tratamento farmacológico , Transtornos do Desenvolvimento Sexual/genética , Transtornos do Desenvolvimento Sexual/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/efeitos da radiação , Hipotálamo/citologia , Modelos Moleculares , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Mutação Puntual/genética , Ratos , Ratos Transgênicos , Receptor ErbB-4/metabolismo , Fator de Crescimento Transformador alfa/metabolismo
3.
Reprod Toxicol ; 44: 73-84, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24211603

RESUMO

Endocrine disrupting chemicals have been shown to alter the pubertal process. The controlling levels of the Gonadotropin releasing hormone (GnRH) network involve GnRH itself, KiSS1, and the transcriptional regulators enhanced at puberty 1 (EAP1), Thyroid Transcription Factor 1 (TTF1), and Yin Yang 1 (YY1). While Genistein and Bisphenol A (BPA) have been shown to advance the advent of puberty, exposure to Dioxin delayed pubertal onset. Utilizing in vitro approaches, we observed that Genistein and BPA suppress inhibitory and activate stimulatory components of the GnRH network, while Dioxin exhibit an inhibitory effect at all regulatory hierarchical levels of the GnRH network. It repressed KiSS1, Gnrh, Ttf1 and Yy1 transcription via the xenobiotic response element (XRE), while EAP1 was not affected. Therefore, EDCs alter the neuroendocrine GnRH regulatory network at all hierarchical levels.


Assuntos
Disruptores Endócrinos/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Kisspeptinas/genética , Fitoestrógenos/toxicidade , Securina/genética , Animais , Compostos Benzidrílicos/toxicidade , Linhagem Celular , Células Cultivadas , Genes Reporter , Genisteína/toxicidade , Humanos , Hipotálamo/citologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenóis/toxicidade , Dibenzodioxinas Policloradas/toxicidade , Regiões Promotoras Genéticas , Ratos Sprague-Dawley , Fator de Transcrição YY1/genética
4.
Reprod Toxicol ; 44: 63-72, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24316331

RESUMO

We studied the effects of neonatal exposure to diethylstilbestrol (DES) on pubertal timing in female rats. We examined associated neuroendocrine changes and effects of prenatal food restriction. Age at vaginal opening was advanced after exposure to 10 µg/kg/d of DES and delayed after 1 µg/kg/d (subcutaneous injections). Using this lower dose, pulsatile GnRH secretion was slower at 25 days of age. Both doses reduced KiSS1 mRNA levels at 15 days of age. Using functional Kisspeptin promoter assay, 1 or 10 µM DES reduced or increased KISS1 transcription, respectively. Leptin stimulatory effect on GnRH secretion in vitro (15 days of age) was reduced after prenatal food restriction and neonatal DES exposure (higher dose), both effects being cumulative. Thus, alterations in pubertal timing by DES neonatally are not unequivocally toward precocity, the level of exposure being critical. We provide evidence of neuroendocrine disruption and interaction with prenatal food availability.


Assuntos
Dietilestilbestrol/toxicidade , Disruptores Endócrinos/toxicidade , Estrogênios não Esteroides/toxicidade , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/genética , Maturidade Sexual/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Linhagem Celular , Ciclo Estral/efeitos dos fármacos , Feminino , Privação de Alimentos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Leptina/sangue , Camundongos , Gravidez , Fenômenos Fisiológicos da Nutrição Pré-Natal , RNA Mensageiro/metabolismo , Ratos Wistar , Vagina/efeitos dos fármacos , Vagina/crescimento & desenvolvimento
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Int J Androl ; 29(1): 256-63; discussion 286-90, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16466547

RESUMO

Sexual development and mature reproductive function are controlled by a handful of neurones that, located in the basal forebrain, produce the decapeptide luteinizing hormone releasing hormone (LHRH). LHRH is released into the portal system that connects the hypothalamus to the pituitary gland and act on the latter to stimulate the synthesis and release of gonadotrophin hormones. The pubertal activation of LHRH release requires coordinated changes in excitatory and inhibitory inputs to LHRH-secreting neurones. These inputs are provided by both transsynaptic and glia-to-neurone communication pathways. Using cellular and molecular approaches, in combination with transgenic animal models and high-throughput procedures for gene discovery, we are gaining new insight into the basic mechanisms underlying this dual control of LHRH secretion and, hence, the initiation of mammalian puberty. Our results suggest that the initiation of puberty requires reciprocal neurone-glia communication involving excitatory amino acids and growth factors, and the coordinated actions of a group of transcriptional regulators that appear to represent a higher level of control governing the pubertal process.


Assuntos
Sistemas Neurossecretores/fisiologia , Puberdade , Maturidade Sexual/fisiologia , Animais , Animais Geneticamente Modificados , Astrócitos , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hipotálamo , Neuroglia , Neurônios , Maturidade Sexual/genética
11.
Endocrinology ; 147(3): 1166-74, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16373420

RESUMO

The initiation of mammalian puberty requires an increase in pulsatile release of GnRH from the hypothalamus. This increase is brought about by coordinated changes in transsynaptic and glial-neuronal communication. As the neuronal and glial excitatory inputs to the GnRH neuronal network increase, the transsynaptic inhibitory tone decreases, leading to the pubertal activation of GnRH secretion. The excitatory neuronal systems most prevalently involved in this process use glutamate and the peptide kisspeptin for neurotransmission/neuromodulation, whereas the most important inhibitory inputs are provided by gamma-aminobutyric acid (GABA)ergic and opiatergic neurons. Glial cells, on the other hand, facilitate GnRH secretion via growth factor-dependent cell-cell signaling. Coordination of this regulatory neuronal-glial network may require a hierarchical arrangement. One level of coordination appears to be provided by a host of unrelated genes encoding proteins required for cell-cell communication. A second, but overlapping, level might be provided by a second tier of genes engaged in specific cell functions required for productive cell-cell interaction. A third and higher level of control involves the transcriptional regulation of these subordinate genes by a handful of upper echelon genes that, operating within the different neuronal and glial subsets required for the initiation of the pubertal process, sustain the functional integration of the network. The existence of functionally connected genes controlling the pubertal process is consistent with the concept that puberty is under genetic control and that the genetic underpinnings of both normal and deranged puberty are polygenic rather than specified by a single gene. The availability of improved high-throughput techniques and computational methods for global analysis of mRNAs and proteins will allow us to not only initiate the systematic identification of the different components of this neuroendocrine network but also to define their functional interactions.


Assuntos
Sistema Endócrino/fisiologia , Neurônios/metabolismo , Sistemas Neurossecretores , Puberdade , Biologia de Sistemas/métodos , Animais , Comunicação Celular , DNA/metabolismo , Genes Supressores de Tumor , Hormônio Liberador de Gonadotropina/metabolismo , Substâncias de Crescimento/metabolismo , Humanos , Hipotálamo/metabolismo , Kisspeptinas , Modelos Biológicos , Neuroglia/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas , RNA Mensageiro/metabolismo , Transdução de Sinais , Software , Sinapses , Transcrição Gênica , Proteínas Supressoras de Tumor
12.
Endocr Dev ; 8: 94-125, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15722620

RESUMO

Central precocious puberty (CPP) is the premature onset of puberty due to a precocious activation of gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus. This condition results in accelerated development of secondary sex characteristics, accelerated bone maturation, impaired final height with disproportioned body appearance and can have a disturbing impact on the psychosocial behavior of children suffering from CPP. It is therefore necessary to assess the hormonal status of children who show pubertal signs before the age 8 years in girls and 9 years in boys. The indication for treatment should be made after evaluating pubertal progression, progression of bone age maturation and final height prognosis, development of reproductive function, and psychosocial adjustment and well-being. This paper summarizes the experience of GnRH agonist treatment, which is momentarily the treatment of choice for central precocious puberty in children.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Puberdade Precoce/diagnóstico , Puberdade Precoce/terapia , Adolescente , Tamanho Corporal , Criança , Feminino , Hormônio Liberador de Gonadotropina/análise , Humanos , Hipotálamo/fisiologia , Masculino , Psicologia , Puberdade/fisiologia , Puberdade Precoce/fisiopatologia , Puberdade Precoce/psicologia , Maturidade Sexual/fisiologia , Resultado do Tratamento
13.
Endocrinology ; 144(6): 2566-79, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12746320

RESUMO

gamma-Aminobutyric acid (GABA) inhibits the embryonic migration of GnRH neurons and regulates hypothalamic GnRH release. A subset of GnRH neurons expresses GABA along their migratory route in the nasal compartment before entering the brain, suggesting that GABA produced by GnRH neurons may help regulate the migratory process. To examine this hypothesis and the possibility that persistence of GABA production by GnRH neurons may affect subsequent reproductive function, we generated transgenic mice in which the expression of glutamic acid decarboxylase-67 (GAD-67), a key enzyme in GABA synthesis, is targeted to GnRH neurons under the control of the GnRH gene promoter. On embryonic d 15, when GnRH neurons are still migrating, the transgenic animals had more GnRH neurons in aberrant locations in the cerebral cortex and fewer neurons reaching the hypothalamic-preoptic region, whereas migration into the brain was not affected. Hypothalamic GnRH content in mutant mice was low during the first week of postnatal life, increasing to normal values during infantile development (second week after birth) in the presence of increased pulsatile GnRH release. Consistent with these changes, serum LH and FSH levels were also elevated. Gonadotropin release returned to normal values by the time steroid negative feedback became established (fourth week of life). Ovariectomy at this time demonstrated an enhanced gonadotropin response in transgenic animals. Although the onset of puberty, as assessed by the age at vaginal opening and first ovulation, was not affected in the mutant mice, estrous cyclicity and adult reproductive capacity were disrupted. Mutant mice had reduced litter sizes, increased time intervals between deliveries of litters, and a shorter reproductive life span. Thus, GABA produced within GnRH neurons does not delay GnRH neuronal migration, but instead serves as a developmental cue that increases the positional diversity of these neurons within the basal forebrain. In addition, the results suggest that the timely termination of GABA production within the GnRH neuronal network is a prerequisite for normal reproductive function. The possibility arises that similar abnormalities in GABA homeostasis may contribute to syndromes of hypothalamic amenorrhea/oligomenorrhea in humans.


Assuntos
Movimento Celular/fisiologia , Glutamato Descarboxilase/genética , Hormônio Liberador de Gonadotropina/metabolismo , Isoenzimas/genética , Neurônios/citologia , Reprodução/fisiologia , Animais , Animais Recém-Nascidos , Ciclo Estral/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Glutamato Descarboxilase/metabolismo , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/fisiologia , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/enzimologia , Regiões Promotoras Genéticas/genética , Fluxo Pulsátil , Ratos , Ácido gama-Aminobutírico/biossíntese , Ácido gama-Aminobutírico/metabolismo
14.
J Neurosci ; 23(1): 230-9, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12514220

RESUMO

The initiation of mammalian puberty requires the activation of hypothalamic neurons secreting the neuropeptide luteinizing hormone-releasing hormone (LHRH). It is thought that this activation is caused by changes in trans-synaptic input to LHRH neurons. More recently, it has been postulated that the pubertal increase in LHRH secretion in female animals also requires neuron-glia signaling mediated by growth factors of the epidermal growth factor (EGF) family and their astrocytic erbB receptors. Although it appears clear that functional astrocytic erbB1 receptors are necessary for the timely advent of puberty, the physiological contribution that erbB4 receptors may make to this process has not been established. To address this issue, we generated transgenic mice expressing a dominant-negative erbB4 receptor (DN-erbB4) under the control of the GFAP promoter, which targets transgene expression to astrocytes. DN-erbB4 expression is most abundant in hypothalamic astrocytes, where it blocks the ligand-dependent activation of glial erbB4 and erbB2 receptors, without affecting erbB1 (EGF) receptor signaling. Mice carrying the transgene exhibit delayed sexual maturation and a diminished reproductive capacity in early adulthood. These abnormalities are related to a deficiency in pituitary gonadotropin hormone secretion, caused by impaired release of LHRH, the hypothalamic neuropeptide that controls sexual development. In turn, the reduction in LHRH release is caused by the inability of hypothalamic astrocytes to respond to neuregulin (NRG) with production of prostaglandin E(2), which in wild-type animals mediates the stimulatory effect of astroglial erbB receptor activation on neuronal LHRH release. Thus, neuron-astroglia communication via NRG-erbB4/2 receptor signaling appears to be essential for the timely unfolding of the developmental program by which the brain controls mammalian sexual maturation.


Assuntos
Astrócitos/fisiologia , Receptores ErbB/fisiologia , Hipotálamo/citologia , Neurregulinas/fisiologia , Maturidade Sexual , Animais , Astrócitos/metabolismo , Células Cultivadas , Dinoprostona/metabolismo , Receptores ErbB/genética , Feminino , Fertilidade , Hormônio Foliculoestimulante/metabolismo , Proteína Glial Fibrilar Ácida/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/fisiologia , Cinética , Camundongos , Camundongos Transgênicos , Mutação , Neurregulinas/farmacologia , Regiões Promotoras Genéticas , Receptor ErbB-2/metabolismo , Receptor ErbB-4 , Caracteres Sexuais , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA