Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38126277

RESUMO

Evidence suggests that estradiol-sensing preoptic area GABA neurons are involved in the preovulatory surge mechanism necessary for ovulation. In vivo CRISPR-Cas9 editing was used to achieve a 60-70% knockdown in estrogen receptor alpha (ESR1) expression by GABA neurons located within the regions of the rostral periventricular area of the third ventricle (RP3V) and medial preoptic nuclei (MPN) in adult female mice. Mice exhibited variable reproductive phenotypes with the only significant finding being mice with bilateral ESR1 deletion in RP3V GABA neurons having reduced cFos expression in gonadotropin-releasing hormone (GnRH) neurons at the time of the surge. One sub-population of RP3V GABA neurons expresses kisspeptin. Re-grouping ESR1-edited mice on the basis of their RP3V kisspeptin expression revealed a highly consistent phenotype; mice with a near-complete loss of kisspeptin immunoreactivity displayed constant estrus and failed to exhibit surge activation but retained pulsatile luteinizing hormone (LH) secretion. These observations demonstrate that ESR1-expressing GABA-kisspeptin neurons in the RP3V are essential for the murine preovulatory LH surge mechanism.


Assuntos
Sistemas CRISPR-Cas , Kisspeptinas , Camundongos , Feminino , Animais , Kisspeptinas/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios GABAérgicos/metabolismo , Ciclo Estral/fisiologia , Ácido gama-Aminobutírico/metabolismo
2.
Nat Commun ; 13(1): 7433, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36460649

RESUMO

The mechanisms underlying the homeostatic estrogen negative feedback pathway central to mammalian fertility have remained unresolved. Direct measurement of gonadotropin-releasing hormone (GnRH) pulse generator activity in freely behaving mice with GCaMP photometry demonstrated striking estradiol-dependent plasticity in the frequency, duration, amplitude, and profile of pulse generator synchronization events. Mice with Cre-dependent deletion of ESR1 from all kisspeptin neurons exhibited pulse generator activity identical to that of ovariectomized wild-type mice. An in vivo CRISPR-Cas9 approach was used to knockdown ESR1 expression selectively in arcuate nucleus (ARN) kisspeptin neurons. Mice with >80% deletion of ESR1 in ARN kisspeptin neurons exhibited the ovariectomized pattern of GnRH pulse generator activity and high frequency LH pulses but with very low amplitude due to reduced responsiveness of the pituitary. Together, these studies demonstrate that estrogen utilizes ESR1 in ARN kisspeptin neurons to achieve estrogen negative feedback of the GnRH pulse generator in mice.


Assuntos
Hormônio Liberador de Gonadotropina , Kisspeptinas , Feminino , Camundongos , Animais , Kisspeptinas/genética , Retroalimentação , Estrogênios , Núcleo Arqueado do Hipotálamo , Mamíferos
3.
Endocrinology ; 159(11): 3822-3833, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30304401

RESUMO

Developments in optical imaging and optogenetics are transforming the functional investigation of neuronal networks throughout the brain. Recent studies in the neuroendocrine field have used genetic mouse models combined with a variety of light-activated optical tools as well as GCaMP calcium imaging to interrogate the neural circuitry controlling hormone secretion. The present review highlights the benefits and caveats of these approaches for undertaking both acute brain slice and functional studies in vivo. We focus on the use of channelrhodopsin and the inhibitory optogenetic tools, archaerhodopsin and halorhodopsin, in addition to GCaMP imaging of individual cells in vitro and neural populations in vivo using fiber photometry. We also address issues around the use of genetic vs viral delivery of encoded proteins to specific Cre-expressing cell populations, their quantification, and the use of conscious vs anesthetized animal models. To date, optogenetics and GCaMP imaging have proven useful in dissecting functional circuitry within the brain and are likely to become essential investigative tools for deciphering the different neural networks controlling hormone secretion.


Assuntos
Encéfalo/metabolismo , Hormônios/metabolismo , Neurônios/metabolismo , Animais , Proteínas Arqueais , Encéfalo/diagnóstico por imagem , Channelrhodopsins , Dependovirus , Técnicas de Transferência de Genes , Halorrodopsinas , Camundongos , Modelos Animais , Vias Neurais/diagnóstico por imagem , Vias Neurais/metabolismo , Imagem Óptica , Optogenética , Fotometria
4.
J Neuroendocrinol ; : e12600, 2018 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-29656508

RESUMO

The 3rd World Conference on Kisspeptin, "Kisspeptin 2017: Brain and Beyond" was held March 30-31 at the Rosen Centre Hotel in Orlando, Florida, providing an international forum for multidisciplinary scientists to meet and share cutting-edge research on kisspeptin biology and its relevance to human health and disease. The meeting built upon previous world conferences focused on the role of kisspeptin and associated peptides in the control of gonadotropin-releasing hormone (GnRH) secretion and reproduction. Based on recent discoveries, the scope of this meeting was expanded to include functions of kisspeptin and related peptides in other physiological systems including energy homeostasis, pregnancy, ovarian and uterine function, and thermoregulation. In addition, discussions addressed the translation of basic knowledge of kisspeptin biology to the treatment of disease, with the goal of seeking consensus about the best approaches to improve human health. The two-day meeting featured a non-traditional structure, with each day starting with poster sessions followed by lunch discussions and facilitated large-group sessions with short presentations to maximize the exchange of new, unpublished data. Topics were identified by a survey prior to the meeting, and focused on major unresolved questions, important controversies, and future directions in the field. Finally, career development activities provided mentoring for trainees and junior investigators, and networking opportunities for those individuals with established researchers in the field. Overall, the meeting was rated as a success by attendees and covered a wide range of lively and provocative discussion topics on the changing nature of the field of "kisspeptinology" and its future. This article is protected by copyright. All rights reserved.

5.
Proc Natl Acad Sci U S A ; 114(47): E10216-E10223, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29109258

RESUMO

The pulsatile release of luteinizing hormone (LH) is critical for mammalian fertility. However, despite several decades of investigation, the identity of the neuronal network generating pulsatile reproductive hormone secretion remains unproven. We use here a variety of optogenetic approaches in freely behaving mice to evaluate the role of the arcuate nucleus kisspeptin (ARNKISS) neurons in LH pulse generation. Using GCaMP6 fiber photometry, we find that the ARNKISS neuron population exhibits brief (∼1 min) synchronized episodes of calcium activity occurring as frequently as every 9 min in gonadectomized mice. These ARNKISS population events were found to be near-perfectly correlated with pulsatile LH secretion. The selective optogenetic activation of ARNKISS neurons for 1 min generated pulses of LH in freely behaving mice, whereas inhibition with archaerhodopsin for 30 min suppressed LH pulsatility. Experiments aimed at resetting the activity of the ARNKISS neuron population with halorhodopsin were found to reset ongoing LH pulsatility. These observations indicate the ARNKISS neurons as the long-elusive hypothalamic pulse generator driving fertility.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/metabolismo , Rede Nervosa/metabolismo , Neurônios/metabolismo , Potenciais de Ação , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/diagnóstico por imagem , Feminino , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Optogenética/métodos , Periodicidade , Fotometria/métodos , Imagens com Corantes Sensíveis à Voltagem
6.
Endocrinology ; 157(12): 4794-4802, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27715255

RESUMO

Using a new tail-tip bleeding procedure and a sensitive ELISA, we describe here the patterns of LH secretion throughout the mouse estrous cycle; in ovariectomized mice; in ovariectomized, estradiol-treated mice that model estrogen-negative and -positive feedback; and in transgenic GNR23 mice that exhibit allele-dependent reductions in GnRH neuron number. Pulsatile LH secretion was evident at all stages of the estrous cycle, with LH pulse frequency being approximately one pulse per hour in metestrous, diestrous, and proestrous mice but much less frequent at estrus (less than one pulse per 4 h). Ovariectomy resulted in substantial increases in basal and pulsatile LH secretion with pulses occurring approximately every 21 minutes. Chronic treatment with negative-feedback, estradiol-filled capsules returned LH pulse frequency to intact follicular phase levels, although pulse amplitude remained elevated. On the afternoon of proestrus, the LH surge was found to begin in a highly variable manner over a 4-hour range, lasting for more than 3 hours. In contrast, ovariectomized, estradiol-treated, positive-feedback mice exhibited a relatively uniform surge onset at approximately 0.5 hour prior to lights out. Gonadectomized wild-type and heterozygous GNR23 (∼200 GnRH neurons) male mice exhibited an LH pulse every 60 minutes. Homozygous GNR23 mice (∼80 GnRH neurons) had very low basal LH concentrations but continued to exhibit small amplitude LH pulses every 90 minutes. These studies provide the first characterization in mice of pulse and surge modes of LH secretion across the estrous cycle and demonstrate that very few GnRH neurons are required for pulsatile LH secretion.


Assuntos
Retroalimentação Fisiológica/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/metabolismo , Neurônios/metabolismo , Animais , Ensaio de Imunoadsorção Enzimática , Estradiol/farmacologia , Ciclo Estral/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Feminino , Hipotálamo/citologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Ovariectomia
7.
Philos Trans R Soc Lond B Biol Sci ; 371(1688): 20150115, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26833836

RESUMO

Sex differences in brain neuroanatomy and neurophysiology underpin considerable physiological and behavioural differences between females and males. Sexual differentiation of the brain is regulated by testosterone secreted by the testes predominantly during embryogenesis in humans and the neonatal period in rodents. Despite huge advances in understanding how testosterone, and its metabolite oestradiol, sexually differentiate the brain, little is known about the mechanism that actually generates the male-specific neonatal testosterone surge. This review examines the evidence for the role of the hypothalamus, and particularly the gonadotropin-releasing hormone (GnRH) neurons, in generating the neonatal testosterone surge in rodents and primates. Kisspeptin-GPR54 signalling is well established as a potent and critical regulator of GnRH neuron activity during puberty and adulthood, and we argue here for an equally important role at birth in driving the male-specific neonatal testosterone surge in rodents. The presence of a male-specific population of preoptic area kisspeptin neurons that appear transiently in the perinatal period provide one possible source of kisspeptin drive to neonatal GnRH neurons in the mouse.


Assuntos
Animais Recém-Nascidos/fisiologia , Hipotálamo/fisiologia , Testosterona/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Masculino , Testosterona/sangue
8.
J Neurosci ; 34(46): 15297-305, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25392497

RESUMO

Sex differences in brain function underlie robust differences between males and females in both normal and disease states. Although alternative mechanisms exist, sexual differentiation of the male mammalian brain is initiated predominantly by testosterone secreted by the testes during the perinatal period. Despite considerable advances in understanding how testosterone and its metabolite estradiol sexually differentiate the brain, little is known about the mechanism that generates the male-specific perinatal testosterone surge. In mice, we show that a male-specific activation of GnRH neurons occurs 0-2 h following birth and that this correlates with the male-specific surge of testosterone occurring up to 5 h after birth. The necessity of GnRH signaling for the sexually differentiating effects of the perinatal testosterone surge was demonstrated by the persistence of female-like brain characteristics in adult male, GnRH receptor knock-out mice. Kisspeptin neurons have recently been identified to be potent, direct activators of GnRH neurons. We demonstrate that a population of kisspeptin neurons appears in the preoptic area of only the male between E19 and P1. The importance of kisspeptin inputs to GnRH neurons for the process of sexual differentiation was demonstrated by the lack of a normal neonatal testosterone surge, and disordered brain sexual differentiation of male mice in which the kisspeptin receptor was deleted selectively from GnRH neurons. These observations demonstrate the necessity of perinatal GnRH signaling for driving brain sexual differentiation and indicate that kisspeptin inputs to GnRH neurons are essential for this process to occur.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Neurônios/fisiologia , Área Pré-Óptica/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Diferenciação Sexual/fisiologia , Transdução de Sinais , Animais , Animais Recém-Nascidos , Feminino , Hormônio Liberador de Gonadotropina/genética , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Gravidez , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Receptores LHRH/genética , Receptores LHRH/fisiologia , Caracteres Sexuais , Testosterona/sangue , Tirosina 3-Mono-Oxigenase/metabolismo , Vasopressinas/metabolismo
9.
Neuroendocrinology ; 100(2-3): 191-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25301053

RESUMO

Kisspeptin-Gpr54 signaling is critical for regulating the activity of gonadotropin-releasing hormone (GnRH) neurons in mammals. Previous studies have shown that the negative feedback mechanism is disrupted in global Gpr54-null mutants. The present investigation aimed to determine (1) if a lack of cyclical estrogen exposure of the GnRH neuronal network in the life-long hypogonadotropic Gpr54-null mice contributed to their failed negative feedback mechanism and (2) the cellular location of disrupted kisspeptin-Gpr54 signaling. Plasma luteinizing hormone (LH) concentrations were determined in individual adult female mice when intact, following ovariectomy (OVX) and in response to an acute injection of 17ß-estradiol (E2). Control mice exhibited a characteristic rise in LH after OVX that was suppressed by acute E2. Global Gpr54-null mice failed to exhibit any post-OVX increase in LH or response to E2. Adult female global Gpr54-null mice given a cyclical regimen of estradiol for three cycles prior to OVX also failed to exhibit any post-OVX increase in LH or response to E2. To address whether Gpr54 signaling at the GnRH neuron itself was necessary for the failed response to OVX in global Gpr54-null animals, adult female mice with a GnRH neuron-selective deletion of Gpr54 were examined. These mice also failed to exhibit any post-OVX increase in LH or response to E2. These experiments demonstrate defective negative feedback in global Gpr54-null mice that cannot be attributed to a lack of prior exposure of the GnRH neuronal network to cyclical estradiol. The absence of negative feedback in GnRH neuron-selective Gpr54-null mice demonstrates the necessity of direct kisspeptin signaling at the GnRH neuron for this mechanism to occur.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Neurônios/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Análise Química do Sangue , Estradiol/administração & dosagem , Estrogênios/administração & dosagem , Estrogênios/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Feminino , Hormônio Luteinizante/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Ovariectomia , Periodicidade , Hipófise/efeitos dos fármacos , Hipófise/fisiologia , Radioimunoensaio , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1
10.
Nat Commun ; 4: 2492, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24051579

RESUMO

Signaling between kisspeptin and its receptor, G-protein-coupled receptor 54 (Gpr54), is now recognized as being essential for normal fertility. However, the key cellular location of kisspeptin-Gpr54 signaling is unknown. Here we create a mouse with a GnRH neuron-specific deletion of Gpr54 to assess the role of gonadotropin-releasing hormone (GnRH) neurons. Mutant mice are infertile, fail to go through puberty and exhibit markedly reduced gonadal size and follicle-stimulating hormone levels alongside GnRH neurons that are unresponsive to kisspeptin. In an attempt to rescue the infertile phenotype of global Gpr54⁻/⁻ mutants, we use BAC transgenesis to target Gpr54 to the GnRH neurons. This results in mice with normal puberty onset, estrous cyclicity, fecundity and a recovery of kisspeptin's stimulatory action upon GnRH neurons. Using complimentary cell-specific knockout and knockin approaches we demonstrate here that the GnRH neuron is the key site of kisspeptin-Gpr54 signaling for fertility.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Infertilidade/genética , Kisspeptinas/genética , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Animais , Feminino , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/patologia , Infertilidade/metabolismo , Infertilidade/patologia , Kisspeptinas/metabolismo , Camundongos , Camundongos Knockout , Neurônios/patologia , Tamanho do Órgão , Ovário/metabolismo , Ovário/patologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Maturidade Sexual
11.
Front Neuroendocrinol ; 34(2): 120-31, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23500175

RESUMO

The activation of the gonadotropin-releasing hormone (GnRH) neurons from a state of relative quiescence is critical for initiating puberty in mammals. Kisspeptin and its G-protein coupled receptor Gpr54 are essential for puberty, with disruption to either resulting in failed puberty in humans and mice. Robust data from several species indicate that Kiss1 mRNA and/or kisspeptin peptide expression within the hypothalamus increases during pubertal development. Kisspeptin fiber innervation of GnRH neurons and kisspeptin release within the hypothalamus also increase during pubertal development, indicating that there is increased kisspeptinergic drive to GnRH neurons during pubertal development. It is becoming increasingly apparent that gonadal steroids play important roles in the regulation of kisspeptin expression during pubertal development, and in particular, estradiol signaling through estrogen receptor alpha appears to be necessary for these changes to occur. This review focuses on the role that estradiol plays in the regulation of kisspeptin expression during pubertal development.


Assuntos
Estradiol/farmacologia , Hormônio Liberador de Gonadotropina/fisiologia , Kisspeptinas/fisiologia , Neurônios/fisiologia , Puberdade/fisiologia , Animais , Estradiol/fisiologia , Receptor alfa de Estrogênio/fisiologia , Feminino , Humanos , Hipotálamo/metabolismo , Kisspeptinas/biossíntese , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Puberdade/efeitos dos fármacos , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Kisspeptina-1
12.
Endocrinology ; 153(8): 3758-69, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22719049

RESUMO

The GnRH neurons exhibit long dendrites and project to the median eminence. The aim of the present study was to generate an acute brain slice preparation that enabled recordings to be undertaken from GnRH neurons maintaining the full extent of their dendrites or axons. A thick, horizontal brain slice was developed, in which it was possible to record from the horizontally oriented GnRH neurons located in the anterior hypothalamic area (AHA). In vivo studies showed that the majority of AHA GnRH neurons projected outside the blood-brain barrier and expressed c-Fos at the time of the GnRH surge. On-cell recordings compared AHA GnRH neurons in the horizontal slice (AHAh) with AHA and preoptic area (POA) GnRH neurons in coronal slices [POA coronal (POAc) and AHA coronal (AHAc), respectively]. AHAh GnRH neurons exhibited tighter burst firing compared with other slice orientations. Although α-Amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) excited GnRH neurons in all preparations, γ-aminobutyric acid (GABA) was excitatory in AHAc and POAc but inhibitory in AHAh slices. GABA(A) receptor postsynaptic currents were the same in AHAh and AHAc slices. Intriguingly, direct activation of GABA(A) or GABA(B) receptors respectively stimulated and inhibited GnRH neurons regardless of slice orientation. Subsequent experiments indicated that net GABA effects were determined by differences in the ratio of GABA(A) and GABA(B) receptor-mediated effects in "long" and "short" dendrites of GnRH neurons in the different slice orientations. These studies document a new brain slice preparation for recording from GnRH neurons with their extensive dendrites/axons and highlight the importance of GnRH neuron orientation relative to the angle of brain slicing in studying these neurons in vitro.


Assuntos
Encéfalo/citologia , Encéfalo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Ácido gama-Aminobutírico/farmacologia , Animais , Núcleo Hipotalâmico Anterior/citologia , Núcleo Hipotalâmico Anterior/efeitos dos fármacos , Núcleo Hipotalâmico Anterior/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Área Pré-Óptica/citologia , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Receptores de GABA/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
13.
Mol Cell Endocrinol ; 324(1-2): 45-50, 2010 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-20109523

RESUMO

Studies undertaken in many species indicate that kisspeptin-Gpr54 signaling is essential for the activation of gonadotropin-releasing hormone (GnRH) neurons to bring about puberty. Investigations in transgenic mouse models, in particular, have highlighted the importance of kisspeptin signaling at the level of the GnRH neuron itself in this process. This review aims to highlight current understanding of the neurobiological mechanisms underlying the kisspeptin activation of postnatal GnRH neurons. The three key features of the kisspeptin-Gpr54-GnRH neuron axis leading up to puberty are (i) the expression of adult-like levels of Gpr54 mRNA in GnRH neurons well in advance of puberty, (ii) a modest increase in the electrical response of GnRH neurons to Gpr54 activation across postnatal development and (iii), the "sudden" appearance of kisspeptin fibers surrounding GnRH neuron cell bodies/proximal dendrites just prior to puberty onset. These kisspeptin fibers are likely to originate from the kisspeptin population located in the rostral periventricular region of the third ventricle (RP3V). Together, available data suggest that the key step in the kisspeptin control of puberty lies in the control of kisspeptin synthesis within RP3V kisspeptin neurons that innervate GnRH neurons. This has recently been shown to be dependent upon circulating estradiol concentrations. As such, we propose that RP3V kisspeptin neurons represent a critical estradiol-dependent amplification mechanism brought into play relatively late in pubertal development to activate GnRH neurons and complete the process of puberty onset. Subsequently, in the adult female, this same circuitry is used to activate GnRH neurons to generate the cyclical preovulatory GnRH/LH surge.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Maturidade Sexual/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Encéfalo/metabolismo
14.
Biol Sex Differ ; 1(1): 7, 2010 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-21208471

RESUMO

BACKGROUND: Reproductive functions controlled by the hypothalamus are highly sexually differentiated. One of the most dramatic differences involves estrogen positive feedback, which leads to ovulation. A crucial feature of this positive feedback is the ability of estradiol to facilitate progesterone synthesis in female hypothalamic astrocytes. Conversely, estradiol fails to elevate hypothalamic progesterone levels in male rodents, which lack the estrogen positive feedback-induced luteinizing hormone (LH) surge. To determine whether hypothalamic astrocytes are sexually differentiated, we examined the cellular responses of female and male astrocytes to estradiol stimulation. METHODS: Primary adult hypothalamic astrocyte cultures were established from wild type rats and mice, estrogen receptor-α knockout (ERKO) mice, and four core genotype (FCG) mice, with the sex determining region of the Y chromosome (Sry) deleted and inserted into an autosome. Astrocytes were analyzed for Sry expression with reverse transcription PCR. Responses to estradiol stimulation were tested by measuring free cytoplasmic calcium concentration ([Ca2+]i) with fluo-4 AM, and progesterone synthesis with column chromatography and radioimmunoassay. Membrane estrogen receptor-α (mERα) levels were examined using surface biotinylation and western blotting. RESULTS: Estradiol stimulated both [Ca2+]i release and progesterone synthesis in hypothalamic astrocytes from adult female mice. Male astrocytes had a significantly elevated [Ca2+]i response but it was significantly lower than in females, and progesterone synthesis was not enhanced. Surface biotinylation demonstrated mERα in both female and male astrocytes, but only in female astrocytes did estradiol treatment increase insertion of the receptor into the membrane, a necessary step for maximal [Ca2+]i release. Regardless of the chromosomal sex, estradiol facilitated progesterone synthesis in astrocytes from mice with ovaries (XX and XY-), but not in mice with testes (XY-Sry and XXSry). CONCLUSIONS: Astrocytes are sexually differentiated, and in adulthood reflect the actions of sex steroids during development. The response of hypothalamic astrocytes to estradiol stimulation was determined by the presence or absence of ovaries, regardless of chromosomal sex. The trafficking of mERα in female, but not male, astrocytes further suggests that cell signaling mechanisms are sexually differentiated.

15.
Endocrinology ; 150(7): 3214-20, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19299459

RESUMO

The regulation of GnRH neurons by kisspeptin is critical for normal puberty onset in mammals. In the rodent the kisspeptin neurons innervating GnRH neurons are thought to reside in the rostral periventricular area of the third ventricle (RP3V). Using kisspeptin immunocytochemistry we show that kisspeptin peptide expression in the RP3V of female mice begins around postnatal d 15 (P15) and rapidly increases to achieve adult-like levels by P30, the time of puberty onset. Ovariectomy of female pups at P15 resulted in a 70-90% reduction (P < 0.01) in kisspeptin peptide expression within the RP3V of P30 or P60 mice. Replacement of 17-beta-estradiol (E2) in P15-ovariectomized mice from P15-30 or P22-30 resulted in a complete restoration of kisspeptin peptide expression in the RP3V (P < 0.01). Kisspeptin-immunoreactive fibers throughout the hypothalamus, including the arcuate nucleus, followed the same pattern of estrogen-dependent expression. To test the absolute necessity of estrogen for kisspeptin expression in the RP3V, aromatase knockout mice were examined. Kisspeptin-immunoreactive cells were detected in the arcuate nucleus, but there was a complete absence of kisspeptin peptide in RP3V neurons of aromatase knockout adult females. These results demonstrate that E2 is essential for the prepubertal development of kisspeptin peptide within RP3V neurons and suggest that an E2-kisspeptin positive feedback mechanism exists before puberty. This implies that RP3V kisspeptin neurons are E2-dependent amplifiers of GnRH neuron activity in the prepubertal period.


Assuntos
Estradiol/fisiologia , Maturidade Sexual/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Estradiol/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas , Camundongos , Neurônios/fisiologia , Ovariectomia , Área Pré-Óptica/fisiologia , Terceiro Ventrículo/fisiologia , Proteínas Supressoras de Tumor/biossíntese
16.
J Neurosci ; 28(35): 8691-7, 2008 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-18753370

RESUMO

Kisspeptin and its receptor GPR54 have recently been identified as key signaling partners in the neural control of fertility in animal models and humans. The gonadotropin-releasing hormone (GnRH) neurons represent the final output neurons of the neural network controlling fertility and are suspected to be the primary locus of kisspeptin-GPR54 signaling. Using mouse models, the present study addressed whether kisspeptin and GPR54 have a key role in the activation of GnRH neurons to generate the luteinizing hormone (LH) surge responsible for ovulation. Dual-label immunocytochemistry experiments showed that 40-60% of kisspeptin neurons in the rostral periventricular area of the third ventricle (RP3V) expressed estrogen receptor alpha and progesterone receptors. Using an ovariectomized, gonadal steroid-replacement regimen, which reliably generates an LH surge, approximately 30% of RP3V kisspeptin neurons were found to express c-FOS in surging mice compared with 0% in nonsurging controls. A strong correlation was found between the percentage of c-FOS-positive kisspeptin neurons and the percentage of c-FOS-positive GnRH neurons. To evaluate whether kisspeptin and/or GPR54 were essential for GnRH neuron activation and the LH surge, Gpr54- and Kiss1-null mice were examined. Whereas wild-type littermates all exhibited LH surges and c-FOS in approximately 50% of their GnRH neurons, none of the mutant mice from either line showed an LH surge or any GnRH neurons with c-FOS. These observations provide the first evidence that kisspeptin-GPR54 signaling is essential for GnRH neuron activation that initiates ovulation. This broadens considerably the potential roles and therapeutic possibilities for kisspeptin and GPR54 in fertility regulation.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/metabolismo , Neurônios/metabolismo , Proteínas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Encéfalo/citologia , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Kisspeptinas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Ovariectomia/métodos , Progesterona/farmacologia , Progestinas/farmacologia , Proteínas/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Receptores de Kisspeptina-1 , Transdução de Sinais/efeitos dos fármacos
17.
Endocrinology ; 147(12): 5817-25, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16959837

RESUMO

The neuropeptide kisspeptin has recently been implicated as having a critical role in the activation of the GnRH neurons to bring about puberty. We examined here the postnatal development of kisspeptin neuronal populations and their projections to GnRH neurons in the mouse. Three populations of kisspeptin neurons located in the 1) anteroventral periventricular nucleus (AVPV) and the preoptic periventricular nucleus (PeN), 2) dorsomedial hypothalamus, and 3) arcuate nucleus were identified using an antisera raised against mouse kisspeptin-10. A marked 10-fold (P<0.01), female-dominant sex difference in the numbers of kisspeptin neurons existed in the AVPV/PeN but not elsewhere. Kisspeptin neurons in the AVPV/PeN of both sexes displayed a similar pattern of postnatal development with no cells detected at postnatal day (P) 10, followed by increases from P25 to reach adult levels by puberty onset (P<0.01; P31 females and P45 males). This pattern was not found in the dorsomedial hypothalamus or arcuate nucleus. Dual immunofluorescence experiments demonstrated close appositions between kisspeptin fibers and GnRH neuron cell bodies that were first apparent at P25 and increased across postnatal development in both sexes. These studies demonstrate kisspeptin peptide expression in the mouse hypothalamus and reveal the postnatal development of a sexually dimorphic continuum of kisspeptin neurons within the AVPV and PeN. This periventricular population of kisspeptin neurons reaches adult-like proportions at the time of puberty onset and is the likely source of the kisspeptin inputs to GnRH neurons.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/crescimento & desenvolvimento , Neurônios/metabolismo , Oligopeptídeos/metabolismo , Caracteres Sexuais , Animais , Animais Recém-Nascidos , Núcleos Anteriores do Tálamo/metabolismo , Feminino , Hipotálamo/citologia , Hipotálamo/metabolismo , Kisspeptinas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleos da Linha Média do Tálamo/metabolismo , Modelos Biológicos , Vias Neurais/crescimento & desenvolvimento , Oligopeptídeos/imunologia , Área Pré-Óptica/metabolismo , Distribuição Tecidual
18.
Mol Cell Endocrinol ; 254-255: 32-8, 2006 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-16781054

RESUMO

The gonadotropin-releasing hormone (GnRH) neurons represent the critical cell type activated to induce puberty in mammals. However, the mechanisms underlying their activation remain unclear. As the principal amino acid neurotransmitters in the brain, GABA and glutamate are known to have critical roles in the development of neuronal networks. This review provides an update on what is known about GABA and glutamate signaling at the GnRH neuron across development. An examination of morphological, receptor subunit expression, and electrophysiological data suggest that GABAA receptor signaling develops in advance of glutamatergic signaling. However, compared with other networks, the switch from GABAA receptor depolarization to hyperpolarization of GnRH neurons is delayed until the time of puberty. These observations suggest that developing GnRH neurons exhibit a sequence of GABA-->glutamate signaling similar to that of other neuronal networks but that it is significantly elongated so as to only be complete by the time of puberty onset.


Assuntos
Ácido Glutâmico/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Ácido gama-Aminobutírico/fisiologia , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Eletrofisiologia , Feminino , Ácido Glutâmico/metabolismo , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiologia , Masculino , Subunidades Proteicas/metabolismo , Receptores de GABA-A/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...