Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(27): e2113749119, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35763574

RESUMO

Kisspeptin neurons in the mediobasal hypothalamus (MBH) are critical targets of ovarian estrogen feedback regulating mammalian fertility. To reveal molecular mechanisms underlying this signaling, we thoroughly characterized the estrogen-regulated transcriptome of kisspeptin cells from ovariectomized transgenic mice substituted with 17ß-estradiol or vehicle. MBH kisspeptin neurons were harvested using laser-capture microdissection, pooled, and subjected to RNA sequencing. Estrogen treatment significantly (p.adj. < 0.05) up-regulated 1,190 and down-regulated 1,139 transcripts, including transcription factors, neuropeptides, ribosomal and mitochondrial proteins, ion channels, transporters, receptors, and regulatory RNAs. Reduced expression of the excitatory serotonin receptor-4 transcript (Htr4) diminished kisspeptin neuron responsiveness to serotonergic stimulation. Many estrogen-regulated transcripts have been implicated in puberty/fertility disorders. Patients (n = 337) with congenital hypogonadotropic hypogonadism (CHH) showed enrichment of rare variants in putative CHH-candidate genes (e.g., LRP1B, CACNA1G, FNDC3A). Comprehensive characterization of the estrogen-dependent kisspeptin neuron transcriptome sheds light on the molecular mechanisms of ovary-brain communication and informs genetic research on human fertility disorders.


Assuntos
Núcleo Arqueado do Hipotálamo , Estrogênios , Fertilidade , Kisspeptinas , Neurônios , Ovário , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Estrogênios/metabolismo , Feminino , Fertilidade/genética , Perfilação da Expressão Gênica , Humanos , Hipogonadismo/congênito , Hipogonadismo/genética , Kisspeptinas/genética , Kisspeptinas/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Ovário/metabolismo
2.
J Neurosci ; 41(44): 9177-9191, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34561233

RESUMO

Sex steroid hormones act on hypothalamic kisspeptin neurons to regulate reproductive neural circuits in the brain. Kisspeptin neurons start to express estrogen receptors in utero, suggesting steroid hormone action on these cells early during development. Whether neurosteroids are locally produced in the embryonic brain and impinge onto kisspeptin/reproductive neural circuitry is not known. To address this question, we analyzed aromatase expression, a key enzyme in estrogen synthesis, in male and female mouse embryos. We identified an aromatase neuronal network comprising ∼6000 neurons in the hypothalamus and amygdala. By birth, this network has become sexually dimorphic in a cluster of aromatase neurons in the arcuate nucleus adjacent to kisspeptin neurons. We demonstrate that male arcuate aromatase neurons convert testosterone to estrogen to regulate kisspeptin neuron activity. We provide spatiotemporal information on aromatase neuronal network development and highlight a novel mechanism whereby aromatase neurons regulate the activity of distinct neuronal populations expressing estrogen receptors.SIGNIFICANCE STATEMENT Sex steroid hormones, such as estradiol, are important regulators of neural circuits controlling reproductive physiology in the brain. Embryonic kisspeptin neurons in the hypothalamus express steroid hormone receptors, suggesting hormone action on these cells in utero Whether neurosteroids are locally produced in the brain and impinge onto reproductive neural circuitry is insufficiently understood. To address this question, we analyzed aromatase expression, a key enzyme in estradiol synthesis, in mouse embryos and identified a network comprising ∼6000 neurons in the brain. By birth, this network has become sexually dimorphic in a cluster of aromatase neurons in the arcuate nucleus adjacent to kisspeptin neurons. We demonstrate that male aromatase neurons convert testosterone to estradiol to regulate kisspeptin neuron activity.


Assuntos
Tonsila do Cerebelo/metabolismo , Aromatase/metabolismo , Estrogênios/biossíntese , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neurônios/metabolismo , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/fisiologia , Animais , Aromatase/genética , Feminino , Hipotálamo/citologia , Hipotálamo/fisiologia , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia
3.
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
4.
J Physiol ; 596(5): 885-899, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29214635

RESUMO

KEY POINTS: Neurons in the hypothalamus of the brain which secrete the peptide kisspeptin are important regulators of reproduction, and normal reproductive development. Electrical activity, in the form of action potentials, or spikes, leads to secretion of peptides and neurotransmitters, influencing the activity of downstream neurons; in kisspeptin neurons, this activity is highly irregular, but the mechanism of this is not known. In this study, we show that irregularity depends on the presence of a particular type of potassium ion channel in the membrane, which opens transiently in response to electrical excitation. The results contribute to understanding how kisspeptin neurons generate and time their membrane potential spikes, and how reliable this process is. Improved understanding of the activity of kisspeptin neurons, and how it shapes their secretion of peptides, is expected to lead to better treatment for reproductive dysfunction and disorders of reproductive development. ABSTRACT: Kisspeptin neurons in the hypothalamus are critically involved in reproductive function, via their effect on GnRH neuron activity and consequent gonadotropin release. Kisspeptin neurons show an intrinsic irregularity of firing, but the mechanism of this remains unclear. To address this, we carried out targeted whole-cell patch-clamp recordings of kisspeptin neurons in the arcuate nucleus (Kiss1Arc ), in brain slices isolated from adult male Kiss-Cre:tdTomato mice. Cells fired irregularly in response to constant current stimuli, with a wide range of spike time variability, and prominent subthreshold voltage fluctuations. In voltage clamp, both a persistent sodium (NaP) current and a fast transient (A-type) potassium current were apparent, activating at potentials just below the threshold for spiking. These currents have also previously been described in irregular-spiking cortical interneurons, in which the A-type current, mediated by Kv4 channels, interacts with NaP current to generate complex dynamics of the membrane potential, and irregular firing. In Kiss1Arc neurons, A-type current was blocked by phrixotoxin, a specific blocker of Kv4.2/4.3 channels, and consistent expression of Kv4.2 transcripts was detected by single-cell RT-PCR. In addition, firing irregularity was correlated to the density of A-type current in the membrane. Using conductance injection, we demonstrated that adding Kv4-like potassium conductance (gKv4 ) to a cell produces a striking increase in firing irregularity, and excitability is reduced, while subtracting gKv4 has the opposite effects. Thus, we propose that Kv4 interacting dynamically with NaP is a key determinant of the irregular firing behaviour of Kiss1Arc neurons, shaping their physiological function in gonadotropin release.


Assuntos
Potenciais de Ação , Núcleo Arqueado do Hipotálamo/fisiologia , Kisspeptinas/fisiologia , Neurônios/fisiologia , Canais de Potássio Shal/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/citologia
5.
Proc Natl Acad Sci U S A ; 111(30): 11133-8, 2014 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-25028498

RESUMO

The gut endocrine system is emerging as a central player in the control of appetite and glucose homeostasis, and as a rich source of peptides with therapeutic potential in the field of diabetes and obesity. In this study we have explored the physiology of insulin-like peptide 5 (Insl5), which we identified as a product of colonic enteroendocrine L-cells, better known for their secretion of glucagon-like peptide-1 and peptideYY. i.p. Insl5 increased food intake in wild-type mice but not mice lacking the cognate receptor Rxfp4. Plasma Insl5 levels were elevated by fasting or prolonged calorie restriction, and declined with feeding. We conclude that Insl5 is an orexigenic hormone released from colonic L-cells, which promotes appetite during conditions of energy deprivation.


Assuntos
Colo/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Células Enteroendócrinas/metabolismo , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/farmacologia , Animais , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Peptídeo YY/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo
6.
J Clin Invest ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38861336

RESUMO

Reproduction is safeguarded by multiple, often cooperative regulatory networks. Kisspeptin signaling, via KISS1R, plays a fundamental role in reproductive control, primarily by regulation of hypothalamic GnRH neurons. We disclose herein a pathway for direct kisspeptin actions in astrocytes that contributes to central reproductive modulation. Protein-protein-interaction and ontology analyses of hypothalamic proteomic profiles after kisspeptin stimulation revealed that glial/astrocyte markers are regulated by kisspeptin in mice. This glial-kisspeptin pathway was validated by the demonstrated expression of Kiss1r in mouse astrocytes in vivo and astrocyte cultures from humans, rats and mice, where kisspeptin activated canonical intracellular signaling-pathways. Cellular co-expression of Kiss1r with the astrocyte markers, GFAP and S100-ß, occurred in different brain regions, with higher percentage in Kiss1- and GnRH-enriched areas. Conditional ablation of Kiss1r in GFAP-positive cells, in the G-KiRKO mouse, altered gene expression of key factors in PGE2 synthesis in astrocytes, and perturbed astrocyte-GnRH neuronal appositions, as well as LH responses to kisspeptin and LH pulsatility, as surrogate marker of GnRH secretion. G-KiRKO mice also displayed changes in reproductive responses to metabolic stress induced by high-fat diet, affecting female pubertal onset, estrous cyclicity and LH-secretory profiles. Our data unveil a non-neuronal pathway for kisspeptin actions in astrocytes, which cooperates in fine-tuning the reproductive axis and its responses to metabolic stress.

7.
J Neurosci ; 32(3): 932-45, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22262891

RESUMO

Reproduction is controlled in the brain by a neural network that drives the secretion of gonadotropin-releasing hormone (GnRH). Various permissive homeostatic signals must be integrated to achieve ovulation in mammals. However, the neural events controlling the timely activation of GnRH neurons are not completely understood. Here we show that kisspeptin, a potent activator of GnRH neuronal activity, directly communicates with neurons that synthesize the gaseous transmitter nitric oxide (NO) in the preoptic region to coordinate the progression of the ovarian cycle. Using a transgenic Gpr54-null IRES-LacZ knock-in mouse model, we demonstrate that neurons containing neuronal NO synthase (nNOS), which are morphologically associated with kisspeptin fibers, express the kisspeptin receptor GPR54 in the preoptic region, but not in the tuberal region of the hypothalamus. The activation of kisspeptin signaling in preoptic neurons promotes the activation of nNOS through its phosphorylation on serine 1412 via the AKT pathway and mimics the positive feedback effects of estrogens. Finally, we show that while NO release restrains the reproductive axis at stages of the ovarian cycle during which estrogens exert their inhibitory feedback, it is required for the kisspeptin-dependent preovulatory activation of GnRH neurons. Thus, interactions between kisspeptin and nNOS neurons may play a central role in regulating the hypothalamic-pituitary-gonadal axis in vivo.


Assuntos
Hipotálamo/citologia , Kisspeptinas/metabolismo , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Ovulação/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipotálamo/efeitos dos fármacos , Kisspeptinas/deficiência , Kisspeptinas/farmacologia , Hormônio Luteinizante/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/deficiência , Ovulação/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Receptores Acoplados a Proteínas G/deficiência , Receptores de Kisspeptina-1 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esteroides/farmacologia
8.
Am J Physiol Cell Physiol ; 304(9): C858-72, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23407879

RESUMO

We have isolated a K(+)-selective, Ca(2+)-dependent whole cell current and single-channel correlate in the human erythroleukemia (HEL) cell line. The whole cell current was inhibited by the intermediate-conductance KCa3.1 inhibitors clotrimazole, TRAM-34, and charybdotoxin, unaffected by the small-conductance KCa2 family inhibitor apamin and the large-conductance KCa1.1 inhibitors paxilline and iberiotoxin, and augmented by NS309. The single-channel correlate of the whole cell current was blocked by TRAM-34 and clotrimazole, insensitive to paxilline, and augmented by NS309 and had a single-channel conductance in physiological K(+) gradients of ~9 pS. RT-PCR revealed that the KCa3.1 gene, but not the KCa1.1 gene, was expressed in HEL cells. The KCa3.1 current, isolated in HEL cells under whole cell patch-clamp conditions, displayed an activated current component during depolarizing voltage steps from hyperpolarized holding potentials and tail currents upon repolarization, consistent with voltage-dependent modulation. This activated current increased with increasing voltage steps above -40 mV and was sensitive to inhibition by clotrimazole, TRAM-34, and charybdotoxin and insensitive to apamin, paxilline, and iberiotoxin. In single-channel experiments, depolarization resulted in an increase in open channel probability (Po) of KCa3.1, with no increase in channel number. The voltage modulation of Po was an increasing monotonic function of voltage. In the absence of elevated Ca(2+), voltage was ineffective at inducing channel activity in whole cell and single-channel experiments. These data indicate that KCa3.1 in HEL cells displays a unique form of voltage dependence modulating Po.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Ativação do Canal Iônico , Cálcio/fisiologia , Linhagem Celular Tumoral , Charibdotoxina/farmacologia , Clotrimazol/farmacologia , Estimulação Elétrica , Expressão Gênica , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Leucemia Eritroblástica Aguda , Magnésio/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Potássio/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Pirazóis/farmacologia
9.
Adv Exp Med Biol ; 784: 481-503, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23550020

RESUMO

Kisspeptins are a family of overlapping neuropeptides, encoded by the Kiss1 gene, that are required for activation and maintenance of the mammalian reproductive axis. Kisspeptins act within the hypothalamus to stimulate release of gonadotrophic releasing hormone and activation of the pituitary-gonadal axis. Robust model systems are required to dissect the regulatory mechanisms that control Kiss1 neuronal activity and to examine the molecular consequences of kisspeptin signalling. While studies in normal animals have been important in this, transgenic mice with targeted mutations affecting the kisspeptin signalling pathway have played a significant role in extending our understanding of kisspeptin physiology. Knock-out mice recapitulate the reproductive defects associated with mutations in humans and provide an experimentally tractable model system to interrogate regulatory feedback mechanisms. In addition, transgenic mice with cell-specific expression of modulator proteins such as the CRE recombinase or fluorescent reporter proteins such as GFP allow more sophisticated analyses such as cell or gene ablation or electrophysiological profiling. At a less complex level, immortalized cell lines have been useful for studying the role of kisspeptin in cell migration and metastasis and examining the intracellular signalling events associated with kisspeptin signalling.


Assuntos
Movimento Celular/fisiologia , Kisspeptinas/metabolismo , Reprodução/fisiologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Transformada , Kisspeptinas/genética , Camundongos , Camundongos Knockout , Mutação
10.
Pharmacol Rev ; 62(4): 565-78, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21079036

RESUMO

Kisspeptins are members of the Arg-Phe amide family of peptides, which have been identified as endogenous ligands for a G-protein-coupled receptor encoded by a gene originally called GPR54 (also known as AXOR12 or hOT7T175). After this pairing, the gene has been renamed KISS1R. The International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification recommends that the official name for the receptor is the kisspeptin receptor to follow the convention of naming the receptor protein after the endogenous ligand. The endogenous ligand was initially called metastin, after its role as a metastasis suppressor, and is now referred to as kisspeptin-54 (KP-54), a C-terminally amidated 54-amino acid peptide cleaved from the 145-amino acid gene product. Shorter C-terminal cleavage fragments [KP-14, KP-13 and KP-10 (the smallest active fragment)] are also biologically active. Both receptor and peptide are widely expressed in human, rat, and mouse; the receptor sequence shares more than 80% homology in these species. Activation of the kisspeptin receptor by kisspeptin is via coupling to G(q/11) and the phospholipase C pathway, causing Ca(2+) mobilization. Mutations in the KISS1R gene result in hypogonadotropic hypogonadotropism, and targeted disruption of Kiss1r in mice reproduces this phenotype, which led to the discovery of the remarkable ability of the kisspeptin receptor to act as a molecular switch for puberty. In addition to regulating the reproductive axis, the kisspeptin receptor is also implicated in cancer, placentation, diabetes, and the cardiovascular system.


Assuntos
Receptores Acoplados a Proteínas G , Terminologia como Assunto , Animais , Humanos , Especificidade de Órgãos , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/classificação , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Kisspeptina-1 , Relação Estrutura-Atividade
11.
Dev Dyn ; 241(10): 1591-602, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22837050

RESUMO

BACKGROUND: Adhesion G protein-coupled receptors (aGPCR) constitute a structurally and functionally diverse class of seven-transmembrane receptor proteins. Although for some of the members important roles in immunology, neurology, as well as developmental biology have been suggested, most receptors have been poorly characterized. RESULTS: We have studied evolution, expression, and function of an entire receptor group containing four uncharacterized aGPCR: Gpr110, Gpr111, Gpr115, and Gpr116. We show that the genomic loci of these four receptors are clustered tightly together in mouse and human genomes and that this cluster likely derives from a single common ancestor gene. Using transcriptional profiling on wild-type and knockout/LacZ reporter knockin mice strains, we have obtained detailed expression maps that show ubiquitous expression of Gpr116, co-expression of Gpr111 and Gpr115 in developing skin, and expression of Gpr110 in adult kidney. Loss of Gpr110, Gpr111, or Gpr115 function did not result in detectable defects, indicating that genes of this aGPCR group might function redundantly. CONCLUSIONS: The aGPCR cluster Gpr110, Gpr111, Gpr115, and Gpr116 developed from one common ancestor in vertebrates. Expression suggests a role in epithelia, and one can speculate about a possible redundant function of GPR111 and GPR115.


Assuntos
Evolução Molecular , Loci Gênicos/genética , Família Multigênica/genética , Receptores Acoplados a Proteínas G/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Primers do DNA/genética , Epitélio/metabolismo , Galactosídeos , Perfilação da Expressão Gênica , Humanos , Indóis , Rim/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Pele/metabolismo , Especificidade da Espécie
12.
Nat Commun ; 14(1): 3076, 2023 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-37248237

RESUMO

Coupling the release of pituitary hormones to the developmental stage of the oocyte is essential for female fertility. It requires estrogen to restrain kisspeptin (KISS1)-neuron pulsatility in the arcuate hypothalamic nucleus, while also exerting a surge-like effect on KISS1-neuron activity in the AVPV hypothalamic nucleus. However, a mechanistic basis for this region-specific effect has remained elusive. Our genomic analysis in female mice demonstrate that some processes, such as restraint of KISS1-neuron activity in the arcuate nucleus, may be explained by region-specific estrogen receptor alpha (ERα) DNA binding at gene regulatory regions. Furthermore, we find that the Kiss1-locus is uniquely regulated in these hypothalamic nuclei, and that the nuclear receptor co-repressor NR0B1 (DAX1) restrains its transcription specifically in the arcuate nucleus. These studies provide mechanistic insight into how ERα may control the KISS1-neuron, and Kiss1 gene expression, to couple gonadotropin release to the developmental stage of the oocyte.


Assuntos
Receptor Nuclear Órfão DAX-1 , Receptor alfa de Estrogênio , Hipotálamo , Kisspeptinas , Animais , Feminino , Camundongos , Núcleo Arqueado do Hipotálamo/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Receptor Nuclear Órfão DAX-1/genética , Receptor Nuclear Órfão DAX-1/metabolismo
13.
J Neurosci ; 31(7): 2421-30, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21325509

RESUMO

The anteroventral periventricular nucleus (AVPV) is thought to play a key role in regulating the excitability of gonadotropin-releasing hormone (GnRH) neurons that control fertility. Using an angled, parahorizontal brain slice preparation we have undertaken a series of electrophysiological experiments to examine how the AVPV controls GnRH neurons in adult male and female mice. More than half (59%) of GnRH neurons located in the rostral preoptic area were found to receive monosynaptic inputs from the AVPV in a sex-dependent manner. AVPV stimulation frequencies <1 Hz generated short-latency action potentials in GnRH neurons with GABA and glutamate mediating >90% of the evoked fast synaptic currents. The AVPV GABA input was dominant and found to excite or inhibit GnRH neurons in a cell-dependent manner. Increasing the AVPV stimulation frequency to 5-10 Hz resulted in the appearance of additional poststimulus inhibitory as well as delayed excitatory responses in GnRH neurons that were independent of ionotropic amino acid receptors. The inhibition observed immediately following the end of the stimulation period was mediated partly by GABA(B) receptors, while the delayed activation was mediated by the neuropeptide kisspeptin. The latter response was essentially absent in Gpr54 knock-out mice and abolished by a Gpr54 antagonist. Together, these studies show that AVPV neurons provide direct amino acid and neuropeptidergic inputs to GnRH neurons. Low-frequency activation generates predominant GABA/glutamate release with higher frequency activation recruiting release of kisspeptin. This frequency-dependent release of amino acid and neuropeptide neurotransmitters greatly expands the range of AVPV control of GnRH neuron excitability.


Assuntos
Aminoácidos/metabolismo , Núcleos Anteriores do Tálamo/citologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Biofísica , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Ácido Glutâmico/farmacologia , Hormônio Liberador de Gonadotropina/genética , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurotransmissores/metabolismo , Técnicas de Patch-Clamp , Tempo de Reação/fisiologia , Receptores Acoplados a Proteínas G/deficiência , Receptores de Kisspeptina-1 , Estatísticas não Paramétricas , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Valina/análogos & derivados , Valina/farmacologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/farmacologia
14.
Front Endocrinol (Lausanne) ; 13: 960769, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36093104

RESUMO

Kisspeptin neurons residing in the rostral periventricular area of the third ventricle (KPRP3V) and the arcuate nucleus (KPARC) mediate positive and negative estrogen feedback, respectively. Here, we aim to compare transcriptional responses of KPRP3V and KPARC neurons to estrogen. Transgenic mice were ovariectomized and supplemented with either 17ß-estradiol (E2) or vehicle. Fluorescently tagged KPRP3V neurons collected by laser-capture microdissection were subjected to RNA-seq. Bioinformatics identified 222 E2-dependent genes. Four genes encoding neuropeptide precursors (Nmb, Kiss1, Nts, Penk) were robustly, and Cartpt was subsignificantly upregulated, suggesting putative contribution of multiple neuropeptides to estrogen feedback mechanisms. Using overrepresentation analysis, the most affected KEGG pathways were neuroactive ligand-receptor interaction and dopaminergic synapse. Next, we re-analyzed our previously obtained KPARC neuron RNA-seq data from the same animals using identical bioinformatic criteria. The identified 1583 E2-induced changes included suppression of many neuropeptide precursors, granins, protein processing enzymes, and other genes related to the secretory pathway. In addition to distinct regulatory responses, KPRP3V and KPARC neurons exhibited sixty-two common changes in genes encoding three hormone receptors (Ghsr, Pgr, Npr2), GAD-65 (Gad2), calmodulin and its regulator (Calm1, Pcp4), among others. Thirty-four oppositely regulated genes (Kiss1, Vgf, Chrna7, Tmem35a) were also identified. The strikingly different transcriptional responses in the two neuron populations prompted us to explore the transcriptional mechanism further. We identified ten E2-dependent transcription factors in KPRP3V and seventy in KPARC neurons. While none of the ten transcription factors interacted with estrogen receptor-α, eight of the seventy did. We propose that an intricate, multi-layered transcriptional mechanism exists in KPARC neurons and a less complex one in KPRP3V neurons. These results shed new light on the complexity of estrogen-dependent regulatory mechanisms acting in the two functionally distinct kisspeptin neuron populations and implicate additional neuropeptides and mechanisms in estrogen feedback.


Assuntos
Núcleo Arqueado do Hipotálamo , Kisspeptinas , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Estrogênios/metabolismo , Estrogênios/farmacologia , Kisspeptinas/genética , Kisspeptinas/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Fatores de Transcrição/metabolismo
15.
J Neuroendocrinol ; 34(11): e13207, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36305576

RESUMO

Kisspeptin neurons in the arcuate nucleus of the hypothalamus generate gonadotrophin-releasing hormone (GnRH) pulses, and act as critical initiators of functional gonadotrophin secretion and reproductive competency. However, kisspeptin in other brain regions, most notably the posterodorsal subnucleus of the medial amygdala (MePD), plays a significant modulatory role over the hypothalamic kisspeptin population; our recent studies using optogenetics have shown that low-frequency light stimulation of MePD kisspeptin results in increased luteinsing hormone pulse frequency. Nonetheless, the neurochemical pathways that underpin this regulatory function remain unknown. To study this, we have utilised an optofluid technology, precisely combining optogenetic stimulation with intra-nuclear pharmacological receptor antagonism, to investigate the neurotransmission involved in this circuitry. We have shown experimentally and verified using a mathematical model that functional neurotransmission of both GABA and glutamate is a requirement for effective modulation of the GnRH pulse generator by amygdala kisspeptin neurons.


Assuntos
Hormônio Liberador de Gonadotropina , Kisspeptinas , Feminino , Camundongos , Animais , Kisspeptinas/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Ácido Glutâmico/metabolismo , Hormônio Luteinizante/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Tonsila do Cerebelo/metabolismo , Ácido gama-Aminobutírico/metabolismo
16.
J Neurosci ; 30(25): 8581-90, 2010 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-20573904

RESUMO

NMDA and kisspeptins can stimulate gonadotropin-releasing hormone (GnRH) release after peripheral or central administration in mice. To determine whether these agonists act independently or through a common pathway, we have examined their ability to stimulate GnRH/luteinizing hormone (LH) release after peripheral or central administration in Kiss1- or Gpr54 (Kiss1r)-null mutant mice. Peripheral injection of NMDA failed to stimulate GnRH/LH release in prepubertal or gonadally intact mutant male mice. Dual-labeling experiments indicated a direct activation of Kiss1-expressing neurons in the arcuate nucleus. In contrast, central injection of NMDA into the lateral ventricle increased plasma LH levels in both Kiss1 and Gpr54 mutant male mice similar to the responses in wild-type mice. Central injection of NMDA stimulated c-Fos expression throughout the hypothalamus but not in GnRH neurons, suggesting an action at the nerve terminals only. In contrast, kisspeptin-10 stimulated LH release after both central and peripheral injection but induced c-Fos expression in GnRH neurons only after central administration. Finally, central injection of NMDA induces c-Fos expression in catecholamine- and nitric oxide-producing neurons in the hypothalamus of mutant mice, indicating a possible kisspeptin-independent GnRH/LH release by NMDA through activation of these neurons. Thus, NMDA may act at both GnRH cell bodies (kisspeptin-independent) and nerve terminals (kisspeptin-dependent) in a dual way to participate in the GnRH/LH secretion in the male mouse.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , N-Metilaspartato/administração & dosagem , Neurônios/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Análise de Variância , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Imuno-Histoquímica , Injeções Intraperitoneais , Injeções Intraventriculares , Kisspeptinas , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Proteínas Supressoras de Tumor/genética
17.
BMC Genomics ; 12: 209, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21527035

RESUMO

BACKGROUND: Humans and mice with loss of function mutations in GPR54 (KISS1R) or kisspeptin do not progress through puberty, caused by a failure to release GnRH. The transcriptional networks regulated by these proteins in the hypothalamus have yet to be explored by genome-wide methods. RESULTS: We show here, using 1 million exon mouse arrays (Exon 1.0 Affymetrix) and quantitative polymerase chain reaction (QPCR) validation to analyse microdissected hypothalamic tissue from Gpr54 and Kiss1 knockout mice, the extent of transcriptional regulation in the hypothalamus. The sensitivity to detect important transcript differences in microdissected RNA was confirmed by the observation of counter-regulation of Kiss1 expression in Gpr54 knockouts and confirmed by immunohistochemistry (IHC). Since Gpr54 and Kiss1 knockout animals are effectively pre-pubertal with low testosterone (T) levels, we also determined which of the validated transcripts were T-responsive and which varied according to genotype alone. We observed four types of transcriptional regulation (i) genotype only dependent regulation, (ii) T only dependent regulation, (iii) genotype and T-dependent regulation with interaction between these variables, (iv) genotype and T-dependent regulation with no interaction between these variables. The results implicate for the first time several transcription factors (e.g. Npas4, Esr2), proteases (Klk1b22), and the orphan 10-transmembrane transporter TMEM144 in the biology of GPR54/kisspeptin function in the hypothalamus. We show for the neuronal activity regulated transcription factor NPAS4, that distinct protein over-expression is seen in the hypothalamus and hippocampus in Gpr54 knockout mice. This links for the first time the hypothalamic-gonadal axis with this important regulator of inhibitory synapse formation. Similarly we confirm TMEM144 up-regulation in the hypothalamus by RNA in situ hybridization and western blot. CONCLUSIONS: Taken together, global transcriptional profiling shows that loss of GPR54 and kisspeptin are not fully equivalent in the mouse hypothalamus.


Assuntos
Redes Reguladoras de Genes , Hipotálamo/metabolismo , Proteínas/genética , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Testosterona/metabolismo , Animais , Genótipo , Kisspeptinas , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Kisspeptina-1 , Transcrição Gênica
18.
Mol Pain ; 7: 90, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22112588

RESUMO

BACKGROUND: Kisspeptin is a neuropeptide known for its role in the hypothalamic regulation of the reproductive axis. Following the recent description of kisspeptin and its 7-TM receptor, GPR54, in the dorsal root ganglia and dorsal horns of the spinal cord, we examined the role of kisspeptin in the regulation of pain sensitivity in mice. RESULTS: Immunofluorescent staining in the mouse skin showed the presence of GPR54 receptors in PGP9.5-positive sensory fibers. Intraplantar injection of kisspeptin (1 or 3 nmol/5 µl) induced a small nocifensive response in naive mice, and lowered thermal pain threshold in the hot plate test. Both intraplantar and intrathecal (0.5 or 1 nmol/3 µl) injection of kisspeptin caused hyperalgesia in the first and second phases of the formalin test, whereas the GPR54 antagonist, p234 (0.1 or 1 nmol), caused a robust analgesia. Intraplantar injection of kisspeptin combined with formalin enhanced TRPV1 phosphorylation at Ser800 at the injection site, and increased ERK1/2 phosphorylation in the ipsilateral dorsal horn as compared to naive mice and mice treated with formalin alone. CONCLUSION: These data demonstrate for the first time that kisspeptin regulates pain sensitivity in rodents and suggest that peripheral GPR54 receptors could be targeted by novel drugs in the treatment of inflammatory pain.


Assuntos
Hiperalgesia/metabolismo , Kisspeptinas/metabolismo , Neuropeptídeos/metabolismo , Animais , Técnica Indireta de Fluorescência para Anticorpo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos , Limiar da Dor , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1
19.
Reproduction ; 141(3): 357-66, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21156837

RESUMO

Mice with mutations in the kisspeptin signaling pathway (Kiss1(-/-) or Gpr54(-/-)) have low gonadotrophic hormone levels, small testes, and impaired spermatogenesis. Between 2 and 7 months of age, however, the testes of the mutant mice increase in weight and in Gpr54(-/-) mice, the number of seminiferous tubules containing spermatids/spermatozoa increases from 17 to 78%. In contrast, the Kiss1(-/-) mice have a less severe defect in spermatogenesis and larger testes than Gpr54(-/-) mice at both 2 and 7 months of age. The reason for the improved spermatogenesis was investigated. Plasma testosterone and FSH levels did not increase with age in the mutant mice and remained much lower than in wild-type (WT) mice. In contrast, intratesticular testosterone levels were similar between mutant and WT mice. These data indicate that age-related spermatogenesis can be completed under conditions of low plasma testosterone and FSH and that intratesticular testosterone may contribute to this process. In addition, however, when the Gpr54(-/-) mice were fed a phytoestrogen-free diet, they showed no age-related increase in testes weight or improved spermatogenesis. Thus, both genetic and environmental factors are involved in the improved spermatogenesis in the mutant mice as they age although the mice still remain infertile. These data show that the possible impact of dietary phytoestrogens should be taken into account when studying the phenotype of mutant mice with defects in the reproductive axis.


Assuntos
Infertilidade Masculina/genética , Fitoestrógenos/farmacologia , Receptores Acoplados a Proteínas G/genética , Espermatogênese/efeitos dos fármacos , Espermatogênese/genética , Fatores Etários , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Dieta , Avaliação Pré-Clínica de Medicamentos , Fertilidade/efeitos dos fármacos , Fertilidade/genética , Infertilidade Masculina/patologia , Masculino , Camundongos , Camundongos Knockout , Fitoestrógenos/administração & dosagem , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Kisspeptina-1 , Índice de Gravidade de Doença , Espermatogênese/fisiologia , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/patologia
20.
Trends Endocrinol Metab ; 20(3): 115-21, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19097915

RESUMO

Kisspeptin binding to its G-protein-coupled receptor KISS1R (also known as GPR54), which is expressed by gonadotropin-releasing hormone (GnRH) neurons, stimulates GnRH release and activation of the mammalian reproductive axis. Kisspeptin neurons make close contact with GnRH neurons acting at both the cell body and the nerve terminals. Kisspeptin can act directly on GnRH neurons and/or indirectly via synaptic input from other neurons to inhibit inwardly rectifying potassium channels and activate non-specific cation channels, with the effect of long-lasting depolarization and increased action potential firing rate. This review covers the recent advances in the molecular consequences of kisspeptin action on GnRH neurons and how these neuronal circuits are integrated in different species. These studies provide insight into the mechanism by which kisspeptin regulates the reproductive axis.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Humanos , Kisspeptinas , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA