Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
J Neuroendocrinol ; 33(12): e13058, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34748236

RESUMO

Prenatal exposure to excess androgens is associated with the development of polycystic ovary syndrome (PCOS). In prenatally androgenised (PNA) mice, a model of PCOS, progesterone receptor (PR) protein expression is reduced in arcuate nucleus (ARC) GABA neurons. This suggests a mechanism for PCOS-related impaired steroid hormone feedback and implicates androgen excess with respect to inducing transcriptional repression of the PR-encoding gene Pgr in the ARC. However, the androgen sensitivity of ARC neurons and the relative gene expression of PRs over development and following prenatal androgen exposure remain unknown. Here, we used a quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) of microdissected ARC to determine the relative androgen receptor (Ar) and progesterone receptor (Pgr) gene expression in PNA and control mice at five developmental timepoints. In a two-way analysis of variance, none of the genes examined showed expression changes with a statistically significant interaction between treatment and age, although PgrA showed a borderline interaction. For all genes, there was a statistically significant main effect of age on expression levels, reflecting a general increase in expression with increasing age, regardless of treatment. For PgrB and Ar, there was a statistically significant main effect of treatment, indicating a change in expression following PNA (increased for PgrB and decreased for Ar), regardless of age. For PgrA, there was a borderline main effect of treatment, suggesting a possible change in expression following PNA, regardless of age. PgrAB gene expression changes showed no significant main effect of treatment. We additionally examined androgen and progesterone responsiveness specifically in P60 ARC GABA neurons using RNAScope® (Advanced Cell Diagnostics, Inc.) in situ hybridization. This analysis revealed that Pgr and Ar were expressed in the majority of ARC GABA neurons in normal adult females. However, our RNAScope® analysis did not show significant changes in Pgr or Ar expression within ARC GABA neurons following PNA. Lastly, because GABA drive to gonadotropin-releasing hormone neurons is increased in PNA, we hypothesised that PNA mice would show increased expression of glutamic acid decarboxylase (GAD), the rate-limiting enzyme in GABA production. However, the RT-qPCR showed that the expression of GAD encoding genes (Gad1 and Gad2) was unchanged in adult PNA mice compared to controls. Our findings indicate that PNA treatment can impact Pgr and Ar mRNA expression in adulthood. This may reflect altered circulating steroid hormones in PNA mice or PNA-induced epigenetic changes in the regulation of Pgr and Ar gene expression in ARC neurons.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética , Receptores Androgênicos/genética , Receptores de Progesterona/genética , Virilismo , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Crescimento e Desenvolvimento/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos/metabolismo , Receptores de Progesterona/metabolismo , Virilismo/embriologia , Virilismo/genética , Virilismo/metabolismo
2.
J Neuroendocrinol ; 33(9): e13020, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34423876

RESUMO

The arcuate nucleus of the hypothalamus is central in the regulation of body weight homeostasis through its ability to sense peripheral metabolic signals and relay them, through neural circuits, to other brain areas, ultimately affecting physiological and behavioural changes. The early postnatal development of these neural circuits is critical for normal body weight homeostasis, such that perturbations during this critical period can lead to obesity. The role for peripheral regulators of body weight homeostasis, including leptin, insulin and ghrelin, in this postnatal development is well described, yet some of the fundamental processes underpinning axonal and dendritic growth remain unclear. Here, we hypothesised that molecules known to regulate axonal and dendritic growth processes in other areas of the developing brain would be expressed in the postnatal arcuate nucleus and/or target nuclei where they would function to mediate the development of this circuitry. Using state-of-the-art RNAscope® technology, we have revealed the expression patterns of genes encoding Dcc/Netrin-1, Robo1/Slit1 and Fzd5/Wnt5a receptor/ligand pairs in the early postnatal mouse hypothalamus. We found that individual genes had unique expression patterns across developmental time in the arcuate nucleus, paraventricular nucleus of the hypothalamus, ventromedial nucleus of the hypothalamus, dorsomedial nucleus of the hypothalamus, median eminence and, somewhat unexpectedly, the third ventricle epithelium. These observations indicate a number of new molecular players in the development of neural circuits regulating body weight homeostasis, as well as novel molecular markers of tanycyte heterogeneity.


Assuntos
Genes Controladores do Desenvolvimento/fisiologia , Hipotálamo/metabolismo , Rede Nervosa/embriologia , Terceiro Ventrículo/metabolismo , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Especificidade de Órgãos/genética , Gravidez , Terceiro Ventrículo/citologia , Terceiro Ventrículo/crescimento & desenvolvimento
3.
Mol Biol Rep ; 48(4): 3773-3784, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33877530

RESUMO

The prevalence of obesity is increasing in nowadays societies and, despite being a multifactorial disease, it has a significant correlation with food intake. The control of food intake is performed by neurons of the arcuate nucleus of the hypothalamus (ARC), which secret orexigenic and anorexigenic neuropeptides, such as proopiomelanocortin (POMC), under stimulation of, e.g., ghrelin, insulin, and leptin. Insulin, uses inositol 1,4,5-trisphosphate/serine-threonine kinase (IP3/Akt) pathways and stimulates the exclusion of (Forkhead box protein O1) FOXO1 from the nucleus and thereby does the inactivation of the inhibition of POMC expression, while Leptin stimulates signal transducer and activator of transcription 3 (STAT3) phosphorylation and POMC expression. Epigenetic modifications of the synthesis of these neuropeptides can lead to an increased caloric intake, which, in turn, is an important risk factor for obesity and its comorbidities. Epigenetic modifications are reversible, so the search for epigenetic targets has significant scientific and therapeutic appeal. In this review, we synthesize the effect of food intake on the epigenetic modifications of Neuropeptide Y and Pro-opiomelanocortin of ARC and its relationships with obesity development and comorbidities. We found that there is no consensus on the methylation of neuropeptides when the evaluations are carried out in different promoters. Based on reports carried on in the early life in laboratory animals, which is the timeline that the vast majority of author used to study this topic, chronic inflammation, defects in insulin and leptin signaling may be linked to changes occurring in the phosphoinositide 3-kinase/Akt (PI3K/Akt) and/or STAT3/SOCS3 (cytokine signaling 3) pathways. In its turn, the epigenetic modifications related to increased food intake and reduced energy expenditure may be associated with PI3K/Akt and STAT3/SOCS3 signaling disruption and Pro-opiomelanocortin expression.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Ingestão de Alimentos , Epigênese Genética , Pró-Opiomelanocortina/genética , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Estresse Oxidativo , Pró-Opiomelanocortina/metabolismo , Transdução de Sinais
4.
Nutrients ; 12(11)2020 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-33138074

RESUMO

Maternal high-fat (HF) is associated with offspring hyperphagia and obesity. We hypothesized that maternal HF alters fetal neuroprogenitor cell (NPC) and hypothalamic arcuate nucleus (ARC) development with preferential differentiation of neurons towards orexigenic (NPY/AgRP) versus anorexigenic (POMC) neurons, leading to offspring hyperphagia and obesity. Furthermore, these changes may involve hypothalamic bHLH neuroregulatory factors (Hes1, Mash1, Ngn3) and energy sensor AMPK. Female mice were fed either a control or a high fat (HF) diet prior to mating, and during pregnancy and lactation. HF male newborns were heavier at birth and exhibited decreased protein expression of hypothalamic bHLH factors, pAMPK/AMPK and POMC with increased AgRP. As adults, these changes persisted though with increased ARC pAMPK/AMPK. Importantly, the total NPY neurons were increased, which was consistent with the increased food intake and adult fat mass. Further, NPCs from HF newborn hypothalamic tissue showed similar changes with preferential NPC neuronal differentiation towards NPY. Lastly, the role of AMPK was further confirmed with in vitro treatment of Control NPCs with pharmacologic AMPK modulators. Thus, the altered ARC development of HF offspring results in excess appetite and reduced satiety leading to obesity. The underlying mechanism may involve AMPK/bHLH pathways.


Assuntos
Animais Recém-Nascidos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Hiperfagia/etiologia , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/etiologia , Efeitos Tardios da Exposição Pré-Natal/etiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Apetite/fisiologia , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Feminino , Hipotálamo/metabolismo , Masculino , Camundongos , Neurogênese/fisiologia , Neurônios/metabolismo , Gravidez , Saciação/fisiologia
5.
Mol Cells ; 43(7): 600-606, 2020 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-32489185

RESUMO

Numerous physiological processes in nature have multiple oscillations within 24 h, that is, ultradian rhythms. Compared to the circadian rhythm, which has a period of approximately one day, these short oscillations range from seconds to hours, and the mechanisms underlying ultradian rhythms remain largely unknown. This review aims to explore and emphasize the implications of ultradian rhythms and their underlying regulations. Reproduction and developmental processes show ultradian rhythms, and these physiological systems can be regulated by short biological rhythms. Specifically, we recently uncovered synchronized calcium oscillations in the organotypic culture of hypothalamic arcuate nucleus (ARN) kisspeptin neurons that regulate reproduction. Synchronized calcium oscillations were dependent on voltage-gated ion channel-mediated action potentials and were repressed by chemogenetic inhibition, suggesting that the network within the ARN and between the kisspeptin population mediates the oscillation. This minireview describes that ultradian rhythms are a general theme that underlies biological features, with special reference to calcium oscillations in the hypothalamic ARN from a developmental perspective. We expect that more attention to these oscillations might provide insight into physiological or developmental mechanisms, since many oscillatory features in nature still remain to be explored.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Sinalização do Cálcio , Kisspeptinas/metabolismo , Neurônios/metabolismo , Ritmo Ultradiano , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/fisiologia , Sinalização do Cálcio/genética , Sinalização do Cálcio/fisiologia , Humanos , Recém-Nascido , Kisspeptinas/genética , Neurônios/citologia , Ritmo Ultradiano/genética , Ritmo Ultradiano/fisiologia
6.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R855-R869, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32186897

RESUMO

Angiotensin II (ANG II) Agtr1a receptor (AT1A) is expressed in cells of the arcuate nucleus of the hypothalamus that express the leptin receptor (Lepr) and agouti-related peptide (Agrp). Agtr1a expression in these cells is required to stimulate resting energy expenditure in response to leptin and high-fat diets (HFDs), but the mechanism activating AT1A signaling by leptin remains unclear. To probe the role of local paracrine/autocrine ANG II generation and signaling in this mechanism, we bred mice harboring a conditional allele for angiotensinogen (Agt, encoding AGT) with mice expressing Cre-recombinase via the Lepr or Agrp promoters to cause cell-specific deletions of Agt (AgtLepr-KO and AgtAgrp-KO mice, respectively). AgtLepr-KO mice were phenotypically normal, arguing against a paracrine/autocrine AGT signaling mechanism for metabolic control. In contrast, AgtAgrp-KO mice exhibited reduced preweaning survival, and surviving adults exhibited altered renal structure and steroid flux, paralleling previous reports of animals with whole body Agt deficiency or Agt disruption in albumin (Alb)-expressing cells (thought to cause liver-specific disruption). Surprisingly, adult AgtAgrp-KO mice exhibited normal circulating AGT protein and hepatic Agt mRNA expression but reduced Agt mRNA expression in adrenal glands. Reanalysis of RNA-sequencing data sets describing transcriptomes of normal adrenal glands suggests that Agrp and Alb are both expressed in this tissue, and fluorescent reporter gene expression confirms Cre activity in adrenal gland of both Agrp-Cre and Alb-Cre mice. These findings lead to the iconoclastic conclusion that extrahepatic (i.e., adrenal) expression of Agt is critically required for normal renal development and survival.


Assuntos
Glândulas Suprarrenais/metabolismo , Proteína Relacionada com Agouti/metabolismo , Angiotensinogênio/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético , Rim/metabolismo , Receptores para Leptina/metabolismo , Glândulas Suprarrenais/crescimento & desenvolvimento , Proteína Relacionada com Agouti/deficiência , Proteína Relacionada com Agouti/genética , Angiotensinogênio/deficiência , Angiotensinogênio/genética , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Comunicação Autócrina , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Rim/crescimento & desenvolvimento , Masculino , Camundongos Knockout , Miocárdio/metabolismo , Comunicação Parácrina , Receptores para Leptina/deficiência , Receptores para Leptina/genética , Albumina Sérica/genética , Albumina Sérica/metabolismo , Transdução de Sinais
7.
Elife ; 82019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30694175

RESUMO

Leptin regulates energy balance and also exhibits neurotrophic effects during critical developmental periods. However, the actual role of leptin during development is not yet fully understood. To uncover the importance of leptin in early life, the present study restored leptin signaling either at the fourth or tenth week of age in mice formerly null for the leptin receptor (LepR) gene. We found that some defects previously considered irreversible due to neonatal deficiency of leptin signaling, including the poor development of arcuate nucleus neural projections, were recovered by LepR reactivation in adulthood. However, LepR deficiency in early life led to irreversible obesity via suppression of energy expenditure. LepR reactivation in adulthood also led to persistent reduction in hypothalamic Pomc, Cartpt and Prlh mRNA expression and to defects in the reproductive system and brain growth. Our findings revealed that early defects in leptin signaling cause permanent metabolic, neuroendocrine and developmental problems.


Assuntos
Envelhecimento/genética , Regulação da Expressão Gênica no Desenvolvimento , Leptina/genética , Obesidade/genética , Receptores para Leptina/genética , Envelhecimento/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Metabolismo Energético/genética , Feminino , Gônadas/crescimento & desenvolvimento , Gônadas/metabolismo , Gônadas/patologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Hipotálamo/patologia , Leptina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Obesidade/metabolismo , Obesidade/patologia , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Hormônio Liberador de Prolactina/genética , Hormônio Liberador de Prolactina/metabolismo , Receptores para Leptina/deficiência , Transdução de Sinais
8.
Neuroscience ; 371: 1-15, 2018 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-29203230

RESUMO

Studies show that maternal consumption of a high-fat diet (HFD) can impair the formation of hypothalamic neuronal circuits in mouse offspring. This damage can be mediated by Notch1/Hes5 signaling activation, leading to repression of proneural factors such as Mash1 and Ngn2/3, which are essential for neuronal differentiation and neurogenesis. Thus, we aimed to investigate the effects of maternal HFD consumption during gestation and lactation on the Notch1/Mash1 pathway in the hypothalamus and arcuate nucleus (ARC) of mouse offspring (neonates and 28 days old). Our results showed that maternal HFD consumption increases body weight and adiposity of mouse offspring, accompanied by increased levels of Il-1ß mRNA compared to those in control offspring. We noticed high mRNA levels of Hes5 accompanied by diminished mRNA levels of Ascl1 (Mash1). The number of Mash1-labeled cells in the ARC was diminished in HFD-O. Additionally, the population of NPY neurons was increased in these animals. Mash1 is important for the development of POMC and NPY neurons in the ARC. Therefore, the reduction in Mash1-labeled cells could be related to modification of the NPY neuron population in the ARC. This scenario favors hyperphagia and weight gain, and could be responsible for the development of obesity in adulthood.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Ingestão de Alimentos , Hipotálamo/crescimento & desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna , Neurônios/metabolismo , Receptor Notch1/metabolismo , Adiposidade , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Peso Corporal , Ingestão de Alimentos/fisiologia , Feminino , Hipotálamo/metabolismo , Hipotálamo/patologia , Interleucina-1beta/metabolismo , Masculino , Camundongos , Neurônios/patologia , Neuropeptídeo Y/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Proteínas Repressoras/metabolismo , Transdução de Sinais
9.
J Neuroendocrinol ; 29(2)2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28071834

RESUMO

Recent data suggest that common genetic risks for metabolic disorders such as obesity may be human-specific and exert effects via the central nervous system. To overcome the limitation of human tissue access for study, we have generated induced human pluripotent stem cell (hiPSC)-derived neuronal cultures that recapture many features of hypothalamic neurones within the arcuate nucleus. In the present study, we have comprehensively characterised this model across development, benchmarked these neurones to in vivo events, and demonstrate a link between obesity risk variants and hypothalamic development. The dynamic transcriptome across neuronal maturation was examined using microarray and RNA sequencing methods at nine time points. K-means clustering of the longitudinal data was conducted to identify co-regulation and microRNA control of biological processes. The transcriptomes were compared with those of 103 samples from 13 brain regions reported in the Genotype-Tissue Expression database (GTEx) using principal components analysis. Genes with proximity to body mass index (BMI)-associated genetic variants were mapped to the developmentally expressed genesets, and enrichment significance was assessed with Fisher's exact test. The human neuronal cultures have a transcriptional and physiological profile of neuropeptide Y/agouti-related peptide arcuate nucleus neurones. The neuronal transcriptomes were highly correlated with adult hypothalamus compared to any other brain region from the GTEx. Also, approximately 25% of the transcripts showed substantial changes in expression across neuronal development and potential co-regulation of biological processes that mirror neuronal development in vivo. These developmentally expressed genes were significantly enriched for genes in proximity to BMI-associated variants. We confirmed the utility of this in vitro human model for studying the development of key hypothalamic neurones involved in energy balance and show that genes at loci associated with body weight regulation may share a pattern of developmental regulation. These data support the need to investigate early development to elucidate the human-specific central nervous system pathophysiology underlying obesity susceptibility.


Assuntos
Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/fisiologia , Loci Gênicos/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/fisiologia , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Índice de Massa Corporal , Células Cultivadas , Humanos , Neurônios/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Obesidade/genética , Transcriptoma/fisiologia
10.
Brain Res ; 1635: 27-40, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26790345

RESUMO

The hypothalamus, which plays a critical role in regulation of energy homeostasis, is formed during the perinatal period and thus vulnerable to fetal/newborn environmental conditions. We investigated synaptogenesis and neurotransmission of neurons in arcuate nucleus of the hypothalamus (ARH) during the postnatal period using immunohistochemical and electrophysiological methods. Our results show that the density of neuropeptide Y (NPY) fibers increases abruptly after the second postnatal week. NPY and proopiomelanocortin (POMC) immunoreactive fibers/varicosities puncta are mutually juxtaposed to perikarya of both neurons with increasing NPY and decreasing POMC apposition until the third postnatal week. The frequencies of spontaneous GABAergic inhibitory and glutamatergic excitatory postsynaptic currents (sIPSC and sEPSC) increase with age, with action potential dependent sIPSCs predominant during first postnatal week and sEPSCs thereafter. The presynaptic function of ARH synapses appears to reach adult levels around the age of weaning, while the postsynaptic receptors are still undergoing modification, evidenced by changes of frequencies, amplitudes and deactivation kinetics of PSCs. The number of NPY fibers juxtaposed to NPY neurons is correlated with the frequency of postsynaptic currents, suggesting that NPY/GABA release may facilitate maturation of synapses on their innervated neurons. Our results indicate that a neural circuit in ARH with a stronger NPY/GABAergic tone undergoes significant development during the postnatal period, which may be important for the maturation and/or remodeling of ARH neural circuits.


Assuntos
Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/fisiologia , Neurônios/fisiologia , Neuropeptídeo Y/metabolismo , Ácido gama-Aminobutírico/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Masculino , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley , Potenciais Sinápticos , alfa-MSH/metabolismo
11.
Nutr Neurosci ; 19(10): 467-474, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25087680

RESUMO

OBJECTIVES: Genistein is a plant-derived estrogenic isoflavone commonly found in dietary and therapeutic supplements, due to its potential health benefits. Growth hormone-releasing hormone (GHRH) and somatostatin (SS) are neurosecretory peptides synthesized in neurons of the hypothalamus and regulate the growth hormone secretion. Early reports indicate that estrogens have highly involved in the regulation of GHRH and SS secretions. Since little is known about the potential effects of genistein on GHRH and SS neurons, we exposed rats to genistein. METHODS: Genistein were administered to adult rats in dose of 30 mg/kg, for 3 weeks. The estradiol-dipropionate treatment was used as the adequate controls to genistein. Using applied stereology on histological sections of hypothalamus, we obtained the quantitative information on arcuate (Arc) and periventricular (Pe) nucleus volume and volume density of GHRH neurons and SS neurons. Image analyses were used to obtain GHRH and SS contents in the median eminence (ME). RESULTS: Administration of estradiol-dipropionate caused the increase of Arc and Pe nucleus volume, SS neuron volume density, GHRH and SS staining intensity in the ME, when compared with control. Genistein treatment increased: Arc nucleus volume and the volume density of GHRH neurons (by 26%) and SS neurons (1.5 fold), accompanied by higher GHRH and SS staining intensity in the ME, when compared to the orhidectomized group. DISCUSSION: These results suggest that genistein has a significant effect on hypothalamic region, involved in the regulation of somatotropic system function, and could contribute to the understanding of genistein as substance that alter the hormonal balance.


Assuntos
Genisteína/farmacologia , Hormônio Liberador de Hormônio do Crescimento/agonistas , Hipotálamo/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fitoestrógenos/farmacologia , Somatostatina/agonistas , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Tamanho Celular/efeitos dos fármacos , Suplementos Nutricionais/efeitos adversos , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Estradiol/análogos & derivados , Estradiol/farmacologia , Estrogênios/administração & dosagem , Estrogênios/efeitos adversos , Estrogênios/farmacologia , Genisteína/administração & dosagem , Genisteína/efeitos adversos , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Injeções Subcutâneas , Masculino , Eminência Mediana/citologia , Eminência Mediana/efeitos dos fármacos , Eminência Mediana/crescimento & desenvolvimento , Eminência Mediana/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Orquiectomia , Tamanho do Órgão/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/crescimento & desenvolvimento , Núcleo Hipotalâmico Paraventricular/metabolismo , Fitoestrógenos/administração & dosagem , Fitoestrógenos/efeitos adversos , Ratos Wistar , Somatostatina/metabolismo , Técnicas Estereotáxicas
12.
Neuroendocrinology ; 103(3-4): 369-82, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26338351

RESUMO

Neuropeptides of the hypothalamic arcuate nucleus (ARC) regulate important homeostatic and endocrine functions and also play critical roles in pubertal development. The altered peptidergic and aminoacidergic neurotransmission accompanying pubertal maturation of the ARC is not fully understood. Here we studied the developmental shift in the gene expression profile of the ARC of male mice. RNA samples for quantitative RT-PCR studies were isolated from the ARC of 14-day-old infantile and 60-day-old adult male mice with laser capture microdissection. The expression of 18 neuropeptide, 15 neuropeptide receptor, 4 sex steroid receptor and 6 classic neurotransmitter marker mRNAs was compared between the two time points. The adult animals showed increased mRNA levels encoding cocaine- and amphetamine-regulated transcripts, galanin-like peptide, dynorphin, kisspeptin, proopiomelanocortin, proenkephalin and galanin and a reduced expression of mRNAs for pituitary adenylate cyclase-activating peptide, calcitonin gene-related peptide, neuropeptide Y, substance P, agouti-related protein, neurotensin and growth hormone-releasing hormone. From the neuropeptide receptors tested, melanocortin receptor-4 showed the most striking increase (5-fold). Melanocortin receptor-3 and the Y1 and Y5 neuropeptide Y receptors increased 1.5- to 1.8-fold, whereas δ-opioid receptor and neurotensin receptor-1 transcripts were reduced by 27 and 21%, respectively. Androgen receptor, progesterone receptor and α-estrogen receptor transcripts increased by 54-72%. The mRNAs of glutamic acid decarboxylases-65 and -67, vesicular GABA transporter and choline acetyltransferase remained unchanged. Tyrosine hydroxylase mRNA increased by 44%, whereas type-2 vesicular glutamate transporter mRNA decreased by 43% by adulthood. Many of the developmental changes we revealed in this study suggest a reduced inhibitory and/or enhanced excitatory neuropeptidergic drive on fertility in adult animals.


Assuntos
Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Fatores Etários , Animais , Masculino , Camundongos , Neuropeptídeos/genética , RNA Mensageiro/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Transmissão Sináptica/genética
13.
Neuroendocrinology ; 103(3-4): 248-58, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26138474

RESUMO

Kisspeptin, a regulator of reproductive function and puberty in mammals, is expressed in the rostral (anteroventral) periventricular nucleus (AVPV) and arcuate nucleus (Arc), and its expression is at least partially regulated by estradiol in rodents. The aim of the present study was to determine contributions of genetic factors and gonadal steroid hormones to the sexual differentiation of kisspeptin-immunoreactive (kisspeptin-ir) cell populations in the AVPV and Arc during postnatal development using agonadal steroidogenic factor 1 (SF-1) knockout (KO) mice. To examine the effects of gonadal hormones on pubertal development of kisspeptin neurons, SF-1 KO mice were treated with estradiol benzoate (EB) from postnatal day (P)25 to P36, and their brains were examined at P36. No sex differences were observed in the SF-1 KO mice during postnatal development and after treatment with EB - which failed to increase the number of kisspeptin-ir cells at P36 to the levels found in wild-type (WT) control females. This suggests that specific time periods of estradiol actions or other factors are needed for sexual differentiation of the pattern of immunoreactive kisspeptin in the AVPV. Kisspeptin immunoreactivity in the Arc was significantly higher in gonadally intact WT and SF-1 KO females than in male mice at P36 during puberty. Further, in WT and SF-1 KO females, but not in males, adult levels were reached at P36. This suggests that maturation of the kisspeptin system in the Arc differs between sexes and is regulated by gonad-independent mechanisms.


Assuntos
Núcleo Arqueado do Hipotálamo , Regulação da Expressão Gênica no Desenvolvimento , Hormônios Esteroides Gonadais/farmacologia , Kisspeptinas/metabolismo , Área Pré-Óptica , Caracteres Sexuais , Fator Esteroidogênico 1/genética , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Castração , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/crescimento & desenvolvimento , Área Pré-Óptica/metabolismo , Maturidade Sexual/efeitos dos fármacos , Maturidade Sexual/genética , Fator Esteroidogênico 1/deficiência
14.
J Neurosci ; 35(22): 8558-69, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26041922

RESUMO

Neurons coexpressing neuropeptide Y, agouti-related peptide, and GABA (NAG) play an important role in ingestive behavior and are located in the arcuate nucleus of the hypothalamus. NAG neurons receive both GABAergic and glutamatergic synaptic inputs, however, the developmental time course of synaptic input organization of NAG neurons in mice is unknown. In this study, we show that these neurons have low numbers of GABAergic synapses and that GABA is inhibitory to NAG neurons during early postnatal period. In contrast, glutamatergic inputs onto NAG neurons are relatively abundant by P13 and are comparatively similar to the levels observed in the adult. As mice reach adulthood (9-10 weeks), GABAergic tone onto NAG neurons increases. At this age, NAG neurons received similar numbers of inhibitory and EPSCs. To further differentiate age-associated changes in synaptic distribution, 17- to 18-week-old lean and diet-induced obesity (DIO) mice were studied. Surprisingly, NAG neurons from lean adult mice exhibit a reduction in the GABAergic synapses compared with younger adults. Conversely, DIO mice display reductions in the number of GABAergic and glutamatergic inputs onto NAG neurons. Based on these experiments, we propose that synaptic distribution in NAG neurons is continuously restructuring throughout development to accommodate the animals' energy requirements.


Assuntos
Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Neurônios/fisiologia , Sinapses/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Fatores Etários , Animais , Animais Recém-Nascidos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Feminino , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/genética , Tetrodotoxina/farmacologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/farmacologia
15.
Dev Cogn Neurosci ; 13: 68-74, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26011750

RESUMO

Structural coherence across the arcuate fasciculus has previously been related to reading skill, but the arcuate may be divisible into distinct subtracts which support different functions. Here, we examine longitudinal data from 30 children between the ages of 8 and 14 to determine whether initial coherence in any of the arcuate's subsections is predictive of changes in reading across a longitudinal interval of approximately three years. The arcuate was divided using probabilistic tractography; mean fractional anisotropy across each subtract was extracted for each participant. Time 1 to Time 2 change in reading skill (identification, fluency score average) was significantly and uniquely predicted by only direct fronto-temporal arcuate segment coherence. Participants with lower direct segment FA demonstrated decreases in reading scores, potentially reflecting lessened improvements due to continued inefficient processing. These results were consistent in the older and younger halves of the sample. As such, we demonstrate that it is specifically the direct segment of the arcuate that may support and be predictive of reading skill both initially and longitudinally across development.


Assuntos
Envelhecimento , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Leitura , Adolescente , Envelhecimento/fisiologia , Anisotropia , Núcleo Arqueado do Hipotálamo/anatomia & histologia , Núcleo Arqueado do Hipotálamo/fisiologia , Criança , Imagem de Tensor de Difusão , Feminino , Humanos , Masculino , Rede Nervosa/crescimento & desenvolvimento , Valor Preditivo dos Testes
16.
Hum Mol Genet ; 24(15): 4276-83, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25926624

RESUMO

Prader-Willi syndrome (PWS) is a multigene disorder associated with neonatal failure to thrive, developmental delay and endocrine abnormalities suggestive of hypothalamic dysfunction. Children with PWS typically develop overt hyperphagia and obesity ∼8 years of age, later than children with other genetic forms of obesity. This suggests a postnatal developmental or degenerative component to PWS-associated obesity. De novo inactivating mutations in one PWS candidate gene, MAGEL2, have been identified in children with features of PWS. Adult mice lacking Magel2 are insensitive to the anorexic effect of leptin treatment, and their hypothalamic pro-opiomelanocortin (POMC) neurons fail to depolarize in response to leptin. However, it is unclear whether this leptin insensitivity is congenital, or whether normal leptin sensitivity in neonatal Magel2-null mice is lost postnatally. We used in vitro cytosolic calcium imaging to follow the postnatal development of leptin responses in POMC neurons in these mice. Leptin caused an activation of POMC neurons in wild-type acute hypothalamic slice preparations at all ages, reflecting their normal leptin-invoked depolarization. Normal leptin responses were found in Magel2-null mice up to 4 weeks of age, but the proportion of leptin-responsive POMC neurons was reduced in 6-week-old Magel2-null mice. The number of α-melanocyte-stimulating hormone immunoreactive fibers in the paraventricular hypothalamic nucleus was also reduced in mutant mice at 6 weeks of age. A similar progressive loss of leptin sensitivity caused by loss of MAGEL2 in children with PWS could explain the delayed onset of increased appetite and weight gain in this complex disorder.


Assuntos
Antígenos de Neoplasias/genética , Leptina/metabolismo , Neurônios/metabolismo , Síndrome de Prader-Willi/genética , Proteínas/genética , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Modelos Animais de Doenças , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Hipotálamo/patologia , Leptina/administração & dosagem , Camundongos , Neurônios/patologia , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Pró-Opiomelanocortina/metabolismo , Aumento de Peso/genética
17.
J Neurosci ; 34(30): 9982-94, 2014 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25057200

RESUMO

Leptin is well known for its role in the regulation of energy homeostasis in adults, a mechanism that at least partially results from the inhibition of the activity of NPY/AgRP/GABA neurons (NAG) in the arcuate nucleus of the hypothalamus (ARH). During early postnatal development in the rodent, leptin promotes axonal outgrowth from ARH neurons, and preautonomic NAG neurons are particularly responsive to leptin's trophic effects. To begin to understand how leptin could simultaneously promote axonal outgrowth from and inhibit the activity of NAG neurons, we characterized the electrochemical effects of leptin on NAG neurons in mice during early development. Here, we show that NAG neurons do indeed express a functional leptin receptor throughout the early postnatal period in the mouse; however, at postnatal days 13-15, leptin causes membrane depolarization in NAG neurons, rather than the expected hyperpolarization. Leptin action on NAG neurons transitions from stimulatory to inhibitory in the periweaning period, in parallel with the acquisition of functional ATP-sensitive potassium channels. These findings are consistent with the idea that leptin provides an orexigenic drive through the NAG system to help rapidly growing pups meet their energy requirements.


Assuntos
Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Leptina/fisiologia , Neurônios/fisiologia , Receptores para Leptina/fisiologia , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Masculino , Camundongos , Camundongos Transgênicos , Receptores para Leptina/biossíntese
18.
J Reprod Dev ; 60(4): 312-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24909600

RESUMO

Puberty is associated with an increase in gonadotropin secretion as a result of an increase in gonadotropin-releasing hormone (GnRH) secretion. Kisspeptin is considered to play a key role in puberty onset in many mammalian species, including rodents, ruminants and primates. The present study aimed to determine if changes in hypothalamic expression of the KISS1 gene, encoding kisspeptin, are associated with the onset of puberty in pigs. The animals (n=4 in each group) were perfused with 4% paraformaldehyde at 0, 1, 2, 3 and 4 months old, as prepubertal stages, and at 5 months old, as the peripubertal stage, following each blood sampling. KISS1 gene expressions in coronal sections of brains were visualized by in situ hybridization. Plasma luteinizing hormone (LH) was measured by radioimmunoassay. KISS1 mRNA signals were observed in the arcuate nucleus (ARC) at all ages examined without any significant difference in the number of KISS1-expressing cells, indicating that the KISS1 gene is constantly expressed in the ARC throughout pubertal development in pigs. The plasma LH concentration was the highest in 0-month-old piglets and significantly decreased in the 1- and 2 month-old groups (P<0.05), suggesting a developing negative feedback mechanism affecting gonadotropin release during the prepubertal period. Considering the potent stimulating effect of kisspeptin on gonadotropin release in prepubertal pigs, kisspeptin secretion rather than kisspeptin synthesis may be responsible for the onset of puberty in pigs.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Maturidade Sexual , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Kisspeptinas/genética , Hormônio Luteinizante/sangue , Ovário/crescimento & desenvolvimento , Progesterona/sangue , Suínos
19.
Behav Pharmacol ; 25(2): 173-81, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24561499

RESUMO

Prescription opiate use by adolescent girls has increased significantly in the past decade. Preclinical studies using rats report alterations in morphine sensitivity in the adult offspring of adolescent morphine-exposed females (MOR-F1) when compared with the offspring of adolescent saline-exposed females (SAL-F1). To begin to elucidate the development of these next generation modifications, the present study examined the effects of acute morphine administration on sedation and corticosterone secretion in prepubescent SAL-F1 and MOR-F1 male and female rats. In addition, alterations in proopiomelanocortin (POMC) gene expression in the arcuate nucleus, as well as in tyrosine hydroxylase (TH) and µ-opioid receptor (OPRM1) gene expressions in the ventral tegmental area, were analyzed using quantitative PCR, to determine whether differential regulation of these genes was correlated with the observed behavioral and/or endocrine effects. Increased morphine-induced sedation, coupled with an attenuation of morphine-induced corticosterone secretion, was observed in MOR-F1 males. Significant alterations in both POMC and OPRM1 gene expressions were also observed in MOR-F1 males, with no change in TH mRNA expression. Overall, these data suggest that the transgenerational effects of adolescent morphine exposure can be discerned before pubertal development and are more pronounced in males, and suggest dysregulation of the hypothalamic-pituitary-adrenal axis in the offspring of adolescent morphine-exposed females.


Assuntos
Exposição Materna , Morfina/farmacologia , Entorpecentes/farmacologia , Maturidade Sexual/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/fisiologia , Corticosterona/sangue , Corticosterona/metabolismo , Relação Dose-Resposta a Droga , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Masculino , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Caracteres Sexuais , Maturidade Sexual/fisiologia , Tirosina 3-Mono-Oxigenase/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/crescimento & desenvolvimento , Área Tegmentar Ventral/fisiologia
20.
Mol Endocrinol ; 28(1): 99-115, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24284824

RESUMO

Gestational exposures to estrogenic compounds, both endogenous hormones and exogenous endocrine-disrupting chemicals (EDCs), have long-term effects on reproductive physiology and behavior. We tested the hypothesis that prenatal treatment of rats with low doses of Aroclor 1221 (A1221), a weakly estrogenic polychlorinated biphenyl mix previously used in industry, or estradiol benzoate (EB), alters development of the hypothalamus in a sexually dimorphic manner and subsequently perturbs reproductive function. Pregnant Sprague-Dawley rats were injected on embryonic days 16 and 18 with vehicle (dimethylsulfoxide), A1221 (1 mg/kg), or EB (50 µg/kg). Developmental milestones were monitored, and on postnatal days 15, 30, 45, and 90, 1 male and 1 female per litter were euthanized. Because of their key roles in the mediation of steroid actions on reproductive function, the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC) were punched for a low-density quantitative PCR array of 48 neuroendocrine genes and analysis of DNA methylation of a subset of genes. Gestational exposure to A1221 or EB delayed the timing of puberty in males and disrupted estrous cyclicity in females. In the AVPV, 28 genes were affected by treatment in a developmental stage-specific manner, mostly in females, which exhibited a masculinized expression profile. This included 2 clock genes, Per2 and Arntl, implicating circadian circuits as being vulnerable to endocrine disruption. DNA methylation analysis of 2 genes, Per2 and Ar, showed no effect of EDCs and suggested alternative mechanisms for the altered mRNA levels. In the ARC, 12 genes were affected by treatment, mostly in males, again with dynamic developmental changes. Bionetwork analysis of relationships among genes, hormones, and physiological markers showed sexually dimorphic effects of estrogenic EDC exposures, with the female AVPV and the male ARC being most vulnerable, and provided novel relationships among hypothalamic genes and postnatal reproductive maturation.


Assuntos
Arocloros/toxicidade , Disruptores Endócrinos/toxicidade , Estradiol/análogos & derivados , Transtornos do Crescimento/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Análise por Conglomerados , Metilação de DNA/efeitos dos fármacos , Estradiol/toxicidade , Ciclo Estral/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Redes Reguladoras de Genes , Hormônios Esteroides Gonadais/sangue , Transtornos do Crescimento/metabolismo , Masculino , Exposição Materna , Núcleos da Linha Média do Tálamo/efeitos dos fármacos , Núcleos da Linha Média do Tálamo/crescimento & desenvolvimento , Proteínas Circadianas Period/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Regiões Promotoras Genéticas , Puberdade Tardia/induzido quimicamente , Puberdade Tardia/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/genética , Caracteres Sexuais , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...