Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 263
Filtrar
1.
Cell Rep ; 34(1): 108587, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33406432

RESUMO

Despite a growing appreciation for microglial influences on the developing brain, the responsiveness of microglia to insults during gestation remains less well characterized, especially in the embryo when microglia themselves are still maturing. Here, we asked if fetal microglia could coordinate an innate immune response to an exogenous insult. Using time-lapse imaging, we showed that hypothalamic microglia actively surveyed their environment by near-constant "touching" of radial glia projections. However, following an insult (i.e., IUE or AAV transduction), this seemingly passive touching became more intimate and long lasting, ultimately resulting in the retraction of radial glial projections and degeneration into small pieces. Mechanistically, the TAM receptors MERTK and AXL were upregulated in microglia following the insult, and Annexin V treatment inhibited radial glia breakage and engulfment by microglia. These data demonstrate a remarkable responsiveness of embryonic microglia to insults during gestation, a critical window for neurodevelopment.


Assuntos
Embrião de Mamíferos/metabolismo , Células Ependimogliais/fisiologia , Hipotálamo/embriologia , Hipotálamo/fisiologia , Microglia/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , c-Mer Tirosina Quinase/metabolismo , Animais , Encéfalo/embriologia , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Imunidade Inata , Camundongos , Camundongos Transgênicos , Imagem Óptica/métodos , Receptor Tirosina Quinase Axl
2.
Med Sci Monit ; 26: e927334, 2020 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-33191393

RESUMO

BACKGROUND This study utilized CRISPR/Cas9 gene editing technology to construct a Mex3c gene-deficient mouse model, and studied C-FOS expression in hypothalamic nuclei. MATERIAL AND METHODS Thirty Mex3c-/+ mice, 30 mice in the normal group, and 30 Mex3c-/+ mice were randomly divided into control, leptin, and ghrelin groups according to different intraperitoneal injections. HE and Nissl staining were performed to observe the morphology of hypothalamic nerve cells. The C-FOS expression in hypothalamic nuclei of each group was analyzed by immunohistochemical techniques. HE staining was used to observe neural tube morphology, and LFB staining was used to observe nerve myelin sheath morphology. TEM was used to observe neuronal ultrastructure and immunohistochemical techniques were utilized to analyze nestin expression. RESULTS C-FOS expression was lower in the normal control group than in the leptin and ghrelin groups. The Mex3c control group and the leptin group had higher C-FOS expression than the ghrelin group. In neural tube studies, no significant differences were found in the neural tube pathological sections of E14.5-day embryos in each group. Nestin results demonstrated lower expression in the normal group and there was little difference between the HD and Mex3c groups. CONCLUSIONS Mex3c appears to participate in the regulation of energy metabolism by inducing C-FOS expression in the hypothalamus. The neural tubes of the offspring of Mex3c-/+ mice had defects during development.


Assuntos
Núcleo Celular/metabolismo , Embrião de Mamíferos/anatomia & histologia , Hipotálamo/embriologia , Tubo Neural/anatomia & histologia , Tubo Neural/embriologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Genótipo , Hipotálamo/anatomia & histologia , Camundongos Knockout , Modelos Animais , Proteínas de Ligação a RNA/metabolismo
3.
Mol Cell Endocrinol ; 518: 111030, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32931849

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons control mammalian reproduction and migrate from their birthplace in the nasal placode to the hypothalamus during development. Despite much work on the origin and migration of GnRH neurons, the processes that control GnRH lineage formation are not fully understood. Here, we demonstrate that Nhlh genes control vomeronasal receptor expression in the developing murine olfactory placode associated with the generation of the first GnRH neurons at embryonic days (E)10-12. Inactivation of ß2-microglobulin (ß2-m), which selectively affects surface expression of V2Rs, dramatically decreased the number of GnRH neurons in the Nhlh2 mutant background, preventing rescue of fertility in female Nhlh2 mutant mice by male pheromones. In addition, we show that GnRH neurons generated after E12 fail to establish synaptic connections to the vomeronasal amygdala, suggesting the existence of functionally specialized subpopulations of GnRH neurons, which process pheromonal information.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurogênese/genética , Neurônios/metabolismo , Receptores de Feromônios/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Cultivadas , Fatores Quimiotáticos/genética , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/embriologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Feromônios/metabolismo , Gravidez , Receptores de Feromônios/metabolismo
4.
Brain Res ; 1712: 167-179, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30776325

RESUMO

Gonadal hormones contribute to brain sexual differentiation. We analyzed expression of progesterone receptor (PR), estrogen receptor-α (ERα), ERß, and kisspeptin, in the preoptic area (POA) and/or the arcuate nucleus (ARC), in gonad-lacking steroidogenic factor-1 knockout (KO) mice during perinatal development. At postnatal-day (P) 0-P7, POA PR levels were higher in wild-type (WT) males compared with WT females, while those in KO males were lower than in WT males and similar to those in WT and KO females. At P14-P21, PR levels in all groups increased similarly. POA ERα levels were similar in all groups at embryonic-day (E) 15.5-P14. Those in WT but not KO males reduced during postnatal development to be significantly lower compared with females at P21. POA ERß levels were higher in WT males than in WT females, while those in KO males were lower than in WT males and similar to those in WT and KO females at P0-P21. POA kisspeptin expression was female-biased in WT mice, while levels in KO females were lower compared with WT females and similar to those in WT and KO males. ARC kisspeptin levels were equivalent among groups at E15.5-P0. At P7-P21, ARC levels in WT but not KO males became lower compared with WT females. Diethylstilbestrol exposure during P0-P6 and P7-P13 increased POA PR and ERß, and decreased POA ERα and ARC kisspeptin levels at P7 and/or P14 in both sexes of KO mice. These data further understanding of gonadal hormone action on neuronal marker expression during brain sexual development.


Assuntos
Kisspeptinas/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Gônadas , Hipotálamo/embriologia , Hipotálamo/metabolismo , Kisspeptinas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Gravidez , Área Pré-Óptica/metabolismo , Receptores de Estrogênio/genética , Receptores de Progesterona/genética , Caracteres Sexuais , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo
5.
J Neurosci ; 39(13): 2398-2415, 2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30692221

RESUMO

Primary cilia are essential for CNS development. In the mouse, they play a critical role in patterning the spinal cord and telencephalon via the regulation of Hedgehog/Gli signaling. However, despite the frequent disruption of this signaling pathway in human forebrain malformations, the role of primary cilia in forebrain morphogenesis has been little investigated outside the telencephalon. Here we studied development of the diencephalon, hypothalamus and eyes in mutant mice in which the Ftm/Rpgrip1l ciliopathy gene is disrupted. At the end of gestation, Ftm-/- fetuses displayed anophthalmia, a reduction of the ventral hypothalamus and a disorganization of diencephalic nuclei and axonal tracts. In Ftm-/- embryos, we found that the ventral forebrain structures and the rostral thalamus were missing. Optic vesicles formed but lacked the optic cups. In Ftm-/- embryos, Sonic hedgehog (Shh) expression was virtually lost in the ventral forebrain but maintained in the zona limitans intrathalamica (ZLI), the mid-diencephalic organizer. Gli activity was severely downregulated but not lost in the ventral forebrain and in regions adjacent to the Shh-expressing ZLI. Reintroduction of the repressor form of Gli3 into the Ftm-/- background restored optic cup formation. Our data thus uncover a complex role of cilia in development of the diencephalon, hypothalamus and eyes via the region-specific control of the ratio of activator and repressor forms of the Gli transcription factors. They call for a closer examination of forebrain defects in severe ciliopathies and for a search for ciliopathy genes as modifiers in other human conditions with forebrain defects.SIGNIFICANCE STATEMENT The Hedgehog (Hh) signaling pathway is essential for proper forebrain development as illustrated by a human condition called holoprosencephaly. The Hh pathway relies on primary cilia, cellular organelles that receive and transduce extracellular signals and whose dysfunctions lead to rare inherited diseases called ciliopathies. To date, the role of cilia in the forebrain has been poorly studied outside the telencephalon. In this paper we study the role of the Ftm/Rpgrip1l ciliopathy gene in mouse forebrain development. We uncover complex functions of primary cilia in forebrain morphogenesis through region-specific modulation of the Hh pathway. Our data call for further examination of forebrain defects in ciliopathies and for a search for ciliopathy genes as modifiers in human conditions affecting forebrain development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Proteína Gli3 com Dedos de Zinco/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Olho/embriologia , Olho/metabolismo , Hipotálamo/embriologia , Hipotálamo/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Tálamo/embriologia , Tálamo/metabolismo
6.
Development ; 145(21)2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30291164

RESUMO

Sonic hedgehog (Shh) plays well characterized roles in brain and spinal cord development, but its functions in the hypothalamus have been more difficult to elucidate owing to the complex neuroanatomy of this brain area. Here, we use fate mapping and conditional deletion models in mice to define requirements for dynamic Shh activity at distinct developmental stages in the tuberal hypothalamus, a brain region with important homeostatic functions. At early time points, Shh signaling regulates dorsoventral patterning, neurogenesis and the size of the ventral midline. Fate-mapping experiments demonstrate that Shh-expressing and -responsive progenitors contribute to distinct neuronal subtypes, accounting for some of the cellular heterogeneity in tuberal hypothalamic nuclei. Conditional deletion of the hedgehog transducer smoothened (Smo), after dorsoventral patterning has been established, reveals that Shh signaling is necessary to maintain proliferation and progenitor identity during peak periods of hypothalamic neurogenesis. We also find that mosaic disruption of Smo causes a non-cell autonomous gain in Shh signaling activity in neighboring wild-type cells, suggesting a mechanism for the pathogenesis of hypothalamic hamartomas, benign tumors that form during hypothalamic development.


Assuntos
Proteínas Hedgehog/metabolismo , Hipotálamo/embriologia , Hipotálamo/metabolismo , Transdução de Sinais , Animais , Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/metabolismo , Padronização Corporal , Núcleo Celular/metabolismo , Proliferação de Células , Embrião de Mamíferos/metabolismo , Camundongos , Neurogênese , Neurônios/metabolismo , Receptor Smoothened/metabolismo , Fatores de Tempo , Proteína GLI1 em Dedos de Zinco/metabolismo
7.
Physiol Rep ; 6(18): e13871, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30221477

RESUMO

Estradiol plays a critical role in stimulating the fetal hypothalamus-pituitary-adrenal axis at the end of gestation. Estradiol action is mediated through nuclear and membrane receptors that can be modulated by ICI 182,780, a pure antiestrogen compound. The objective of this study was to evaluate the transcriptomic profile of estradiol and ICI 182,780, testing the hypothesis that ICI 182,780 antagonizes the action of estradiol in the fetal hypothalamus. Chronically catheterized ovine fetuses were infused for 48 h with: vehicle (Control, n = 6), 17ß-estradiol 500 µg/kg/day (Estradiol, n = 4), ICI 182,780 5 µg/kg/day (ICI 5 µg, n = 4) and ICI 182,780 5 mg/kg/day (ICI 5 mg, n = 5). Fetal hypothalami were collected afterward, and gene expression was measured through microarray. Statistical analysis of transcriptomic data was performed with Bioconductor-R and Cytoscape software. Unexpectedly, 35% and 15.5% of the upregulated differentially expressed genes (DEG) by Estradiol significantly overlapped (P < 0.05) with upregulated DEG by ICI 5 mg and ICI 5 µg, respectively. For the downregulated DEG, these percentages were 29.9% and 15.5%, respectively. There was almost no overlap for DEG following opposite directions between Estradiol and ICI ICI 5 mg or ICI 5 µg. Furthermore, most of the genes in the estrogen signaling pathway - after activation of the epidermal growth factor receptor - followed the same direction in Estradiol, ICI 5 µg or ICI 5 mg compared to Control. In conclusion, estradiol and ICI 182,780 have estrogenic genomic effects in the developing brain, suggesting the possibility that the major action of estradiol on the fetal hypothalamus involves another receptor system rather than estrogen receptors.


Assuntos
Antagonistas do Receptor de Estrogênio/administração & dosagem , Estrogênios/administração & dosagem , Feto/efeitos dos fármacos , Fulvestranto/administração & dosagem , Hipotálamo/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Animais , Feminino , Feto/embriologia , Feto/fisiologia , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/genética , Hipotálamo/embriologia , Hipotálamo/fisiologia , Infusões Intravenosas , Gravidez , Ovinos , Transcriptoma/genética
8.
J Anat ; 232(4): 540-553, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29280147

RESUMO

The circumventricular organs (CVOs) are specialised neuroepithelial structures found in the midline of the brain, grouped around the third and fourth ventricles. They mediate the communication between the brain and the periphery by performing sensory and secretory roles, facilitated by increased vascularisation and the absence of a blood-brain barrier. Surprisingly little is known about the origins of the CVOs (both developmental and evolutionary), but their functional and organisational similarities raise the question of the extent of their relationship. Here, I review our current knowledge of the embryonic development of the seven major CVOs (area postrema, median eminence, neurohypophysis, organum vasculosum of the lamina terminalis, pineal organ, subcommissural organ, subfornical organ) in embryos of different vertebrate species. Although there are conspicuous similarities between subsets of CVOs, no unifying feature characteristic of their development has been identified. Cross-species comparisons suggest that CVOs also display a high degree of evolutionary flexibility. Thus, the term 'CVO' is merely a functional definition, and features shared by multiple CVOs may be the result of homoplasy rather than ontogenetic or phylogenetic relationships.


Assuntos
Barreira Hematoencefálica/embriologia , Órgãos Circunventriculares/embriologia , Animais , Área Postrema/anatomia & histologia , Área Postrema/fisiologia , Órgãos Circunventriculares/anatomia & histologia , Humanos , Hipotálamo/embriologia , Filogenia , Glândula Pineal/anatomia & histologia , Glândula Pineal/embriologia , Neuro-Hipófise/embriologia , Órgão Subcomissural/anatomia & histologia , Órgão Subcomissural/fisiologia , Órgão Subfornical/embriologia
9.
Neuromolecular Med ; 19(1): 136-146, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27567921

RESUMO

Reelin is an extracellular glycoprotein which contributes to synaptic plasticity and function of memory in the adult brain. It has been indicated that the Reelin signaling cascade participates in Alzheimer's disease (AD). Besides the neurons, glial cells such as astrocytes also express Reelin protein. While functional loss of astrocytes has been reported to be associated with AD, dysfunction of astrocytic Reelin signaling pathway has not received much attention. Therefore, we investigated the effects of α-boswellic acid (ABA) as one of the major component of Boswellia serrata resin on primary fetal human astrocytes under a stress paradigm as a possible model for AD through study on Reelin cascade. For this aim, we used streptozotocin (STZ), in which from an outlook generates Alzheimer's hallmarks in astrocytes, and assayed Reelin expression, Tau and Akt phosphorylation as well as reactive oxygen species (ROS) generation and apoptosis in the presences of ABA. Our results indicated that while STZ (100 µM) down-regulated the expression of Reelin, ABA (25 µM) up-regulated its expression (p < 0.01) for 24 h. ABA efficiently reduced hyperphosphorylated Tau (Ser404) in STZ-treated astrocytes (p < 0.01). Furthermore, STZ-induced apoptosis by increasing cleaved caspase three (p < 0.01) and ROS generation (p < 0.01), a further pathological hallmark of Tauopathy. On the other hand, ABA decreased ROS generation and promoted proliferation of astrocytes through elevating Survivin expression (p < 0.01). These results showed that ABA could be considered as a potent therapeutic agent for prevention and decreasing the progression of Alzheimer's hallmarks in astrocytes; however, more in vivo studies would be needed.


Assuntos
Astrócitos/efeitos dos fármacos , Moléculas de Adesão Celular Neuronais/biossíntese , Proteínas da Matriz Extracelular/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Triterpenos Pentacíclicos/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Serina Endopeptidases/biossíntese , Proteínas tau/metabolismo , Apoptose/efeitos dos fármacos , Astrócitos/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hipotálamo/citologia , Hipotálamo/embriologia , Proteínas Inibidoras de Apoptose/biossíntese , Proteínas Inibidoras de Apoptose/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Estresse Oxidativo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Reelina , Serina Endopeptidases/genética , Estreptozocina/farmacologia , Survivina
10.
Neuroendocrinology ; 105(4): 357-371, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27871073

RESUMO

BACKGROUND: Kisspeptins are important regulators of the development and function of the hypothalamic-pituitary-gonadal axis. However, the importance of kisspeptin at the pituitary level is unclear. METHODS: We examined the expression profile of kisspeptin in the mouse pituitary during development and in adulthood using RT-PCR, quantitative PCR and immunohistochemistry. RESULTS: Kiss1 mRNA was detected in both embryonic and postnatal pituitaries. Kisspeptin-immunoreactive (+) cells were detected from embryonic day (E) 13.5 throughout adulthood, being localized to the rostroventral portion in the anterior pituitary (AP) in embryos, and also to the dorsocaudal AP postnatally. A large proportion of kisspeptin+ cells were double-labeled with gonadotrope markers including Foxl2, SF-1, and LHß, and the percentage of LHß+ cells in kisspeptin+ cells increased during development. No kisspeptin+ cells were positive for the proliferating cell marker MCM7 (minichromosome maintenance protein 7), but a few kisspeptin+ cells co-expressed the stem/progenitor cell marker Sox2. Kisspeptin expression was similar between sexes and between agonadal SF-1 knockout embryos and wild-type littermates. Kiss1 mRNA levels were not significantly different between sexes or during early postnatal development, but levels in females increased when puberty began and were significantly higher than in males at postpubertal ages. CONCLUSIONS: These results suggest that kisspeptin is expressed in gonadotrope precursors during gonadotrope differentiation, and that kisspeptin expression begins soon after the initiation of αGSU production and is extinguished soon after the initiation of LH production. Furthermore, pituitary kisspeptin expression may be regulated in a gonad-independent manner during development, but may be associated with gonadotrope function in adulthood.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Gonadotrofos/metabolismo , Hipotálamo , Kisspeptinas/metabolismo , Hipófise , Fatores Etários , Animais , Animais Recém-Nascidos , Embrião de Mamíferos , Feminino , Hipotálamo/embriologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Hipófise/embriologia , Hipófise/crescimento & desenvolvimento , Hipófise/metabolismo , RNA Mensageiro/metabolismo , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Tireotropina Subunidade beta/metabolismo
11.
Mol Cell Neurosci ; 77: 87-94, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27989735

RESUMO

Deficits in neuronal migration during development in the central nervous system may contribute to psychiatric diseases. The ligand neuregulin1 (NRG1) and its receptor ErbB4 are genes conferring susceptibility to schizophrenia, playing a key role in the control of neuronal migration both during development and adulthood. Several NRG1 and ErbB4 isoforms were identified, which deeply differ in their characteristics. Here we focused on the four ErbB4 isoforms and the two NRG1 isoforms differing in their EGF-like domain, namely α and ß. We hypothesized that these isoforms, which are differently regulated in schizophrenic patients, could play different roles in neuronal migration. Our hypothesis was strengthened by the observation that both NRG1α and NRG1ß and the four ErbB4 isoforms are expressed in the medial and lateral ganglionic eminences and in the cortex during development in rat. We analysed in vitro the signal transduction pathways activated by the different ErbB4 isoforms following the treatment with soluble recombinant NRG1α or NRG1ß and the ability to stimulate migration. Our data show that two ErbB4 isoforms, namely JMa-cyt2 and JMb-cyt1, following NRG1α and NRG1ß treatment, strongly activate AKT phosphorylation, conferring high migratory activity to neuronal progenitors, thus demonstrating that both NRG1α and NRG1ß can play a role in neuronal migration.


Assuntos
Movimento Celular , Células-Tronco Neurais/metabolismo , Neuregulina-1/metabolismo , Receptor ErbB-4/metabolismo , Animais , Linhagem Celular , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Hipotálamo/citologia , Hipotálamo/embriologia , Hipotálamo/metabolismo , Células-Tronco Neurais/fisiologia , Neuregulina-1/genética , Neurogênese , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptor ErbB-4/genética
12.
Hum Mol Genet ; 25(15): 3208-3215, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27288456

RESUMO

Prader-Willi syndrome (PWS) is a genetic disorder characterized by a variety of physiological and behavioral dysregulations, including hyperphagia, a condition that can lead to life-threatening obesity. Feeding behavior is a highly complex process with multiple feedback loops that involve both peripheral and central systems. The arcuate nucleus of the hypothalamus (ARH) is critical for the regulation of homeostatic processes including feeding, and this nucleus develops during neonatal life under of the influence of both environmental and genetic factors. Although much attention has focused on the metabolic and behavioral outcomes of PWS, an understanding of its effects on the development of hypothalamic circuits remains elusive. Here, we show that mice lacking Magel2, one of the genes responsible for the etiology of PWS, display an abnormal development of ARH axonal projections. Notably, the density of anorexigenic α-melanocyte-stimulating hormone axons was reduced in adult Magel2-null mice, while the density of orexigenic agouti-related peptide fibers in the mutant mice appeared identical to that in control mice. On the basis of previous findings showing a pivotal role for metabolic hormones in hypothalamic development, we also measured leptin and ghrelin levels in Magel2-null and control neonates and found that mutant mice have normal leptin and ghrelin levels. In vitro experiments show that Magel2 directly promotes axon growth. Together, these findings suggest that a loss of Magel2 leads to the disruption of hypothalamic feeding circuits, an effect that appears to be independent of the neurodevelopmental effects of leptin and ghrelin and likely involves a direct neurotrophic effect of Magel2.


Assuntos
Antígenos de Neoplasias/metabolismo , Grelina/metabolismo , Hipotálamo/embriologia , Leptina/metabolismo , Proteínas/metabolismo , Animais , Antígenos de Neoplasias/genética , Grelina/genética , Leptina/genética , Camundongos , Camundongos Mutantes , Síndrome de Prader-Willi/embriologia , Síndrome de Prader-Willi/genética , Proteínas/genética
13.
Int J Dev Neurosci ; 53: 18-25, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27326907

RESUMO

Maternal obesity during pregnancy is associated with chronic maternal, placental, and fetal inflammation; and it elevates the risk for offspring obesity. Changes in the development of the hypothalamus, a brain region that regulates body weight and energy balance, are emerging as important determinants of offspring risk, but such changes are only beginning to be defined. Here we focused on the hypothesis that the pathological exposure of developing hypothalamic astrocytes to cytokines would alter their development. A maternal high-fat diet (mHFD) mouse model was used to investigate changes in hypothalamic astrocytes in the fetus during late gestation and in early neonates by using immunochemistry, confocal microscopy, and qPCR. The number of astrocytes and the proportion of proliferating astrocytes was significantly higher in the arcuate nucleus (ARC) and the supraoptic nucleus (SON) of the hypothalamus at both ages compared to control offspring from normal weight pregnancies. Supplemental to this we found that cultured fetal hypothalamic astrocytes proliferated significantly in response to IL6 (10ng/ml), one of the cytokines significantly elevated in fetuses of obese dams, via the JAK/STAT3 signaling pathway. Thus, maternal obesity during pregnancy stimulated the proliferation and thereby increased numbers of astrocytes in the fetal as well as early neonatal hypothalamus, which may be driven, during fetal life, by IL6.


Assuntos
Astrócitos/metabolismo , Desenvolvimento Fetal/fisiologia , Hipotálamo/embriologia , Hipotálamo/patologia , Neurônios/fisiologia , Obesidade , Animais , Astrócitos/efeitos dos fármacos , Peso Corporal , Bromodesoxiuridina/metabolismo , Proliferação de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Proteína Glial Fibrilar Ácida/metabolismo , Interleucina-6/farmacologia , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Proteína Oncogênica v-akt/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
14.
Nat Commun ; 7: 11121, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27079169

RESUMO

The anterior pituitary harbours five distinct hormone-producing cell types, and their cellular differentiation is a highly regulated and coordinated process. Here we show that ZBTB20 is essential for anterior pituitary development and lactotrope specification in mice. In anterior pituitary, ZBTB20 is highly expressed by all the mature endocrine cell types, and to some less extent by somatolactotropes, the precursors of prolactin (PRL)-producing lactotropes. Disruption of Zbtb20 leads to anterior pituitary hypoplasia, hypopituitary dwarfism and a complete loss of mature lactotropes. In ZBTB20-null mice, although lactotrope lineage commitment is normally initiated, somatolactotropes exhibit profound defects in lineage specification and expansion. Furthermore, endogenous ZBTB20 protein binds to Prl promoter, and its knockdown decreases PRL expression and secretion in a lactotrope cell line MMQ. In addition, ZBTB20 overexpression enhances the transcriptional activity of Prl promoter in vitro. In conclusion, our findings point to ZBTB20 as a critical regulator of anterior pituitary development and lactotrope specification.


Assuntos
Linhagem da Célula/genética , Lactotrofos/metabolismo , Adeno-Hipófise/metabolismo , Fatores de Transcrição/genética , Animais , Western Blotting , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Hipotálamo/embriologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Lactotrofos/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Adeno-Hipófise/embriologia , Adeno-Hipófise/crescimento & desenvolvimento , Prolactina/genética , Prolactina/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo
15.
Behav Brain Res ; 304: 125-38, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26778786

RESUMO

Embryonic exposure to ethanol is known to affect neurochemical systems in rodents and increase alcohol drinking and related behaviors in humans and rodents. With zebrafish emerging as a powerful tool for uncovering neural mechanisms of numerous diseases and exhibiting similarities to rodents, the present report building on our rat studies examined in zebrafish the effects of embryonic ethanol exposure on hypothalamic neurogenesis, expression of orexigenic neuropeptides, and voluntary ethanol consumption and locomotor behaviors in larval and adult zebrafish, and also effects of central neuropeptide injections on these behaviors affected by ethanol. At 24h post-fertilization, zebrafish embryos were exposed for 2h to ethanol, at low concentrations of 0.25% and 0.5%, in the tank water. Embryonic ethanol compared to control dose-dependently increased hypothalamic neurogenesis and the proliferation and expression of the orexigenic peptides, galanin (GAL) and orexin (OX), in the anterior hypothalamus. These changes in hypothalamic peptide neurons were accompanied by an increase in voluntary consumption of 10% ethanol-gelatin and in novelty-induced locomotor and exploratory behavior in adult zebrafish and locomotor activity in larvae. After intracerebroventricular injection, these peptides compared to vehicle had specific effects on these behaviors altered by ethanol, with GAL stimulating consumption of 10% ethanol-gelatin more than plain gelatin food and OX stimulating novelty-induced locomotor behavior while increasing intake of food and ethanol equally. These results, similar to those obtained in rats, suggest that the ethanol-induced increase in genesis and expression of these hypothalamic peptide neurons contribute to the behavioral changes induced by embryonic exposure to ethanol.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Embrião não Mamífero/efeitos dos fármacos , Etanol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo , Neuropeptídeos/metabolismo , Fatores Etários , Consumo de Bebidas Alcoólicas/patologia , Análise de Variância , Animais , Bromodesoxiuridina/metabolismo , Proteína Semelhante a ELAV 3/metabolismo , Proteína Semelhante a ELAV 4/metabolismo , Feminino , Galanina/genética , Galanina/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/embriologia , Hipotálamo/crescimento & desenvolvimento , Larva , Neurogênese/efeitos dos fármacos , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , Orexinas/genética , Orexinas/metabolismo , Gravidez , Peixe-Zebra
16.
Endocrinology ; 157(3): 1211-21, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26779746

RESUMO

The hypothalamic-pituitary-thyroid axis is governed by hypophysiotropic TRH-synthesizing neurons located in the hypothalamic paraventricular nucleus under control of the negative feedback of thyroid hormones. The mechanisms underlying the ontogeny of this phenomenon are poorly understood. We aimed to determine the onset of thyroid hormone-mediated hypothalamic-negative feedback and studied how local hypothalamic metabolism of thyroid hormones could contribute to this process in developing chicken. In situ hybridization revealed that whereas exogenous T4 did not induce a statistically significant inhibition of TRH expression in the paraventricular nucleus at embryonic day (E)19, T4 treatment was effective at 2 days after hatching (P2). In contrast, TRH expression responded to T3 treatment in both age groups. TSHß mRNA expression in the pituitary responded to T4 in a similar age-dependent manner. Type 2 deiodinase (D2) was expressed from E13 in tanycytes of the mediobasal hypothalamus, and its activity increased between E15 and P2 both in the mediobasal hypothalamus and in tanycyte-lacking hypothalamic regions. Nkx2.1 was coexpressed with D2 in E13 and P2 tanycytes and transcription of the cdio2 gene responded to Nkx2.1 in U87 glioma cells, indicating its potential role in the developmental regulation of D2 activity. The T3-degrading D3 enzyme was also detected in tanycytes, but its level was not markedly changed before and after the period of negative feedback acquisition. These findings suggest that increasing the D2-mediated T3 generation during E18-P2 could provide the sufficient local T3 concentration required for the onset of T3-dependent negative feedback in the developing chicken hypothalamus.


Assuntos
Retroalimentação Fisiológica/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Sistema Hipotálamo-Hipofisário/metabolismo , Iodeto Peroxidase/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , RNA Mensageiro/metabolismo , Glândula Tireoide/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Tiroxina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/metabolismo , Linhagem Celular Tumoral , Embrião de Galinha , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Sistema Hipotálamo-Hipofisário/embriologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/embriologia , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Iodeto Peroxidase/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/embriologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , RNA Mensageiro/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator Nuclear 1 de Tireoide , Tireotropina Subunidade beta/genética , Tiroxina/farmacologia , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Tri-Iodotironina/efeitos dos fármacos , Tri-Iodotironina/metabolismo , Iodotironina Desiodinase Tipo II
17.
Endocrine ; 52(3): 618-31, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26676302

RESUMO

Progesterone receptor (PR) presents two main isoforms (PR-A and PR-B) that are regulated by two specific promoters and transcribed from alternative transcriptional start sites. The molecular regulation of PR isoforms expression in embryonic hypothalamus is poorly understood. The aim of the present study was to assess estradiol regulation of PR isoforms in a mouse embryonic hypothalamic cell line (mHypoE-N42), as well as the transcriptional status of their promoters. MHypoE-N42 cells were treated with estradiol for 6 and 12 h. Then, Western blot, real-time quantitative reverse transcription polymerase chain reaction, and chromatin and DNA immunoprecipitation experiments were performed. PR-B expression was transiently induced by estradiol after 6 h of treatment in an estrogen receptor alpha (ERα)-dependent manner. This induction was associated with an increase in ERα phosphorylation (serine 118) and its recruitment to PR-B promoter. After 12 h of estradiol exposure, a downregulation of this PR isoform was associated with a decrease of specific protein 1, histone 3 lysine 4 trimethylation, and RNA polymerase II occupancy on PR-B promoter, without changes in DNA methylation and hydroxymethylation. In contrast, there were no estradiol-dependent changes in PR-A expression that could be related with the epigenetic marks or the transcription factors evaluated. We demonstrate that PR isoforms are differentially regulated by estradiol and that the induction of PR-B expression is associated to specific transcription factors interactions and epigenetic changes in its promoter in embryonic hypothalamic cells.


Assuntos
Estradiol/farmacologia , Hipotálamo/embriologia , Células-Tronco Neurais/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores de Progesterona/genética , Animais , Linhagem Celular , Metilação de DNA/efeitos dos fármacos , Embrião de Mamíferos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/citologia , Hipotálamo/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Isoformas de Proteínas/genética , Receptores de Progesterona/metabolismo
18.
Neurosci Lett ; 600: 85-90, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26067405

RESUMO

The nuclear receptor steroidogenic factor-1 (SF-1) plays essential roles in the development and function of the endocrine and reproductive systems. During embryogenesis, SF-1 is expressed in the ventromedial hypothalamic nucleus (VMH) and regulates the migration and terminal differentiation of the VMH neurons. Additionally, in situ hybridization data indicated SF-1 expression in the dorsal telencephalon at embryonic day (E) 13.5. In this study, we investigated the neocortical development in SF-1 knockout (KO) mouse embryos. The number of neurons was increased in the intermediate/subventricular zones and decreased in the cortical plate in the SF-1 KO embryos. SF-1 KO embryos produced more neural stem/progenitor cells, especially apical progenitor cells, and showed abnormal radial glial fiber morphology. The increase in neural stem/progenitor cells was caused by an increased S-phase fraction in the proliferative cells and the inhibition of cell cycle exit in these cells. The mRNA expression of the estrogen receptor ESRα was up-regulated and that of the estrogen synthetase Cyp19a1 was down-regulated in the dorsal telencephalon of SF-1 KO embryos. We showed that SF-1 is expressed in the dorsal telencephalon at E15.5 and E18.5, but not in adult animals. Our data demonstrated that SF-1 is involved in cell cycle regulation, neurogenesis, and neuronal migration via controlling the estrogen signaling for proper neocortical development.


Assuntos
Neocórtex/citologia , Neurogênese , Neurônios/fisiologia , Fator Esteroidogênico 1/metabolismo , Animais , Aromatase/genética , Aromatase/metabolismo , Contagem de Células , Ciclo Celular , Movimento Celular , Proliferação de Células , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Hipotálamo/citologia , Hipotálamo/embriologia , Camundongos , Camundongos Knockout , Neocórtex/embriologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurônios/citologia , Fator Esteroidogênico 1/genética
19.
Proc Natl Acad Sci U S A ; 112(15): E1861-70, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25825735

RESUMO

Food intake and body weight regulation depend on proper expression of the proopiomelanocortin gene (Pomc) in a group of neurons located in the mediobasal hypothalamus of all vertebrates. These neurons release POMC-encoded melanocortins, which are potent anorexigenic neuropeptides, and their absence from mice or humans leads to hyperphagia and severe obesity. Although the pathophysiology of hypothalamic POMC neurons is well understood, the genetic program that establishes the neuronal melanocortinergic phenotype and maintains a fully functional neuronal POMC phenotype throughout adulthood remains unknown. Here, we report that the early expression of the LIM-homeodomain transcription factor Islet 1 (ISL1) in the developing hypothalamus promotes the terminal differentiation of melanocortinergic neurons and is essential for hypothalamic Pomc expression since its initial onset and throughout the entire lifetime. We detected ISL1 in the prospective hypothalamus just before the onset of Pomc expression and, from then on, Pomc and Isl1 coexpress. ISL1 binds in vitro and in vivo to critical homeodomain binding DNA motifs present in the neuronal Pomc enhancers nPE1 and nPE2, and mutations of these sites completely disrupt the ability of these enhancers to drive reporter gene expression to hypothalamic POMC neurons in transgenic mice and zebrafish. ISL1 is necessary for hypothalamic Pomc expression during mouse and zebrafish embryogenesis. Furthermore, conditional Isl1 inactivation from POMC neurons impairs Pomc expression, leading to hyperphagia and obesity. Our results demonstrate that ISL1 specifies the identity of hypothalamic melanocortin neurons and is required for melanocortin-induced satiety and normal adiposity throughout the entire lifespan.


Assuntos
Adiposidade/fisiologia , Ingestão de Alimentos/fisiologia , Proteínas com Homeodomínio LIM/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Fatores de Transcrição/metabolismo , Adiposidade/genética , Animais , Sequência de Bases , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Ingestão de Alimentos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Hiperfagia/genética , Hiperfagia/fisiopatologia , Hipotálamo/citologia , Hipotálamo/embriologia , Hipotálamo/metabolismo , Proteínas com Homeodomínio LIM/genética , Masculino , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Dados de Sequência Molecular , Neurônios/citologia , Obesidade/genética , Obesidade/fisiopatologia , Pró-Opiomelanocortina/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/genética , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
20.
J Neuroendocrinol ; 27(4): 264-76, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25599767

RESUMO

Sex steroid hormones act on developing neural circuits regulating the hypothalamic-pituitary-gonadal axis and are involved in hormone-sensitive behaviours. These hormones act mainly via nuclear receptors, such as oestrogen receptor (ER)-α and androgen receptor (AR). By using immunohistochemistry, we analysed the expression level of ERα and AR throughout perinatal life [at embryonic (E) day 19 and postnatal (P) days 5, 15 and 25] and in adulthood in several hypothalamic nuclei controlling reproduction in both wild-type and aromatase knockout (ArKO) (i.e. which cannot convert testosterone into oestradiol) mice to determine whether there are sex differences in hypothalamic ERα and AR expression and, if so, whether these are established by the action of oestradiol. As early as E19, ERα immunoreactivity (-IR) was observed at same expression levels in both sexes in the anteroventral periventricular nucleus (AVPv), the medial preoptic area (MPOA), the bed nucleus of the stria terminalis (BnST), the ventrolateral part of the ventromedial hypothalamic nucleus and the arcuate nucleus (ARC). Sex differences (female > male) in ERα-IR were observed not only during the prepubertal period in the BnST (P5 to P25) and the MPOA (P15), but also in adulthood in these two brain regions. Sex differences in AR-IR (male > female) were observed at P5 in the AVPv and ARC, and at P25 in the MPOA and ARC, as well as in adulthood in all hypothalamic regions analysed. In adulthood, gonadectomy and hormonal treatment (oestradiol or dihydrotestosterone) also strongly modulated ERα-IR and AR, respectively. Taken together, sex differences in ERα-IR and AR-IR were observed in all hypothalamic regions analysed, although they most likely do not reflect the action of oestradiol because ArKO mice of both sexes showed expression levels very similar to wild-type mice throughout perinatal development.


Assuntos
Envelhecimento/metabolismo , Receptor alfa de Estrogênio/metabolismo , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Receptores Androgênicos/metabolismo , Animais , Embrião de Mamíferos , Feminino , Hipotálamo/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Caracteres Sexuais , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA