Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Development ; 143(20): 3763-3773, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27578785

RESUMO

Neurons in the hypothalamic arcuate nucleus relay and translate important cues from the periphery into the central nervous system. However, the gene regulatory program directing their development remains poorly understood. Here, we report that the LIM-homeodomain transcription factor Isl1 is expressed in several subpopulations of developing arcuate neurons and plays crucial roles in their fate specification. Mice with conditional deletion of the Isl1 gene in developing hypothalamus display severe deficits in both feeding and linear growth. Consistent with these results, their arcuate nucleus fails to express key fate markers of Isl1-expressing neurons that regulate feeding and growth. These include the orexigenic neuropeptides AgRP and NPY for specifying AgRP-neurons, the anorexigenic neuropeptide αMSH for POMC-neurons, and two growth-stimulatory peptides, growth hormone-releasing hormone (GHRH) for GHRH-neurons and somatostatin (Sst) for Sst-neurons. Finally, we show that Isl1 directly enhances the expression of AgRP by cooperating with the key orexigenic transcription factors glucocorticoid receptor and brain-specific homeobox factor. Our results identify Isl1 as a crucial transcription factor that plays essential roles in the gene regulatory program directing development of multiple arcuate neuronal subpopulations.


Assuntos
Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Células HEK293 , Humanos , Imuno-Histoquímica , Imunoprecipitação , Proteínas com Homeodomínio LIM/genética , Camundongos , Gravidez , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética
3.
Am J Physiol Regul Integr Comp Physiol ; 316(6): R791-R801, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30943041

RESUMO

Amylin acts in the area postrema (AP) and arcuate nucleus (ARC) to control food intake. Amylin also increases axonal fiber outgrowth from the AP→nucleus tractus solitarius and from ARC→hypothalamic paraventricular nucleus. More recently, exogenous amylin infusion for 4 wk was shown to increase neurogenesis in adult rats in the AP. Furthermore, amylin has been shown to enhance leptin signaling in the ARC and ventromedial nucleus of the hypothalamus (VMN). Thus, we hypothesized that endogenous amylin could be a critical factor in regulating cell birth in the ARC and AP and that amylin could also be involved in the birth of leptin-sensitive neurons. Amylin+/- dams were injected with BrdU at embryonic day 12 and at postnatalday 2; BrdU+ cells were quantified in wild-type (WT) and amylin knockout (KO) mice. The number of BrdU+HuC/D+ neurons was similar in ARC and AP, but the number of BrdU+Iba1+ microglia was significantly decreased in both nuclei. Five-week-old WT and KO littermates were injected with leptin to test whether amylin is involved in the birth of leptin-sensitive neurons. Although there was no difference in the number of BrdU+c-Fos+ neurons in the ARC and dorsomedial nucleus, an increase in BrdU+c-Fos+ neurons was seen in VMN and lateral hypothalamus (LH) in amylin KO mice. In conclusion, these data suggest that during fetal development, endogenous amylin favors the birth of microglial cells in the ARC and AP and that it decreases the birth of leptin-sensitive neurons in the VMN and LH.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Área Postrema/metabolismo , Linhagem da Célula , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Microglia/metabolismo , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/embriologia , Área Postrema/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Região Hipotalâmica Lateral/embriologia , Região Hipotalâmica Lateral/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Leptina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Fenótipo , Gravidez , Proteínas Proto-Oncogênicas c-fos/metabolismo , Núcleo Hipotalâmico Ventromedial/embriologia , Núcleo Hipotalâmico Ventromedial/metabolismo
4.
Dev Biol ; 406(2): 235-46, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26318021

RESUMO

The mammalian arcuate nucleus (ARC) houses neurons critical for energy homeostasis and sexual maturation. Proopiomelanocortin (POMC) and Neuropeptide Y (NPY) neurons function to balance energy intake and Kisspeptin neurons are critical for the onset of puberty and reproductive function. While the physiological roles of these neurons have been well established, their development remains unclear. We have previously shown that Notch signaling plays an important role in cell fate within the ARC of mice. Active Notch signaling prevented neural progenitors from differentiating into feeding circuit neurons, whereas conditional loss of Notch signaling lead to a premature differentiation of these neurons. Presently, we hypothesized that Kisspeptin neurons would similarly be affected by Notch manipulation. To address this, we utilized mice with a conditional deletion of the Notch signaling co-factor Rbpj-κ (Rbpj cKO), or mice persistently expressing the Notch1 intracellular domain (NICD tg) within Nkx2.1 expressing cells of the developing hypothalamus. Interestingly, we found that in both models, a lack of Kisspeptin neurons are observed. This suggests that Notch signaling must be properly titrated for formation of Kisspeptin neurons. These results led us to hypothesize that Kisspeptin neurons of the ARC may arise from a different lineage of intermediate progenitors than NPY neurons and that Notch was responsible for the fate choice between these neurons. To determine if Kisspeptin neurons of the ARC differentiate similarly through a Pomc intermediate, we utilized a genetic model expressing the tdTomato fluorescent protein in all cells that have ever expressed Pomc. We observed some Kisspeptin expressing neurons labeled with the Pomc reporter similar to NPY neurons, suggesting that these distinct neurons can arise from a common progenitor. Finally, we hypothesized that temporal differences leading to premature depletion of progenitors in cKO mice lead to our observed phenotype. Using a BrdU birthdating paradigm, we determined the percentage of NPY and Kisspeptin neurons born on embryonic days 11.5, 12.5, and 13.5. We found no difference in the timing of differentiation of either neuronal subtype, with a majority occurring at e11.5. Taken together, our findings suggest that active Notch signaling is an important molecular switch involved in instructing subpopulations of progenitor cells to differentiate into Kisspeptin neurons.


Assuntos
Núcleo Arqueado do Hipotálamo/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Kisspeptinas/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Bromodesoxiuridina , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Modelos Neurológicos , Pró-Opiomelanocortina/metabolismo , Receptores Notch/metabolismo
5.
J Neurosci ; 34(10): 3756-66, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24599473

RESUMO

Puberty is a transition period of reproductive development from juvenile stages to adulthood and depends upon the activity of gonadotropin-releasing hormone (GnRH) neurons. GnRH neurons are initially activated in utero but remain quiescent throughout the juvenile period. Premature reactivation of GnRH neurons results in precocious puberty in mice and humans, but the mechanisms underlying developmental control of GnRH neuron activity remain unknown. The neuropeptide kisspeptin, a potent activator of GnRH neurons that is implicated as a critical permissive signal triggering puberty and a major regulator of the adult female hypothalamus-pituitary-gonadal axis, is paradoxically produced by neurons in the developing brain well before puberty onset. Thus, the neural circuits controlling the timing of reproductive maturation remain elusive. Here, we delineate the underlying neural circuitry using conditional genetic transsynaptic tracing in female mouse embryos. We find that kisspeptin-producing neurons in the arcuate nucleus (ARC) already communicate with a specific subset of GnRH neurons in utero. We show that ARC kisspeptin neurons are upstream of GnRH neurons, and that GnRH neuron connectivity to ARC kisspeptin neurons does not depend on their spatial position in the brain. Furthermore, we demonstrate that the neural circuits between ARC kisspeptin and GnRH neurons are fully established and operative before birth. Finally, we find that most GnRH neurons express the kisspeptin receptor GPR54 upon circuit formation, suggesting that the signaling system implicated in gatekeeping puberty becomes operative in the embryo.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina/biossíntese , Rede Nervosa/metabolismo , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Animais , Núcleo Arqueado do Hipotálamo/embriologia , Feminino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Rede Nervosa/embriologia , Gravidez , Receptores de Kisspeptina-1
6.
Adv Exp Med Biol ; 784: 27-62, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23550001

RESUMO

Our understanding of kisspeptin and its actions depends, in part, on a detailed knowledge of the neuroanatomy of the kisspeptin signaling system in the brain. In this chapter, we will review our current knowledge of the distribution of kisspeptin cells, fibers, and receptors in the mammalian brain, including the development, phenotype, and projections of different kisspeptin subpopulations. A fairly consistent picture emerges from this analysis. There are two major groups of kisspeptin cell bodies: a large number in the arcuate nucleus (ARC) and a smaller collection in the rostral periventricular area of the third ventricle (RP3V) of rodents and preoptic area (POA) of non-rodents. Both sets of neurons project to GnRH cell bodies, which contain Kiss1r, and the ARC kisspeptin population also projects to GnRH axons in the median eminence. ARC kisspeptin neurons contain neurokinin B and dynorphin, while a variable percentage of those cells in the RP3V of rodents contain galanin and/or dopamine. Neurokinin B and dynorphin have been postulated to contribute to the control of GnRH pulses and sex steroid negative feedback, while the role of galanin and dopamine in rostral kisspeptin neurons is not entirely clear. Kisspeptin neurons, fibers, and Kiss1r are found in other areas, including widespread areas outside the hypothalamus, but their physiological role(s) in these regions remains to be determined.


Assuntos
Núcleo Arqueado do Hipotálamo/embriologia , Kisspeptinas/metabolismo , Área Pré-Óptica/embriologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Terceiro Ventrículo/embriologia , Animais , Núcleo Arqueado do Hipotálamo/anatomia & histologia , Axônios/metabolismo , Dinorfinas/metabolismo , Galanina/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Neurocinina B/metabolismo , Área Pré-Óptica/anatomia & histologia , Receptores de Kisspeptina-1 , Terceiro Ventrículo/anatomia & histologia
7.
Biol Reprod ; 85(4): 650-60, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21677307

RESUMO

Kisspeptins, a family of neuropeptides encoded by the Kiss1 gene that are mainly expressed in discrete neuronal populations of the hypothalamus, have recently emerged as essential upstream regulatory elements of GnRH (gonadotropin-releasing hormone) neurons and, thereby, potent elicitors of gonadotropin secretion. Indeed, kisspeptins are now recognized as important regulators of key aspects of the maturation and function of the reproductive axis, including the sexual differentiation of the brain, the timing of puberty, the adult regulation of gonadotropin secretion by gonadal hormones, and the control of fertility by metabolic and environmental (e.g., photoperiod) cues. Appreciation of these fundamental biological features has led to the contention that kisspeptins are indispensable elements of the reproductive brain whose relevance goes beyond their crucial physiological roles and may pose potential pathophysiological and therapeutic interest. In spite of such a consensus, recent developments in the field have helped to expand, and somewhat challenged, our current understanding of the neuroendocrine and molecular mechanisms whereby some of the effects of kisspeptins are conducted. This review aims to provide a synoptic and balanced account of the consensus knowledge and recent findings in the field of kisspeptin physiology, which we predict will be crucial in shaping the progress of our understanding of the roles played by this family of neuropeptides in reproductive biology.


Assuntos
Kisspeptinas/fisiologia , Reprodução , Desenvolvimento Sexual , Animais , Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético , Retroalimentação Fisiológica , Feminino , Hormônios Esteroides Gonadais/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Leptina/metabolismo , Masculino , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Ovulação , Isoformas de Proteínas/fisiologia , Transmissão Sináptica , Núcleos Talâmicos/embriologia , Núcleos Talâmicos/metabolismo
8.
Neuron ; 109(7): 1150-1167.e6, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33600763

RESUMO

The hypothalamus plays crucial roles in regulating endocrine, autonomic, and behavioral functions via its diverse nuclei and neuronal subtypes. The developmental mechanisms underlying ontogenetic establishment of different hypothalamic nuclei and generation of neuronal diversity remain largely unknown. Here, we show that combinatorial T-box 3 (TBX3), orthopedia homeobox (OTP), and distal-less homeobox (DLX) expression delineates all arcuate nucleus (Arc) neurons and defines four distinct subpopulations, whereas combinatorial NKX2.1/SF1 and OTP/DLX expression identifies ventromedial hypothalamus (VMH) and tuberal nucleus (TuN) neuronal subpopulations, respectively. Developmental analysis indicates that all four Arc subpopulations are mosaically and simultaneously generated from embryonic Arc progenitors, whereas glutamatergic VMH neurons and GABAergic TuN neurons are sequentially generated from common embryonic VMH progenitors. Moreover, clonal lineage-tracing analysis reveals that diverse lineages from multipotent radial glia progenitors orchestrate Arc and VMH-TuN establishment. Together, our study reveals cellular mechanisms underlying generation and organization of diverse neuronal subtypes and ontogenetic establishment of individual nuclei in the mammalian hypothalamus.


Assuntos
Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Neurônios/fisiologia , Animais , Animais Geneticamente Modificados , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/embriologia , Linhagem da Célula , Ácido Glutâmico/fisiologia , Proteínas de Homeodomínio/metabolismo , Hipotálamo/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/fisiologia , Células-Tronco/fisiologia , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/embriologia , Núcleo Hipotalâmico Ventromedial/metabolismo , Ácido gama-Aminobutírico/fisiologia
9.
Nat Commun ; 10(1): 3696, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31420539

RESUMO

Despite the crucial physiological processes governed by neurons in the hypothalamic arcuate nucleus (ARC), such as growth, reproduction and energy homeostasis, the developmental pathways and regulators for ARC neurons remain understudied. Our single cell RNA-seq analyses of mouse embryonic ARC revealed many cell type-specific markers for developing ARC neurons. These markers include transcription factors whose expression is enriched in specific neuronal types and often depleted in other closely-related neuronal types, raising the possibility that these transcription factors play important roles in the fate commitment or differentiation of specific ARC neuronal types. We validated this idea with the two transcription factors, Foxp2 enriched for Ghrh-neurons and Sox14 enriched for Kisspeptin-neurons, using Foxp2- and Sox14-deficient mouse models. Taken together, our single cell transcriptome analyses for the developing ARC uncovered a panel of transcription factors that are likely to form a gene regulatory network to orchestrate fate specification and differentiation of ARC neurons.


Assuntos
Núcleo Arqueado do Hipotálamo/citologia , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/embriologia , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Kisspeptinas/metabolismo , Camundongos , Camundongos Knockout , Neurogênese/genética , Proteínas Repressoras/genética , Fatores de Transcrição SOXB2/genética , Análise de Célula Única
10.
Nat Commun ; 9(1): 2026, 2018 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-29795232

RESUMO

Despite critical roles of the hypothalamic arcuate neurons in controlling the growth and energy homeostasis, the gene regulatory network directing their development remains unclear. Here we report that the transcription factors Dlx1/2 and Otp coordinate the balanced generation of the two functionally related neurons in the hypothalamic arcuate nucleus, GHRH-neurons promoting the growth and AgRP-neurons controlling the feeding and energy expenditure. Dlx1/2-deficient mice show a loss-of-GHRH-neurons and an increase of AgRP-neurons, and consistently develop dwarfism and consume less energy. These results indicate that Dlx1/2 are crucial for specifying the GHRH-neuronal identity and, simultaneously, for suppressing AgRP-neuronal fate. We further show that Otp is required for the generation of AgRP-neurons and that Dlx1/2 repress the expression of Otp by directly binding the Otp gene. Together, our study demonstrates that the identity of GHRH- and AgRP-neurons is synchronously specified and segregated by the Dlx1/2-Otp gene regulatory axis.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Fatores de Transcrição/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/embriologia , Embrião de Galinha , Nanismo/genética , Embrião de Mamíferos , Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Feminino , Células HEK293 , Proteínas de Homeodomínio/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição/genética
11.
J Neuroendocrinol ; 19(8): 575-82, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17620099

RESUMO

Energy homeostasis is achieved by the integration of peripheral metabolic signals by neural circuits. The organisation and function of neural circuits regulating energy homeostasis has been the subject of intense investigation and has led to the definition of a core circuitry in the hypothalamus that interacts with key regions in the brain stem, which appear to mediate many of the effects of the adipocyte-derived hormone leptin on feeding and energy balance. Recent data on the ontogeny of these pathways indicate that, in rodents, these feeding circuits primarily form during neonatal life and remain structurally and functionally immature until 3 weeks of life. Our understanding of the mechanisms promoting the formation of these critical circuits has been advanced significantly by recent evidence showing that neonatal leptin acts as a neurotrophic factor promoting the development of projections from the arcuate nucleus of the hypothalamus. Together with an expanding literature on the role of nutritional factors to affect health, these discoveries may contribute to our understanding on perinatally acquired predisposition to later disease, such as obesity and diabetes.


Assuntos
Metabolismo Energético/fisiologia , Leptina/fisiologia , Vias Neurais/embriologia , Vias Neurais/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/embriologia , Tronco Encefálico/embriologia , Tronco Encefálico/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Humanos , Hipotálamo/embriologia , Hipotálamo/metabolismo , Leptina/deficiência , Leptina/farmacologia , Modelos Biológicos , Modelos Neurológicos , Vias Neurais/metabolismo , Neurônios/efeitos dos fármacos
12.
Mol Endocrinol ; 20(7): 1623-32, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16469766

RESUMO

The neuroendocrine hypothalamus regulates a number of critical biological processes and underlies a range of diseases from growth failure to obesity. Although the elucidation of hypothalamic function has progressed well, knowledge of hypothalamic development is poor. In particular, little is known about the processes underlying the neurogenesis and specification of neurons of the ventral nuclei, the arcuate and ventromedial nuclei. The proneural gene Mash1 is expressed throughout the basal retrochiasmatic neuroepithelium and loss of Mash1 results in hypoplasia of both the arcuate and ventromedial nuclei. These defects are due to a failure of neurogenesis and apoptosis, a defect that can be rescued by ectopic Ngn2 under the control of the Mash1 promoter. In addition to its role in neurogenesis, analysis of Mash1(-/-), Mash1(+/-), Mash1(KINgn2/KINgn2), and Mash1(KINgn2/+) mice demonstrates that Mash1 is specifically required for Gsh1 expression and subsequent GHRH expression, positively regulates SF1 expression, and suppresses both tyrosine hydroxylase (TH) and neuropeptide Y (NPY) expression. Although Mash1 is not required for propiomelanocortin (POMC) expression, it is required for normal development of POMC(+) neurons. These data demonstrate that Mash1 is both required for the generation of ventral neuroendocrine neurons as well as playing a central role in subtype specification of these neurons.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Diferenciação Celular/genética , Hipotálamo/embriologia , Animais , Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hipotálamo/anatomia & histologia , Perda de Heterozigosidade , Camundongos , Células Neuroepiteliais/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Quiasma Óptico/anatomia & histologia , Especificidade de Órgãos/genética , Pró-Opiomelanocortina/metabolismo , Fatores de Processamento de RNA , Fatores de Transcrição/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Regulação para Cima/genética , Núcleos Ventrais do Tálamo/anatomia & histologia , Núcleos Ventrais do Tálamo/embriologia , Núcleo Hipotalâmico Ventromedial/embriologia , Núcleo Hipotalâmico Ventromedial/metabolismo
13.
J Neuroendocrinol ; 29(12)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29121420

RESUMO

When individuals undergo gestation in an obese dam, they are at increased risk for impairments in the ability of the brain to regulate body weight. In rodents, gestation in an obese dam leads to a number of changes to the development of the hypothalamic neurones that regulate body weight, including reduced neuronal connectivity at birth. In the present study, we aimed to clarify how this neural circuitry develops normally, as well as to explore the mechanism underpinning the deficiency in connectivity seen in foetuses developing in obese dams. First, we developed an in vitro model for observing and manipulating the axonal growth of foetal arcuate nucleus (ARN) neuropeptide (NPY) neurones. We then used this model to test 2 hypotheses: (i) ARN NPY neurones respond to Netrin-1, one of a small number of axon growth and guidance factors that regulate neural circuit formation throughout the developing brain; and (ii) Netrin-1 responsiveness would be lost upon exposure to the inflammatory cytokine interleukin (IL)-6, which is elevated in foetuses developing in obese dams. We observed that ARN NPY neurones responded to Netrin-1 with a significant expansion of their growth cones, comprising the terminal apparatus that neurones use to navigate. Unexpectedly, we found further that NPY neurones from obese pregnancies had a reduced responsiveness to Netrin-1, raising the possibility that ARN NPY neurones from foetuses developing in obese dams were phenotypically different from normal NPY neurones. Finally, we observed that IL-6 treatment of normal NPY neurones in vitro led to a reduced growth cone responsiveness to Netrin-1, essentially causing them to behave similarly to NPY neurones from obese pregnancies. These results support the hypothesis that IL-6 can disrupt the normal process of axon growth from NPY neurones, and suggest one possible mechanism for how the body weight regulating circuitry fails to develop properly in the offspring of obese dams.


Assuntos
Núcleo Arqueado do Hipotálamo/embriologia , Netrina-1/fisiologia , Neurônios/fisiologia , Neuropeptídeo Y/metabolismo , Obesidade/fisiopatologia , Complicações na Gravidez/fisiopatologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Células Cultivadas , Feminino , Cones de Crescimento/fisiologia , Interleucina-6/administração & dosagem , Interleucina-6/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/metabolismo , Obesidade/complicações , Gravidez
14.
Endocrinology ; 147(8): 3681-91, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16675520

RESUMO

Humans are routinely exposed to bisphenol A (BPA), an estrogenic chemical present in food and beverage containers, dental composites, and many products in the home and workplace. BPA binds both classical nuclear estrogen receptors and facilitates membrane-initiated estrogenic effects. Here we explore the ability of environmentally relevant exposure to BPA to affect anatomical and functional measures of brain development and sexual differentiation. Anatomical evidence of alterations in brain sexual differentiation were examined in male and female offspring born to mouse dams exposed to 0, 25, or 250 ng BPA/kg body weight per day from the evening of d 8 of gestation through d 16 of lactation. These studies examined the sexually dimorphic population of tyrosine hydroxylase (TH) neurons in the rostral periventricular preoptic area, an important brain region for estrous cyclicity and estrogen-positive feedback. The significant sex differences in TH neuron number observed in control offspring were diminished or obliterated in offspring exposed to BPA primarily because of a decline in TH neuron number in BPA-exposed females. As a functional endpoint of BPA action on brain sexual differentiation, we examined the effects of perinatal BPA exposure on sexually dimorphic behaviors in the open field. Data from these studies revealed significant sex differences in the vehicle-exposed offspring that were not observed in the BPA-exposed offspring. These data indicate that BPA may be capable of altering important events during critical periods of brain development.


Assuntos
Comportamento Animal/efeitos dos fármacos , Estrogênios não Esteroides/farmacologia , Hipotálamo Anterior , Fenóis/farmacologia , Caracteres Sexuais , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Compostos Benzidrílicos , Contagem de Células , Período Crítico Psicológico , Ciclo Estral/fisiologia , Comportamento Exploratório/fisiologia , Feminino , Hipotálamo Anterior/efeitos dos fármacos , Hipotálamo Anterior/embriologia , Hipotálamo Anterior/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos , Neurônios/citologia , Neurônios/enzimologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/embriologia , Núcleo Hipotalâmico Paraventricular/crescimento & desenvolvimento , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/embriologia , Área Pré-Óptica/crescimento & desenvolvimento , Núcleos Septais/efeitos dos fármacos , Núcleos Septais/embriologia , Núcleos Septais/crescimento & desenvolvimento , Comportamento Sexual Animal/efeitos dos fármacos , Maturidade Sexual , Tirosina 3-Mono-Oxigenase/metabolismo
15.
Neuroscience ; 143(4): 975-86, 2006 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-17029798

RESUMO

In the rodent, arcuate nucleus of the hypothalamus (ARH)-derived neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons have efferent projections throughout the hypothalamus that do not fully mature until the second and third postnatal weeks. Since this process is likely completed by birth in primates we characterized the ontogeny of NPY and melanocortin systems in the fetal Japanese macaque during the late second (G100), early third (G130) and late third trimesters (G170). NPY mRNA was expressed in the ARH, paraventricular nucleus (PVH), and dorsomedial nucleus of the hypothalamus (DMH) as early as G100. ARH-derived NPY projections to the PVH were initiated at G100 but were limited and variable; however, there was a modest increase in density and number by G130. ARH-NPY/agouti-related peptide (AgRP) fiber projections to efferent target sites were completely developed by G170, but the density continued to increase in the postnatal period. In contrast to NPY/AgRP projections, alphaMSH fibers were minimal at G100 and G130 but were moderate at G170. This study also revealed several significant species differences between rodent and the nonhuman primate (NHP). There were few NPY/catecholamine projections to the PVH and ARH prior to birth, while projections were increased in the adult. A substantial proportion of the catecholamine fibers did not coexpress NPY. In addition, cocaine and amphetamine-related transcript (CART) and alpha-melanocyte stimulating hormone (alphaMSH) were not colocalized in fibers or cell bodies. As a consequence of the prenatal development of these neuropeptide systems in the NHP, the maternal environment may critically influence these circuits. Additionally, because differences exist in the neuroanatomy of NPY and melanocortin circuitry the regulation of these systems may be different in primates than in rodents.


Assuntos
Hipotálamo/embriologia , Hipotálamo/metabolismo , Macaca/embriologia , Macaca/metabolismo , Neuropeptídeos/metabolismo , Proteína Relacionada com Agouti , Animais , Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/metabolismo , Catecolaminas/metabolismo , Núcleo Hipotalâmico Dorsomedial/embriologia , Núcleo Hipotalâmico Dorsomedial/metabolismo , Feminino , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/embriologia , Vias Neurais/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Neuropeptídeos/genética , Núcleo Hipotalâmico Paraventricular/embriologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Roedores/embriologia , Roedores/metabolismo , Especificidade da Espécie , alfa-MSH/metabolismo
16.
Mol Endocrinol ; 4(8): 1257-63, 1990 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1705658

RESUMO

Insulin-like growth factor-II (IGF-II), the predominant form of IGF in fetal and neonatal serum and tissues, is found in vivo complexed with IGF-binding proteins. One of these binding proteins, IGFBP-2, is present at high levels in fetal rat plasma and binds both IGF-I and IGF-II with high affinity. We here have used in situ hybridization to compare the distribution of IGFBP-2 mRNA with that of IGF-II mRNA in embryonic day 13.5-15 rat embryos. The spatial patterns of IGF-II and IGFBP-2 expression in the fetal trunk were distinct and, in general, nonoverlapping. Most mesoderm derivatives that express IGF-II at high levels contained little, if any, IGFBP-2 mRNA. Instead, IGFBP-2 mRNA was expressed at high levels in many cell types derived from ectoderm and endoderm. The expression of IGFBP-2 mRNA in the central nervous system (CNS) during this developmental period was examined in particular detail. The three most prominent sites of IGFBP-2 expression in the CNS were comprised of cells with nonneuronal phenotypes: 1) the epithelium of the choroid plexus, a tissue that produces cerebrospinal fluid; 2) the floor plate, an area that can guide axonal outgrowth from commissural neurons of the spinal cord in vitro; and 3) the infundibulum, the progenitor of the posterior pituitary that is believed to influence differentiation of the adjacent intermediate pituitary.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Proteínas de Transporte/genética , Feto/metabolismo , Expressão Gênica , Idade Gestacional , Fator de Crescimento Insulin-Like II/genética , Animais , Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/metabolismo , Plexo Corióideo/embriologia , Plexo Corióideo/metabolismo , Ectoderma/metabolismo , Epitélio/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Mesoderma/metabolismo , Hibridização de Ácido Nucleico , Hipófise/embriologia , Hipófise/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos , Medula Espinal/embriologia , Medula Espinal/metabolismo
17.
Neurosci Behav Physiol ; 35(8): 809-13, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16132261

RESUMO

The aim of the present work was to verify the hypothesis that non-dopaminergic neurons expressing individual complementary dopamine synthesis enzymes can perform the co-located synthesis of dopamine. According to this hypothesis, neurons expressing tyrosine hydroxylase use L-tyrosine for the synthesis of L-dihydroxyphenylalanine (L-DOPA), which then enters neurons expressing aromatic amino acid decarboxylase, which converts L-DOPA to dopamine. Experiments were performed using the mediobasal hypothalamus of rat fetuses, which mostly contains single-enzyme neurons (>99%) and occasional double-enzyme neurons (<1%). Controls were obtained from the fetal substantia nigra, which is enriched with dopaminergic neurons. High-performance liquid chromatography was used to measure levels of dopamine and L-DOPA in cell extracts and the incubation medium after incubation in the presence and absence of exogenous L-tyrosine. Addition of L-tyrosine to the medium led to increases in the level of synthesis and release of L-DOPA in the mediobasal hypothalamus and substantia nigra. In addition, L-tyrosine increased dopamine synthesis in the substantia nigra and decreased dopamine synthesis in the mediobasal hypothalamus. This regional difference in levels of dopamine synthesis is probably due to inhibition of the uptake of L-DOPA from the intercellular medium by neurons in the mediobasal hypothalamus containing aromatic amino acid decarboxylase, due to the competitive binding of the L-DOPA transporter by L-tyrosine. Thus, these results provide the first evidence for the co-located synthesis of dopamine by non-dopaminergic neurons expressing single complementary enzymes involved in the synthesis of this neurotransmitter.


Assuntos
Núcleo Arqueado do Hipotálamo/enzimologia , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Dopamina/biossíntese , Neurônios/enzimologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/embriologia , Feminino , Feto/citologia , Feto/enzimologia , Levodopa/metabolismo , Masculino , Gravidez , Ratos , Ratos Wistar , Substância Negra/citologia , Substância Negra/embriologia , Substância Negra/metabolismo , Tirosina/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
18.
Endocrinology ; 156(9): 3084-90, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26172029

RESUMO

The neuropeptide kisspeptin is a major regulator of the hypothalamus-pituitary-gonadal axis. Although it has long been known that kisspeptin and its receptor G protein-coupled receptor 54 (GPR54) are expressed in the developing brain well before puberty onset, the potential role of kisspeptin/GPR54 signaling in the embryonic brain has remained mysterious. Recent studies in female mice have shown that kisspeptin neurons in the arcuate nucleus of the hypothalamus (ARC) already communicate with a subset of GnRH neurons in utero. Whether this specific neural circuit is also formed in the developing male brain is not known. Here, we used a combination of different genetic strategies to analyze the ontogeny and development of the kisspeptin/GPR54 system in the male mouse brain. We demonstrate orchestrated onset of kisspeptin and GPR54 expression in the male embryonic mouse brain and find that androgen receptor and estrogen receptor-α immunoreactivity within the male brain delineate the birthplace of kisspeptin neurons in the ARC. Using conditional transsynaptic tracing from kisspeptin neurons, we find that ARC kisspeptin neurons already communicate with a subset of GnRH neurons in utero and that the neural circuits between ARC kisspeptin and GnRH neurons in the male mouse brain are established before birth. Furthermore, we also show that the connectivity between kisspeptin and GnRH neurons does not depend on the spatial position of GnRH neurons. Our data delineate the maturing neural circuits underlying control of the reproductive axis in the male embryonic mouse brain.


Assuntos
Núcleo Arqueado do Hipotálamo/embriologia , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Gravidez , Receptores Androgênicos/metabolismo , Receptores de Kisspeptina-1
19.
J Neuropathol Exp Neurol ; 56(5): 509-22, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9143264

RESUMO

The arcuate nucleus (ARC) at the ventral surface of the human medulla has been historically considered a precerebellar nucleus. More recently, it has been implicated in central chemoreception, cardiopulmonary coupling and blood pressure responses. A deficiency of the ARC has been reported in a subset of putative human developmental disorders of ventilatory function. To investigate anatomic relationships of the ARC with brainstem regions involved in cardiorespiratory control, we applied crystals of DiI, a lipophilic dye which labels cells and cell processes by lateral diffusion along cell membranes, to 23 paraformaldehyde-fixed human fetal brainstems at 19 to 22 weeks postconceptional age. After 7 to 15.5 months diffusion, serial frozen sections were examined by florescence microscopy. DiI diffusion from the ARC labeled fibers and cell bodies in the medullary raphé, and the external arcuate fibers. Diffusion from the medullary raphé [corrected] labeled the reticular formation, medullary raphé, and the ARC. Diffusion from the pyramid and the basis pontis (negative control) labeled the corticospinal tract, with no labeling of the medullary raphé or ARC. The results suggest the existence of cellular connections between the ARC and the caudal raphé, a region implicated in cardiorespiratory control.


Assuntos
Núcleo Arqueado do Hipotálamo/embriologia , Feto/anatomia & histologia , Carbocianinas , Corantes Fluorescentes , Humanos , Bulbo/embriologia , Microscopia de Fluorescência , Vias Neurais/embriologia , Tratos Piramidais/embriologia , Núcleos da Rafe/embriologia , Formação Reticular/embriologia
20.
Endocrinology ; 139(4): 1738-45, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9528957

RESUMO

Testosterone and its metabolites are the principal gonadal hormones responsible for sexual differentiation of the brain. However, the relative roles of the androgen receptor (AR) vs. the estrogen receptor in specific aspects of this process remain unclear due to the intracellular metabolism of testosterone to active androgenic and estrogenic compounds. In this study, we used an 35S-labeled riboprobe and in situ hybridization to analyze steady state, relative levels of AR messenger RNA (mRNA) expression in the developing bed nucleus of the stria terminalis, medial preoptic area, and lateral septum, as well as the ventromedial and arcuate nuclei of the hypothalamus. Each area was examined on embryonic day 20 and postnatal days 0, 4, 10, and 20 to produce a developmental profile of AR mRNA expression. AR mRNA hybridization was present on embryonic day 20 in all areas analyzed. In addition, AR mRNA expression increased throughout the perinatal period in all areas examined in both males and females. However, between postnatal days 4 and 10, sharp increases in AR mRNA expression in the principal portion of the bed nucleus of the stria terminalis and the medial preoptic area occurred in the male that were not paralleled in the female. Subsequently, males exhibited higher levels of AR mRNA than females in these areas by postnatal day 10. There was no sex difference in AR mRNA content in the lateral septum, ventromedial nucleus, or arcuate nucleus at any age. These results suggest that sex differences in AR mRNA expression during development may lead to an early sex difference in sensitivity to the potential masculinizing effects of androgen.


Assuntos
Expressão Gênica , Prosencéfalo/embriologia , Prosencéfalo/crescimento & desenvolvimento , RNA Mensageiro/análise , Receptores Androgênicos/genética , Caracteres Sexuais , Animais , Núcleo Arqueado do Hipotálamo/química , Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Feminino , Humanos , Hibridização In Situ , Masculino , Gravidez , Área Pré-Óptica/química , Área Pré-Óptica/embriologia , Área Pré-Óptica/crescimento & desenvolvimento , Prosencéfalo/química , Ratos , Ratos Sprague-Dawley , Septo Pelúcido/química , Septo Pelúcido/embriologia , Septo Pelúcido/crescimento & desenvolvimento , Tálamo/química , Tálamo/embriologia , Tálamo/crescimento & desenvolvimento , Núcleo Hipotalâmico Ventromedial/química , Núcleo Hipotalâmico Ventromedial/embriologia , Núcleo Hipotalâmico Ventromedial/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa