Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Chem Neuroanat ; 123: 102117, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35680104

RESUMO

The hypothalamus is a large brain region made of nuclei and areas involved in the control of behaviors and physiological regulations. Among them, the arcuate nucleus (ARH) and the lateral hypothalamic area (LHA) contain key neuronal populations expressing the pro-opiomelanocortin (POMC), the agouti-related peptide (AgRP), and the melanin-concentrating hormone (MCH), respectively, that are involved in goal-oriented behaviors (such as feeding behavior) and glucose homeostasis. These neuronal populations are generated from distinct parts of the germinative neuroepithelium during embryonic life, and acquire their cell fate under the influence of morphogen proteins, specific transcription factors, and epigenetic modulators. POMC and MCH neuronal development continues by sending long descending axonal projections before birth under the control of axon guidance molecules such as Netrin1 and Slit2. Later, during the postnatal period, POMC and AgRP neurons develop intra-hypothalamic projections notably to the paraventricular nucleus of the hypothalamus through the influence of other axon guidance cues such as the class3 Semaphorins. Other cellular processes, such as autophagy and primary cilia function, and hormonal cues also appear critical for the proper development of POMC neurons.


Assuntos
Hipotálamo , Pró-Opiomelanocortina , Proteína Relacionada com Agouti/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo
2.
Exp Mol Med ; 54(4): 403-413, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35474338

RESUMO

The melanocortin system plays a critical role in the central regulation of food intake and energy balance. This system consists of neurons producing pro-opiomelanocortin (POMC), melanocortin receptors (MC4Rs), and the endogenous antagonist agouti-related peptide (AgRP). Pomc and Mc4r deficiency in rodents and humans causes early onset of obesity, whereas a loss of Agrp function is associated with leanness. Accumulating evidence shows that many chronic diseases, including obesity, might originate during early life. The melanocortin system develops during a relatively long period beginning during embryonic life with the birth of POMC and AgRP neurons and continuing postnatally with the assembly of their neuronal circuitry. The development of the melanocortin system requires the tight temporal regulation of molecular factors, such as transcription factors and axon guidance molecules, and cellular mechanisms, such as autophagy. It also involves a complex interplay of endocrine and nutritional factors. The disruption of one or more of these developmental factors can lead to abnormal maturation and function of the melanocortin system and has profound metabolic consequences later in life.


Assuntos
Melanocortinas , Pró-Opiomelanocortina , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Humanos , Hipotálamo/metabolismo , Melanocortinas/metabolismo , Obesidade/metabolismo , Peptídeos/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo
3.
Int J Obes (Lond) ; 46(6): 1138-1144, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35173277

RESUMO

BACKGROUND/OBJECTIVES: Alteration of the perinatal nutritional environment is an important risk factor for the development of metabolic diseases in later life. The hormone leptin plays a critical role in growth and development. Previous studies reported that postnatal overnutrition increases leptin secretion during the pre-weaning period. However, a direct link between leptin, neonatal overnutrition, and lifelong metabolic regulation has not been investigated. METHODS: We used the small litter mouse model combined with neonatal leptin antagonist injections to examine whether attenuating leptin during early life improves lifelong metabolic regulation in postnatally overnourished mice. RESULTS: Postnatally overnourished mice displayed rapid weight gain during lactation and remained overweight as adults. These mice also showed increased adiposity and perturbations in glucose homeostasis in adulthood. Neonatal administration of a leptin antagonist normalized fat mass and insulin sensitivity in postnatally overnourished mice. These metabolic improvements were associated with enhanced sensitivity of hypothalamic neurons to leptin. CONCLUSIONS: Early postnatal overnutrition causes metabolic alterations that can be permanently attenuated with the administration of a leptin antagonist during a restricted developmental window.


Assuntos
Leptina , Hipernutrição , Animais , Feminino , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos , Obesidade/metabolismo , Hipernutrição/metabolismo , Gravidez , Aumento de Peso
4.
Med Sci (Paris) ; 37(4): 366-371, 2021 Apr.
Artigo em Francês | MEDLINE | ID: mdl-33908854

RESUMO

The survival of the species depends on two closely interlinked processes: the correct functioning of the reproductive system, and the balance between the energy needs of an individual and the supply of energy sources through feeding. These two processes are regulated in the hypothalamus, which produces neurohormones that control various physiological functions. Among these neurohormones, GnRH controls not only the maturation and function of the reproductive organs, including the ovaries and the testes, during puberty and in adulthood, but also sexual attraction. Recent evidence suggest that neuropilin-1-mediated signaling in GnRH-synthesizing neurons could be a linchpin that holds together various neuroanatomical, physiological and behavioral adaptations involved in triggering puberty and achieving reproductive function.


TITLE: Signalisation impliquant la neuropiline dans les neurones sécrétant la GnRH - Son rôle dans le déclenchement de la puberté. ABSTRACT: La survie d'une espèce dépend de deux processus intimement liés : la reproduction, d'une part, et l'équilibre entre les besoins énergétiques et l'approvisionnement en sources d'énergie par l'alimentation, d'autre part. Ces deux processus sont contrôlés dans le cerveau par l'hypothalamus, qui produit des neurohormones agissant sur l'hypophyse pour piloter diverses fonctions physiologiques. L'une de ces neurohormones, la GnRH, contrôle non seulement la maturation et le fonctionnement des organes reproducteurs, incluant les ovaires et les testicules, lors de la puberté et à l'âge adulte, mais aussi l'attirance sexuelle. De récentes découvertes suggèrent que la signalisation impliquant la neuropiline-1 dans les neurones sécrétant la GnRH jouerait un rôle charnière dans la coordination du neurodéveloppement et des adaptations physiologiques et comportementales nécessaires au déclenchement de la puberté et à l'acquisition de la fonction de reproduction. Dans cet article de synthèse, nous replaçons ces découvertes dans le contexte de récents travaux montrant que les voies de signalisation des sémaphorines de classe 3 sont impliquées dans la physiopathologie non seulement de l'infertilité, mais aussi de l'obésité. Nous discutons également l'implication potentielle des neurones produisant la GnRH dans la perception des odeurs sociales et dans la précocité de la maturation sexuelle. L'hypothèse selon laquelle l'activité de ces neurones au cours du développement postnatal constituerait le chaînon manquant entre la prise de poids, le déclenchement de la puberté et le comportement sexuel, ouvre la voie à une meilleure compréhension de l'implication de l'homéostasie énergétique dans la maturation sexuelle, et pourrait aussi avoir des implications thérapeutiques pour la puberté précoce.


Assuntos
Hormônio Liberador de Gonadotropina/biossíntese , Neurônios/metabolismo , Neuropilina-1/metabolismo , Puberdade Precoce/etiologia , Puberdade/fisiologia , Animais , Ingestão de Energia , Metabolismo Energético/fisiologia , Feminino , Genitália/fisiologia , Humanos , Hipotálamo/fisiologia , Masculino , Camundongos , Reprodução/fisiologia , Caracteres Sexuais , Excitação Sexual
5.
Mol Metab ; 47: 101186, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33571700

RESUMO

OBJECTIVE: The ventromedial nucleus of the hypothalamus (VMH) is a critical component of the forebrain pathways that regulate energy homeostasis. It also plays an important role in the metabolic response to fasting. However, the mechanisms contributing to these physiological processes remain elusive. Autophagy is an evolutionarily conserved mechanism that maintains cellular homeostasis by turning over cellular components and providing nutrients to the cells during starvation. Here, we investigated the importance of the autophagy-related gene Atg7 in Sf1-expressing neurons of the VMH in control and fasted conditions. METHODS: We generated Sf1-Cre; Atg7loxP/loxP mice and examined their metabolic and cellular response to fasting. RESULTS: Fasting induces autophagy in the VMH, and mice lacking Atg7 in Sf1-expressing neurons display altered leptin sensitivity and impaired energy expenditure regulation in response to fasting. Moreover, loss of Atg7 in Sf1 neurons causes alterations in the central response to fasting. Furthermore, alterations in mitochondria morphology and activity are observed in mutant mice. CONCLUSION: Together, these data show that autophagy is nutritionally regulated in VMH neurons and that VMH autophagy participates in the control of energy homeostasis during fasting.


Assuntos
Autofagia , Jejum , Mitocôndrias/metabolismo , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Animais , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Metabolismo Energético , Feminino , Homeostase , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Transcriptoma
6.
Nat Commun ; 11(1): 1914, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32313051

RESUMO

Obesity is associated with the activation of cellular responses, such as endoplasmic reticulum (ER) stress. Here, we show that leptin-deficient ob/ob mice display elevated hypothalamic ER stress as early as postnatal day 10, i.e., prior to the development of obesity in this mouse model. Neonatal treatment of ob/ob mice with the ER stress-relieving drug tauroursodeoxycholic acid (TUDCA) causes long-term amelioration of body weight, food intake, glucose homeostasis, and pro-opiomelanocortin (POMC) projections. Cells exposed to ER stress often activate autophagy. Accordingly, we report that in vitro induction of ER stress and neonatal leptin deficiency in vivo activate hypothalamic autophagy-related genes. Furthermore, genetic deletion of autophagy in pro-opiomelanocortin neurons of ob/ob mice worsens their glucose homeostasis, adiposity, hyperphagia, and POMC neuronal projections, all of which are ameliorated with neonatal TUDCA treatment. Together, our data highlight the importance of early life ER stress-autophagy pathway in influencing hypothalamic circuits and metabolic regulation.


Assuntos
Autofagia/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Leptina/metabolismo , Neurogênese/fisiologia , Adiposidade , Animais , Antivirais/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína 7 Relacionada à Autofagia/genética , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Colagogos e Coleréticos/farmacologia , Modelos Animais de Doenças , Ingestão de Alimentos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Comportamento Alimentar , Homeostase , Hiperfagia/metabolismo , Leptina/genética , Masculino , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Neuroendocrinologia , Neurogênese/efeitos dos fármacos , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Ácido Tauroquenodesoxicólico
7.
PLoS Biol ; 18(3): e3000296, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32163401

RESUMO

The steady increase in the prevalence of obesity and associated type II diabetes mellitus is a major health concern, particularly among children. Maternal obesity represents a risk factor that contributes to metabolic perturbations in the offspring. Endoplasmic reticulum (ER) stress has emerged as a critical mechanism involved in leptin resistance and type 2 diabetes in adult individuals. Here, we used a mouse model of maternal obesity to investigate the importance of early life ER stress in the nutritional programming of this metabolic disease. Offspring of obese dams developed glucose intolerance and displayed increased body weight, adiposity, and food intake. Moreover, maternal obesity disrupted the development of melanocortin circuits associated with neonatal hyperleptinemia and leptin resistance. ER stress-related genes were up-regulated in the hypothalamus of neonates born to obese mothers. Neonatal treatment with the ER stress-relieving drug tauroursodeoxycholic acid improved metabolic and neurodevelopmental deficits and reversed leptin resistance in the offspring of obese dams.


Assuntos
Estresse do Retículo Endoplasmático , Hipotálamo/crescimento & desenvolvimento , Obesidade Materna/metabolismo , Animais , Animais Recém-Nascidos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Composição Corporal , Peso Corporal , Dieta/efeitos adversos , Estresse do Retículo Endoplasmático/genética , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/embriologia , Hipotálamo/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Pâncreas/crescimento & desenvolvimento , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Pró-Opiomelanocortina/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , alfa-MSH/metabolismo
8.
Cell ; 176(4): 729-742.e18, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30661757

RESUMO

Hypothalamic melanocortin neurons play a pivotal role in weight regulation. Here, we examined the contribution of Semaphorin 3 (SEMA3) signaling to the development of these circuits. In genetic studies, we found 40 rare variants in SEMA3A-G and their receptors (PLXNA1-4; NRP1-2) in 573 severely obese individuals; variants disrupted secretion and/or signaling through multiple molecular mechanisms. Rare variants in this set of genes were significantly enriched in 982 severely obese cases compared to 4,449 controls. In a zebrafish mutagenesis screen, deletion of 7 genes in this pathway led to increased somatic growth and/or adiposity demonstrating that disruption of Semaphorin 3 signaling perturbs energy homeostasis. In mice, deletion of the Neuropilin-2 receptor in Pro-opiomelanocortin neurons disrupted their projections from the arcuate to the paraventricular nucleus, reduced energy expenditure, and caused weight gain. Cumulatively, these studies demonstrate that SEMA3-mediated signaling drives the development of hypothalamic melanocortin circuits involved in energy homeostasis.


Assuntos
Metabolismo Energético/genética , Melanocortinas/metabolismo , Semaforinas/genética , Adolescente , Adulto , Animais , Peso Corporal , Linhagem Celular , Criança , Pré-Escolar , Modelos Animais de Doenças , Ingestão de Alimentos , Feminino , Variação Genética/genética , Homeostase , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Obesidade/genética , Obesidade/metabolismo , Receptores de Superfície Celular/metabolismo , Semaforinas/metabolismo , Adulto Jovem , Peixe-Zebra
9.
Cell Rep ; 22(13): 3401-3408, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590610

RESUMO

Transcriptional analysis of brain tissue from people with molecularly defined causes of obesity may highlight disease mechanisms and therapeutic targets. We performed RNA sequencing of hypothalamus from individuals with Prader-Willi syndrome (PWS), a genetic obesity syndrome characterized by severe hyperphagia. We found that upregulated genes overlap with the transcriptome of mouse Agrp neurons that signal hunger, while downregulated genes overlap with the expression profile of Pomc neurons activated by feeding. Downregulated genes are expressed mainly in neuronal cells and contribute to neurogenesis, neurotransmitter release, and synaptic plasticity, while upregulated, predominantly microglial genes are involved in inflammatory responses. This transcriptional signature may be mediated by reduced brain-derived neurotrophic factor expression. Additionally, we implicate disruption of alternative splicing as a potential molecular mechanism underlying neuronal dysfunction in PWS. Transcriptomic analysis of the human hypothalamus may identify neural mechanisms involved in energy homeostasis and potential therapeutic targets for weight loss.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Jejum/fisiologia , Hipotálamo/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Camundongos , Obesidade/metabolismo , Síndrome de Prader-Willi/patologia , Transcriptoma
10.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1032-R1044, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27629888

RESUMO

Selectively bred diet-induced obese (DIO) rats become obese on a high-fat diet and are leptin resistant before becoming obese. Compared with diet-resistant (DR) neonates, DIO neonates have impaired leptin-dependent arcuate (ARC) neuropeptide Y/agouti-related peptide (NPY/AgRP) and α-melanocyte-stimulating hormone (α-MSH; from proopiomelanocortin (POMC) neurons) axon outgrowth to the paraventricular nucleus (PVN). Using phosphorylation of STAT3 (pSTAT3) as a surrogate, we show that reduced DIO ARC leptin signaling develops by postnatal day 7 (P7) and is reduced within POMC but not NPY/AgRP neurons. Since amylin increases leptin signaling in adult rats, we treated DIO neonates with amylin during postnatal hypothalamic development and assessed leptin signaling, leptin-dependent ARC-PVN pathway development, and metabolic changes. DIO neonates treated with amylin from P0-6 and from P0-16 increased ARC leptin signaling and both AgRP and α-MSH ARC-PVN pathway development, but increased only POMC neuron number. Despite ARC-PVN pathway correction, P0-16 amylin-induced reductions in body weight did not persist beyond treatment cessation. Since amylin enhances adult DIO ARC signaling via an IL-6-dependent mechanism, we assessed ARC-PVN pathway competency in IL-6 knockout mice and found that the AgRP, but not the α-MSH, ARC-PVN pathway was reduced. These results suggest that both leptin and amylin are important neurotrophic factors for the postnatal development of the ARC-PVN pathway. Amylin might act as a direct neurotrophic factor in DIO rats to enhance both the number of POMC neurons and their α-MSH ARC-PVN pathway development. This suggests important and selective roles for amylin during ARC hypothalamic development.


Assuntos
Hipotálamo/fisiopatologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/administração & dosagem , Leptina/metabolismo , Obesidade/tratamento farmacológico , Obesidade/fisiopatologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/patologia , Núcleo Arqueado do Hipotálamo/fisiopatologia , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Gorduras na Dieta , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/farmacologia , Masculino , Neurogênese/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Cuidado Pós-Natal , Ratos , Resultado do Tratamento
11.
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
12.
Cell Metab ; 19(2): 293-301, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24506870

RESUMO

Leptin secreted by adipocytes acts on the brain to reduce food intake by regulating neuronal activity in the mediobasal hypothalamus (MBH). Obesity is associated with resistance to high circulating leptin levels. Here, we demonstrate that peripherally administered leptin activates its receptor (LepR) in median eminence tanycytes followed by MBH neurons, a process requiring tanycytic ERK signaling and the passage of leptin through the cerebrospinal fluid. In mice lacking the signal-transducing LepRb isoform or with diet-induced obesity, leptin taken up by tanycytes accumulates in the median eminence and fails to reach the MBH. Triggering ERK signaling in tanycytes with EGF reestablishes leptin transport, elicits MBH neuron activation and energy expenditure in obese animals, and accelerates the restoration of leptin sensitivity upon the return to a normal-fat diet. ERK-dependent leptin transport by tanycytes could thus play a critical role in the pathophysiology of leptin resistance, and holds therapeutic potential for treating obesity.


Assuntos
Encéfalo/metabolismo , Células Ependimogliais/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Animais , Western Blotting , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
13.
Front Neuroendocrinol ; 34(1): 18-26, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23357643

RESUMO

Brain development is a complex and dynamic process, and many environmental factors have been found to influence the normal development of neural pathways. Cumulative evidence suggests that metabolic hormones that regulate the hypothalamic circuits that control energy homeostasis function in much the same way that sex steroids act on sexually dimorphic circuits. For example, although the effects of the adipocyte-derived hormone leptin were originally thought to be limited to the neural control of energy homeostasis in adult animals, it is now becoming increasingly clear that leptin can also determine patterns of neurogenesis, axon growth, and synaptic plasticity in the developing hypothalamus. More recent studies have also extended the role of the metabolic hormones ghrelin and insulin in various aspects of brain development. Examining how metabolic hormones control hypothalamic development will help our understanding of the developmental origin of adult metabolic diseases and, hopefully, improve our ability to predict adverse outcomes.


Assuntos
Grelina/fisiologia , Hipotálamo/metabolismo , Insulina/fisiologia , Leptina/fisiologia , Neurogênese/fisiologia , Animais , Grelina/metabolismo , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/fisiologia , Insulina/metabolismo , Leptina/metabolismo , Camundongos , Vias Neurais/fisiologia , Ratos
14.
J Neurosci ; 32(33): 11486-94, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22895731

RESUMO

It is increasingly accepted that alterations of the early life environment may have lasting impacts on physiological functions. In particular, epidemiological and animal studies have indicated that changes in growth and nutrition during childhood and adolescence can impair reproductive function. However, the precise biological mechanisms that underlie these programming effects of neonatal nutrition on reproduction are still poorly understood. Here, we used a mouse model of divergent litter size to investigate the effects of early postnatal overnutrition and undernutrition on the maturation of hypothalamic circuits involved in reproductive function. Neonatally undernourished females display attenuated postnatal growth associated with delayed puberty and defective development of axonal projections from the arcuate nucleus to the preoptic region. These alterations persist into adulthood and specifically affect the organization of neural projections containing kisspeptin, a key neuropeptide involved in pubertal activation and fertility. Neonatal overfeeding also perturbs the development of neural projections from the arcuate nucleus to the preoptic region, but it does not result in alterations in kisspeptin projections. These studies indicate that alterations in the early nutritional environment cause lasting and deleterious effects on the organization of neural circuits involved in the control of reproduction, and that these changes are associated with lifelong functional perturbations.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipotálamo/citologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Estado Nutricional/fisiologia , Reprodução/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Peso Corporal , Mapeamento Encefálico , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/crescimento & desenvolvimento , Kisspeptinas/metabolismo , Tamanho da Ninhada de Vivíparos , Hormônio Luteinizante/metabolismo , Masculino , Desnutrição/metabolismo , Desnutrição/patologia , Camundongos , Fibras Nervosas/fisiologia , Neurocinina B/metabolismo , Neurônios/metabolismo , Ovariectomia , Hipernutrição/metabolismo , Hipernutrição/patologia , Fatores Sexuais
15.
Endocrinology ; 153(8): 3657-67, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22621961

RESUMO

The hypothalamus plays a critical role in the regulation of energy balance. Neuroanatomical and mouse genetic data have defined a core circuitry in the hypothalamus that mediates many of the effects of leptin on feeding and energy balance regulation. The present study used 5-bromo-2'-deoxyuridine (a marker of dividing cells) and a neuronal marker to systematically examine neurogenesis in the mouse embryonic hypothalamus, particularly the birth of neurons that relay leptin signaling. The vast majority of neurons in hypothalamic nuclei known to control energy balance is generated between embryonic days (E) 12 and E16, with a sharp peak of neurogenesis occurring on E12. Neurons in the dorsomedial and paraventricular nuclei and the lateral hypothalamic area are born between E12 and E14. The arcuate and ventromedial nuclei exhibit a relatively longer neurogenic period. Many neurons in these nuclei are born on E12, but some neurons are generated as late as E16. We also examined the birth of leptin-activated cells by coupling the 5-bromo-2'-deoxyuridine staining with cFos immunohistochemistry. Remarkably, the majority of leptin-activated cells in the adult hypothalamus were also born during a discrete developmental window on E12. These results provide new insight into the development of hypothalamic neurons that control feeding and identify important developmental periods when alterations in the intrauterine environment may affect hypothalamic neurogenesis and produce long-term consequences on hypothalamic cell numbers.


Assuntos
Hipotálamo/citologia , Hipotálamo/embriologia , Leptina/farmacologia , Neurônios/citologia , Neurônios/metabolismo , Animais , Bromodesoxiuridina/farmacologia , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Gravidez
16.
J Neurosci ; 32(4): 1244-52, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22279209

RESUMO

Circulating hormones influence multiple aspects of hypothalamic development and play a role in directing formation of neural circuits. Leptin is secreted by adipocytes and functions as a key developmental signal that promotes axon outgrowth from the arcuate nucleus (ARH) during a discrete developmental critical period. To determine the cellular mechanisms by which leptin impacts development of hypothalamic circuits, we examined roles for leptin receptor (LepRb) signals in neonatal mice. LepRb, ERK, and STAT3 signaling were required for leptin-stimulated neurite outgrowth from ARH explants in vitro. Neonatal mice with disrupted LepRb→ERK signaling displayed impaired ARH projections but were able to compensate by adulthood. LepRb→STAT3 signaling also plays a role in early circuit formation and controls the ultimate architecture of POMC, but not AgRP, projections. Thus, the developmental actions of leptin on feeding circuits are dependent on LepRb, and distinct signaling pathways are responsible for directing formation of NPY and POMC projections.


Assuntos
Comportamento Alimentar/fisiologia , Hipotálamo/crescimento & desenvolvimento , Rede Nervosa/crescimento & desenvolvimento , Receptores para Leptina/fisiologia , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Humanos , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/fisiologia , Vias Neurais/fisiologia , Técnicas de Cultura de Órgãos
17.
Endocrinology ; 152(11): 4171-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21862611

RESUMO

Maternal diabetes is a common complication of pregnancy, and the offspring of diabetic mothers have a higher risk of developing obesity and type 2 diabetes later in life. Despite these observations, the precise biological processes mediating this metabolic programming are not well understood. Here, we explored the consequences of maternal diabetes on the organization of hypothalamic neural circuits involved in the regulation of energy balance. To accomplish this aim, we used a mouse model of maternal insulin deficiency induced by streptozotocin injections. Maternal diabetes was found to be associated with changes in offspring growth as revealed by a significantly higher pre- and postweaning body weight in the offspring of insulin-deficient dams relative to those of control mice. Mice born to diabetic dams also showed increased fasting glucose levels, increased insulin levels, and increased food intake during their adult lives. These impairments in metabolic regulation were associated with leptin resistance during adulthood. Importantly, the ability of leptin to activate intracellular signaling in arcuate neurons was also significantly reduced in neonates born to diabetic dams. Furthermore, neural projections from the arcuate nucleus to the paraventricular nucleus were markedly reduced in the offspring of insulin-deficient dams. Together, these data show that insulin deficiency during gestation has long-term consequences for metabolic regulation. They also indicate that animals born to diabetic dams display abnormally organized hypothalamic feeding pathways that could result from the attenuated responsiveness of hypothalamic neurons to the neurotrophic actions of leptin during neonatal development.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Diabetes Gestacional/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Rede Nervosa/metabolismo , Neurônios/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiopatologia , Animais , Diabetes Mellitus Tipo 2/fisiopatologia , Diabetes Gestacional/fisiopatologia , Ingestão de Alimentos/fisiologia , Feminino , Hipotálamo/fisiopatologia , Insulina/metabolismo , Camundongos , Rede Nervosa/fisiopatologia , Obesidade/metabolismo , Obesidade/fisiopatologia , Fosforilação , Gravidez , Fator de Transcrição STAT3/metabolismo
18.
Peptides ; 32(11): 2362-6, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21736909

RESUMO

Ghrelin is a pleiotropic hormone that was originally described as promoting feeding and stimulating growth hormone release in adults. A growing body of evidence suggests that ghrelin may also exert developmental and organizational effects during perinatal life. The perinatal actions of ghrelin include the regulation of early developmental events such as blastocyst development and perinatal growth. Moreover, alterations in perinatal ghrelin levels result in structural differences in various peripheral organs, such as the pancreas and gastrointestinal tract. Recent data have also suggested that ghrelin acts on appetite-related brain centers in early life. Together, these observations indicate that exposure to factors that alter how ghrelin impacts development may induce lasting effects on physiological regulation.


Assuntos
Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Grelina/metabolismo , Hipotálamo/metabolismo , Neurogênese/fisiologia , Pâncreas/metabolismo , Receptores de Grelina/metabolismo , Animais , Apetite/fisiologia , Regulação do Apetite/fisiologia , Feto , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/crescimento & desenvolvimento , Grelina/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Hipotálamo/embriologia , Hipotálamo/crescimento & desenvolvimento , Recém-Nascido , Camundongos , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Perinatologia , Ratos , Receptores de Grelina/genética
19.
J Mol Neurosci ; 43(3): 478-84, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21086065

RESUMO

We tested the hypothesis that astrocytic activity modulates neuronal uptake and signaling of leptin in the adult-onset obese agouti viable yellow (A vy) mouse. In the immunohistochemical study, A vy mice were pretreated with the astrocyte metabolic inhibitor fluorocitrate or phosphate-buffered saline (PBS) vehicle intracerebroventricularly (icv) followed 1 h later by Alexa568-leptin. Confocal microscopy showed that fluorocitrate pretreatment reduced astrocytic uptake of Alexa568-leptin 30 min after icv while increasing neuronal uptake in the arcuate nucleus and dorsomedial hypothalamus. Fluorocitrate also induced mild astrogliosis and moderately increased pSTAT3 immunopositive neurons in response to Alexa568-leptin in the dorsomedial hypothalamus. In the Western blotting study, A vy mice were pretreated with either PBS or fluorocitrate, and received PBS or leptin 1 h later followed by determination of pSTAT3 and GFAP expression an additional 30 min afterward. The results show that fluorocitrate induced a mild pSTAT3 activation but attenuated leptin-induced pSTAT3 activation and decreased GFAP expression independently of leptin treatment. We conclude that inhibition of astrocytic activity resulted in enhanced neuronal leptin uptake and signaling. This suggests opposite roles of astrocytes and neurons in leptin's actions in the A vy mouse with adult-onset obesity.


Assuntos
Astrócitos/metabolismo , Hipotálamo/citologia , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos Obesos , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Animais , Astrócitos/citologia , Corantes Fluorescentes/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Infusões Intraventriculares , Leptina/administração & dosagem , Masculino , Camundongos , Neurônios/citologia , Fator de Transcrição STAT3/metabolismo
20.
Nestle Nutr Workshop Ser Pediatr Program ; 65: 25-35; discussion 35-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20139672

RESUMO

A large body of epidemiological data suggests that adverse early environments, including obesity during pregnancy or early postnatal life, are linked to an elevated prevalence of metabolic disease in adult offspring. The mechanisms underlying these effects are still poorly understood, but recent data from rodents provide insight into a potential role for the brain in this 'metabolic programming.' This review summarizes the developmental changes that have been observed in the hypothalamus in response to changes in the early nutritional and hormonal environment. It also discusses how resetting a diverse array of neuroendocrine systems may have long-term effects on the regulation of metabolism and energy balance.


Assuntos
Desenvolvimento Infantil , Hipotálamo/fisiologia , Fenômenos Fisiológicos da Nutrição do Lactente , Leptina/fisiologia , Fenômenos Fisiológicos da Nutrição Materna , Doenças Metabólicas/etiologia , Animais , Metabolismo Energético , Feminino , Humanos , Hipotálamo/crescimento & desenvolvimento , Lactente , Sistemas Neurossecretores/crescimento & desenvolvimento , Obesidade/etiologia , Obesidade/fisiopatologia , Gravidez , Complicações na Gravidez , Efeitos Tardios da Exposição Pré-Natal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA