Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Ultrastruct Pathol ; 47(4): 292-303, 2023 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-36992558

RESUMO

Obesity is a serious health issue. As regard, the central nervous system, obesity induces neuronal damage. Vitamin D has well-known anti-inflammatory and neuroprotective effects. To detect if vitamin D protects against damage in the arcuate nucleus induced by a high fat-high fructose diet. Forty adult rats were used, and four groups were formed. Group I (negative control) kept on a standard chow diet for six weeks, Group II (positive control) received vitamin D orally once every other day for six weeks, Group III (high fat-high fructose treated group) was given high fat-high fructose diets for six weeks and Group IV (high fat-high fructose and vitamin D treated group) were given high fat-high fructose diets concomitantly with vitamin D for six weeks. High fat-high fructose diet markedly caused histological changes in arcuate neurons as nuclei appeared darkly stained and shrunken with condensed chromatin, and the nucleolus became less prominent. The cytoplasm appeared rarefied with loss of most of the organelles. An increase in neuroglial cells was noticed. The synaptic area showed sparse degenerated mitochondria and a disrupted presynaptic membrane. A high-fat diet has a damaging effect on arcuate neurons and vitamin D alleviates these effects.


Assuntos
Dieta Hiperlipídica , Vitamina D , Ratos , Animais , Vitamina D/farmacologia , Dieta Hiperlipídica/efeitos adversos , Núcleo Arqueado do Hipotálamo/patologia , Obesidade/etiologia , Obesidade/patologia , Frutose/toxicidade
2.
Proc Natl Acad Sci U S A ; 116(30): 15236-15243, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31285329

RESUMO

Dopamine neurons of the hypothalamic arcuate nucleus (ARC) tonically inhibit the release of the protein hormone prolactin from lactotropic cells in the anterior pituitary gland and thus play a central role in prolactin homeostasis of the body. Prolactin, in turn, orchestrates numerous important biological functions such as maternal behavior, reproduction, and sexual arousal. Here, we identify the canonical transient receptor potential channel Trpc5 as an essential requirement for normal function of dopamine ARC neurons and prolactin homeostasis. By analyzing female mice carrying targeted mutations in the Trpc5 gene including a conditional Trpc5 deletion, we show that Trpc5 is required for maintaining highly stereotyped infraslow membrane potential oscillations of dopamine ARC neurons. Trpc5 is also required for eliciting prolactin-evoked tonic plateau potentials in these neurons that are part of a regulatory feedback circuit. Trpc5 mutant females show severe prolactin deficiency or hypoprolactinemia that is associated with irregular reproductive cyclicity, gonadotropin imbalance, and impaired reproductive capabilities. These results reveal a previously unknown role for the cation channel Trpc5 in prolactin homeostasis of female mice and provide strategies to explore the genetic basis of reproductive disorders and other malfunctions associated with defective prolactin regulation in humans.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Neurônios Dopaminérgicos/metabolismo , Doenças Genéticas Inatas/genética , Transtornos da Lactação/genética , Prolactina/deficiência , Prolactina/genética , Canais de Cátion TRPC/genética , Animais , Núcleo Arqueado do Hipotálamo/patologia , Nível de Alerta/fisiologia , Neurônios Dopaminérgicos/patologia , Retroalimentação Fisiológica , Feminino , Regulação da Expressão Gênica , Doenças Genéticas Inatas/metabolismo , Doenças Genéticas Inatas/patologia , Gonadotropinas/sangue , Gonadotropinas/genética , Homeostase/genética , Humanos , Transtornos da Lactação/metabolismo , Transtornos da Lactação/patologia , Potenciais da Membrana/fisiologia , Camundongos , Mutação , Prolactina/sangue , Prolactina/metabolismo , Reprodução/fisiologia , Transdução de Sinais , Canais de Cátion TRPC/deficiência
3.
Am J Physiol Endocrinol Metab ; 320(3): E467-E474, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33356996

RESUMO

The arcuate nucleus (ARC) of the hypothalamus comprises two antagonistic neuron populations critical for energy balance, namely, the anorexigenic pro-opiomelanocortin (POMC) and the orexigenic agouti-related peptide (AgRP) neurons that act as agonists and antagonists, respectively, for neurons expressing the type IV melanocortin receptor (MC4R) (Andermann ML and Lowell BB. Neuron 95: 757-778, 2017). MC4R activation increases energy expenditure and decreases food intake during positive energy balance states to prevent diet-induced obesity (DIO). Work from our group identified aberrant neuronal cell cycle events both as a novel biomarker and druggable target in the ARC for the treatment of DIO, demonstrating pharmacological restoration of retinoblastoma protein function in the ARC using cyclin-dependent kinase 4/6 (CDK4/6) inhibitors could treat DIO in mice by increasing lipid oxidation to selectively decrease fat mass. However, the role of CDK4/6 inhibitors on food intake was not examined. Four-week-old Mc4r-loxTB mice were continuously administered high-fat diet (60% kcal fat). At 8 wk of age, animals were administered 60 mg/kg abemaciclib orally or a saline control and monitored every 2 wk for fat mass changes by MRI. At 11 wk of age, all animals were injected bilaterally in the paraventricular hypothalamus with AAV8 serotype virus expressing a Cre-mCherry and monitored for another 5 wk. Restoration of Mc4r expression in the paraventricular hypothalamic nucleus (PVN/PVH) reduced food intake in hyperphagic obese mice when given CDK4/6 inhibitor therapy. The reduced food intake was responsible for reduced fat mass in mice treated with abemaciclib. These results indicate that targeting POMC neurons could be an effective strategy in treating diet-related obesity.NEW & NOTEWORTHY We have defined some of the necessary components to prevent high-fat diet-induced obesity at the molecular and cellular level. Within POMC neurons, the retinoblastoma protein must remain active and prevented from phosphoinactivation by cyclin-dependent kinases. The downstream neurons within the PVH must also properly express MC4R for the circuit to appropriately regulate feeding behavior.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Melanocortinas/metabolismo , Rede Nervosa/efeitos dos fármacos , Obesidade/tratamento farmacológico , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Dieta Hiperlipídica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Camundongos Transgênicos , Rede Nervosa/metabolismo , Rede Nervosa/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Inibidores de Proteínas Quinases/farmacologia , Receptor Tipo 4 de Melanocortina/genética , Transdução de Sinais/efeitos dos fármacos
4.
Mol Psychiatry ; 25(5): 1006-1021, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31485012

RESUMO

Chronic stress causes dysregulations of mood and energy homeostasis, but the neurocircuitry underlying these alterations remain to be fully elucidated. Here we demonstrate that chronic restraint stress in mice results in hyperactivity of pro-opiomelanocortin neurons in the arcuate nucleus of the hypothalamus (POMCARH neurons) associated with decreased neural activities of dopamine neurons in the ventral tegmental area (DAVTA neurons). We further revealed that POMCARH neurons project to the VTA and provide an inhibitory tone to DAVTA neurons via both direct and indirect neurotransmissions. Finally, we show that photoinhibition of the POMCARH→VTA circuit in mice increases body weight and food intake, and reduces depression-like behaviors and anhedonia in mice exposed to chronic restraint stress. Thus, our results identified a novel neurocircuitry regulating feeding and mood in response to stress.


Assuntos
Anedonia , Depressão/metabolismo , Transtornos da Alimentação e da Ingestão de Alimentos/etiologia , Transtornos da Alimentação e da Ingestão de Alimentos/metabolismo , Vias Neurais , Pró-Opiomelanocortina/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Transtornos da Alimentação e da Ingestão de Alimentos/psicologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/patologia
5.
Exp Cell Res ; 389(1): 111848, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31954693

RESUMO

Loss of neuron homeostasis in the arcuate nucleus (ARC) is responsible for diet-induced-obesity (DIO). We previously reported that loss of Rb1 gene compromised the homeostasis of anorexigenic POMC neurons in ARC and induced obesity in mice. To evaluate the development of DIO, we propose to analyze the transcriptomic alteration of POMC neurons in mice following high fat diet (HFD) feeding. We isolated these neurons from established DIO mice and performed transcriptomic profiling using RNA-seq. In total, 1066 genes (628 upregulated and 438 downregulated) were identified as differentially expressed genes (DEGs). Pathway enrichment analysis with these DEGs further revealed that "cell cycle," "apoptosis," "chemokine signaling," and "sphingolipid metabolism" pathways were correlated with DIO development. Moreover, we validated that the pRb protein, a key regulator of "cell cycle pathway," was inactivated by phosphorylation in POMC neurons by HFD feeding. Importantly, the reversal of deregulated cell cycle by stereotaxic delivering of the unphosphorylated pRbΔP in ARC significantly meliorated the DIO. Collectively, our study provides insights into the mechanisms related to the loss of homeostasis of POMC neurons in DIO, and suggests pRb phosphorylation as a potential intervention target to treat DIO.


Assuntos
Dieta Hiperlipídica , Neurônios/metabolismo , Obesidade/genética , Pró-Opiomelanocortina/metabolismo , Transcriptoma , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Neurônios/patologia , Obesidade/etiologia , Obesidade/patologia , Pró-Opiomelanocortina/genética
6.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34360816

RESUMO

Hypothalamic dysfunction is an initial event following diet-induced obesity, primarily involving areas regulating energy balance such as arcuate nucleus (Arc) and median eminence (ME). To gain insights into the early hypothalamic diet-induced alterations, adult CD1 mice fed a high-fat diet (HFD) for 6 weeks were studied and compared with normo-fed controls. Transmission and scanning electron microscopy and histological staining were employed for morphological studies of the ME, while Raman spectroscopy was applied for the biochemical analysis of the Arc-ME complex. In HFD mice, ME ß2-tanycytes, glial cells dedicated to blood-liquor crosstalk, exhibited remarkable ultrastructural anomalies, including altered alignment, reduced junctions, degenerating organelles, and higher content of lipid droplets, lysosomes, and autophagosomes. Degenerating tanycytes also displayed an electron transparent cytoplasm filled with numerous vesicles, and they were surrounded by dilated extracellular spaces extending up to the subependymal layer. Consistently, Raman spectroscopy analysis of the Arc-ME complex revealed higher glycogen, collagen, and lipid bands in HFD mice compared with controls, and there was also a higher band corresponding to the cyanide group in the former compared to the last. Collectively, these data show that ME ß2-tanycytes exhibit early structural and chemical alterations due to HFD and reveal for the first-time hypothalamic cyanide presence following high dietary lipids consumption, which is a novel aspect with potential implications in the field of obesity.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Eminência Mediana/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/patologia , Metabolismo Energético , Masculino , Eminência Mediana/patologia , Camundongos , Obesidade/patologia
7.
Horm Behav ; 120: 104675, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31923417

RESUMO

Consumption of a high fat diet (HFD) increases circulating free fatty acids, which can enter the brain and promote a state of microgliosis, as defined by a change in microglia number and/or morphology. Most studies investigating diet-induced microgliosis have been conducted in male rodents despite well-documented sex differences in the neural control of food intake and neuroimmune signaling. This highlights the need to investigate how sex hormones may modulate the behavioral and cellular response to HFD consumption. Estradiol is of particular interest since it exerts a potent anorexigenic effect and has both anti-inflammatory and neuroprotective effects in the brain. As such, the aim of the current study was to investigate whether estradiol attenuates the development of HFD-induced microgliosis in female rats. Estradiol- and vehicle-treated ovariectomized rats were fed either a low-fat chow diet or a 60% HFD for 4 days, after which they were perfused and brain sections were processed via immunohistochemistry for microglia-specific Iba1 protein. Four days of HFD consumption promoted microgliosis, as measured via an increase in the number of microglia in the arcuate nucleus (ARC) of the hypothalamus and nucleus of the solitary tract (NTS), and a decrease in microglial branching in the ARC, NTS, lateral hypothalamus (LH), and ventromedial hypothalamus. Estradiol replacement attenuated the HFD-induced changes in microglia accumulation and morphology in the ARC, LH, and NTS. We conclude that estradiol has protective effects against HFD-induced microgliosis in a region-specific manner in hypothalamic and hindbrain areas implicated in the neural control of food intake.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Estradiol/farmacologia , Gliose/prevenção & controle , Microglia/efeitos dos fármacos , Ovariectomia/efeitos adversos , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/patologia , Encefalopatias/etiologia , Encefalopatias/patologia , Encefalopatias/prevenção & controle , Contagem de Células , Tamanho Celular/efeitos dos fármacos , Gorduras na Dieta/efeitos adversos , Estradiol/deficiência , Feminino , Gliose/etiologia , Gliose/patologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Microglia/patologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/patologia
8.
Neuroendocrinology ; 110(6): 552-562, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31484186

RESUMO

The hypothalamus contains integrative systems that support life, including physiological processes such as food intake, energy expenditure, and reproduction. Here, we show that anorexia nervosa (AN) patients, contrary to normal weight and constitutionally lean individuals, respond with a paradoxical reduction in hypothalamic levels of glutamate/glutamine (Glx) upon feeding. This reversal of the Glx response is associated with decreased wiring in the arcuate nucleus and increased connectivity in the lateral hypothalamic area, which are involved in the regulation on a variety of physiological and behavioral functions including the control of food intake and energy balance. The identification of distinct hypothalamic neurochemical dysfunctions and associated structural variations in AN paves the way for the development of new diagnostic and treatment strategies in conditions associated with abnormal body mass index and a maladaptive response to negative energy balance.


Assuntos
Anorexia Nervosa , Núcleo Arqueado do Hipotálamo , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Região Hipotalâmica Lateral , Adulto , Anorexia Nervosa/diagnóstico por imagem , Anorexia Nervosa/metabolismo , Anorexia Nervosa/patologia , Anorexia Nervosa/fisiopatologia , Núcleo Arqueado do Hipotálamo/diagnóstico por imagem , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Núcleo Arqueado do Hipotálamo/fisiopatologia , Feminino , Humanos , Região Hipotalâmica Lateral/diagnóstico por imagem , Região Hipotalâmica Lateral/metabolismo , Região Hipotalâmica Lateral/patologia , Região Hipotalâmica Lateral/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Espectroscopia de Prótons por Ressonância Magnética , Adulto Jovem
9.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33255553

RESUMO

Obesity-associated low-grade inflammation favors weight gain, whereas systemic infection frequently leads to anorexia. Thus, inflammatory signals can either induce positive or negative energy balance. In this study, we used whole-cell patch-clamp to investigate the acute effects of three important proinflammatory cytokines, tumor necrosis factor α (TNF-α), interleukin-6, and interleukin-1ß (IL-1ß) on the membrane excitability of agouti-related peptide (AgRP)- or proopiomelanocortin (POMC)-producing neurons. We found that both TNF-α and IL-1ß acutely inhibited the activity of 35-42% of AgRP-producing neurons, whereas very few POMC neurons were depolarized by TNF-α. Interleukin-6 induced no acute changes in the activity of AgRP or POMC neurons. Our findings indicate that the effect of TNF-α and IL-1ß, especially on the activity of AgRP-producing neurons, may contribute to inflammation-induced anorexia observed during acute inflammatory conditions.


Assuntos
Proteína Relacionada com Agouti/genética , Inflamação/genética , Interleucina-1beta/genética , Obesidade/genética , Fator de Necrose Tumoral alfa/genética , Animais , Anorexia/genética , Anorexia/metabolismo , Anorexia/patologia , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Metabolismo Energético , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-6/genética , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Neuropeptídeo Y/genética , Obesidade/metabolismo , Obesidade/patologia , Técnicas de Patch-Clamp , Pró-Opiomelanocortina/genética
10.
J Neurochem ; 149(1): 73-97, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30615192

RESUMO

The hypothalamus is essential for regulation of energy homeostasis and metabolism. Feeding hypercaloric, high-fat (HF) diet induces hypothalamic arcuate nucleus injury and alters metabolism more severely in male than in female mice. The site(s) and extent of hypothalamic injury in male and female mice are not completely understood. In the paraventricular nucleus (PVN) of the hypothalamus, single-minded family basic helix-loop helix transcription factor 1 (Sim1) neurons are essential to control energy homeostasis. We tested the hypothesis that exposure to HF diet induces injury to Sim1 neurons in the PVN of male and female mice. Mice expressing membrane-bound enhanced green fluorescent protein (mEGFP) in Sim1 neurons (Sim1-Cre:Rosa-mEGFP mice) were generated to visualize the effects of exposure to HF diet on these neurons. Male and female Sim1-Cre:Rosa-mEGFP mice exposed to HF diet had increased weight, hyperleptinemia, and developed hepatosteatosis. In male and female mice exposed to HF diet, expression of mEGFP was reduced by > 40% in Sim1 neurons of the PVN, an effect paralleled by cell apoptosis and neuronal loss, but not by microgliosis. In the arcuate nucleus of the Sim1-Cre:Rosa-mEGFP male mice, there was decreased alpha-melanocyte-stimulating hormone in proopiomelanocortin neurons projecting to the PVN, with increased cell apoptosis, neuronal loss, and microgliosis. These defects were undetectable in the arcuate nucleus of female mice exposed to the HF diet. Thus, injury to Sim1 neurons of the PVN is a shared feature of exposure to HF diet in mice of both sexes, while injury to proopiomelanocortin neurons in arcuate nucleus is specific to male mice. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Neurônios/patologia , Núcleo Hipotalâmico Paraventricular/patologia , Proteínas Repressoras/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/patologia , Feminino , Masculino , Camundongos , Neurônios/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Pró-Opiomelanocortina/metabolismo
11.
BMC Neurosci ; 20(1): 33, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31291887

RESUMO

BACKGROUND: Hypothalamic inflammation including astrogliosis and microglia activation occurs after intake of high fat diet (HFD) in rodent models or in obese individuals. However, the effect of chronic HFD feeding on oligodendrocytes (OLGs), a myelin-producing glial population in the central nervous system (CNS), remains unclear. In this study, we used 8-week old male C57BL/6 mice fed by HFD for 3-6 months to induce chronic obesity. RESULTS: The transmission electron microscopy imaging analysis showed that the integrity of hypothalamic myelin was disrupted after HFD feeding for 4 and 6 months. Moreover, the accumulation of Iba1+-microglia with an amoeboid hypertrophic form was continually observed in arcuate nucleus of HFD-fed mice during the entire feeding time period. Interleukin-33 (IL-33), a tissue alarmin upon injury to the CNS, was detected with an increased level in hypothalamus after HFD feeding for 3 and 4 months. Furthermore, the in vitro study indicated that exposure of mature OLGs to IL-33 impaired OLG cell structure along with a decline in the expression of myelin basic protein. CONCLUSIONS: Altogether, our findings demonstrate that chronic HFD feeding triggers hypothalamic myelin disruption in accompany with IL-33 upregulation and prolonged microglial activation in hypothalamus. Given that the addition of exogenous IL-33 was harmful for the maturation of OLGs, an increase in IL-33 by chronic HFD feeding might contribute to the induction of hypothalamic myelin disruption.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hipotálamo/metabolismo , Interleucina-33/metabolismo , Bainha de Mielina/patologia , Regulação para Cima , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Hipotálamo/patologia , Masculino , Camundongos , Proteína Básica da Mielina/biossíntese , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Cultura Primária de Células , Ratos , Fatores de Tempo
12.
Proc Natl Acad Sci U S A ; 112(2): 596-601, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25550522

RESUMO

Polycystic ovarian syndrome (PCOS), the leading cause of female infertility, is associated with an increase in luteinizing hormone (LH) pulse frequency, implicating abnormal steroid hormone feedback to gonadotropin-releasing hormone (GnRH) neurons. This study investigated whether modifications in the synaptically connected neuronal network of GnRH neurons could account for this pathology. The PCOS phenotype was induced in mice following prenatal androgen (PNA) exposure. Serial blood sampling confirmed that PNA elicits increased LH pulse frequency and impaired progesterone negative feedback in adult females, mimicking the neuroendocrine abnormalities of the clinical syndrome. Imaging of GnRH neurons revealed greater dendritic spine density that correlated with increased putative GABAergic but not glutamatergic inputs in PNA mice. Mapping of steroid hormone receptor expression revealed that PNA mice had 59% fewer progesterone receptor-expressing cells in the arcuate nucleus of the hypothalamus (ARN). To address whether increased GABA innervation to GnRH neurons originates in the ARN, a viral-mediated Cre-lox approach was taken to trace the projections of ARN GABA neurons in vivo. Remarkably, projections from ARN GABAergic neurons heavily contacted and even bundled with GnRH neuron dendrites, and the density of fibers apposing GnRH neurons was even greater in PNA mice (56%). Additionally, this ARN GABA population showed significantly less colocalization with progesterone receptor in PNA animals compared with controls. Together, these data describe a robust GABAergic circuit originating in the ARN that is enhanced in a model of PCOS and may underpin the neuroendocrine pathophysiology of the syndrome.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiopatologia , Neurônios GABAérgicos/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Síndrome do Ovário Policístico/fisiopatologia , Androgênios/administração & dosagem , Animais , Núcleo Arqueado do Hipotálamo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Hormônio Luteinizante/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sistemas Neurossecretores/fisiopatologia , Síndrome do Ovário Policístico/etiologia , Síndrome do Ovário Policístico/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Receptores de Progesterona/fisiologia
13.
Neurobiol Dis ; 101: 16-26, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28108291

RESUMO

Migraine is a disabling brain disorder involving abnormal trigeminovascular activation and sensitization. Fasting or skipping meals is considered a migraine trigger and altered fasting glucose and insulin levels have been observed in migraineurs. Therefore peptides involved in appetite and glucose regulation including insulin, glucagon and leptin could potentially influence migraine neurobiology. We aimed to determine the effect of insulin (10U·kg-1), glucagon (100µg·200µl-1) and leptin (0.3, 1 and 3mg·kg-1) signaling on trigeminovascular nociceptive processing at the level of the trigeminocervical-complex and hypothalamus. Male rats were anesthetized and prepared for craniovascular stimulation. In vivo electrophysiology was used to determine changes in trigeminocervical neuronal responses to dural electrical stimulation, and phosphorylated extracellular signal-regulated kinases 1 and 2 (pERK1/2) immunohistochemistry to determine trigeminocervical and hypothalamic neural activity; both in response to intravenous administration of insulin, glucagon, leptin or vehicle control in combination with blood glucose analysis. Blood glucose levels were significantly decreased by insulin (p<0.001) and leptin (p<0.01) whereas glucagon had the opposite effect (p<0.001). Dural-evoked neuronal firing in the trigeminocervical-complex was significantly inhibited by insulin (p<0.001), glucagon (p<0.05) and leptin (p<0.01). Trigeminocervical-complex pERK1/2 cell expression was significantly decreased by insulin and leptin (both p<0.001), and increased by glucagon (p<0.001), when compared to vehicle control. However, only leptin affected pERK1/2 expression in the hypothalamus, significantly decreasing pERK1/2 immunoreactive cell expression in the arcuate nucleus (p<0.05). These findings demonstrate that insulin, glucagon and leptin can alter the transmission of trigeminal nociceptive inputs. A potential neurobiological link between migraine and impaired metabolic homeostasis may occur through disturbed glucose regulation and a transient hypothalamic dysfunction.


Assuntos
Glucagon/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Transtornos de Enxaqueca/metabolismo , Neurônios/metabolismo , Núcleos do Trigêmeo/metabolismo , Analgésicos não Narcóticos/administração & dosagem , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Glucagon/administração & dosagem , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hipotálamo/patologia , Insulina/administração & dosagem , Leptina/administração & dosagem , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Transtornos de Enxaqueca/patologia , Transtornos de Enxaqueca/prevenção & controle , Vias Neurais/metabolismo , Vias Neurais/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Dor/metabolismo , Dor/patologia , Dor/prevenção & controle , Ratos Sprague-Dawley , Núcleos do Trigêmeo/patologia
14.
Hum Mol Genet ; 24(15): 4276-83, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25926624

RESUMO

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


Assuntos
Antígenos de Neoplasias/genética , Leptina/metabolismo , Neurônios/metabolismo , Síndrome de Prader-Willi/genética , Proteínas/genética , Animais , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Modelos Animais de Doenças , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Hipotálamo/patologia , Leptina/administração & dosagem , Camundongos , Neurônios/patologia , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Pró-Opiomelanocortina/metabolismo , Aumento de Peso/genética
15.
Int J Obes (Lond) ; 41(1): 149-158, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27773938

RESUMO

BACKGROUND: Consumption of dietary fat is one of the key factors leading to obesity. High-fat diet (HFD)-induced obesity is characterized by induction of inflammation in the hypothalamus; however, the temporal regulation of proinflammatory markers and their impact on hypothalamic appetite-regulating neuropeptide Y/agouti-related peptide (NPY/AgRP) neurons remains undefined. METHODS: Mice were injected with an acute lipid infusion for 24 h or fed a HFD over 8-20 weeks. Characterized mouse NPY/AgRP hypothalamic cell lines were used for in vitro experimentation. Immunohistochemistry in brain slices or quantitative real-time PCR in cell lines, was performed to determine changes in the expression of key inflammatory markers and neuropeptides. RESULTS: Hypothalamic inflammation, indicated by tumor necrosis factor (TNF)-α expression and astrocytosis in the arcuate nucleus, was evident following acute lipid infusion. HFD for 8 weeks suppressed TNF-α, while significantly increasing heat-shock protein 70 and ciliary neurotrophic factor, both neuroprotective components. HFD for 20 weeks induced TNF-α expression in NPY/AgRP neurons, suggesting a detrimental temporal regulatory mechanism. Using NPY/AgRP hypothalamic cell lines, we found that palmitate provoked a mixed inflammatory response on a panel of inflammatory and endoplasmic reticulum (ER) stress genes, whereas TNF-α significantly upregulated IκBα, nuclear factor (NF)-κB and interleukin-6 mRNA levels. Palmitate and TNF-α exposure predominantly induced NPY mRNA levels. Utilizing an I kappa B kinase ß (IKKß) inhibitor, we demonstrated that these effects potentially occur via the inflammatory IKKß/NF-κB pathway. CONCLUSIONS: These findings indicate that acute lipid and chronic HFD feeding in vivo, as well as acute palmitate and TNF-α exposure in vitro, induce markers of inflammation or ER stress in the hypothalamic appetite-stimulating NPY/AgRP neurons over time, which may contribute to a dramatic alteration in NPY/AgRP content or expression. Acute and chronic HFD feeding in vivo temporally regulates arcuate TNF-α expression with reactive astrocytosis, which suggests a time-dependent neurotrophic or neurotoxic role of lipids.


Assuntos
Apetite/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Hipotálamo/patologia , Inflamação/induzido quimicamente , Neurônios/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Palmitatos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hipotálamo/efeitos dos fármacos , Inflamação/patologia , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Obesidade/patologia
16.
Diabetes Obes Metab ; 19(12): 1740-1750, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28544245

RESUMO

AIMS: Ghrelin is implicated in the control of energy balance and glucose homeostasis. The ghrelin receptor exhibits ligand-independent constitutive activity, which can be pharmacologically exploited to induce inverse ghrelin actions. Because ghrelin receptor inverse agonists (GHSR-IA) might be effective for the treatment of obesity-related metabolic disease, we tested 2 novel synthetic compounds GHSR-IA1 and GHSR-IA2. MATERIALS AND METHODS: In functional cell assays, electrophysiogical and immunohistochemical experiments, we demonstrated inverse agonist activity for GHSR-IA1 and GHSR-IA2. We used healthy mice, Zucker diabetic fatty (ZDF) rats and diet-induced obese (DIO) mice to explore effects on food intake (FI), body weight (BW), conditioned taste aversion (CTA), oral glucose tolerance (OGT), pancreatic islet morphology, hepatic steatosis (HS), and blood lipids. RESULTS: Both compounds acutely reduced FI in mice without inducing CTA. Chronic GHSR-IA1 increased metabolic rate in chow-fed mice, suppressed FI, and improved OGT in ZDF rats. Moreover, the progression of islet hyperplasia to fibrosis in ZDF rats slowed down. GHSR-IA2 reduced FI and BW in DIO mice, and reduced fasting and stimulated glucose levels compared with pair-fed and vehicle-treated mice. GHSR-IA2-treated DIO mice showed decreased blood lipids. GHSR-IA1 treatment markedly decreased HS in DIO mice. CONCLUSIONS: Our study demonstrates therapeutic actions of novel ghrelin receptor inverse agonists, suggesting a potential to treat obesity-related metabolic disorders including diabetes mellitus.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hiperlipidemias/prevenção & controle , Hipoglicemiantes/uso terapêutico , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Obesidade/tratamento farmacológico , Receptores de Grelina/agonistas , Animais , Fármacos Antiobesidade/efeitos adversos , Fármacos Antiobesidade/farmacologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Agonismo Inverso de Drogas , Ingestão de Energia/efeitos dos fármacos , Células HEK293 , Humanos , Hiperlipidemias/etiologia , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Hepatopatia Gordurosa não Alcoólica/etiologia , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Distribuição Aleatória , Ratos , Ratos Zucker , Receptores de Grelina/antagonistas & inibidores , Receptores de Grelina/metabolismo , Aumento de Peso/efeitos dos fármacos
17.
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
18.
PLoS Genet ; 9(1): e1003207, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23341784

RESUMO

Prader-Willi Syndrome is the most common syndromic form of human obesity and is caused by the loss of function of several genes, including MAGEL2. Mice lacking Magel2 display increased weight gain with excess adiposity and other defects suggestive of hypothalamic deficiency. We demonstrate Magel2-null mice are insensitive to the anorexic effect of peripherally administered leptin. Although their excessive adiposity and hyperleptinemia likely contribute to this physiological leptin resistance, we hypothesized that Magel2 may also have an essential role in intracellular leptin responses in hypothalamic neurons. We therefore measured neuronal activation by immunohistochemistry on brain sections from leptin-injected mice and found a reduced number of arcuate nucleus neurons activated after leptin injection in the Magel2-null animals, suggesting that most but not all leptin receptor-expressing neurons retain leptin sensitivity despite hyperleptinemia. Electrophysiological measurements of arcuate nucleus neurons expressing the leptin receptor demonstrated that although neurons exhibiting hyperpolarizing responses to leptin are present in normal numbers, there were no neurons exhibiting depolarizing responses to leptin in the mutant mice. Additional studies demonstrate that arcuate nucleus pro-opiomelanocortin (POMC) expressing neurons are unresponsive to leptin. Interestingly, Magel2-null mice are hypersensitive to the anorexigenic effects of the melanocortin receptor agonist MT-II. In Prader-Willi Syndrome, loss of MAGEL2 may likewise abolish leptin responses in POMC hypothalamic neurons. This neural defect, together with increased fat mass, blunted circadian rhythm, and growth hormone response pathway defects that are also linked to loss of MAGEL2, could contribute to the hyperphagia and obesity that are hallmarks of this disorder.


Assuntos
Antígenos de Neoplasias , Leptina , Neurônios , Síndrome de Prader-Willi , Pró-Opiomelanocortina , Proteínas , Adiposidade/genética , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Ritmo Circadiano/genética , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Humanos , Hiperfagia/genética , Hiperfagia/metabolismo , Leptina/administração & dosagem , Leptina/metabolismo , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Obesidade/genética , Obesidade/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Proteínas/genética , Proteínas/metabolismo , Receptores para Leptina/metabolismo , Aumento de Peso/efeitos dos fármacos
19.
J Biol Chem ; 288(24): 17675-88, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23640886

RESUMO

It was shown previously that abnormal prohormone processing or inactive proconverting enzymes that are responsible for this processing cause profound obesity. Our laboratory demonstrated earlier that in the diet-induced obesity (DIO) state, the appetite-suppressing neuropeptide α-melanocyte-stimulating hormone (α-MSH) is reduced, yet the mRNA of its precursor protein proopiomelanocortin (POMC) remained unaltered. It was also shown that the DIO condition promotes the development of endoplasmic reticulum (ER) stress and leptin resistance. In the current study, using an in vivo model combined with in vitro experiments, we demonstrate that obesity-induced ER stress obstructs the post-translational processing of POMC by decreasing proconverting enzyme 2, which catalyzes the conversion of adrenocorticotropin to α-MSH, thereby decreasing α-MSH peptide production. This novel mechanism of ER stress affecting POMC processing in DIO highlights the importance of ER stress in regulating central energy balance in obesity.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Estresse do Retículo Endoplasmático , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Processamento de Proteína Pós-Traducional , Hormônio Adrenocorticotrópico/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/patologia , Linhagem Celular , Dieta Hiperlipídica/efeitos adversos , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica , Leptina/fisiologia , Masculino , Camundongos , Obesidade/etiologia , Obesidade/patologia , Pró-Opiomelanocortina/genética , Pró-Proteína Convertase 2/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , alfa-MSH/metabolismo , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
20.
BMC Neurol ; 14: 46, 2014 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-24607148

RESUMO

BACKGROUND: The role of the arcuate fasciculus (AF) in the dominant hemisphere in stroke patients with aphasia has not been clearly elucidated. We investigated the relation between language function and diffusion tensor tractography (DTT) findings for the left AF in chronic stroke patients with aphasia. METHOD: Twenty five consecutive right-handed stroke patients with aphasia following lesions in the left hemisphere were recruited for this study. The aphasia quotient (AQ) of Korean-Western Aphasia Battery was used for assessment of language function. We measured values of fractional anisotropy (FA), apparent diffusion coefficient (ADC), voxel number of the left AF. We classified patients into three groups: type A--the left AF was not reconstructed, type B--the left AF was discontinued between Wernicke's and Broca's areas, and type C--the left AF was preserved around the stroke lesion. RESULTS: Moderate positive correlation was observed between AQ and voxel number of the left AF (r = 0.471, p < 0.05). However, no correlation was observed between AQ and FA (r = 0.275, p > 0.05) and ADC values (r = -0.286, p > 0.05). Significant differences in AQ scores were observed between the three types (p < 0.05); the AQ score of type C was higher than those of type A and B, and that of type B was also higher than that of type A (p < 0.05). CONCLUSION: According to our findings, the remaining volume of the left AF, irrespective of directionality and diffusivity, showed moderate positive correlation with language function in chronic stroke patients with aphasia. Discontinuation or non-construction of the left AF was also an important factor for language function.


Assuntos
Afasia/diagnóstico , Afasia/epidemiologia , Núcleo Arqueado do Hipotálamo/patologia , Via Perfurante/patologia , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/epidemiologia , Adulto , Idoso , Afasia/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Doença Crônica , Imagem de Tensor de Difusão/métodos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Via Perfurante/metabolismo , Acidente Vascular Cerebral/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA