Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuron ; 102(3): 653-667.e6, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-30879785

RESUMO

SIM1-expressing paraventricular hypothalamus (PVH) neurons are key regulators of energy balance. Within the PVHSIM1 population, melanocortin-4 receptor-expressing (PVHMC4R) neurons are known to regulate satiety and bodyweight, yet they account for only half of PVHSIM1 neuron-mediated regulation. Here we report that PVH prodynorphin-expressing (PVHPDYN) neurons, which notably lack MC4Rs, function independently and additively with PVHMC4R neurons to account for the totality of PVHSIM1 neuron-mediated satiety. Moreover, PVHPDYN neurons are necessary for prevention of obesity in an independent but equipotent manner to PVHMC4R neurons. While PVHPDYN and PVHMC4R neurons both project to the parabrachial complex (PB), they synaptically engage distinct efferent nodes, the pre-locus coeruleus (pLC), and central lateral parabrachial nucleus (cLPBN), respectively. PB-projecting PVHPDYN neurons, like PVHMC4R neurons, receive input from interoceptive ARCAgRP neurons, respond to caloric state, and are sufficient and necessary to control food intake. This expands the CNS satiety circuitry to include two non-overlapping PVH to hindbrain circuits.


Assuntos
Comportamento Alimentar/fisiologia , Neurônios/citologia , Obesidade/fisiopatologia , Núcleo Hipotalâmico Paraventricular/citologia , Resposta de Saciedade/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Metabolismo Energético , Encefalinas/metabolismo , Locus Cerúleo/citologia , Locus Cerúleo/metabolismo , Locus Cerúleo/fisiologia , Camundongos , Neurônios/metabolismo , Neurônios/fisiologia , Núcleos Parabraquiais/citologia , Núcleos Parabraquiais/metabolismo , Núcleos Parabraquiais/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Precursores de Proteínas/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Proteínas Repressoras/metabolismo
2.
Proc Natl Acad Sci U S A ; 113(43): 12298-12303, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27791019

RESUMO

Melanocortin-4 receptor (Mc4r)-expressing neurons in the autonomic nervous system, particularly in the paraventricular nucleus of the hypothalamus (PVH), play an essential role in blood pressure (BP) control. Mc4r-deficient (Mc4rKO) mice are severely obese but lack obesity-related hypertension; they also show a reduced pressor response to salt loading. We have previously reported that lean juvenile offspring born to diet-induced obese rats (OffOb) exhibit sympathetic-mediated hypertension, and we proposed a role for postnatally raised leptin in its etiology. Here, we test the hypothesis that neonatal hyperleptinemia due to maternal obesity induces persistent changes in the central melanocortin system, thereby contributing to offspring hypertension. Working on the OffOb paradigm in both sexes and using transgenic technology to restore Mc4r in the PVH of Mc4rKO (Mc4rPVH) mice, we have now shown that these mice develop higher BP than Mc4rKO or WT mice. We have also found that experimental hyperleptinemia induced in the neonatal period in Mc4rPVH and WT mice, but not in the Mc4rKO mice, leads to heightened BP and severe renal dysfunction. Thus, Mc4r in the PVH appears to be required for early-life programming of hypertension arising from either maternal obesity or neonatal hyperleptinemia. Early-life exposure of the PVH to maternal obesity through postnatal elevation of leptin may have long-term consequences for cardiovascular health.


Assuntos
Hipertensão/genética , Leptina/metabolismo , Obesidade/genética , Efeitos Tardios da Exposição Pré-Natal/genética , Receptor Tipo 4 de Melanocortina/genética , Animais , Pressão Sanguínea/genética , Dieta/efeitos adversos , Modelos Animais de Doenças , Feminino , Humanos , Hipertensão/complicações , Hipertensão/fisiopatologia , Leptina/genética , Masculino , Relações Materno-Fetais/fisiologia , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Obesidade/complicações , Obesidade/fisiopatologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/patologia
3.
PLoS One ; 11(4): e0153187, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27077912

RESUMO

Opioids are widely used medicinally as analgesics and abused for hedonic effects, actions that are each complicated by substantial risks such as cardiorespiratory depression. These drugs mimic peptides such as ß-endorphin, which has a key role in endogenous analgesia. The ß-endorphin in the central nervous system originates from pro-opiomelanocortin (POMC) neurons in the arcuate nucleus and nucleus of the solitary tract (NTS). Relatively little is known about the NTSPOMC neurons but their position within the sensory nucleus of the vagus led us to test the hypothesis that they play a role in modulation of cardiorespiratory and nociceptive control. The NTSPOMC neurons were targeted using viral vectors in a POMC-Cre mouse line to express either opto-genetic (channelrhodopsin-2) or chemo-genetic (Pharmacologically Selective Actuator Modules). Opto-genetic activation of the NTSPOMC neurons in the working heart brainstem preparation (n = 21) evoked a reliable, titratable and time-locked respiratory inhibition (120% increase in inter-breath interval) with a bradycardia (125±26 beats per minute) and augmented respiratory sinus arrhythmia (58% increase). Chemo-genetic activation of NTSPOMC neurons in vivo was anti-nociceptive in the tail flick assay (latency increased by 126±65%, p<0.001; n = 8). All effects of NTSPOMC activation were blocked by systemic naloxone (opioid antagonist) but not by SHU9119 (melanocortin receptor antagonist). The NTSPOMC neurons were found to project to key brainstem structures involved in cardiorespiratory control (nucleus ambiguus and ventral respiratory group) and endogenous analgesia (periaqueductal gray and midline raphe). Thus the NTSPOMC neurons may be capable of tuning behaviour by an opioidergic modulation of nociceptive, respiratory and cardiac control.


Assuntos
Analgesia , Bradicardia/metabolismo , Tronco Encefálico/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Insuficiência Respiratória/metabolismo , Analgésicos Opioides/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Tronco Encefálico/citologia , Tronco Encefálico/efeitos dos fármacos , Channelrhodopsins , Feminino , Masculino , Hormônios Estimuladores de Melanócitos/farmacologia , Camundongos Transgênicos , Microscopia Confocal , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Núcleo Solitário/citologia , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/metabolismo
4.
Mol Cell Neurosci ; 68: 258-71, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26292267

RESUMO

The neuropeptide galanin has diverse roles in the central and peripheral nervous systems, by activating the G protein-coupled receptors Gal1, Gal2 and the less studied Gal3 (GalR1-3 gene products). There is a wealth of data on expression of Gal1-3 at the mRNA level, but not at the protein level due to the lack of specificity of currently available antibodies. Here we report the generation of knock-in mice expressing Gal1 or Gal2 receptor fluorescently tagged at the C-terminus with, respectively, mCherry or hrGFP (humanized Renilla green fluorescent protein). In dorsal root ganglia (DRG) neurons expressing the highest levels of Gal1-mCherry, localization to the somatic cell membrane was detected by live-cell fluorescence and immunohistochemistry, and that fluorescence decreased upon addition of galanin. In spinal cord, abundant Gal1-mCherry immunoreactive processes were detected in the superficial layers of the dorsal horn, and highly expressing intrinsic neurons of the lamina III/IV border showed both somatic cell membrane localization and outward transport of receptor from the cell body, detected as puncta within cell processes. In brain, high levels of Gal1-mCherry immunofluorescence were detected within thalamus, hypothalamus and amygdala, with a high density of nerve endings in the external zone of the median eminence, and regions with lesser immunoreactivity included the dorsal raphe nucleus. Gal2-hrGFP mRNA was detected in DRG, but live-cell fluorescence was at the limits of detection, drawing attention to both the much lower mRNA expression than to Gal1 in mice and the previously unrecognized potential for translational control by upstream open reading frames (uORFs).


Assuntos
Neurônios/fisiologia , Receptor Tipo 1 de Galanina/metabolismo , Receptor Tipo 2 de Galanina/metabolismo , Animais , Encéfalo/metabolismo , Células Cultivadas , Gânglios Espinais/citologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Confocal , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Galanina/genética , Receptor Tipo 2 de Galanina/genética , Medula Espinal/metabolismo , Proteína Vermelha Fluorescente
5.
Obesity (Silver Spring) ; 22(10): 2252-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25044758

RESUMO

OBJECTIVE: Genome-wide association studies (GWAS) of BMI are mostly undertaken under the assumption that "kg/m(2) " is an index of weight fully adjusted for height, but in general this is not true. The aim here was to assess the contribution of common genetic variation to a adjusted version of that phenotype which appropriately accounts for covariation in height in children. METHODS: A GWAS of height-adjusted BMI (BMI[x] = weight/height(x) ), calculated to be uncorrelated with height, in 5809 participants (mean age 9.9 years) from the Avon Longitudinal Study of Parents and Children (ALSPAC) was performed. RESULTS: GWAS based on BMI[x] yielded marked differences in genomewide results profile. SNPs in ADCY3 (adenylate cyclase 3) were associated at genome-wide significance level (rs11676272 (0.28 kg/m(3.1) change per allele G (0.19, 0.38), P = 6 × 10(-9) ). In contrast, they showed marginal evidence of association with conventional BMI [rs11676272 (0.25 kg/m(2) (0.15, 0.35), P = 6 × 10(-7) )]. Results were replicated in an independent sample, the Generation R study. CONCLUSIONS: Analysis of BMI[x] showed differences to that of conventional BMI. The association signal at ADCY3 appeared to be driven by a missense variant and it was strongly correlated with expression of this gene. Our work highlights the importance of well understood phenotype use (and the danger of convention) in characterising genetic contributions to complex traits.


Assuntos
Adenilil Ciclases/genética , Estatura , Índice de Massa Corporal , Peso Corporal/genética , Polimorfismo de Nucleotídeo Único , Adolescente , Alelos , Estatura/genética , Criança , Pré-Escolar , Feminino , Estudo de Associação Genômica Ampla , Humanos , Lactente , Recém-Nascido , Estudos Longitudinais , Masculino , Fenótipo
6.
Endocrinology ; 155(6): 2144-54, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24684305

RESUMO

A wealth of animal and human studies demonstrate that early life environment significantly influences adult metabolic balance, however the etiology for offspring metabolic misprogramming remains incompletely understood. Here, we determine the effect of maternal diet per se on offspring sex-specific outcomes in metabolic health and hypothalamic transcriptome regulation in mice. Furthermore, to define developmental periods of maternal diet misprogramming aspects of offspring metabolic balance, we investigated offspring physiological and transcriptomic consequences of maternal high-fat/high-sugar diet feeding during pregnancy and/or lactation. We demonstrate that female offspring of high-fat/high-sugar diet-fed dams are particularly vulnerable to metabolic perturbation with body weight increases due to postnatal processes, whereas in utero effects of the diet ultimately lead to glucose homeostasis dysregulation. Furthermore, glucose- and maternal-diet sensitive gene expression modulation in the paraventricular hypothalamus is strikingly sexually dimorphic. In summary, we uncover female-specific, maternal diet-mediated in utero misprogramming of offspring glucose homeostasis and a striking sexual dimorphism in glucose- and maternal diet-sensitive paraventricular hypothalamus gene expression adjustment. Notably, female offspring metabolic vulnerability to maternal high-fat/high-sugar diet propagates a vicious cycle of obesity and type 2 diabetes in subsequent generations.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Glucose/farmacologia , Hipotálamo/metabolismo , Caracteres Sexuais , Adiposidade/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Hipotálamo/efeitos dos fármacos , Insulina/sangue , Masculino , Camundongos , Edulcorantes/farmacologia
7.
Physiol Rep ; 2(3): e00240, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24760503

RESUMO

Abstract Growth hormone secretagogue receptor (GHS-R) signaling has been associated with growth hormone release, increases in food intake and pleiotropic cardiovascular effects. Recent data demonstrated that acute GHS-R antagonism leads to increases in mean arterial pressure mediated by the sympathetic nervous system in rats; a highly undesirable effect if GHS-R antagonism was to be used as a therapeutic approach to reducing food intake in an already obese, hypertensive patient population. However, our data in conscious, freely moving GHS-R deficient mice demonstrate that chronic absence of GHS-R signaling is protective against obesity-induced hypertension. GHS-R deficiency leads to reduced systolic blood pressure variability (SBPV); in response to acute high-fat diet (HFD)-feeding, increases in the sympathetic control of SBPV are suppressed in GHS-R KO mice. Our data further suggest that GHS-R signaling dampens the immediate HFD-mediated increase in spontaneous baroreflex sensitivity. In diet-induced obesity, absence of GHS-R signaling leads to reductions in obesity-mediated hypertension and tachycardia. Collectively, our findings thus suggest that chronic blockade of GHS-R signaling may not result in adverse cardiovascular effects in obesity.

8.
Genes Cancer ; 4(3-4): 118-24, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24020003

RESUMO

SIRT3 is a NAD(+)-dependent deacetylase that regulates the function of numerous mitochondrial proteins with roles in metabolism, oxidative stress, and cell survival. It is emerging as an instrumental regulator of the mitochondrial adaptive responses to stress, including metabolic reprogramming and enhancing antioxidant defense mechanisms. Here, we discuss the role that SIRT3 plays at both a cellular and physiological level and consider its involvement in disease. Mitochondrial dysfunction is a key contributing factor in many diseases; however, the mechanisms involved are often not well understood, and few targeted therapies exist. If manipulation of SIRT3 proves to be beneficial in disease states, then it could be a promising target for novel therapies.

9.
Cell ; 152(3): 612-9, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23374353

RESUMO

Melanocortin 4 receptors (MC4Rs) in the central nervous system are key regulators of energy and glucose homeostasis. Notably, obese patients with MC4R mutations are hyperinsulinemic and resistant to obesity-induced hypertension. Although these effects are probably dependent upon the activity of the autonomic nervous system, the cellular effects of MC4Rs on parasympathetic and sympathetic neurons remain undefined. Here, we show that MC4R agonists inhibit parasympathetic preganglionic neurons in the brainstem. In contrast, MC4R agonists activate sympathetic preganglionic neurons in the spinal cord. Deletion of MC4Rs in cholinergic neurons resulted in elevated levels of insulin. Furthermore, re-expression of MC4Rs specifically in cholinergic neurons (including sympathetic preganglionic neurons) restores obesity-associated hypertension in MC4R null mice. These findings provide a cellular correlate of the autonomic side effects associated with MC4R agonists and demonstrate a role for MC4Rs expressed in cholinergic neurons in the regulation of insulin levels and in the development of obesity-induced hypertension.


Assuntos
Tronco Encefálico/metabolismo , Insulina/metabolismo , Neurônios/metabolismo , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Pressão Sanguínea , Tronco Encefálico/citologia , Neurônios Colinérgicos/metabolismo , AMP Cíclico/metabolismo , Fenômenos Eletrofisiológicos , Humanos , Canais KATP/metabolismo , Masculino , Camundongos , Obesidade/metabolismo , Obesidade/fisiopatologia , Sistema Nervoso Parassimpático/metabolismo , Receptor Tipo 4 de Melanocortina/genética , Medula Espinal/metabolismo , Sistema Nervoso Simpático/metabolismo
10.
PLoS One ; 7(11): e48225, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23139766

RESUMO

Progressive mitochondrial dysfunction contributes to neuronal degeneration in age-mediated disease. An essential regulator of mitochondrial function is the deacetylase, sirtuin 3 (SIRT3). Here we investigate a role for CNS Sirt3 in mitochondrial responses to reactive oxygen species (ROS)- and Alzheimer's disease (AD)-mediated stress. Pharmacological augmentation of mitochondrial ROS increases Sirt3 expression in primary hippocampal culture with SIRT3 over-expression being neuroprotective. Furthermore, Sirt3 expression mirrors spatiotemporal deposition of ß-amyloid in an AD mouse model and is also upregulated in AD patient temporal neocortex. Thus, our data suggest a role for SIRT3 in mechanisms sensing and tackling ROS- and AD-mediated mitochondrial stress.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Espécies Reativas de Oxigênio/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Transporte de Elétrons , Células HEK293 , Células HeLa , Humanos , Lentivirus , Camundongos , Mitocôndrias/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Sirtuína 3/genética , Sirtuína 3/metabolismo , Frações Subcelulares/metabolismo , Regulação para Cima/genética
11.
PLoS One ; 6(4): e19155, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21544206

RESUMO

The neurotransmitter glutamate is released by excitatory projection neurons throughout the brain. However, non-glutamatergic cells, including cholinergic and monoaminergic neurons, express markers that suggest that they are also capable of vesicular glutamate release. Striatal cholinergic interneurons (CINs) express the Type-3 vesicular glutamate transporter (VGluT3), although whether they form functional glutamatergic synapses is unclear. To examine this possibility, we utilized mice expressing Cre-recombinase under control of the endogenous choline acetyltransferase locus and conditionally expressed light-activated Channelrhodopsin2 in CINs. Optical stimulation evoked action potentials in CINs and produced postsynaptic responses in medium spiny neurons that were blocked by glutamate receptor antagonists. CIN-mediated glutamatergic responses exhibited a large contribution of NMDA-type glutamate receptors, distinguishing them from corticostriatal inputs. CIN-mediated glutamatergic responses were insensitive to antagonists of acetylcholine receptors and were not seen in mice lacking VGluT3. Our results indicate that CINs are capable of mediating fast glutamatergic transmission, suggesting a new role for these cells in regulating striatal activity.


Assuntos
Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Corpo Estriado/metabolismo , Interneurônios/metabolismo , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Channelrhodopsins , Corpo Estriado/efeitos dos fármacos , Potenciais Evocados/efeitos dos fármacos , Feminino , Interneurônios/efeitos dos fármacos , Masculino , Mecamilamina/farmacologia , Camundongos , Picrotoxina/farmacologia , Receptores Colinérgicos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Escopolamina/farmacologia
12.
Cell Metab ; 13(2): 195-204, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21284986

RESUMO

Melanocortin-4 receptor (MC4R) mutations cause dysregulation of energy balance and hyperinsulinemia. We have used mouse models to study the physiological roles of extrahypothalamic MC4Rs. Re-expression of MC4Rs in cholinergic neurons (ChAT-Cre, loxTB MC4R mice) modestly reduced body weight gain without altering food intake and was sufficient to normalize energy expenditure and attenuate hyperglycemia and hyperinsulinemia. In contrast, restoration of MC4R expression in brainstem neurons including those in the dorsal motor nucleus of the vagus (Phox2b-Cre, loxTB MC4R mice) was sufficient to attenuate hyperinsulinemia, while the hyperglycemia and energy balance were not normalized. Additionally, hepatic insulin action and insulin-mediated suppression of hepatic glucose production were improved in ChAT-Cre, loxTB MC4R mice. These findings suggest that MC4Rs expressed by cholinergic neurons regulate energy expenditure and hepatic glucose production. Our results also provide further evidence of the dissociation in pathways mediating the effects of melanocortins on energy balance and glucose homeostasis.


Assuntos
Glucose/metabolismo , Neurônios/metabolismo , Receptor Tipo 4 de Melanocortina/fisiologia , Animais , Colina O-Acetiltransferase/genética , Ingestão de Alimentos , Metabolismo Energético , Homeostase , Hiperglicemia/metabolismo , Hiperinsulinismo/metabolismo , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo
13.
J Neurosci ; 30(44): 14630-4, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21048120

RESUMO

D-Fenfluramine (D-Fen) increases serotonin (5-HT) content in the synaptic cleft and exerts anorexigenic effects in animals and humans. However, the neural circuits that mediate these effects are not fully identified. To address this issue, we assessed the efficacy of D-Fen-induced hypophagia in mouse models with manipulations of several genes in selective populations of neurons. Expectedly, we found that global deletion of 5-HT 2C receptors (5-HT(2C)Rs) significantly attenuated D-Fen-induced anorexia. These anorexigenic effects were restored in mice with 5-HT(2C)Rs expressed only in pro-opiomelanocortin (POMC) neurons. Further, we found that deletion of melanocortin 4 receptors (MC4Rs), a downstream target of POMC neurons, abolished anorexigenic effects of D-Fen. Reexpression of MC4Rs only in SIM1 neurons in the hypothalamic paraventricular nucleus and neurons in the amygdala was sufficient to restore the hypophagic property of D-Fen. Thus, our results identify a neurochemically defined neural circuit through which D-Fen influences appetite and thereby indicate that this 5-HT(2C)R/POMC-MC4R/SIM1 circuit may yield a more refined target to exploit for weight loss.


Assuntos
Anorexia/metabolismo , Anorexia/fisiopatologia , Fenfluramina/farmacologia , Melanocortinas/fisiologia , Serotonina/fisiologia , Animais , Anorexia/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Pró-Opiomelanocortina/fisiologia , Receptor Tipo 4 de Melanocortina/deficiência , Receptor Tipo 4 de Melanocortina/genética , Receptor 5-HT2C de Serotonina/deficiência , Receptor 5-HT2C de Serotonina/genética , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Redução de Peso/genética , Redução de Peso/fisiologia
14.
Endocrinology ; 150(11): 4874-82, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19819947

RESUMO

Recent studies demonstrated a role for hypothalamic insulin and leptin action in the regulation of glucose homeostasis. This regulation involves proopiomelanocortin (POMC) neurons because suppression of phosphatidyl inositol 3-kinase (PI3K) signaling in these neurons blunts the acute effects of insulin and leptin on POMC neuronal activity. In the current study, we investigated whether disruption of PI3K signaling in POMC neurons alters normal glucose homeostasis using mouse models designed to both increase and decrease PI3K-mediated signaling in these neurons. We found that deleting p85alpha alone induced resistance to diet-induced obesity. In contrast, deletion of the p110alpha catalytic subunit of PI3K led to increased weight gain and adipose tissue along with reduced energy expenditure. Independent of these effects, increased PI3K activity in POMC neurons improved insulin sensitivity, whereas decreased PI3K signaling resulted in impaired glucose regulation. These studies show that activity of the PI3K pathway in POMC neurons is involved in not only normal energy regulation but also glucose homeostasis.


Assuntos
Glucose/metabolismo , Homeostase , Hipotálamo/metabolismo , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Opiomelanocortina/metabolismo , Transdução de Sinais , Animais , Metabolismo Energético , Feminino , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Fosfatidilinositol 3-Quinases/genética , Pró-Opiomelanocortina/genética
15.
EMBO Rep ; 10(10): 1175-81, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19713961

RESUMO

Within the central nervous system (CNS), the hypothalamus senses and integrates information on the nutrient state of the body. However, the molecular mechanisms translating nutrient sensing into changes in gene expression and, ultimately, nutrient intake remain unclear. A crucial function for the cyclic AMP-response element binding protein (CREB) co-activator CREB-regulated transcription co-activator 2 (CRTC2) in maintaining glucose homeostasis has been shown in the liver. Here, we report CRTC2 expression in distinct areas of the CNS, including hypothalamic neurons. We show that hypothalamic CRTC2 phosphorylation and subcellular localization is altered by nutrient state. Specifically, glucose regulates hypothalamic CRTC2 activity via AMP-activated protein kinase (AMPK)-mediated phosphorylation of CRTC2. Hypothalamic AMPK controls the expression of the cAMP response element (CRE) gene, insulin receptor substrate 2 (Irs2), by regulating CRTC2 occupancy of the Irs2 promoter. Indeed, CRTC2 is required for the appropriate expression of specific hypothalamic CRE genes. Our data identify CRTC2 as a new hypothalamic AMPK target and highlight a role for CRTC2 in the mechanisms linking hypothalamic glucose sensing with CRE gene regulation.


Assuntos
Regulação da Expressão Gênica , Glucose/metabolismo , Hipotálamo/metabolismo , Transativadores/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Camundongos , Ratos , Técnicas de Cultura de Tecidos , Fatores de Transcrição
16.
Exp Physiol ; 94(8): 857-66, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19542189

RESUMO

Obesity, due to its associated co-morbidities, including type 2 diabetes and cardiovascular disease, is at the forefront of today's health care concerns. Our need for novel, multifaceted approaches to tackle the global increase of waistlines is urgent, and understanding the physiological processes underlying our vulnerability to weight gain is an important one of them. Evidence for considerable heritability of body weight indicates genetic influences in the susceptibility to our obesogenic environment. Here, we will focus on neurons in brain structures such as the hypothalamus, which sense the body's metabolic state and, through an intricate cascade of events, elicit an appropriate response. We will explore the use of genetically modified mouse models in the investigation of physiological functions of genes and pathways in neuronal regulation of metabolic balance. Use of these techniques allows us to make manipulations at the molecular level (e.g. in the neuronal metabolic sensing mechanism) and combine this with systems-level physiological analysis (e.g. body weight). Recent technological advances also enable the investigation of the contributions of genes to the co-morbidities of obesity, such as obesity-induced hypertension. Reviewing examples of improvements as well as large gaps in our knowledge, this lecture aims to incite interest in whole body physiological research.


Assuntos
Metabolismo Energético/fisiologia , Fome , Hipotálamo/fisiologia , Neurônios/fisiologia , Obesidade/genética , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Metabolismo Energético/genética , Homeostase/genética , Homeostase/fisiologia , Hipertensão/etiologia , Camundongos , Obesidade/complicações , Pró-Opiomelanocortina/fisiologia , Receptor Tipo 4 de Melanocortina/fisiologia
17.
J Clin Invest ; 118(5): 1796-805, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18382766

RESUMO

Normal food intake and body weight homeostasis require the direct action of leptin on hypothalamic proopiomelanocortin (POMC) neurons. It has been proposed that leptin action requires PI3K activity. We therefore assessed the contribution of PI3K signaling to leptin's effects on POMC neurons and organismal energy balance. Leptin caused a rapid depolarization of POMC neurons and an increase in action potential frequency in patch-clamp recordings of hypothalamic slices. Pharmacologic inhibition of PI3K prevented this depolarization and increased POMC firing rate, indicating a PI3K-dependent mechanism of leptin action. Mice with genetically disrupted PI3K signaling in POMC cells failed to undergo POMC depolarization or increased firing frequency in response to leptin. Insulin's ability to hyperpolarize POMC neurons was also abolished in these mice. Moreover, targeted disruption of PI3K blunted the suppression of feeding elicited by central leptin administration. Despite these differences, mice with impaired PI3K signaling in POMC neurons exhibited normal long-term body weight regulation. Collectively, these results suggest that PI3K signaling in POMC neurons is essential for leptin-induced activation and insulin-induced inhibition of POMC cells and for the acute suppression of food intake elicited by leptin, but is not a major contributor to the regulation of long-term organismal energy homeostasis.


Assuntos
Hipotálamo/citologia , Leptina/metabolismo , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Opiomelanocortina/metabolismo , Transdução de Sinais/fisiologia , Animais , Peso Corporal , Ingestão de Alimentos , Metabolismo Energético , Homeostase , Humanos , Camundongos , Camundongos Knockout , Neurônios/citologia , Técnicas de Patch-Clamp
18.
Endocrinology ; 149(4): 1773-85, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18162515

RESUMO

Two known types of leptin-responsive neurons reside within the arcuate nucleus: the agouti gene-related peptide (AgRP)/neuropeptide Y (NPY) neuron and the proopiomelanocortin (POMC) neuron. By deleting the leptin receptor gene (Lepr) specifically in AgRP/NPY and/or POMC neurons of mice, we examined the several and combined contributions of these neurons to leptin action. Body weight and adiposity were increased by Lepr deletion from AgRP and POMC neurons individually, and simultaneous deletion in both neurons (A+P LEPR-KO mice) further increased these measures. Young (periweaning) A+P LEPR-KO mice exhibit hyperphagia and decreased energy expenditure, with increased weight gain, oxidative sparing of triglycerides, and increased fat accumulation. Interestingly, however, many of these abnormalities were attenuated in adult animals, and high doses of leptin partially suppress food intake in the A+P LEPR-KO mice. Although mildly hyperinsulinemic, the A+P LEPR-KO mice displayed normal glucose tolerance and fertility. Thus, AgRP/NPY and POMC neurons each play mandatory roles in aspects of leptin-regulated energy homeostasis, high leptin levels in adult mice mitigate the importance of leptin-responsiveness in these neurons for components of energy balance, suggesting the presence of other leptin-regulated pathways that partially compensate for the lack of leptin action on the POMC and AgRP/NPY neurons.


Assuntos
Proteína Relacionada com Agouti/fisiologia , Ingestão de Alimentos , Metabolismo Energético , Pró-Opiomelanocortina/fisiologia , Receptores para Leptina/fisiologia , Animais , Composição Corporal , Fertilidade , Hiperinsulinismo/etiologia , Hiperfagia , Lactação , Masculino , Camundongos , Neuropeptídeo Y/fisiologia
19.
Nature ; 449(7159): 228-32, 2007 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-17728716

RESUMO

A subset of neurons in the brain, known as 'glucose-excited' neurons, depolarize and increase their firing rate in response to increases in extracellular glucose. Similar to insulin secretion by pancreatic beta-cells, glucose excitation of neurons is driven by ATP-mediated closure of ATP-sensitive potassium (K(ATP)) channels. Although beta-cell-like glucose sensing in neurons is well established, its physiological relevance and contribution to disease states such as type 2 diabetes remain unknown. To address these issues, we disrupted glucose sensing in glucose-excited pro-opiomelanocortin (POMC) neurons via transgenic expression of a mutant Kir6.2 subunit (encoded by the Kcnj11 gene) that prevents ATP-mediated closure of K(ATP) channels. Here we show that this genetic manipulation impaired the whole-body response to a systemic glucose load, demonstrating a role for glucose sensing by POMC neurons in the overall physiological control of blood glucose. We also found that glucose sensing by POMC neurons became defective in obese mice on a high-fat diet, suggesting that loss of glucose sensing by neurons has a role in the development of type 2 diabetes. The mechanism for obesity-induced loss of glucose sensing in POMC neurons involves uncoupling protein 2 (UCP2), a mitochondrial protein that impairs glucose-stimulated ATP production. UCP2 negatively regulates glucose sensing in POMC neurons. We found that genetic deletion of Ucp2 prevents obesity-induced loss of glucose sensing, and that acute pharmacological inhibition of UCP2 reverses loss of glucose sensing. We conclude that obesity-induced, UCP2-mediated loss of glucose sensing in glucose-excited neurons might have a pathogenic role in the development of type 2 diabetes.


Assuntos
Glucose/metabolismo , Homeostase , Neurônios/metabolismo , Obesidade/fisiopatologia , Pró-Opiomelanocortina/metabolismo , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/farmacologia , Humanos , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/genética , Canais Iônicos/metabolismo , Glicosídeos Iridoides , Iridoides/farmacologia , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Obesidade/induzido quimicamente , Obesidade/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteína Desacopladora 2
20.
Science ; 317(5834): 94-9, 2007 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-17556551

RESUMO

Forming distinct representations of multiple contexts, places, and episodes is a crucial function of the hippocampus. The dentate gyrus subregion has been suggested to fulfill this role. We have tested this hypothesis by generating and analyzing a mouse strain that lacks the gene encoding the essential subunit of the N-methyl-d-aspartate (NMDA) receptor NR1, specifically in dentate gyrus granule cells. The mutant mice performed normally in contextual fear conditioning, but were impaired in the ability to distinguish two similar contexts. A significant reduction in the context-specific modulation of firing rate was observed in the CA3 pyramidal cells when the mutant mice were transferred from one context to another. These results provide evidence that NMDA receptors in the granule cells of the dentate gyrus play a crucial role in the process of pattern separation.


Assuntos
Giro Denteado/fisiologia , Hipocampo/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Plasticidade Neuronal , Reconhecimento Fisiológico de Modelo , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Comportamento Animal , Condicionamento Psicológico , Sinais (Psicologia) , Giro Denteado/citologia , Discriminação Psicológica , Potenciais Pós-Sinápticos Excitadores , Medo , Hipocampo/citologia , Aprendizagem em Labirinto , Camundongos , Camundongos Knockout , Via Perfurante , Células Piramidais/fisiologia , Receptores de N-Metil-D-Aspartato/genética , Recombinação Genética , Transmissão Sináptica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...