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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 152(3): 612-9, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374353

RESUMEN

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.


Asunto(s)
Tronco Encefálico/metabolismo , Insulina/metabolismo , Neuronas/metabolismo , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Presión Sanguínea , Tronco Encefálico/citología , Neuronas Colinérgicas/metabolismo , AMP Cíclico/metabolismo , Fenómenos Electrofisiológicos , Humanos , Canales KATP/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Obesidad/fisiopatología , Sistema Nervioso Parasimpático/metabolismo , Receptor de Melanocortina Tipo 4/genética , Médula Espinal/metabolismo , Sistema Nervioso Simpático/metabolismo
2.
Proc Natl Acad Sci U S A ; 113(43): 12298-12303, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27791019

RESUMEN

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.


Asunto(s)
Hipertensión/genética , Leptina/metabolismo , Obesidad/genética , Efectos Tardíos de la Exposición Prenatal/genética , Receptor de Melanocortina Tipo 4/genética , Animales , Presión Sanguínea/genética , Dieta/efectos adversos , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertensión/complicaciones , Hipertensión/fisiopatología , Leptina/genética , Masculino , Relaciones Materno-Fetales/fisiología , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Obesidad/complicaciones , Obesidad/fisiopatología , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/patología
3.
Mol Cell Neurosci ; 68: 258-71, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26292267

RESUMEN

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).


Asunto(s)
Neuronas/fisiología , Receptor de Galanina Tipo 1/metabolismo , Receptor de Galanina Tipo 2/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Ganglios Espinales/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Microscopía Confocal , ARN Mensajero/metabolismo , Receptor de Galanina Tipo 1/genética , Receptor de Galanina Tipo 2/genética , Médula Espinal/metabolismo , Proteína Fluorescente Roja
4.
Nature ; 449(7159): 228-32, 2007 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-17728716

RESUMEN

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.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Neuronas/metabolismo , Obesidad/fisiopatología , Proopiomelanocortina/metabolismo , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Humanos , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/genética , Canales Iónicos/metabolismo , Glicósidos Iridoides , Iridoides/farmacología , Ratones , Ratones Obesos , Ratones Transgénicos , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Obesidad/inducido químicamente , Obesidad/metabolismo , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Proteína Desacopladora 2
5.
J Neurosci ; 30(44): 14630-4, 2010 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21048120

RESUMEN

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.


Asunto(s)
Anorexia/metabolismo , Anorexia/fisiopatología , Fenfluramina/farmacología , Melanocortinas/fisiología , Serotonina/fisiología , Animales , Anorexia/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vías Nerviosas/citología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Proopiomelanocortina/fisiología , Receptor de Melanocortina Tipo 4/deficiencia , Receptor de Melanocortina Tipo 4/genética , Receptor de Serotonina 5-HT2C/deficiencia , Receptor de Serotonina 5-HT2C/genética , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Pérdida de Peso/genética , Pérdida de Peso/fisiología
6.
Cell Metab ; 4(2): 123-32, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16890540

RESUMEN

Suppressor of cytokine signaling-3 (Socs-3) negatively regulates the action of various cytokines, as well as the metabolic hormones leptin and insulin. Mice with haploinsufficiency of Socs-3, or those with neuronal deletion of Socs-3, are lean and more leptin and insulin sensitive. To examine the role of Socs-3 within specific neurons critical to energy balance, we created mice with selective deletion of Socs-3 within pro-opiomelanocortin (POMC)-expressing cells. These mice had enhanced leptin sensitivity, measured by weight loss and food intake after leptin infusion. On chow diet, glucose homeostasis was improved despite normal weight gain. On a high-fat diet, the rate of weight gain was reduced, due to increased energy expenditure rather than decreased food intake; glucose homeostasis and insulin sensitivity were substantially improved. These studies demonstrate that Socs-3 within POMC neurons regulates leptin sensitivity and glucose homeostasis, and plays a key role in linking high-fat diet to disordered metabolism.


Asunto(s)
Grasas de la Dieta/farmacología , Glucosa/metabolismo , Homeostasis , Leptina/farmacología , Proopiomelanocortina/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Animales , Células Cultivadas , Hígado Graso/prevención & control , Ratones , Ratones Noqueados , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Pérdida de Peso
7.
J Clin Invest ; 118(5): 1796-805, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18382766

RESUMEN

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.


Asunto(s)
Hipotálamo/citología , Leptina/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proopiomelanocortina/metabolismo , Transducción de Señal/fisiología , Animales , Peso Corporal , Ingestión de Alimentos , Metabolismo Energético , Homeostasis , Humanos , Ratones , Ratones Noqueados , Neuronas/citología , Técnicas de Placa-Clamp
8.
EMBO Rep ; 10(10): 1175-81, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19713961

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Glucosa/metabolismo , Hipotálamo/metabolismo , Transactivadores/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ratones , Ratas , Técnicas de Cultivo de Tejidos , Factores de Transcripción
9.
Neuron ; 49(2): 191-203, 2006 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-16423694

RESUMEN

Leptin, an adipocyte-derived hormone, acts directly on the brain to control food intake and energy expenditure. An important question is the identity of first-order neurons initiating leptin's anti-obesity effects. A widely held view is that most, if not all, of leptin's effects are mediated by neurons located in the arcuate nucleus of the hypothalamus. However, leptin receptors (LEPRs) are expressed in other sites as well, including the ventromedial hypothalamus (VMH). The possible role of leptin acting in "nonarcuate" sites has largely been ignored. In the present study, we show that leptin depolarizes and increases the firing rate of steroidogenic factor-1 (SF1)-positive neurons in the VMH. We also show, by generating mice that lack LEPRs on SF1-positive neurons, that leptin action at this site plays an important role in reducing body weight and, of note, in resisting diet-induced obesity. These results reveal a critical role for leptin action on VMH neurons.


Asunto(s)
Peso Corporal/fisiología , Proteínas de Homeodominio/fisiología , Homeostasis/fisiología , Leptina/farmacología , Neuronas/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/fisiología , Animales , Composición Corporal/efectos de los fármacos , Composición Corporal/genética , Composición Corporal/fisiología , Dieta , Electrofisiología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Homeostasis/efectos de los fármacos , Inmunohistoquímica , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Obesidad/fisiopatología , Técnicas de Placa-Clamp , Fenotipo , Sondas ARN , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Leptina , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor Esteroidogénico 1 , Factores de Transcripción/genética , Núcleo Hipotalámico Ventromedial/citología
10.
Neuron ; 51(2): 239-49, 2006 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-16846858

RESUMEN

The neural pathways through which central serotonergic systems regulate food intake and body weight remain to be fully elucidated. We report that serotonin, via action at serotonin1B receptors (5-HT1BRs), modulates the endogenous release of both agonists and antagonists of the melanocortin receptors, which are a core component of the central circuitry controlling body weight homeostasis. We also show that serotonin-induced hypophagia requires downstream activation of melanocortin 4, but not melanocortin 3, receptors. These results identify a primary mechanism underlying the serotonergic regulation of energy balance and provide an example of a centrally derived signal that reciprocally regulates melanocortin receptor agonists and antagonists in a similar manner to peripheral adiposity signals.


Asunto(s)
Ingestión de Alimentos/fisiología , Neuronas/fisiología , Receptor de Melanocortina Tipo 3/fisiología , Receptor de Serotonina 5-HT1B/fisiología , Receptores de Melanocortina/fisiología , Serotonina/fisiología , Animales , Ingestión de Alimentos/efectos de los fármacos , Estimulación Eléctrica , Masculino , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Neuronas/efectos de los fármacos , Piridinas/farmacología , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/antagonistas & inhibidores , Receptor de Melanocortina Tipo 4/fisiología , Receptores de Melanocortina/agonistas , Receptores de Melanocortina/antagonistas & inhibidores , Serotonina/farmacología , Agonistas del Receptor de Serotonina 5-HT1
11.
J Clin Invest ; 116(7): 1886-901, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16794735

RESUMEN

Leptin and insulin have been identified as fuel sensors acting in part through their hypothalamic receptors to inhibit food intake and stimulate energy expenditure. As their intracellular signaling converges at the PI3K pathway, we directly addressed the role of phosphatidylinositol3,4,5-trisphosphate-mediated (PIP3-mediated) signals in hypothalamic proopiomelanocortin (POMC) neurons by inactivating the gene for the PIP3 phosphatase Pten specifically in this cell type. Here we show that POMC-specific disruption of Pten resulted in hyperphagia and sexually dimorphic diet-sensitive obesity. Although leptin potently stimulated Stat3 phosphorylation in POMC neurons of POMC cell-restricted Pten knockout (PPKO) mice, it failed to significantly inhibit food intake in vivo. POMC neurons of PPKO mice showed a marked hyperpolarization and a reduction in basal firing rate due to increased ATP-sensitive potassium (KATP) channel activity. Leptin was not able to elicit electrical activity in PPKO POMC neurons, but application of the PI3K inhibitor LY294002 and the KATP blocker tolbutamide restored electrical activity and leptin-evoked firing of POMC neurons in these mice. Moreover, icv administration of tolbutamide abolished hyperphagia in PPKO mice. These data indicate that PIP3-mediated signals are critical regulators of the melanocortin system via modulation of KATP channels.


Asunto(s)
Neuronas/metabolismo , Obesidad , Fosfohidrolasa PTEN/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Canales de Potasio/metabolismo , Proopiomelanocortina/metabolismo , Sistemas de Mensajero Secundario/fisiología , Animales , Cromonas/metabolismo , Dieta , Ingestión de Alimentos/efectos de los fármacos , Femenino , Hipoglucemiantes/farmacología , Hipotálamo/citología , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Noqueados , Morfolinas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Tolbutamida/farmacología
12.
Exp Physiol ; 94(8): 857-66, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19542189

RESUMEN

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.


Asunto(s)
Metabolismo Energético/fisiología , Hambre , Hipotálamo/fisiología , Neuronas/fisiología , Obesidad/genética , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Metabolismo Energético/genética , Homeostasis/genética , Homeostasis/fisiología , Hipertensión/etiología , Ratones , Obesidad/complicaciones , Proopiomelanocortina/fisiología , Receptor de Melanocortina Tipo 4/fisiología
13.
Nat Neurosci ; 8(10): 1289-91, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16158063

RESUMEN

Multiple hormones controlling energy homeostasis regulate the expression of neuropeptide Y (NPY) and agouti-related peptide (AgRP) in the arcuate nucleus of the hypothalamus. Nevertheless, inactivation of the genes encoding NPY and/or AgRP has no impact on food intake in mice. Here we demonstrate that induced selective ablation of AgRP-expressing neurons in adult mice results in acute reduction of feeding, demonstrating direct evidence for a critical role of these neurons in the regulation of energy homeostasis.


Asunto(s)
Núcleo Arqueado del Hipotálamo/citología , Conducta Alimentaria/fisiología , Regulación de la Expresión Génica/fisiología , Neuronas/metabolismo , Proteínas/metabolismo , Proteína Relacionada con Agouti , Animales , Anorexia/metabolismo , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Recuento de Células/métodos , Toxina Diftérica/farmacología , Ingestión de Alimentos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Proopiomelanocortina/deficiencia , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Proteínas/genética , Factores de Tiempo , beta-Galactosidasa/biosíntesis
14.
Neuron ; 102(3): 653-667.e6, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30879785

RESUMEN

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.


Asunto(s)
Conducta Alimentaria/fisiología , Neuronas/citología , Obesidad/fisiopatología , Núcleo Hipotalámico Paraventricular/citología , Respuesta de Saciedad/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metabolismo Energético , Encefalinas/metabolismo , Locus Coeruleus/citología , Locus Coeruleus/metabolismo , Locus Coeruleus/fisiología , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiología , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Precursores de Proteínas/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Proteínas Represoras/metabolismo
15.
Neuron ; 42(6): 983-91, 2004 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-15207242

RESUMEN

Neuroanatomical and electrophysiological studies have shown that hypothalamic POMC neurons are targets of the adipostatic hormone leptin. However, the physiological relevance of leptin signaling in these neurons has not yet been directly tested. Here, using the Cre/loxP system, we critically test the functional importance of leptin action on POMC neurons by deleting leptin receptors specifically from these cells in mice. Mice lacking leptin signaling in POMC neurons are mildly obese, hyperleptinemic, and have altered expression of hypothalamic neuropeptides. In summary, leptin receptors on POMC neurons are required but not solely responsible for leptin's regulation of body weight homeostasis.


Asunto(s)
Peso Corporal/fisiología , Homeostasis/fisiología , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Receptores de Superficie Celular/fisiología , Transducción de Señal/fisiología , Factores de Edad , Animales , Composición Corporal/genética , Ingestión de Alimentos/genética , Femenino , Expresión Génica/genética , Proteínas Fluorescentes Verdes , Hipotálamo/citología , Hipotálamo/metabolismo , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Leptina/sangre , Leptina/farmacología , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuropéptidos/metabolismo , Consumo de Oxígeno/genética , Proopiomelanocortina/genética , ARN Mensajero/metabolismo , Receptores de Leptina , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores Sexuales , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Factores de Transcripción/metabolismo , alfa-MSH/metabolismo
16.
Endocrinology ; 149(4): 1773-85, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18162515

RESUMEN

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.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Ingestión de Alimentos , Metabolismo Energético , Proopiomelanocortina/fisiología , Receptores de Leptina/fisiología , Animales , Composición Corporal , Fertilidad , Hiperinsulinismo/etiología , Hiperfagia , Lactancia , Masculino , Ratones , Neuropéptido Y/fisiología
17.
J Clin Invest ; 115(12): 3564-72, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16322794

RESUMEN

Ghrelin is the endogenous ligand for the growth hormone secretagogue receptor (GHSR; ghrelin receptor). Since its discovery, accumulating evidence has suggested that ghrelin may play a role in signaling and reversing states of energy insufficiency. For example, ghrelin levels rise following food deprivation, and ghrelin administration stimulates feeding and increases body weight and adiposity. However, recent loss-of-function studies have raised questions regarding the physiological significance of ghrelin in regulating these processes. Here, we present results of a study using a novel GHSR-null mouse model, in which ghrelin administration fails to acutely stimulate food intake or activate arcuate nucleus neurons. We show that when fed a high-fat diet, both female and male GHSR-null mice eat less food, store less of their consumed calories, preferentially utilize fat as an energy substrate, and accumulate less body weight and adiposity than control mice. Similar effects on body weight and adiposity were also observed in female, but not male, GHSR-null mice fed standard chow. GHSR deletion also affected locomotor activity and levels of glycemia. These findings support the hypothesis that ghrelin-responsive pathways are an important component of coordinated body weight control. Moreover, our data suggest that ghrelin signaling is required for development of the full phenotype of diet-induced obesity.


Asunto(s)
Dieta , Obesidad/genética , Hormonas Peptídicas/fisiología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Tejido Adiposo/metabolismo , Alelos , Análisis de Varianza , Alimentación Animal , Animales , Glucemia/metabolismo , Southern Blotting , Western Blotting , Composición Corporal , Peso Corporal , Cruzamientos Genéticos , ADN/metabolismo , Femenino , Eliminación de Gen , Predisposición Genética a la Enfermedad , Genotipo , Ghrelina , Heterocigoto , Homeostasis , Hiperglucemia/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leptina/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Neuronas/metabolismo , Obesidad/metabolismo , Hormonas Peptídicas/química , Fenotipo , ARN Mensajero/metabolismo , Receptores de Ghrelina , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinación Genética , Transducción de Señal , Tinción con Nitrato de Plata , Factores de Tiempo
18.
PLoS One ; 11(4): e0153187, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27077912

RESUMEN

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.


Asunto(s)
Analgesia , Bradicardia/metabolismo , Tronco Encefálico/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Insuficiencia Respiratoria/metabolismo , Analgésicos Opioides/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Tronco Encefálico/citología , Tronco Encefálico/efectos de los fármacos , Channelrhodopsins , Femenino , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones Transgénicos , Microscopía Confocal , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas/efectos de los fármacos , Núcleo Solitario/citología , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo
19.
J Comp Neurol ; 493(1): 63-71, 2005 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-16254991

RESUMEN

The past decade has greatly increased our understanding and appreciation of the ability of the central nervous system (CNS) to regulate food intake and body weight. This was spearheaded by the discovery of key molecules regulating body weight homeostasis. It is now also apparent that the CNS, especially the hypothalamus, plays a primary role in directly regulating glucose homeostasis, independently of effects on body weight. These discoveries are important given the increasing incidences of obesity and type II diabetes in Western societies. In this article, we will highlight recent data from genetically modified mice. These data and other models have helped to dissect the CNS pathways regulating body weight and glucose homeostasis. Finally, although these studies have been illustrative, they also underscore our relative lack of knowledge and highlight the need for more definitive approaches to unravel the functional significance of these pathways.


Asunto(s)
Glucemia/metabolismo , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Hipotálamo/fisiología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Homeostasis/fisiología , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Hipotálamo/metabolismo , Inmunohistoquímica , Leptina/metabolismo , Leptina/farmacología , Leptina/fisiología , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Proopiomelanocortina/metabolismo , Proopiomelanocortina/fisiología , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/fisiología , Receptores de Leptina
20.
Endocrinology ; 145(4): 1602-11, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14701677

RESUMEN

The type 1A GH secretagogue (GHS) receptor (GHSR) has been proposed to mediate the effects of ghrelin on GH release, food intake, and body composition. We have overexpressed GHSR in GH-producing GC cells and GHRH neurons in an attempt to enhance signaling via this pathway selectively, in the GH axis. Constitutive overexpression of human GHSR in rat GC cell lines resulted in increased basal phosphoinositol turnover and rendered them responsive to GHS ligands. We then generated transgenic mice overexpressing human GHSR in GHRH neurons using a 38-kb rat GHRH cosmid promoter. GHRH-GHSR transgenic mice showed increased hypothalamic GHRH expression, pituitary GH contents, and postweaning growth rates. Body weights of the transgenic mice became similar in adulthood, whereas adipose mass was reduced, particularly so in female GHRH-GHSR mice. Organ and muscle weights of transgenic mice were increased despite chronic exposure to a high fat diet. These results suggest that constitutive overexpression of GHSR in GHRH neurons up-regulates basal activity in the GHRH-GH axis. However, GHRH-GHSR mice showed no evidence of increased sensitivity to acute or chronic treatment with exogenous GHS ligands. Food intake and adipose tissue responses to chronic high fat feeding and treatment with GHS ligands were unaffected, as were locomotor and anxiety behaviors, although GHRH-GHSR mice remained significantly leaner than wild-type littermates. Thus, constitutive overexpression of GHSR can up-regulate basal signaling activity in the GHRH/GH axis and reduce adiposity without affecting other GHSR-mediated signals.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Tejido Adiposo/patología , Animales , Ansiedad , Conducta Animal/efectos de los fármacos , Línea Celular , Grasas de la Dieta/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Hormona del Crecimiento/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/crecimiento & desarrollo , Actividad Motora , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Oligopéptidos/farmacología , Prolactina/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Receptores de Ghrelina , Delgadez
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA