Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Metabolism ; 123: 154839, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34331964

RESUMEN

BACKGROUND AND AIMS: Serotonergic and dopaminergic systems in the brain are essential for homeostatic and reward-associated regulation of food intake and systemic energy metabolism. It is largely unknown how fasting influences these systems or if such effects are altered in humans with obesity. We therefore aimed to evaluate the effects of fasting on hypothalamic/thalamic serotonin transporter (SERT) and striatal dopamine transporter (DAT) availability in lean subjects and subjects with obesity. METHODS: In this randomized controlled cross-over trial, we assessed the effects of 12 vs 24 h of fasting on SERT and DAT availability in the hypothalamus/thalamus and striatum, respectively, using SPECT imaging in 10 lean men and 10 men with obesity. RESULTS: As compared with the 12-h fast, a 24-h fast increased hypothalamic SERT availability in lean men, but not in men with obesity. We observed high inter-individual variation in the effects of fasting on thalamic SERT and striatal DAT, with no differences between lean men and those with obesity. In all subjects, fasting-induced increases in circulating free fatty acid (FFA) concentrations were associated with an increase in hypothalamic SERT availability and a decrease in striatal DAT availability. Multiple regression analysis showed that changes in plasma insulin and FFAs together accounted for 44% of the observed variation in striatal DAT availability. CONCLUSION: Lean men respond to prolonged fasting by increasing hypothalamic SERT availability, whereas this response is absent in men with obesity. Inter-individual differences in the adaptations of the cerebral serotonergic and dopaminergic systems to fasting may, in part, be explained by changes in peripheral metabolic signals of fasting, including FFAs and insulin.


Asunto(s)
Ayuno , Hipotálamo/fisiopatología , Obesidad/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Anciano , Estudios de Casos y Controles , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/metabolismo , Estudios Cruzados , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Humanos , Hipotálamo/diagnóstico por imagen , Hipotálamo/metabolismo , Insulina/metabolismo , Masculino , Persona de Mediana Edad , Tomografía Computarizada de Emisión de Fotón Único
2.
Nature ; 585(7825): 351-352, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32879476
3.
JCI Insight ; 3(17)2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30185666

RESUMEN

When obesity is caused by consumption of a high-fat diet, the tumor suppressor pRb is phosphoinactivated in the neurons of the mediobasal hypothalamus, a brain area critical for energy-balance regulation. However, the functional relevance of pRb phosphoinactivation in the mediobasal hypothalamus to diet-induced obesity remains unknown. Here, we show that inhibiting pRb phosphorylation in the mediobasal hypothalamus can prevent and treat diet-induced obesity in mice. Expressing an unphosphorylable pRb nonselectively in the mediobasal hypothalamus or conditionally in anorexigenic POMC neurons inhibits diet-induced obesity. Intracerebroventricular delivery of US Food and Drug Administration-approved (FDA-approved) cyclin-dependent kinase 4 (CDK4) inhibitor abemaciclib inhibits pRb phosphorylation in the mediobasal hypothalamus and prevents diet-induced obesity. Oral administration of abemaciclib at doses approved for human use reduces fat mass in diet-induced obese mice by increasing lipid oxidation without significantly reducing lean mass. With analysis of recent literature identifying CDK4 as the most abundantly expressed neuronal CDK in the mediobasal hypothalamus, our work uncovers CDK4 as the major kinase for hypothalamic pRb phosphoinactivation and a highly effective central antiobesity target. As three CDK4/6 inhibitors have recently received FDA approval for life-long breast cancer therapy, our study provides a preclinical basis for their expedient repurposing for obesity management.


Asunto(s)
Aminopiridinas/farmacología , Bencimidazoles/farmacología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Dieta Alta en Grasa/efectos adversos , Obesidad/metabolismo , Obesidad/prevención & control , Animales , Modelos Animales de Enfermedad , Aprobación de Drogas , Metabolismo Energético , Hipotálamo/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Estados Unidos , United States Food and Drug Administration
4.
Mol Metab ; 8: 37-50, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29290621

RESUMEN

OBJECTIVE: Bile acids have been implicated as important regulators of glucose metabolism via activation of FXR and GPBAR1. We have previously shown that FGF19 can modulate glucose handling by suppressing the activity of hypothalamic AGRP/NPY neurons. As bile acids stimulate the release of FGF19/FGF15 into the circulation, we pursued the potential of bile acids to improve glucose tolerance via a gut-brain axis involving FXR and FGF15/FGF19 within enterocytes and FGF receptors on hypothalamic AGRP/NPY neurons. METHODS: A 5-day gavage of taurocholic acid, mirroring our previous protocol of a 5-day FGF19 treatment, was performed. Oral glucose tolerance tests in mice with genetic manipulations of FGF signaling and melanocortin signaling were used to define a gut-brain axis responsive to bile acids. RESULTS: The taurocholic acid gavage led to increased serum concentrations of taurocholic acid as well as increases of FGF15 mRNA in the ileum and improved oral glucose tolerance in obese (ob/ob) mice. In contrast, lithocholic acid, an FXR antagonist but a potent agonist for GPBAR1, did not improve glucose tolerance. The positive response to taurocholic acid is dependent upon an intact melanocortinergic system as obese MC4R-null mice or ob/ob mice without AGRP did not show improvements in glucose tolerance after taurocholate gavage. We also tested the FGF receptor isoform necessary for the bile acid response, using AGRP:Fgfr1-/- and AGRP:Fgfr2-/- mice. While the absence of FGFR1 in AGRP/NPY neurons did not alter glucose tolerance after taurocholate gavage, manipulations of Fgfr2 caused bidirectional changes depending upon the experimental model. We hypothesized the existence of an endogenous hypothalamic FGF, most likely FGF17, that acted as a chronic activator of AGRP/NPY neurons. We developed two short peptides based on FGF8 and FGF17 that should antagonize FGF17 action. Both of these peptides improved glucose homeostasis after a 4-day course of central and peripheral injections. Significantly, daily average blood glucose from continuous glucose monitoring was reduced in all tested animals but glucose concentrations remained in the euglycemia range. CONCLUSIONS: We have defined a gut-brain axis that regulates glucose metabolism mediated by antagonistic fibroblast growth factors. From the intestine, bile acids stimulate FGF15 secretion, leading to activation of the FGF receptors in hypothalamic AGRP/NPY neurons. FGF receptor intracellular signaling subsequently silences AGRP/NPY neurons, leading to improvements of glucose tolerance that are likely mediated by the autonomic nervous system. Finally, short peptides that antagonize homodimeric FGF receptor signaling within the hypothalamus have beneficial effects on glucose homeostasis without inducing hypoglycemia. These peptides could provide a new mode of regulating glucose metabolism.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Glucemia/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Intolerancia a la Glucosa/metabolismo , Hipotálamo/metabolismo , Animales , Hipotálamo/fisiología , Ratones , Ratones Obesos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
5.
Cell Metab ; 23(1): 113-27, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26698918

RESUMEN

The integrative physiology of inter-organ communication in lipophagy regulation is not well understood. Lipophagy and the cytosolic lipases ATGL and HSL contribute to lipid droplet (LD) mobilization; however, whether autophagy proteins engage with lipases to promote lipid utilization remains unknown. Here, we show that cold induces autophagy in proopiomelanocortin (POMC) neurons and activates lipophagy in brown adipose tissue (BAT) and liver in mice. Targeted activation of autophagy in POMC neurons via intra-hypothalamic rapamycin is sufficient to trigger lipid utilization in room temperature-housed mice. Conversely, inhibiting autophagy in POMC neurons or in peripheral tissues or denervating BAT blocks lipid utilization. Unexpectedly, the autophagosome marker LC3 is mechanistically coupled to ATGL-mediated lipolysis. ATGL exhibits LC3-interacting region (LIR) motifs, and mutating a single LIR motif on ATGL displaces ATGL from LD and disrupts lipolysis. Thus, cold-induced activation of central autophagy activates lipophagy and cytosolic lipases in a complementary manner to mediate lipolysis in peripheral tissues.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Autofagia , Hipotálamo/citología , Lipólisis , Hígado/metabolismo , Adipocitos Marrones/fisiología , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/inervación , Secuencia de Aminoácidos , Animales , Frío , Femenino , Lipasa/metabolismo , Gotas Lipídicas/metabolismo , Hígado/citología , Lisosomas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Datos de Secuencia Molecular , Neuronas/fisiología , Consumo de Oxígeno , Proopiomelanocortina/metabolismo
6.
Appetite ; 71: 466-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23624294

RESUMEN

The adiposity hormone leptin has been implicated in the regulation of behavioral and metabolic controls of body weight. Leptin receptors are found in multiple peripheral and central tissues, particularly within hypothalamic and brainstem neuronal populations. Central leptinergic signaling acts as an indirect control to modulate the feeding inhibitory potency of the direct controls of meal size. Mouse models of neuronal leptin loss and gain of function have helped to identify and characterize how central leptin contributes to the central control of food intake.


Asunto(s)
Comidas , Tamaño de la Porción , Receptores de Leptina/metabolismo , Adiposidad/fisiología , Animales , Peso Corporal , Ingestión de Alimentos/fisiología , Conducta Alimentaria , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Modelos Animales , Obesidad/metabolismo , Transducción de Señal
7.
Cell Metab ; 16(5): 579-87, 2012 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23123165

RESUMEN

Direct detection of circulating nutrients by the central nervous system has been implicated in the regulation of energy balance, and the mediobasal hypothalamus is considered as the primary sensing site mediating these effects. Neurons sensitive to energyrelated signals have also been identified outside the hypothalamus, particularly within the caudomedial nucleus of the solitary tract (cmNTS) in brainstem, but the consequences of direct cmNTS nutrient detection on energy balance remain poorly characterized. Here we determined the behavioral and metabolic consequences of direct L-leucine detection by the cmNTS and investigated the intracellular signaling and neurochemical pathways implicated in cmNTS L-leucine sensing in rats. Our results support the distributed nature of central nutrient detection, evidence a role for the cmNTS S6K1 pathway in the regulation of meal size and body weight, and suggest that the cmNTS integrates direct cmNTS nutrient detection with gut-derived, descending forebrain, and adiposity signals of energy availability to regulate food intake.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Leucina/farmacología , Núcleo Solitario/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Metabolismo Energético , Hipotálamo/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Ratas , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de los fármacos , Núcleo Solitario/enzimología
8.
J Neurosci ; 32(29): 9870-7, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22815502

RESUMEN

Thioredoxin interacting protein (TXNIP) has recently been described as a key regulator of energy metabolism through pleiotropic actions that include nutrient sensing in the mediobasal hypothalamus (MBH). However, the role of TXNIP in neurochemically specific hypothalamic subpopulations and the circuits downstream from MBH TXNIP engaged to regulate energy homeostasis remain unexplored. To evaluate the metabolic role of TXNIP activity specifically within arcuate Agrp neurons, we generated Agrp-specific TXNIP gain-of-function and loss-of-function mouse models using Agrp-Ires-cre mice, TXNIP (flox/flox) mice, and a lentivector expressing the human TXNIP isoform conditionally in the presence of Cre recombinase. Overexpression of TXNIP in Agrp neurons predisposed to diet-induced obesity and adipose tissue storage by decreasing energy expenditure and spontaneous locomotion, without affecting food intake. Conversely, Agrp neuronal TXNIP deletion protected against diet-induced obesity and adipose tissue storage by increasing energy expenditure and spontaneous locomotion, also without affecting food intake. TXNIP overexpression in Agrp neurons did not primarily affect glycemic control, whereas deletion of TXNIP in Agrp neurons improved fasting glucose levels and glucose tolerance independently of its effects on body weight and adiposity. Bidirectional manipulation of TXNIP expression induced reciprocal changes in central leptin sensitivity and the neural regulation of lipolysis. Together, these results identify a critical role for TXNIP in Agrp neurons in mediating diet-induced obesity through the regulation of energy expenditure and adipose tissue metabolism, independently of food intake. They also reveal a previously unidentified role for Agrp neurons in the brain-adipose axis.


Asunto(s)
Adiposidad/fisiología , Proteína Relacionada con Agouti/metabolismo , Proteínas Portadoras/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/farmacología , Neuronas/metabolismo , Tiorredoxinas/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Glucemia/metabolismo , Proteínas Portadoras/genética , Dieta , Ingestión de Alimentos/fisiología , Homeostasis/fisiología , Hipotálamo/efectos de los fármacos , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Obesidad/genética , Obesidad/metabolismo , Tiorredoxinas/genética
9.
EMBO Rep ; 13(3): 258-65, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22249165

RESUMEN

Autophagy degrades cytoplasmic contents to achieve cellular homeostasis. We show that selective loss of autophagy in hypothalamic proopiomelanocortin (POMC) neurons decreases α-melanocyte-stimulating hormone (MSH) levels, promoting adiposity, impairing lipolysis and altering glucose homeostasis. Ageing reduces hypothalamic autophagy and α-MSH levels, and aged-mice phenocopy, the adiposity and lipolytic defect observed in POMC neuron autophagy-null mice. Intraperitoneal isoproterenol restores lipolysis in both models, demonstrating normal adipocyte catecholamine responsiveness. We propose that an unconventional, autophagosome-mediated form of secretion in POMC neurons controls energy balance by regulating α-MSH production. Modulating hypothalamic autophagy might have implications for preventing obesity and metabolic syndrome of ageing.


Asunto(s)
Autofagia/genética , Hipotálamo/metabolismo , Lipólisis/genética , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Adiposidad/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proopiomelanocortina/genética , alfa-MSH/metabolismo
10.
J Clin Invest ; 121(12): 4916-20, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22056385

RESUMEN

Increased endogenous glucose production (EGP) is a hallmark of type 2 diabetes mellitus. While there is evidence for central regulation of EGP by activation of hypothalamic ATP-sensitive potassium (K(ATP)) channels in rodents, whether these central pathways contribute to regulation of EGP in humans remains to be determined. Here we present evidence for central nervous system regulation of EGP in humans that is consistent with complementary rodent studies. Oral administration of the K(ATP) channel activator diazoxide under fixed hormonal conditions substantially decreased EGP in nondiabetic humans and Sprague Dawley rats. In rats, comparable doses of oral diazoxide attained appreciable concentrations in the cerebrospinal fluid, and the effects of oral diazoxide were abolished by i.c.v. administration of the K(ATP) channel blocker glibenclamide. These results suggest that activation of hypothalamic K(ATP) channels may be an important regulator of EGP in humans and that this pathway could be a target for treatment of hyperglycemia in type 2 diabetes mellitus.


Asunto(s)
Diazóxido/farmacología , Gluconeogénesis/efectos de los fármacos , Hipotálamo/metabolismo , Canales de Potasio/fisiología , Adulto , Animales , Glucemia/análisis , Barrera Hematoencefálica , Bloqueadores de los Canales de Calcio/administración & dosificación , Bloqueadores de los Canales de Calcio/farmacología , Diazóxido/administración & dosificación , Diazóxido/líquido cefalorraquídeo , Diazóxido/farmacocinética , Diazóxido/uso terapéutico , Método Doble Ciego , Inducción Enzimática/efectos de los fármacos , Femenino , Gluconeogénesis/fisiología , Técnica de Clampeo de la Glucosa , Glucosa-6-Fosfatasa/antagonistas & inhibidores , Glucosa-6-Fosfatasa/biosíntesis , Glucosa-6-Fosfatasa/genética , Gliburida/administración & dosificación , Gliburida/farmacología , Humanos , Hipotálamo/fisiopatología , Inyecciones Intraventriculares , Insulina/sangre , Activación del Canal Iónico/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Fosforilación/efectos de los fármacos , Canales de Potasio/agonistas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo
11.
Cell Metab ; 14(2): 173-83, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21803288

RESUMEN

Macroautophagy is a lysosomal degradative pathway that maintains cellular homeostasis by turning over cellular components. Here we demonstrate a role for autophagy in hypothalamic agouti-related peptide (AgRP) neurons in the regulation of food intake and energy balance. We show that starvation-induced hypothalamic autophagy mobilizes neuron-intrinsic lipids to generate endogenous free fatty acids, which in turn regulate AgRP levels. The functional consequences of inhibiting autophagy are the failure to upregulate AgRP in response to starvation, and constitutive increases in hypothalamic levels of pro-opiomelanocortin and its cleavage product α-melanocyte-stimulating hormone that typically contribute to a lean phenotype. We propose a conceptual framework for considering how autophagy-regulated lipid metabolism within hypothalamic neurons may modulate neuropeptide levels to have immediate effects on food intake, as well as long-term effects on energy homeostasis. Regulation of hypothalamic autophagy could become an effective intervention in conditions such as obesity and the metabolic syndrome.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Autofagia/fisiología , Ingestión de Alimentos , Metabolismo Energético , Hipotálamo/metabolismo , Neuronas/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia , Células Cultivadas , Ácidos Grasos/biosíntesis , Hipotálamo/fisiología , Lípidos/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Inanición , alfa-MSH/biosíntesis
12.
J Neurosci ; 31(16): 6019-27, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21508227

RESUMEN

Nutrient excess in obesity and diabetes is emerging as a common putative cause for multiple deleterious effects across diverse cell types, responsible for a variety of metabolic dysfunctions. The hypothalamus is acknowledged as an important regulator of whole-body energy homeostasis, through both detection of nutrient availability and coordination of effectors that determine nutrient intake and utilization, thus preventing cellular and whole-body nutrient excess. However, the mechanisms underlying hypothalamic nutrient detection and its impact on peripheral nutrient utilization remain poorly understood. Recent data suggest a role for thioredoxin-interacting protein (TXNIP) as a molecular nutrient sensor important in the regulation of energy metabolism, but the role of hypothalamic TXNIP in the regulation of energy balance has not been evaluated. Here we show in mice that TXNIP is expressed in nutrient-sensing neurons of the mediobasal hypothalamus, responds to hormonal and nutrient signals, and regulates adipose tissue metabolism, fuel partitioning, and glucose homeostasis. Hypothalamic expression of TXNIP is induced by acute nutrient excess and in mouse models of obesity and diabetes, and downregulation of mediobasal hypothalamic TXNIP expression prevents diet-induced obesity and insulin resistance. Thus, mediobasal hypothalamic TXNIP plays a critical role in nutrient sensing and the regulation of fuel utilization.


Asunto(s)
Proteínas Portadoras/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Neuronas/metabolismo , Tiorredoxinas/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Animales , Western Blotting , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/fisiología , Proteínas Portadoras/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Ingestión de Energía/efectos de los fármacos , Glucosa/metabolismo , Glucosa/farmacología , Técnica de Clampeo de la Glucosa , Homeostasis/fisiología , Hipotálamo/fisiopatología , Inmunohistoquímica , Insulina/metabolismo , Insulina/farmacología , Resistencia a la Insulina , Leptina/metabolismo , Leptina/farmacología , Masculino , Ratones , Tiorredoxinas/genética
13.
PLoS One ; 6(2): e17058, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21379576

RESUMEN

MKR mice, lacking insulin-like growth factor 1 receptor (IGF-1R) signaling in skeletal muscle, are lean yet hyperlipidemic, hyperinsulinemic, and hyperglycemic, with severe insulin resistance and elevated hepatic and skeletal muscle levels of triglycerides. We have previously shown that chronic peripheral administration of the adipokine leptin improves hepatic insulin sensitivity in these mice independently of its effects on food intake. As central leptin signaling has been implicated in the control of peripheral glucose homeostasis, here we examined the ability of central intracerebroventricular leptin administration to affect energy balance and peripheral glucose homeostasis in non-obese diabetic male MKR mice. Central leptin significantly reduced food intake, body weight gain and adiposity, as well as serum glucose, insulin, leptin, free fatty acid and triglyceride levels relative to ACSF treated controls. These reductions were accompanied by increased fat oxidation as measured by indirect calorimetry, as well as increased oxygen consumption. Central leptin also improved glucose tolerance and hepatic insulin sensitivity determined using the euglycemic-hyperinsulinemic clamps relative to pair fed vehicle treated controls, as well as increasing the rate of glucose disappearance. Hepatic vagotomy only partially reversed the ability of central leptin to improve glucose tolerance. These results demonstrate that central leptin dramatically improves insulin sensitivity independently of its effects on food intake, in a lean mouse model of type 2 diabetes. The findings also suggest that: 1) both hepatic vagal and non-vagal pathways contribute to this improvement, and 2) central leptin alters glucose disposal in skeletal muscle in this model.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Leptina/administración & dosificación , Hígado/inervación , Nervio Vago/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Evaluación Preclínica de Medicamentos , Infusiones Intraventriculares , Leptina/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/fisiopatología , Masculino , Ratones , Ratones Noqueados , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Delgadez/metabolismo , Delgadez/patología , Nervio Vago/metabolismo , Nervio Vago/fisiología
14.
Behav Brain Res ; 209(1): 1-12, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20035790

RESUMEN

The hypothalamus is a center of convergence and integration of multiple nutrient-related signals. It can sense changes in circulating adiposity hormones, gastric hormones and nutrients, and receives neuroanatomical projections from other nutrient sensors, mainly within the brainstem. The hypothalamus also integrates these signals with various cognitive forebrain-descending information and reward/motivation-related signals coming from the midbrain-dopamine system, to coordinate neuroendocrine, behavioral and metabolic effectors of energy balance. Some of the key nutrient-sensing hypothalamic neurons have been identified in the arcuate, the ventro-medial and the lateral nuclei of the hypothalamus, and the molecular mechanisms underlying intracellular integration of nutrient-related signals in these neurons are currently under intensive investigation. However, little is known about the neural pathways downstream from hypothalamic nutrient sensors, and how they drive effectors of energy homeostasis under physiological conditions. This manuscript will review recent progress from molecular, genetic and neurophysiological studies that identify and characterize the critical intracellular signalling pathways and neurocircuits involved in determining hypothalamic nutrient detection, and link these circuits to behavioral and metabolic effectors of energy balance. We will provide a critical analysis of current data to identify ongoing challenges for future research in this field.


Asunto(s)
Metabolismo Energético/fisiología , Homeostasis/fisiología , Hipotálamo/fisiología , Fenómenos Fisiológicos de la Nutrición/fisiología , Tejido Adiposo/metabolismo , Animales , Conducta Alimentaria/fisiología , Glucosa/metabolismo , Humanos , Hipotálamo/citología , Modelos Biológicos , Red Nerviosa/fisiología , Vías Nerviosas/fisiología , Neuronas/fisiología
15.
J Neurosci ; 29(26): 8302-11, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19571121

RESUMEN

In response to nutrient stimuli, the mediobasal hypothalamus (MBH) drives multiple neuroendocrine and behavioral mechanisms to regulate energy balance. While central leucine reduces food intake and body weight, the specific neuroanatomical sites of leucine sensing, downstream neural substrates, and neurochemical effectors involved in this regulation remain largely unknown. Here we demonstrate that MBH leucine engages a neural energy regulatory circuit by stimulating POMC (proopiomelanocortin) neurons of the MBH, oxytocin neurons of the paraventricular hypothalamus, and neurons within the brainstem nucleus of the solitary tract to acutely suppress food intake by reducing meal size. We identify central p70 S6 kinase and Erk1/2 pathways as intracellular effectors required for this response. Activation of endogenous leucine intracellular metabolism produced longer-term reductions in meal number. Our data identify a novel, specific hypothalamus-brainstem circuit that links amino acid availability and nutrient sensing to the control of food intake.


Asunto(s)
Tronco Encefálico/fisiología , Ingestión de Alimentos/fisiología , Hipotálamo/metabolismo , Leucina/administración & dosificación , Animales , Anorexia/metabolismo , Peso Corporal/efectos de los fármacos , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Butadienos/farmacología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inhibidores Enzimáticos/farmacología , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Proteínas Fluorescentes Verdes/genética , Hipotálamo/anatomía & histología , Técnicas In Vitro , Inyecciones Intraventriculares/métodos , Cetoácidos/farmacología , Leucina/sangre , Leucina/líquido cefalorraquídeo , Masculino , Melanocortinas/metabolismo , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nitrilos/farmacología , Oxitocina/antagonistas & inhibidores , Oxitocina/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Treonina/metabolismo , Factores de Tiempo , Tirosina/metabolismo , Vasotocina/farmacología
16.
Cell Metab ; 8(6): 459-67, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19041762

RESUMEN

p70 S6 kinase 1 (S6K) is a major downstream effector of the mammalian target of rapamycin (mTOR), primarily implicated in the control of protein synthesis, cell growth, and proliferation. Here we demonstrate that specific bidirectional molecular targeting of mediobasal hypothalamic (MBH) S6K activity in rats is sufficient to significantly alter food intake, body weight, hypothalamic orexigenic neuropeptide expression, hypothalamic leptin sensitivity, and the metabolic and feeding responses to a fast. In addition, adenoviral-mediated constitutive activation of MBH S6K improved cold tolerance and protected against high-fat diet-induced overeating, fat deposition, and insulin resistance. Our results provide direct evidence that MBH S6K activity bidirectionally drives behavioral and metabolic determinants of energy balance and promote the assessment of MBH S6K activity as a therapeutic target in metabolic diseases.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/enzimología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Adenoviridae/genética , Adipocitos Marrones/metabolismo , Animales , Peso Corporal , Calorimetría , Frío , Ingestión de Alimentos , Ayuno , Leptina/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR , Factores de Tiempo
17.
J Clin Invest ; 118(8): 2959-68, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18618016

RESUMEN

Prolonged activation of p70 S6 kinase (S6K) by insulin and nutrients leads to inhibition of insulin signaling via negative feedback input to the signaling factor IRS-1. Systemic deletion of S6K protects against diet-induced obesity and enhances insulin sensitivity in mice. Herein, we present evidence suggesting that hypothalamic S6K activation is involved in the pathogenesis of diet-induced hepatic insulin resistance. Extending previous findings that insulin suppresses hepatic glucose production (HGP) partly via its effect in the hypothalamus, we report that this effect was blunted by short-term high-fat diet (HFD) feeding, with concomitant suppression of insulin signaling and activation of S6K in the mediobasal hypothalamus (MBH). Constitutive activation of S6K in the MBH mimicked the effect of the HFD in normal chow-fed animals, while suppression of S6K by overexpression of dominant-negative S6K or dominant-negative raptor in the MBH restored the ability of MBH insulin to suppress HGP after HFD feeding. These results suggest that activation of hypothalamic S6K contributes to hepatic insulin resistance in response to short-term nutrient excess.


Asunto(s)
Dieta , Hipotálamo/metabolismo , Resistencia a la Insulina , Hígado/fisiología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Adenoviridae/genética , Animales , Activación Enzimática , Masculino , Ratas , Ratas Sprague-Dawley
18.
Nat Med ; 14(6): 667-75, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18516053

RESUMEN

Leptin (encoded by Lep) controls body weight by regulating food intake and fuel partitioning. Obesity is characterized by leptin resistance and increased endocannabinoid tone. Here we show that leptin infused into the mediobasal hypothalamus (MBH) of rats inhibits white adipose tissue (WAT) lipogenesis, which occurs independently of signal transducer and activator of transcription-3 (STAT3) signaling. Correspondingly, transgenic inactivation of STAT3 signaling by mutation of the leptin receptor (s/s mice) leads to reduced adipose mass compared to db/db mice (complete abrogation of leptin receptor signaling). Conversely, the ability of hypothalamic leptin to suppress WAT lipogenesis in rats is lost when hypothalamic phosphoinositide 3-kinase signaling is prevented or when sympathetic denervation of adipose tissue is performed. MBH leptin suppresses the endocannabinoid anandamide in WAT, and, when this suppression of endocannabinoid tone is prevented by systemic CB1 receptor activation, MBH leptin fails to suppress WAT lipogenesis. These data suggest that the increased endocannabinoid tone observed in obesity is linked to a failure of central leptin signaling to restrain peripheral endocannabinoids.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Leptina/metabolismo , Lipogénesis , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Glucemia/análisis , Moduladores de Receptores de Cannabinoides/análisis , Catecolaminas/análisis , Hipotálamo/metabolismo , Infusiones Intravenosas , Insulina/sangre , Leptina/administración & dosificación , Leptina/fisiología , Masculino , Modelos Biológicos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
19.
Diabetes ; 57(8): 2061-5, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18511848

RESUMEN

OBJECTIVE: A selective rise in hypothalamic lipid metabolism and the subsequent activation of SUR1/Kir6.2 ATP-sensitive K(+) (K(ATP)) channels inhibit hepatic glucose production. The mechanisms that link the ability of hypothalamic lipid metabolism to the activation of K(ATP) channels remain unknown. RESEARCH DESIGN AND METHODS: To examine whether hypothalamic protein kinase C (PKC) mediates the ability of central nervous system lipids to activate K(ATP) channels and regulate glucose production in normal rodents, we first activated hypothalamic PKC in the absence or presence of K(ATP) channel inhibition. We then inhibited hypothalamic PKC in the presence of lipids. Tracer-dilution methodology in combination with the pancreatic clamp technique was used to assess the effect of hypothalamic administrations on glucose metabolism in vivo. RESULTS: We first reported that direct activation of hypothalamic PKC via direct hypothalamic delivery of PKC activator 1-oleoyl-2-acetyl-sn-glycerol (OAG) suppressed glucose production. Coadministration of hypothalamic PKC-delta inhibitor rottlerin with OAG prevented the ability of OAG to activate PKC-delta and lower glucose production. Furthermore, hypothalamic dominant-negative Kir6.2 expression or the delivery of the K(ATP) channel blocker glibenclamide abolished the glucose production-lowering effects of OAG. Finally, inhibition of hypothalamic PKC eliminated the ability of lipids to lower glucose production. CONCLUSIONS: These studies indicate that hypothalamic PKC activation is sufficient and necessary for lowering glucose production.


Asunto(s)
Glucosa/biosíntesis , Hipotálamo/metabolismo , Proteína Quinasa C/metabolismo , Acetofenonas/administración & dosificación , Acetofenonas/farmacología , Animales , Benzopiranos/administración & dosificación , Benzopiranos/farmacología , Diglicéridos/administración & dosificación , Diglicéridos/farmacología , Activación Enzimática/efectos de los fármacos , Gliburida/administración & dosificación , Gliburida/farmacología , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/enzimología , Canales KATP/antagonistas & inhibidores , Masculino , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
20.
Nat Neurosci ; 8(5): 579-84, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15856066

RESUMEN

Selective regions of the brain, including the hypothalamus, are capable of gathering information on the body's nutritional status in order to implement appropriate behavioral and metabolic responses to changes in fuel availability. This review focuses on direct metabolic signaling within the hypothalamus. There is growing evidence supporting the idea that fatty acid metabolism within discrete hypothalamic regions can function as a sensor for nutrient availability that can integrate multiple nutritional and hormonal signals.


Asunto(s)
Regulación del Apetito/fisiología , Metabolismo Energético/fisiología , Ácidos Grasos/metabolismo , Retroalimentación Fisiológica/fisiología , Hipotálamo/fisiología , Acilcoenzima A/metabolismo , Animales , Humanos , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Obesidad/fisiopatología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA