Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell Rep ; 41(13): 111894, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36577374

RESUMEN

Paradoxically, glucose, the primary driver of satiety, activates a small population of anorexigenic pro-opiomelanocortin (POMC) neurons. Here, we show that lactate levels in the circulation and in the cerebrospinal fluid are elevated in the fed state and the addition of lactate to glucose activates the majority of POMC neurons while increasing cytosolic NADH generation, mitochondrial respiration, and extracellular pyruvate levels. Inhibition of lactate dehydrogenases diminishes mitochondrial respiration, NADH production, and POMC neuronal activity. However, inhibition of the mitochondrial pyruvate carrier has no effect. POMC-specific downregulation of Ucp2 (Ucp2PomcKO), a molecule regulated by fatty acid metabolism and shown to play a role as transporter in the malate-aspartate shuttle, abolishes lactate- and glucose-sensing of POMC neurons. Ucp2PomcKO mice have impaired glucose metabolism and are prone to obesity on a high-fat diet. Altogether, our data show that lactate through redox signaling and blocking mitochondrial glucose utilization activates POMC neurons to regulate feeding and glucose metabolism.


Asunto(s)
NAD , Proopiomelanocortina , Ratones , Animales , Proopiomelanocortina/metabolismo , NAD/metabolismo , Glucosa/metabolismo , Neuronas/metabolismo , Lactatos/metabolismo , Hipotálamo/metabolismo , Proteína Desacopladora 2/metabolismo
2.
J Clin Invest ; 132(19)2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-36189793

RESUMEN

The TET family of dioxygenases promote DNA demethylation by oxidizing 5-methylcytosine to 5-hydroxymethylcytosine (5hmC). Hypothalamic agouti-related peptide-expressing (AGRP-expressing) neurons play an essential role in driving feeding, while also modulating nonfeeding behaviors. Besides AGRP, these neurons produce neuropeptide Y (NPY) and the neurotransmitter GABA, which act in concert to stimulate food intake and decrease energy expenditure. Notably, AGRP, NPY, and GABA can also elicit anxiolytic effects. Here, we report that in adult mouse AGRP neurons, CRISPR-mediated genetic ablation of Tet3, not previously known to be involved in central control of appetite and metabolism, induced hyperphagia, obesity, and diabetes, in addition to a reduction of stress-like behaviors. TET3 deficiency activated AGRP neurons, simultaneously upregulated the expression of Agrp, Npy, and the vesicular GABA transporter Slc32a1, and impeded leptin signaling. In particular, we uncovered a dynamic association of TET3 with the Agrp promoter in response to leptin signaling, which induced 5hmC modification that was associated with a chromatin-modifying complex leading to transcription inhibition, and this regulation occurred in both the mouse models and human cells. Our results unmasked TET3 as a critical central regulator of appetite and energy metabolism and revealed its unexpected dual role in the control of feeding and other complex behaviors through AGRP neurons.


Asunto(s)
Ansiolíticos , Dioxigenasas , 5-Metilcitosina/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Ansiolíticos/farmacología , Cromatina/metabolismo , Dioxigenasas/genética , Dioxigenasas/metabolismo , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Ratones , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Ácido gamma-Aminobutírico/genética , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología
3.
Sci Adv ; 8(35): eabn8092, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36044565

RESUMEN

The ventromedial hypothalamus (VMH) is known to regulate body weight and counterregulatory response. However, how VMH neurons regulate lipid metabolism and energy balance remains unknown. O-linked ß-d-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation), catalyzed by O-GlcNAc transferase (OGT), is considered a cellular sensor of nutrients and hormones. Here, we report that genetic ablation of OGT in VMH neurons inhibits neuronal excitability. Mice with VMH neuron-specific OGT deletion show rapid weight gain, increased adiposity, and reduced energy expenditure, without significant changes in food intake or physical activity. The obesity phenotype is associated with adipocyte hypertrophy and reduced lipolysis of white adipose tissues. In addition, OGT deletion in VMH neurons down-regulates the sympathetic activity and impairs the sympathetic innervation of white adipose tissues. These findings identify OGT in the VMH as a homeostatic set point that controls body weight and underscore the importance of the VMH in regulating lipid metabolism through white adipose tissue-specific innervation.


Asunto(s)
Lipólisis , N-Acetilglucosaminiltransferasas , Obesidad , Tejido Adiposo/metabolismo , Animales , Peso Corporal , Hipotálamo/metabolismo , Lipólisis/genética , Ratones , Obesidad/genética , Obesidad/metabolismo
4.
Mol Psychiatry ; 27(10): 3951-3960, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35906488

RESUMEN

Hypothalamic agouti-related peptide and neuropeptide Y-expressing (AgRP) neurons have a critical role in both feeding and non-feeding behaviors of newborn, adolescent, and adult mice, suggesting their broad modulatory impact on brain functions. Here we show that constitutive impairment of AgRP neurons or their peripubertal chemogenetic inhibition resulted in both a numerical and functional reduction of neurons in the medial prefrontal cortex (mPFC) of mice. These changes were accompanied by alteration of oscillatory network activity in mPFC, impaired sensorimotor gating, and altered ambulatory behavior that could be reversed by the administration of clozapine, a non-selective dopamine receptor antagonist. The observed AgRP effects are transduced to mPFC in part via dopaminergic neurons in the ventral tegmental area and may also be conveyed by medial thalamic neurons. Our results unmasked a previously unsuspected role for hypothalamic AgRP neurons in control of neuronal pathways that regulate higher-order brain functions during development and in adulthood.


Asunto(s)
Hipotálamo , Neuropéptido Y , Animales , Ratones , Proteína Relacionada con Agouti/metabolismo , Neuronas Dopaminérgicas/metabolismo , Hipotálamo/metabolismo , Neuropéptido Y/metabolismo , Corteza Prefrontal/metabolismo
5.
J Clin Invest ; 131(10)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33848272

RESUMEN

Hypothalamic feeding circuits have been identified as having innate synaptic plasticity, mediating adaption to the changing metabolic milieu by controlling responses to feeding and obesity. However, less is known about the regulatory principles underlying the dynamic changes in agouti-related protein (AgRP) perikarya, a region crucial for gating of neural excitation and, hence, feeding. Here we show that AgRP neurons activated by food deprivation, ghrelin administration, or chemogenetics decreased their own inhibitory tone while triggering mitochondrial adaptations in neighboring astrocytes. We found that it was the inhibitory neurotransmitter GABA released by AgRP neurons that evoked this astrocytic response; this in turn resulted in increased glial ensheetment of AgRP perikarya by glial processes and increased excitability of AgRP neurons. We also identified astrocyte-derived prostaglandin E2, which directly activated - via EP2 receptors - AgRP neurons. Taken together, these observations unmasked a feed-forward, self-exciting loop in AgRP neuronal control mediated by astrocytes, a mechanism directly relevant for hunger, feeding, and overfeeding.


Asunto(s)
Proteína Relacionada con Agouti , Astrocitos/metabolismo , Hambre , Hipotálamo/metabolismo , Plasticidad Neuronal , Neuronas/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Masculino , Ratones , Ratones Transgénicos , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo
6.
J Clin Invest ; 130(9): 4985-4998, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32516139

RESUMEN

The brain has evolved in an environment where food sources are scarce, and foraging for food is one of the major challenges for survival of the individual and species. Basic and clinical studies show that obesity or overnutrition leads to overwhelming changes in the brain in animals and humans. However, the exact mechanisms underlying the consequences of excessive energy intake are not well understood. Neurons expressing the neuropeptide hypocretin/orexin (Hcrt) in the lateral/perifonical hypothalamus (LH) are critical for homeostatic regulation, reward seeking, stress response, and cognitive functions. In this study, we examined adaptations in Hcrt cells regulating behavioral responses to salient stimuli in diet-induced obese mice. Our results demonstrated changes in primary cilia, synaptic transmission and plasticity, cellular responses to neurotransmitters necessary for reward seeking, and stress responses in Hcrt neurons from obese mice. Activities of neuronal networks in the LH and hippocampus were impaired as a result of decreased hypocretinergic function. The weakened Hcrt system decreased reward seeking while altering responses to acute stress (stress-coping strategy), which were reversed by selectively activating Hcrt cells with chemogenetics. Taken together, our data suggest that a deficiency in Hcrt signaling may be a common cause of behavioral changes (such as lowered arousal, weakened reward seeking, and altered stress response) in obese animals.


Asunto(s)
Conducta Alimentaria , Hipotálamo , Red Nerviosa , Neuronas , Obesidad , Orexinas , Animales , Hipotálamo/metabolismo , Hipotálamo/patología , Hipotálamo/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Red Nerviosa/metabolismo , Red Nerviosa/patología , Red Nerviosa/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Obesidad/fisiopatología , Orexinas/genética , Orexinas/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología
7.
Cell ; 159(2): 306-17, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25303527

RESUMEN

Induction of beige cells causes the browning of white fat and improves energy metabolism. However, the central mechanism that controls adipose tissue browning and its physiological relevance are largely unknown. Here, we demonstrate that fasting and chemical-genetic activation of orexigenic AgRP neurons in the hypothalamus suppress the browning of white fat. O-linked ß-N-acetylglucosamine (O-GlcNAc) modification of cytoplasmic and nuclear proteins regulates fundamental cellular processes. The levels of O-GlcNAc transferase (OGT) and O-GlcNAc modification are enriched in AgRP neurons and are elevated by fasting. Genetic ablation of OGT in AgRP neurons inhibits neuronal excitability through the voltage-dependent potassium channel, promotes white adipose tissue browning, and protects mice against diet-induced obesity and insulin resistance. These data reveal adipose tissue browning as a highly dynamic physiological process under central control, in which O-GlcNAc signaling in AgRP neurons is essential for suppressing thermogenesis to conserve energy in response to fasting.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Dieta , N-Acetilglucosaminiltransferasas/metabolismo , Neuronas/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Ayuno , Femenino , Ghrelina/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Resistencia a la Insulina , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , N-Acetilglucosaminiltransferasas/genética , Obesidad/metabolismo , Obesidad/prevención & control
8.
Nat Neurosci ; 17(7): 908-10, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24880214

RESUMEN

We found that leptin receptors were expressed in hypothalamic astrocytes and that their conditional deletion led to altered glial morphology and synaptic inputs onto hypothalamic neurons involved in feeding control. Leptin-regulated feeding was diminished, whereas feeding after fasting or ghrelin administration was elevated in mice with astrocyte-specific leptin receptor deficiency. These data reveal an active role of glial cells in hypothalamic synaptic remodeling and control of feeding by leptin.


Asunto(s)
Astrocitos/fisiología , Ingestión de Alimentos/fisiología , Hipotálamo/fisiología , Leptina/fisiología , Red Nerviosa/fisiología , Neuronas/fisiología , Transducción de Señal/fisiología , Animales , Recuento de Células , Potenciales Postsinápticos Excitadores/fisiología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipotálamo/citología , Inmunohistoquímica , Hibridación in Situ , Leptina/genética , Masculino , Melanocortinas/fisiología , Ratones , Ratones Noqueados , Microscopía Electrónica , Cultivo Primario de Células , Proopiomelanocortina/fisiología , Intercambio Gaseoso Pulmonar/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
J Physiol ; 591(7): 1951-66, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23318871

RESUMEN

Hypocretin (orexin), a neuropeptide synthesized exclusively in the perifornical/lateral hypothalamus, is critical for drug seeking and relapse, but it is not clear how the circuitry centred on hypocretin-producing neurons (hypocretin neurons) is modified by drugs of abuse and how changes in this circuit might alter behaviours related to drug addiction. In this study, we show that repeated, but not single, in vivo cocaine administration leads to a long-lasting, experience-dependent potentiation of glutamatergic synapses on hypocretin neurons in mice following a cocaine-conditioned place preference (CPP) protocol. The synaptic potentiation occurs postsynaptically and probably involves up-regulation of AMPA-type glutamate receptors on hypocretin neurons. Phosphorylation of cAMP response element-binding protein (CREB) is also significantly increased in hypocretin neurons in cocaine-treated animals, suggesting that CREB-mediated pathways may contribute to synaptic potentiation in these cells. Furthermore, the potentiation of synaptic efficacy in hypocretin neurons persists during cocaine withdrawal, but reverses to baseline levels after prolonged abstinence. Finally, the induction of long-term potentiation (LTP) triggered by a high-frequency stimulation is facilitated in hypocretin neurons in cocaine-treated mice, suggesting that long-lasting changes in synapses onto hypocretin neurons would probably be further potentiated by other stimuli (such as concurrent environmental cues) paired with the drug. In summary, we show here that hypocretin neurons undergo experience-dependent synaptic potentiation that is distinct from that reported in other reward systems, such as the ventral tegmental area, following exposure to cocaine. These findings support the idea that the hypocretin system is important for behavioural changes associated with cocaine administration in animals and humans.


Asunto(s)
Cocaína/administración & dosificación , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neuronas/efectos de los fármacos , Neuropéptidos/fisiología , Sinapsis/efectos de los fármacos , Animales , Condicionamiento Psicológico , Potenciales Postsinápticos Excitadores , Hipotálamo/fisiología , Potenciación a Largo Plazo , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Neuronas/fisiología , Orexinas , Sinapsis/fisiología
10.
Elife ; 2: e01462, 2013 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-24381247

RESUMEN

Sugars that contain glucose, such as sucrose, are generally preferred to artificial sweeteners owing to their post-ingestive rewarding effect, which elevates striatal dopamine (DA) release. While the post-ingestive rewarding effect, which artificial sweeteners do not have, signals the nutrient value of sugar and influences food preference, the neural circuitry that mediates the rewarding effect of glucose is unknown. In this study, we show that optogenetic activation of melanin-concentrating hormone (MCH) neurons during intake of the artificial sweetener sucralose increases striatal dopamine levels and inverts the normal preference for sucrose vs sucralose. Conversely, animals with ablation of MCH neurons no longer prefer sucrose to sucralose and show reduced striatal DA release upon sucrose ingestion. We further show that MCH neurons project to reward areas and are required for the post-ingestive rewarding effect of sucrose in sweet-blind Trpm5(-/-) mice. These studies identify an essential component of the neural pathways linking nutrient sensing and food reward. DOI: http://dx.doi.org/10.7554/eLife.01462.001.


Asunto(s)
Glucosa/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Valor Nutritivo , Animales , Hipotálamo/química , Ratones , Recompensa
11.
J Clin Invest ; 122(11): 3900-13, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23064363

RESUMEN

Glial cells perform critical functions that alter the metabolism and activity of neurons, and there is increasing interest in their role in appetite and energy balance. Leptin, a key regulator of appetite and metabolism, has previously been reported to influence glial structural proteins and morphology. Here, we demonstrate that metabolic status and leptin also modify astrocyte-specific glutamate and glucose transporters, indicating that metabolic signals influence synaptic efficacy and glucose uptake and, ultimately, neuronal function. We found that basal and glucose-stimulated electrical activity of hypothalamic proopiomelanocortin (POMC) neurons in mice were altered in the offspring of mothers fed a high-fat diet. In adulthood, increased body weight and fasting also altered the expression of glucose and glutamate transporters. These results demonstrate that whole-organism metabolism alters hypothalamic glial cell activity and suggest that these cells play an important role in the pathology of obesity.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Astrocitos/metabolismo , Grasas de la Dieta/efectos adversos , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Animales , Astrocitos/patología , Grasas de la Dieta/farmacología , Hipotálamo/patología , Ratones , Neuronas/metabolismo , Neuronas/patología , Obesidad/inducido químicamente , Obesidad/patología , Proopiomelanocortina/metabolismo , Ratas , Ratas Wistar
12.
Nat Med ; 17(9): 1121-7, 2011 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-21873987

RESUMEN

Previous studies have proposed roles for hypothalamic reactive oxygen species (ROS) in the modulation of circuit activity of the melanocortin system. Here we show that suppression of ROS diminishes pro-opiomelanocortin (POMC) cell activation and promotes the activity of neuropeptide Y (NPY)- and agouti-related peptide (AgRP)-co-producing (NPY/AgRP) neurons and feeding, whereas ROS-activates POMC neurons and reduces feeding. The levels of ROS in POMC neurons were positively correlated with those of leptin in lean and ob/ob mice, a relationship that was diminished in diet-induced obese (DIO) mice. High-fat feeding resulted in proliferation of peroxisomes and elevated peroxisome proliferator-activated receptor γ (PPAR-γ) mRNA levels within the hypothalamus. The proliferation of peroxisomes in POMC neurons induced by the PPAR-γ agonist rosiglitazone decreased ROS levels and increased food intake in lean mice on high-fat diet. Conversely, the suppression of peroxisome proliferation by the PPAR antagonist GW9662 increased ROS concentrations and c-fos expression in POMC neurons. Also, it reversed high-fat feeding-triggered elevated NPY/AgRP and low POMC neuronal firing, and resulted in decreased feeding of DIO mice. Finally, central administration of ROS alone increased c-fos and phosphorylated signal transducer and activator of transcription 3 (pStat3) expression in POMC neurons and reduced feeding of DIO mice. These observations unmask a previously unknown hypothalamic cellular process associated with peroxisomes and ROS in the central regulation of energy metabolism in states of leptin resistance.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Neuronas/metabolismo , PPAR gamma/metabolismo , Peroxisomas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Proteína Relacionada con Agouti/metabolismo , Anilidas/farmacología , Animales , Línea Celular , Ingestión de Alimentos/fisiología , Electrofisiología , Proteínas Fluorescentes Verdes , Hipotálamo/citología , Ratones , Ratones Obesos , Neuropéptido Y/metabolismo , PPAR gamma/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa , Proopiomelanocortina/metabolismo
13.
Cell ; 138(5): 976-89, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19737523

RESUMEN

Leptin inhibition of bone mass accrual requires the integrity of specific hypothalamic neurons but not expression of its receptor on these neurons. The same is true for its regulation of appetite and energy expenditure. This suggests that leptin acts elsewhere in the brain to achieve these three functions. We show here that brainstem-derived serotonin (BDS) favors bone mass accrual following its binding to Htr2c receptors on ventromedial hypothalamic neurons and appetite via Htr1a and 2b receptors on arcuate neurons. Leptin inhibits these functions and increases energy expenditure because it reduces serotonin synthesis and firing of serotonergic neurons. Accordingly, while abrogating BDS synthesis corrects the bone, appetite and energy expenditure phenotypes caused by leptin deficiency, inactivation of the leptin receptor in serotonergic neurons recapitulates them fully. This study modifies the map of leptin signaling in the brain and identifies a molecular basis for the common regulation of bone and energy metabolisms. For a video summary of this article, see the PaperFlick file with the Supplemental Data available online.


Asunto(s)
Apetito , Densidad Ósea , Metabolismo Energético , Leptina/metabolismo , Serotonina/metabolismo , Tronco Encefálico/metabolismo , Hipotálamo/metabolismo , Receptores de Leptina/metabolismo , Transducción de Señal
14.
J Clin Invest ; 119(8): 2291-303, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19620781

RESUMEN

The anorexigenic neuromodulator alpha-melanocyte-stimulating hormone (alpha-MSH; referred to here as alpha-MSH1-13) undergoes extensive posttranslational processing, and its in vivo activity is short lived due to rapid inactivation. The enzymatic control of alpha-MSH1-13 maturation and inactivation is incompletely understood. Here we have provided insight into alpha-MSH1-13 inactivation through the generation and analysis of a subcongenic mouse strain with reduced body fat compared with controls. Using positional cloning, we identified a maximum of 6 coding genes, including that encoding prolylcarboxypeptidase (PRCP), in the donor region. Real-time PCR revealed a marked genotype effect on Prcp mRNA expression in brain tissue. Biochemical studies using recombinant PRCP demonstrated that PRCP removes the C-terminal amino acid of alpha-MSH1-13, producing alpha-MSH1-12, which is not neuroactive. We found that Prcp was expressed in the hypothalamus in neuronal populations that send efferents to areas where alpha-MSH1-13 is released from axon terminals. The inhibition of PRCP activity by small molecule protease inhibitors administered peripherally or centrally decreased food intake in both wild-type and obese mice. Furthermore, Prcp-null mice had elevated levels of alpha-MSH1-13 in the hypothalamus and were leaner and shorter than the wild-type controls on a regular chow diet; they were also resistant to high-fat diet-induced obesity. Our results suggest that PRCP is an important component of melanocortin signaling and weight maintenance via control of active alpha-MSH1-13 levels.


Asunto(s)
Carboxipeptidasas/fisiología , Ingestión de Alimentos , alfa-MSH/antagonistas & inhibidores , Animales , Carboxipeptidasas/antagonistas & inhibidores , Carboxipeptidasas/genética , Ingestión de Alimentos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Hipotálamo/metabolismo , Masculino , Hormonas Estimuladoras de los Melanocitos/metabolismo , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Endogámicos BALB C , Obesidad/etiología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Reacción en Cadena de la Polimerasa , Pirimidinas/farmacología , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptores de Melanocortina/fisiología , alfa-MSH/fisiología
15.
Nature ; 454(7206): 846-51, 2008 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-18668043

RESUMEN

The gut-derived hormone ghrelin exerts its effect on the brain by regulating neuronal activity. Ghrelin-induced feeding behaviour is controlled by arcuate nucleus neurons that co-express neuropeptide Y and agouti-related protein (NPY/AgRP neurons). However, the intracellular mechanisms triggered by ghrelin to alter NPY/AgRP neuronal activity are poorly understood. Here we show that ghrelin initiates robust changes in hypothalamic mitochondrial respiration in mice that are dependent on uncoupling protein 2 (UCP2). Activation of this mitochondrial mechanism is critical for ghrelin-induced mitochondrial proliferation and electric activation of NPY/AgRP neurons, for ghrelin-triggered synaptic plasticity of pro-opiomelanocortin-expressing neurons, and for ghrelin-induced food intake. The UCP2-dependent action of ghrelin on NPY/AgRP neurons is driven by a hypothalamic fatty acid oxidation pathway involving AMPK, CPT1 and free radicals that are scavenged by UCP2. These results reveal a signalling modality connecting mitochondria-mediated effects of G-protein-coupled receptors on neuronal function and associated behaviour.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Ghrelina/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ghrelina/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Canales Iónicos/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Proteínas Mitocondriales/genética , Neuronas/efectos de los fármacos , Neuropéptido Y/genética , Fosforilación/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Proteína Desacopladora 2
16.
J Clin Invest ; 117(12): 4022-33, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18060037

RESUMEN

Sleep is a natural process that preserves energy, facilitates development, and restores the nervous system in higher animals. Sleep loss resulting from physiological and pathological conditions exerts tremendous pressure on neuronal circuitry responsible for sleep-wake regulation. It is not yet clear how acute and chronic sleep loss modify neuronal activities and lead to adaptive changes in animals. Here, we show that acute and chronic prolonged wakefulness in mice induced by modafinil treatment produced long-term potentiation (LTP) of glutamatergic synapses on hypocretin/orexin neurons in the lateral hypothalamus, a well-established arousal/wake-promoting center. A similar potentiation of synaptic strength at glutamatergic synapses on hypocretin/orexin neurons was also seen when mice were sleep deprived for 4 hours by gentle handling. Blockade of dopamine D1 receptors attenuated prolonged wakefulness and synaptic plasticity in these neurons, suggesting that modafinil functions through activation of the dopamine system. Also, activation of the cAMP pathway was not able to further induce LTP at glutamatergic synapses in brain slices from mice treated with modafinil. These results indicate that synaptic plasticity due to prolonged wakefulness occurs in circuits responsible for arousal and may contribute to changes in the brain and body of animals experiencing sleep loss.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Plasticidad Neuronal , Neuronas/metabolismo , Neuropéptidos , Privación de Sueño/metabolismo , Sinapsis/metabolismo , Vigilia , Animales , Compuestos de Bencidrilo/efectos adversos , Compuestos de Bencidrilo/farmacología , Estimulantes del Sistema Nervioso Central/efectos adversos , Estimulantes del Sistema Nervioso Central/farmacología , AMP Cíclico/metabolismo , Dopamina/metabolismo , Femenino , Hipotálamo/metabolismo , Hipotálamo/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Potenciación a Largo Plazo , Masculino , Ratones , Modafinilo , Plasticidad Neuronal/efectos de los fármacos , Neuronas/patología , Neuropéptidos/metabolismo , Orexinas , Receptores de Dopamina D1/metabolismo , Privación de Sueño/inducido químicamente , Privación de Sueño/patología , Sinapsis/patología , Vigilia/efectos de los fármacos
17.
Cell Metab ; 5(1): 21-33, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17189204

RESUMEN

The active thyroid hormone, triiodothyronine (T3), regulates mitochondrial uncoupling protein activity and related thermogenesis in peripheral tissues. Type 2 deiodinase (DII), an enzyme that catalyzes active thyroid hormone production, and mitochondrial uncoupling protein 2 (UCP2) are also present in the hypothalamic arcuate nucleus, where their interaction and physiological significance have not been explored. Here, we report that DII-producing glial cells are in direct apposition to neurons coexpressing neuropeptide Y (NPY), agouti-related protein (AgRP), and UCP2. Fasting increased DII activity and local thyroid hormone production in the arcuate nucleus in parallel with increased GDP-regulated UCP2-dependent mitochondrial uncoupling. Fasting-induced T3-mediated UCP2 activation resulted in mitochondrial proliferation in NPY/AgRP neurons, an event that was critical for increased excitability of these orexigenic neurons and consequent rebound feeding following food deprivation. These results reveal a physiological role for a thyroid-hormone-regulated mitochondrial uncoupling in hypothalamic neuronal networks.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Ayuno , Conducta Alimentaria , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Termogénesis , Triyodotironina/metabolismo , Proteína Relacionada con Agouti , Animales , Núcleo Arqueado del Hipotálamo/citología , Ingestión de Alimentos , Proteínas Fluorescentes Verdes , Guanosina Difosfato/metabolismo , Hipotálamo/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Yoduro Peroxidasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Neuroglía/metabolismo , Neuropéptido Y/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteína Desacopladora 2 , Yodotironina Deyodinasa Tipo II
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA