Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Am J Physiol Endocrinol Metab ; 318(5): E655-E666, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32045262

RESUMEN

Excessive alcohol consumption, including binge drinking, is a common cause of fatty liver disease. Binge drinking rapidly induces hepatic steatosis, an early step in the pathogenesis of chronic liver injury. Despite its prevalence, the process by which excessive alcohol consumption promotes hepatic lipid accumulation remains unclear. Alcohol exerts potent effects on the brain, including hypothalamic neurons crucial for metabolic regulation. However, whether or not the brain plays a role in alcohol-induced hepatic steatosis is unknown. In the brain, alcohol increases extracellular levels of adenosine, a potent neuromodulator, and previous work implicates adenosine signaling as being important for the development of alcoholic fatty liver disease. Acute alcohol exposure also increases both the activity of agouti-related protein (AgRP)-expressing neurons and AgRP immunoreactivity. Here, we show that adenosine receptor A2B signaling in the brain modulates the extent of alcohol-induced fatty liver in mice and that both the AgRP neuropeptide and the sympathetic nervous system are indispensable for hepatic steatosis induced by bingelike alcohol consumption. Together, these results indicate that the brain plays an integral role in alcohol-induced hepatic lipid accumulation and that central adenosine signaling, hypothalamic AgRP, and the sympathetic nervous system are crucial mediators of this process.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Hígado Graso Alcohólico/metabolismo , Hipotálamo/metabolismo , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Neuronas/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Masculino , Ratones
2.
Annu Rev Physiol ; 77: 131-60, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25668019

RESUMEN

Diet-induced obesity leads to devastating and common chronic diseases, fueling ongoing interest in determining new mechanisms underlying both obesity and its consequences. It is now well known that chronic overnutrition produces a unique form of inflammation in peripheral insulin target tissues, and efforts to limit this inflammation have met with some success in preserving insulin sensitivity in obese individuals. Recently, the activation of inflammatory pathways by dietary excess has also been observed among cells located in the mediobasal hypothalamus, a brain area that exerts central control over peripheral glucose, fat, and energy metabolism. Here we review progress in the field of diet-induced hypothalamic inflammation, drawing key distinctions between metabolic inflammation in the hypothalamus and that occurring in peripheral tissues. We focus on specific stimuli of the inflammatory response, the roles of individual hypothalamic cell types, and the links between hypothalamic inflammation and metabolic function under normal and pathophysiological circumstances. Finally, we explore the concept of controlling hypothalamic inflammation to mitigate metabolic disease.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/fisiopatología , Inflamación/fisiopatología , Metabolismo/fisiología , Animales , Dieta/efectos adversos , Humanos , Hipotálamo/patología , Enfermedades Metabólicas/fisiopatología , Microglía/fisiología , Obesidad/etiología , Obesidad/fisiopatología
3.
Proc Natl Acad Sci U S A ; 110(8): E697-706, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23386726

RESUMEN

Chronic consumption of a fat-rich diet leads to attenuation of leptin signaling in hypothalamic neurons, a hallmark feature of cellular leptin resistance. To date, little is known about the temporal and spatial dysregulation of neuronal function under conditions of nutrient excess. We show that agouti-related protein (AgRP)-expressing neurons precede proopiomelanocortin neurons in developing diet-induced cellular leptin resistance. High-fat diet-induced up-regulation of suppressor of cytokine signaling-3 (SOCS3) occurs in AgRP neurons before proopiomelanocortin and other hypothalamic neurons. SOCS3 expression in AgRP neurons increases after 2 d of high-fat feeding, but reduces after switching to a low-fat diet for 1 d. Consistently, transgenic overexpression of SOCS3 in AgRP neurons produces metabolic phenotypes resembling those observed after short-term high-fat feeding. We further show that AgRP neurons are the predominant cell type situated outside the blood-brain barrier in the mediobasal hypothalamus. AgRP neurons are more responsive to low levels of circulating leptin, but they are also more prone to development of leptin resistance in response to a small increase in blood leptin concentrations. Collectively, these results suggest that AgRP neurons are able to sense slight changes in plasma metabolic signals, allowing them to serve as first-line responders to fluctuation of energy intake. Furthermore, modulation of SOCS3 expression in AgRP neurons may play a dynamic and physiological role in metabolic fine tuning in response to short-term changes of nutritional status.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Hipotálamo/fisiopatología , Leptina/fisiología , Neuronas/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Animales , Hipotálamo/citología , Ratones , Neuronas/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Regulación hacia Arriba
4.
J Neurosci ; 33(29): 11972-85, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23864684

RESUMEN

Like obesity, prolonged food deprivation induces severe hepatic steatosis; however, the functional significance of this phenomenon is not well understood. In this study, we show that the fall in plasma leptin concentration during fasting is required for the development of hepatic steatosis in mice. Removal of leptin receptors from AGRP neurons diminishes fasting-induced hepatic steatosis. Furthermore, the suppressive effects of leptin on fasting-induced hepatic steatosis are absent in mice lacking the gene encoding agouti-related protein (Agrp), suggesting that this function of leptin is mediated by AGRP. Prolonged fasting leads to suppression of hepatic sympathetic activity, increased expression of acyl CoA:diacylglycerol acyltransferase-2 in the liver, and elevation of hepatic triglyceride content and all of these effects are blunted in the absence of AGRP. AGRP deficiency, despite having no effects on feeding or body adiposity in the free-fed state, impairs triglyceride and ketone body release from the liver during prolonged fasting. Furthermore, reducing CNS Agrp expression in wild-type mice by RNAi protected against the development of hepatic steatosis not only during starvation, but also in response to consumption of a high-fat diet. These findings identify the leptin-AGRP circuit as a critical modulator of hepatic triglyceride stores in starvation and suggest a vital role for this circuit in sustaining the supply of energy from the liver to extrahepatic tissues during periods of prolonged food deprivation.


Asunto(s)
Proteína Relacionada con Agouti/genética , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Hígado/metabolismo , Receptores de Leptina/genética , Acilcoenzima A/genética , Acilcoenzima A/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Composición Corporal/efectos de los fármacos , Composición Corporal/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Metabolismo Energético/efectos de los fármacos , Hígado Graso/genética , Hígado Graso/metabolismo , Privación de Alimentos/fisiología , Hipotálamo/efectos de los fármacos , Leptina/farmacología , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Norepinefrina/metabolismo , Obesidad/genética , Obesidad/metabolismo , Receptores de Leptina/metabolismo
5.
Mol Metab ; 80: 101886, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38246589

RESUMEN

OBJECTIVE: The central melanocortin system is essential for the regulation of food intake and body weight. Agouti-related protein (AgRP) is the sole orexigenic component of the central melanocortin system and is conserved across mammalian species. AgRP is currently known to be expressed exclusively in the mediobasal hypothalamus, and hypothalamic AgRP-expressing neurons are essential for feeding. Here we characterized a previously unknown population of AgRP cells in the mouse hindbrain. METHODS: Expression of AgRP in the hindbrain was investigated using gene expression analysis, single-cell RNA sequencing, immunofluorescent analysis and multiple transgenic mice with reporter expressions. Activation of AgRP neurons was achieved by Designer Receptors Exclusively Activated by Designer Drugs (DREADD) and by transcranial focal photo-stimulation using a step-function opsin with ultra-high light sensitivity (SOUL). RESULTS: AgRP expressing cells were present in the area postrema (AP) and the adjacent subpostrema area (SubP) and commissural nucleus of the solitary tract (cNTS) of the mouse hindbrain (termed AgRPHind herein). AgRPHind cells consisted of locally projecting neurons as well as tanycyte-like cells. Food deprivation stimulated hindbrain Agrp expression as well as neuronal activity of subsets of AgRPHind cells. In adult mice that lacked hypothalamic AgRP neurons, chemogenetic activation of AgRP neurons resulted in hyperphagia and weight gain. In addition, transcranial focal photo-stimulation of hindbrain AgRP cells increased food intake in adult mice with or without hypothalamic AgRP neurons. CONCLUSIONS: Our study indicates that the central melanocortin system in the hindbrain possesses an orexigenic component, and that AgRPHind neurons stimulate feeding independently of hypothalamic AgRP neurons.


Asunto(s)
Hipotálamo , Melanocortinas , Ratones , Animales , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Ratones Transgénicos , Melanocortinas/metabolismo , Rombencéfalo/metabolismo , Mamíferos/metabolismo
6.
Nat Med ; 12(5): 534-40, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16604086

RESUMEN

Leptin controls food intake by regulating the transcription of key neuropeptides in the hypothalamus. The mechanism by which leptin regulates gene expression is unclear, however. Here we show that delivery of adenovirus encoding a constitutively nuclear mutant FoxO1, a transcription factor known to control liver metabolism and pancreatic beta-cell function, to the hypothalamic arcuate nucleus of rodents results in a loss of the ability of leptin to curtail food intake and suppress expression of Agrp. Conversely, a transactivation-deficient FoxO1 mutant prevents induction of Agrp by fasting. We also find that FoxO1 and the transcription factor Stat3 exert opposing actions on the expression of Agrp and Pomc through transcriptional squelching. FoxO1 promotes opposite patterns of coactivator-corepressor exchange at the Pomc and Agrp promoters, resulting in activation of Agrp and inhibition of Pomc. Thus, FoxO1 represents a shared component of pathways integrating food intake and peripheral metabolism.


Asunto(s)
Ingestión de Alimentos , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Leptina/metabolismo , Proteínas/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Proteína Relacionada con Agouti , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Células Cultivadas , Ayuno , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Técnicas de Transferencia de Gen , Péptidos y Proteínas de Señalización Intercelular , Ratones , Neuronas/citología , Neuronas/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Regiones Promotoras Genéticas , Proteínas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
7.
Cell Biosci ; 13(1): 20, 2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36732847

RESUMEN

BACKGROUND: Foraging for food precedes food consumption and is an important component of the overall metabolic programming that regulates feeding. Foraging is governed by central nervous system neuronal circuits but how it is influenced by diet and hormonal signals is still not well understood. RESULTS: In this study, we show that dietary cholesterol exerted suppressive effects on locomotor activity and that these effects were partially mediated by the neuropeptide Agouti-related protein (AgRP). High dietary cholesterol stimulated intestinal expression of fibroblast growth factor 15 (Fgf15), an ortholog of the human fibroblast growth factor 19 (FGF19). Intracerebroventricular infusion of FGF19 peptide reduced exploratory activity in the open field test paradigm. On the other hand, the lack of dietary cholesterol enhanced exploratory activity in the open field test, but this effect was abolished by central administration of FGF19. CONCLUSIONS: Experiments in this study show that dietary cholesterol suppresses locomotor activity and foraging-like behaviors, and this regulation is in part mediated by AgRP neurons. Dietary cholesterol or the central action of FGF19 suppresses exploratory behaviors, and the anxiogenic effects of dietary cholesterol may be mediated by the effect of FGF19 in the mouse brain. This study suggests that dietary cholesterol and intestinal hormone FGF15/19 signal a satiating state to the brain, thereby suppressing foraging-like behaviors.

8.
Proc Natl Acad Sci U S A ; 106(37): 15932-7, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19805233

RESUMEN

In female mammals including rodents and humans, feeding decreases during the periovulatory period of the ovarian cycle, which coincides with a surge in circulating estrogen levels. Ovariectomy increases food intake, which can be normalized by estrogen treatment at a dose and frequency mimicking those during the estrous cycle. Furthermore, administration of estrogen to rodents potently inhibits food intake. Despite these well-known effects of estrogen, neuronal subtypes that mediate estrogen's anorexigenic effects have not been identified. In this study, we show that changes in hypothalamic expression of agouti-related protein (Agrp) and neuropeptide Y (Npy) coincide with the cyclic changes in feeding across the estrous cycle. These cyclic changes in feeding are abolished in mice with degenerated AgRP neurons even though these mice cycle normally. Central administration of 17beta-estradiol (E2) decreases food intake in controls but not in mice lacking the AgRP neurons. Furthermore, E2 treatment suppresses fasting-induced c-Fos activation in AgRP and NPY neurons and blunts the refeeding response. Surprisingly, although estrogen receptor alpha (ERalpha) is the key mediator of estrogen's anorexigenic effects, we find that expression of ERalpha is completely excluded from AgRP and NPY neurons in the mouse hypothalamus, suggesting that estrogen may regulate these neurons indirectly via presynaptic neurons that express ERalpha. This study indicates that neurons coexpressing AgRP and NPY are functionally required for the cyclic changes in feeding across estrous cycle and that AgRP and NPY neurons are essential mediators of estrogen's anorexigenic function.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Regulación del Apetito/fisiología , Ciclo Estral/fisiología , Neuropéptido Y/fisiología , Proteína Relacionada con Agouti/deficiencia , Proteína Relacionada con Agouti/genética , Animales , Regulación del Apetito/efectos de los fármacos , Peso Corporal/genética , Peso Corporal/fisiología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Ciclo Estral/genética , Femenino , Expresión Génica , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Ovariectomía , Ovario/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Sci Signal ; 15(733): eabj8204, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35536884

RESUMEN

Variants in the gene encoding ankyrin repeat and SOCS box-containing 4 (ASB4) are linked to human obesity. Here, we characterized the pathways underlying the metabolic functions of ASB4. Hypothalamic Asb4 expression was suppressed by fasting in wild-type mice but not in mice deficient in AgRP, which encodes Agouti-related protein (AgRP), an appetite-stimulating hormone, suggesting that ASB4 is a negative target of AgRP. Many ASB4 neurons in the brain were adjacent to AgRP terminals, and feeding induced by AgRP neuronal activation was disrupted in Asb4-deficient mice. Acute knockdown of Asb4 in the brain caused marked hyperphagia due to increased meal size, and Asb4 deficiency led to increased meal size and food intake at the onset of refeeding, when very large meals were consumed. Asb4-deficient mice were resistant to the meal-terminating effects of exogenously administered calcitonin and showed decreased neuronal expression of Calcr, which encodes the calcitonin receptor. Pro-opiomelanocortin (POMC) neurons in the arcuate nucleus in mice are involved in glucose homeostasis, and Asb4 deficiency specifically in POMC neurons resulted in glucose intolerance that was independent of obesity. Furthermore, individuals with type 2 diabetes showed reduced ASB4 abundance in the infundibular nuclei, the human equivalent of the arcuate nucleus. Together, our results indicate that ASB4 acts in the brain to improve glucose homeostasis and to induce satiety after substantial meals, particularly those after food deprivation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Neuropéptidos , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Proteína Relacionada con Agouti/farmacología , Animales , Calcitonina/metabolismo , Calcitonina/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Homeostasis , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Neuropéptidos/metabolismo , Obesidad/genética , Obesidad/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Proopiomelanocortina/farmacología
10.
Mol Metab ; 58: 101442, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35051651

RESUMEN

OBJECTIVE: Preference for dietary fat vs. carbohydrate varies markedly across free-living individuals. It is recognized that food choice is under genetic and physiological regulation, and that the central melanocortin system is involved. However, how genetic and dietary factors interact to regulate relative macronutrient intake is not well understood. METHODS: We investigated how the choice for food rich in carbohydrate vs. fat is influenced by dietary cholesterol availability and agouti-related protein (AGRP), the orexigenic component of the central melanocortin system. We assessed how macronutrient intake and different metabolic parameters correlate with plasma AGRP in a cohort of obese humans. We also examined how both dietary cholesterol levels and inhibiting de novo cholesterol synthesis affect carbohydrate and fat intake in mice, and how dietary cholesterol deficiency during the postnatal period impacts macronutrient intake patterns in adulthood. RESULTS: In obese human subjects, plasma levels of AGRP correlated inversely with consumption of carbohydrates over fats. Moreover, AgRP-deficient mice preferred to consume more calories from carbohydrates than fats, more so when each diet lacked cholesterol. Intriguingly, inhibiting cholesterol biosynthesis (simvastatin) promoted carbohydrate intake at the expense of fat without altering total caloric consumption, an effect that was remarkably absent in AgRP-deficient mice. Finally, feeding lactating C57BL/6 dams and pups a cholesterol-free diet prior to weaning led the offspring to prefer fats over carbohydrates as adults, indicating that altered cholesterol metabolism early in life programs adaptive changes to macronutrient intake. CONCLUSIONS: Together, our study illustrates a specific gene-diet interaction in modulating food choice.


Asunto(s)
Colesterol en la Dieta , Carbohidratos de la Dieta , Adulto , Proteína Relacionada con Agouti , Animales , Dieta , Femenino , Humanos , Lactancia , Melanocortinas , Ratones , Ratones Endogámicos C57BL , Obesidad
11.
J Neurosci ; 30(2): 723-30, 2010 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20071537

RESUMEN

The ability to develop counter-regulatory mechanisms to maintain energy balance in response to environmental and physiologic insults is essential for survival, but the mechanisms underlying these compensatory regulations are poorly understood. Agouti-related peptide (AGRP) and Neuropeptide Y are potent orexigens and are coexpressed in neurons in the arcuate nucleus of the hypothalamus. Acute ablation of these neurons leads to severe anorexia and weight loss, whereas progressive degeneration of these neurons has minimal impact on food intake and body weight, suggesting that compensatory mechanisms are developed to maintain orexigenic drive. In this study, we show that cell proliferation is increased in the hypothalamus of adult mutant animals in which AgRP neurons undergo progressive neurodegeneration due to deletion of mitochondrial transcription factor A, and that a subset of these newly generated cells differentiate into AgRP neurons along with other resident neuronal subtypes. Furthermore, some of the newly generated cells are capable of responding to leptin, and a central blockade of cell proliferation in adult animals results in decreases in food intake and body adiposity in mutant but not in control animals. Our study indicates that neurons important for energy homeostasis can be regenerated in adult feeding centers under neurodegenerative conditions. It further suggests that de novo neurogenesis might serve as a compensatory mechanism contributing to the plastic control of energy balance in response to environmental and physiologic insults.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Absorciometría de Fotón/métodos , Hormona Adrenocorticotrópica/metabolismo , Proteína Relacionada con Agouti/genética , Análisis de Varianza , Animales , Animales Recién Nacidos , Antimetabolitos Antineoplásicos/farmacología , Composición Corporal/efectos de los fármacos , Composición Corporal/genética , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Proliferación Celular/efectos de los fármacos , Citarabina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Hipotálamo/citología , Inyecciones Intraventriculares/métodos , Antígeno Ki-67/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Neuronas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo
12.
J Neurosci ; 30(35): 11815-25, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20810901

RESUMEN

Sirt1 has been associated with various effects of calorie restriction, including an increase in lifespan. Here we show in mice that a central regulatory component in energy metabolism, the hypothalamic melanocortin system, is affected by Sirt1, which promotes the activity and connectivity of this system resulting in negative energy balance. In adult mice, the pharmacological inhibition of brain Sirt1 activity decreased Agrp neuronal activity and the inhibitory tone on the anorexigenic POMC neurons, as measured by the number of synaptic inputs to these neurons. When a Sirt1 inhibitor (EX-527) was injected either peripherally (i.p., 10 mg/kg) or directly into the brain (i.c.v., 1.5 nmol/mouse), it decreased both food intake during the dark cycle and ghrelin-induced food intake. This effect on feeding is mediated by upstream melanocortin receptors, because the MC4R antagonist, SHU9119, reversed Sirt1's effect on food intake. This action of Sirt1 required an appropriate shift in the mitochondrial redox state: in the absence of such an adaptation enabled by the mitochondrial protein, UCP2, Sirt1-induced cellular and behavioral responses were impaired. In accordance with the pharmacological results, the selective knock-out of Sirt1 in hypothalamic Agrp neurons through the use of Cre-Lox technology decreased electric responses of Agrp neurons to ghrelin and decreased food intake, leading to decreased lean mass, fat mass, and body weight. The present data indicate that Sirt1 has a central mode of action by acting on the NPY/Agrp neurons to affect body metabolism.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Metabolismo Energético/fisiología , Melanocortinas/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Sirtuina 1/fisiología , Sinapsis/fisiología , Potenciales Sinápticos/fisiología , Proteína Relacionada con Agouti/biosíntesis , Animales , Carbazoles/administración & dosificación , Combinación de Medicamentos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Metabolismo Energético/efectos de los fármacos , Femenino , Masculino , Melanocortinas/metabolismo , Hormonas Estimuladoras de los Melanocitos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidación-Reducción/efectos de los fármacos , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/deficiencia , Sinapsis/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos
13.
Dev Biol ; 339(1): 38-50, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20025866

RESUMEN

The hypothalamic neuronal circuits that modulate energy homeostasis become mature and functional during early postnatal life. However, the molecular mechanism underlying this developmental process remains largely unknown. Here we use a mouse genetic approach to investigate the role of gamma-protocadherins (Pcdh-gammas) in hypothalamic neuronal circuits. First, we show that rat insulin promoter (RIP)-Cre conditional knockout mice lacking Pcdh-gammas in a broad subset of hypothalamic neurons are obese and hyperphagic. Second, specific deletion of Pcdh-gammas in anorexigenic proopiomelanocortin (POMC) expressing neurons also leads to obesity. Using cell lineage tracing, we show that POMC and RIP-Cre expressing neurons do not overlap but interact with each other in the hypothalamus. Moreover, excitatory synaptic inputs are reduced in Pcdh-gamma deficient POMC neurons. Genetic evidence from both knockout models shows that Pcdh-gammas can regulate POMC neuronal function autonomously and non-autonomously through cell-cell interaction. Taken together, our data demonstrate that Pcdh-gammas regulate the formation and functional integrity of hypothalamic feeding circuitry in mice.


Asunto(s)
Cadherinas/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Animales , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Linaje de la Célula , Metabolismo Energético , Hipotálamo/citología , Inmunohistoquímica , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Inmunoelectrónica , Neuronas/citología , Reacción en Cadena de la Polimerasa
14.
J Clin Invest ; 117(8): 2325-36, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17671657

RESUMEN

Hypothalamic AMP-activated protein kinase (AMPK) has been suggested to act as a key sensing mechanism, responding to hormones and nutrients in the regulation of energy homeostasis. However, the precise neuronal populations and cellular mechanisms involved are unclear. The effects of long-term manipulation of hypothalamic AMPK on energy balance are also unknown. To directly address such issues, we generated POMC alpha 2KO and AgRP alpha 2KO mice lacking AMPK alpha2 in proopiomelanocortin- (POMC-) and agouti-related protein-expressing (AgRP-expressing) neurons, key regulators of energy homeostasis. POMC alpha 2KO mice developed obesity due to reduced energy expenditure and dysregulated food intake but remained sensitive to leptin. In contrast, AgRP alpha 2KO mice developed an age-dependent lean phenotype with increased sensitivity to a melanocortin agonist. Electrophysiological studies in AMPK alpha2-deficient POMC or AgRP neurons revealed normal leptin or insulin action but absent responses to alterations in extracellular glucose levels, showing that glucose-sensing signaling mechanisms in these neurons are distinct from those pathways utilized by leptin or insulin. Taken together with the divergent phenotypes of POMC alpha 2KO and AgRP alpha 2KO mice, our findings suggest that while AMPK plays a key role in hypothalamic function, it does not act as a general sensor and integrator of energy homeostasis in the mediobasal hypothalamus.


Asunto(s)
Metabolismo Energético/fisiología , Homeostasis/fisiología , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Complejos Multienzimáticos/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Proteína Relacionada con Agouti , Animales , Ingestión de Alimentos/fisiología , Glucosa/metabolismo , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Leptina/metabolismo , Ratones , Ratones Noqueados , Complejos Multienzimáticos/deficiencia , Proopiomelanocortina/deficiencia , Proteínas Serina-Treonina Quinasas/deficiencia , Transducción de Señal/fisiología
15.
Mol Endocrinol ; 22(3): 751-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18096691

RESUMEN

Leptin is a fat-derived hormone that exerts pleiotropic effects on energy balance and neuroendocrine functions. Mice defective in leptin or its receptor [leptin receptor, isoform b (LepRb)] exhibit profound obesity, infertility, and reduced linear growth. Leptin binding to its receptor triggers multiple signaling pathways, including signal transducer and activator of transcription 3 (Stat 3), phosphatidylinositol-3-kinase, and ERK. A considerable amount of effort has been focused on how these signaling pathways mediate diverse leptin functions. Mice containing a mutant LepRb incapable of Stat3 signaling are obese but remain fertile with enhanced linear growth. In contrast, deletion of Stat3 in the whole brain with Nestin-Cre results in infertility and decreased linear growth, in addition to obesity. The additional phenotypes of the Nestin-mediated deletion could reflect Stat3 action in non-LepRb neurons or leptin-independent Stat3 actions in LepRb neurons. To resolve this discrepancy and to gain more insight into the metabolic actions of Stat3, we have generated mice in which Stat3 is disrupted specifically in LepRb neurons after the onset of leptin receptor expression. We show that mutant mice exhibit profound obesity with increased linear growth and normal fertility. In addition, impaired glycemic control in these animals correlates with their degree of obesity. These results demonstrate that Stat3 in LepRb neurons does not regulate linear growth or fertility. These results further suggest that leptin's effects on growth and reproduction are mediated by other signaling pathways, and that Stat3-mediated control of these functions is mediated independently of leptin and LepRb neurons.


Asunto(s)
Hipotálamo/fisiología , Leptina/fisiología , Neuronas/fisiología , Receptores de Leptina/fisiología , Factor de Transcripción STAT3/fisiología , Proteína Relacionada con Agouti/biosíntesis , Proteína Relacionada con Agouti/genética , Animales , Glucemia/metabolismo , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Femenino , Hipotálamo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Neuronas/metabolismo , Neuropéptido Y/biosíntesis , Neuropéptido Y/genética , Fosforilación , Proopiomelanocortina/biosíntesis , Proopiomelanocortina/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Leptina/biosíntesis , Receptores de Leptina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
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
17.
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
18.
J Clin Invest ; 115(4): 940-50, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15841180

RESUMEN

Insulin receptor substrate 2 (Irs2) plays complex roles in energy homeostasis. We generated mice lacking Irs2 in beta cells and a population of hypothalamic neurons (RIPCreIrs2KO), in all neurons (NesCreIrs2KO), and in proopiomelanocortin neurons (POMCCreIrs2KO) to determine the role of Irs2 in the CNS and beta cell. RIPCreIrs2KO mice displayed impaired glucose tolerance and reduced beta cell mass. Overt diabetes did not ensue, because beta cells escaping Cre-mediated recombination progressively populated islets. RIPCreIrs2KO and NesCreIrs2KO mice displayed hyperphagia, obesity, and increased body length, which suggests altered melanocortin action. POMCCreIrs2KO mice did not display this phenotype. RIPCreIrs2KO and NesCreIrs2KO mice retained leptin sensitivity, which suggests that CNS Irs2 pathways are not required for leptin action. NesCreIrs2KO and POMCCreIrs2KO mice did not display reduced beta cell mass, but NesCreIrs2KO mice displayed mild abnormalities of glucose homeostasis. RIPCre neurons did not express POMC or neuropeptide Y. Insulin and a melanocortin agonist depolarized RIPCre neurons, whereas leptin was ineffective. Insulin hyperpolarized and leptin depolarized POMC neurons. Our findings demonstrate a critical role for IRS2 in beta cell and hypothalamic function and provide insights into the role of RIPCre neurons, a distinct hypothalamic neuronal population, in growth and energy homeostasis.


Asunto(s)
Metabolismo Energético , Homeostasis , Hipotálamo/metabolismo , Islotes Pancreáticos/metabolismo , Neuronas/metabolismo , Fosfoproteínas/metabolismo , Animales , Peso Corporal , Electrofisiología , Genotipo , Glucosa/metabolismo , Hipotálamo/citología , Insulina/administración & dosificación , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Islotes Pancreáticos/citología , Leptina/administración & dosificación , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Fosfoproteínas/genética , Proopiomelanocortina/metabolismo , Receptor de Insulina/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
19.
Trends Endocrinol Metab ; 29(6): 363-366, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29477281

RESUMEN

Bile acids facilitate dietary fat absorption upon release into the small intestine after a meal. A recent study by Liu and colleagues identifies a gut-brain axis wherein bile acids signal an energy-replete state to hypothalamic AgRP neurons via activation of neuronal FGF receptors, which orchestrate whole-body glucose metabolism.


Asunto(s)
Ácidos y Sales Biliares , Factores de Crecimiento de Fibroblastos , Encéfalo , Glucosa , Hipotálamo
20.
Endocrinology ; 159(6): 2408-2420, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29750244

RESUMEN

Proper regulation of energy metabolism requires neurons in the central nervous system to respond dynamically to signals that reflect the body's energy reserve, and one such signal is leptin. Agouti-related protein (AgRP) is a hypothalamic neuropeptide that is markedly upregulated in leptin deficiency, a condition that is associated with severe obesity, diabetes, and hepatic steatosis. Because deleting AgRP in mice does not alter energy balance, we sought to determine whether AgRP plays an indispensable role in regulating energy and hepatic lipid metabolism in the sensitized background of leptin deficiency. We generated male mice that are deficient for both leptin and AgRP [double-knockout (DKO)]. DKO mice and ob/ob littermates had similar body weights, food intake, energy expenditure, and plasma insulin levels, although DKO mice surprisingly developed heightened hyperglycemia with advancing age. Overall hepatic lipid content was reduced in young prediabetic DKO mice, but not in the older diabetic counterparts. Intriguingly, however, both young and older DKO mice had an altered zonal distribution of hepatic lipids with reduced periportal lipid deposition. Moreover, leptin stimulated, whereas AgRP inhibited, hepatic sympathetic activity. Ablating sympathetic nerves to the liver, which primarily innervate the portal regions, produced periportal lipid accumulation in wild-type mice. Collectively, our results highlight AgRP as a regulator of hepatic sympathetic activity and metabolic zonation.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad Mórbida/genética , Obesidad Mórbida/metabolismo , Obesidad Mórbida/patología , Distribución Tisular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA