Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell Rep ; 28(11): 2905-2922.e5, 2019 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-31509751

RESUMEN

The importance of hypothalamic leptin and insulin resistance in the development and maintenance of obesity remains unclear. The tyrosine phosphatases protein tyrosine phosphatase 1B (PTP1B) and T cell protein tyrosine phosphatase (TCPTP) attenuate leptin and insulin signaling and are elevated in the hypothalami of obese mice. We report that elevated PTP1B and TCPTP antagonize hypothalamic leptin and insulin signaling and contribute to the maintenance of obesity. Deletion of PTP1B and TCPTP in the hypothalami of obese mice enhances CNS leptin and insulin sensitivity, represses feeding, and increases browning, to decrease adiposity and improve glucose metabolism. The daily intranasal administration of a PTP1B inhibitor, plus the glucocorticoid antagonist RU486 that decreases TCPTP expression, represses feeding, increases browning, promotes weight loss, and improves glucose metabolism in obese mice. Our findings causally link heightened hypothalamic PTP1B and TCPTP with leptin and insulin resistance and the maintenance of obesity and define a viable pharmacological approach by which to promote weight loss in obesity.


Asunto(s)
Hipotálamo/metabolismo , Resistencia a la Insulina/genética , Leptina/metabolismo , Obesidad/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Pérdida de Peso/genética , Tejido Adiposo Blanco/metabolismo , Administración Intranasal , Animales , Barrera Hematoencefálica/metabolismo , Colestanos/administración & dosificación , Dieta Alta en Grasa , Conducta Alimentaria/efectos de los fármacos , Gliosis/genética , Gliosis/metabolismo , Glucocorticoides/farmacología , Hipotálamo/efectos de los fármacos , Leptina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mifepristona/administración & dosificación , Obesidad/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Espermina/administración & dosificación , Espermina/análogos & derivados
2.
Neuroendocrinology ; 109(4): 374-390, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30995667

RESUMEN

BACKGROUND: Obesity is associated with reduced physiological responses to leptin and insulin, leading to the concept of obesity-associated hormonal resistance. OBJECTIVES: Here, we demonstrate that contrary to expectations, leptin signaling not only remains functional but also is constantly activated in the arcuate nucleus of the hypothalamus (ARH) neurons of obese mice. This state of persistent response to endogenous leptin underpins the lack of response to exogenous leptin. METHODS AND RESULTS: The study of combined leptin and insulin signaling demonstrates that there is a common pool of ARH neurons responding to both hormones. More importantly, we show that the constant activation of leptin receptor neurons in the ARH prevents insulin signaling in these neurons, leading to impaired glucose tolerance. Accordingly, antagonising leptin signaling in diet-induced obese (DIO) mice restores insulin signaling in the ARH and improves glucose homeostasis. Direct inhibition of PTP1B in the CNS restores arcuate insulin signaling similarly to leptin inhibition; this effect is likely to be mediated by AgRP neurons since PTP1B deletion specifically in AgRP neurons restores glucose and insulin tolerance in DIO mice. CONCLUSIONS: Finally, our results suggest that the constant activation of arcuate leptin signaling in DIO mice increases PTP1B expression, which exerts an inhibitory effect on insulin signaling leading to impaired glucose homeostasis.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiopatología , Glucosa/metabolismo , Homeostasis , Hipotálamo/fisiopatología , Insulina , Leptina , Obesidad/fisiopatología , Animales , Dieta , Dieta Alta en Grasa , Genotipo , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Endogámicos C57BL , Neuronas , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Factor de Transcripción STAT3/biosíntesis , Transducción de Señal
3.
J Med Chem ; 61(24): 11144-11157, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30525586

RESUMEN

Celastrol is a natural pentacyclic triterpene used in traditional Chinese medicine with significant weight-lowering effects. Celastrol-administered mice at 100 µg/kg decrease food consumption and body weight via a leptin-dependent mechanism, yet its molecular targets in this pathway remain elusive. Here, we demonstrate in vivo that celastrol-induced weight loss is largely mediated by the inhibition of leptin negative regulators protein tyrosine phosphatase (PTP) 1B (PTP1B) and T-cell PTP (TCPTP) in the arcuate nucleus (ARC) of the hypothalamus. We show in vitro that celastrol binds reversibly and inhibits noncompetitively PTP1B and TCPTP. NMR data map the binding site to an allosteric site in the catalytic domain that is in proximity of the active site. By using a panel of PTPs implicated in hypothalamic leptin signaling, we show that celastrol additionally inhibited PTEN and SHP2 but had no activity toward other phosphatases of the PTP family. These results suggest that PTP1B and TCPTP in the ARC are essential for celastrol's weight lowering effects in adult obese mice.


Asunto(s)
Fármacos Antiobesidad/farmacología , Obesidad/tratamiento farmacológico , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 2/antagonistas & inhibidores , Triterpenos/farmacología , Sitio Alostérico , Animales , Fármacos Antiobesidad/metabolismo , Dominio Catalítico , Dieta Alta en Grasa/efectos adversos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratones Transgénicos , Obesidad/etiología , Triterpenos Pentacíclicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/química , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Relación Estructura-Actividad , Triterpenos/química , Triterpenos/metabolismo , Pérdida de Peso/efectos de los fármacos
4.
Elife ; 72018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30230471

RESUMEN

Hypothalamic neurons respond to nutritional cues by altering gene expression and neuronal excitability. The mechanisms that control such adaptive processes remain unclear. Here we define populations of POMC neurons in mice that are activated or inhibited by insulin and thereby repress or inhibit hepatic glucose production (HGP). The proportion of POMC neurons activated by insulin was dependent on the regulation of insulin receptor signaling by the phosphatase TCPTP, which is increased by fasting, degraded after feeding and elevated in diet-induced obesity. TCPTP-deficiency enhanced insulin signaling and the proportion of POMC neurons activated by insulin to repress HGP. Elevated TCPTP in POMC neurons in obesity and/or after fasting repressed insulin signaling, the activation of POMC neurons by insulin and the insulin-induced and POMC-mediated repression of HGP. Our findings define a molecular mechanism for integrating POMC neural responses with feeding to control glucose metabolism.


Asunto(s)
Glucosa/metabolismo , Insulina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Animales , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacología , Hipotálamo/citología , Insulina/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plasticidad Neuronal/genética , Proopiomelanocortina/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
5.
Diabetes ; 67(11): 2456-2465, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30158241

RESUMEN

Celastrol, a plant-derived constituent of traditional Chinese medicine, has been proposed to offer significant potential as an antiobesity drug. However, the molecular mechanism for this activity is unknown. We show that the weight-lowering effects of celastrol are driven by decreased food consumption. Although young Lep ob mice respond with a decrease in food intake and body weight, adult Lep db and Lep ob mice are unresponsive to celastrol, suggesting that functional leptin signaling in adult mice is required to elicit celastrol's catabolic actions. Protein tyrosine phosphatase 1 (PTP1B), a leptin negative-feedback regulator, has been previously reported to be one of celastrol's targets. However, we found that global PTP1B knockout (KO) and wild-type (WT) mice have comparable weight loss and hypophagia when treated with celastrol. Increased levels of uncoupling protein 1 (UCP1) in subcutaneous white and brown adipose tissue suggest celastrol-induced thermogenesis as a further mechanism. However, diet-induced obese UCP1 WT and KO mice have comparable weight loss upon celastrol treatment, and celastrol treatment has no effect on energy expenditure under ambient housing or thermoneutral conditions. Overall, our results suggest that celastrol-induced weight loss is hypophagia driven and age-dependently mediated by functional leptin signaling. Our data encourage reconsideration of therapeutic antiobesity strategies built on leptin sensitization.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Obesidad/metabolismo , Extractos Vegetales/farmacología , Triterpenos/farmacología , Proteína Desacopladora 1/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Ratones Noqueados , Obesidad/genética , Triterpenos Pentacíclicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Desacopladora 1/genética
6.
Cell Metab ; 26(2): 375-393.e7, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28768176

RESUMEN

Beige adipocytes can interconvert between white and brown-like states and switch between energy storage versus expenditure. Here we report that beige adipocyte plasticity is important for feeding-associated changes in energy expenditure and is coordinated by the hypothalamus and the phosphatase TCPTP. A fasting-induced and glucocorticoid-mediated induction of TCPTP, inhibited insulin signaling in AgRP/NPY neurons, repressed the browning of white fat and decreased energy expenditure. Conversely feeding reduced hypothalamic TCPTP, to increase AgRP/NPY neuronal insulin signaling, white adipose tissue browning and energy expenditure. The feeding-induced repression of hypothalamic TCPTP was defective in obesity. Mice lacking TCPTP in AgRP/NPY neurons were resistant to diet-induced obesity and had increased beige fat activity and energy expenditure. The deletion of hypothalamic TCPTP in obesity restored feeding-induced browning and increased energy expenditure to promote weight loss. Our studies define a hypothalamic switch that coordinates energy expenditure with feeding for the maintenance of energy balance.


Asunto(s)
Ingestión de Alimentos/psicología , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/biosíntesis , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Ratones , Ratones Transgénicos , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Obesidad/genética , Obesidad/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética
7.
Neuroendocrinology ; 104(4): 364-381, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28122381

RESUMEN

The central link between obesity and type 2 diabetes is the development of insulin resistance. To date, it is still not clear whether hyperinsulinemia causes insulin resistance, which underlies the pathogenesis of obesity-associated type 2 diabetes, owing to the sophisticated regulatory mechanisms that exist in the periphery and in the brain. In recent years, accumulating evidence has demonstrated the existence of insulin resistance within the hypothalamus. In this review, we have integrated the recent discoveries surrounding both central and peripheral insulin resistance to provide a comprehensive overview of insulin resistance in obesity and the regulation of systemic glucose homeostasis. In particular, this review will discuss how hyperinsulinemia and hyperleptinemia in obesity impair insulin sensitivity in tissues such as the liver, skeletal muscle, adipose tissue, and the brain. In addition, this review highlights insulin transport into the brain, signaling pathways associated with hypothalamic insulin receptor expression in the regulation of hepatic glucose production, and finally the perturbation of systemic glucose homeostasis as a consequence of central insulin resistance. We also suggest future approaches to overcome both central and peripheral insulin resistance to treat obesity and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Glucosa/metabolismo , Homeostasis/fisiología , Hipotálamo/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Obesidad/fisiopatología , Animales , Humanos , Leptina/fisiología , Transducción de Señal/fisiología
8.
Curr Opin Nephrol Hypertens ; 26(1): 20-25, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27798459

RESUMEN

PURPOSE OF REVIEW: Cardiovascular diseases (CVDs) are the number one cause of death globally. The risk for the development of CVDs is significantly increased in obesity. Leptin, the product of white adipose tissue, appears to contribute to the development of CVDs in obesity. Here, we discuss the premise that leptin engages the sympathetic nervous system and contributes to elevated blood pressure (BP) developing in obesity. RECENT FINDINGS: The long-term regulation of BP is dependent on the activity of the autonomic nervous system and specifically the sympathetic nervous system. Sympathetic nerve activity is significantly increased in obese rodents and humans. Leptin increases sympathetic nerve activity in rodents and humans; however, leptin only consistently increases BP chronically in rodents. The ability of leptin to increase BP in rodents is via both hypothalamic and extrahypothalamic regions. In leptin-deficient and leptin receptor-deficient humans, leptin appears to be the key reason for decreased systolic BP. However, in other research conducted in humans, chronic administration of leptin does not elevate BP. SUMMARY: Further research into the role of leptin in the development of CVDs, especially in humans, needs to be conducted.


Asunto(s)
Presión Sanguínea , Hipertensión/fisiopatología , Hipotálamo/fisiopatología , Leptina/metabolismo , Obesidad/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Animales , Presión Sanguínea/efectos de los fármacos , Humanos , Hipotálamo/fisiología , Leptina/deficiencia , Leptina/farmacología , Receptores de Leptina
9.
Mol Cell Endocrinol ; 428: 148-60, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27032712

RESUMEN

When 60-day-old tammar wallaby pouch young (Macropus eugenii) are fostered to mothers at 120 days of lactation, their growth, developmental rate and maturation of their GH/IGF axes are markedly accelerated. To determine the effect of fostering on energy intake, body composition and fat accretion, we first measured total body fat and lean mass in these young. Next, we mimicked the triglyceride oleic and palmitic acid composition of 120-day milk by supplementing 60 day young with these fatty acids and comparing their growth with that of growth accelerated young. There was no difference in the weight or growth axis maturation of supplemented young but there was significantly more body fat in these and in the growth-accelerated fostered young than in controls. We conclude that the accelerated growth and GH/IGF axis maturation observed previously in fostered young is most likely due to increased milk consumption and earlier access to specific nutrients.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Composición Corporal , Macropodidae/crecimiento & desarrollo , Macropodidae/fisiología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Tejido Adiposo/anatomía & histología , Tejido Adiposo/efectos de los fármacos , Fenómenos Fisiológicos Nutricionales de los Animales/efectos de los fármacos , Animales , Composición Corporal/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Suplementos Dietéticos , Metabolismo Energético/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormona del Crecimiento/sangre , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lactancia/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Macropodidae/sangre , Macropodidae/genética , Leche/metabolismo , Aceites/farmacología , Tamaño de los Órganos/efectos de los fármacos , Periodo Posparto/sangre , Periodo Posparto/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
Front Neuroendocrinol ; 39: 59-65, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26410445

RESUMEN

Leptin resistance is one of the main challenges of obesity. To date, two levels of resistance have been identified, first a decreased rate of leptin uptake into the brain and secondly a diminished central response to leptin. New findings have identified the mechanisms of leptin transport and demonstrated that it can be rescued in obesity, but it did not overcome the problem of central resistance. Alteration in the actions of leptin following diet-induced obesity (DIO) appears to be a multifactorial condition. Several phosphatases are inhibiting leptin signaling pathways in a pathological way. Besides, hypothalamic inflammation alters the neuronal circuits that control metabolism. Recent studies describing both mechanisms (inhibition of leptin signaling and inflammation), have provided key insights to potential new targets for treatment. However, recent data showing that DIO mice may conserve a cellular and physiological response to endogenous leptin, highlights the need to redefine the concept of "leptin resistance".


Asunto(s)
Leptina/metabolismo , Obesidad/fisiopatología , Animales , Femenino , Hipotálamo/crecimiento & desarrollo , Inflamación/fisiopatología , Ratones , Embarazo
11.
Nature ; 519(7541): 45-50, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25707796

RESUMEN

Hypothalamic pro-opiomelanocortin (POMC) neurons promote satiety. Cannabinoid receptor 1 (CB1R) is critical for the central regulation of food intake. Here we test whether CB1R-controlled feeding in sated mice is paralleled by decreased activity of POMC neurons. We show that chemical promotion of CB1R activity increases feeding, and notably, CB1R activation also promotes neuronal activity of POMC cells. This paradoxical increase in POMC activity was crucial for CB1R-induced feeding, because designer-receptors-exclusively-activated-by-designer-drugs (DREADD)-mediated inhibition of POMC neurons diminishes, whereas DREADD-mediated activation of POMC neurons enhances CB1R-driven feeding. The Pomc gene encodes both the anorexigenic peptide α-melanocyte-stimulating hormone, and the opioid peptide ß-endorphin. CB1R activation selectively increases ß-endorphin but not α-melanocyte-stimulating hormone release in the hypothalamus, and systemic or hypothalamic administration of the opioid receptor antagonist naloxone blocks acute CB1R-induced feeding. These processes involve mitochondrial adaptations that, when blocked, abolish CB1R-induced cellular responses and feeding. Together, these results uncover a previously unsuspected role of POMC neurons in the promotion of feeding by cannabinoids.


Asunto(s)
Cannabinoides/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Hipotálamo/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Animales , Metabolismo Energético/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Naloxona/farmacología , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Respuesta de Saciedad/efectos de los fármacos , Respuesta de Saciedad/fisiología , Proteína Desacopladora 2 , alfa-MSH/metabolismo , betaendorfina/metabolismo
12.
J Clin Invest ; 124(10): 4473-88, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25202980

RESUMEN

Liraglutide is a glucagon-like peptide-1 (GLP-1) analog marketed for the treatment of type 2 diabetes. Besides lowering blood glucose, liraglutide also reduces body weight. It is not fully understood how liraglutide induces weight loss or to what degree liraglutide acts directly in the brain. Here, we determined that liraglutide does not activate GLP-1-producing neurons in the hindbrain, and liraglutide-dependent body weight reduction in rats was independent of GLP-1 receptors (GLP-1Rs) in the vagus nerve, area postrema, and paraventricular nucleus. Peripheral injection of fluorescently labeled liraglutide in mice revealed the presence of the drug in the circumventricular organs. Moreover, labeled liraglutide bound neurons within the arcuate nucleus (ARC) and other discrete sites in the hypothalamus. GLP-1R was necessary for liraglutide uptake in the brain, as liraglutide binding was not seen in Glp1r(-/-) mice. In the ARC, liraglutide was internalized in neurons expressing proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART). Electrophysiological measurements of murine brain slices revealed that GLP-1 directly stimulates POMC/CART neurons and indirectly inhibits neurotransmission in neurons expressing neuropeptide Y (NPY) and agouti-related peptide (AgRP) via GABA-dependent signaling. Collectively, our findings indicate that the GLP-1R on POMC/CART-expressing ARC neurons likely mediates liraglutide-induced weight loss.


Asunto(s)
Péptido 1 Similar al Glucagón/análogos & derivados , Receptores de Glucagón/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Electrofisiología , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Hipotálamo/metabolismo , Liraglutida , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Vago/metabolismo
13.
J Neurosci ; 33(38): 15306-17, 2013 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-24048859

RESUMEN

Neuropeptide Y (NPY) neurons in both the arcuate nucleus of the hypothalamus (ARH) and the dorsomedial hypothalamus (DMH) have been implicated in food intake and obesity. However, while ARH NPY is highly expressed in the lean animal, DMH NPY mRNA expression is observed only after diet-induced obesity (DIO). Furthermore, while ARH NPY neurons are inhibited by leptin, the effect of this adipokine on DMH NPY neurons is unknown. In this study we show that in contrast to the consistent expression in the ARH, DMH NPY mRNA expression was undetectable until after 10 weeks in mice fed a high-fat diet, and peaked at 20 weeks. Surprisingly, electrophysiological experiments demonstrated that leptin directly depolarized and increased the firing rate of DMH NPY neurons in DIO mice. To further differentiate the regulation of DMH and ARH NPY populations, fasting decreased expression of DMH NPY expression, while it increased ARH NPY expression. However, treatment with a leptin receptor antagonist failed to alter DMH NPY expression, indicating that leptin may not be the critical factor regulating mRNA expression. Importantly, we also demonstrated that DMH NPY neurons coexpress cocaine amphetamine-regulated transcript (CART); however, CART mRNA expression in the DMH peaked earlier in the progression of DIO. This study demonstrates novel and important findings. First, NPY and CART are coexpressed in the same neurons within the DMH, and second, leptin stimulates DMH NPY neurons. These studies suggest that during the progression of DIO, there is an unknown signal that drives the expression of the orexigenic NPY signal within the DMH, and that the chronic hyperleptinemia increases the activity of these NPY/CART neurons.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Leptina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Obesidad/patología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Análisis de Varianza , Animales , Dieta/efectos adversos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipotálamo/patología , Técnicas In Vitro , Insulina/sangre , Leptina/antagonistas & inhibidores , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuropéptido Y/genética , Obesidad/sangre , Obesidad/etiología , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Radioinmunoensayo , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo
14.
Cell Metab ; 14(5): 684-99, 2011 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22000926

RESUMEN

In obesity, anorectic responses to leptin are diminished, giving rise to the concept of "leptin resistance." Increased expression of protein tyrosine phosphatase 1B (PTP1B) has been associated with the attenuation of leptin signaling and development of cellular leptin resistance. Here we report that hypothalamic levels of the tyrosine phosphatase TCPTP are also elevated in obesity to attenuate the leptin response. We show that mice that lack TCPTP in neuronal cells have enhanced leptin sensitivity and are resistant to high-fat-diet-induced weight gain and the development of leptin resistance. Also, intracerebroventricular administration of a TCPTP inhibitor enhances leptin signaling and responses in mice. Moreover, the combined deletion of TCPTP and PTP1B in neuronal cells has additive effects in the prevention of diet-induced obesity. Our results identify TCPTP as a critical negative regulator of hypothalamic leptin signaling and causally link elevated TCPTP to the development of cellular leptin resistance in obesity.


Asunto(s)
Hipotálamo/metabolismo , Leptina/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 2/deficiencia , Transducción de Señal , Animales , Glucemia/análisis , Composición Corporal/efectos de los fármacos , Dieta Alta en Grasa , Inhibidores Enzimáticos/farmacología , Femenino , Expresión Génica , Hipotálamo/citología , Infusiones Intraventriculares , Insulina/sangre , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Obesidad/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Receptores de Leptina/metabolismo , Técnicas de Cultivo de Tejidos
15.
Proc Natl Acad Sci U S A ; 107(33): 14875-80, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20679202

RESUMEN

The neuronal circuits involved in the regulation of feeding behavior and energy expenditure are soft-wired, reflecting the relative activity of the postsynaptic neuronal system, including the anorexigenic proopiomelanocortin (POMC)-expressing cells of the arcuate nucleus. We analyzed the synaptic input organization of the melanocortin system in lean rats that were vulnerable (DIO) or resistant (DR) to diet-induced obesity. We found a distinct difference in the quantitative and qualitative synaptology of POMC cells between DIO and DR animals, with a significantly greater number of inhibitory inputs in the POMC neurons in DIO rats compared with DR rats. When exposed to a high-fat diet (HFD), the POMC cells of DIO animals lost synapses, whereas those of DR rats recruited connections. In both DIO rats and mice, the HFD-triggered loss of synapses on POMC neurons was associated with increased glial ensheathment of the POMC perikarya. The altered synaptic organization of HFD-fed animals promoted increased POMC tone and a decrease in the stimulatory connections onto the neighboring neuropeptide Y (NPY) cells. Exposure to HFD was associated with reactive gliosis, and this affected the structure of the blood-brain barrier such that the POMC and NPY cell bodies and dendrites became less accessible to blood vessels. Taken together, these data suggest that consumption of an HFD has a major impact on the cytoarchitecture of the arcuate nucleus in vulnerable subjects, with changes that might be irreversible due to reactive gliosis.


Asunto(s)
Dieta , Gliosis/metabolismo , Melanocortinas/metabolismo , Obesidad/metabolismo , Sinapsis/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/patología , Núcleo Arqueado del Hipotálamo/fisiopatología , Grasas de la Dieta/efectos adversos , Femenino , Gliosis/etiología , Hipotálamo/metabolismo , Hipotálamo/patología , Hipotálamo/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica , Neuronas/metabolismo , Neuronas/ultraestructura , Neuropéptido Y/metabolismo , Obesidad/etiología , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/fisiología
16.
Obesity (Silver Spring) ; 17(1): 16-24, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19107124

RESUMEN

The administration of antipsychotic drugs to human patients or experimental animals leads to significant weight gain, which is widely presumed to be driven by hyperphagia; however, the contribution from energy expenditure remains unclear. These studies aim to examine the contribution of shifts in energy expenditure, particularly those involving centrally mediated changes in thermogenesis, to the body weight gain associated with the administration of olanzapine to female Sprague Dawley rats. Olanzapine (6 mg/kg/day orally) caused a transient increase in food intake but a maintained increase in body weight. When pair-fed rats were treated with olanzapine, body weight continued to rise compared to vehicle-treated rats, consistent with a reduction in energy expenditure. Brown adipose tissue (BAT) temperature, measured using biotelemetry devices, decreased immediately after the onset of olanzapine treatment and remained depressed, as did physical activity. UCP1 expression in interscapular BAT was reduced following chronic olanzapine treatment. An acute injection of olanzapine was preceded by an injection of a retrograde tracer into the spinal cord to evaluate the nature of the olanzapine-activated neural pathway. Levels of Fos protein in a number of spinally projecting neurons within discrete hypothalamic and brainstem sites were elevated in olanzapine-treated rats. Some of these neurons in the perifornical region of the lateral hypothalamus (LHA) were also Orexin A positive. These data collectively show a significant impact of thermogenesis (and physical activity) on the weight gain associated with olanzapine treatment. The anatomical studies provide an insight into the central neuroanatomical substrate that may subserve the altered thermogenic responses brought about by olanzapine.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Antipsicóticos/efectos adversos , Benzodiazepinas/farmacología , Peso Corporal/efectos de los fármacos , Termogénesis/fisiología , Aumento de Peso/efectos de los fármacos , Tejido Adiposo Pardo/efectos de los fármacos , Animales , Ingestión de Energía/efectos de los fármacos , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiología , Actividad Motora/efectos de los fármacos , Olanzapina , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/fisiología , Ratas , Ratas Sprague-Dawley , Termogénesis/efectos de los fármacos
17.
PLoS One ; 3(5): e2202, 2008 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-18493584

RESUMEN

BACKGROUND: Cannabinoids, the active components of marijuana, stimulate appetite, and cannabinoid receptor-1 (CB1-R) antagonists suppress appetite and promote weight loss. Little is known about how CB1-R antagonists affect the central neurocircuitry, specifically the melanocortin system that regulates energy balance. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that peripherally administered CB1-R antagonist (AM251) or agonist equally suppressed or stimulated feeding respectively in A(y) , which lack a functional melanocortin system, and wildtype mice, demonstrating that cannabinoid effects on feeding do not require melanocortin circuitry. CB1-R antagonist or agonist administered into the ventral tegmental area (VTA) equally suppressed or stimulated feeding respectively, in both genotypes. In addition, peripheral and central cannabinoid administration similarly induced c-Fos activation in brain sites suggesting mediation via motivational dopaminergic circuitry. Amperometry-detected increases in evoked dopamine (DA) release by the CB1-R antagonist in nucleus accumbens slices indicates that AM251 modulates DA release from VTA terminals. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that the effects of cannabinoids on energy balance are independent of hypothalamic melanocortin circuitry and is primarily driven by the reward system.


Asunto(s)
Cannabinoides/farmacología , Conducta Alimentaria/efectos de los fármacos , Melanocortinas/fisiología , Animales , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
18.
J Clin Invest ; 118(5): 1796-805, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18382766

RESUMEN

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


Asunto(s)
Hipotálamo/citología , Leptina/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proopiomelanocortina/metabolismo , Transducción de Señal/fisiología , Animales , Peso Corporal , Ingestión de Alimentos , Metabolismo Energético , Homeostasis , Humanos , Ratones , Ratones Noqueados , Neuronas/citología , Técnicas de Placa-Clamp
19.
Annu Rev Physiol ; 70: 537-56, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17937601

RESUMEN

The adipose tissue-derived hormone leptin acts via its receptor (LRb) in the brain to regulate energy balance and neuroendocrine function. LRb signaling via STAT3 and a number of other pathways is required for the totality of leptin action. The failure of elevated leptin levels to suppress feeding and mediate weight loss in common forms of obesity defines a state of so-called leptin resistance. A number of mechanisms, including the leptin-stimulated phosphorylation of Tyr(985) on LRb and the suppressor of cytokine signaling 3, attenuate leptin signaling and promote a cellular leptin resistance in obesity. Several unique features of the arcuate nucleus of the hypothalamus may contribute to the severity of cellular leptin resistance in this region. Other mechanisms that govern feeding behavior and food reward may also underlie the inception of obesity.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Hipotálamo/fisiología , Leptina/metabolismo , Obesidad/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Leptina/genética , Obesidad/fisiopatología
20.
Cell Metab ; 5(6): 438-49, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17550779

RESUMEN

Insulin action in the central nervous system regulates energy homeostasis and glucose metabolism. To define the insulin-responsive neurons that mediate these effects, we generated mice with selective inactivation of the insulin receptor (IR) in either pro-opiomelanocortin (POMC)- or agouti-related peptide (AgRP)-expressing neurons of the arcuate nucleus of the hypothalamus. While neither POMC- nor AgRP-restricted IR knockout mice exhibited altered energy homeostasis, insulin failed to normally suppress hepatic glucose production during euglycemic-hyperinsulinemic clamps in AgRP-IR knockout (IR(DeltaAgRP)) mice. These mice also exhibited reduced insulin-stimulated hepatic interleukin-6 expression and increased hepatic expression of glucose-6-phosphatase. These results directly demonstrate that insulin action in POMC and AgRP cells is not required for steady-state regulation of food intake and body weight. However, insulin action specifically in AgRP-expressing neurons does play a critical role in controlling hepatic glucose production and may provide a target for the treatment of insulin resistance in type 2 diabetes.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Glucosa/metabolismo , Insulina/farmacología , Hígado/metabolismo , Neuronas/efectos de los fármacos , Animales , Western Blotting , Peso Corporal , Electrofisiología , Femenino , Prueba de Tolerancia a la Glucosa , Glucosa-6-Fosfatasa/metabolismo , Homeostasis , Hiperinsulinismo/metabolismo , Hipotálamo/metabolismo , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Interleucina-6/metabolismo , Hígado/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proopiomelanocortina/metabolismo , Receptor de Insulina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA