Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Metab ; 6(3): 473-493, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38378998

RESUMEN

Agouti-related peptide (AgRP)-expressing and proopiomelanocortin (POMC)-expressing neurons reciprocally regulate food intake. Here, we combine non-interacting recombinases to simultaneously express functionally opposing chemogenetic receptors in AgRP and POMC neurons for comparing metabolic responses in male and female mice with simultaneous activation of AgRP and inhibition of POMC neurons with isolated activation of AgRP neurons or isolated inhibition of POMC neurons. We show that food intake is regulated by the additive effect of AgRP neuron activation and POMC neuron inhibition, while systemic insulin sensitivity and gluconeogenesis are differentially modulated by isolated-versus-simultaneous regulation of AgRP and POMC neurons. We identify a neurocircuit engaging Npy1R-expressing neurons in the paraventricular nucleus of the hypothalamus, where activated AgRP neurons and inhibited POMC neurons cooperate to promote food consumption and activate Th+ neurons in the nucleus tractus solitarii. Collectively, these results unveil how food intake is precisely regulated by the simultaneous bidirectional interplay between AgRP and POMC neurocircuits.


Asunto(s)
Neuronas , Proopiomelanocortina , Ratones , Masculino , Femenino , Animales , Proopiomelanocortina/metabolismo , Proteína Relacionada con Agouti/metabolismo , Neuronas/metabolismo , Hipotálamo/metabolismo
2.
Nat Neurosci ; 24(7): 913-929, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34002087

RESUMEN

Pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus represent key regulators of metabolic homeostasis. Electrophysiological and single-cell sequencing experiments have revealed a remarkable degree of heterogeneity of these neurons. However, the exact molecular basis and functional consequences of this heterogeneity have not yet been addressed. Here, we have developed new mouse models in which intersectional Cre/Dre-dependent recombination allowed for successful labeling, translational profiling and functional characterization of distinct POMC neurons expressing the leptin receptor (Lepr) and glucagon like peptide 1 receptor (Glp1r). Our experiments reveal that POMCLepr+ and POMCGlp1r+ neurons represent largely nonoverlapping subpopulations with distinct basic electrophysiological properties. They exhibit a specific anatomical distribution within the arcuate nucleus and differentially express receptors for energy-state communicating hormones and neurotransmitters. Finally, we identify a differential ability of these subpopulations to suppress feeding. Collectively, we reveal a notably distinct functional microarchitecture of critical metabolism-regulatory neurons.


Asunto(s)
Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Neuronas/fisiología , Proopiomelanocortina/metabolismo , Animales , Metabolismo Energético/fisiología , Homeostasis/fisiología , Hipotálamo/citología , Ratones , Ratones Transgénicos , Neuronas/citología
3.
Cell Rep ; 25(2): 383-397.e10, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30304679

RESUMEN

Mitochondrial oxidative phosphorylation (OXPHOS) and substrate utilization critically regulate the function of hypothalamic proopiomelanocortin (POMC)-expressing neurons. Here, we demonstrate that inactivation of apoptosis-inducing factor (AIF) in POMC neurons mildly impairs mitochondrial respiration and decreases firing of POMC neurons in lean mice. In contrast, under diet-induced obese conditions, POMC-Cre-specific inactivation of AIF prevents obesity-induced silencing of POMC neurons, translating into improved glucose metabolism, improved leptin, and insulin sensitivity, as well as increased energy expenditure in AIFΔPOMC mice. On a cellular level, AIF deficiency improves mitochondrial morphology, facilitates the utilization of fatty acids for mitochondrial respiration, and increases reactive oxygen species (ROS) formation in POMC neurons from obese mice, ultimately leading to restored POMC firing upon HFD feeding. Collectively, partial impairment of mitochondrial function shifts substrate utilization of POMC neurons from glucose to fatty acid metabolism and restores their firing properties, resulting in improved systemic glucose and energy metabolism in obesity.


Asunto(s)
Ácidos Grasos/metabolismo , Glucosa/metabolismo , Homeostasis , Mitocondrias/patología , Neuronas/metabolismo , Obesidad/prevención & control , Fosforilación Oxidativa , Proopiomelanocortina/metabolismo , Animales , Factor Inductor de la Apoptosis/fisiología , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Intolerancia a la Glucosa , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Mitocondrias/metabolismo , Neuronas/patología , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología
4.
Nat Commun ; 9(1): 3432, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143607

RESUMEN

p53 is a well-known tumor suppressor that has emerged as an important player in energy balance. However, its metabolic role in the hypothalamus remains unknown. Herein, we show that mice lacking p53 in agouti-related peptide (AgRP), but not proopiomelanocortin (POMC) or steroidogenic factor-1 (SF1) neurons, are more prone to develop diet-induced obesity and show reduced brown adipose tissue (BAT) thermogenic activity. AgRP-specific ablation of p53 resulted in increased hypothalamic c-Jun N-terminal kinase (JNK) activity before the mice developed obesity, and central inhibition of JNK reversed the obese phenotype of these mice. The overexpression of p53 in the ARC or specifically in AgRP neurons of obese mice decreased body weight and stimulated BAT thermogenesis, resulting in body weight loss. Finally, p53 in AgRP neurons regulates the ghrelin-induced food intake and body weight. Overall, our findings provide evidence that p53 in AgRP neurons is required for normal adaptations against diet-induced obesity.


Asunto(s)
Dieta/efectos adversos , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Tejido Adiposo Pardo/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/genética , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Ratas Sprague-Dawley , Factor Esteroidogénico 1/metabolismo , Proteína p53 Supresora de Tumor/genética
5.
Cell Rep ; 23(6): 1728-1741, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29742429

RESUMEN

Anorexigenic pro-opiomelanocortin (Pomc)/alpha-melanocyte stimulating hormone (αMSH) neurons of the hypothalamic melanocortin system function as key regulators of energy homeostasis, also controlling somatic growth across different species. However, the mechanisms of melanocortin-dependent growth control still remain ill-defined. Here, we reveal a thus-far-unrecognized structural and functional connection between Pomc neurons and the somatotropic hypothalamo-pituitary axis. Excessive feeding of larval zebrafish causes leptin resistance and reduced levels of the hypothalamic satiety mediator pomca. In turn, this leads to reduced activation of hypophysiotropic somatostatin (Sst)-neurons that express the melanocortin receptor Mc4r, elevated growth hormone (GH) expression in the pituitary, and enhanced somatic growth. Mc4r expression and αMSH responsiveness are conserved in Sst-expressing hypothalamic neurons of mice. Thus, acquired leptin resistance and attenuation of pomca transcription in response to excessive caloric intake may represent an ancient mechanism to promote somatic growth when food resources are plentiful.


Asunto(s)
Dieta , Hormona del Crecimiento/metabolismo , Crecimiento y Desarrollo/efectos de los fármacos , Leptina/farmacología , Proopiomelanocortina/metabolismo , Somatostatina/metabolismo , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Larva/efectos de los fármacos , Larva/fisiología , Metabolismo de los Lípidos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Neural/efectos de los fármacos , Adenohipófisis/efectos de los fármacos , Adenohipófisis/metabolismo , Pez Cebra , alfa-MSH/metabolismo
6.
Cell Rep ; 17(10): 2512-2521, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27926856

RESUMEN

Melanin-concentrating-hormone (MCH)-expressing neurons (MCH neurons) in the lateral hypothalamus (LH) are critical regulators of energy and glucose homeostasis. Here, we demonstrate that insulin increases the excitability of these neurons in control mice. In vivo, insulin promotes phosphatidylinositol 3-kinase (PI3K) signaling in MCH neurons, and cell-type-specific deletion of the insulin receptor (IR) abrogates this response. While lean mice lacking the IR in MCH neurons (IRΔMCH) exhibit no detectable metabolic phenotype under normal diet feeding, they present with improved locomotor activity and insulin sensitivity under high-fat-diet-fed, obese conditions. Similarly, obesity promotes PI3 kinase signaling in these neurons, and this response is abrogated in IRΔMCH mice. In turn, acute chemogenetic activation of MCH neurons impairs locomotor activity but not insulin sensitivity. Collectively, our experiments reveal an insulin-dependent activation of MCH neurons in obesity, which contributes via distinct mechanisms to the manifestation of impaired locomotor activity and insulin resistance.


Asunto(s)
Hormonas Hipotalámicas/genética , Resistencia a la Insulina/genética , Insulina/metabolismo , Melaninas/genética , Obesidad/metabolismo , Hormonas Hipofisarias/genética , Animales , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Humanos , Hipotálamo/metabolismo , Insulina/administración & dosificación , Locomoción/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Obesidad/tratamiento farmacológico , Obesidad/patología , Fosfatidilinositol 3-Quinasas/genética
7.
Cell ; 162(6): 1404-17, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26359991

RESUMEN

Activation of orexigenic AgRP-expressing neurons in the arcuate nucleus of the hypothalamus potently promotes feeding, thus defining new regulators of AgRP neuron activity could uncover potential novel targets for obesity treatment. Here, we demonstrate that AgRP neurons express the purinergic receptor 6 (P2Y6), which is activated by uridine-diphosphate (UDP). In vivo, UDP induces ERK phosphorylation and cFos expression in AgRP neurons and promotes action potential firing of these neurons in brain slice recordings. Consequently, central application of UDP promotes feeding, and this response is abrogated upon pharmacologic or genetic inhibition of P2Y6 as well as upon pharmacogenetic inhibition of AgRP neuron activity. In obese animals, hypothalamic UDP content is elevated as a consequence of increased circulating uridine concentrations. Collectively, these experiments reveal a potential regulatory pathway in obesity, where peripheral uridine increases hypothalamic UDP concentrations, which in turn can promote feeding via PY6-dependent activation of AgRP neurons.


Asunto(s)
Regulación del Apetito , Hipotálamo/metabolismo , Obesidad/metabolismo , Receptores Purinérgicos P2/metabolismo , Uridina Difosfato/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Cell ; 156(3): 495-509, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24462248

RESUMEN

Maternal metabolic homeostasis exerts long-term effects on the offspring's health outcomes. Here, we demonstrate that maternal high-fat diet (HFD) feeding during lactation predisposes the offspring for obesity and impaired glucose homeostasis in mice, which is associated with an impairment of the hypothalamic melanocortin circuitry. Whereas the number and neuropeptide expression of anorexigenic proopiomelanocortin (POMC) and orexigenic agouti-related peptide (AgRP) neurons, electrophysiological properties of POMC neurons, and posttranslational processing of POMC remain unaffected in response to maternal HFD feeding during lactation, the formation of POMC and AgRP projections to hypothalamic target sites is severely impaired. Abrogating insulin action in POMC neurons of the offspring prevents altered POMC projections to the preautonomic paraventricular nucleus of the hypothalamus (PVH), pancreatic parasympathetic innervation, and impaired glucose-stimulated insulin secretion in response to maternal overnutrition. These experiments reveal a critical timing, when altered maternal metabolism disrupts metabolic homeostasis in the offspring via impairing neuronal projections, and show that abnormal insulin signaling contributes to this effect.


Asunto(s)
Dieta Alta en Grasa , Hiperglucemia/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Lactancia , Obesidad/metabolismo , Animales , Axones/metabolismo , Femenino , Masculino , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Embarazo , Proopiomelanocortina/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal
9.
Cell Calcium ; 54(2): 71-85, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23787148

RESUMEN

Calcium imaging has become a routine technique in neuroscience for subcellular to network level investigations. The fast progresses in the development of new indicators and imaging techniques call for dedicated reliable analysis methods. In particular, efficient and quantitative background fluorescence subtraction routines would be beneficial to most of the calcium imaging research field. A background-subtracted fluorescence transients estimation method that does not require any independent background measurement is therefore developed. This method is based on a fluorescence model fitted to single-trial data using a classical nonlinear regression approach. The model includes an appropriate probabilistic description of the acquisition system's noise leading to accurate confidence intervals on all quantities of interest (background fluorescence, normalized background-subtracted fluorescence time course) when background fluorescence is homogeneous. An automatic procedure detecting background inhomogeneities inside the region of interest is also developed and is shown to be efficient on simulated data. The implementation and performances of the proposed method on experimental recordings from the mouse hypothalamus are presented in details. This method, which applies to both single-cell and bulk-stained tissues recordings, should help improving the statistical comparison of fluorescence calcium signals between experiments and studies.


Asunto(s)
Calcio/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Hipotálamo/metabolismo , Modelos Biológicos , Neuronas/metabolismo , Imagen Óptica/métodos , Animales , Señalización del Calcio/fisiología , Hipotálamo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neuronas/citología , Proopiomelanocortina/metabolismo , Análisis de Regresión , Reproducibilidad de los Resultados , Factores de Tiempo
10.
J Neurosci ; 29(37): 11582-93, 2009 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-19759305

RESUMEN

Leptin-stimulated Stat3 activation in proopiomelanocortin (POMC)-expressing neurons of the hypothalamus plays an important role in maintenance of energy homeostasis. While Stat3 activation in POMC neurons is required for POMC expression, the role of elevated basal Stat3 activation as present in the development of obesity has not been directly addressed. Here, we have generated and characterized mice expressing a constitutively active version of Stat3 (Stat3-C) in POMC neurons (Stat3-C(POMC) mice). On normal chow diet, these animals develop obesity as a result of hyperphagia and decreased POMC expression accompanied by central leptin and insulin resistance. This unexpected finding coincides with POMC-cell-specific, Stat3-mediated upregulation of SOCS3 expression inhibiting both leptin and insulin signaling as insulin-stimulated PIP(3) (phosphatidylinositol-3,4,5 triphosphate) formation and protein kinase B (AKT) activation in POMC neurons as well as with the fact that insulin's ability to hyperpolarize POMC neurons is largely reduced in POMC cells of Stat3-C(POMC) mice. These data indicate that constitutive Stat3 activation is not sufficient to promote POMC expression but requires simultaneous PI3K (phosphoinositide 3-kinase)-dependent release of FOXO1 repression. In contrast, upon exposure to a high-fat diet, food intake and body weight were unaltered in Stat3-C(POMC) mice compared with control mice. Taken together, these experiments directly demonstrate that enhanced basal Stat3 activation in POMC neurons as present in control mice upon high-fat feeding contributes to the development of hypothalamic leptin and insulin resistance.


Asunto(s)
Insulina/metabolismo , Leptina/metabolismo , Proteínas de la Membrana/metabolismo , Inhibición Neural/fisiología , Neuronas/fisiología , Obesidad/fisiopatología , Proopiomelanocortina/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Composición Corporal/genética , Peso Corporal/genética , Modelos Animales de Enfermedad , Ingestión de Alimentos/genética , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática/métodos , Retroalimentación/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Prueba de Tolerancia a la Glucosa , Proteínas Fluorescentes Verdes/genética , Hipotálamo/patología , Técnicas In Vitro , Resistencia a la Insulina/genética , Factor Inhibidor de Leucemia/farmacología , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Inhibición Neural/efectos de los fármacos , Inhibición Neural/genética , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Obesidad/genética , Obesidad/metabolismo , Técnicas de Placa-Clamp/métodos , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Transfección
11.
Cell Metab ; 7(4): 291-301, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18396135

RESUMEN

Insulin- and leptin-stimulated phosphatidylinositol-3 kinase (PI3K) activation has been demonstrated to play a critical role in central control of energy homeostasis. To delineate the importance of pathways downstream of PI3K specifically in pro-opiomelanocortin (POMC) cell regulation, we have generated mice with selective inactivation of 3-phosphoinositide-dependent protein kinase 1 (PDK1) in POMC-expressing cells (PDK1(DeltaPOMC) mice). PDK1(DeltaPOMC) mice initially display hyperphagia, increased body weight, and impaired glucose metabolism caused by reduced hypothalamic POMC expression. On the other hand, PDK1(DeltaPOMC) mice exhibit progressive, severe hypocortisolism caused by loss of POMC-expressing corticotrophs in the pituitary. Expression of a dominant-negative mutant of FOXO1 specifically in POMC cells is sufficient to ameliorate positive energy balance in PDK1(DeltaPOMC) mice but cannot restore regular pituitary function. These results reveal important but differential roles for PDK1 signaling in hypothalamic and pituitary POMC cells in the control of energy homeostasis and stress response.


Asunto(s)
Metabolismo Energético , Factores de Transcripción Forkhead/metabolismo , Proopiomelanocortina/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Estrés Fisiológico , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Peso Corporal/efectos de los fármacos , Corticosterona/metabolismo , Corticosterona/farmacología , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Regulación de la Expresión Génica , Hiperfagia/genética , Hipotálamo/citología , Hipotálamo/metabolismo , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Hipófisis/metabolismo , Proopiomelanocortina/deficiencia , Proopiomelanocortina/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA