Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Neuroendocrinology ; 110(1-2): 92-104, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31104058

RESUMEN

In outbred mice, susceptibility or resistance to diet-induced obesity is associated with rapid changes in hypothalamic proopiomelanocortin (POMC) levels. Here, we evaluated 3 hypotheses that potentially explain the development of the different obesity phenotypes in outbred Swiss mice. First, rapid and differential changes in the gut microbiota in obesity-prone (OP) and obesity-resistant (OR) mice fed on a high-fat diet (HFD) might cause differential efficiencies in fatty acid harvesting leading to changes in systemic fatty acid concentrations that in turn affect POMC expression and processing. Second, independently of the gut microbiota, OP mice might have increased blood fatty acid levels after the introduction of a HFD, which could affect POMC expression and processing. Third, fatty acids might act directly in the hypothalamus to differentially regulate POMC expression and/or processing in OP and OR mice. We evaluated OP and OR male Swiss mice using 16S rRNA sequencing for the determination of gut microbiota; gas chromatography for blood lipid determination; and immunoblot and real-time polymerase chain reaction for protein and transcript determination and indirect calorimetry. Some experiments were performed with human pluripotent stem cells differentiated into hypothalamic neurons. We did not find evidence supporting the first 2 hypotheses. However, we found that in OP but not in OR mice, palmitate induces a rapid increase in hypothalamic POMC, which is followed by increased expression of proprotein convertase subtilisin/kexin type 1 PC1/3. Lentiviral inhibition of hypothalamic PC1/3 increased caloric intake and body mass in both OP and OR mice. In human stem cell-derived hypothalamic cells, we found that palmitate potently suppressed the production of POMC-derived peptides. Palmitate directly regulates PC1/3 in OP mice and likely has a functional impact on POMC processing.


Asunto(s)
Microbioma Gastrointestinal , Hipotálamo/metabolismo , Inflamación/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Palmitatos/farmacología , Proopiomelanocortina/metabolismo , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Humanos , Ácido Linoleico/farmacología , Masculino , Ratones , Obesidad/sangre , Obesidad/etiología , Células Madre Pluripotentes , ARN Ribosómico 16S
2.
Am J Physiol Endocrinol Metab ; 305(2): E230-42, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23695212

RESUMEN

Melatonin can contribute to glucose homeostasis either by decreasing gluconeogenesis or by counteracting insulin resistance in distinct models of obesity. However, the precise mechanism through which melatonin controls glucose homeostasis is not completely understood. Male Wistar rats were administered an intracerebroventricular (icv) injection of melatonin and one of following: an icv injection of a phosphatidylinositol 3-kinase (PI3K) inhibitor, an icv injection of a melatonin receptor (MT) antagonist, or an intraperitoneal (ip) injection of a muscarinic receptor antagonist. Anesthetized rats were subjected to pyruvate tolerance test to estimate in vivo glucose clearance after pyruvate load and in situ liver perfusion to assess hepatic gluconeogenesis. The hypothalamus was removed to determine Akt phosphorylation. Melatonin injections in the central nervous system suppressed hepatic gluconeogenesis and increased hypothalamic Akt phosphorylation. These effects of melatonin were suppressed either by icv injections of PI3K inhibitors and MT antagonists and by ip injection of a muscarinic receptor antagonist. We conclude that melatonin activates hypothalamus-liver communication that may contribute to circadian adjustments of gluconeogenesis. These data further suggest a physiopathological relationship between the circadian disruptions in metabolism and reduced levels of melatonin found in type 2 diabetes patients.


Asunto(s)
Antioxidantes/farmacología , Gluconeogénesis/efectos de los fármacos , Hipotálamo/metabolismo , Hígado/metabolismo , Melatonina/farmacología , Proteína Oncogénica v-akt/metabolismo , Receptor de Melatonina MT1/efectos de los fármacos , Receptor de Melatonina MT2/efectos de los fármacos , Animales , Western Blotting , Técnica del Anticuerpo Fluorescente , Prueba de Tolerancia a la Glucosa , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Hígado/efectos de los fármacos , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Ácido Pirúvico/metabolismo , Ratas , Ratas Wistar , Receptores Muscarínicos/efectos de los fármacos
3.
Sci Rep ; 9(1): 13068, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31506541

RESUMEN

Leptin regulates both feeding and glycaemia primarily through its receptors expressed on agouti-related peptide (AgRP) and pro-opiomelanocortin-expressing (POMC) neurons; however, it is unknown whether activity of these neuronal populations mediates the regulation of these processes. To determine this, we injected Cre-dependent designer receptors exclusively activated by designer drugs (DREADD) viruses into the hypothalamus of normoglycaemic and diabetic AgRP-ires-cre and POMC-cre mice to chemogenetically activate or inhibit these neuronal populations. Despite robust changes in food intake, activation or inhibition of AgRP neurons did not affect glycaemia, while activation caused significant (P = 0.014) impairment in insulin sensitivity. Stimulation of AgRP neurons in diabetic mice reversed leptin's ability to inhibit feeding but did not counter leptin's ability to lower blood glucose levels. Notably, the inhibition of POMC neurons stimulated feeding while decreasing glucose levels in normoglycaemic mice. The findings suggest that leptin's effects on feeding by AgRP neurons are mediated by changes in neuronal firing, while the control of glucose balance by these cells is independent of chemogenetic activation or inhibition. The firing-dependent glucose lowering mechanism within POMC neurons is a potential target for the development of novel anti-diabetic medicines.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Glucemia , Glucosa/metabolismo , Neuronas/metabolismo , Proproteína Convertasas/metabolismo , Animales , Diabetes Mellitus Experimental , Ingestión de Alimentos , Intolerancia a la Glucosa , Resistencia a la Insulina , Leptina/metabolismo , Ratones , Modelos Biológicos
4.
J Biomed Nanotechnol ; 9(6): 1098-106, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23858976

RESUMEN

The aim of this work was to develop an efficient, biodegradable, biocompatible and safe controlled release system using insulin-loaded poly(epsilon-caprolactone) (PCL) nanoparticles. The insulin-loaded PCL nanoparticles were prepared by double emulsion method (water-in-oil-in-water) using Pluronic F68 as emulsifier. Using the double emulsion method a high insulin encapsulation efficiency (90.6 +/-1.6%) with a zeta potential of -29 +/-2.7 mV and average particle size of 796 +/-10.5 nm was obtained. Insulin-loaded PCL nanoparticles showed no toxicity to MIN6 cells. Insulin nanoparticles administered subcutaneously and intraperitoneally in rats reduced glycaemia of basal levels after 15 minutes, and presented a sustainable hypoglycemic effect on insulin-dependent type 1 diabetic rats, showing to be more efficient than unencapsulated insulin. Furthermore, these nanoparticles were not hepatotoxic, as evaluated by the effect over liver cell-death and oxidative stress scavenger system in rats. These results suggest that insulin-loaded PCL nanoparticles prepared by water-in-oil-in-water emulsion method are biocompatible, efficient and safe insulin-delivering system with controlled insulin release, which indicates that it may be a powerful tool for insulin-dependent patients care.


Asunto(s)
Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/síntesis química , Diabetes Mellitus Experimental/tratamiento farmacológico , Insulina/administración & dosificación , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Poliésteres/química , Animales , Diabetes Mellitus Experimental/diagnóstico , Difusión , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/química , Insulina/química , Masculino , Ensayo de Materiales , Ratas , Ratas Wistar , Estreptozocina
5.
Endocrinology ; 153(8): 3633-45, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22585831

RESUMEN

Fructose consumption causes insulin resistance and favors hepatic gluconeogenesis through mechanisms that are not completely understood. Recent studies demonstrated that the activation of hypothalamic 5'-AMP-activated protein kinase (AMPK) controls dynamic fluctuations in hepatic glucose production. Thus, the present study was designed to investigate whether hypothalamic AMPK activation by fructose would mediate increased gluconeogenesis. Both ip and intracerebroventricular (icv) fructose treatment stimulated hypothalamic AMPK and acetyl-CoA carboxylase phosphorylation, in parallel with increased hepatic phosphoenolpyruvate carboxy kinase (PEPCK) and gluconeogenesis. An increase in AMPK phosphorylation by icv fructose was observed in the lateral hypothalamus as well as in the paraventricular nucleus and the arcuate nucleus. These effects were mimicked by icv 5-amino-imidazole-4-carboxamide-1-ß-d-ribofuranoside treatment. Hypothalamic AMPK inhibition with icv injection of compound C or with injection of a small interfering RNA targeted to AMPKα2 in the mediobasal hypothalamus (MBH) suppressed the hepatic effects of ip fructose. We also found that fructose increased corticosterone levels through a mechanism that is dependent on hypothalamic AMPK activation. Concomitantly, fructose-stimulated gluconeogenesis, hepatic PEPCK expression, and glucocorticoid receptor binding to the PEPCK gene were suppressed by pharmacological glucocorticoid receptor blockage. Altogether the data presented herein support the hypothesis that fructose-induced hypothalamic AMPK activation stimulates hepatic gluconeogenesis by increasing corticosterone levels.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Corticosterona/metabolismo , Fructosa/farmacología , Gluconeogénesis/efectos de los fármacos , Hipotálamo/metabolismo , Hígado/metabolismo , Animales , Inmunoprecipitación de Cromatina , Activación Enzimática/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Hipotálamo/efectos de los fármacos , Immunoblotting , Hígado/efectos de los fármacos , Masculino , Fosfoenolpiruvato Carboxiquinasa (ATP) , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda