Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Metab ; 28: 120-134, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31446151

RESUMEN

OBJECTIVE: The long-acting glucagon-like peptide-1 receptor (GLP-1R) agonist, liraglutide, stimulates insulin secretion and efficiently suppresses food intake to reduce body weight. As such, liraglutide is growing in popularity in the treatment of diabetes and chronic weight management. Within the brain, liraglutide has been shown to alter the activity of hypothalamic proopiomelanocortin (POMC) and Neuropeptide Y/Agouti-related peptide (NPY/AgRP) neurons. Moreover, the acute activities of POMC and NPY neurons have been directly linked to feeding behavior, body weight, and glucose metabolism. Despite the increased usage of liraglutide and other GLP-1 analogues as diabetic and obesity interventions, the cellular mechanisms by which liraglutide alters the activity of metabolically relevant neuronal populations are poorly understood. METHODS: In order to resolve this issue, we utilized neuron-specific transgenic mouse models to identify POMC and NPY neurons for patch-clamp electrophysiology experiments. RESULTS: We found that liraglutide directly activated arcuate POMC neurons via TrpC5 channels, sharing a similar mechanistic pathway to the adipose-derived peptide leptin. Liraglutide also indirectly increases excitatory tone to POMC neurons. In contrast, liraglutide inhibited NPY/AgRP neurons through post-synaptic GABAA receptors and enhanced activity of pre-synaptic GABAergic neurons, which required both TrpC5 subunits and K-ATP channels. In support of an additive role of leptin and liraglutide in suppressing food intake, leptin potentiated the acute effects of liraglutide to activate POMC neurons. TrpC5 subunits in POMC neurons were also required for the intact pharmacological effects of liraglutide on food intake and body weight. Thus, the current study adds to recent work from our group and others, which highlight potential mechanisms to amplify the effects of GLP-1 agonists in vivo. Moreover, these data highlight multiple sites of action (both pre- and post-synaptic) for GLP-1 agonists on this circuit. CONCLUSIONS: Taken together, our results identify critical molecular mechanisms linking GLP-1 analogues in arcuate POMC and NPY/AgRP neurons with metabolism.


Asunto(s)
Proteína Relacionada con Agouti/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Hipotálamo/efectos de los fármacos , Liraglutida/farmacología , Neuronas/efectos de los fármacos , Neuropéptido Y/antagonistas & inhibidores , Proopiomelanocortina/antagonistas & inhibidores , Proteína Relacionada con Agouti/metabolismo , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Metabolismo Energético/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo
2.
Neuroendocrinology ; 98(3): 233-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24080898

RESUMEN

Clusterin is a sulfated glycoprotein abundantly expressed in the pituitary gland and hypothalamus of mammals. However, its physiological role in neuroendocrine function is largely unknown. In the present study, we investigated the effects of intracerebroventricular (ICV) administration of clusterin on plasma pituitary hormone levels in normal rats. Single ICV injection of clusterin provoked neurohormonal changes seen under acute stress condition: increased plasma adrenocorticotropic hormone (ACTH), corticosterone, GH and prolactin levels and decreased LH and FSH levels. Consistently, hypothalamic and pituitary clusterin expression levels were upregulated following a restraint stress, suggesting an involvement of endogenous clusterin in stress-induced neurohormonal changes. In the pituitary intermediate lobe, clusterin was coexpressed with proopiomelanocortin (POMC), a precursor of ACTH. Treatment of clusterin in POMC expressing AtT-20 pituitary cells increased basal and corticotropin-releasing hormone (CRH)-stimulated POMC promoter activities and intracellular cAMP levels. Furthermore, clusterin treatment triggered ACTH secretion from AtT-20 cells in a CRH-dependent manner, indicating that increased clusterin under stressful conditions may augment CRH-stimulated ACTH production and release. In summary, hypothalamic and pituitary clusterin may function as a modulator of neurohormonal responses under stressful conditions.


Asunto(s)
Clusterina/fisiología , Hipotálamo/metabolismo , Neurotransmisores/biosíntesis , Hipófisis/metabolismo , Hormona Adrenocorticotrópica/antagonistas & inhibidores , Hormona Adrenocorticotrópica/biosíntesis , Hormona Adrenocorticotrópica/metabolismo , Animales , Clusterina/administración & dosificación , Clusterina/sangre , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Masculino , Neurotransmisores/antagonistas & inhibidores , Neurotransmisores/metabolismo , Hipófisis/efectos de los fármacos , Proopiomelanocortina/antagonistas & inhibidores , Proopiomelanocortina/biosíntesis , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/sangre , Estrés Psicológico/prevención & control , Estrés Psicológico/psicología , Regulación hacia Arriba/fisiología
3.
Alcohol Clin Exp Res ; 37(7): 1133-42, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23413810

RESUMEN

BACKGROUND: Prenatal exposure to ethanol (EtOH) reduces the expression of hypothalamic proopiomelanocortin (POMC) gene, known to control various physiological functions including the organismal stress response. In this study, we determined whether the changes in POMC neuronal functions are associated with altered expressions of histone-modifying and DNA-methylating enzymes in POMC-producing neurons, because these enzymes are known to be involved in regulation of gene expression. In addition, we tested whether gestational choline supplementation prevents the adverse effects of EtOH on these neurons. METHODS: Pregnant rat dams were fed with alcohol-containing liquid diet or control diet during gestational days 7 and 21 with or without choline, and their male offspring rats were used during the adult period. Using double-immunohistochemistry, real-time reverse transcription polymerase chain reaction (RT-PCR) and methylation-specific RT-PCR, we determined protein and mRNA levels of histone-modifying and DNA-methylating enzymes and the changes in POMC gene methylation and expression in the hypothalamus of adult male offspring rats. Additionally, we measured the basal- and lipopolysaccharide (LPS)-induced corticosterone levels in plasma by enzyme-linked immunosorbent assay. RESULTS: Prenatal EtOH treatment suppressed hypothalamic levels of protein and mRNA of histone activation marks (H3K4me3, Set7/9, acetylated H3K9, phosphorylated H3S10), and increased the repressive marks (H3K9me2, G9a, Setdb1), DNA-methylating enzyme (Dnmt1), and the methyl-CpG-binding protein (MeCP2). The treatment also elevated the level of POMC gene methylation, while it reduced levels of POMC mRNA and ß-EP and elevated corticosterone response to LPS. Gestational choline normalized the EtOH-altered protein and the mRNA levels of H3K4me3, Set7/9, H3K9me2, G9a, Setdb1, Dnmt1, and MeCP2. It also normalizes the changes in POMC gene methylation and gene expression, ß-EP production, and the corticosterone response to LPS. CONCLUSIONS: These data suggest that prenatal EtOH modulates histone and DNA methylation in POMC neurons that may be resulting in hypermethylation of POMC gene and reduction in POMC gene expression. Gestational choline supplementation prevents the adverse effects of EtOH on these neurons.


Asunto(s)
Colina/administración & dosificación , Metilación de ADN/efectos de los fármacos , Histonas/antagonistas & inhibidores , Efectos Tardíos de la Exposición Prenatal/prevención & control , Proopiomelanocortina/antagonistas & inhibidores , betaendorfina/antagonistas & inhibidores , Animales , Metilación de ADN/fisiología , Suplementos Dietéticos , Femenino , Histonas/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proopiomelanocortina/biosíntesis , Ratas , betaendorfina/biosíntesis
4.
J Neurosci ; 33(8): 3624-32, 2013 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-23426689

RESUMEN

POMC-derived melanocortins inhibit food intake. In the adult rodent brain, POMC-expressing neurons are located in the arcuate nucleus (ARC) and the nucleus tractus solitarius (NTS), but it remains unclear how POMC neurons in these two brain nuclei regulate feeding behavior and metabolism differentially. Using pharmacogenetic methods to activate or deplete neuron groups in separate brain areas, in the present study, we show that POMC neurons in the ARC and NTS suppress feeding behavior at different time scales. Neurons were activated using the DREADD (designer receptors exclusively activated by designer drugs) method. The evolved human M3-muscarinic receptor was expressed in a selective population of POMC neurons by stereotaxic infusion of Cre-recombinase-dependent, adeno-associated virus vectors into the ARC or NTS of POMC-Cre mice. After injection of the human M3-muscarinic receptor ligand clozapine-N-oxide (1 mg/kg, i.p.), acute activation of NTS POMC neurons produced an immediate inhibition of feeding behavior. In contrast, chronic stimulation was required for ARC POMC neurons to suppress food intake. Using adeno-associated virus delivery of the diphtheria toxin receptor gene, we found that diphtheria toxin-induced ablation of POMC neurons in the ARC but not the NTS, increased food intake, reduced energy expenditure, and ultimately resulted in obesity and metabolic and endocrine disorders. Our results reveal different behavioral functions of POMC neurons in the ARC and NTS, suggesting that POMC neurons regulate feeding and energy homeostasis by integrating long-term adiposity signals from the hypothalamus and short-term satiety signals from the brainstem.


Asunto(s)
Tronco Encefálico/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Inhibición Neural/fisiología , Neuronas/fisiología , Proopiomelanocortina/fisiología , Adiposidad/genética , Animales , Tronco Encefálico/virología , Dependovirus/genética , Femenino , Vectores Genéticos/administración & dosificación , Células HEK293 , Homeostasis/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Inhibición Neural/genética , Vías Nerviosas/fisiopatología , Neuronas/virología , Proopiomelanocortina/antagonistas & inhibidores
5.
Endocrinology ; 148(2): 507-11, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17095593

RESUMEN

The oral antidiabetic agent metformin acts at least partially via an activation of AMP-activated kinase (AMPK) in liver and muscle cells. It has appeared recently that hypothalamic AMPK is a key regulator of feeding in mammals. Because metformin also exhibits anorectic effects in animal models as well as in humans, we hypothesized that AMPK may be a target of metformin in hypothalamic neurons. In this study, we show that, in primary cultures of rat hypothalamic neurons, low glucose levels stimulate the phosphorylation of AMPK, thus increasing neuropeptide Y (NPY) gene expression. The addition of metformin in low glucose conditions was found to block AMPK phosphorylation. Consistently, the stimulation of NPY observed in low glucose conditions was also inhibited by the drug. Proopiomelanocortin gene expression measured in parallel was inhibited under low glucose conditions, but in contrast to NPY, it was not dependent upon AMPK and not affected by metformin. Taken together, our data demonstrate that metformin can inhibit AMPK activity in hypothalamic neurons, thus modulating the expression of the orexigenic peptide NPY. These results provide, for the first time, a potential mechanism of action for the anorectic effects of metformin, a widely used drug that could represent a valuable adjunct to novel therapies aimed at modulating central feeding pathways.


Asunto(s)
Hipoglucemiantes/farmacología , Hipotálamo/metabolismo , Metformina/farmacología , Complejos Multienzimáticos/antagonistas & inhibidores , Neuronas/metabolismo , Neuropéptido Y/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Glucosa/administración & dosificación , Glucosa/farmacología , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Neuronas/efectos de los fármacos , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Concentración Osmolar , Fosforilación/efectos de los fármacos , Proopiomelanocortina/antagonistas & inhibidores , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas
6.
J Pharm Sci ; 87(5): 616-25, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9572914

RESUMEN

An oligodeoxynucleic sequence of 30 bases (30-mer ODN), complementary to a region of beta-endorphin mRNA, was synthesized to have an antisense effect with regard to the expression of this oligopeptide. Following the solid-phase synthesis of the oligodeoxynucleotide, the 30-mer ODN was encapsulated within liposomes to provide a higher resistance against DNases and an improved entrance into cells. The most suitable liposome formulation as a 30-mer ODN carrier consisted of small unilamellar vesicles (50 nm) with an encapsulation capacity of 4.76 microL/micromol. The liposomal formulations containing dipalmitoyl-DL-alpha-phosphatidyl-L-serine presented fusogenic properties, which are of great importance for the delivery of antisense compounds. The antisense activity of 30-mer ODN-loaded liposomes was evaluated by the determination of beta-endorphin levels in AtT-20 cells. The free 30-mer ODN did not provide any lowering of the beta-endorphin production, whereas the liposomally entrapped compound elicited a concentration-dependent inhibition. The inhibition was determined by a sequence-specific binding of the 30-mer ODN with the target mRNA.


Asunto(s)
Expresión Génica/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Proopiomelanocortina/antagonistas & inhibidores , 1,2-Dipalmitoilfosfatidilcolina/química , Animales , Rastreo Diferencial de Calorimetría , Línea Celular , Colesterol/química , Dimiristoilfosfatidilcolina/química , Portadores de Fármacos , Liposomas , Ratones , Oligonucleótidos Antisentido/síntesis química , Oligonucleótidos Antisentido/química , Fosfatidilserinas/química , Proopiomelanocortina/química , ARN Mensajero/química , Espectrometría de Fluorescencia , betaendorfina/antagonistas & inhibidores , betaendorfina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA