Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Biochim Biophys Acta Gen Subj ; 1862(3): 403-413, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29154902

RESUMEN

BACKGROUND: During the development of obesity the expansion of white adipose tissue (WAT) leads to a dysregulation and an excessive remodeling of extracellular matrix (ECM), leading to fibrosis formation. These ECM changes have high impact on WAT physiology and may change obesity progression. Blocking WAT fibrosis may have beneficial effects on the efficacy of diet regimen or therapeutical approaches in obesity. Since dipeptidyl peptidase IV (DPP-IV) inhibitors prevent fibrosis in tissues, such as heart, liver and kidney, the objective of this study was to assess whether vildagliptin, a DPP-IV inhibitor, prevents fibrosis in WAT in a mouse model of obesity, and to investigate the mechanisms underlying this effect. METHODS: We evaluated the inhibitory effect of vildagliptin on fibrosis markers on WAT of high-fat diet (HFD)-induced obese mice and on 3T3-L1 cell line of mouse adipocytes treated with a fibrosis inducer, transforming growth factor beta 1 (TGFß1). RESULTS: Vildagliptin prevents the increase of fibrosis markers in WAT of HFD-fed mice and reduces blood glucose, serum triglycerides, total cholesterol and leptin levels. In the in vitro study, the inhibition of DPP-IV with vildagliptin, neuropeptide Y (NPY) treatment and NPY Y1 receptor activation prevents ECM deposition and fibrosis markers increase induced by TGFß1 treatment. CONCLUSIONS: Vildagliptin prevents fibrosis formation in adipose tissue in obese mice, at least partially through NPY and NPY Y1 receptor activation. GENERAL SIGNIFICANCE: This study highlights the importance of vildagliptin in the treatment of fibrosis that occur in obesity.


Asunto(s)
Adamantano/análogos & derivados , Tejido Adiposo Blanco/efectos de los fármacos , Dipeptidil Peptidasa 4/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Hipolipemiantes/uso terapéutico , Nitrilos/uso terapéutico , Obesidad/tratamiento farmacológico , Pirrolidinas/uso terapéutico , Células 3T3-L1 , Adamantano/farmacología , Adamantano/uso terapéutico , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo Blanco/patología , Animales , Glucemia/análisis , Colágeno/metabolismo , Dieta Alta en Grasa , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrosis , Hipolipemiantes/farmacología , Leptina/sangre , Leptina/fisiología , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Neuropéptido Y/agonistas , Neuropéptido Y/farmacología , Neuropéptido Y/fisiología , Nitrilos/farmacología , Obesidad/patología , Pirrolidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/fisiología , Factor de Crecimiento Transformador beta1/farmacología , Vildagliptina
2.
Proc Natl Acad Sci U S A ; 112(13): E1642-51, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25775546

RESUMEN

Aging is characterized by autophagy impairment that contributes to age-related disease aggravation. Moreover, it was described that the hypothalamus is a critical brain area for whole-body aging development and has impact on lifespan. Neuropeptide Y (NPY) is one of the major neuropeptides present in the hypothalamus, and it has been shown that, in aged animals, the hypothalamic NPY levels decrease. Because caloric restriction (CR) delays aging, at least in part, by stimulating autophagy, and also increases hypothalamic NPY levels, we hypothesized that NPY could have a relevant role on autophagy modulation in the hypothalamus. Therefore, the aim of this study was to investigate the role of NPY on autophagy in the hypothalamus. Using both hypothalamic neuronal in vitro models and mice overexpressing NPY in the hypothalamus, we observed that NPY stimulates autophagy in the hypothalamus. Mechanistically, in rodent hypothalamic neurons, NPY increases autophagy through the activation of NPY Y1 and Y5 receptors, and this effect is tightly associated with the concerted activation of PI3K, MEK/ERK, and PKA signaling pathways. Modulation of hypothalamic NPY levels may be considered a potential strategy to produce protective effects against hypothalamic impairments associated with age and to delay aging.


Asunto(s)
Autofagia , Hipotálamo/citología , Neuronas/citología , Neuropéptido Y/fisiología , Envejecimiento , Animales , Encéfalo/metabolismo , Restricción Calórica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Transducción de Señal
3.
Brain ; 134(Pt 5): 1400-15, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21478185

RESUMEN

Machado-Joseph disease, also known as spinocerebellar ataxia type 3, is the most common of the dominantly inherited ataxias worldwide and is characterized by mutant ataxin-3 misfolding, intracellular accumulation of aggregates and neuronal degeneration. Here we investigated the implication of autophagy, the major pathway for organelle and protein turnover, in the accumulation of mutant ataxin-3 aggregates and neurodegeneration found in Machado-Joseph disease and we assessed whether specific stimulation of this pathway could mitigate the disease. Using tissue from patients with Machado-Joseph disease, transgenic mice and a lentiviral-based rat model, we found an abnormal expression of endogenous autophagic markers, accumulation of autophagosomes and decreased levels of beclin-1, a crucial protein in the early nucleation step of autophagy. Lentiviral vector-mediated overexpression of beclin-1 led to stimulation of autophagic flux, mutant ataxin-3 clearance and overall neuroprotective effects in neuronal cultures and in a lentiviral-based rat model of Machado-Joseph disease. These data demonstrate that autophagy is a key degradation pathway, with beclin-1 playing a significant role in alleviating Machado-Joseph disease pathogenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/genética , Enfermedad de Machado-Joseph/genética , Proteínas de la Membrana/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Anciano , Animales , Proteínas Reguladoras de la Apoptosis/genética , Ataxina-3 , Proteínas Relacionadas con la Autofagia , Beclina-1 , Encéfalo/metabolismo , Encéfalo/patología , Proteínas Portadoras/genética , Línea Celular Tumoral , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/genética , Humanos , Enfermedad de Machado-Joseph/patología , Enfermedad de Machado-Joseph/fisiopatología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Ratas , Ratas Wistar , Proteína Sequestosoma-1 , Transfección/métodos , Expansión de Repetición de Trinucleótido/genética
4.
Trends Endocrinol Metab ; 25(2): 80-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24231724

RESUMEN

The recently described generation of new neurons in the adult hypothalamus, the center for energy regulation, suggests that hypothalamic neurogenesis is a crucial part of the mechanisms that regulate food intake. Accordingly, neurogenesis in both the adult and embryonic hypothalamus is affected by nutritional cues and metabolic disorders such as obesity, with consequent effects on energy-balance. This review critically discusses recent findings on the contribution of adult hypothalamic neurogenesis to feeding regulation, the impact of energy-balance disorders on adult hypothalamic neurogenesis, and the influence of embryonic hypothalamic neurogenesis upon feeding regulation in the adult. Understanding how hypothalamic neurogenesis contributes to food intake control will change the paradigm on how we perceive energy-balance regulation.


Asunto(s)
Ingestión de Alimentos/fisiología , Hipotálamo/fisiología , Neurogénesis , Adulto , Animales , Metabolismo Energético/fisiología , Humanos , Hipotálamo/embriología , Células-Madre Neurales/fisiología , Ratas , Factores de Transcripción/fisiología
5.
PLoS One ; 9(3): e88917, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24598761

RESUMEN

A significant number of children undergo maternal exposure to antidepressants and they often present low birth weight. Therefore, it is important to understand how selective serotonin reuptake inhibitors (SSRIs) affect the development of the hypothalamus, the key center for metabolism regulation. In this study we investigated the proliferative actions of fluoxetine in fetal hypothalamic neuroprogenitor cells and demonstrate that fluoxetine induces the proliferation of these cells, as shown by increased neurospheres size and number of proliferative cells (Ki-67+ cells). Moreover, fluoxetine inhibits the differentiation of hypothalamic neuroprogenitor cells, as demonstrated by decreased number of mature neurons (Neu-N+ cells) and increased number of undifferentiated cells (SOX-2+ cells). Additionally, fluoxetine-induced proliferation and maintenance of hypothalamic neuroprogenitor cells leads to changes in the mRNA levels of appetite regulator neuropeptides, including Neuropeptide Y (NPY) and Cocaine-and-Amphetamine-Regulated-Transcript (CART). This study provides the first evidence that SSRIs affect the development of hypothalamic neuroprogenitor cells in vitro with consequent alterations on appetite neuropeptides.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Fluoxetina/farmacología , Hipotálamo/citología , Células-Madre Neurales/efectos de los fármacos , Animales , Antidepresivos de Segunda Generación/efectos adversos , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Fluoxetina/efectos adversos , Expresión Génica/efectos de los fármacos , Humanos , Hipotálamo/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Embarazo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Ratas , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Esferoides Celulares/efectos de los fármacos
6.
PLoS One ; 6(5): e19745, 2011 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-21589937

RESUMEN

Some pathological conditions with feeding pattern alterations, including obesity and Huntington disease (HD) are associated with hypothalamic dysfunction and neuronal cell death. Additionally, the hypothalamus is a neurogenic region with the constitutive capacity to generate new cells of neuronal lineage, in adult rodents. The aim of the present work was to evaluate the expression of feeding-related neuropeptides in hypothalamic progenitor cells and their capacity to differentiate to functional neurons which have been described to be affected by hypothalamic dysfunction. Our study shows that hypothalamic progenitor cells from rat embryos grow as floating neurospheres and express the feeding-related neuropeptides Neuropeptide Y (NPY), Agouti-related Protein (AGRP), Pro-OpioMelanocortin (POMC), Cocaine-and-Amphetamine Responsive Transcript (CART) and Orexin-A/Hypocretin-1. Moreover the relative mRNA expression of NPY and POMC increases during the expansion of hypothalamic neurospheres in proliferative conditions.Mature neurons were obtained from the differentiation of hypothalamic progenitor cells including NPY, AGRP, POMC, CART and Orexin-A positive neurons. Furthermore the relative mRNA expression of NPY, CART and Orexin-A increases after the differentiation of hypothalamic neurospheres. Similarly to the adult hypothalamic neurons the neurospheres-derived neurons express the glutamate transporter EAAT3. The orexigenic and anorexigenic phenotype of these neurons was identified by functional response to ghrelin and leptin hormones, respectively. This work demonstrates the presence of appetite-related neuropeptides in hypothalamic progenitor cells and neurons obtained from the differentiation of hypothalamic neurospheres, including the neuronal phenotypes that have been described by others as being affected by hypothalamic neurodegeneration. These in vitro models can be used to study hypothalamic progenitor cells aiming a therapeutic intervention to mitigate feeding dysfunction that are associated with hypothalamic neurodegeneration.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Neuropéptidos/metabolismo , Proopiomelanocortina/metabolismo , Animales , Diferenciación Celular , Femenino , Hipotálamo/citología , Neuronas/citología , Orexinas , Ratas , Ratas Wistar
7.
PLoS One ; 6(7): e22333, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21799827

RESUMEN

Neuropeptide Y (NPY) produced by arcuate nucleus (ARC) neurons has a strong orexigenic effect on target neurons. Hypothalamic NPY levels undergo wide-ranging oscillations during the circadian cycle and in response to fasting and peripheral hormones (from 0.25 to 10-fold change). The aim of the present study was to evaluate the impact of a moderate long-term modulation of NPY within the ARC neurons on food consumption, body weight gain and hypothalamic neuropeptides. We achieved a physiological overexpression (3.6-fold increase) and down-regulation (0.5-fold decrease) of NPY in the rat ARC by injection of AAV vectors expressing NPY and synthetic microRNA that target the NPY, respectively. Our work shows that a moderate overexpression of NPY was sufficient to induce diurnal over-feeding, sustained body weight gain and severe obesity in adult rats. Additionally, the circulating levels of leptin were elevated but the immunoreactivity (ir) of ARC neuropeptides was not in accordance (POMC-ir was unchanged and AGRP-ir increased), suggesting a disruption in the ability of ARC neurons to response to peripheral metabolic alterations. Furthermore, a dysfunction in adipocytes phenotype was observed in these obese rats. In addition, moderate down-regulation of NPY did not affect basal feeding or normal body weight gain but the response to food deprivation was compromised since fasting-induced hyperphagia was inhibited and fasting-induced decrease in locomotor activity was absent.These results highlight the importance of the physiological ARC NPY levels oscillations on feeding regulation, fasting response and body weight preservation, and are important for the design of therapeutic interventions for obesity that include the NPY.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético , Neuropéptido Y/metabolismo , Adipocitos/citología , Adipogénesis/genética , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/fisiología , Tamaño de la Célula , Dependovirus/genética , Regulación hacia Abajo , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Ayuno/metabolismo , Retroalimentación Fisiológica , Vectores Genéticos/genética , Células HEK293 , Humanos , Hiperfagia/genética , Masculino , MicroARNs/genética , Neuronas/metabolismo , Neuropéptido Y/sangre , Neuropéptido Y/genética , Obesidad/sangre , PPAR gamma/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo , Aumento de Peso/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA