Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 12(8)2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37190103

RESUMEN

Stimulation of hepatic sympathetic nerves increases glucose production and glycogenolysis. Activity of pre-sympathetic neurons in the paraventricular nucleus (PVN) of the hypothalamus and in the ventrolateral and ventromedial medulla (VLM/VMM) largely influence the sympathetic output. Increased activity of the sympathetic nervous system (SNS) plays a role in the development and progression of metabolic diseases; however, despite the importance of the central circuits, the excitability of pre-sympathetic liver-related neurons remains to be determined. Here, we tested the hypothesis that the activity of liver-related neurons in the PVN and VLM/VMM is altered in diet-induced obese mice, as well as their response to insulin. Patch-clamp recordings were conducted from liver-related PVN neurons, VLM-projecting PVN neurons, and pre-sympathetic liver-related neurons in the ventral brainstem. Our data demonstrate that the excitability of liver-related PVN neurons increased in high-fat diet (HFD)-fed mice compared to mice fed with control diet. Insulin receptor expression was detected in a population of liver-related neurons, and insulin suppressed the firing activity of liver-related PVN and pre-sympathetic VLM/VMM neurons in HFD mice; however, it did not affect VLM-projecting liver-related PVN neurons. These findings further suggest that HFD alters the excitability of pre-autonomic neurons as well as their response to insulin.


Asunto(s)
Dieta Alta en Grasa , Insulinas , Ratones , Animales , Neuronas/metabolismo , Hígado , Encéfalo , Insulinas/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-36656675

RESUMEN

Aims: Part of hypothalamic (mediobasal hypothalamus [MBH]) neurons detect changes in blood glucose levels that in turn coordinate the vagal control of insulin secretion. This control cascade requires the production of mitochondrial reactive oxygen species (mROS), which is altered in models of obesity and insulin resistance. Obese, insulin-resistant Zücker rats are characterized by hypothalamic hypersensitivity to glucose. This initiates an abnormal vagus-induced insulin secretion, associated with an overproduction of mROS in response to a low glucose dose. Here, we hypothesized that ghrelin, known to buffer reactive oxygen species (ROS) via mitochondrial function, may be a major component of the hypothalamic glucose hypersensitivity in the hypoghrelinemic obese Zücker rat. Results: Hypothalamic glucose hypersensitivity-induced insulin secretion of Zücker obese rats was reversed by ghrelin pretreatment. The overproduction of MBH mROS in response to a low glucose load no longer occurred in obese rats that had previously received the cerebral ghrelin infusion. This decrease in mROS production was accompanied by a normalization of oxidative phosphorylation (OXPHOS). Conversely, blocking the action of ghrelin with a growth hormone secretagogue receptor antagonist in a model of hyperghrelinemia (fasted rats) completely restored hypothalamic glucose sensing-induced insulin secretion that was almost absent in this physiological situation. Accordingly, ROS signaling and mitochondrial activity were increased by the ghrelin receptor antagonist. Innovation: These results demonstrate for the first time that ghrelin addressed only to the brain could have a protective effect on the defective control of insulin secretion in the insulin-resistant, hypoghrelinemic obese subject. Conclusions: Ghrelin, through its action on OXPHOS, modulates mROS signaling in response to cerebral hyperglycemia and the consequent vagal control of insulin secretion. In insulin-resistant obese states, brain hypoghrelinemia could be responsible for the nervous defect in insulin secretion.

3.
Auton Neurosci ; 245: 103058, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36538864

RESUMEN

Brown adipose tissue (BAT) contributes to energy homeostasis via nonshivering thermogenesis. The BAT is densely innervated by the sympathetic nervous system (SNS) and activity of pre-autonomic neurons modulates the sympathetic outflow. Leptin, an adipocyte hormone, alters energy homeostasis and thermogenesis of BAT via several neuronal circuits; however, the cellular effects of leptin on interscapular BAT (iBAT)-related neurons in the hypothalamus remain to be determined. In this study, we used pseudorabies virus (PRV) to identify iBAT-related neurons in the paraventricular nucleus (PVN) of the hypothalamus and test the hypothesis that iBAT-related PVN neurons are modulated by leptin. Inoculation of iBAT with PRV in leptin receptor reporter mice (Lepr:EGFP) demonstrated that a population of iBAT-related PVN neurons expresses Lepr receptors. Our electrophysiological findings revealed that leptin application caused hyperpolarization in some of iBAT-related PVN neurons. Bath application of leptin also modulated excitatory and inhibitory neurotransmission to most of iBAT-related PVN neurons. Using channel rhodopsin assisted circuit mapping we found that GABAergic and glutamatergic Lepr-expressing neurons in the dorsomedial hypothalamus/dorsal hypothalamic area (dDMH/DHA) project to PVN neurons; however, connected iBAT-related PVN neurons receive exclusively inhibitory signals from Lepr-expressing dDMH/DHA neurons.


Asunto(s)
Leptina , Núcleo Hipotalámico Paraventricular , Ratones , Animales , Leptina/metabolismo , Leptina/farmacología , Receptores de Leptina , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/fisiología , Hipotálamo/metabolismo , Neuronas/metabolismo , Termogénesis/fisiología , Sistema Nervioso Simpático/fisiología
4.
J Neurophysiol ; 121(1): 140-151, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30461371

RESUMEN

Transient receptor potential vanilloid type 1 (TRPV1) is a ligand-gated ion channel expressed in the peripheral and central nervous systems. TRPV1-dependent mechanisms take part in a wide range of physiological and pathophysiological pathways including the regulation of homeostatic functions. TRPV1 expression in the hypothalamus has been described as well as evidence that TRPV1-dependent excitatory inputs to hypothalamic preautonomic neurons are diminished in diabetic conditions. Here we aimed to determine the functional expression of TRPV1 in two hypothalamic nuclei known to be involved in the central control of metabolism and to test the hypothesis that TRPV1-expressing neurons receive TRPV1-expressing inputs. A mouse model (TRPV1Cre/tdTom) was generated to identify TRPV1-expressing cells and determine the cellular properties of TRPV1-expressing neurons in adult mice. Our study demonstrated the functional expression of TRPV1 in the dorsomedial hypothalamic nucleus and paraventricular nucleus in adult mice. Our findings revealed that a subset of TRPV1Cre/tdTom neurons receive TRPV1-expressing excitatory inputs, indicating direct interaction between TRPV1-expressing neurons. In addition, astrocytes likely play a role in the modulation of TRPV1-expressing neurons. In summary, this study identified specific hypothalamic regions where TRPV1 is expressed and functional in adult mice and the existence of direct connections between TRPV1Cre/tdTom neurons. NEW & NOTEWORTHY Transient receptor potential vanilloid type 1 (TRPV1) is expressed in the hypothalamus, and TRPV1-dependent regulation of preautonomic neurons is decreased in hyperglycemic conditions. Our study demonstrated functional expression of TRPV1 in two hypothalamic nuclei involved in the control of energy homeostasis. Our results also revealed that a subset of TRPV1-expressing neurons receive TRPV1-expressing excitatory inputs. These findings suggest direct interaction between TRPV1-expressing neurons.


Asunto(s)
Hipotálamo/metabolismo , Neuronas/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Dependovirus , Femenino , Hipotálamo/citología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Ratones Transgénicos , Neuronas/citología , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Canales Catiónicos TRPV/genética , Técnicas de Cultivo de Tejidos , Proteína Fluorescente Roja
5.
Diabetes ; 68(3): 490-501, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30305367

RESUMEN

Estrogens favor glucose homeostasis primarily through the estrogen receptor-α (ERα), but the respective importance of nuclear ERα (NOER) and membrane ERα (MOER) pools to glucose homeostasis are unknown. We studied glucose homeostasis, insulin secretion, and insulin sensitivity in male and female mice expressing either the NOER or the MOER. Male and female MOER mice exhibited fasting and fed hyperglycemia and glucose intolerance. Female MOER mice displayed impaired central insulin signaling associated with hyperinsulinemia and insulin resistance due to unrestrained hepatic gluconeogenesis, without alterations in glucose-stimulated insulin secretion (GSIS). In contrast, male MOER mice did not exhibit detectable insulin resistance, but showed impaired GSIS associated with reduced brain glucose sensing. Female NOER mice exhibited milder hepatic insulin resistance and glucose intolerance. In conclusion, nuclear ERα signaling is predominant in maintaining glucose homeostasis in mice of both sexes. Lack of nuclear ERα alters the central control of insulin sensitivity in females and predominantly impairs the central regulation of insulin secretion in males.


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Glucemia/metabolismo , Femenino , Inmunohistoquímica , Insulina/sangre , Resistencia a la Insulina/fisiología , Secreción de Insulina/fisiología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Mol Metab ; 20: 166-177, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30553770

RESUMEN

OBJECTIVE: Hypothalamic glucose sensing (HGS) initiates insulin secretion (IS) via a vagal control, participating in energy homeostasis. This requires mitochondrial reactive oxygen species (mROS) signaling, dependent on mitochondrial fission, as shown by invalidation of the hypothalamic DRP1 protein. Here, our objectives were to determine whether a model with a HGS defect induced by a short, high fat-high sucrose (HFHS) diet in rats affected the fission machinery and mROS signaling within the mediobasal hypothalamus (MBH). METHODS: Rats fed a HFHS diet for 3 weeks were compared with animals fed a normal chow. Both in vitro (calcium imaging) and in vivo (vagal nerve activity recordings) experiments to measure the electrical activity of isolated MBH gluco-sensitive neurons in response to increased glucose level were performed. In parallel, insulin secretion to a direct glucose stimulus in isolated islets vs. insulin secretion resulting from brain glucose stimulation was evaluated. Intra-carotid glucose load-induced hypothalamic DRP1 translocation to mitochondria and mROS (H2O2) production were assessed in both groups. Finally, compound C was intracerebroventricularly injected to block the proposed AMPK-inhibited DRP1 translocation in the MBH to reverse the phenotype of HFHS fed animals. RESULTS: Rats fed a HFHS diet displayed a decreased HGS-induced IS. Responses of MBH neurons to glucose exhibited an alteration of their electrical activity, whereas glucose-induced insulin secretion in isolated islets was not affected. These MBH defects correlated with a decreased ROS signaling and glucose-induced translocation of the fission protein DRP1, as the vagal activity was altered. AMPK-induced inhibition of DRP1 translocation increased in this model, but its reversal through the injection of the compound C, an AMPK inhibitor, failed to restore HGS-induced IS. CONCLUSIONS: A hypothalamic alteration of DRP1-induced fission and mROS signaling in response to glucose was observed in HGS-induced IS of rats exposed to a 3 week HFHS diet. Early hypothalamic modifications of the neuronal activity could participate in a primary defect of the control of IS and ultimately, the development of diabetes.


Asunto(s)
Glucemia/metabolismo , Dinaminas/metabolismo , Hipotálamo/metabolismo , Mitocondrias/metabolismo , Células Receptoras Sensoriales/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Arterias Carótidas/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Proteínas Quinasas/metabolismo , Transporte de Proteínas , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
7.
Sci Rep ; 6: 34011, 2016 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-27659313

RESUMEN

Type 2 Diabetes (T2D), a major public health issue reaching worldwide epidemic, has been correlated with lower olfactory abilities in humans. As olfaction represents a major component of feeding behavior, its alteration may have drastic consequences on feeding behaviors that may in turn aggravates T2D. In order to decipher the impact of T2D on the olfactory epithelium, we fed mice with a high fructose diet (HFruD) inducing early diabetic state in 4 to 8 weeks. After only 4 weeks of this diet, mice exhibited a dramatic decrease in olfactory behavioral capacities. Consistently, this decline in olfactory behavior was correlated to decreased electrophysiological responses of olfactory neurons recorded as a population and individually. Our results demonstrate that, in rodents, olfaction is modified by HFruD-induced diabetes. Functional, anatomical and behavioral changes occurred in the olfactory system at a very early stage of the disease.

8.
Sci Rep ; 6: 30817, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27465434

RESUMEN

We aimed to determine whether moderate diet restriction could restore cardiac, oxidative and metabolic alterations induced by postnatal overfeeding (PNOF). Litters of C57BL/6 male mice were either maintained at 9 (normal litter, NL), or reduced to 3 (small litter, SL) in order to induce PNOF. At 6 months, half of the NL and SL mice were subjected to 20% calorie-restriction (CR: NLCR, SLCR) for one month, while the other half continued to eat ad libitum (AL: NLAL, SLAL). Six-month old SL mice presented overweight, fat accumulation, hyperleptinemia, glucose intolerance, insulin resistance, increased cardiac ROS production and decreased left ventricular ejection fraction (LVEF). After CR, SL mice body weight was normalized; however, their fat mass and leptinemia were not decreased, glucose metabolism was improved and LVEF was increased. In SL mice, CR increased the cardiac mitochondrial respiratory rate and decreased cardiac ROS production. Hearts from SLCR mice showed better recovery and smaller postischemic infarct size. Intriguingly, no difference was observed between NLAL and NLCR mice for most of the parameters investigated. Short-term moderate CR not only normalized body weight in SL mice but also improved metabolic programming and reversed oxidative and cardiac dysfunction induced by PNOF.


Asunto(s)
Restricción Calórica/métodos , Enfermedades Metabólicas/dietoterapia , Mitocondrias Cardíacas/fisiología , Animales , Animales Recién Nacidos , Composición Corporal , Peso Corporal , Resistencia a la Insulina , Tamaño de la Camada , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
9.
Behav Brain Res ; 268: 40-7, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24675157

RESUMEN

Learned association between odor, taste and further post-ingestive consequence is known as flavor nutrient conditioned preference. Amygdala is supposed to be one of the areas involved in these associations. In the present study, one flavor was associated with a 16% glucose (CS(+)) whereas another flavor was paired with less reinforcing 4% glucose (CS(-)). We showed that CS(+) presentation after conditioning increased Fos expression in the basolateral nucleus of amygdala (BLA). Furthermore, we performed electrophysiological recordings in the BLA in free moving rats. After preference acquisition, rats were exposed to either the CS(+) or the CS(-). The proportion of neurons showing a decreased activity during the CS(-) presentation was significantly higher in conditioned rats compared to controls. Among this neuronal population recorded in conditioned rats, we noticed a significant proportion of neurons that also showed a decreased activity during the CS(+) presentation. Our data indicate an involvement of BLA during retrieval of learned flavors. It also suggests that both flavors might have acquired a biological value through conditioning.


Asunto(s)
Complejo Nuclear Basolateral/fisiología , Condicionamiento Psicológico/fisiología , Preferencias Alimentarias/fisiología , Neuronas/fisiología , Percepción del Gusto/fisiología , Potenciales de Acción , Animales , Ingestión de Alimentos , Electrodos Implantados , Privación de Alimentos , Glucosa/administración & dosificación , Inmunohistoquímica , Masculino , Proteínas Oncogénicas v-fos/metabolismo , Ratas Wistar , Procesamiento de Señales Asistido por Computador
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...