Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 24(8): e57344, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37314252

RESUMEN

The counterregulatory response to hypoglycemia (CRR), which ensures a sufficient glucose supply to the brain, is an essential survival function. It is orchestrated by incompletely characterized glucose-sensing neurons, which trigger a coordinated autonomous and hormonal response that restores normoglycemia. Here, we investigate the role of hypothalamic Tmem117, identified in a genetic screen as a regulator of CRR. We show that Tmem117 is expressed in vasopressin magnocellular neurons of the hypothalamus. Tmem117 inactivation in these neurons increases hypoglycemia-induced vasopressin secretion leading to higher glucagon secretion in male mice, and this effect is estrus cycle phase dependent in female mice. Ex vivo electrophysiological analysis, in situ hybridization, and in vivo calcium imaging reveal that Tmem117 inactivation does not affect the glucose-sensing properties of vasopressin neurons but increases ER stress, ROS production, and intracellular calcium levels accompanied by increased vasopressin production and secretion. Thus, Tmem117 in vasopressin neurons is a physiological regulator of glucagon secretion, which highlights the role of these neurons in the coordinated response to hypoglycemia.


Asunto(s)
Glucagón , Hipoglucemia , Ratones , Masculino , Femenino , Animales , Glucagón/efectos adversos , Calcio , Hipoglucemia/genética , Hipoglucemia/inducido químicamente , Vasopresinas/efectos adversos , Glucosa , Neuronas/fisiología , Glucemia , Insulina
2.
Nat Commun ; 13(1): 5761, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36180454

RESUMEN

The counterregulatory response to hypoglycemia that restores normal blood glucose levels is an essential physiological function. It is initiated, in large part, by incompletely characterized brain hypoglycemia sensing neurons that trigger the secretion of counterregulatory hormones, in particular glucagon, to stimulate hepatic glucose production. In a genetic screen of recombinant inbred BXD mice we previously identified Agpat5 as a candidate regulator of hypoglycemia-induced glucagon secretion. Here, using genetic mouse models, we demonstrate that Agpat5 expressed in agouti-related peptide neurons is required for their activation by hypoglycemia, for hypoglycemia-induced vagal nerve activity, and glucagon secretion. We find that inactivation of Agpat5 leads to increased fatty acid oxidation and ATP production and that suppressing Cpt1a-dependent fatty acid import into mitochondria restores hypoglycemia sensing. Collectively, our data show that AgRP neurons are involved in the control of glucagon secretion and that Agpat5, by partitioning fatty acyl-CoAs away from mitochondrial fatty acid oxidation and ATP generation, ensures that the fall in intracellular ATP, which triggers neuronal firing, faithfully reflects changes in glycemia.


Asunto(s)
Glucagón , Hipoglucemia , Adenosina Trifosfato , Proteína Relacionada con Agouti/genética , Animales , Glucemia , Ácidos Grasos , Glucosa , Insulina , Lípidos/efectos adversos , Ratones , Neuronas
3.
Mol Metab ; 61: 101479, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35339728

RESUMEN

OBJECTIVES: Glucagon secretion to stimulate hepatic glucose production is the first line of defense against hypoglycemia. This response is triggered by so far incompletely characterized central hypoglycemia-sensing mechanisms, which control autonomous nervous activity and hormone secretion. The objective of this study was to identify novel hypothalamic genes controlling insulin-induced glucagon secretion. METHODS: To obtain new information on the mechanisms of hypothalamic hypoglycemia sensing, we combined genetic and transcriptomic analysis of glucagon response to insulin-induced hypoglycemia in a panel of BXD recombinant inbred mice. RESULTS: We identified two QTLs on chromosome 8 and chromosome 15. We further investigated the role of Irak4 and Cpne8, both located in the QTL on chromosome 15, in C57BL/6J and DBA/2J mice, the BXD mouse parental strains. We found that the poor glucagon response of DBA/2J mice was associated with higher hypothalamic expression of Irak4, which encodes a kinase acting downstream of the interleukin-1 receptor (Il-1R), and of Il-ß when compared with C57BL/6J mice. We showed that intracerebroventricular administration of an Il-1R antagonist in DBA/2J mice restored insulin-induced glucagon secretion; this was associated with increased c-fos expression in the arcuate and paraventricular nuclei of the hypothalamus and with higher activation of both branches of the autonomous nervous system. Whole body inactivation of Cpne8, which encodes a Ca++-dependent regulator of membrane trafficking and exocytosis, however, had no impact on insulin-induced glucagon secretion. CONCLUSIONS: Collectively, our data identify Irak4 as a genetically controlled regulator of hypoglycemia-activated hypothalamic neurons and glucagon secretion.


Asunto(s)
Glucagón , Hipoglucemia , Hipotálamo , Quinasas Asociadas a Receptores de Interleucina-1 , Animales , Glucagón/metabolismo , Hipoglucemia/genética , Hipoglucemia/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA
4.
Diabetes ; 70(7): 1443-1457, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33883213

RESUMEN

The counterregulatory response to hypoglycemia is an essential survival function. It is controlled by an integrated network of glucose-responsive neurons, which trigger endogenous glucose production to restore normoglycemia. The complexity of this glucoregulatory network is, however, only partly characterized. In a genetic screen of a panel of recombinant inbred mice we previously identified Fgf15, expressed in neurons of the dorsomedial hypothalamus (DMH), as a negative regulator of glucagon secretion. Here, we report on the generation of Fgf15CretdTomato mice and their use to further characterize these neurons. We show that they were glutamatergic and comprised glucose-inhibited and glucose-excited neurons. When activated by chemogenetics, Fgf15 neurons prevented the increase in vagal nerve firing and the secretion of glucagon normally triggered by insulin-induced hypoglycemia. On the other hand, they increased the activity of the sympathetic nerve in the basal state and prevented its silencing by glucose overload. Higher sympathetic tone increased hepatic Creb1 phosphorylation, Pck1 mRNA expression, and hepatic glucose production leading to glucose intolerance. Thus, Fgf15 neurons of the DMH participate in the counterregulatory response to hypoglycemia by a direct adrenergic stimulation of hepatic glucose production while suppressing vagally induced glucagon secretion. This study provides new insights into the complex neuronal network that prevents the development of hypoglycemia.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Glucagón/metabolismo , Gluconeogénesis/fisiología , Hipotálamo/metabolismo , Hígado/metabolismo , Neuronas/fisiología , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Femenino , Hipoglucemia/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Nervioso Simpático/fisiología
5.
PLoS Biol ; 18(11): e3000680, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33253166

RESUMEN

Proopiomelanocortin (POMC) neurons are major regulators of energy balance and glucose homeostasis. In addition to being regulated by hormones and nutrients, POMC neurons are controlled by glutamatergic input originating from multiple brain regions. However, the factors involved in the formation of glutamatergic inputs and how they contribute to bodily functions remain largely unknown. Here, we show that during the development of glutamatergic inputs, POMC neurons exhibit enriched expression of the Efnb1 (EphrinB1) and Efnb2 (EphrinB2) genes, which are known to control excitatory synapse formation. In vivo loss of Efnb1 in POMC-expressing progenitors decreases the amount of glutamatergic inputs, associated with a reduced number of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) receptor subunits and excitability of these cells. We found that mice lacking Efnb1 in POMC-expressing progenitors display impaired glucose tolerance due to blunted vagus nerve activity and decreased insulin secretion. However, despite reduced excitatory inputs, mice lacking Efnb2 in POMC-expressing progenitors showed no deregulation of insulin secretion and only mild alterations in feeding behavior and gluconeogenesis. Collectively, our data demonstrate the role of ephrins in controlling excitatory input amount into POMC-expressing progenitors and show an isotype-specific role of ephrins on the regulation of glucose homeostasis and feeding.


Asunto(s)
Efrina-B1/metabolismo , Glucosa/metabolismo , Proopiomelanocortina/metabolismo , Animales , Encéfalo/metabolismo , Metabolismo Energético/fisiología , Efrina-B1/fisiología , Efrina-B2/metabolismo , Efrina-B2/fisiología , Fármacos actuantes sobre Aminoácidos Excitadores/metabolismo , Homeostasis/fisiología , Masculino , Ratones , Ratones Noqueados , N-Metilaspartato/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo
6.
Mol Metab ; 35: 100958, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32244185

RESUMEN

OBJECTIVES: In the pathogenesis of type 2 diabetes, development of insulin resistance triggers an increase in pancreatic ß-cell insulin secretion capacity and ß-cell number. Failure of this compensatory mechanism is caused by a dedifferentiation of ß-cells, which leads to insufficient insulin secretion and diabetic hyperglycemia. The ß-cell factors that normally protect against dedifferentiation remain poorly defined. Here, through a systems biology approach, we identify the transcription factor Klf6 as a regulator of ß-cell adaptation to metabolic stress. METHODS: We used a ß-cell specific Klf6 knockout mouse model to investigate whether Klf6 may be a potential regulator of ß-cell adaptation to a metabolic stress. RESULTS: We show that inactivation of Klf6 in ß-cells blunts their proliferation induced by the insulin resistance of pregnancy, high-fat high-sucrose feeding, and insulin receptor antagonism. Transcriptomic analysis showed that Klf6 controls the expression of ß-cell proliferation genes and, in the presence of insulin resistance, it prevents the down-expression of genes controlling mature ß-cell identity and the induction of disallowed genes that impair insulin secretion. Its expression also limits the transdifferentiation of ß-cells into α-cells. CONCLUSION: Our study identifies a new transcription factor that protects ß-cells against dedifferentiation, and which may be targeted to prevent diabetes development.


Asunto(s)
Desdiferenciación Celular/genética , Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina/genética , Células Secretoras de Insulina/metabolismo , Factor 6 Similar a Kruppel/genética , Factor 6 Similar a Kruppel/metabolismo , Animales , Proliferación Celular/genética , Transdiferenciación Celular , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Insulina/metabolismo , Secreción de Insulina/genética , Masculino , Ratones , Ratones Noqueados , Transcriptoma
7.
J Biol Chem ; 292(31): 12860-12873, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28559285

RESUMEN

Virus-related type 2 diabetes is commonly observed in individuals infected with the hepatitis C virus (HCV); however, the underlying molecular mechanisms remain unknown. Our aim was to unravel these mechanisms using FL-N/35 transgenic mice expressing the full HCV ORF. We observed that these mice displayed glucose intolerance and insulin resistance. We also found that Glut-2 membrane expression was reduced in FL-N/35 mice and that hepatocyte glucose uptake was perturbed, partly accounting for the HCV-induced glucose intolerance in these mice. Early steps of the hepatic insulin signaling pathway, from IRS2 to PDK1 phosphorylation, were constitutively impaired in FL-N/35 primary hepatocytes via deregulation of TNFα/SOCS3. Higher hepatic glucose production was observed in the HCV mice, despite higher fasting insulinemia, concomitant with decreased expression of hepatic gluconeogenic genes. Akt kinase activity was higher in HCV mice than in WT mice, but Akt-dependent phosphorylation of the forkhead transcription factor FoxO1 at serine 256, which triggers its nuclear exclusion, was lower in HCV mouse livers. These findings indicate an uncoupling of the canonical Akt/FoxO1 pathway in HCV protein-expressing hepatocytes. Thus, the expression of HCV proteins in the liver is sufficient to induce insulin resistance by impairing insulin signaling and glucose uptake. In conclusion, we observed a complete set of events leading to a prediabetic state in HCV-transgenic mice, providing a valuable mechanistic explanation for HCV-induced diabetes in humans.


Asunto(s)
Hepacivirus/patogenicidad , Hepatitis C/fisiopatología , Hepatocitos/virología , Resistencia a la Insulina , Estado Prediabético/etiología , Absorción Fisiológica , Animales , Línea Celular Tumoral , Células Cultivadas , Regulación de la Expresión Génica , Gluconeogénesis , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatitis C/metabolismo , Hepatitis C/patología , Hepatitis C/virología , Hepatocitos/metabolismo , Hepatocitos/patología , Masculino , Ratones Transgénicos , Músculo Estriado/metabolismo , Músculo Estriado/virología , Sistemas de Lectura Abierta , Fosforilación , Estado Prediabético/virología , Procesamiento Proteico-Postraduccional , ARN/metabolismo , Organismos Libres de Patógenos Específicos , Proteínas Virales/genética , Proteínas Virales/metabolismo
8.
Cell Rep ; 17(7): 1795-1806, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27829151

RESUMEN

The counterregulatory response to hypoglycemia, which restores normal blood glucose levels to ensure sufficient provision of glucose to the brain, is critical for survival. To discover underlying brain regulatory systems, we performed a genetic screen in recombinant inbred mice for quantitative trait loci (QTL) controlling glucagon secretion in response to neuroglucopenia. We identified a QTL on the distal part of chromosome 7 and combined this genetic information with transcriptomic analysis of hypothalami. This revealed Fgf15 as the strongest candidate to control the glucagon response. Fgf15 was expressed by neurons of the dorsomedial hypothalamus and the perifornical area. Intracerebroventricular injection of FGF19, the human ortholog of Fgf15, reduced activation by neuroglucopenia of dorsal vagal complex neurons, of the parasympathetic nerve, and lowered glucagon secretion. In contrast, silencing Fgf15 in the dorsomedial hypothalamus increased neuroglucopenia-induced glucagon secretion. These data identify hypothalamic Fgf15 as a regulator of glucagon secretion.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Pruebas Genéticas , Glucagón/metabolismo , Hipotálamo/metabolismo , Envejecimiento , Animales , Cromosomas de los Mamíferos/metabolismo , Desoxiglucosa/farmacología , Silenciador del Gen/efectos de los fármacos , Genoma , Hipotálamo/efectos de los fármacos , Ratones Endogámicos C57BL , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/metabolismo , Sitios de Carácter Cuantitativo/genética
9.
Sci Rep ; 6: 36937, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27853235

RESUMEN

Nuclear receptor PPARγ has been proven to affect metabolism in multiple tissues, and has received considerable attention for its involvement in colon cancer and inflammatory disease. However, its role in intestinal metabolism has been largely ignored. To investigate this potential aspect of PPARγ function, we submitted intestinal epithelium-specific PPARγ knockout mice (iePPARγKO) to a two-week period of 25% caloric restriction (CR), following which iePPARγKO mice retained more fat than their wild type littermates. In attempting to explain this discrepancy, we analysed the liver, skeletal muscle, intestinal lipid trafficking, and the microbiome, none of which appeared to contribute to the adiposity phenotype. Interestingly, under conditions of CR, iePPARγKO mice failed to activate their sympathetic nervous system (SNS) and increase CR-specific locomotor activity. These KO mice also manifested a defective control of their body temperature, which was overly reduced. Furthermore, the white adipose tissue of iePPARγKO CR mice showed lower levels of both hormone-sensitive lipase, and its phosphorylated form. This would result from impaired SNS signalling and possibly cause reduced lipolysis. We conclude that intestinal epithelium PPARγ plays an essential role in increasing SNS activity under CR conditions, thereby contributing to energy mobilization during metabolically stressful episodes.


Asunto(s)
PPAR gamma/metabolismo , Sistema Nervioso Simpático/metabolismo , Tejido Adiposo Blanco/metabolismo , Adiposidad/fisiología , Animales , Restricción Calórica/métodos , Mucosa Intestinal/metabolismo , Lipólisis/fisiología , Hígado/metabolismo , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo
10.
Diabetes ; 65(10): 2920-31, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27422385

RESUMEN

Glucokinase (Gck) is a critical regulator of glucose-induced insulin secretion by pancreatic ß-cells. It has been suggested to also play an important role in glucose signaling in neurons of the ventromedial hypothalamic nucleus (VMN), a brain nucleus involved in the control of glucose homeostasis and feeding. To test the role of Gck in VMN glucose sensing and physiological regulation, we studied mice with genetic inactivation of the Gck gene in Sf1 neurons of the VMN (Sf1Gck(-/-) mice). Compared with control littermates, Sf1Gck(-/-) mice displayed increased white fat mass and adipocyte size, reduced lean mass, impaired hypoglycemia-induced glucagon secretion, and a lack of parasympathetic and sympathetic nerve activation by neuroglucopenia. However, these phenotypes were observed only in female mice. To determine whether Gck was required for glucose sensing by Sf1 neurons, we performed whole-cell patch clamp analysis of brain slices from control and Sf1Gck(-/-) mice. Absence of Gck expression did not prevent the glucose responsiveness of glucose-excited or glucose-inhibited Sf1 neurons in either sex. Thus Gck in the VMN plays a sex-specific role in the glucose-dependent control of autonomic nervous activity; this is, however, unrelated to the control of the firing activity of classical glucose-responsive neurons.


Asunto(s)
Glucoquinasa/metabolismo , Hipotálamo/enzimología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Femenino , Glucagón/metabolismo , Glucoquinasa/genética , Glucosa/farmacología , Homeostasis/efectos de los fármacos , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Mutantes , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/enzimología , Núcleo Hipotalámico Ventromedial/metabolismo
11.
Med Sci (Paris) ; 31(4): 397-403, 2015 Apr.
Artículo en Francés | MEDLINE | ID: mdl-25958758

RESUMEN

Fatty acid sensitive neurons located in hypothalamus, hippocampus or striatum are able to detect daily variations of plasma fatty acid levels. Thus, these neurons play a role to regulate energy balance by controling food intake, insulin secretion or hepatic glucose production. Molecular mechanisms that mediate fatty acid effects include receptor FAT (fatty acid transporter)/CD36. Deregulation of this brain lipid sensing may be an early event leading to further dysfunction of energy balance leading to obesity and type 2 diabetes.


Asunto(s)
Encéfalo/metabolismo , Metabolismo Energético , Metabolismo de los Lípidos/fisiología , Animales , Ácidos Grasos/metabolismo , Humanos , Hipotálamo/metabolismo , Neuronas/metabolismo , Neurotransmisores/metabolismo , Receptores de Neurotransmisores/metabolismo
12.
Diabetologia ; 58(4): 749-57, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25403481

RESUMEN

AIMS/HYPOTHESIS: Nutrient homeostasis requires integration of signals generated by glucose metabolism and hormones. Expression of the calcium-stimulated adenylyl cyclase ADCY8 is regulated by glucose and the enzyme is capable of integrating signals from multiple pathways. It may thus have an important role in glucose-induced signalling and glucose homeostasis. METHODS: We used pharmacological and genetic approaches in beta cells to determine secretion and calcium metabolism. Furthermore, Adcy8 knockout mice were characterised. RESULTS: In clonal beta cells, inhibitors of adenylyl cyclases or their downstream targets reduced the glucose-induced increase in cytosolic calcium and insulin secretion. This was reproduced by knock-down of ADCY8, but not of ADCY1. These agents also inhibited glucose-induced increase in cytosolic calcium and electrical activity in primary beta cells and similar effects were observed after ADCY8 knock-down. Moreover, insulin secretion was diminished in islets from Adcy8 knockout mice. These mice were glucose intolerant after oral or intraperitoneal administration of glucose whereas their levels of glucagon-like peptide-1 remained unaltered. Finally, we knocked down ADCY8 in the ventromedial hypothalamus to evaluate the need for ADCY8 in the central regulation of glucose homeostasis. Whereas mice fed a standard diet had normal glucose levels, high-fat diet exacerbated glucose intolerance and knock-down mice were incapable of raising their plasma insulin levels. Finally we confirmed that ADCY8 is expressed in human islets. CONCLUSIONS/INTERPRETATIONS: Collectively, our findings demonstrate that ADCY8 is required for the physiological activation of glucose-induced signalling pathways in beta cells, for glucose tolerance and for hypothalamic adaptation to a high-fat diet via regulation of islet insulin secretion.


Asunto(s)
Adenilil Ciclasas/metabolismo , Glucemia/metabolismo , Células Secretoras de Insulina/enzimología , Adenilil Ciclasas/deficiencia , Adenilil Ciclasas/genética , Animales , Calcio/metabolismo , Línea Celular , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Genotipo , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/enzimología , Homeostasis , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Potenciales de la Membrana , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección , Núcleo Hipotalámico Ventromedial/enzimología
13.
Cell Metab ; 19(3): 527-38, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24606905

RESUMEN

Glucose-sensing neurons in the brainstem participate in the regulation of energy homeostasis but have been poorly characterized because of the lack of specific markers to identify them. Here we show that GLUT2-expressing neurons of the nucleus of the tractus solitarius form a distinct population of hypoglycemia-activated neurons. Their response to low glucose is mediated by reduced intracellular glucose metabolism, increased AMP-activated protein kinase activity, and closure of leak K(+) channels. These are GABAergic neurons that send projections to the vagal motor nucleus. Light-induced stimulation of channelrhodospin-expressing GLUT2 neurons in vivo led to increased parasympathetic nerve firing and glucagon secretion. Thus GLUT2 neurons of the nucleus tractus solitarius link hypoglycemia detection to counterregulatory response. These results may help identify the cause of hypoglycemia-associated autonomic failure, a major threat in the insulin treatment of diabetes.


Asunto(s)
Neuronas GABAérgicas/fisiología , Glucagón/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Núcleo Solitario/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Channelrhodopsins , Desoxiglucosa/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Glucosamina/farmacología , Glucosa/farmacología , Hipoglucemia/metabolismo , Hipoglucemia/patología , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo
14.
Mol Metab ; 3(2): 167-76, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24634821

RESUMEN

Brain lipid sensing is necessary to regulate energy balance. Lipoprotein lipase (LPL) may play a role in this process. We tested if hippocampal LPL regulated energy homeostasis in rodents by specifically attenuating LPL activity in the hippocampus of rats and mice, either by infusing a pharmacological inhibitor (tyloxapol), or using a genetic approach (adeno-associated virus expressing Cre-GFP injected into Lpl (lox/lox) mice). Decreased LPL activity by either method led to increased body weight gain due to decreased locomotor activity and energy expenditure, concomitant with increased parasympathetic tone (unchanged food intake). Decreased LPL activity in both models was associated with increased de novo ceramide synthesis and neurogenesis in the hippocampus, while intrahippocampal infusion of de novo ceramide synthesis inhibitor myriocin completely prevented body weight gain. We conclude that hippocampal lipid sensing might represent a core mechanism for energy homeostasis regulation through de novo ceramide synthesis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...