Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Metab ; 5(11): 1969-1985, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37884694

RESUMEN

T cell activation is associated with a profound and rapid metabolic response to meet increased energy demands for cell division, differentiation and development of effector function. Glucose uptake and engagement of the glycolytic pathway are major checkpoints for this event. Here we show that the low-affinity, concentration-dependent glucose transporter 2 (Glut2) regulates the development of CD8+ T cell effector responses in mice by promoting glucose uptake, glycolysis and glucose storage. Expression of Glut2 is modulated by environmental factors including glucose and oxygen availability and extracellular acidification. Glut2 is highly expressed by circulating, recently primed T cells, allowing efficient glucose uptake and storage. In glucose-deprived inflammatory environments, Glut2 becomes downregulated, thus preventing passive loss of intracellular glucose. Mechanistically, Glut2 expression is regulated by a combination of molecular interactions involving hypoxia-inducible factor-1 alpha, galectin-9 and stomatin. Finally, we show that human T cells also rely on this glucose transporter, thus providing a potential target for therapeutic immunomodulation.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa , Glucosa , Ratones , Humanos , Animales , Glucosa/metabolismo , Transporte Biológico/fisiología , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Diferenciación Celular , Linfocitos T CD8-positivos/metabolismo
2.
Science ; 381(6657): 515-524, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37535729

RESUMEN

Tumor microenvironments (TMEs) influence cancer progression but are complex and often differ between patients. Considering that microenvironment variations may reveal rules governing intratumoral cellular programs and disease outcome, we focused on tumor-to-tumor variation to examine 52 head and neck squamous cell carcinomas. We found that macrophage polarity-defined by CXCL9 and SPP1 (CS) expression but not by conventional M1 and M2 markers-had a noticeably strong prognostic association. CS macrophage polarity also identified a highly coordinated network of either pro- or antitumor variables, which involved each tumor-associated cell type and was spatially organized. We extended these findings to other cancer indications. Overall, these results suggest that, despite their complexity, TMEs coordinate coherent responses that control human cancers and for which CS macrophage polarity is a relevant yet simple variable.


Asunto(s)
Polaridad Celular , Quimiocina CXCL9 , Neoplasias de Cabeza y Cuello , Macrófagos , Osteopontina , Carcinoma de Células Escamosas de Cabeza y Cuello , Microambiente Tumoral , Humanos , Quimiocina CXCL9/análisis , Quimiocina CXCL9/metabolismo , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Macrófagos/inmunología , Osteopontina/análisis , Osteopontina/metabolismo , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Polaridad Celular/inmunología
3.
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
4.
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
5.
iScience ; 24(10): 103122, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34622169

RESUMEN

The paraventricular nucleus of the thalamus (PVT) controls goal-oriented behavior through its connections to the nucleus accumbens (NAc). We previously characterized Glut2aPVT neurons that are activated by hypoglycemia, and which increase sucrose seeking behavior through their glutamatergic projections to the NAc. Here, we identified glucokinase (Gck)-expressing neurons of the PVT (GckaPVT) and generated a mouse line expressing the Cre recombinase from the glucokinase locus (Gck Cre/+ mice). Ex vivo calcium imaging and whole-cell patch clamp recordings revealed that GckaPVT neurons that project to the NAc were mostly activated by hyperglycemia. Their chemogenetic inhibition or optogenetic stimulation, respectively, enhanced food intake or decreased sucrose-seeking behavior. Collectively, our results describe a neuronal population of Gck-expressing neurons in the PVT, which has opposite glucose sensing properties and control over feeding behavior than the previously characterized Glut2aPVT neurons. This study allows a better understanding of the complex regulation of feeding behavior by the PVT.

7.
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
8.
NPJ Vaccines ; 6(1): 36, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33723260

RESUMEN

T cells are important for controlling ovarian cancer (OC). We previously demonstrated that combinatorial use of a personalized whole-tumor lysate-pulsed dendritic cell vaccine (OCDC), bevacizumab (Bev), and cyclophosphamide (Cy) elicited neoantigen-specific T cells and prolonged OC survival. Here, we hypothesize that adding acetylsalicylic acid (ASA) and low-dose interleukin (IL)-2 would increase the vaccine efficacy in a recurrent advanced OC phase I trial (NCT01132014). By adding ASA and low-dose IL-2 to the OCDC-Bev-Cy combinatorial regimen, we elicited vaccine-specific T-cell responses that positively correlated with patients' prolonged time-to-progression and overall survival. In the ID8 ovarian model, animals receiving the same regimen showed prolonged survival, increased tumor-infiltrating perforin-producing T cells, increased neoantigen-specific CD8+ T cells, and reduced endothelial Fas ligand expression and tumor-infiltrating T-regulatory cells. This combinatorial strategy was efficacious and also highlighted the predictive value of the ID8 model for future ovarian trial development.

9.
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
10.
Nat Commun ; 9(1): 546, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416045

RESUMEN

Glucagon secretion by pancreatic α-cells is triggered by hypoglycemia and suppressed by high glucose levels; impaired suppression of glucagon secretion is a hallmark of both type 1 and type 2 diabetes. Here, we show that α-cell glucokinase (Gck) plays a role in the control of glucagon secretion. Using mice with α-cell-specific inactivation of Gck (αGckKO mice), we find that glucokinase is required for the glucose-dependent increase in intracellular ATP/ADP ratio and the closure of KATP channels in α-cells and the suppression of glucagon secretion at euglycemic and hyperglycemic levels. αGckKO mice display hyperglucagonemia in the fed state, which is associated with increased hepatic gluconeogenic gene expression and hepatic glucose output capacity. In adult mice, fed hyperglucagonemia is further increased and glucose intolerance develops. Thus, glucokinase governs an α-cell metabolic pathway that suppresses secretion at or above normoglycemic levels; abnormal suppression of glucagon secretion deregulates hepatic glucose metabolism and, over time, induces a pre-diabetic phenotype.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Glucoquinasa/genética , Intolerancia a la Glucosa/metabolismo , Glucosa/metabolismo , Hipoglucemia/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico , Femenino , Expresión Génica , Células Secretoras de Glucagón/patología , Glucoquinasa/deficiencia , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/patología , Hipoglucemia/genética , Hipoglucemia/patología , Insulina/metabolismo , Canales KATP/genética , Canales KATP/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados
11.
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
12.
Nat Neurosci ; 19(8): 999-1002, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27322418

RESUMEN

Feeding behavior is governed by homeostatic needs and motivational drive to obtain palatable foods. Here, we identify a population of glutamatergic neurons in the paraventricular thalamus of mice that express the glucose transporter Glut2 (encoded by Slc2a2) and project to the nucleus accumbens. These neurons are activated by hypoglycemia and, in freely moving mice, their activation by optogenetics or Slc2a2 inactivation increases motivated sucrose-seeking but not saccharin-seeking behavior. These neurons may control sugar overconsumption in obesity and diabetes.


Asunto(s)
Conducta Animal/fisiología , Glucosa/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Tálamo/metabolismo , Animales , Conducta Alimentaria , Hipoglucemia/metabolismo , Ratones Transgénicos , Motivación/fisiología , Núcleo Accumbens/metabolismo , Obesidad/metabolismo , Autoadministración/métodos , Sacarosa/metabolismo
13.
Diabetes ; 64(12): 4148-57, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26384384

RESUMEN

Insulin-like growth factor 2 (IGF2), produced and secreted by adult ß-cells, functions as an autocrine activator of the ß-cell insulin-like growth factor 1 receptor signaling pathway. Whether this autocrine activity of IGF2 plays a physiological role in ß-cell and whole-body physiology is not known. Here, we studied mice with ß-cell-specific inactivation of Igf2 (ßIGF2KO mice) and assessed ß-cell mass and function in aging, pregnancy, and acute induction of insulin resistance. We showed that glucose-stimulated insulin secretion (GSIS) was markedly reduced in old female ßIGF2KO mice; glucose tolerance was, however, normal because of increased insulin sensitivity. While on a high-fat diet, both male and female ßIGF2KO mice displayed lower GSIS compared with control mice, but reduced ß-cell mass was observed only in female ßIGF2KO mice. During pregnancy, there was no increase in ß-cell proliferation and mass in ßIGF2KO mice. Finally, ß-cell mass expansion in response to acute induction of insulin resistance was lower in ßIGF2KO mice than in control mice. Thus, the autocrine action of IGF2 regulates adult ß-cell mass and function to preserve in vivo GSIS in aging and to adapt ß-cell mass in response to metabolic stress, pregnancy hormones, and acute induction of insulin resistance.


Asunto(s)
Envejecimiento , Resistencia a la Insulina , Factor II del Crecimiento Similar a la Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptor IGF Tipo 1/agonistas , Transducción de Señal , Alostasis , Animales , Apoptosis , Proliferación Celular , Cruzamientos Genéticos , Dieta Alta en Grasa/efectos adversos , Femenino , Regulación del Desarrollo de la Expresión Génica , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Secreción de Insulina , Factor II del Crecimiento Similar a la Insulina/genética , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/patología , Masculino , Ratones Noqueados , Ratones Transgénicos , Embarazo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Caracteres Sexuales , Técnicas de Cultivo de Tejidos
14.
Diabetologia ; 58(3): 558-65, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25500700

RESUMEN

AIMS/HYPOTHESIS: Pancreatic beta cells play a central role in the control of glucose homeostasis by secreting insulin to stimulate glucose uptake by peripheral tissues. Understanding the molecular mechanisms that control beta cell function and plasticity has critical implications for the pathophysiology and therapy of major forms of diabetes. Selective gene inactivation in pancreatic beta cells, using the Cre-lox system, is a powerful approach to assess the role of particular genes in beta cells and their impact on whole body glucose homeostasis. Several Cre recombinase (Cre) deleter mice have been established to allow inactivation of genes in beta cells, but many show non-specific recombination in other cell types, often in the brain. METHODS: We describe the generation of Ins1(Cre) and Ins1(CreERT2) mice in which the Cre or Cre-oestrogen receptor fusion protein (CreERT2) recombinases have been introduced at the initiation codon of the Ins1 gene. RESULTS: We show that Ins1(Cre) mice induce efficient and selective recombination of floxed genes in beta cells from the time of birth, with no recombination in the central nervous system. These mice have normal body weight and glucose homeostasis. Furthermore, we show that tamoxifen treatment of adult Ins1(CreERT2) mice crossed with Rosa26-tdTomato mice induces efficient recombination in beta cells. CONCLUSIONS/INTERPRETATION: These two strains of deleter mice are useful new resources to investigate the molecular physiology of pancreatic beta cells.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Femenino , Prueba de Tolerancia a la Glucosa , Insulina/genética , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Mutantes
15.
J Clin Invest ; 124(1): 413-24, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24334455

RESUMEN

How glucose sensing by the nervous system impacts the regulation of ß cell mass and function during postnatal development and throughout adulthood is incompletely understood. Here, we studied mice with inactivation of glucose transporter 2 (Glut2) in the nervous system (NG2KO mice). These mice displayed normal energy homeostasis but developed late-onset glucose intolerance due to reduced insulin secretion, which was precipitated by high-fat diet feeding. The ß cell mass of adult NG2KO mice was reduced compared with that of WT mice due to lower ß cell proliferation rates in NG2KO mice during the early postnatal period. The difference in proliferation between NG2KO and control islets was abolished by ganglionic blockade or by weaning the mice on a carbohydrate-free diet. In adult NG2KO mice, first-phase insulin secretion was lost, and these glucose-intolerant mice developed impaired glucagon secretion when fed a high-fat diet. Electrophysiological recordings showed reduced parasympathetic nerve activity in the basal state and no stimulation by glucose. Furthermore, sympathetic activity was also insensitive to glucose. Collectively, our data show that GLUT2-dependent control of parasympathetic activity defines a nervous system/endocrine pancreas axis that is critical for ß cell mass establishment in the postnatal period and for long-term maintenance of ß cell function.


Asunto(s)
Proliferación Celular , Transportador de Glucosa de Tipo 2/genética , Glucosa/metabolismo , Homeostasis , Células Secretoras de Insulina/metabolismo , Potenciales de Acción , Animales , Fibras Autónomas Preganglionares/fisiología , Metabolismo Energético , Femenino , Ganglios Parasimpáticos/metabolismo , Ganglios Parasimpáticos/fisiopatología , Intolerancia a la Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/deficiencia , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/inervación , Páncreas/patología
16.
J Clin Invest ; 123(4): 1662-76, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23549084

RESUMEN

Liver glucose metabolism plays a central role in glucose homeostasis and may also regulate feeding and energy expenditure. Here we assessed the impact of glucose transporter 2 (Glut2) gene inactivation in adult mouse liver (LG2KO mice). Loss of Glut2 suppressed hepatic glucose uptake but not glucose output. In the fasted state, expression of carbohydrate-responsive element-binding protein (ChREBP) and its glycolytic and lipogenic target genes was abnormally elevated. Feeding, energy expenditure, and insulin sensitivity were identical in LG2KO and control mice. Glucose tolerance was initially normal after Glut2 inactivation, but LG2KO mice exhibited progressive impairment of glucose-stimulated insulin secretion even though ß cell mass and insulin content remained normal. Liver transcript profiling revealed a coordinated downregulation of cholesterol biosynthesis genes in LG2KO mice that was associated with reduced hepatic cholesterol in fasted mice and reduced bile acids (BAs) in feces, with a similar trend in plasma. We showed that chronic BAs or farnesoid X receptor (FXR) agonist treatment of primary islets increases glucose-stimulated insulin secretion, an effect not seen in islets from Fxr(-/-) mice. Collectively, our data show that glucose sensing by the liver controls ß cell glucose competence and suggest BAs as a potential mechanistic link.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Hígado/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Ácidos y Sales Biliares/metabolismo , Glucemia , Células Cultivadas , Colesterol/sangre , Colesterol/metabolismo , Regulación hacia Abajo , Metabolismo Energético , Heces/química , Fluorodesoxiglucosa F18/metabolismo , Técnicas de Inactivación de Genes , Glucosa/fisiología , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/genética , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Homeostasis , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Metabolismo de los Lípidos , Hígado/diagnóstico por imagen , Hígado/fisiopatología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Cintigrafía , Radiofármacos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
17.
FASEB J ; 24(6): 1747-58, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20097878

RESUMEN

The physiological contribution of glucose in thermoregulation is not completely established nor whether this control may involve a regulation of the melanocortin pathway. Here, we assessed thermoregulation and leptin sensitivity of hypothalamic arcuate neurons in mice with inactivation of glucose transporter type 2 (Glut2)-dependent glucose sensing. Mice with inactivation of Glut2-dependent glucose sensors are cold intolerant and show increased susceptibility to food deprivation-induced torpor and abnormal hypothermic response to intracerebroventricular administration of 2-deoxy-d-glucose compared to control mice. This is associated with a defect in regulated expression of brown adipose tissue uncoupling protein I and iodothyronine deiodinase II and with a decreased leptin sensitivity of neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons, as observed during the unfed-to-refed transition or following i.p. leptin injection. Sites of central Glut-2 expression were identified by a genetic tagging approach and revealed that glucose-sensitive neurons were present in the lateral hypothalamus, the dorsal vagal complex, and the basal medulla but not in the arcuate nucleus. NPY and POMC neurons were, however, connected to nerve terminals from Glut2-expressing neurons. Thus, our data suggest that glucose controls thermoregulation and the leptin sensitivity of NPY and POMC neurons through activation of Glut2-dependent glucose-sensing neurons located outside of the arcuate nucleus.


Asunto(s)
Regulación de la Temperatura Corporal , Transportador de Glucosa de Tipo 2/fisiología , Glucosa/metabolismo , Leptina/farmacología , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Western Blotting , Femenino , Glucosa/análisis , Humanos , Técnicas para Inmunoenzimas , Integrasas , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Neuropéptido Y/genética , Proopiomelanocortina/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Desacopladora 1 , Yodotironina Deyodinasa Tipo II
18.
PLoS One ; 3(10): e3385, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19030233

RESUMEN

BACKGROUND: The visceral (VAT) and subcutaneous (SCAT) adipose tissues play different roles in physiology and obesity. The molecular mechanisms underlying their expansion in obesity and following body weight reduction are poorly defined. METHODOLOGY: C57Bl/6 mice fed a high fat diet (HFD) for 6 months developed low, medium, or high body weight as compared to normal chow fed mice. Mice from each groups were then treated with the cannabinoid receptor 1 antagonist rimonabant or vehicle for 24 days to normalize their body weight. Transcriptomic data for visceral and subcutaneous adipose tissues from each group of mice were obtained and analyzed to identify: i) genes regulated by HFD irrespective of body weight, ii) genes whose expression correlated with body weight, iii) the biological processes activated in each tissue using gene set enrichment analysis (GSEA), iv) the transcriptional programs affected by rimonabant. PRINCIPAL FINDINGS: In VAT, "metabolic" genes encoding enzymes for lipid and steroid biosynthesis and glucose catabolism were down-regulated irrespective of body weight whereas "structure" genes controlling cell architecture and tissue remodeling had expression levels correlated with body weight. In SCAT, the identified "metabolic" and "structure" genes were mostly different from those identified in VAT and were regulated irrespective of body weight. GSEA indicated active adipogenesis in both tissues but a more prominent involvement of tissue stroma in VAT than in SCAT. Rimonabant treatment normalized most gene expression but further reduced oxidative phosphorylation gene expression in SCAT but not in VAT. CONCLUSION: VAT and SCAT show strikingly different gene expression programs in response to high fat diet and rimonabant treatment. Our results may lead to identification of therapeutic targets acting on specific fat depots to control obesity.


Asunto(s)
Grasa Abdominal/metabolismo , Matriz Extracelular/metabolismo , Obesidad/genética , Obesidad/metabolismo , Grasa Subcutánea/metabolismo , Transcripción Genética , Adipocitos/efectos de los fármacos , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Antagonistas de Receptores de Cannabinoides , Grasas de la Dieta/administración & dosificación , Regulación de la Expresión Génica , Insulina/sangre , Leptina/sangre , Lipoproteínas VLDL/sangre , Ratones , Ratones Endogámicos C57BL , Piperidinas/farmacología , Pirazoles/farmacología , Rimonabant
19.
Diabetes ; 55(4): 988-95, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16567520

RESUMEN

A role for glucose in the control of feeding has been proposed, but its precise physiological importance is unknown. Here, we evaluated feeding behavior in glut2-null mice, which express a transgenic glucose transporter in their beta-cells to rescue insulin secretion (ripglut1;glut2-/- mice). We showed that in the absence of GLUT2, daily food intake was increased and feeding initiation and termination following a fasting period were abnormal. This was accompanied by suppressed regulation of hypothalamic orexigenic and anorexigenic neuropeptides expression during the fast-to-refed transition. In these conditions, however, there was normal regulation of the circulating levels of insulin, leptin, or glucose but a loss of regulation of plasma ghrelin concentrations. To evaluate whether the abnormal feeding behavior was due to suppressed glucose sensing, we evaluated feeding in response to intraperitoneal or intracerebroventricular glucose or 2-deoxy-D-glucose injections. We showed that in GLUT2-null mice, feeding was no longer inhibited by glucose or activated by 2-deoxy-D-glucose injections and the regulation of hypothalamic neuropeptide expression by intracerebroventricular glucose administration was lost. Together, these data demonstrate that absence of GLUT2 suppressed the function of central glucose sensors, which control feeding probably by regulating the hypothalamic melanocortin pathway. Furthermore, inactivation of these glucose sensors causes overeating.


Asunto(s)
Conducta Alimentaria/fisiología , Transportador de Glucosa de Tipo 2/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Ghrelina , Transportador de Glucosa de Tipo 2/deficiencia , Transportador de Glucosa de Tipo 2/genética , Hipotálamo/fisiología , Insulina/sangre , Leptina/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/genética , Hormonas Peptídicas/sangre , Proopiomelanocortina/genética , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , alfa-MSH/fisiología
20.
J Clin Invest ; 115(12): 3545-53, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16322792

RESUMEN

Ripglut1;glut2-/- mice have no endogenous glucose transporter type 2 (glut2) gene expression but rescue glucose-regulated insulin secretion. Control of glucagon plasma levels is, however, abnormal, with fed hyperglucagonemia and insensitivity to physiological hypo- or hyperglycemia, indicating that GLUT2-dependent sensors control glucagon secretion. Here, we evaluated whether these sensors were located centrally and whether GLUT2 was expressed in glial cells or in neurons. We showed that ripglut1;glut2-/- mice failed to increase plasma glucagon levels following glucoprivation induced either by i.p. or intracerebroventricular 2-deoxy-D-glucose injections. This was accompanied by failure of 2-deoxy-D-glucose injections to activate c-Fos-like immunoreactivity in the nucleus of the tractus solitarius and the dorsal motor nucleus of the vagus. When glut2 was expressed by transgenesis in glial cells but not in neurons of ripglut1;glut2-/- mice, stimulated glucagon secretion was restored as was c-Fos-like immunoreactive labeling in the brainstem. When ripglut1;glut2-/- mice were backcrossed into the C57BL/6 genetic background, fed plasma glucagon levels were also elevated due to abnormal autonomic input to the alpha cells; glucagon secretion was, however, stimulated by hypoglycemic stimuli to levels similar to those in control mice. These studies identify the existence of central glucose sensors requiring glut2 expression in glial cells and therefore functional coupling between glial cells and neurons. These sensors may be activated at different glycemic levels depending on the genetic background.


Asunto(s)
Astrocitos/metabolismo , Regulación de la Expresión Génica , Glucagón/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/fisiología , Glucosa/metabolismo , Animales , Glucemia/metabolismo , Northern Blotting , Southern Blotting , Western Blotting , ADN Complementario/metabolismo , Desoxiglucosa/química , Glucagón/sangre , Glucagón/química , Células Secretoras de Glucagón/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Hipoglucemia/patología , Inmunohistoquímica , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Modelos Genéticos , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factores de Tiempo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...