Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Biol Chem ; 291(25): 13063-75, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27137930

RESUMEN

Efficient energy storage in adipose tissues requires optimal function of the insulin-producing ß-cell, whereas its dysfunction promotes diabetes. The associated paradox related to ß-cell efficiency is that excessive accumulation of fat in adipose tissue predisposes for type 2 diabetes. Insulin exocytosis is regulated by intracellular metabolic signal transduction, with glutamate dehydrogenase playing a key role in the amplification of the secretory response. Here, we used mice with ß-cell-selective glutamate dehydrogenase deletion (ßGlud1(-/-)), lacking an amplifying pathway of insulin secretion. As opposed to control mice, ßGlud1(-/-) animals fed a high calorie diet maintained glucose tolerance and did not develop diet-induced obesity. Islets of ßGlud1(-/-) mice did not increase their secretory response upon high calorie feeding, as did islets of control mice. Inhibited adipose tissue expansion observed in knock-out mice correlated with lower expression of genes responsible for adipogenesis. Rather than being efficiently stored, lipids were consumed at a higher rate in ßGlud1(-/-) mice compared with controls, in particular during food intake periods. These results show that reduced ß-cell function prior to high calorie feeding prevented diet-induced obesity.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Células Secretoras de Insulina/fisiología , Obesidad/etiología , Transducción de Señal , Animales , Metabolismo Basal , Células Cultivadas , Intolerancia a la Glucosa , Glutamato Deshidrogenasa/genética , Metabolismo de los Lípidos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/patología
2.
J Clin Med ; 13(7)2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38610833

RESUMEN

Background: The current study aims to assess the psychological conditions of Italian adolescents with obesity seeking an in-hospital multidisciplinary body weight reduction program, by exploring their psychological adjustment, emotional states, and co-occurring eating disorder symptoms. Methods: The study involved ninety-two consecutive Italian adolescents with obesity (31 males, 61 females), with a mean age ± SD: 16.4 ± 1.1 years and body mass index (BMI): 38.3 ± 6.04 kg/m2). The Strengths and Difficulties Questionnaire (SDQ), Beck Depression Inventory (BDI), State-Trait Anxiety Inventory (STAI), and Eating Attitude Test-26 (EAT-26) were used for the evaluations. Differences between genders, degrees of obesity (Group 1 = BMI SDS 2-2.99 and Group 2: BMI SDS > 3), and those with or without eating disorder symptoms (Group 1: EAT-26 ≤ 20 and Group 2: EAT-26 > 20) were explored. Results: The results showed that females reported higher scores on the Emotional Symptoms, Prosocial Behaviors, Total Difficulties, and Total Impact subscales of the SDQ, the BDI, both subscales of the STAI, and the Bulimia subscales of the EAT-26 than males, independently from the degrees of obesity. Participants with eating disorder symptoms (Group 2: EAT-26 > 20) showed higher scores on the Emotional Symptoms and Total Difficulties subscales of the SDQ, the BDI, and both subscales of the STAI than those of Group 1 (EAT-26 ≤ 20). Conclusions: The study explores the psychological conditions of adolescents with obesity. The results can inform appropriate treatment approaches for the management of obesity in developmental age groups, which not only take into account the medical and physical aspects of obesity, but also the behavioral, emotional, and social difficulties expressed by adolescents, in addition to specific eating disorder symptoms.

3.
Ther Clin Risk Manag ; 19: 645-656, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37560130

RESUMEN

Purpose: Hepatitis C virus (HCV) spreads from contact with blood of an infected person. HCV infections are common among people who use drugs (PWUDs), when sharing needles, syringes, or other equipment for injected drugs. The advent of pangenotypic direct-antiviral agents (DAA) in 2017 transformed the treatment landscape for HCV, but PWUDs remain a complex and hard-to-treat population with high risk of HCV reinfection. The aim of this real-world analysis was to characterize the demographic and clinical features of PWUDs in Italy, also focusing on comorbidity profile, treatment with DAAs, resource consumptions for the National Health System (NHS). Patients and Methods: During 01/2011-06/2020, administrative databases of Italian healthcare entities, covering 3,900,000 individuals, were browsed to identify PWUDs with or without HCV infection. Among HCV+ patients, a further stratification was made into treated and untreated with DAAs. The date of PWUD or HCV first diagnosis or DAA first prescription was considered as index-date. Patients were then followed-up for one year. Alcohol-dependency was also investigated. Results: Total 3690 PWUDs were included, of whom 1141 (30.9%) PWUD-HCV+ and 2549 (69.1%) PWUD-HCV-. HCV-positive were significantly older (43.6 vs 38.5 years, p < 0.001), had a worse comorbidity profile (Charlson-index: 0.8 vs 0.4, p < 0.001), and high rates of psychiatric, respiratory, dermatological, musculoskeletal diseases and genitourinary (sexually transmitted) infections. Moreover, they received more drug prescriptions (other than DAAs, like anti-acids, antiepileptics, psycholeptics) and had undergone more frequent hospitalization, predominantly for hepatobiliary, respiratory system and mental disorders. DDA-untreated had significantly higher Charlson-index than DAA-treated (0.9 vs 0.6, p = 0.003). Alcoholism was found in 436 (11.8%) cases. Conclusion: This Italian real-world analysis suggests that PWUDs with HCV infection, especially those untreated with DAAs, show an elevated drug consumption due to their complex clinical profile. These findings could help to ameliorate the healthcare interventions on PWUDs with HCV infection.

4.
Ther Clin Risk Manag ; 19: 57-65, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36699017

RESUMEN

Purpose: The study aims at investigating the impact of polymedication and aging in the prevalence of multiple drug-drug interactions (DDIs) on HCV patients treated with sofosbuvir/velpatasvir (SOF/VEL) or glecaprevir/pibrentasvir (GLE/PIB). Patients and Methods: This is a retrospective analysis based on administrative data covering around 6.9 million individuals. Patients treated with SOF/VEL or GLE/PIB over November 2017-March 2020 were included. Index date corresponded to SOF/VEL or GLE/PIB first prescription during such period; patients were followed up for treatment duration. Analyses were then focused on patients with ≥2 comedications at risk of multiple DDIs. The severity and the effect of multiple DDI were identified using the Liverpool University tool. Results: A total of 2057 patients with SOF/VEL and 2128 with GLE/PIB were selected. Mean age of SOF/VEL patients was 58.5 years, higher than GLE/PIB ones (52.5 years) (p < 0.001), and patients >50 years were more present in SOF/VEL vs GLE/PIB cohorts: 72% vs 58%, (p < 0.001). Most prescribed co-medications were cardiovascular, alimentary and nervous system drugs. Proportion of patients with ≥2 comedications was higher in SOF/VEL compared to GLE/PIB cohort (56.5% vs 32.3%, p < 0.001). Those at high-risk of multiple DDIs accounted for 11.6% (N = 135) of SOF/VEL and 19.6% (N = 135) of GLE/PIB (p < 0.001) patients with ≥2 comedications. Among them, the potential effect of DDI was a decrease of DAA serum levels (11% of SOF/VEL and GLE/PIB patients) and an increased concentration of comedication serum levels (14% of SOF/VEL and 42% of GLE/PIB patients). Conclusion: This real-world analysis provided a thorough characterization on the burden of polymedication regimens in HCV patients treated with SOF/VEL or GLE/PIB that expose such patients to an increased risk of DDIs. In our sample population, SOF/VEL regimen was more frequently detected on elderly patients and on those with ≥2 comedications at risk of multi-DDI, ie, among patients characterized by higher rates of comorbidities and polypharmacy.

5.
J Biol Chem ; 286(15): 13214-25, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282101

RESUMEN

Chronic exposure to elevated levels of glucose and fatty acids leads to dysfunction of pancreatic ß-cells by mechanisms that are only partly understood. The transcription factor peroxisome proliferator-activated receptor α (PPARα) is an important regulator of genes involved in fatty acid metabolism and has been shown to protect against lipid-induced ß-cell dysfunction. We and others have previously shown that expression of the PPARα gene in ß-cells is rapidly repressed by glucose. Here we show that the PPARα gene is transcribed from five alternative transcription start sites, resulting in three alternative first exons that are spliced to exon 2. Expression of all PPARα transcripts is repressed by glucose both in insulinoma cells and in isolated pancreatic islets. The observation that the dynamics of glucose repression of PPARα transcription are very similar to those of glucose activation of target genes by the carbohydrate response element-binding protein (ChREBP) prompted us to investigate the potential role of ChREBP in the regulation of PPARα expression. We show that a constitutively active ChREBP lacking the N-terminal domain efficiently represses PPARα expression in insulinoma cells and in rodent and human islets. In addition, we demonstrate that siRNA-mediated knockdown of ChREBP abrogates glucose repression of PPARα expression as well as induction of well established ChREBP target genes in insulinoma cells. In conclusion, this work shows that ChREBP is a critical and direct mediator of glucose repression of PPARα gene expression in pancreatic ß-cells, suggesting that ChREBP may be important for glucose suppression of the fatty acid oxidation capacity of ß-cells.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Nucleares/metabolismo , PPAR alfa/biosíntesis , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Línea Celular Tumoral , Exones/fisiología , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Glucosa/farmacología , Células HEK293 , Humanos , Células Secretoras de Insulina/citología , Ratones , Proteínas Nucleares/genética , Oxidación-Reducción , PPAR alfa/genética , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Proteínas Represoras/genética , Edulcorantes/metabolismo , Edulcorantes/farmacología , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología
6.
J Neurochem ; 123(3): 342-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22924626

RESUMEN

Glutamate dehydrogenase (GDH), encoded by GLUD1, participates in the breakdown and synthesis of glutamate, the main excitatory neurotransmitter. In the CNS, besides its primary signaling function, glutamate is also at the crossroad of metabolic and neurotransmitter pathways. Importance of brain GDH was questioned here by generation of CNS-specific GDH-null mice (CnsGlud1(-/-)); which were viable, fertile and without apparent behavioral problems. GDH immunoreactivity as well as enzymatic activity were absent in Cns-Glud1(-/-) brains. Immunohistochemical analyses on brain sections revealed that the pyramidal cells of control animals were positive for GDH, whereas the labeling was absent in hippocampal sections of Cns-Glud1(-/-) mice. Electrophysiological recordings showed that deletion of GDH within the CNS did not alter synaptic transmission in standard conditions. Cns-Glud1(-/-) mice exhibited deficient oxidative catabolism of glutamate in astrocytes, showing that GDH is required for Krebs cycle pathway. As revealed by NMR studies, brain glutamate levels remained unchanged, whereas glutamine levels were increased. This pattern was favored by up-regulation of astrocyte-type glutamate and glutamine transporters and of glutamine synthetase. Present data show that the lack of GDH in the CNS modifies the metabolic handling of glutamate without altering synaptic transmission.


Asunto(s)
Encéfalo/enzimología , Eliminación de Gen , Ácido Glutámico/metabolismo , Receptores de Glutamato/deficiencia , Receptores de Glutamato/genética , Transmisión Sináptica/genética , Animales , Encéfalo/patología , Encéfalo/fisiología , Células Cultivadas , Femenino , Glutamato Deshidrogenasa , Glutamina/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Vías Nerviosas/fisiopatología , Técnicas de Cultivo de Órganos , Receptores de Glutamato/fisiología , Transmisión Sináptica/fisiología
7.
J Lipid Res ; 51(6): 1370-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19965574

RESUMEN

The peroxisome proliferator-activated receptor delta (PPARdelta) is implicated in regulation of mitochondrial processes in a number of tissues, and PPARdelta activation is associated with decreased susceptibility to ectopic lipid deposition and metabolic disease. Here, we show that PPARdelta is the PPAR subtype expressed at the highest level in insulinoma cells and rat pancreatic islets. Furthermore, PPARdelta displays high transcriptional activity and acts in pronounced synergy with retinoid-X-receptor (RXR). Interestingly, unsaturated fatty acids mimic the effects of synthetic PPARdelta agonists. Using short hairpin RNA-mediated knockdown, we demonstrate that the ability of unsaturated fatty acids to stimulate fatty acid metabolism is dependent on PPARdelta. Activation of PPARdelta increases the fatty acid oxidation capacity in INS-1E beta-cells, enhances glucose-stimulated insulin secretion (GSIS) from islets, and protects GSIS against adverse effects of prolonged fatty acid exposure. The presented results indicate that the nuclear receptor PPARdelta is a fatty acid sensor that adapts beta-cell mitochondrial function to long-term changes in unsaturated fatty acid levels. As maintenance of mitochondrial metabolism is essential to preserve beta-cell function, these data indicate that dietary or pharmacological activation of PPARdelta and RXR may be beneficial in the prevention of beta-cell dysfunction.


Asunto(s)
Ácidos Grasos/metabolismo , Ácidos Grasos/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , PPAR delta/metabolismo , Animales , Línea Celular Tumoral , Células Clonales , Sinergismo Farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/patología , Masculino , Mitocondrias/efectos de los fármacos , Ácido Oléico/farmacología , Oxidación-Reducción , PPAR delta/agonistas , PPAR delta/deficiencia , PPAR delta/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Receptores X Retinoide/agonistas , Receptores X Retinoide/metabolismo , Transcripción Genética/efectos de los fármacos
8.
Nature ; 431(7005): 200-5, 2004 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-15306821

RESUMEN

Elucidating the signalling mechanisms by which obesity leads to impaired insulin action is critical in the development of therapeutic strategies for the treatment of diabetes. Recently, mice deficient for S6 Kinase 1 (S6K1), an effector of the mammalian target of rapamycin (mTOR) that acts to integrate nutrient and insulin signals, were shown to be hypoinsulinaemic, glucose intolerant and have reduced beta-cell mass. However, S6K1-deficient mice maintain normal glucose levels during fasting, suggesting hypersensitivity to insulin, raising the question of their metabolic fate as a function of age and diet. Here, we report that S6K1-deficient mice are protected against obesity owing to enhanced beta-oxidation. However on a high fat diet, levels of glucose and free fatty acids still rise in S6K1-deficient mice, resulting in insulin receptor desensitization. Nevertheless, S6K1-deficient mice remain sensitive to insulin owing to the apparent loss of a negative feedback loop from S6K1 to insulin receptor substrate 1 (IRS1), which blunts S307 and S636/S639 phosphorylation; sites involved in insulin resistance. Moreover, wild-type mice on a high fat diet as well as K/K A(y) and ob/ob (also known as Lep/Lep) mice-two genetic models of obesity-have markedly elevated S6K1 activity and, unlike S6K1-deficient mice, increased phosphorylation of IRS1 S307 and S636/S639. Thus under conditions of nutrient satiation S6K1 negatively regulates insulin signalling.


Asunto(s)
Envejecimiento/fisiología , Grasas de la Dieta/farmacología , Resistencia a la Insulina/fisiología , Obesidad/genética , Obesidad/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/deficiencia , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Adipocitos/metabolismo , Animales , Glucemia/análisis , Activación Enzimática , Ayuno/sangre , Ayuno/fisiología , Ácidos Grasos no Esterificados/sangre , Eliminación de Gen , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina , Resistencia a la Insulina/genética , Lipólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/sangre , Obesidad/inducido químicamente , Oxidación-Reducción , Fosfoproteínas/metabolismo , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Insulina/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Transducción de Señal/efectos de los fármacos
9.
Biochem J ; 424(3): 459-66, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19764902

RESUMEN

Transfer of reducing equivalents between cytosolic compartments and the mitochondrial matrix is mediated by NADH shuttles. Among these, the malate-aspartate shuttle has been proposed to play a major role in beta-cells for the control of glucose-stimulated insulin secretion. AGC1 or Aralar1 (aspartate-glutamate carrier 1) is a key component of the malate-aspartate shuttle. Overexpression of AGC1 increases the capacity of the malate-aspartate shuttle, resulting in enhanced metabolism-secretion coupling, both in INS-1E cells and rat islets. In the present study, knockdown of AGC1 was achieved in the same beta-cell models, using adenovirus-mediated delivery of shRNA (small-hairpin RNA). Compared with control INS-1E cells, down-regulation of AGC1 blunted NADH formation (-57%; P<0.05), increased lactate production (+16%; P<0.001) and inhibited glucose oxidation (-22%; P<0.01). This correlated with a reduced secretory response at 15 mM glucose (-25%; P<0.05), while insulin release was unchanged at intermediate 7.5 mM and basal 2.5 mM glucose. In isolated rat islets, efficient AGC1 knockdown did not alter insulin exocytosis evoked by 16.7 mM glucose. However, 4 mM amino-oxyacetate, commonly used to block transaminases of the malate-aspartate shuttle, inhibited glucose-stimulated insulin secretion to similar extents in INS-1E cells (-66%; P<0.01) and rat islets (-56%; P<0.01). These results show that down-regulation of the key component of the malate-aspartate shuttle AGC1 reduced glucose-induced oxidative metabolism and insulin secretion in INS-1E cells, whereas similar AGC1 knockdown in rat islets did not affect their secretory response.


Asunto(s)
Islotes Pancreáticos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Interferencia de ARN , Adenosina Trifosfato/metabolismo , Ácido Aminooxiacético/farmacología , Animales , Calcio/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Glucosa/farmacología , Ácido Glutámico/metabolismo , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Lactatos/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de Transporte de Membrana/genética , Mitocondrias/metabolismo , Mitocondrias/fisiología , Proteínas de Transporte de Membrana Mitocondrial , Proteínas Mitocondriales/genética , Ratas
10.
Adv Exp Med Biol ; 654: 193-216, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20217499

RESUMEN

Pancreatic beta-cells are poised to sense glucose and other nutrient secretagogues to regulate insulin exocytosis, thereby maintaining glucose homeostasis. This process requires translation of metabolic substrates into intracellular messengers recognized by the exocytotic machinery. Central to this metabolism-secretion coupling, mitochondria integrate and generate metabolic signals, thereby connecting glucose recognition to insulin exocytosis. In response to a glucose rise, nucleotides and metabolites are generated by mitochondria and participate, together with cytosolic calcium, to the stimulation of insulin release. This review describes the mitochondrion-dependent pathways of regulated insulin secretion. Mitochondrial defects, such as mutations and reactive oxygen species production, are discussed in the context of beta-cell failure that may participate to the etiology of diabetes.


Asunto(s)
Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/citología , Mitocondrias/fisiología , Adenosina Trifosfato/metabolismo , Animales , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Glutamato Deshidrogenasa/metabolismo , Ácido Glutámico/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Mitocondrias/metabolismo , Modelos Biológicos , NAD/metabolismo , Especies Reactivas de Oxígeno
11.
Riv Psichiatr ; 55(5): 297-300, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33078021

RESUMEN

INTRODUCTION: The term pro-ana (pro-anorexia) means the spread of restrictive eating behaviors and anorectic advices in virtual spaces written by teenagers. The purpose of this pilot study consists in a qualitative and quantitative analysis of foods contained in a linguistic corpus made up of users' comments on pro-ana websites. METHOD: The corpus of pro-ana websites was analyzed through the T2K tool based on word-frequency processing. RESULTS: The results show conversations regarding beverages, products of vegetable origin (fruit, vegetables) and low-calorie foods, with a tendency to limit the fear linked to the choice of high-calorie foods through reassuring and reconcilable language labels ("light", "sugar free"). CONCLUSIONS: These findings specify the food semantics on pro-ana websites associated to an anorectic vocabulary with restrictive diets. The results could be used to characterize the most common food as risk factors within the eating disorders framework.


Asunto(s)
Anorexia , Trastornos de Alimentación y de la Ingestión de Alimentos , Adolescente , Humanos , Italia , Proyectos Piloto , Semántica
12.
Biochim Biophys Acta ; 1777(7-8): 965-72, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18486589

RESUMEN

Glutamate is implicated in numerous metabolic and signalling functions that vary according to specific tissues. Glutamate metabolism is tightly controlled by activities of mitochondrial enzymes and transmembrane carriers, in particular glutamate dehydrogenase and mitochondrial glutamate carriers that have been identified in recent years. It is remarkable that, although glutamate-specific enzymes and transporters share similar properties in most tissues, their regulation varies greatly according to particular organs in order to achieve tissue specific functions. This is illustrated in this review when comparing glutamate handling in liver, brain, and pancreatic beta-cells. We describe the main cellular glutamate pathways and their specific functions in different tissues, ultimately contributing to the control of metabolic homeostasis at the organism level.


Asunto(s)
Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/metabolismo , Ácido Glutámico/metabolismo , Mitocondrias/metabolismo , Animales , Ácido Aspártico/metabolismo , Proteínas Portadoras/metabolismo , Diabetes Mellitus/metabolismo , Homeostasis , Humanos , Especificidad de Órganos
13.
Schizophr Bull ; 45(1): 127-137, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29471549

RESUMEN

Brain imaging has revealed that the CA1 subregion of the hippocampus is hyperactive in prodromal and diagnosed patients with schizophrenia (SCZ), and that glutamate is a driver of this hyperactivity. Strikingly, mice deficient in the glutamate synthetic enzyme glutaminase have CA1 hypoactivity and a SCZ-resilience profile, implicating glutamate-metabolizing enzymes. To address this further, we examined mice with a brain-wide deficit in the glutamate-metabolizing enzyme glutamate dehydrogenase (GDH), encoded by Glud1, which should lead to glutamate excess due to reduced glutamate metabolism in astrocytes. We found that Glud1-deficient mice have behavioral abnormalities in the 3 SCZ symptom domains, with increased baseline and amphetamine-induced hyperlocomotion as a positive symptom proxy, nest building and social preference as a negative symptom proxy, and reversal/extradimensional set shifting in the water T-maze and contextual fear conditioning as a cognitive symptom proxy. Neuroimaging of cerebral blood volume revealed hippocampal hyperactivity in CA1, which was associated with volume reduction. Parameters of hippocampal synaptic function revealed excess glutamate release and an elevated excitatory/inhibitory balance in CA1. Finally, in a direct clinical correlation using imaging-guided microarray, we found a significant SCZ-associated postmortem reduction in GLUD1 expression in CA1. These findings advance GLUD1 deficiency as a driver of excess hippocampal excitatory transmission and SCZ symptoms, and identify GDH as a target for glutamate modulation pharmacotherapy for SCZ. More broadly, these findings point to the likely involvement of alterations in glutamate metabolism in the pathophysiology of SCZ.


Asunto(s)
Conducta Animal/fisiología , Región CA1 Hipocampal , Glutamato Deshidrogenasa/deficiencia , Receptores de Glutamato/metabolismo , Esquizofrenia , Animales , Región CA1 Hipocampal/diagnóstico por imagen , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiopatología , Volumen Sanguíneo Cerebral/fisiología , Modelos Animales de Enfermedad , Femenino , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Esquizofrenia/diagnóstico por imagen , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología
14.
J Neurol Sci ; 405: 116423, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31520869

RESUMEN

Fingolimod exerts its therapeutic effect in multiple sclerosis by modulating sphingosine-1P receptors which are expressed in the heart mediating fingolimod first dose effects. Understanding potential interactions of baseline characteristics and autonomic profile with fingolimod first dose effects may add novel safety information and help explain cases requiring extension of the 6-hour ECG monitoring period. We aimed at characterizing the patient population treated with the first dose of fingolimod in clinical practice in an observational, multicenter, prospective 6-hours (up to 24) study. ECG was recorded for 15 min before first fingolimod administration and for 6 h after. Heart rate (HR) and HR variability in the frequency domain were derived from ECG traces. Out of the 625 enrolled patients, 580 (92.8%) were discharged at the sixth hour after fingolimod first dose; 45 (7.2%) required monitoring extension. Data confirm the well characterized cardiovascular fingolimod profile upon treatment initiation. Ten (1.6%) patients showed an atrioventricular block, all asymptomatic and self-resolving. Normalized spectral power in the High Frequency band (marking vagal modulation) and previous annualized relapse rate were independently correlated with the probability of undergoing extended monitoring. Our results could provide useful information for the stratification and individualized monitoring of MS patients prescribed with fingolimod.


Asunto(s)
Fármacos del Sistema Nervioso Autónomo/farmacología , Monitoreo de Drogas/normas , Clorhidrato de Fingolimod/efectos adversos , Adolescente , Adulto , Anciano , Monitoreo de Drogas/estadística & datos numéricos , Electrocardiografía , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Estudios Prospectivos , Adulto Joven
15.
Biochem Pharmacol ; 72(8): 965-73, 2006 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-16934763

RESUMEN

Topiramate (Topamax), primarily prescribed against epilepsy, was reported to reduce body weight and to ameliorate glycemic control in obese patients with diabetes. In rodent models of obesity and diabetes, topiramate treatment counteracts hyperglycemia and increases insulin levels upon glucose tolerance test. These observations suggest that topiramate might exert direct action on insulin secreting cells, in particular regarding obesity associated beta-cell dysfunction. In this study, INS-1E beta-cells were exposed for 3 days to the fatty acid oleate (0.4mM) and concomitantly treated with therapeutic concentrations of topiramate before measurements of insulin secretion and metabolic parameters. In healthy cells, topiramate had no acute or chronic effects on insulin release. Exposure of INS-1E cells to oleate for 3 days increased insulin release at basal 2.5mM glucose and blunted the response to stimulatory glucose concentration (15mM). Such lipotoxic effects were associated with impaired mitochondrial function, as evidenced by partial loss of resting mitochondrial membrane potential and reduced hyperpolarization in response to glucose. Oil-red-O staining and triglyceride measurements revealed lipid accumulation in oleate treated cells. Topiramate treatment counteracted oleate-induced lipid load and partially protected against mitochondrial membrane dysfunction. In particular, topiramate restored glucose stimulated insulin secretion, essentially by maintaining low insulin release at basal glucose. Topiramate increased expression of the nutrient sensor PPARalpha and of the mitochondrial fatty acid carrier CPT-1, correlating with enhancement of beta-oxidation rate. The data demonstrate that a drug originally used as mood stabilizer exerts a direct action on beta-cells, protecting against lipid-induced dysfunction.


Asunto(s)
Anticonvulsivantes/farmacología , Fructosa/análogos & derivados , Células Secretoras de Insulina/efectos de los fármacos , Ácido Oléico/farmacología , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular , Fructosa/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/fisiología , PPAR alfa/genética , PPAR alfa/metabolismo , ARN Mensajero/metabolismo , Ratas , Topiramato , Triglicéridos/metabolismo
16.
Cell Rep ; 13(2): 365-75, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26440896

RESUMEN

Glucose, the main energy substrate used in the CNS, is continuously supplied by the periphery. Glutamate, the major excitatory neurotransmitter, is foreseen as a complementary energy contributor in the brain. In particular, astrocytes actively take up glutamate and may use it through oxidative glutamate dehydrogenase (GDH) activity. Here, we investigated the significance of glutamate as energy substrate for the brain. Upon glutamate exposure, astrocytes generated ATP in a GDH-dependent way. The observed lack of glutamate oxidation in brain-specific GDH null CnsGlud1(-/-) mice resulted in a central energy-deprivation state with increased ADP/ATP ratios and phospho-AMPK in the hypothalamus. This induced changes in the autonomous nervous system balance, with increased sympathetic activity promoting hepatic glucose production and mobilization of substrates reshaping peripheral energy stores. Our data reveal the importance of glutamate as necessary energy substrate for the brain and the role of central GDH in the regulation of whole-body energy homeostasis.


Asunto(s)
Metabolismo Energético , Ácido Glutámico/metabolismo , Hipotálamo/metabolismo , Receptores de Glutamato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Astrocitos/metabolismo , Células Cultivadas , Glucosa/metabolismo , Glutamato Deshidrogenasa , Hipotálamo/citología , Hígado/metabolismo , Masculino , Ratones , Oxidación-Reducción , Receptores de Glutamato/genética
17.
Neurochem Int ; 59(4): 510-7, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21600947

RESUMEN

Glutamate dehydrogenase (GDH) is a mitochondrial enzyme linking the Krebs cycle to the multifunctional amino acid glutamate. Thereby, GDH plays a pivotal role between carbohydrate and protein metabolisms, controlling production and consumption of the messenger molecule glutamate in neuroendocrine cells. GDH activity is under the control of several regulators, conferring to this enzyme energy-sensor property. Indeed, GDH directly depends on the provision of the co-factor NADH/NAD(+), rendering the enzyme sensitive to the redox status of the cell. Moreover, GDH is allosterically regulated by GTP and ADP. GDH is also regulated by ADP-ribosylation, mediated by a member of the energy-sensor family sirtuins, namely SIRT4. In the brain, GDH ensures the cycling of the neurotransmitter glutamate between neurons and astrocytes. GDH also controls ammonia metabolism and detoxification, mainly in the liver and kidney. In pancreatic ß-cells, the importance of GDH as a key enzyme in the regulation of insulin secretion is now well established. Inhibition of GDH activity decreases insulin release, while activating mutations are associated with a hyperinsulinism syndrome. Although GDH enzyme catalyzes the same reaction in every tissue, its function regarding metabolic homeostasis varies greatly according to specific organs. In this review, we will discuss specificities of GDH regulation in neuroendocrine cells, in particular pancreatic islets and central nervous system.


Asunto(s)
Sistema Nervioso Central/enzimología , Metabolismo Energético/fisiología , Glutamato Deshidrogenasa/metabolismo , Homeostasis/fisiología , Islotes Pancreáticos/enzimología , Animales , Glutamato Deshidrogenasa/antagonistas & inhibidores , Glutamato Deshidrogenasa/fisiología , Humanos , Mitocondrias/metabolismo
18.
J Endocrinol ; 205(1): 79-86, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20061513

RESUMEN

Sclerocarya birrea is a plant widely used as traditional medication for the treatment of diabetes in sub-Saharan regions. However, the mechanism of action is unknown and only hypoglycaemic effects of S. birrea extract (SBE) in diabetic rats have been reported to date. Here, we tested aqueous extracts of S. birrea on insulin-secreting INS-1E cells and isolated rat islets. Following 24 h of treatment at 5 microg/ml, the extract markedly potentiated glucose-stimulated insulin secretion. Neither basal insulin release nor non-nutrient stimulation was affected. The potentiation of the secretory response at stimulatory glucose appeared after 12 h of treatment. No acute effects were observed and, at the effective concentration, SBE was safe regarding cell integrity and differentiation. The mechanism of action of the SBE was related to glucose metabolism as both ATP generation and glucose oxidation were enhanced following the 24-h treatment. In streptozotocin-induced diabetic rats, SBE administration corrected glycaemia and restored plasma insulin levels after 2 weeks of treatment. These data show direct action of S. birrea on insulin-secreting cells and favour further delineation for use of the plant in the management of diabetes.


Asunto(s)
Anacardiaceae , Diabetes Mellitus Experimental/tratamiento farmacológico , Glucosa/farmacología , Hiperglucemia/tratamiento farmacológico , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Extractos Vegetales/uso terapéutico , Adenosina Trifosfato/metabolismo , Animales , Línea Celular Tumoral , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Glucosa/metabolismo , Hiperglucemia/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/metabolismo , Insulinoma/patología , Masculino , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Corteza de la Planta , Extractos Vegetales/farmacología , Tallos de la Planta , Ratas , Ratas Wistar , Estreptozocina
19.
Dev Cell ; 18(5): 763-74, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20493810

RESUMEN

Earlier, we reported that S6K1(-/-) mice have reduced body fat mass, have elevated rates of lipolysis, have severely decreased adipocyte size, and are resistant to high fat diet (HFD)-induced obesity. Here we report that adipocytes of S6K1(-/-) mice on a HFD have the capacity to increase in size to a degree comparable to that of wild-type (WT) mice, but not in number, indicating an unexpected lesion in adipogenesis. Tracing this lesion revealed that S6K1 is dispensable for terminal adipocyte differentiation, but is involved in the commitment of embryonic stem cells to early adipocyte progenitors. We further show that absence of S6K1 attenuates the upregulation of transcription factors critical for commitment to adipogenesis. These results led to the conclusion that a lack of S6K1 impairs the generation of de novo adipocytes when mice are challenged with a HFD, consistent with a reduction in early adipocyte progenitors.


Asunto(s)
Adipocitos/citología , Diferenciación Celular/fisiología , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/fisiología , Adipocitos/patología , Adipogénesis/genética , Adipogénesis/fisiología , Tejido Adiposo Blanco/anatomía & histología , Animales , Humanos , Hiperplasia/genética , Masculino , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/prevención & control , ARN Mensajero/genética
20.
J Biol Chem ; 284(2): 921-9, 2009 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-19015267

RESUMEN

Insulin exocytosis is regulated in pancreatic ss-cells by a cascade of intracellular signals translating glucose levels into corresponding secretory responses. The mitochondrial enzyme glutamate dehydrogenase (GDH) is regarded as a major player in this process, although its abrogation has not been tested yet in animal models. Here, we generated transgenic mice, named betaGlud1(-/-), with ss-cell-specific GDH deletion. Our results show that GDH plays an essential role in the full development of the insulin secretory response. In situ pancreatic perfusion revealed that glucose-stimulated insulin secretion was reduced by 37% in betaGlud1(-/-). Furthermore, isolated islets with either constitutive or acute adenovirus-mediated knock-out of GDH showed a 49 and 38% reduction in glucose-induced insulin release, respectively. Adenovirus-mediated re-expression of GDH in betaGlud1(-/-) islets fully restored glucose-induced insulin release. Thus, GDH appears to account for about 40% of glucose-stimulated insulin secretion and to lack redundant mechanisms. In betaGlud1(-/-) mice, the reduced secretory capacity resulted in lower plasma insulin levels in response to both feeding and glucose load, while body weight gain was preserved. The results demonstrate that GDH is essential for the full development of the secretory response in beta-cells. However, maximal secretory capacity is not required for maintenance of glucose homeostasis in normo-caloric conditions.


Asunto(s)
Glucosa/metabolismo , Glutamato Deshidrogenasa/deficiencia , Glutamato Deshidrogenasa/metabolismo , Homeostasis , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Envejecimiento/fisiología , Animales , Separación Celular , Eliminación de Gen , Glutamato Deshidrogenasa/genética , Secreción de Insulina , Ratones , Ratones Noqueados , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA