Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Physiol Pharmacol ; 68(5): 669-681, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29375041

RESUMEN

Orexin regulates food intake and energy expenditure. Here, we test the ability of orexin-A (OXA, hypocretin-1) at improving metabolic control in type 2 diabetic animals and elaborate potential mechanisms of action. Rats with experimentally induced type 2 diabetes by a combination of streptozotocin injection and high-fat diet feeding were chronically infused with OXA. In vitro experiments were conducted on isolated pancreatic islets, primary adipocytes and insulin secreting INS-1E cells. OXA improved glucose control, enhanced insulin sensitivity and attenuated pancreatic ß-cell loss in type 2 diabetic rats. Ex vivo, apoptotic death of pancreatic islets isolated from OXA-treated type 2 diabetic animals as well as the impairment of glucose-stimulated insulin secretion were attenuated, as compared to islets derived from vehicle-treated rats. OXA reduced plasma tumor necrosis factor-α (TNF-α) and non-esterified fatty acids (NEFA) levels in type 2 diabetic rats. OXA decreased palmitate- and TNF-α-induced apoptosis of INS-1E cells. OXA improves glucose control by enhancing insulin sensitivity and protecting ß-cells from apoptotic cell death in type 2 diabetic animals.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Células Secretoras de Insulina/efectos de los fármacos , Orexinas/uso terapéutico , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 2/sangre , Células Secretoras de Insulina/metabolismo , Masculino , Orexinas/farmacología , Ratas , Resultado del Tratamiento
2.
Biochim Biophys Acta ; 1850(2): 343-51, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25445712

RESUMEN

BACKGROUND: Plasma levels of pancreatic polypeptide (PP) rise upon food intake. Although other pancreatic islet hormones, such as insulin and glucagon, have been extensively investigated, PP secretion and actions are still poorly understood. METHODS: The release of PP upon glucose stimulation and the effects of PP on glucagon and insulin secretion were analyzed in isolated pancreatic islets. Expression of PP receptor (PPYR1) was investigated by immunoblotting, quantitative RT-PCR on sorted pancreatic islet cells, and immunohistochemistry. RESULTS: In isolated mouse pancreatic islets, glucose stimulation increased PP release, while insulin secretion was up and glucagon release was down. Direct exposure of islets to PP inhibited glucagon release. In mouse islets, PPYR1 protein was observed by immunoblotting and quantitative RT-PCR revealed PPYR1 expression in the FACS-enriched glucagon alpha-cell fraction. Immunohistochemistry on pancreatic sections showed the presence of PPYR1 in alpha-cells of both mouse and human islets, while the receptor was absent in other islet cell types and exocrine pancreas. CONCLUSIONS: Glucose stimulates PP secretion and PP inhibits glucagon release in mouse pancreatic islets. PP receptors are present in alpha-cells of mouse and human pancreatic islets. GENERAL SIGNIFICANCE: These data demonstrate glucose-regulated secretion of PP and its effects on glucagon release through PPYR1 receptors expressed by alpha-cells.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Polipéptido Pancreático/metabolismo , Receptores de Neuropéptido Y/biosíntesis , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Secretoras de Glucagón/citología , Glucosa/farmacología , Humanos , Inmunohistoquímica , Masculino , Ratones , Edulcorantes/farmacología
3.
Biochem Pharmacol ; 79(12): 1827-36, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20138024

RESUMEN

Treatment with the dopamine receptor D2 (DRD2) agonist bromocriptine improves metabolic features in obese patients with type 2 diabetes by a still unknown mechanism. In the present study, we investigated the acute effect of bromocriptine and its underlying mechanism(s) on insulin secretion both in vivo and in vitro. For this purpose, C57Bl6/J mice were subjected to an intraperitoneal glucose tolerance test (ipGTT) and a hyperglycemic (HG) clamp 60min after a single injection of bromocriptine or placebo. The effects of bromocriptine on glucose-stimulated insulin secretion (GSIS), cell membrane potential and intracellular cAMP levels were also determined in INS-1E beta cells. We report here that bromocriptine increased glucose levels during ipGTT in vivo, an effect associated with a dose-dependent decrease in GSIS. During the HG clamp, bromocriptine reduced both first-phase and second-phase insulin response. This inhibitory effect was also observed in INS-1E beta cells, in which therapeutic concentrations of bromocriptine (0.5-50nM) decreased GSIS. Mechanistically, neither cellular energy state nor cell membrane depolarization was affected by bromocriptine whereas intracellular cAMP levels were significantly reduced, suggesting involvement of G-protein-coupled receptors. Surprisingly, the DRD2 antagonist domperidone did not counteract the effect of bromocriptine on GSIS, whereas yohimbine, an antagonist of the alpha2-adrenergic receptors, completely abolished bromocriptine-induced inhibition of GSIS. In conclusion, acute administration of bromocriptine inhibits GSIS by a DRD2-independent mechanism involving direct activation of the pancreatic alpha2-adrenergic receptors. We suggest that treatment with bromocriptine promotes beta cells rest, thereby preventing long-lasting hypersecretion of insulin and subsequent beta cell failure.


Asunto(s)
Bromocriptina/farmacología , Agonistas de Dopamina/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Receptores Adrenérgicos alfa/metabolismo , Receptores de Dopamina D2/agonistas , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Glucosa/farmacología , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Insulinoma , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas
4.
Diabetologia ; 53(2): 331-40, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19908022

RESUMEN

AIMS/HYPOTHESIS: Pancreatic beta cells chronically exposed to fatty acids may lose specific functions and even undergo apoptosis. Generally, lipotoxicity is triggered by saturated fatty acids, whereas unsaturated fatty acids induce lipodysfunction, the latter being characterised by elevated basal insulin release and impaired glucose responses. The peroxisome proliferator-activated receptor alpha (PPARalpha) has been proposed to play a protective role in this process, although the cellular mechanisms involved are unclear. METHODS: We modulated PPARalpha production in INS-1E beta cells and investigated key metabolic pathways and genes responsible for metabolism-secretion coupling during a culture period of 3 days in the presence of 0.4 mmol/l oleate. RESULTS: In INS-1E cells, the secretory dysfunction primarily induced by oleate was aggravated by silencing of PPARalpha. Conversely, PPARalpha upregulation preserved glucose-stimulated insulin secretion, essentially by increasing the response at a stimulatory concentration of glucose (15 mmol/l), a protection we also observed in human islets. The protective effect was associated with restored glucose oxidation rate and upregulation of the anaplerotic enzyme pyruvate carboxylase. PPARalpha overproduction increased both beta-oxidation and fatty acid storage in the form of neutral triacylglycerol, revealing overall induction of lipid metabolism. These observations were substantiated by expression levels of associated genes. CONCLUSIONS/INTERPRETATION: PPARalpha protected INS-1E beta cells from oleate-induced dysfunction, promoting both preservation of glucose metabolic pathways and fatty acid turnover.


Asunto(s)
Carbohidratos/fisiología , Células Secretoras de Insulina/fisiología , Ácido Oléico/toxicidad , PPAR alfa/fisiología , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Antígenos CD36/genética , Carnitina O-Palmitoiltransferasa/genética , Técnicas de Cultivo de Célula , Ácidos Grasos no Esterificados/farmacología , Regulación de la Expresión Génica , Glucosa/farmacología , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , PPAR alfa/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tubulina (Proteína)/genética
5.
Diabetologia ; 51(8): 1429-39, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18385973

RESUMEN

AIMS/HYPOTHESIS: The expression of several neuronal genes in pancreatic beta cells is due to the absence of the transcription factor repressor element 1 (RE-1) silencing transcription factor (REST). The identification of these traits and their functional significance in beta cells has only been partly elucidated. Herein, we investigated the biological consequences of a repression of REST target genes by expressing REST in beta cells. METHODS: The effect of REST expression on glucose homeostasis, insulin content and release, and beta cell mass was analysed in transgenic mice selectively expressing REST in beta cells. Relevant target genes were identified in INS-1E and primary beta cells expressing REST. RESULTS: Transgenic mice featuring a beta cell-targeted expression of REST exhibited glucose intolerance and reduced beta cell mass. In primary beta cells, REST repressed several proteins of the exocytotic machinery, including synaptosomal-associated protein (SNAP) 25, synaptotagmin (SYT) IV, SYT VII, SYT IX and complexin II; it impaired first and second phases of insulin secretion. Using RNA interference in INS-1E cells, we showed that SYT IV and SYT VII were implicated in the control of insulin release. CONCLUSIONS/INTERPRETATION: The data document the critical role of REST target genes in pancreatic beta cells. Specifically, we provide evidence that the downregulation of these genes is detrimental for the exocytosis of large dense core vesicles, thus contributing to beta cell dysfunction and impaired glucose homeostasis.


Asunto(s)
Silenciador del Gen , Células Secretoras de Insulina/fisiología , Proteínas Represoras/genética , Animales , Cromatina/fisiología , Secuencia de Consenso , Citosol/fisiología , Genes Reporteros , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Ratones Transgénicos , Membranas Mitocondriales/fisiología , Proteínas Represoras/fisiología
6.
Biochem Soc Trans ; 34(Pt 5): 811-4, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17052204

RESUMEN

The coupling of cytosolic glycolytic NADH production with the mitochondrial electron transport chain is crucial for pancreatic beta-cell function and energy metabolism. The activity of lactate dehydrogenase in the beta-cell is low, thus glycolysis-derived electrons are transported towards the mitochondrial matrix by a NADH shuttle system, which in turn regenerates cytosolic NAD+. Mitochondrial electron transport then produces ATP, the main coupling factor for insulin secretion. Aralar1, a Ca2+-sensitive member of the malate-aspartate shuttle expressed in beta-cells, has been found to play a significant role in nutrient-stimulated insulin secretion and beta-cell function. Increased capacity of Aralar1 enhances the responsiveness of the cell to glucose. Conversely, inhibition of the malate-aspartate shuttle results in impaired glucose metabolism and insulin secretion. Current research investigates potentiating or attenuating activities of various amino acids on insulin secretion, mitochondrial membrane potential and NADH production in Aralar1-overexpressing beta-cells. This work may provide evidence for a central role of Aralar1 in the regulation of nutrient metabolism in the beta-cells.


Asunto(s)
Metabolismo Energético , Células Secretoras de Insulina/fisiología , Animales , Ácido Aspártico/metabolismo , Citosol/fisiología , Flavina-Adenina Dinucleótido/metabolismo , Glucosa/metabolismo , Glicerofosfatos/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Malatos/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , NAD/metabolismo , Oxidación-Reducción
7.
Biochem Soc Trans ; 34(Pt 5): 824-7, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17052207

RESUMEN

Pancreatic beta-cells are able to respond to nutrients, principally glucose, as the primary stimulus for insulin exocytosis. This unique feature requires translation of metabolic substrates into intracellular messengers recognized by the exocytotic machinery. Central to this signal transduction mechanism, mitochondria integrate and generate metabolic signals, thereby coupling glucose recognition with insulin secretion. In response to a glucose rise, nucleotides and metabolites are generated by mitochondria and participate, together with cytosolic Ca2+, in the stimulation of insulin exocytosis. Mitochondrial defects, such as mutations and ROS (reactive oxygen species) production, might be associated with beta-cell failure in the course of diabetes. mtDNA (mitochondrial DNA) mutation A3243G is associated with MIDD (mitochondrial inherited diabetes and deafness). A common hypothesis to explain the link between the genotype and the phenotype is that the mutation might impair mitochondrial metabolism expressly required for beta-cell functions, although this assumption lacks direct demonstration. mtDNA-deficient cellular models are glucose-unresponsive and are defective in mitochondrial function. Recently, we used clonal cytosolic hybrid cells (namely cybrids) harbouring mitochondria derived from MIDD patients. Compared with control mtDNA from the same patient, the A3243G mutation markedly modified metabolic pathways. Moreover, cybrid cells carrying patient-derived mutant mtDNA exhibited deranged cell Ca2+ handling and elevated ROS under metabolic stress. In animal models, transgenic mice lacking expression of the mitochondrial genome specifically in beta-cells are diabetic and their islets are incable of releasing insulin in response to glucose. These various models demonstrate the fragility of nutrient-stimulated insulin secretion, caused primarily by defective mitochondrial function.


Asunto(s)
ADN Mitocondrial/genética , Insulina/metabolismo , Mitocondrias/patología , Glucosa/metabolismo , Humanos , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/patología , Mutación , Especies Reactivas de Oxígeno/metabolismo
8.
J Thromb Haemost ; 4(7): 1588-95, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16839358

RESUMEN

OBJECTIVE: von Willebrand factor (VWF) is acutely released from endothelial cells in response to numerous calcium-raising agents (e.g. thrombin, histamine) and cAMP-raising agents (e.g. epinephrine, adenosine, vasopressin). In contrast, very few inhibitors of endothelial VWF secretion have been described. The neurotransmitter dopamine is a modulator of exocytosis in several endocrine cells, and is possibly involved in the regulation of several endothelial cell functions. We therefore investigated the effect of dopamine on endothelial VWF secretion. RESULTS: Dopamine, D2/D3- and D4-specific agonists inhibited histamine- but not thrombin-induced VWF secretion. Expression of dopamine D2, D3 and D4 receptors was demonstrated by reverse transcription polymerase chain reaction (RT-PCR) in both human aortic (HAEC) and umbilical vein (HUVEC) endothelial cells. D2-D4 agonists did not inhibit histamine-induced rise in [Ca(2+)](i): they inhibited histamine-induced secretion even in the absence of extracellular calcium. Thus, the dopamine effects are not mediated by [Ca(2+)](i)-dependent signalling. D2/D3- and D4-specific agonists inhibited neither the rise in cAMP nor VWF secretion in response to epinephrine and adenosine, arguing against an effect on cAMP-mediated signalling. D1 and D5 receptors were not detected in HAEC or HUVEC by RT-PCR, and the D1/D5-specific agonist SKF 38 393 failed to modulate VWF secretion, arguing against a role for these receptors in endothelial exocytosis. CONCLUSIONS: Dopamine inhibits histamine-induced endothelial exocytosis by activating D2-D4 receptor, via a mechanism distinct from [Ca(2+)](i)-or cAMP-mediated signaling. In contrast, D1 and D5 receptors are not functionally expressed in cultured endothelial cells. Dopamine agonists may be useful as inhibitors of endothelial activation in inflammation and cardiovascular disease.


Asunto(s)
Dopamina/fisiología , Células Endoteliales/metabolismo , Receptores Dopaminérgicos/fisiología , Factor de von Willebrand/metabolismo , Aorta/citología , Células Cultivadas , Agonistas de Dopamina/farmacología , Endotelio Vascular/citología , Exocitosis , Histamina , Humanos , Receptores de Dopamina D2/fisiología , Receptores de Dopamina D3/fisiología , Receptores de Dopamina D4/fisiología , Transducción de Señal , Venas Umbilicales/citología
9.
Diabetologia ; 49(8): 1816-26, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16736129

RESUMEN

AIMS/HYPOTHESIS: Mitochondrial DNA (mtDNA) mutations cause several diseases, including mitochondrial inherited diabetes and deafness (MIDD), typically associated with the mtDNA A3243G point mutation on tRNALeu gene. The common hypothesis to explain the link between the genotype and the phenotype is that the mutation might impair mitochondrial metabolism expressly required for beta cell functions. However, this assumption has not yet been tested. METHODS: We used clonal osteosarcoma cytosolic hybrid cells (namely cybrids) harbouring mitochondria derived from MIDD patients and containing either exclusively wild-type or mutated (A3243G) mtDNA. According to the importance of mitochondrial metabolism in beta cells, we studied the impact of the mutation on key parameters by comparing stimulation of these cybrids by the main insulin secretagogue glucose and the mitochondrial substrate pyruvate. RESULTS: Compared with control mtDNA from the same patient, the A3243G mutation markedly modified metabolic pathways leading to a high glycolytic rate (2.8-fold increase), increased lactate production (2.5-fold), and reduced glucose oxidation (-83%). We also observed impaired NADH responses (-56%), negligible mitochondrial membrane potential, and reduced, only transient ATP generation. Moreover, cybrid cells carrying patient-derived mutant mtDNA exhibited deranged cell calcium handling with increased cytosolic loads (1.4-fold higher), and elevated reactive oxygen species (2.6-fold increase) under glucose deprivation. CONCLUSIONS/INTERPRETATION: The present study demonstrates that the mtDNA A3243G mutation impairs crucial metabolic events required for proper cell functions, such as coupling of glucose recognition to insulin secretion.


Asunto(s)
ADN Mitocondrial/genética , Diabetes Mellitus/genética , Islotes Pancreáticos/metabolismo , Mutación , Polimorfismo de Nucleótido Simple , Adenosina Trifosfato/metabolismo , Línea Celular , Glucosa/metabolismo , Glucólisis/genética , Humanos , Insulina/metabolismo , Secreción de Insulina
11.
Diabetologia ; 47(2): 266-76, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14689183

RESUMEN

AIMS/HYPOTHESIS: Glutamate dehydrogenase (GDH) is a mitochondrial enzyme playing a key role in the control of insulin secretion. However, it is not known whether GDH expression levels in beta cells are rate-limiting for the secretory response to glucose. GDH also controls glutamine and glutamate oxidative metabolism, which is only weak in islets if GDH is not allosterically activated by L-leucine or (+/-)-2-aminobicyclo-[2,2,1]heptane-2-carboxylic acid (BCH). METHODS: We constructed an adenovirus encoding for GDH to overexpress the enzyme in the beta-cell line INS-1E, as well as in isolated rat and mouse pancreatic islets. The secretory responses to glucose and glutamine were studied in static and perifusion experiments. Amino acid concentrations and metabolic parameters were measured in parallel. RESULTS: GDH overexpression in rat islets did not change insulin release at basal or intermediate glucose (2.8 and 8.3 mmol/l respectively), but potentiated the secretory response at high glucose concentrations (16.7 mmol/l) compared to controls (+35%). Control islets exposed to 5 mmol/l glutamine at basal glucose did not increase insulin release, unless BCH was added with a resulting 2.5-fold response. In islets overexpressing GDH glutamine alone stimulated insulin secretion (2.7-fold), which was potentiated 2.2-fold by adding BCH. The secretory responses evoked by glutamine under these conditions correlated with enhanced cellular metabolism. CONCLUSIONS/INTERPRETATION: GDH could be rate-limiting in glucose-induced insulin secretion, as GDH overexpression enhanced secretory responses. Moreover, GDH overexpression made islets responsive to glutamine, indicating that under physiological conditions this enzyme acts as a gatekeeper to prevent amino acids from being inappropriate efficient secretagogues.


Asunto(s)
Glutamato Deshidrogenasa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Adenosina Trifosfato/metabolismo , Aminoácidos/efectos de los fármacos , Aminoácidos/metabolismo , Aminoácidos Cíclicos/farmacología , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Glucosa/farmacología , Glutamina/farmacología , Humanos , Inmunohistoquímica , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Leucina/análogos & derivados , Leucina/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Mitocondrias/química , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Oxidación-Reducción/efectos de los fármacos , Cloruro de Potasio/farmacología , Ratas , Ratas Wistar
12.
Diabetologia ; 46(10): 1394-401, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-13680126

RESUMEN

AIMS/HYPOTHESIS: The electrons of the glycolysis-derived reduced form of NADH are transferred to mitochondria through the NADH shuttle system. There are two NADH shuttles: the glycerol phosphate and malate-aspartate shuttle. Mice with a targeted disruption of mitochondrial glycerol-3-phosphate dehydrogenase, a rate-limiting enzyme of the glycerol phosphate shuttle, are not diabetic and have normal islet glucose-induced secretion. In this study, we analyzed if environmental factors, such as a high carbohydrate diet could contribute to the development of Type 2 diabetes mellitus in mice with a specific defective genetic background. METHODS: The mice were fed with a high carbohydrate diet for 1 and 6 months, and several biochemical parameters were analysed. The mitochondrial respiratory activity was assayed by polarography; and the islet function was studied by islet perifusion and pancreas perfusion. RESULTS: The high carbohydrate diet induced hyperglycaemia, hyperinsulinaemia, and islet hyperplasia in the wild-type and heterozygote mice. Activity of the respiratory chain complex I also increased in these mice. In contrast, these effects were not observed in the null mice fed with the diet; in addition, these null mice had an increased insulin sensitivity compared to wild-type mice. CONCLUSION/INTERPRETATION: The phenotype of the mice with an impairment of NADH shuttles does not worsen when fed a high carbohydrate diet; moreover, the diet does not compromise islet function.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Carbohidratos de la Dieta/administración & dosificación , Glicerolfosfato Deshidrogenasa/deficiencia , Hiperglucemia/etiología , Mitocondrias/enzimología , Animales , Glucemia/análisis , Respiración de la Célula/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Lípidos/sangre , Ratones , Ratones Noqueados , Mitocondrias/fisiología , Factores de Tiempo
13.
Cell Calcium ; 31(2): 97-104, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11969250

RESUMEN

Rapid formation of high-Ca2+ perimitochondrial cytoplasmic microdomains has been shown to evoke mitochondrial Ca2+ signal and activate mitochondrial dehydrogenases, however, the significance of submicromolar cytoplasmic Ca2+ concentrations in the control of mitochondrial metabolism has not been sufficiently elucidated. Here we studied the mitochondrial response to application of Ca2+ at buffered concentrations in permeabilized rat adrenal glomerulosa cells, in an insulin-producing cell line (INS-1/EK-3) and in an osteosarcoma cell line (143BmA-13). Mitochondrial Ca2+ concentration was measured with the fluorescent dye rhod-2 and, using an in situ calibration method, with the mitochondrially targeted luminescent protein mt-aequorin. In both endocrine cell types, mitochondrial Ca2+ concentration increased in response to elevated cytoplasmic Ca2+ concentration (between 60 and 740 nM) and an increase in mitochondrial Ca2+ concentration could be revealed already at a cytoplasmic Ca2+ concentration step from 60-140 nM. Similar responses were observed in the osteosarcoma cell line, although a clearcut response was first observed at 280 nM extramitochondrial Ca2+ only. As examined in glomerulosa cells, graded increases in cytoplasmic Ca2+ concentration were associated with graded increases in the reduction of mitochondrial pyridine nucleotides, consistent with Ca2+-dependent activation of mitochondrial dehydrogenases. Our data indicate that in addition to the recognized role of high-Ca2+ cytoplasmic microdomains, also small Ca2+ signals may influence mitochondrial metabolism.


Asunto(s)
Calcio/metabolismo , Mitocondrias/metabolismo , Aequorina , Animales , Señalización del Calcio , Línea Celular , Colorantes Fluorescentes , Compuestos Heterocíclicos con 3 Anillos , Masculino , NADP/metabolismo , Oxidación-Reducción , Ratas , Ratas Wistar , Células Tumorales Cultivadas , Zona Glomerular/citología , Zona Glomerular/metabolismo
14.
Cell Mol Life Sci ; 59(11): 1803-18, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12530515

RESUMEN

Mitochondrial metabolism is crucial for the coupling of glucose recognition to the exocytosis of the insulin granules. This is illustrated by in vitro and in vivo observations discussed in the present review. Mitochondria generate ATP, which is the main coupling messenger in insulin secretion. However, the subsequent Ca2+ signal in the cytosol is necessary but not sufficient for full development of sustained insulin secretion. Hence, mitochondria generate ATP and other coupling factors serving as fuel sensors for the control of the exocytotic process. Numerous studies have sought to identify the factors that mediate the amplifying pathway over the Ca2+ signal in glucose-stimulated insulin secretion. Predominantly, these factors are nucleotides (GTP, ATP, cAMP, NADPH), although metabolites have also been proposed, such as long-chain acyl-CoA derivatives and glutamate. Hence, the classical neurotransmitter glutamate receives a novel role, that of an intracellular messenger or co-factor in insulin secretion. This scenario further highlights the importance of glutamate dehydrogenase, a mitochondrial enzyme well recognized to play a key role in the control of insulin secretion. Therefore, additional putative messengers of mitochondrial origin are likely to participate in insulin secretion.


Asunto(s)
Exocitosis/fisiología , Insulina/metabolismo , Islotes Pancreáticos/fisiología , Islotes Pancreáticos/ultraestructura , Mitocondrias/fisiología , Animales , ADN Mitocondrial/metabolismo , Glucosa/metabolismo , Glutamato Deshidrogenasa/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Secreción de Insulina , Transducción de Señal/fisiología
15.
Nature ; 414(6865): 807-12, 2001 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-11742413

RESUMEN

The aetiology of type 2, or non-insulin-dependent, diabetes mellitus has been characterized in only a limited number of cases. Among these, mitochondrial diabetes, a rare subform of the disease, is the consequence of pancreatic beta-cell dysfunction caused by mutations in mitochondrial DNA, which is distinct from the nuclear genome. The impact of such mutations on beta-cell function reflects the importance of mitochondria in the control of insulin secretion. The beta-cell mitochondria serve as fuel sensors, generating factors that couple nutrient metabolism to the exocytosis of insulin-containing vesicles. The latter process requires an increase in cytosolic Ca2+, which depends on ATP synthesized by the mitochondria. This organelle also generates other factors, of which glutamate has been proposed as a potential intracellular messenger.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Islotes Pancreáticos/fisiología , Mitocondrias/fisiología , Enfermedades Mitocondriales/fisiopatología , Animales , ADN Mitocondrial , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Exocitosis , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/fisiopatología , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/genética , Mutación , Transducción de Señal
16.
J Biol Chem ; 276(39): 36391-6, 2001 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-11473118

RESUMEN

Mitochondrial metabolism plays a pivotal role in the pancreatic beta cell by generating signals that couple glucose sensing to insulin secretion. We have demonstrated previously that mitochondrially derived glutamate participates directly in the stimulation of insulin exocytosis. The aim of the present study was to impose altered cellular glutamate levels by overexpression of glutamate decarboxylase (GAD) to repress elevation of cytosolic glutamate. INS-1E cells infected with a recombinant adenovirus vector encoding GAD65 showed efficient overexpression of the GAD protein with a parallel increase in enzyme activity. In control cells glutamate levels were slightly increased by 7.5 mm glucose (1.4-fold) compared with the effect at 15 mm (2.3-fold) versus basal 2.5 mm glucose. Upon GAD overexpression, glutamate concentrations were no longer elevated by 15 mm glucose as compared with controls (-40%). Insulin secretion was stimulated in control cells by glucose at 7.5 mm (2.5-fold) and more efficiently at 15 mm (5.2-fold). INS-1E cells overexpressing GAD exhibited impaired insulin secretion on stimulation with 15 mm glucose (-37%). The secretory response to 30 mm KCl, used to raise cytosolic Ca(2+) levels, was unaffected. Similar results were obtained in perifused rat pancreatic islets following adenovirus transduction. This GAD65-mediated glutamate decarboxylation correlating with impaired glucose-induced insulin secretion is compatible with a role for glutamate as a glucose-derived factor participating in insulin exocytosis.


Asunto(s)
Glucosa/metabolismo , Glutamato Descarboxilasa/metabolismo , Ácido Glutámico/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Isoenzimas/metabolismo , Adenosina Trifosfato/metabolismo , Adenoviridae/genética , Animales , Calcio/metabolismo , Células Cultivadas , Citosol/metabolismo , Exocitosis , Glutamatos/metabolismo , Humanos , Immunoblotting , Secreción de Insulina , Islotes Pancreáticos/citología , Potenciales de la Membrana , Microscopía Fluorescente , Mitocondrias/metabolismo , Unión Proteica , Ratas , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Transducción Genética , Células Tumorales Cultivadas
17.
J Biol Chem ; 276(27): 25279-86, 2001 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-11309388

RESUMEN

The absence of Pdx1 and the expression of brain-4 distinguish alpha-cells from other pancreatic endocrine cell lineages. To define the transcription factor responsible for pancreatic cell differentiation, we employed the reverse tetracycline-dependent transactivator system in INS-I cell-derived subclones INSralphabeta and INSrbeta to achieve tightly controlled and conditional expression of wild type Pdx1 or its dominant-negative mutant, as well as brain-4. INSralphabeta cells express not only insulin but also glucagon and brain-4, while INSrbeta cells express only insulin. Overexpression of Pdx1 eliminated glucagon mRNA and protein in INSralphabeta cells and promoted the expression of beta-cell-specific genes in INSrbeta cells. Induction of dominant-negative Pdx1 in INSralphabeta cells resulted in differentiation of insulin-producing beta-cells into glucagon-containing alpha-cells without altering brain4 expression. Loss of Pdx1 function alone in INSrbeta cells, which do not express endogenous brain-4 and glucagon, was also sufficient to abolish the expression of genes restricted to beta-cells and to cause alpha-cell differentiation. In contrast, induction of brain-4 in INSrbeta cells initiated detectable expression of glucagon but did not affect beta-cell-specific gene expression. In conclusion, Pdx1 confers the expression of pancreatic beta-cell-specific genes, such as genes encoding insulin, islet amyloid polypeptide, Glut2, and Nkx6.1. Pdx1 defines pancreatic cell lineage differentiation. Loss of Pdx1 function rather than expression of brain4 is a prerequisite for alpha-cell differentiation.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Homeodominio , Páncreas/citología , Transactivadores/genética , Animales , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Glucagón/biosíntesis , Insulinoma/química , Islotes Pancreáticos/metabolismo , Peso Molecular , Neoplasias Pancreáticas/química , Ratas , Células Tumorales Cultivadas
18.
J Biol Chem ; 276(6): 3791-7, 2001 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-11036072

RESUMEN

Mitochondrial Ca(2+) concentration ([Ca(2+)](m)) was monitored in C2C12 skeletal muscle cells stably expressing the Ca(2+)-sensitive photoprotein aequorin targeted to mitochondria. In myotubes, KCl-induced depolarization caused a peak of 3.03 +/- 0.14 micrometer [Ca(2+)](m) followed by an oscillatory second phase (5.1 +/- 0.1 per min). Chelation of extracellular Ca(2+) or blockade of the voltage-operated Ca(2+) channel attenuated both phases of the KCl response. The inhibitor of the sarcoplasmic reticulum Ca(2+)-ATPase, cyclopiazonic acid, reduced the amplitude of the KCl-induced [Ca(2+)](m) peak and prevented the oscillations, suggesting that these were generated intracellularly. No such [Ca(2+)](m) oscillations occurred with the nicotinic agonist carbachol, cyclopiazonic acid alone, or the purinergic agonist ATP. In contrast, caffeine produced an oscillatory behavior, indicating a role of ryanodine receptors as mediators of the oscillations. The [Ca(2+)](m) response was desensitized when cells were exposed to two consecutive challenges with KCl separated by a 5-min wash, whereas a second pulse of carbachol potentiated [Ca(2+)](m), indicating differences in intracellular Ca(2+) redistribution. Cross-desensitization between KCl and carbachol and cross-potentiation between carbachol and KCl were observed. These results suggest that close contacts between mitochondria and sarcoplasmic reticulum exist permitting Ca(2+) exchanges during KCl depolarization. These newly demonstrated dynamic changes in [Ca(2+)](m) in stimulated skeletal muscle cells might contribute to the understanding of physiological and pathological processes in muscular disorders.


Asunto(s)
Calcio/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Animales , Línea Celular , Ratones , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Cloruro de Potasio/farmacología
19.
J Physiol ; 529 Pt 1: 49-56, 2000 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11080250

RESUMEN

Glucose-induced insulin secretion is determined by signals generated in the mitochondria. The elevation of ATP is necessary for the membrane-dependent increase in cytosolic Ca2+, the main trigger of insulin exocytosis. Beta cells depleted of mitochondrial DNA fail to respond to glucose while still secreting insulin in response to membrane depolarisation. This cell model resembles the situation of defective insulin secretion in patients with mitochondrial diabetes. On the other hand, infants with activating mutations in the mitochondrial enzyme glutamate dehydrogenase are characterised by hyperinsulinism and hypoglycaemia. We have recently proposed that glutamate, generated by this enzyme, participates in insulin secretion as a glucose-derived metabolic messenger. In this model, glutamate acts downstream of the mitochondria by sensitising the exocytotic process to Ca2+. The evidence in favour of such a role for glutamate is discussed in the present review.


Asunto(s)
Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Mitocondrias/fisiología , Transducción de Señal/fisiología , Animales , Exocitosis/efectos de los fármacos , Humanos , Islotes Pancreáticos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Estimulación Química
20.
IUBMB Life ; 50(1): 27-31, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11087117

RESUMEN

The mitochondria play a pivotal role in regulating glucose-induced insulin secretion in the pancreatic beta cell. We have recently demonstrated that glutamate derived from mitochondria participates directly in the stimulation of insulin exocytosis. In the present study, mitochondria isolated from the beta cell line INS-1E generated glutamate when incubated with the tricarboxylic acid cycle intermediate succinate. The generation of glutamate correlated with stimulated mitochondrial activity monitored as oxygen consumption and was inhibited by the mitochondrial uncoupler carbonyl cyanide p-trifluoromethoxyphenylhydrazone. Glutamate is formed by the mitochondrial enzyme glutamate dehydrogenase from alpha-ketoglutarate. Transient overexpression of glutamate dehydrogenase in INS-1E cells resulted in potentiation of glucose-stimulated hormone secretion without affecting basal release. These results further point to glutamate as an intracellular messenger playing a key role in the control of insulin exocytosis.


Asunto(s)
Ácido Glutámico/metabolismo , Hormonas/metabolismo , Islotes Pancreáticos/metabolismo , Mitocondrias/metabolismo , Animales , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Glucosa/metabolismo , Glucosa/farmacología , Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/metabolismo , Glicerofosfatos/farmacología , Hormona de Crecimiento Humana/metabolismo , Insulina/metabolismo , Secreción de Insulina , Insulinoma , Islotes Pancreáticos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Oxígeno/metabolismo , Neoplasias Pancreáticas , Ratas , Succinatos/farmacología , Células Tumorales Cultivadas , Desacopladores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA