Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 33(4): 804-817.e5, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33321098

RESUMEN

Metabolic fuels regulate insulin secretion by generating second messengers that drive insulin granule exocytosis, but the biochemical pathways involved are incompletely understood. Here we demonstrate that stimulation of rat insulinoma cells or primary rat islets with glucose or glutamine + 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (Gln + BCH) induces reductive, "counter-clockwise" tricarboxylic acid (TCA) cycle flux of glutamine to citrate. Molecular or pharmacologic suppression of isocitrate dehydrogenase-2 (IDH2), which catalyzes reductive carboxylation of 2-ketoglutarate to isocitrate, results in impairment of glucose- and Gln + BCH-stimulated reductive TCA cycle flux, lowering of NADPH levels, and inhibition of insulin secretion. Pharmacologic suppression of IDH2 also inhibits insulin secretion in living mice. Reductive TCA cycle flux has been proposed as a mechanism for generation of biomass in cancer cells. Here we demonstrate that reductive TCA cycle flux also produces stimulus-secretion coupling factors that regulate insulin secretion, including in non-dividing cells.


Asunto(s)
Ciclo del Ácido Cítrico/fisiología , Glucosa/farmacología , Glutamina/farmacología , Secreción de Insulina/efectos de los fármacos , Animales , Células Cultivadas , Glucosa/metabolismo , Glutamina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Lipogénesis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Compuestos de Fenilurea/farmacología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Sulfonamidas/farmacología , Sumoilación/efectos de los fármacos
2.
Diabetes Obes Metab ; 19 Suppl 1: 90-94, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28880482

RESUMEN

After multiple decades of investigation, the precise mechanisms involved in fuel-stimulated insulin secretion are still being revealed. One avenue for gaining deeper knowledge is to apply emergent tools of "metabolomics," involving mass spectrometry and nuclear magnetic resonance-based profiling of islet cells in their fuel-stimulated compared with basal states. The current article summarizes recent insights gained from application of metabolomics tools to the specific process of glucose-stimulated insulin secretion, revealing 2 new mechanisms that may provide targets for improving insulin secretion in diabetes.


Asunto(s)
Investigación Biomédica/métodos , Islotes Pancreáticos/metabolismo , Metabolómica/métodos , Modelos Biológicos , Animales , Investigación Biomédica/tendencias , Exocitosis , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/enzimología , Metabolómica/tendencias , Vías Secretoras
3.
Arch Biochem Biophys ; 589: 120-30, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26116790

RESUMEN

Metabolomics, the characterization of the set of small molecules in a biological system, is advancing research in multiple areas of islet biology. Measuring a breadth of metabolites simultaneously provides a broad perspective on metabolic changes as the islets respond dynamically to metabolic fuels, hormones, or environmental stressors. As a result, metabolomics has the potential to provide new mechanistic insights into islet physiology and pathophysiology. Here we summarize advances in our understanding of islet physiology and the etiologies of type-1 and type-2 diabetes gained from metabolomics studies.


Asunto(s)
Islotes Pancreáticos/metabolismo , Metabolómica/métodos , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Islotes Pancreáticos/fisiología , Islotes Pancreáticos/fisiopatología
4.
J Clin Invest ; 125(10): 3847-60, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26389676

RESUMEN

Insulin secretion from ß cells of the pancreatic islets of Langerhans controls metabolic homeostasis and is impaired in individuals with type 2 diabetes (T2D). Increases in blood glucose trigger insulin release by closing ATP-sensitive K+ channels, depolarizing ß cells, and opening voltage-dependent Ca2+ channels to elicit insulin exocytosis. However, one or more additional pathway(s) amplify the secretory response, likely at the distal exocytotic site. The mitochondrial export of isocitrate and engagement with cytosolic isocitrate dehydrogenase (ICDc) may be one key pathway, but the mechanism linking this to insulin secretion and its role in T2D have not been defined. Here, we show that the ICDc-dependent generation of NADPH and subsequent glutathione (GSH) reduction contribute to the amplification of insulin exocytosis via sentrin/SUMO-specific protease-1 (SENP1). In human T2D and an in vitro model of human islet dysfunction, the glucose-dependent amplification of exocytosis was impaired and could be rescued by introduction of signaling intermediates from this pathway. Moreover, islet-specific Senp1 deletion in mice caused impaired glucose tolerance by reducing the amplification of insulin exocytosis. Together, our results identify a pathway that links glucose metabolism to the amplification of insulin secretion and demonstrate that restoration of this axis rescues ß cell function in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Endopeptidasas/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Isocitratos/metabolismo , Animales , Dominio Catalítico , Membrana Celular/metabolismo , Cisteína Endopeptidasas , Diabetes Mellitus Tipo 2/patología , Endopeptidasas/biosíntesis , Endopeptidasas/deficiencia , Endopeptidasas/genética , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Glucosa/farmacología , Glutatión/farmacología , Células HEK293 , Homeostasis , Humanos , Insulina/farmacología , Secreción de Insulina , Islotes Pancreáticos/fisiopatología , Isocitrato Deshidrogenasa/fisiología , Isocitratos/farmacología , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , NADP/metabolismo , Especificidad de Órganos , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Vesículas Secretoras/metabolismo , Transducción de Señal , Sumoilación
5.
Cell Rep ; 13(1): 157-167, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26411681

RESUMEN

Pancreatic islet failure, involving loss of glucose-stimulated insulin secretion (GSIS) from islet ß cells, heralds the onset of type 2 diabetes (T2D). To search for mediators of GSIS, we performed metabolomics profiling of the insulinoma cell line 832/13 and uncovered significant glucose-induced changes in purine pathway intermediates, including a decrease in inosine monophosphate (IMP) and an increase in adenylosuccinate (S-AMP), suggesting a regulatory role for the enzyme that links the two metabolites, adenylosuccinate synthase (ADSS). Inhibition of ADSS or a more proximal enzyme in the S-AMP biosynthesis pathway, adenylosuccinate lyase, lowers S-AMP levels and impairs GSIS. Addition of S-AMP to the interior of patch-clamped human ß cells amplifies exocytosis, an effect dependent upon expression of sentrin/SUMO-specific protease 1 (SENP1). S-AMP also overcomes the defect in glucose-induced exocytosis in ß cells from a human donor with T2D. S-AMP is, thus, an insulin secretagogue capable of reversing ß cell dysfunction in T2D.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología , Adenilosuccinato Liasa/antagonistas & inhibidores , Adenilosuccinato Liasa/genética , Adenilosuccinato Liasa/metabolismo , Adenilosuccinato Sintasa/antagonistas & inhibidores , Adenilosuccinato Sintasa/genética , Adenilosuccinato Sintasa/metabolismo , Animales , Línea Celular Tumoral , Cisteína Endopeptidasas , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Endopeptidasas/genética , Endopeptidasas/metabolismo , Inhibidores Enzimáticos/farmacología , Exocitosis/efectos de los fármacos , Regulación de la Expresión Génica , Glucosa/metabolismo , Guanina/farmacología , Humanos , Inosina Monofosfato/metabolismo , Insulina/biosíntesis , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Metaboloma/genética , Ácido Micofenólico/farmacología , Técnicas de Placa-Clamp , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Transducción de Señal
6.
Proc Natl Acad Sci U S A ; 111(14): 5242-7, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706823

RESUMEN

Loss of functional ß-cell mass is a hallmark of type 1 and type 2 diabetes, and methods for restoring these cells are needed. We have previously reported that overexpression of the homeodomain transcription factor NK6 homeobox 1 (Nkx6.1) in rat pancreatic islets induces ß-cell proliferation and enhances glucose-stimulated insulin secretion, but the pathway by which Nkx6.1 activates ß-cell expansion has not been defined. Here, we demonstrate that Nkx6.1 induces expression of the nuclear receptor subfamily 4, group A, members 1 and 3 (Nr4a1 and Nr4a3) orphan nuclear receptors, and that these factors are both necessary and sufficient for Nkx6.1-mediated ß-cell proliferation. Consistent with this finding, global knockout of Nr4a1 results in a decrease in ß-cell area in neonatal and young mice. Overexpression of Nkx6.1 and the Nr4a receptors results in increased expression of key cell cycle inducers E2F transcription factor 1 and cyclin E1. Furthermore, Nkx6.1 and Nr4a receptors induce components of the anaphase-promoting complex, including ubiquitin-conjugating enzyme E2C, resulting in degradation of the cell cycle inhibitor p21. These studies identify a unique bipartite pathway for activation of ß-cell proliferation, suggesting several unique targets for expansion of functional ß-cell mass.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/fisiología , Proteínas de Homeodominio/fisiología , Islotes Pancreáticos/citología , Proteínas del Tejido Nervioso/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Animales , Animales Recién Nacidos , Inmunoprecipitación de Cromatina , Proteínas de Homeodominio/genética , Masculino , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Ratas , Ratas Wistar , Enzimas Ubiquitina-Conjugadoras/metabolismo , Regulación hacia Arriba
7.
J Biol Chem ; 288(32): 23128-40, 2013 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-23788641

RESUMEN

Recent studies have shown that the pyruvate-isocitrate cycling pathway, involving the mitochondrial citrate/isocitrate carrier and the cytosolic NADP-dependent isocitrate dehydrogenase (ICDc), is involved in control of glucose-stimulated insulin secretion (GSIS). Here we demonstrate that pyruvate-isocitrate cycling regulates expression of the voltage-gated potassium channel family member Kv2.2 in islet ß-cells. siRNA-mediated suppression of ICDc, citrate/isocitrate carrier, or Kv2.2 expression impaired GSIS, and the effect of ICDc knockdown was rescued by re-expression of Kv2.2. Moreover, chronic exposure of ß-cells to elevated fatty acids, which impairs GSIS, resulted in decreased expression of Kv2.2. Surprisingly, knockdown of ICDc or Kv2.2 increased rather than decreased outward K(+) current in the 832/13 ß-cell line. Immunoprecipitation studies demonstrated interaction of Kv2.1 and Kv2.2, and co-overexpression of the two channels reduced outward K(+) current compared with overexpression of Kv2.1 alone. Also, siRNA-mediated knockdown of ICDc enhanced the suppressive effect of the Kv2.1-selective inhibitor stromatoxin1 on K(+) currents. Our data support a model in which a key function of the pyruvate-isocitrate cycle is to maintain levels of Kv2.2 expression sufficient to allow it to serve as a negative regulator of Kv channel activity.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Isocitratos/metabolismo , Ácido Pirúvico/metabolismo , Canales de Potasio Shab/biosíntesis , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/genética , Secreción de Insulina , Células Secretoras de Insulina/citología , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Masculino , Modelos Biológicos , Péptidos/farmacología , Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Canales de Potasio Shab/antagonistas & inhibidores , Canales de Potasio Shab/genética , Venenos de Araña/farmacología
8.
J Pharmacol Exp Ther ; 344(2): 407-16, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23161216

RESUMEN

The voltage-gated potassium channels Kv2.1 and Kv2.2 are highly expressed in pancreatic islets, yet their contribution to islet hormone secretion is not fully understood. Here we investigate the role of Kv2 channels in pancreatic islets using a combination of genetic and pharmacologic approaches. Pancreatic ß-cells from Kv2.1(-/-) mice possess reduced Kv current and display greater glucose-stimulated insulin secretion (GSIS) relative to WT ß-cells. Inhibition of Kv2.x channels with selective peptidyl [guangxitoxin-1E (GxTX-1E)] or small molecule (RY796) inhibitors enhances GSIS in isolated wild-type (WT) mouse and human islets, but not in islets from Kv2.1(-/-) mice. However, in WT mice neither inhibitor improved glucose tolerance in vivo. GxTX-1E and RY796 enhanced somatostatin release in isolated human and mouse islets and in situ perfused pancreata from WT and Kv2.1(-/-) mice. Kv2.2 silencing in mouse islets by adenovirus-small hairpin RNA (shRNA) specifically enhanced islet somatostatin, but not insulin, secretion. In mice lacking somatostatin receptor 5, GxTX-1E stimulated insulin secretion and improved glucose tolerance. Collectively, these data show that Kv2.1 regulates insulin secretion in ß-cells and Kv2.2 modulates somatostatin release in δ-cells. Development of selective Kv2.1 inhibitors without cross inhibition of Kv2.2 may provide new avenues to promote GSIS for the treatment of type 2 diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canales de Potasio Shab/metabolismo , Somatostatina/metabolismo , Adulto , Animales , Proteínas de Artrópodos , Benzamidas/farmacología , Células Cultivadas , Fenómenos Electrofisiológicos , Femenino , Glucosa/farmacología , Humanos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Técnicas de Placa-Clamp , Péptidos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Unión Proteica , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Canales de Potasio Shab/antagonistas & inhibidores , Canales de Potasio Shab/genética , Venenos de Araña/farmacología , Adulto Joven
9.
J Biol Chem ; 285(22): 16530-7, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20356834

RESUMEN

Glucose-stimulated insulin secretion from pancreatic islet beta-cells is dependent in part on pyruvate cycling through the pyruvate/isocitrate pathway, which generates cytosolic alpha-ketoglutarate, also known as 2-oxoglutarate (2OG). Here, we have investigated if mitochondrial transport of 2OG through the 2-oxoglutarate carrier (OGC) participates in control of nutrient-stimulated insulin secretion. Suppression of OGC in clonal pancreatic beta-cells (832/13 cells) and isolated rat islets by adenovirus-mediated delivery of small interfering RNA significantly decreased glucose-stimulated insulin secretion. OGC suppression also reduced insulin secretion in response to glutamine plus the glutamate dehydrogenase activator 2-amino-2-norbornane carboxylic acid. Nutrient-stimulated increases in glucose usage, glucose oxidation, glutamine oxidation, or ATP:ADP ratio were not affected by OGC knockdown, whereas suppression of OGC resulted in a significant decrease in the NADPH:NADP(+) ratio during stimulation with glucose but not glutamine + 2-amino-2-norbornane carboxylic acid. Finally, OGC suppression reduced insulin secretion in response to a membrane-permeant 2OG analog, dimethyl-2OG. These data reveal that the OGC is part of a mechanism of fuel-stimulated insulin secretion that is common to glucose, amino acid, and organic acid secretagogues, involving flux through the pyruvate/isocitrate cycling pathway. Although the components of this pathway must remain intact for appropriate stimulus-secretion coupling, production of NADPH does not appear to be the universal second messenger signal generated by these reactions.


Asunto(s)
Glucosa/metabolismo , Glutamina/metabolismo , Insulina/metabolismo , Ácidos Cetoglutáricos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Mitocondrias/metabolismo , Animales , Citosol/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Modelos Biológicos , NADP/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Am J Physiol Endocrinol Metab ; 295(6): E1287-97, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18728221

RESUMEN

Glucose-stimulated insulin secretion (GSIS) is central to normal control of metabolic fuel homeostasis, and its impairment is a key element of beta-cell failure in type 2 diabetes. Glucose exerts its effects on insulin secretion via its metabolism in beta-cells to generate stimulus/secretion coupling factors, including a rise in the ATP/ADP ratio, which serves to suppress ATP-sensitive K(+) (K(ATP)) channels and activate voltage-gated Ca(2+) channels, leading to stimulation of insulin granule exocytosis. Whereas this K(ATP) channel-dependent mechanism of GSIS has been broadly accepted for more than 30 years, it has become increasingly apparent that it does not fully describe the effects of glucose on insulin secretion. More recent studies have demonstrated an important role for cyclic pathways of pyruvate metabolism in control of insulin secretion. Three cycles occur in islet beta-cells: the pyruvate/malate, pyruvate/citrate, and pyruvate/isocitrate cycles. This review discusses recent work on the role of each of these pathways in control of insulin secretion and builds a case for the particular relevance of byproducts of the pyruvate/isocitrate cycle, NADPH and alpha-ketoglutarate, in control of GSIS.


Asunto(s)
Glucosa/farmacología , Insulina/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Animales , Ácido Cítrico/metabolismo , Exocitosis/efectos de los fármacos , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Redes y Vías Metabólicas/fisiología , Modelos Biológicos , Canales de Potasio/fisiología , Ácido Pirúvico/metabolismo
11.
J Biol Chem ; 283(43): 28909-17, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18755687

RESUMEN

We have previously demonstrated a role for pyruvate cycling in glucose-stimulated insulin secretion (GSIS). Some of the possible pyruvate cycling pathways are completed by conversion of malate to pyruvate by malic enzyme. Using INS-1-derived 832/13 cells, it has recently been shown by other laboratories that NADP-dependent cytosolic malic enzyme (MEc), but not NAD-dependent mitochondrial malic enzyme (MEm), regulates GSIS. In the current study, we show that small interfering RNA-mediated suppression of either MEm or MEc results in decreased GSIS in both 832/13 cells and a new and more glucose- and incretin-responsive INS-1-derived cell line, 832/3. The effect of MEm to suppress GSIS in these cell lines was linked to a substantial decrease in cell growth, whereas MEc suppression resulted in decreased NADPH, shown previously to be correlated with GSIS. However, adenovirus-mediated delivery of small interfering RNAs specific to MEc and MEm to isolated rat islets, while leading to effective suppression of the targets transcripts, had no effect on GSIS. Furthermore, islets isolated from MEc-null MOD1(-/-) mice exhibit normal glucose- and potassium-stimulated insulin secretion. These results indicate that pyruvate-malate cycling does not control GSIS in primary rodent islets.


Asunto(s)
Citosol/metabolismo , Glucosa/química , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Malato Deshidrogenasa/química , Mitocondrias/metabolismo , Animales , Silenciador del Gen , Secreción de Insulina , Masculino , Ratones , Modelos Biológicos , Isoformas de Proteínas , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley
12.
J Biol Chem ; 281(47): 35624-32, 2006 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-17001083

RESUMEN

Glucose-stimulated insulin secretion (GSIS) is mediated in part by glucose metabolism-driven increases in ATP/ADP ratio, but by-products of mitochondrial glucose metabolism also play an important role. Here we investigate the role of the mitochondrial citrate/isocitrate carrier (CIC) in regulation of GSIS. Inhibition of CIC activity in INS-1-derived 832/13 cells or primary rat islets by the substrate analogue 1,2,3-benzenetricarboxylate (BTC) resulted in potent inhibition of GSIS, involving both first and second phase secretion. A recombinant adenovirus containing a CIC-specific siRNA (Ad-siCIC) dose-dependently reduced CIC expression in 832/13 cells and caused parallel inhibitory effects on citrate accumulation in the cytosol. Ad-siCIC treatment did not affect glucose utilization, glucose oxidation, or ATP/ADP ratio but did inhibit glucose incorporation into fatty acids and glucose-induced increases in NADPH/NADP+ ratio relative to cells treated with a control siRNA virus (Ad-siControl). Ad-siCIC also inhibited GSIS in 832/13 cells, whereas overexpression of CIC enhanced GSIS and raised cytosolic citrate levels. In normal rat islets, Ad-siCIC treatment also suppressed CIC mRNA levels and inhibited GSIS. We conclude that export of citrate and/or isocitrate from the mitochondria to the cytosol is an important step in control of GSIS.


Asunto(s)
Antiportadores/química , Antiportadores/fisiología , Proteínas Portadoras/fisiología , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas de Transporte de Membrana/fisiología , Mitocondrias/metabolismo , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/fisiología , Adenoviridae/metabolismo , Animales , Derivados del Benceno/farmacología , Transporte Biológico , Proteínas Portadoras/química , Citosol/metabolismo , Relación Dosis-Respuesta a Droga , Secreción de Insulina , Membranas Intracelulares/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratas , Ácidos Tricarboxílicos/farmacología
13.
J Biol Chem ; 281(41): 30593-602, 2006 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-16912049

RESUMEN

Glucose-stimulated insulin secretion (GSIS) from pancreatic islet beta-cells is central to control of mammalian fuel homeostasis. Glucose metabolism mediates GSIS in part via ATP-regulated K+ (KATP) channels, but multiple lines of evidence suggest participation of other signals. Here we investigated the role of cytosolic NADP-dependent isocitrate dehydrogenase (ICDc) in control of GSIS in beta-cells. Delivery of small interfering RNAs specific for ICDc caused impairment of GSIS in two independent robustly glucose-responsive rat insulinoma (INS-1-derived) cell lines and in primary rat islets. Suppression of ICDc also attenuated the glucose-induced increments in pyruvate cycling activity and in NADPH levels, a predicted by-product of pyruvate cycling pathways, as well as the total cellular NADP(H) content. Metabolic profiling of eight organic acids in cell extracts revealed that suppression of ICDc caused increases in lactate production in both INS-1-derived cell lines and primary islets, consistent with the attenuation of pyruvate cycling, with no significant changes in other intermediates. Based on these studies, we propose that a pyruvate cycling pathway involving ICDc plays an important role in control of GSIS.


Asunto(s)
Citosol/enzimología , Glucosa/metabolismo , Insulina/metabolismo , Isocitrato Deshidrogenasa/química , Ácido Pirúvico/química , Animales , Secreción de Insulina , Islotes Pancreáticos/citología , Lactatos/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Modelos Biológicos , Ratas , Ratas Sprague-Dawley
14.
J Biol Chem ; 281(31): 22342-22351, 2006 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-16740637

RESUMEN

We have previously reported that glucose-stimulated insulin secretion (GSIS) is tightly correlated with pyruvate carboxylase (PC)-catalyzed anaplerotic flux into the tricarboxylic acid cycle and stimulation of pyruvate cycling activity. To further evaluate the role of PC in beta-cell function, we constructed a recombinant adenovirus containing a small interfering RNA (siRNA) specific to PC (Ad-siPC). Ad-siPC reduced PC mRNA levels by 83 and 64% and PC protein by 56 and 35% in INS-1-derived 832/13 cells and primary rat islets, respectively. Surprisingly, this manipulation did not impair GSIS in rat islets. In Ad-siPC-treated 832/13 cells, GSIS was slightly increased, whereas glycolytic rate and glucose oxidation were unaffected. Flux through PC at high glucose was decreased by only 20%, suggesting an increase in PC-specific activity. Acetyl carnitine, a surrogate for acetyl-CoA, an allosteric activator of PC, was increased by 36% in Ad-siPC-treated cells, suggesting a mechanism by which PC enzymatic activity is maintained with suppressed PC protein levels. In addition, the NADPH:NADP ratio, a proposed coupling factor for GSIS, was unaffected in Ad-siPC-treated cells. We conclude that beta-cells activate compensatory mechanisms in response to suppression of PC expression that prevent impairment of anaplerosis, pyruvate cycling, NAPDH production, and GSIS.


Asunto(s)
Regulación Alostérica , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Piruvato Carboxilasa/fisiología , Acetilcarnitina/análisis , Animales , Línea Celular , Secreción de Insulina , Islotes Pancreáticos , NADP/biosíntesis , Piruvato Carboxilasa/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Ratas
15.
J Biol Chem ; 279(26): 27263-71, 2004 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-15073188

RESUMEN

Hyperlipidemia appears to play an integral role in loss of glucose-stimulated insulin secretion (GSIS) in type 2 diabetes. This impairment can be simulated in vitro by chronic culture of 832/13 insulinoma cells with high concentrations of free fatty acids, or by study of lipid-laden islets from Zucker diabetic fatty rats. Here we show that impaired GSIS is not a simple result of saturation of lipid storage pathways, as adenovirus-mediated overexpression of a cytosolically localized variant of malonyl-CoA decarboxylase in either cellular model results in dramatic lowering of cellular triglyceride stores but no improvement in GSIS. Instead, the glucose-induced increment in "pyruvate cycling" activity (pyruvate exchange with tricarboxylic acid cycle intermediates measured by (13)C NMR), previously shown to play an important role in GSIS, is completely ablated in concert with profound suppression of GSIS in lipid-cultured 832/13 cells, whereas glucose oxidation is unaffected. Moreover, GSIS is partially restored in both lipid-cultured 832/13 cells and islets from Zucker diabetic fatty rats by addition of a membrane permeant ester of a pyruvate cycling intermediate (dimethyl malate). We conclude that chronic exposure of islet beta-cells to fatty acids grossly alters a mitochondrial pathway of pyruvate metabolism that is important for normal GSIS.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Malatos/farmacología , Ácido Oléico/metabolismo , Palmitatos/metabolismo , Adenoviridae/genética , Animales , Carboxiliasas/genética , Carboxiliasas/metabolismo , Línea Celular , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Malatos/química , Masculino , Ácido Oléico/toxicidad , Consumo de Oxígeno , Palmitatos/toxicidad , Ácido Pirúvico/metabolismo , Ratas , Ratas Zucker , Transducción Genética , Triglicéridos/deficiencia , Triglicéridos/metabolismo
16.
Proc Natl Acad Sci U S A ; 99(5): 2708-13, 2002 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-11880625

RESUMEN

Cellular metabolism of glucose is required for stimulation of insulin secretion from pancreatic beta cells, but the precise metabolic coupling factors involved in this process are not known. In an effort to better understand mechanisms of fuel-mediated insulin secretion, we have adapted 13C NMR and isotopomer methods to measure influx of metabolic fuels into the tricarboxylic acid (TCA) cycle in insulinoma cells. Mitochondrial metabolism of [U-13C3]pyruvate, derived from [U-13C6]glucose, was compared in four clonal rat insulinoma cell 1-derived cell lines with varying degrees of glucose responsiveness. A 13C isotopomer analysis of glutamate isolated from these cells showed that the fraction of acetyl-CoA derived from [U-13C6]glucose was the same in all four cell lines (44 +/- 5%, 70 +/- 3%, and 84 +/- 4% with 3, 6, or 12 mM glucose, respectively). The 13C NMR spectra also demonstrated the existence of two compartmental pools of pyruvate, one that exchanges with TCA cycle intermediates and a second pool derived from [U-13C6]glucose that feeds acetyl-CoA into the TCA cycle. The 13C NMR spectra were consistent with a metabolic model where the two pyruvate pools do not randomly mix. Flux between the mitochondrial intermediates and the first pool of pyruvate (pyruvate cycling) varied in proportion to glucose responsiveness in the four cell lines. Furthermore, stimulation of pyruvate cycling with dimethylmalate or its inhibition with phenylacetic acid led to proportional changes in insulin secretion. These findings indicate that exchange of pyruvate with TCA cycle intermediates, rather than oxidation of pyruvate via acetyl-CoA, correlates with glucose-stimulated insulin secretion.


Asunto(s)
Ciclo del Ácido Cítrico/fisiología , Glucosa/metabolismo , Insulina/metabolismo , Piruvato Carboxilasa/metabolismo , Ácido Pirúvico/metabolismo , Animales , Isótopos de Carbono , Glucosa/farmacología , Secreción de Insulina , Marcaje Isotópico , Espectroscopía de Resonancia Magnética/métodos , Maleatos/farmacología , Modelos Biológicos , Fenilacetatos/farmacología , Piruvato Carboxilasa/antagonistas & inhibidores , Ratas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...