Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetologia ; 67(3): 528-546, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38127123

RESUMEN

AIMS/HYPOTHESIS: Diabetes mellitus is associated with impaired insulin secretion, often aggravated by oversecretion of glucagon. Therapeutic interventions should ideally correct both defects. Glucagon-like peptide 1 (GLP-1) has this capability but exactly how it exerts its glucagonostatic effect remains obscure. Following its release GLP-1 is rapidly degraded from GLP-1(7-36) to GLP-1(9-36). We hypothesised that the metabolite GLP-1(9-36) (previously believed to be biologically inactive) exerts a direct inhibitory effect on glucagon secretion and that this mechanism becomes impaired in diabetes. METHODS: We used a combination of glucagon secretion measurements in mouse and human islets (including islets from donors with type 2 diabetes), total internal reflection fluorescence microscopy imaging of secretory granule dynamics, recordings of cytoplasmic Ca2+ and measurements of protein kinase A activity, immunocytochemistry, in vivo physiology and GTP-binding protein dissociation studies to explore how GLP-1 exerts its inhibitory effect on glucagon secretion and the role of the metabolite GLP-1(9-36). RESULTS: GLP-1(7-36) inhibited glucagon secretion in isolated islets with an IC50 of 2.5 pmol/l. The effect was particularly strong at low glucose concentrations. The degradation product GLP-1(9-36) shared this capacity. GLP-1(9-36) retained its glucagonostatic effects after genetic/pharmacological inactivation of the GLP-1 receptor. GLP-1(9-36) also potently inhibited glucagon secretion evoked by ß-adrenergic stimulation, amino acids and membrane depolarisation. In islet alpha cells, GLP-1(9-36) led to inhibition of Ca2+ entry via voltage-gated Ca2+ channels sensitive to ω-agatoxin, with consequential pertussis-toxin-sensitive depletion of the docked pool of secretory granules, effects that were prevented by the glucagon receptor antagonists REMD2.59 and L-168049. The capacity of GLP-1(9-36) to inhibit glucagon secretion and reduce the number of docked granules was lost in alpha cells from human donors with type 2 diabetes. In vivo, high exogenous concentrations of GLP-1(9-36) (>100 pmol/l) resulted in a small (30%) lowering of circulating glucagon during insulin-induced hypoglycaemia. This effect was abolished by REMD2.59, which promptly increased circulating glucagon by >225% (adjusted for the change in plasma glucose) without affecting pancreatic glucagon content. CONCLUSIONS/INTERPRETATION: We conclude that the GLP-1 metabolite GLP-1(9-36) is a systemic inhibitor of glucagon secretion. We propose that the increase in circulating glucagon observed following genetic/pharmacological inactivation of glucagon signalling in mice and in people with type 2 diabetes reflects the removal of GLP-1(9-36)'s glucagonostatic action.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hipoglucemia , Islotes Pancreáticos , Fragmentos de Péptidos , Humanos , Glucagón/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Islotes Pancreáticos/metabolismo , Hipoglucemia/metabolismo , Insulina/metabolismo
2.
Diabetes ; 72(10): 1446-1459, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37494670

RESUMEN

Whole-body glucose homeostasis is coordinated through secretion of glucagon and insulin from pancreatic islets. When glucose is low, glucagon is released from α-cells to stimulate hepatic glucose production. However, the mechanisms that regulate glucagon secretion from pancreatic α-cells remain unclear. Here we show that in α-cells, the interaction between fatty acid oxidation and glucose metabolism controls glucagon secretion. The glucose-dependent inhibition of glucagon secretion relies on pyruvate dehydrogenase and carnitine palmitoyl transferase 1a activity and lowering of mitochondrial fatty acid oxidation by increases in glucose. This results in reduced intracellular ATP and leads to membrane repolarization and inhibition of glucagon secretion. These findings provide a new framework for the metabolic regulation of the α-cell, where regulation of fatty acid oxidation by glucose accounts for the stimulation and inhibition of glucagon secretion. ARTICLE HIGHLIGHTS: It has become clear that dysregulation of glucagon secretion and α-cell function plays an important role in the development of diabetes, but we do not know how glucagon secretion is regulated. Here we asked whether glucose inhibits fatty acid oxidation in α-cells to regulate glucagon secretion. We found that fatty acid oxidation is required for the inhibitory effects of glucose on glucagon secretion through reductions in ATP. These findings provide a new framework for the regulation of glucagon secretion by glucose.


Asunto(s)
Células Secretoras de Glucagón , Islotes Pancreáticos , Adenosina Trifosfato/metabolismo , Glucemia/metabolismo , Ácidos Grasos/metabolismo , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucosa/farmacología , Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Humanos , Animales , Ratones
3.
Nat Metab ; 2(1): 32-40, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31993555

RESUMEN

Pancreatic islets are complex micro-organs consisting of at least three different cell types: glucagon-secreting α-, insulin-producing ß- and somatostatin-releasing δ-cells1. Somatostatin is a powerful paracrine inhibitor of insulin and glucagon secretion2. In diabetes, increased somatostatinergic signalling leads to defective counter-regulatory glucagon secretion3. This increases the risk of severe hypoglycaemia, a dangerous complication of insulin therapy4. The regulation of somatostatin secretion involves both intrinsic and paracrine mechanisms5 but their relative contributions and whether they interact remains unclear. Here we show that dapagliflozin-sensitive glucose- and insulin-dependent sodium uptake stimulates somatostatin secretion by elevating the cytoplasmic Na+ concentration ([Na+]i) and promoting intracellular Ca2+-induced Ca2+ release (CICR). This mechanism also becomes activated when [Na+]i is elevated following the inhibition of the plasmalemmal Na+-K+ pump by reductions of the extracellular K+ concentration emulating those produced by exogenous insulin in vivo 6. Islets from some donors with type-2 diabetes hypersecrete somatostatin, leading to suppression of glucagon secretion that can be alleviated by a somatostatin receptor antagonist. Our data highlight the role of Na+ as an intracellular second messenger, illustrate the significance of the intraislet paracrine network and provide a mechanistic framework for pharmacological correction of the hormone secretion defects associated with diabetes that selectively target the δ-cells.


Asunto(s)
Calcio/metabolismo , Sodio/metabolismo , Células Secretoras de Somatostatina/metabolismo , Somatostatina/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Hipoglucemia/metabolismo , Insulina/metabolismo , Ratones
4.
Nat Commun ; 10(1): 139, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30635569

RESUMEN

Hypoglycaemia (low plasma glucose) is a serious and potentially fatal complication of insulin-treated diabetes. In healthy individuals, hypoglycaemia triggers glucagon secretion, which restores normal plasma glucose levels by stimulation of hepatic glucose production. This counterregulatory mechanism is impaired in diabetes. Here we show in mice that therapeutic concentrations of insulin inhibit glucagon secretion by an indirect (paracrine) mechanism mediated by stimulation of intra-islet somatostatin release. Insulin's capacity to inhibit glucagon secretion is lost following genetic ablation of insulin receptors in the somatostatin-secreting δ-cells, when insulin-induced somatostatin secretion is suppressed by dapagliflozin (an inhibitor of sodium-glucose co-tranporter-2; SGLT2) or when the action of secreted somatostatin is prevented by somatostatin receptor (SSTR) antagonists. Administration of these compounds in vivo antagonises insulin's hypoglycaemic effect. We extend these data to isolated human islets. We propose that SSTR or SGLT2 antagonists should be considered as adjuncts to insulin in diabetes therapy.


Asunto(s)
Diabetes Mellitus/patología , Glucagón/metabolismo , Hipoglucemia/patología , Insulina/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Somatostatina/metabolismo , Animales , Compuestos de Bencidrilo/farmacología , Glucemia/análisis , Diabetes Mellitus/tratamiento farmacológico , Femenino , Células Secretoras de Glucagón/efectos de los fármacos , Glucósidos/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Insulina/genética , Receptores de Somatostatina/antagonistas & inhibidores , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología
5.
Cell Metab ; 29(2): 430-442.e4, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30415925

RESUMEN

Diabetes is a bihormonal disorder resulting from combined insulin and glucagon secretion defects. Mice lacking fumarase (Fh1) in their ß cells (Fh1ßKO mice) develop progressive hyperglycemia and dysregulated glucagon secretion similar to that seen in diabetic patients (too much at high glucose and too little at low glucose). The glucagon secretion defects are corrected by low concentrations of tolbutamide and prevented by the sodium-glucose transport (SGLT) inhibitor phlorizin. These data link hyperglycemia, intracellular Na+ accumulation, and acidification to impaired mitochondrial metabolism, reduced ATP production, and dysregulated glucagon secretion. Protein succination, reflecting reduced activity of fumarase, is observed in α cells from hyperglycemic Fh1ßKO and ß-V59M gain-of-function KATP channel mice, diabetic Goto-Kakizaki rats, and patients with type 2 diabetes. Succination is also observed in renal tubular cells and cardiomyocytes from hyperglycemic Fh1ßKO mice, suggesting that the model can be extended to other SGLT-expressing cells and may explain part of the spectrum of diabetic complications.


Asunto(s)
Adenosina Trifosfato/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Hiperglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Línea Celular , Células Secretoras de Glucagón/citología , Humanos , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Canales de Potasio/metabolismo , Ratas , Ratas Wistar , Sodio/metabolismo
6.
Sci Rep ; 8(1): 16994, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451893

RESUMEN

Limited access to human islets has prompted the development of human beta cell models. The human beta cell lines EndoC-ßH1 and EndoC-ßH2 are increasingly used by the research community. However, little is known of their electrophysiological and secretory properties. Here, we monitored parameters that constitute the glucose-triggering pathway of insulin release. Both cell lines respond to glucose (6 and 20 mM) with 2- to 3-fold stimulation of insulin secretion which correlated with an elevation of [Ca2+]i, membrane depolarisation and increased action potential firing. Similar to human primary beta cells, KATP channel activity is low at 1 mM glucose and is further reduced upon increasing glucose concentration; an effect that was mimicked by the KATP channel blocker tolbutamide. The upstroke of the action potentials reflects the activation of Ca2+ channels with some small contribution of TTX-sensitive Na+ channels. The repolarisation involves activation of voltage-gated Kv2.2 channels and large-conductance Ca2+-activated K+ channels. Exocytosis presented a similar kinetics to human primary beta cells. The ultrastructure of these cells shows insulin vesicles composed of an electron-dense core surrounded by a thin clear halo. We conclude that the EndoC-ßH1 and -ßH2 cells share many features of primary human ß-cells and thus represent a useful experimental model.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Exocitosis , Glucosa/farmacología , Secreción de Insulina , Células Secretoras de Insulina/fisiología , Insulina/metabolismo , Células Cultivadas , Fenómenos Electrofisiológicos , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Edulcorantes/farmacología
7.
Cell Rep ; 23(11): 3300-3311, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29898400

RESUMEN

Glucagon, the principal hyperglycemic hormone, is secreted from pancreatic islet α cells as part of the counter-regulatory response to hypoglycemia. Hence, secretory output from α cells is under high demand in conditions of low glucose supply. Many tissues oxidize fat as an alternate energy substrate. Here, we show that glucagon secretion in low glucose conditions is maintained by fatty acid metabolism in both mouse and human islets, and that inhibiting this metabolic pathway profoundly decreases glucagon output by depolarizing α cell membrane potential and decreasing action potential amplitude. We demonstrate, by using experimental and computational approaches, that this is not mediated by the KATP channel, but instead due to reduced operation of the Na+-K+ pump. These data suggest that counter-regulatory secretion of glucagon is driven by fatty acid metabolism, and that the Na+-K+ pump is an important ATP-dependent regulator of α cell function.


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Glucagón/metabolismo , Islotes Pancreáticos/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Glucemia/análisis , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/genética , Ácidos Grasos/química , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Canales KATP/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Redes y Vías Metabólicas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
8.
J Physiol ; 596(9): 1601-1626, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29441586

RESUMEN

KEY POINTS: Na+ current inactivation is biphasic in insulin-secreting cells, proceeding with two voltage dependences that are half-maximal at ∼-100 mV and -60 mV. Inactivation of voltage-gated Na+ (NaV ) channels occurs at ∼30 mV more negative voltages in insulin-secreting Ins1 and primary ß-cells than in HEK, CHO or glucagon-secreting αTC1-6 cells. The difference in inactivation between Ins1 and non-ß-cells persists in the inside-out patch configuration, discounting an involvement of a diffusible factor. In Ins1 cells and primary ß-cells, but not in HEK cells, inactivation of a single NaV subtype is biphasic and follows two voltage dependences separated by 30-40 mV. We propose that NaV channels adopt different inactivation behaviours depending on the local membrane environment. ABSTRACT: Pancreatic ß-cells are equipped with voltage-gated Na+ channels that undergo biphasic voltage-dependent steady-state inactivation. A small Na+ current component (10-15%) inactivates over physiological membrane potentials and contributes to action potential firing. However, the major Na+ channel component is completely inactivated at -90 to -80 mV and is therefore inactive in the ß-cell. It has been proposed that the biphasic inactivation reflects the contribution of different NaV α-subunits. We tested this possibility by expression of TTX-resistant variants of the NaV subunits found in ß-cells (NaV 1.3, NaV 1.6 and NaV 1.7) in insulin-secreting Ins1 cells and in non-ß-cells (including HEK and CHO cells). We found that all NaV subunits inactivated at 20-30 mV more negative membrane potentials in Ins1 cells than in HEK or CHO cells. The more negative inactivation in Ins1 cells does not involve a diffusible intracellular factor because the difference between Ins1 and CHO persisted after excision of the membrane. NaV 1.7 inactivated at 15--20 mV more negative membrane potentials than NaV 1.3 and NaV 1.6 in Ins1 cells but this small difference is insufficient to solely explain the biphasic inactivation in Ins1 cells. In Ins1 cells, but never in the other cell types, widely different components of NaV inactivation (separated by 30 mV) were also observed following expression of a single type of NaV α-subunit. The more positive component exhibited a voltage dependence of inactivation similar to that found in HEK and CHO cells. We propose that biphasic NaV inactivation in insulin-secreting cells reflects insertion of channels in membrane domains that differ with regard to lipid and/or membrane protein composition.


Asunto(s)
Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Canal de Sodio Activado por Voltaje NAV1.3/química , Canal de Sodio Activado por Voltaje NAV1.6/química , Canal de Sodio Activado por Voltaje NAV1.7/química , Bloqueadores de los Canales de Sodio/farmacología , Potenciales de Acción , Secuencia de Aminoácidos , Animales , Cricetinae , Cricetulus , Fenómenos Electrofisiológicos , Células HEK293 , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/tratamiento farmacológico , Insulinoma/patología , Potenciales de la Membrana , Ratones , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.3/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Ratas , Homología de Secuencia , Sodio/metabolismo
9.
Cell Rep ; 20(13): 3135-3148, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-28954230

RESUMEN

We explored the role of the Krebs cycle enzyme fumarate hydratase (FH) in glucose-stimulated insulin secretion (GSIS). Mice lacking Fh1 in pancreatic ß cells (Fh1ßKO mice) appear normal for 6-8 weeks but then develop progressive glucose intolerance and diabetes. Glucose tolerance is rescued by expression of mitochondrial or cytosolic FH but not by deletion of Hif1α or Nrf2. Progressive hyperglycemia in Fh1ßKO mice led to dysregulated metabolism in ß cells, a decrease in glucose-induced ATP production, electrical activity, cytoplasmic [Ca2+]i elevation, and GSIS. Fh1 loss resulted in elevated intracellular fumarate, promoting succination of critical cysteines in GAPDH, GMPR, and PARK 7/DJ-1 and cytoplasmic acidification. Intracellular fumarate levels were increased in islets exposed to high glucose and in islets from human donors with type 2 diabetes (T2D). The impaired GSIS in islets from diabetic Fh1ßKO mice was ameliorated after culture under normoglycemic conditions. These studies highlight the role of FH and dysregulated mitochondrial metabolism in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Fumarato Hidratasa/deficiencia , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Ratones
10.
Cell Rep ; 19(10): 2088-2101, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28591580

RESUMEN

APPL1- and RAB5-positive signaling endosomes play a crucial role in the activation of AKT in response to extracellular stimuli. Myosin VI (MYO6) and two of its cargo adaptor proteins, GIPC and TOM1/TOM1L2, localize to these peripheral endosomes and mediate endosome association with cortical actin filaments. Loss of MYO6 leads to the displacement of these endosomes from the cell cortex and accumulation in the perinuclear space. Depletion of this myosin not only affects endosome positioning, but also induces actin and lipid remodeling consistent with endosome maturation, including accumulation of F-actin and the endosomal lipid PI(3)P. These processes acutely perturb endosome function, as both AKT phosphorylation and RAC-dependent membrane ruffling were markedly reduced by depletion of either APPL1 or MYO6. These results place MYO6 and its binding partners at a central nexus in cellular signaling linking actin dynamics at the cell surface and endosomal signaling in the cell cortex.


Asunto(s)
Actinas/metabolismo , Endosomas/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Actinas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Endosomas/genética , Activación Enzimática , Ratones , Cadenas Pesadas de Miosina/genética , Proteínas Proto-Oncogénicas c-akt/genética
11.
J Clin Invest ; 127(6): 2353-2364, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28481223

RESUMEN

Loss of first-phase insulin secretion is an early sign of developing type 2 diabetes (T2D). Ca2+ entry through voltage-gated L-type Ca2+ channels triggers exocytosis of insulin-containing granules in pancreatic ß cells and is required for the postprandial spike in insulin secretion. Using high-resolution microscopy, we have identified a subset of docked insulin granules in human ß cells and rat-derived clonal insulin 1 (INS1) cells for which localized Ca2+ influx triggers exocytosis with high probability and minimal latency. This immediately releasable pool (IRP) of granules, identified both structurally and functionally, was absent in ß cells from human T2D donors and in INS1 cells cultured in fatty acids that mimic the diabetic state. Upon arrival at the plasma membrane, IRP granules slowly associated with 15 to 20 L-type channels. We determined that recruitment depended on a direct interaction with the synaptic protein Munc13, because expression of the II-III loop of the channel, the C2 domain of Munc13-1, or of Munc13-1 with a mutated C2 domain all disrupted L-type channel clustering at granules and ablated fast exocytosis. Thus, rapid insulin secretion requires Munc13-mediated recruitment of L-type Ca2+ channels in close proximity to insulin granules. Loss of this organization underlies disturbed insulin secretion kinetics in T2D.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Gránulos Citoplasmáticos/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Señalización del Calcio , Células Cultivadas , Diabetes Mellitus Tipo 2/patología , Humanos , Secreción de Insulina , Proteínas del Tejido Nervioso/metabolismo , Transporte de Proteínas
12.
Nat Commun ; 7: 13496, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27882918

RESUMEN

Insulin secretion from pancreatic ß-cells is impaired in all forms of diabetes. The resultant hyperglycaemia has deleterious effects on many tissues, including ß-cells. Here we show that chronic hyperglycaemia impairs glucose metabolism and alters expression of metabolic genes in pancreatic islets. In a mouse model of human neonatal diabetes, hyperglycaemia results in marked glycogen accumulation, and increased apoptosis in ß-cells. Sulphonylurea therapy rapidly normalizes blood glucose levels, dissipates glycogen stores, increases autophagy and restores ß-cell metabolism. Insulin therapy has the same effect but with slower kinetics. Similar changes are observed in mice expressing an activating glucokinase mutation, in in vitro models of hyperglycaemia, and in islets from type-2 diabetic patients. Altered ß-cell metabolism may underlie both the progressive impairment of insulin secretion and reduced ß-cell mass in diabetes.


Asunto(s)
Apoptosis/fisiología , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucógeno/metabolismo , Hiperglucemia/metabolismo , Enfermedades del Recién Nacido/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Autofagia/fisiología , Glucemia/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Glucoquinasa/genética , Humanos , Hipoglucemiantes/farmacología , Técnicas In Vitro , Recién Nacido , Insulina/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Mutación , Ratas , Compuestos de Sulfonilurea/farmacología
13.
Diabetes ; 65(7): 1952-61, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26993066

RESUMEN

The transcription factor Sox4 has been proposed to underlie the increased type 2 diabetes risk linked to an intronic single nucleotide polymorphism in CDKAL1 In a mouse model expressing a mutant form of Sox4, glucose-induced insulin secretion is reduced by 40% despite normal intracellular Ca(2+) signaling and depolarization-evoked exocytosis. This paradox is explained by a fourfold increase in kiss-and-run exocytosis (as determined by single-granule exocytosis measurements) in which the fusion pore connecting the granule lumen to the exterior expands to a diameter of only 2 nm, which does not allow the exit of insulin. Microarray analysis indicated that this correlated with an increased expression of the exocytosis-regulating protein Stxbp6. In a large collection of human islet preparations (n = 63), STXBP6 expression and glucose-induced insulin secretion correlated positively and negatively with SOX4 expression, respectively. Overexpression of SOX4 in the human insulin-secreting cell EndoC-ßH2 interfered with granule emptying and inhibited hormone release, the latter effect reversed by silencing STXBP6 These data suggest that increased SOX4 expression inhibits insulin secretion and increased diabetes risk by the upregulation of STXBP6 and an increase in kiss-and-run exocytosis at the expense of full fusion. We propose that pharmacological interventions promoting fusion pore expansion may be effective in diabetes therapy.


Asunto(s)
Exocitosis/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Factores de Transcripción SOXC/genética , Animales , Calcio/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Silenciador del Gen , Humanos , Secreción de Insulina , Masculino , Ratones , Factores de Transcripción SOXC/metabolismo , Regulación hacia Arriba
14.
J Clin Invest ; 125(12): 4714-28, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26571400

RESUMEN

Strategies aimed at mimicking or enhancing the action of the incretin hormone glucagon-like peptide 1 (GLP-1) therapeutically improve glucose-stimulated insulin secretion (GSIS); however, it is not clear whether GLP-1 directly drives insulin secretion in pancreatic islets. Here, we examined the mechanisms by which GLP-1 stimulates insulin secretion in mouse and human islets. We found that GLP-1 enhances GSIS at a half-maximal effective concentration of 0.4 pM. Moreover, we determined that GLP-1 activates PLC, which increases submembrane diacylglycerol and thereby activates PKC, resulting in membrane depolarization and increased action potential firing and subsequent stimulation of insulin secretion. The depolarizing effect of GLP-1 on electrical activity was mimicked by the PKC activator PMA, occurred without activation of PKA, and persisted in the presence of PKA inhibitors, the KATP channel blocker tolbutamide, and the L-type Ca(2+) channel blocker isradipine; however, depolarization was abolished by lowering extracellular Na(+). The PKC-dependent effect of GLP-1 on membrane potential and electrical activity was mediated by activation of Na(+)-permeable TRPM4 and TRPM5 channels by mobilization of intracellular Ca(2+) from thapsigargin-sensitive Ca(2+) stores. Concordantly, GLP-1 effects were negligible in Trpm4 or Trpm5 KO islets. These data provide important insight into the therapeutic action of GLP-1 and suggest that circulating levels of this hormone directly stimulate insulin secretion by ß cells.


Asunto(s)
Péptido 1 Similar al Glucagón/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteína Quinasa C/metabolismo , Canales Catiónicos TRPM/metabolismo , Animales , Humanos , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/citología , Transporte Iónico/efectos de los fármacos , Transporte Iónico/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Noqueados , Proteína Quinasa C/genética , Canales Catiónicos TRPM/genética , Acetato de Tetradecanoilforbol/farmacología
15.
J Physiol ; 592(21): 4677-96, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25172946

RESUMEN

Mouse pancreatic ß- and α-cells are equipped with voltage-gated Na(+) currents that inactivate over widely different membrane potentials (half-maximal inactivation (V0.5) at -100 mV and -50 mV in ß- and α-cells, respectively). Single-cell PCR analyses show that both α- and ß-cells have Nav1.3 (Scn3) and Nav1.7 (Scn9a) α subunits, but their relative proportions differ: ß-cells principally express Nav1.7 and α-cells Nav1.3. In α-cells, genetically ablating Scn3a reduces the Na(+) current by 80%. In ß-cells, knockout of Scn9a lowers the Na(+) current by >85%, unveiling a small Scn3a-dependent component. Glucagon and insulin secretion are inhibited in Scn3a(-/-) islets but unaffected in Scn9a-deficient islets. Thus, Nav1.3 is the functionally important Na(+) channel α subunit in both α- and ß-cells because Nav1.7 is largely inactive at physiological membrane potentials due to its unusually negative voltage dependence of inactivation. Interestingly, the Nav1.7 sequence in brain and islets is identical and yet the V0.5 for inactivation is >30 mV more negative in ß-cells. This may indicate the presence of an intracellular factor that modulates the voltage dependence of inactivation.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Canal de Sodio Activado por Voltaje NAV1.3/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Sodio/fisiología , Animales , Regulación de la Expresión Génica , Células Secretoras de Glucagón/efectos de los fármacos , Glucosa , Células HEK293 , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.3/genética , Canal de Sodio Activado por Voltaje NAV1.7/genética , Neurotoxinas/farmacología , Isoformas de Proteínas , Subunidades de Proteína
16.
Curr Biol ; 21(17): 1475-9, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21889890

RESUMEN

Sex chromosomes evolved many times independently in many different organisms [1]. According to the currently accepted model, X and Y chromosomes evolve from a pair of autosomes via a series of inversions leading to stepwise expansion of a nonrecombining region on the Y chromosome (NRY) and the consequential degeneration of genes trapped in the NRY [2]. Our results suggest that plants represent an exception to this rule as a result of their unique life-cycle that includes alteration of diploid and haploid generations and widespread haploid expression of genes in plant gametophytes [3]. Using a new high-throughput approach, we identified over 400 new genes expressed from X and Y chromosomes in Silene latifolia, a plant that evolved sex chromosomes about 10 million years ago. Y-linked genes show faster accumulation of amino-acid replacements and loss of expression, compared to X-linked genes. These degenerative processes are significantly less pronounced in more constrained genes and genes that are likely exposed to haploid-phase selection. This may explain why plants retain hundreds of expressed Y-linked genes despite millions of years of Y chromosome degeneration, whereas animal Y chromosomes are almost completely degenerate.


Asunto(s)
Mapeo Cromosómico , Cromosomas de las Plantas , Regulación de la Expresión Génica de las Plantas , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Silene/genética , Evolución Molecular , Genes de Plantas , Ligamiento Genético , Haploidia , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Cromosomas Sexuales/genética , Transcriptoma
17.
Traffic ; 11(10): 1290-303, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20604900

RESUMEN

The polarized trafficking of membrane proteins into the leading edge of the cell is an integral requirement for cell migration. Myosin VI and its interacting protein optineurin have previously been shown to operate in anterograde trafficking pathways, especially for the polarized delivery of cargo to the basolateral domain in epithelial cells. Here we show that in migratory cells ablation of myosin VI or optineurin inhibits the polarized delivery of the epidermal growth factor receptor (EGFR) into the leading edge and leads to profound defects in lamellipodia formation. Depletion of either myosin VI or optineurin, however, does not impair the overall ability of cells to migrate in a random migration assay, but it dramatically reduces directed migration towards a growth factor stimulus. In summary, we identified a specific role for myosin VI and optineurin in directionally persistent cell migration, which involves the polarized delivery of vesicles containing EGFR into the leading edge of the cell.


Asunto(s)
Movimiento Celular , Polaridad Celular , Receptores ErbB/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Factor de Transcripción TFIIIA/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Endocitosis , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana
18.
Biochem Soc Trans ; 37(Pt 5): 966-70, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19754433

RESUMEN

There is now increasing evidence that myosin motor proteins, together with the dynamic actin filament machinery and associated adhesion proteins, play crucial roles in the events leading to motility at the leading edge of migrating cells. Myosins exist as a large superfamily of diverse ATP-dependent motors, and in the present review, we focus on the unique minus-end-directed myosin VI, briefly discussing its potential functions in cell motility.


Asunto(s)
Movimiento Celular/fisiología , Cadenas Pesadas de Miosina/metabolismo , Actinas/metabolismo , Animales , Moléculas de Adhesión Celular/fisiología , Membrana Celular/metabolismo , Polaridad Celular , Proteínas de la Matriz Extracelular/metabolismo , Aparato de Golgi/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Receptores de Superficie Celular , Transducción de Señal/fisiología
19.
Methods Enzymol ; 438: 11-24, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18413238

RESUMEN

The small GTPase Rab8 has been shown to regulate polarized membrane trafficking pathways from the TGN to the cell surface. Optineurin is an effector protein of Rab8 and a binding partner of the actin-based motor protein myosin VI. We used various approaches to study the interactions between myosin VI and its binding partners and to analyze their role(s) in intracellular membrane trafficking pathways. In this chapter, we describe the use of the mammalian two-hybrid assay to demonstrate protein-protein interactions and to identify binding sites. We describe a secretion assay that was used in combination with RNA interference technology to analyze the function of myosin VI, optineurin, and Rab8 in exocytic membrane trafficking pathways.


Asunto(s)
Cadenas Pesadas de Miosina/metabolismo , Factor de Transcripción TFIIIA/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Células CHO , Proteínas de Ciclo Celular , Cricetinae , Cricetulus , Células HeLa , Humanos , Proteínas de Transporte de Membrana , Ratones , ARN Interferente Pequeño , Transfección/métodos , Técnicas del Sistema de Dos Híbridos
20.
Eur J Neurosci ; 27(7): 1626-33, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18380665

RESUMEN

Dopamine signalling is a critically important process in the human brain that controls mood, cognition and motor activity. In order to gain detailed insight into this signalling pathway at the molecular level, we carried out yeast two-hybrid screens with D1-like (D1, D5) and D2-like (D2, D3, D4) dopamine receptors and identified 11 dopamine receptor interacting proteins (DRIPs). Using the C-terminal domain of D1 receptor as bait, we identified AIP1 (ALG-2 interacting protein 1), a known modulator of caspase-dependent and caspase-independent cell death, including neuronal cell death, that is also part of the endosomal transport system. In a separate yeast two-hybrid screen, using the third intracellular cytoplasmic loop of D3 as bait, we again identified AIP1. The interaction of AIP1 with both D1 and D3 was confirmed in vitro and in vivo using a variety of methods, including glutathione S-transferase (GST) pull-down, blot overlay and coimmunoprecipitation from mouse brain lysates. We have also observed colocalization of D1 and D3 with AIP1 in mouse brain tissue. In addition, coexpression of AIP1 with D1 resulted in > 50% reduction in binding capacity of D1 to its antagonist. Finally, AIP1 up-regulates D1 and D3 expression and appears to be important for their stability and trafficking.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Receptores de Dopamina D1/fisiología , Receptores de Dopamina D3/fisiología , Transducción de Señal/fisiología , Animales , Proteínas de Unión al Calcio/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Línea Celular , Complejos de Clasificación Endosomal Requeridos para el Transporte , Humanos , Ratones , Unión Proteica/fisiología , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Ratas , Receptores de Dopamina D1/genética , Receptores de Dopamina D3/genética , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...