Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 241
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Mol Endocrinol ; 62(4): 159-168, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30917339

RESUMEN

Modified lipoproteins can negatively affect beta cell function and survival. However, the mechanisms behind interactions of modified lipoproteins with beta cells - and in particular, relationships to increased uptake - are only partly clarified. By over-expressing the scavenger receptor CD36 (Tet-on), we increased the uptake of fluorescent low-density modified lipoprotein (oxLDL) into insulin-secreting INS-1 cells. The magnitude of uptake followed the degree of CD36 over-expression. CD36 over-expression increased concomitant efflux of 3H-cholesterol in proportion to the cellular contents of 3H-cholesterol. Exposure to concentrations of oxLDL from 20 to 100 µg/mL dose-dependently increased toxicity (evaluated by MTT) as well as apoptosis. However, the increased uptake of oxLDL due to CD36 over-expression did not exert additive effects on oxLDL toxicity - neither on viability, nor on glucose-induced insulin release and cellular content. Reciprocally, blocking CD36 receptors by Sulfo-N-Succinimidyl Oleate decreased the uptake of oxLDL but did not diminish the toxicity. Pancreatic islets of CD36-/- mice displayed reduced uptake of 3H-cholesterol-labeled oxLDL vs wild type but similar toxicity to oxLDL. OxLDL was found to increase the expression of CD36 in islets and INS-1 cells. In summary, given the experimental conditions, our results indicate that (1) increased uptake of oxLDL is not responsible for toxicity of oxLDL, (2) increased efflux of the cholesterol moiety of oxLDL counterbalances, at least in part, increased uptake and (3) oxLDL participates in the regulation of CD36 in pancreatic islets and in INS-1 cells.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Lipoproteínas LDL/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Antígenos CD36/genética , Antígenos CD36/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Colesterol/metabolismo , Doxiciclina/farmacología , Citometría de Flujo , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Lipoproteínas LDL/farmacología , Masculino , Ratones , Microscopía Confocal , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
Nat Commun ; 8: 15652, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28585545

RESUMEN

Type 2 diabetes (T2D) is characterized by insulin resistance and impaired insulin secretion, but the mechanisms underlying insulin secretion failure are not completely understood. Here, we show that a set of co-expressed genes, which is enriched for genes with islet-selective open chromatin, is associated with T2D. These genes are perturbed in T2D and have a similar expression pattern to that of dedifferentiated islets. We identify Sox5 as a regulator of the module. Sox5 knockdown induces gene expression changes similar to those observed in T2D and diabetic animals and has profound effects on insulin secretion, including reduced depolarization-evoked Ca2+-influx and ß-cell exocytosis. SOX5 overexpression reverses the expression perturbations observed in a mouse model of T2D, increases the expression of key ß-cell genes and improves glucose-stimulated insulin secretion in human islets from donors with T2D. We suggest that human islets in T2D display changes reminiscent of dedifferentiation and highlight SOX5 as a regulator of ß-cell phenotype and function.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción SOXD/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Cromatina/metabolismo , Exocitosis , Femenino , Regulación de la Expresión Génica , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Florizina/química , ARN Interferente Pequeño/metabolismo , Ratas , Ácido Valproico/química
3.
Mol Cell Endocrinol ; 447: 52-60, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28237718

RESUMEN

Impaired beta-cell function is key to the development of type 2 diabetes. Cocaine- and amphetamine-regulated transcript (CART) is an islet peptide with insulinotropic and glucagonostatic properties. Here we studied the role of endogenous CART in beta-cell function. CART silencing in INS-1 (832/13) beta-cells reduced insulin secretion and production, ATP levels and beta-cell exocytosis. This was substantiated by reduced expression of several exocytosis genes, as well as reduced expression of genes important for insulin secretion and processing. In addition, CART silencing reduced the expression of a network of transcription factors essential for beta-cell function. Moreover, in RNAseq data from human islet donors, CARTPT expression levels correlated with insulin, exocytosis genes and key beta-cell transcription factors. Thus, endogenous beta-cell CART regulates insulin expression and secretion in INS-1 (832/13) cells, via actions on the exocytotic machinery and a network of beta-cell transcription factors. We conclude that CART is important for maintaining the beta-cell phenotype.


Asunto(s)
Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transcripción Genética , Animales , Apoptosis/genética , Exocitosis/genética , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Secreción de Insulina , Células Secretoras de Insulina/citología , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Factores de Transcripción/metabolismo
4.
Cell Death Dis ; 6: e1976, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26583319

RESUMEN

Pathologic alterations in podocytes lead to failure of an essential component of the glomerular filtration barrier and proteinuria in chronic kidney diseases. Elevated levels of saturated free fatty acid (FFA) are harmful to various tissues, implemented in the progression of diabetes and its complications such as proteinuria in diabetic nephropathy. Here, we investigated the molecular mechanism of palmitate cytotoxicity in cultured mouse podocytes. Incubation with palmitate dose-dependently increased cytosolic and mitochondrial reactive oxygen species, depolarized the mitochondrial membrane potential, impaired ATP synthesis and elicited apoptotic cell death. Palmitate not only evoked mitochondrial fragmentation but also caused marked dilation of the endoplasmic reticulum (ER). Consistently, palmitate upregulated ER stress proteins, oligomerized stromal interaction molecule 1 (STIM1) in the subplasmalemmal ER membrane, abolished the cyclopiazonic acid-induced cytosolic Ca(2+) increase due to depletion of luminal ER Ca(2+). Palmitate-induced ER Ca(2+) depletion and cytotoxicity were blocked by a selective inhibitor of the fatty-acid transporter FAT/CD36. Loss of the ER Ca(2+) pool induced by palmitate was reverted by the phospholipase C (PLC) inhibitor edelfosine. Palmitate-dependent activation of PLC was further demonstrated by following cytosolic translocation of the pleckstrin homology domain of PLC in palmitate-treated podocytes. An inhibitor of diacylglycerol (DAG) kinase, which elevates cytosolic DAG, strongly promoted ER Ca(2+) depletion by low-dose palmitate. GF109203X, a PKC inhibitor, partially prevented palmitate-induced ER Ca(2+) loss. Remarkably, the mitochondrial antioxidant mitoTEMPO inhibited palmitate-induced PLC activation, ER Ca(2+) depletion and cytotoxicity. Palmitate elicited cytoskeletal changes in podocytes and increased albumin permeability, which was also blocked by mitoTEMPO. These data suggest that oxidative stress caused by saturated FFA leads to mitochondrial dysfunction and ER Ca(2+) depletion through FAT/CD36 and PLC signaling, possibly contributing to podocyte injury.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Palmitatos/farmacología , Podocitos/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Retículo Endoplásmico/metabolismo , Ratones , Mitocondrias/metabolismo , Podocitos/metabolismo
5.
Diabetologia ; 55(12): 3262-72, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22965295

RESUMEN

AIMS/HYPOTHESIS: Glucagon-like peptide 1 (GLP-1) is a major incretin, mainly produced by the intestinal L cells, with beneficial actions on pancreatic beta cells. However, while in vivo only very small amounts of GLP-1 reach the pancreas in bioactive form, some observations indicate that GLP-1 may also be produced in the islets. We performed comprehensive morphological, functional and molecular studies to evaluate the presence and various features of a local GLP-1 system in human pancreatic islet cells, including those from type 2 diabetic patients. METHODS: The presence of insulin, glucagon, GLP-1, proconvertase (PC) 1/3 and PC2 was determined in human pancreas by immunohistochemistry with confocal microscopy. Islets were isolated from non-diabetic and type 2 diabetic donors. GLP-1 protein abundance was evaluated by immunoblotting and matrix-assisted laser desorption-ionisation-time of flight (MALDI-TOF) mass spectrometry. Single alpha and beta cell suspensions were obtained by enzymatic dissociation and FACS sorting. Glucagon and GLP-1 release were measured in response to nutrients. RESULTS: Confocal microscopy showed the presence of GLP-1-like and PC1/3 immunoreactivity in subsets of alpha cells, whereas GLP-1 was not observed in beta cells. The presence of GLP-1 in isolated islets was confirmed by immunoblotting, followed by mass spectrometry. Isolated islets and alpha (but not beta) cell fractions released GLP-1, which was regulated by glucose and arginine. PC1/3 (also known as PCSK1) gene expression was shown in alpha cells. GLP-1 release was significantly higher from type 2 diabetic than from non-diabetic isolated islets. CONCLUSIONS/INTERPRETATION: We have shown the presence of a functionally competent GLP-1 system in human pancreatic islets, which resides in alpha cells and might be modulated by type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Insulina/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Páncreas/metabolismo
6.
Diabetes Obes Metab ; 13(4): 326-36, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21205124

RESUMEN

AIM: Glucagon-like peptide-1 (GLP-1) has protective effects on pancreatic ß-cells. We evaluated the effects of a novel, long-acting human GLP-1 analogue, taspoglutide, on ß-cells in vitro and in vivo. METHODS: Proliferation of murine pancreatic ß (MIN6B1) cells and rat islets in culture was assessed by imaging of 5-ethynyl-2'-deoxyuridine-positive cells after culture with taspoglutide. Apoptosis was evaluated with the transferase-mediated 2'-deoxyuridine 5'-triphosphate nick-end labelling assay in rat insulinoma (INS-1E) cells and isolated human islets exposed to cytokines (recombinant interleukin-1ß, interferon-γ, tumour necrosis factor-α) or lipotoxicity (palmitate) in the presence or absence of taspoglutide. Islet morphology and survival and glucose-stimulated insulin secretion in perfused pancreata were assessed 3-4 weeks after a single application of taspoglutide to prediabetic 6-week-old male Zucker diabetic fatty (ZDF) rats. RESULTS: Proliferation was increased in a concentration-dependent manner up to fourfold by taspoglutide in MIN6B1 cells and was significantly stimulated in isolated rat islets. Taspoglutide almost completely prevented cytokine- or lipotoxicity-induced apoptosis in INS-1E cells (control 0.5%, cytokines alone 2.2%, taspoglutide + cytokines 0.6%, p < 0.001; palmitate alone 8.1%, taspoglutide + palmitate 0.5%, p < 0.001) and reduced apoptosis in isolated human islets. Treatment of ZDF rats with taspoglutide significantly prevented ß-cell apoptosis and preserved healthy islet architecture and insulin staining intensity as shown in pancreatic islet cross sections. Basal and glucose-stimulated insulin secretion of in situ perfused ZDF rat pancreata was normalized after taspoglutide treatment. CONCLUSIONS: Taspoglutide promoted ß-cell proliferation, prevented apoptosis in vitro and exerted multiple ß-cell protective effects on islet architecture and function in vivo in ZDF rats.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/administración & dosificación , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Péptidos/administración & dosificación , Receptores de Glucagón/administración & dosificación , Animales , Apoptosis , Células Cultivadas , Desoxiuridina/análogos & derivados , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Humanos , Inmunohistoquímica , Células Secretoras de Insulina/fisiología , Masculino , Péptidos/farmacología , Ratas , Ratas Zucker
7.
Diabetologia ; 54(2): 360-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21104225

RESUMEN

AIMS/HYPOTHESIS: Although recent studies propose that epigenetic factors influence insulin expression, the regulation of the insulin gene in type 2 diabetic islets is still not fully understood. Here, we examined DNA methylation of the insulin gene promoter in pancreatic islets from patients with type 2 diabetes and non-diabetic human donors and related it to insulin expression, HbA(1c) levels, BMI and age. METHODS: DNA methylation was analysed in 25 CpG sites of the insulin promoter and insulin mRNA expression was analysed using quantitative RT-PCR in pancreatic islets from nine donors with type 2 diabetes and 48 non-diabetic donors. RESULTS: Insulin mRNA expression (p = 0.002), insulin content (p = 0.004) and glucose-stimulated insulin secretion (p = 0.04) were reduced in pancreatic islets from patients with type 2 diabetes compared with non-diabetic donors. Moreover, four CpG sites located 234 bp, 180 and 102 bp upstream and 63 bp downstream of the transcription start site (CpG -234, -180, -102 and +63, respectively), showed increased DNA methylation in type 2 diabetic compared with non-diabetic islets (7.8%, p = 0.03; 7.1%, p = 0.02; 4.4%, p = 0.03 and 9.3%, p = 0.03, respectively). While insulin mRNA expression correlated negatively (p < 1 × 10(-6)), the level of HbA(1c) correlated positively (p ≤ 0.01) with the degree of DNA methylation for CpG -234, -180 and +63. Furthermore, DNA methylation for nine additional CpG sites correlated negatively with insulin mRNA expression (p ≤ 0.01). Also, exposure to hyperglycaemia for 72 h increased insulin promoter DNA methylation in clonal rat beta cells (p = 0.005). CONCLUSIONS/INTERPRETATIONS: This study demonstrates that DNA methylation of the insulin promoter is increased in patients with type 2 diabetes and correlates negatively with insulin gene expression in human pancreatic islets.


Asunto(s)
Metilación de ADN/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Regiones Promotoras Genéticas/genética , Animales , Línea Celular , Metilación de ADN/genética , Humanos , Técnicas In Vitro , Insulina/genética , Células Secretoras de Insulina/metabolismo , Ratas
8.
Br J Pharmacol ; 156(8): 1296-304, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19302589

RESUMEN

BACKGROUND AND PURPOSE: Amiloride derivatives are blockers of the Na(+)/H(+) exchanger (NHE) and at micromolar concentrations have protective effects on cardiac and brain ischaemia/reperfusion injury but at higher concentrations also induce apoptosis. Here, we aimed to elucidate the mechanism related to this cytotoxic action. EXPERIMENTAL APPROACH: We quantified the expression of genes associated with endoplasmic reticulum (ER) stress and measured changes in luminal ER Ca(2+) concentration ([Ca(2+)](ER)) with a 'cameleon' indicator, D1ER. KEY RESULTS: Amiloride derivatives induced apoptosis in vascular endothelial cells, an effect that increased at alkaline extracellular pH. The potency order for cytotoxicity was 5-(N,N-hexamethylene)-amiloride (HMA) > 5-(N-methyl-N-isobutyl) amiloride > 5-(N-ethyl-N-isopropyl) amiloride (EIPA) >> amiloride. HMA dose-dependently increased the transcription of the ER stress genes GADD153 and GADD34 and rapidly depleted [Ca(2+)](ER), mimicking the effects of the sarco/endoplasmic reticulum ATPase (SERCA) inhibitor thapsigargin. The NHE1-specific inhibitor HOE 694 inhibited NHE activity by 87% but did not alter [Ca(2+)](ER). The decrease in [Ca(2+)](ER) evoked by amiloride derivatives was also observed in HeLa cells and was mirrored by an increase in cytosolic Ca(2+) concentration. CONCLUSIONS AND IMPLICATIONS: Amiloride derivatives disrupt ER and cytosolic Ca(2+) homeostasis by a mechanism unrelated to NHE inhibition, most likely by interfering with the activity of SERCA. We propose that ER Ca(2+) depletion and subsequent ER stress provide a rationale framework for the apoptotic effects of amiloride derivatives.


Asunto(s)
Amilorida/toxicidad , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Diuréticos/toxicidad , Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Amilorida/análogos & derivados , Antígenos de Diferenciación/genética , Proteínas de Ciclo Celular/genética , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Cinética , Proteína Fosfatasa 1 , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/metabolismo , Sulfonas/farmacología , Tapsigargina/farmacología , Factor de Transcripción CHOP/genética , Transcripción Genética/efectos de los fármacos
9.
Exp Clin Endocrinol Diabetes ; 116 Suppl 1: S46-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18777454

RESUMEN

Mice, deficient for vascular endothelial growth factor VEGF-A in pancreatic islets, have reduced insulin gene expression levels and an impaired glucose tolerance. Here, we investigated whether VEGF-A was required for physiological glucose-stimulated insulin secretion and insulin content. We performed in situ pancreas perfusions and islet perifusions on mice lacking VEGF-A in the pancreatic epithelium in order to study their ability to secrete insulin in response to glucose. We identified insulin secretion defects in the pancreata of VEGF-A deficient mice, including a delayed and blunted response to glucose. Islet perifusion experiments revealed a missing first phase and weaker second phase of insulin secretion, in two of three VEGF-A deficient mice. On average, insulin content in VEGF-A deficient islets was significantly reduced when compared with control islets. We conclude that VEGF-A is required in pancreatic islets for normal glucose-stimulated insulin secretion and physiological insulin content. Thus, VEGF-A is a key factor for pancreatic islet function.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Arginina/farmacología , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Glucosa/farmacología , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/fisiología , Ratones , Ratones Noqueados , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Genes Immun ; 8(3): 232-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17330137

RESUMEN

We recently finemapped a type 1 diabetes (T1D)-linked region on chromosome 21, indicating that one or more T1D-linked genes exist in this region with 33 annotated genes. In the current study, we have taken a novel approach using transcriptional profiling in predicting and prioritizing the most likely candidate genes influencing beta-cell function in this region. Two array-based approaches were used, a rat insulinoma cell line (INS-1alphabeta) overexpressing pancreatic duodenum homeobox 1 (pdx-1) and treated with interleukin 1beta (IL-1beta) as well as human pancreatic islets stimulated with a mixture of cytokines. Several candidate genes with likely functional significance in T1D were identified. Genes showing differential expression in the two approaches were highly similar, supporting the role of these specific gene products in cytokine-induced beta-cell damage. These were genes involved in cytokine signaling, oxidative phosphorylation, defense responses and apoptosis. The analyses, furthermore, revealed several transcription factor binding sites shared by the differentially expressed genes and by genes demonstrating highly similar expression profiles with these genes. Comparable findings in the rat beta-cell line and human islets support the validity of the methods used and support this as a valuable approach for gene mapping and identification of genes with potential functional significance in T1D, within a region of linkage.


Asunto(s)
Cromosomas Humanos Par 21/genética , Diabetes Mellitus Tipo 1/genética , Islotes Pancreáticos/metabolismo , Adolescente , Adulto , Animales , Línea Celular Tumoral , Niño , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Proteínas de Homeodominio/genética , Humanos , Técnicas In Vitro , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/genética , Interleucina-1beta/farmacología , Islotes Pancreáticos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/genética , Ratas , Transactivadores/genética
11.
Oncogene ; 26(29): 4261-71, 2007 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-17260022

RESUMEN

The paired/homeodomain transcription factor Pax4 is essential for islet beta-cell generation during pancreas development and their survival in adulthood. High Pax4 expression was reported in human insulinomas indicating that deregulation of the gene may be associated with tumorigenesis. We report that rat insulinoma INS-1E cells express 25-fold higher Pax4 mRNA levels than rat islets. In contrast to primary beta-cells, activin A but not betacellulin or glucose induced Pax4 mRNA levels indicating dissociation of Pax4 expression from insulinoma cell proliferation. Short hairpin RNA adenoviral constructs targeted to the paired domain or homeodomain (viPax4PD and viPax4HD) were generated. Pax4 mRNA levels were lowered by 73 and 50% in cells expressing either viPax4PD or viPax4HD. Transcript levels of the Pax4 target gene bcl-xl were reduced by 53 and 47%, whereas Pax6 and Pdx1 mRNA levels were unchanged. viPax4PD-infected cells displayed a twofold increase in spontaneous apoptosis and were more susceptible to cytokine-induced cell death. In contrast, proliferation was unaltered. RNA interference-mediated repression of insulin had no adverse effects on either Pax4 or Pdx1 expression as well as on cell replication or apoptosis. These results indicate that Pax4 is redundant for proliferation of insulinoma cells, whereas it is essential for survival through upregulation of the antiapoptotic gene bcl-xl.


Asunto(s)
Proteínas de Homeodominio/genética , Insulinoma/genética , Insulinoma/patología , Factores de Transcripción Paired Box/genética , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , División Celular/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Proteínas de Homeodominio/fisiología , Insulinoma/metabolismo , Factores de Transcripción Paired Box/fisiología , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Regulación hacia Arriba/genética , Proteína bcl-X/biosíntesis , Proteína bcl-X/genética
12.
Diabetologia ; 50(1): 84-93, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17131143

RESUMEN

AIMS/HYPOTHESIS: Levels of uncoupling protein-2 (UCP2) are regulated in the pancreatic beta cells and an increase in the protein level has been associated with mitochondrial uncoupling and alteration in glucose-stimulated insulin secretion. However, it is not clear whether an increase in uncoupling protein-2 per se induces mitochondrial uncoupling and affects ATP generation and insulin secretion. MATERIALS AND METHODS: Transgenic mice with beta cell-specific overexpression of the human UCP2 gene and INS-1 cells with doxycycline-inducible overproduction of the protein were generated and the consequences of increased levels of UCP2 on glucose-induced insulin secretion and on parameters reflecting mitochondrial uncoupling were determined. RESULTS: In transgenic mice, an increase in beta cell UCP2 protein concentration did not significantly modify plasma glucose and insulin levels. Glucose-induced insulin secretion and elevation in the ATP/ADP ratio were unaltered by an increase in UCP2 level. In INS-1 cells, a similar increase in UCP2 level did not modify glucose-induced insulin secretion, cytosolic ATP and ATP/ADP ratio, or glucose oxidation. Increased levels of UCP2 did not modify the mitochondrial membrane potential and oxygen consumption. Increased UCP2 levels decreased cytokine-induced production of reactive oxygen species. CONCLUSION/INTERPRETATION: The results obtained in transgenic mice and in the beta cell line do not support the hypothesis that an increase in UCP2 protein per se uncouples the mitochondria and decreases glucose-induced insulin secretion. In contrast, the observation that increased UCP2 levels decrease cytokine-induced production of reactive oxygen species indicates a potential protective effect of the protein on beta cells, as observed in other cell types.


Asunto(s)
Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Respiración de la Célula/fisiología , Doxiciclina/farmacología , Femenino , Glucosa/metabolismo , Canales Iónicos/genética , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/genética , Proteína Desacopladora 2
13.
Diabetologia ; 50(2): 348-58, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17149590

RESUMEN

AIMS/HYPOTHESIS: Effects of the transcription factor v-maf musculoaponeurotic fibrosarcoma oncogene homologue A (MAFA) on the regulation of beta cell gene expression and function were investigated. MATERIALS AND METHODS: INS-1 stable cell lines permitting inducible up- or downregulation of this transcription factor were established. RESULTS: MAFA overproduction enhanced and its dominant-negative mutant (DN-MAFA) diminished binding of the factor to the insulin promoter, correlating with insulin mRNA levels and cellular protein content. Glucose-stimulated insulin secretion was facilitated by MAFA and blunted by DN-MAFA. This is partly due to alterations in glucokinase production, the glucose sensor of beta cells. In addition, the expression of important beta cell genes, e.g. those encoding solute carrier family 2 (facilitated glucose transporter), member 2 (formerly known as GLUT2), pancreatic and duodenal homeobox factor 1 (PDX1), NK6 transcription factor-related, locus 1 (NKX6-1), glucagon-like peptide 1 receptor (GLP1R), prohormone convertase 1/3 (PCSK1) and pyruvate carboxylase (PC), was regulated positively by MAFA and negatively by DN-MAFA. CONCLUSIONS/INTERPRETATION: The data suggest that MAFA is not only a key activator of insulin transcription, but also a master regulator of genes implicated in maintaining beta cell function, in particular metabolism-secretion coupling, proinsulin processing and GLP1R signalling. Our in vitro study provides molecular targets that explain the phenotype of recently reported Mafa-null mice. We also demonstrate that MAFA is produced specifically in beta cells of human islets. Glucose influenced DNA-binding activity of MAFA in rat islets in a bell-shaped manner. MAFA thus qualifies as a master regulator of beta-cell-specific gene expression and function.


Asunto(s)
Células Secretoras de Insulina/fisiología , Insulina/genética , Lectinas Tipo C/fisiología , Glicoproteínas de Membrana/fisiología , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Insulina/biosíntesis , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Insulinoma , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Neoplasias Pancreáticas , Ratas , Transcripción Genética
14.
Exp Cell Res ; 312(15): 2826-36, 2006 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-16781709

RESUMEN

Despite extensive studies, the hematopoietic versus hepatic origin of liver progenitor oval cells remains controversial. The aim of this study was to determine the origin of such cells after liver injury and to establish an oval cell line. Rat liver injury was induced by subcutaneous insertion of 2-AAF pellets for 7 days with subsequent injection of CCl(4). Livers were removed 9 to 13 days post-CCl(4) treatment. Immunohistochemistry was performed using anti-c-kit, OV6, Thy1, CK19, AFP, vWF and Rab3b. Isolated non-parenchymal cells were grown on mouse embryonic fibroblast, and their gene expression profile was characterized by RT-PCR. We identified a subpopulation of OV6/CK19/Rab3b-expressing cells that was activated in the periportal region of traumatized livers. We also characterized a second subpopulation that expressed the HSCs marker c-kit but not Thy1. Although we successfully isolated both cell types, OV6/CK19/Rab3b(+) cells fail to propagate while c-kit(+)-HSCs appeared to proliferate for up to 7 weeks. Cells formed clusters which expressed c-kit, Thy1 and albumin. Our results indicate that a bona fide oval progenitor cell population resides within the liver and is distinct from c-kit(+)-HSCs. Oval cells require the hepatic niche to proliferate, while cells mobilized from the circulation proliferate and transdifferentiate into hepatocytes without evidence of cell fusion.


Asunto(s)
Células Madre Hematopoyéticas/citología , Hepatocitos/citología , Regeneración Hepática , Hígado/citología , Células Madre/citología , 2-Acetilaminofluoreno/metabolismo , 2-Acetilaminofluoreno/farmacología , Animales , Antígenos de Diferenciación/metabolismo , Médula Ósea/metabolismo , Tetracloruro de Carbono/farmacología , Células Cultivadas , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Inmunohistoquímica , Hígado/efectos de los fármacos , Hígado/crecimiento & desarrollo , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Células Madre/metabolismo , Antígenos Thy-1/inmunología , Antígenos Thy-1/metabolismo
15.
Diabetologia ; 48(5): 900-5, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15834548

RESUMEN

AIMS/HYPOTHESIS: Loss of pancreatic beta cells is the crucial event in the development of type 1 diabetes. It is the result of an imbalance between autoimmune destruction and insufficient regeneration of islet cells. To study the role of islet cell regeneration in the pathogenesis of type 1 diabetes, we focused on PAX4, a paired homeodomain transcriptional repressor that is involved in islet cell growth. METHODS: The study included 379 diabetic children and 1,070 controls from two distinct populations, and a cohort of children who had not developed type 1 diabetes, despite the presence of islet cell antibodies. Genomic DNA analysis of PAX4 was carried out via direct sequencing of PCR-amplified fragments and allelic discrimination. We compared the transrepression potential of the PAX4 variants in betaTC3 cells and analysed their influence on beta cell growth. RESULTS: The type 1 diabetic subjects are different from the normal individuals in terms of the genotype distribution of the A1168C single nucleotide polymorphism in PAX4. The C/C genotype is frequent among type 1 diabetic children (73%) and rare among the control population (32%). Conversely, the A/C genotype is prevalent among control subjects (62%) and antibody-positive children without type 1 diabetes (73.6%), but uncommon among subjects with type 1 diabetes (17.5%). The combination of PAX4A and PAX4C is functionally more active than PAX4C alone (the "diabetic" variant). Beta cells expressing PAX4A and PAX4C efficiently proliferate when stimulated with glucose, whereas cells expressing the PAX4C variant alone do not. CONCLUSIONS/INTERPRETATION: We have identified a link between beta cell regenerative capacity and susceptibility to type 1 diabetes. This finding could explain the fact that not all of the individuals who develop autoimmunity against beta cells actually contract the disease. The C/C genotype of the A1168C polymorphism in PAX4 can be viewed as a predisposition marker that can help to detect individuals prone to develop type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Variación Genética , Proteínas de Homeodominio/genética , Polimorfismo de Nucleótido Simple , Factores de Transcripción/genética , Sustitución de Aminoácidos , Animales , Sitios de Unión , Glucemia/metabolismo , Niño , ADN/sangre , ADN/genética , ADN/aislamiento & purificación , Diabetes Mellitus Tipo 1/sangre , Femenino , Frecuencia de los Genes , Marcadores Genéticos , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Masculino , Mutación Missense , Factores de Transcripción Paired Box , Regiones Promotoras Genéticas , Valores de Referencia , Suiza , Factores de Transcripción/metabolismo
16.
Diabetologia ; 48(4): 720-31, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15756539

RESUMEN

AIMS/HYPOTHESIS: Mutations in genes encoding HNF-4alpha, HNF-1alpha and IPF-1/Pdx-1 are associated with, respectively, MODY subtypes-1, -3 and -4. Impaired glucose-stimulated insulin secretion is the common primary defect of these monogenic forms of diabetes. A regulatory circuit between these three transcription factors has also been suggested. We aimed to explore how Pdx-1 regulates beta cell function and gene expression patterns. METHODS: We studied two previously established INS-1 stable cell lines permitting inducible expression of, respectively, Pdx-1 and its dominant-negative mutant. We used HPLC for insulin processing, adenovirally encoded aequorin for cytosolic [Ca2+], and transient transfection of human growth hormone or patch-clamp capacitance recordings to monitor exocytosis. RESULTS: Induction of DN-Pdx-1 resulted in defective glucose-stimulated and K+-depolarisation-induced insulin secretion in INS-1 cells, while overexpression of Pdx-1 had no effect. We found that DN-Pdx-1 caused down-regulation of fibroblast growth factor receptor 1 (FGFR1), and consequently prohormone convertases (PC-1/3 and -2). As a result, DN-Pdx-1 severely impaired proinsulin processing. In addition, induction of Pdx-1 suppressed the expression of glucagon-like peptide 1 receptor (GLP-1R), which resulted in marked reduction of both basal and GLP-1 agonist exendin-4-stimulated cellular cAMP levels. Induction of DN-Pdx-1 did not affect glucokinase activity, glycolysis, mitochondrial metabolism or ATP generation. The K+-induced cytosolic [Ca2+] rise and Ca2+-evoked exocytosis (membrane capacitance) were not abrogated. CONCLUSIONS/INTERPRETATION: The severely impaired proinsulin processing combined with decreased GLP-1R expression and cellular cAMP content, rather than metabolic defects or altered exocytosis, may contribute to the beta cell dysfunction induced by Pdx-1 deficiency.


Asunto(s)
Proteínas de Homeodominio/fisiología , Insulina/metabolismo , Proinsulina/metabolismo , Receptores de Glucagón/fisiología , Transducción de Señal/fisiología , Transactivadores/fisiología , Adenosina Trifosfato/metabolismo , Animales , Señalización del Calcio/fisiología , Línea Celular Tumoral , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Doxiciclina/farmacología , Exocitosis/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón , Glucoquinasa/genética , Glucosa/metabolismo , Glucosa/farmacología , Glucólisis , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hormona de Crecimiento Humana/genética , Hormona de Crecimiento Humana/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Mitocondrias/metabolismo , Mutación , Proproteína Convertasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Glucagón/genética , Factores de Tiempo , Transactivadores/genética , Transactivadores/metabolismo , Transfección
17.
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
18.
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
19.
Endocrinology ; 142(12): 5311-20, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11713231

RESUMEN

Mutations in the transcription factor hepatocyte nuclear factor-1 alpha (HNF-1 alpha) cause maturity-onset diabetes of the young 3, a severe form of diabetes characterized by pancreatic beta-cell dysfunction. We have used targeted expression of a dominant-negative mutant of HNF-1 alpha to specifically suppress HNF-1 alpha function in beta-cells of transgenic mice. We show that males expressing the mutant protein became overtly diabetic within 6 wk of age, whereas females displayed glucose intolerance. Transgenic males exhibited impaired glucose-stimulated insulin secretion, detected both in vivo and in the perfused pancreas. Pancreatic insulin content was markedly decreased in diabetic animals, whereas the glucagon content was increased. Postnatal islet development was altered, with an increased alpha-cell to beta-cell ratio. beta-Cell ultrastructure showed signs of severe beta-cell damage, including mitochondrial swelling. This animal model of maturity-onset diabetes of the young 3 should be useful for the further elucidation of the mechanism by which HNF-1 alpha deficiency causes beta-cell dysfunction in this disease.


Asunto(s)
Proteínas de Unión al ADN , Diabetes Mellitus Tipo 2/genética , Expresión Génica , Marcación de Gen , Genes Dominantes , Islotes Pancreáticos/fisiología , Proteínas Nucleares , Factores de Transcripción/genética , Animales , Femenino , Glucagón/metabolismo , Intolerancia a la Glucosa/genética , Transportador de Glucosa de Tipo 2 , Factor Nuclear 1 del Hepatocito , Factor Nuclear 1-alfa del Hepatocito , Factor Nuclear 1-beta del Hepatocito , Insulina/metabolismo , Antagonistas de Insulina/farmacología , Islotes Pancreáticos/ultraestructura , Masculino , Ratones , Ratones Transgénicos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Mutación/fisiología , Páncreas/metabolismo , Fenotipo , Caracteres Sexuales , Factores de Transcripción/farmacología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA