Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Diabetologia ; 58(6): 1333-43, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25810039

RESUMEN

AIMS/HYPOTHESIS: Membrane phospholipids are the major intracellular source for fatty acid-derived mediators, which regulate myriad cell functions. We showed previously that high glucose levels triggered the hydrolysis of polyunsaturated fatty acids from beta cell phospholipids. These fatty acids were subjected to free radical-catalysed peroxidation to generate the bioactive aldehyde 4-hydroxy-2E-nonenal (4-HNE). The latter activated the nuclear peroxisome proliferator-activated receptor-δ (PPARδ), which in turn augmented glucose-stimulated insulin secretion. The present study aimed at investigating the combined effects of glucose and fatty acid overload on phospholipid turnover and the subsequent generation of lipid mediators, which affect insulin secretion and beta cell viability. METHODS: INS-1E cells were incubated with increasing glucose concentrations (5-25 mmol/l) without or with palmitic acid (PA; 50-500 µmol/l) and taken for fatty acid-based lipidomic analysis and functional assays. Rat isolated islets of Langerhans were used similarly. RESULTS: PA was incorporated into membrane phospholipids in a concentration- and time-dependent manner; incorporation was highest at 25 mmol/l glucose. This was coupled to a rapid exchange with saturated, mono-unsaturated and polyunsaturated fatty acids. Importantly, released arachidonic acid and linoleic acid were subjected to peroxidation, resulting in the generation of 4-HNE, which further augmented insulin secretion by activating PPARδ in beta cells. However, this adaptive increase in insulin secretion was abolished at high glucose and PA levels, which induced endoplasmic reticulum stress, apoptosis and cell death. CONCLUSIONS/INTERPRETATION: These findings highlight a key role for phospholipid remodelling and fatty acid peroxidation in mediating adaptive and cytotoxic interactions induced by nutrient overload in beta cells.


Asunto(s)
Células Secretoras de Insulina/citología , Peroxidación de Lípido , Fosfolípidos/química , Animales , Apoptosis/efectos de los fármacos , Glucemia/química , Línea Celular , Supervivencia Celular , Ácidos Grasos/química , Radicales Libres , Islotes Pancreáticos/metabolismo , Masculino , PPAR delta/metabolismo , PPAR gamma/metabolismo , Ácido Palmítico/química , Ratas , Ratas Wistar
3.
J Cell Mol Med ; 19(8): 1887-99, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25754218

RESUMEN

Vascular endothelial cell (VEC) senescence is considered an early event in the development of atherosclerotic lesions. Stressful stimuli, in particular oxidative stress, have been linked to premature senescence in the vasculature. Foam cells are a major source of reactive oxygen species and may play a role in the induction of VEC senescence; hence, we investigated their involvement in the induction of VEC senescence in a co-culture transwell system. Primary bovine aortic endothelial cells, exposed to the secretome of THP-1 monocyte-derived foam cells, were analysed for the induction of senescence. Senescence associated ß-galactosidase activity and the expression of p16 and p21 were increased, whereas phosphorylated retinoblastoma protein was reduced. This senescent phenotype was mediated by 4-hydroxnonenal (4-HNE), a lipid peroxidation product secreted from foam cells; scavenging of 4-HNE in the co-culture medium blunted this effect. Furthermore, both foam cells and 4-HNE increased the expression of the pro-oxidant thioredoxin-interacting protein (TXNIP). Molecular manipulation of TXNIP expression confirmed its involvement in foam cell-induced senescence. Previous studies showed that peroxisome proliferator-activated receptor (PPAR)δ was activated by 4-hydroalkenals, such as 4-HNE. Pharmacological interventions supported the involvement of the 4-HNE-PPARδ axis in the induction of TXNIP and VEC senescence. The association of TXNIP with VEC senescence was further supported by immunofluorescent staining of human carotid plaques in which the expression of both TXNIP and p21 was augmented in endothelial cells. Collectively, these findings suggest that foam cell-released 4-HNE activates PPARδ in VEC, leading to increased TXNIP expression and consequently to senescence.


Asunto(s)
Aldehídos/farmacología , Proteínas Portadoras/metabolismo , Senescencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Células Espumosas/metabolismo , Animales , Biomarcadores/metabolismo , Bovinos , Línea Celular , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Células Espumosas/citología , Células Espumosas/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Humanos , Peroxidación de Lípido/efectos de los fármacos , Modelos Biológicos , PPAR delta/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
4.
Free Radic Biol Med ; 65: 978-987, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23973638

RESUMEN

Peroxidation of polyunsaturated fatty acids is intensified in cells subjected to oxidative stress and results in the generation of various bioactive compounds, of which 4-hydroxyalkenals are prominent. During the progression of type 2 diabetes mellitus, the ensuing hyperglycemia promotes the generation of reactive oxygen species (ROS) that contribute to the development of diabetic complications. It has been suggested that ROS-induced lipid peroxidation and the resulting 4-hydroxyalkenals markedly contribute to the development and progression of these pathologies. Recent findings, however, also suggest that noncytotoxic levels of 4-hydroxyalkenals play important signaling functions in the early phase of diabetes and act as hormetic factors to induce adaptive and protective responses in cells, enabling them to function in the hyperglycemic milieu. Our studies and others' have proposed such regulatory functions for 4-hydroxynonenal and 4-hydroxydodecadienal in insulin secreting ß-cells and vascular endothelial cells, respectively. This review presents and discusses the mechanisms regulating the generation of 4-hydroxyalkenals under high glucose conditions and the molecular interactions underlying the reciprocal transition from hormetic to cytotoxic agents.


Asunto(s)
Aldehídos/metabolismo , Peroxidación de Lípido , Transducción de Señal , Animales , Diabetes Mellitus Tipo 2/metabolismo , Progresión de la Enfermedad , Humanos , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
5.
Autophagy ; 9(4): 626-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23380813

RESUMEN

Pancreatic ß-cell dysfunction is central in diabetes. The diabetic milieu may impair proinsulin folding, leading to ß-cell endoplasmic reticulum (ER) stress and apoptosis, and thus a worsening of the diabetes. Autophagy is crucial for the well-being of the ß-cell; however, the impact of stimulating autophagy on ß-cell adaptation to ER stress is unknown. We studied the crosstalk between ER stress and autophagy in a rodent model of diabetes, called Akita, in which proinsulin gene mutation leads to protein misfolding and ß-cell demise. We found that proinsulin misfolding stimulates autophagy and, in symmetry, inhibition of autophagy induces ß-cell stress and apoptosis. Under conditions of excessive proinsulin misfolding, stimulation of autophagy by inhibiting MTORC1 alleviates stress and prevents apoptosis. Moreover, treatment of diabetic Akita mice with the MTORC1 inhibitor rapamycin improves diabetes and prevents ß-cell apoptosis. Thus, autophagy is a central adaptive mechanism in ß-cell stress. Stimulation of autophagy may become a novel therapeutic strategy in diabetes.


Asunto(s)
Autofagia , Diabetes Mellitus/patología , Estrés del Retículo Endoplásmico , Animales , Diabetes Mellitus/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Modelos Biológicos , Mutación/genética , Transducción de Señal
6.
Diabetes ; 62(4): 1227-37, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23274896

RESUMEN

Accumulation of misfolded proinsulin in the ß-cell leads to dysfunction induced by endoplasmic reticulum (ER) stress, with diabetes as a consequence. Autophagy helps cellular adaptation to stress via clearance of misfolded proteins and damaged organelles. We studied the effects of proinsulin misfolding on autophagy and the impact of stimulating autophagy on diabetes progression in Akita mice, which carry a mutation in proinsulin, leading to its severe misfolding. Treatment of female diabetic Akita mice with rapamycin improved diabetes, increased pancreatic insulin content, and prevented ß-cell apoptosis. In vitro, autophagic flux was increased in Akita ß-cells. Treatment with rapamycin further stimulated autophagy, evidenced by increased autophagosome formation and enhancement of autophagosome-lysosome fusion. This was associated with attenuation of cellular stress and apoptosis. The mammalian target of rapamycin (mTOR) kinase inhibitor Torin1 mimicked the rapamycin effects on autophagy and stress, indicating that the beneficial effects of rapamycin are indeed mediated via inhibition of mTOR. Finally, inhibition of autophagy exacerbated stress and abolished the anti-ER stress effects of rapamycin. In conclusion, rapamycin reduces ER stress induced by accumulation of misfolded proinsulin, thereby improving diabetes and preventing ß-cell apoptosis. The beneficial effects of rapamycin in this context strictly depend on autophagy; therefore, stimulating autophagy may become a therapeutic approach for diabetes.


Asunto(s)
Autofagia/efectos de los fármacos , Diabetes Mellitus/tratamiento farmacológico , Retículo Endoplásmico/fisiología , Inmunosupresores/uso terapéutico , Sirolimus/uso terapéutico , Animales , Autofagia/fisiología , Diabetes Mellitus/etiología , Femenino , Inmunosupresores/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Complejos Multiproteicos , Mutación , Naftiridinas/farmacología , Proinsulina/química , Proinsulina/genética , Proinsulina/metabolismo , Pliegue de Proteína , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Sirolimus/farmacología , Estrés Fisiológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
7.
Methods Mol Biol ; 933: 89-102, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22893403

RESUMEN

Insulin deficiency is the underlying cause of hyperglycemia in type 2 diabetes. The gerbil Psammomys obesus (P. obesus) is a naturally insulin resistant rodent with tendency to develop diet-induced hyperglycemia associated with obesity. P. obesus does not exhibit hyperglycemia in its natural desert habitat, feeding on low caloric vegetation. However, when fed regular laboratory chow containing higher caloric density, the animals develop moderate obesity and hyperglycemia. Diabetes development and progression is very fast in P. obesus. The animals reach the irreversible hypoinsulinemic stage of the disease, in which a marked reduction of ß-cell mass is apparent, within 4-6 weeks of high caloric diet. The present review describes the P. obesus of the Hebrew University colony, with emphasis on its use for the study of ß-cell dysfunction in type 2 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Dieta para Diabéticos , Modelos Animales de Enfermedad , Gerbillinae , Animales , Citocinas/inmunología , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Estrés Oxidativo , Ratas
8.
PLoS One ; 6(12): e28804, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22194917

RESUMEN

Thioredoxin-interacting protein (TXNIP) regulates critical biological processes including inflammation, stress and apoptosis. TXNIP is upregulated by glucose and is a critical mediator of hyperglycemia-induced beta-cell apoptosis in diabetes. In contrast, the saturated long-chain fatty acid palmitate, although toxic to the beta-cell, inhibits TXNIP expression. The mechanisms involved in the opposing effects of glucose and fatty acids on TXNIP expression are unknown. We found that both palmitate and oleate inhibited TXNIP in a rat beta-cell line and islets. Palmitate inhibition of TXNIP was independent of fatty acid beta-oxidation or esterification. AMP-activated protein kinase (AMPK) has an important role in cellular energy sensing and control of metabolic homeostasis; therefore we investigated its involvement in nutrient regulation of TXNIP. As expected, glucose inhibited whereas palmitate stimulated AMPK. Pharmacologic activators of AMPK mimicked fatty acids by inhibiting TXNIP. AMPK knockdown increased TXNIP expression in presence of high glucose with and without palmitate, indicating that nutrient (glucose and fatty acids) effects on TXNIP are mediated in part via modulation of AMPK activity. TXNIP is transcriptionally regulated by carbohydrate response element-binding protein (ChREBP). Palmitate inhibited glucose-stimulated ChREBP nuclear entry and recruitment to the Txnip promoter, thereby inhibiting Txnip transcription. We conclude that AMPK is an important regulator of Txnip transcription via modulation of ChREBP activity. The divergent effects of glucose and fatty acids on TXNIP expression result in part from their opposing effects on AMPK activity. In light of the important role of TXNIP in beta-cell apoptosis, its inhibition by fatty acids can be regarded as an adaptive/protective response to glucolipotoxicity. The finding that AMPK mediates nutrient regulation of TXNIP may have important implications for the pathophysiology and treatment of diabetes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Portadoras/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/enzimología , Ácido Oléico/farmacología , Ácido Palmítico/farmacología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proteínas de Ciclo Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Desoxiglucosa/farmacología , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/farmacología , Técnicas de Silenciamiento del Gen , Glucosa/farmacología , Humanos , Células Secretoras de Insulina/citología , Isoenzimas/metabolismo , Metformina/farmacología , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Wistar
9.
Diabetes ; 60(11): 2830-42, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21896929

RESUMEN

OBJECTIVE: Previous studies show that polyunsaturated fatty acids (PUFAs) increase the insulin secretory capacity of pancreatic ß-cells. We aimed at identifying PUFA-derived mediators and their cellular targets that are involved in the amplification of insulin release from ß-cells preexposed to high glucose levels. RESEARCH DESIGN AND METHODS: The content of fatty acids in phospholipids of INS-1E ß-cells was determined by lipidomics analysis. High-performance liquid chromatography was used to identify peroxidation products in ß-cell cultures. Static and dynamic glucose-stimulated insulin secretion (GSIS) assays were performed on isolated rat islets and/or INS-1E cells. The function of peroxisome proliferator-activated receptor-δ (PPAR-δ) in regulating insulin secretion was investigated using pharmacological agents and gene expression manipulations. RESULTS: High glucose activated cPLA(2) and, subsequently, the hydrolysis of arachidonic and linoleic acid (AA and LA, respectively) from phospholipids in INS-1E cells. Glucose also increased the level of reactive oxygen species, which promoted the peroxidation of these PUFAs to generate 4-hydroxy-2E-nonenal (4-HNE). The latter mimicked the GSIS-amplifying effect of high glucose preexposure and of the PPAR-δ agonist GW501516 in INS-1E cells and isolated rat islets. These effects were blocked with GSK0660, a selective PPAR-δ antagonist, and the antioxidant N-acetylcysteine or by silencing PPAR-δ expression. High glucose, 4-HNE, and GW501516 also induced luciferase expression in a PPAR-δ-mediated transactivation assay. Cytotoxic effects of 4-HNE were observed only above the physiologically effective concentration range. CONCLUSIONS: Elevated glucose levels augment the release of AA and LA from phospholipids and their peroxidation to 4-HNE in ß-cells. This molecule is an endogenous ligand for PPAR-δ, which amplifies insulin secretion in ß-cells.


Asunto(s)
Hiperglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Peroxidación de Lípido , PPAR delta/metabolismo , Transducción de Señal , Aldehídos/efectos adversos , Aldehídos/sangre , Aldehídos/metabolismo , Aldehídos/farmacología , Animales , Línea Celular , Diabetes Mellitus Tipo 2/sangre , Ácidos Grasos Insaturados/metabolismo , Silenciador del Gen , Gerbillinae , Fosfolipasas A2 Grupo IV/metabolismo , Humanos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , PPAR delta/agonistas , PPAR delta/antagonistas & inhibidores , PPAR delta/genética , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Técnicas de Cultivo de Tejidos
10.
J Diabetes Investig ; 2(2): 82-91, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-24843466

RESUMEN

Type 2 diabetic patients are insulin resistant as a result of obesity and a sedentary lifestyle. Nevertheless, it has been known for the past five decades that insulin response to nutrients is markedly diminished in type 2 diabetes. There is now a consensus that impaired glucose regulation cannot develop without insulin deficiency. First-phase insulin response to glucose is lost very early in the development of type 2 diabetes. Several prospective studies have shown that impaired insulin response to glucose is a predictor of future impaired glucose tolerance (IGT) and type 2 diabetes. Recently discovered type 2 diabetes-risk gene variants influence ß-cell function, and might represent the molecular basis for the low insulin secretion that predicts future type 2 diabetes. We believe type 2 diabetes develops on the basis of normal but 'weak'ß-cells unable to cope with excessive functional demands imposed by overnutrition and insulin resistance. Several laboratories have shown a reduction in ß-cell mass in type 2 diabetes and IGT, whereas others have found modest reductions and most importantly, a large overlap between ß-cell masses of diabetic and normoglycemic subjects. Therefore, at least initially, the ß-cell dysfunction of type 2 diabetes seems more functional than structural. However, type 2 diabetes is a progressive disorder, and animal models of diabetes show ß-cell apoptosis with prolonged hyperglycemia/hyperlipemia (glucolipotoxicity). ß-Cells exposed in vitro to glucolipotoxic conditions show endoplasmic reticulum (ER) and oxidative stress. ER stress mechanisms might participate in the adaptation of ß-cells to hyperglycemia, unless excessive. ß-Cells are not deficient in anti-oxidant defense, thioredoxin playing a major role. Its inhibitor, thioredoxin-interacting protein (TXNIP), might be important in leading to ß-cell apoptosis and type 2 diabetes. These topics are intensively investigated and might lead to novel therapeutic approaches. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2010.00094.x, 2011).

11.
PLoS One ; 4(3): e4954, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19305497

RESUMEN

BACKGROUND: Palmitate is a potent inducer of endoplasmic reticulum (ER) stress in beta-cells. In type 2 diabetes, glucose amplifies fatty-acid toxicity for pancreatic beta-cells, leading to beta-cell dysfunction and death. Why glucose exacerbates beta-cell lipotoxicity is largely unknown. Glucose stimulates mTORC1, an important nutrient sensor involved in the regulation of cellular stress. Our study tested the hypothesis that glucose augments lipotoxicity by stimulating mTORC1 leading to increased beta-cell ER stress. PRINCIPAL FINDINGS: We found that glucose amplifies palmitate-induced ER stress by increasing IRE1alpha protein levels and activating the JNK pathway, leading to increased beta-cell apoptosis. Moreover, glucose increased mTORC1 activity and its inhibition by rapamycin decreased beta-cell apoptosis under conditions of glucolipotoxicity. Inhibition of mTORC1 by rapamycin did not affect proinsulin and total protein synthesis in beta-cells incubated at high glucose with palmitate. However, it decreased IRE1alpha expression and signaling and inhibited JNK pathway activation. In TSC2-deficient mouse embryonic fibroblasts, in which mTORC1 is constitutively active, mTORC1 regulated the stimulation of JNK by ER stressors, but not in response to anisomycin, which activates JNK independent of ER stress. Finally, we found that JNK inhibition decreased beta-cell apoptosis under conditions of glucolipotoxicity. CONCLUSIONS/SIGNIFICANCE: Collectively, our findings suggest that mTORC1 mediates glucose amplification of lipotoxicity, acting through activation of ER stress and JNK. Thus, mTORC1 is an important transducer of ER stress in beta-cell glucolipotoxicity. Moreover, in stressed beta-cells mTORC1 inhibition decreases IRE1alpha protein expression and JNK activity without affecting ER protein load, suggesting that mTORC1 regulates the beta-cell stress response to glucose and fatty acids by modulating the synthesis and activity of specific proteins involved in the execution of the ER stress response. This novel paradigm may have important implications for understanding beta-cell failure in type 2 diabetes.


Asunto(s)
Retículo Endoplásmico/efectos de los fármacos , Glucosa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Palmitatos/farmacología , Factores de Transcripción/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Cicloheximida/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Retículo Endoplásmico/metabolismo , Activación Enzimática , Gerbillinae , Células Secretoras de Insulina/citología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas de la Membrana/metabolismo , Ratones , Complejos Multiproteicos , Estrés Oxidativo/fisiología , Palmitatos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas , Transducción de Señal/fisiología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factores de Transcripción/genética
12.
Front Biosci (Landmark Ed) ; 14(3): 1099-115, 2009 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-19273118

RESUMEN

The pancreatic beta-cell adapts to increased nutrient availability and insulin resistance by increasing its function and mass. These processes are orchestrated by signals derived from nutrient metabolism, hormones and cytokines. Their end-result is the regulation of insulin secretion and biosynthesis, and beta-cell proliferation and apoptosis. This review focuses on the mechanisms involved in beta-cell nutrient sensing and adaptation and the potential causes of beta-cell dysfunction and death in type 2 diabetes mellitus. Understanding the mechanisms that regulate adequate beta-cell adaptation and the natural history of beta-cell failure is of utmost importance for the development of novel disease modifying treatments.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Islotes Pancreáticos/patología , Animales , Islotes Pancreáticos/metabolismo , Proinsulina/biosíntesis , Proinsulina/metabolismo
13.
Obesity (Silver Spring) ; 16(3): 695-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18239599

RESUMEN

OBJECTIVE: To evaluate the accumulation pattern of intramyocellular lipids (IMCLs) in striated muscle during the development and progression of diabetes, using a novel scanning electron microscopic method. METHODS AND PROCEDURES: Hyperglycemia was induced by feeding diabetes-prone (DP) Psammomys obesus a high-energy (HE) diet. Lipid accumulation within gastrocnemius muscle fibers was assessed in formalin-fixed muscle samples during the development of hyperglycemia using high resolution imaging in a scanning electron microscope. We evaluated the temporal relationship between changes in IMCL quantity and morphology and the altered glucose metabolism and assessed the effect of reversal of hyperglycemia on IMCL level and morphology. Diabetes-resistant (DR) P. obesus served as controls. RESULTS: Lipid accumulation in the muscle fibers of DP animals was increased with the development of hyperglycemia. This was characterized by increased lipid density as well as by an abundance of large lipid droplets. Reversal of the phenotype resulted in the disappearance of large lipid droplets. The IMCL level and the distribution of lipid droplet size were similar in muscles of both the normoglycemic DR and DP animals, with an abundance of small lipid droplets. This profile was changed following a HE diet only in the DP animals. DISCUSSION: Lipid accumulation in the muscle of P. obesus during the development of hyperglycemia is characterized by increased quantity and accumulation of large lipid droplets. These changes were reversible upon normalization of blood glucose. The evaluated methodology is a useful tool for the study of the dynamics of lipid accumulation in different metabolic conditions.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Hiperglucemia/patología , Metabolismo de los Lípidos , Microscopía Electrónica de Rastreo , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/ultraestructura , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ingestión de Energía , Gerbillinae , Hiperglucemia/metabolismo , Insulina/sangre , Masculino , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Fenotipo , Factores de Tiempo
14.
Diabetes ; 57(4): 945-57, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18174523

RESUMEN

OBJECTIVE: Mammalian target of rapamycin (mTOR) and its downstream target S6 kinase 1 (S6K1) mediate nutrient-induced insulin resistance by downregulating insulin receptor substrate proteins with subsequent reduced Akt phosphorylation. Therefore, mTOR/S6K1 inhibition could become a therapeutic strategy in insulin-resistant states, including type 2 diabetes. We tested this hypothesis in the Psammomys obesus (P. obesus) model of nutrition-dependent type 2 diabetes, using the mTOR inhibitor rapamycin. RESEARCH DESIGN AND METHODS: Normoglycemic and diabetic P. obesus were treated with 0.2 mg x kg(-1) x day(-1) i.p. rapamycin or vehicle, and the effects on insulin signaling in muscle, liver and islets, and on different metabolic parameters were analyzed. RESULTS: Unexpectedly, rapamycin worsened hyperglycemia in diabetic P. obesus without affecting glycemia in normoglycemic controls. There was a 10-fold increase of serum insulin in diabetic P. obesus compared with controls; rapamycin completely abolished this increase. This was accompanied by weight loss and a robust increase of serum lipids and ketone bodies. Rapamycin decreased muscle insulin sensitivity paralleled by increased glycogen synthase kinase 3beta activity. In diabetic animals, rapamycin reduced beta-cell mass by 50% through increased apoptosis. Rapamycin increased the stress-responsive c-Jun NH(2)-terminal kinase pathway in muscle and islets, which could account for its effect on insulin resistance and beta-cell apoptosis. Moreover, glucose-stimulated insulin secretion and biosynthesis were impaired in islets treated with rapamycin. CONCLUSIONS: Rapamycin induces fulminant diabetes by increasing insulin resistance and reducing beta-cell function and mass. These findings emphasize the essential role of mTOR/S6K1 in orchestrating beta-cell adaptation to hyperglycemia in type 2 diabetes. It is likely that treatments based on mTOR inhibition will cause exacerbation of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Células Secretoras de Insulina/fisiología , Proteínas Quinasas/fisiología , Sirolimus/toxicidad , Animales , Diabetes Mellitus Tipo 2/inducido químicamente , Modelos Animales de Enfermedad , Gerbillinae , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR
15.
Diabetes ; 57(2): 279-87, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17977959

RESUMEN

OBJECTIVE: The Cohen diabetes-sensitive rat develops postprandial hyperglycemia when fed a high-sucrose, copper-poor diet, whereas the Cohen diabetes-resistant rat maintains normoglycemia. The pathophysiological basis of diabetes was studied in the Cohen diabetic rat centering on the interplay between the exocrine and endocrine compartments of the pancreas. RESEARCH DESIGN AND METHODS: Studies used male Cohen diabetes-sensitive and Cohen diabetes-resistant rats fed 1-month high-sucrose, copper-poor diet. Serum insulin and glucose levels were measured during glucose and insulin tolerance tests. The pancreas was evaluated for weight, insulin content, macrophage, and fat infiltration. Glucose-stimulated insulin secretion (GSIS) was determined in isolated perfused pancreas and in islets. RESULTS: Hyperglycemic Cohen diabetes-sensitive rats exhibited reduced pancreatic weight with lipid deposits and interleukin-1beta-positive macrophage infiltration in the exocrine pancreas. Islet morphology was preserved, and total pancreatic insulin content did not differ from that of Cohen diabetes-resistant rats. Lipids did not accumulate in skeletal muscle, nor was insulin resistance observed in hyperglycemic Cohen diabetes-sensitive rats. Intravenous glucose-tolerance test revealed markedly elevated glucose levels associated with diminished insulin output. Insulin release was induced in vivo by the non-nutrient secretagogues arginine and tolbutamide, suggesting a selective unresponsiveness to glucose. Decreased GSIS was observed in the isolated perfused pancreas of the hyperglycemic Cohen diabetes-sensitive rat, whereas islets isolated from these rats exhibited glucose-dependent insulin secretion and proinsulin biosynthesis. CONCLUSIONS: The association of the in vivo insulin secretory defect with lipid accumulation and activated macrophage infiltration in the exocrine pancreas suggests that changes in the islet microenvironment are the culprit in the insulin secretory malfunction observed in vivo.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Insulina/metabolismo , Páncreas/patología , Sacarosa/farmacología , Alimentación Animal , Animales , Glucemia/metabolismo , Catéteres de Permanencia , Cobre/deficiencia , Diabetes Mellitus Experimental/patología , Dieta para Diabéticos , Carbohidratos de la Dieta , Prueba de Tolerancia a la Glucosa , Hiperglucemia/fisiopatología , Insulina/sangre , Secreción de Insulina , Masculino , Páncreas/efectos de los fármacos , Periodo Posprandial , Ratas
16.
Physiol Genomics ; 29(2): 181-92, 2007 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-17213368

RESUMEN

We investigated the metabolic and genetic basis of diabetes in the Cohen Diabetic rat, a model of diet-induced diabetes, as a means to identify the molecular mechanisms involved. By altering individual components in the diabetogenic diet, we established that the dietary susceptibility that leads to the development of diabetes in this model is directly related to the high casein and low copper content in chow. The development of diabetes is accompanied by depletion of the acini from the exocrine pancreas and replacement with fat cells, while the appearance of the islets of Langerhans remains intact. With reversion back from diabetogenic to regular diet, the diabetic phenotype disappears but the histological changes in the exocrine pancreas prevail. Using positional cloning, we detected a major quantitative trait locus (QTL) on rat chromosome 4 with a chromosomal span of 4.9 cM, and two additional loci on chromosomes 7 and X. A screen for genes within that QTL in the rat and in the syntenic regions in mouse and man revealed only 23 candidate genes. Notable among these genes is Ica1, which has been causally associated with diabetes and bovine casein. We conclude that the development of diabetes in our model is dependent upon high casein and low copper in diet, that it is accompanied by histomorphological changes in the exocrine but not endocrine pancreas, that it is reversible, and that it is associated with a major QTL on chromosome 4 in which we detected Ica1, a high priority candidate gene.


Asunto(s)
Alimentación Animal/análisis , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Dieta , Modelos Animales de Enfermedad , Páncreas/patología , Sitios de Carácter Cuantitativo , Análisis de Varianza , Animales , Autoantígenos/genética , Caseínas/análisis , Mapeo Cromosómico , Cobre/análisis , Diabetes Mellitus Experimental/patología , Pruebas Genéticas , Escala de Lod , Ratas
17.
Endocrinology ; 147(11): 5110-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16916949

RESUMEN

Succinate stimulates insulin secretion and proinsulin biosynthesis. We studied the effects of reduced nicotinamide adenine dinucleotide phosphate (NADPH)-modulating pathways on glucose- and succinate-stimulated insulin secretion and proinsulin biosynthesis in the rat and the insulin-resistant Psammomys obesus. Disruption of the anaplerotic pyruvate/malate shuttle by phenylacetic acid inhibited glucose- and succinate-stimulated insulin secretion and succinate-stimulated proinsulin biosynthesis in both species. In contrast, phenylacetic acid failed to inhibit glucose-stimulated proinsulin biosynthesis in P. obesus islets. Inhibition of the NADPH-consuming enzyme neuronal nitric oxide synthase (nNOS) with l-N(G)-nitro-l-arginine methyl ester or with N(G)-monomethyl-l-arginine(G) doubled succinate-stimulated insulin secretion in rat islets, suggesting that succinate- and nNOS-derived signals interact to regulate insulin secretion. In contrast, nNOS inhibition had no effect on succinate-stimulated proinsulin biosynthesis in both species. In P. obesus islets, insulin secretion was not stimulated by succinate in the absence of glucose, whereas proinsulin biosynthesis was increased 5-fold. Conversely, under stimulating glucose levels, succinate doubled insulin secretion, indicating glucose-dependence. Pyruvate ester and inhibition of nNOS partially mimicked the permissive effect of glucose on succinate-stimulated insulin secretion, suggesting that anaplerosis-derived signals render the beta-cells responsive to succinate. We conclude that beta-cell anaplerosis via pyruvate carboxylase is important for glucose- and succinate-stimulated insulin secretion and for succinate-stimulated proinsulin biosynthesis. In P. obesus, pyruvate/malate shuttle dependent and independent pathways that regulate proinsulin biosynthesis coexist; the latter can maintain fuel stimulated biosynthetic activity when the succinate-dependent pathway is inhibited. nNOS signaling is a negative regulator of insulin secretion, but not of proinsulin biosynthesis.


Asunto(s)
Insulina/metabolismo , Proinsulina/biosíntesis , Ácido Succínico/farmacología , 1-Metil-3-Isobutilxantina/farmacología , Animales , Ciclo del Ácido Cítrico , AMP Cíclico/fisiología , Gerbillinae , Glucosa/farmacología , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Malatos/metabolismo , NADP/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Ácido Pirúvico/metabolismo , Ratas
18.
Diabetes ; 54 Suppl 2: S137-44, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16306331

RESUMEN

Type 2 diabetes is characterized by insulin resistance and progressive beta-cell failure. Deficient insulin secretion, with increased proportions of insulin precursor molecules, is a common feature of type 2 diabetes; this could result from inappropriate beta-cell function and/or reduced beta-cell mass. Most studies using tissues from diabetic patients are retrospective, providing only limited information on the relative contribution of beta-cell dysfunction versus decreased beta-cell mass to the "beta-cell failure" of type 2 diabetes. The gerbil Psammomys obesus is a good model to address questions related to the role of insulin resistance and beta-cell failure in nutritionally induced diabetes. Upon a change from its natural low-calorie diet to the calorie-rich laboratory food, P. obesus develops moderate obesity associated with postprandial hyperglycemia. Continued dietary load, superimposed on its innate insulin resistance, results in depletion of pancreatic insulin stores, with increased proportions of insulin precursor molecules in the pancreas and the blood. Inadequate response of the preproinsulin gene to the increased insulin needs is an important cause of diabetes progression. Changes in beta-cell mass do not correlate with pancreatic insulin stores and are unlikely to play a role in disease initiation and progression. The major culprit is the inappropriate insulin production with depletion of insulin stores as a consequence. Similar mechanisms could operate during the evolution of type 2 diabetes in humans.


Asunto(s)
Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Alimentación Animal , Animales , Dieta , Modelos Animales de Enfermedad , Ambiente , Gerbillinae , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo
19.
Circ Res ; 97(10): 1001-8, 2005 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-16210549

RESUMEN

Substrate autoregulation of glucose transporter-1 (GLUT-1) mRNA and protein expression provides vascular endothelial and smooth muscle cells a sensitive mechanism to adapt their rate of glucose transport in response to changing glycemic conditions. Hyperglycemia-induced downregulation of glucose transport is particularly important in protecting these cells against an excessive influx of glucose and consequently increased intracellular protein glycation and generation of free radicals; both are detrimental in the development of vascular disease in diabetes. We aimed to investigate the molecular mechanism of high glucose-induced downregulation of GLUT-1 mRNA expression in primary bovine aortic vascular endothelial (VEC) and smooth muscle (VSMC) cell cultures. Using RNA mobility shift, UV cross-linking, and in vitro degradation assays, followed by mass-spectrometric analysis, we identified calreticulin as a specific destabilizing trans-acting factor that binds to a 10-nucleotide cis-acting element (CAE(2181-2190)) in the 3'-untranslated region of GLUT-1 mRNA. Pure calreticulin accelerated the rate of GLUT-1 mRNA-probe degradation in vitro, whereas overexpression of calreticulin in vascular cells decreased significantly the total cell content of GLUT-1 mRNA and protein. The expression of calreticulin was augmented in vascular cells exposed to high glucose in comparison with low-glucose conditions. Similarly, increased expression of calreticulin was observed in aortae of diabetic Psammomys obesus in comparison with normoglycemic controls. These data suggest that CAE(2181-2190)-calreticulin complex, which is formed in VSMC and VEC exposed to hyperglycemic conditions, renders GLUT-1 mRNA susceptible to degradation. This interaction underlies the process of downregulation of glucose transport in vascular cells under high-glucose conditions.


Asunto(s)
Calreticulina/fisiología , Células Endoteliales/metabolismo , Transportador de Glucosa de Tipo 1/genética , Hiperglucemia/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Estabilidad del ARN , Regiones no Traducidas 3'/metabolismo , Animales , Bovinos , Células Cultivadas , Regulación hacia Abajo , Músculo Liso Vascular/citología , Óxido Nítrico/biosíntesis , ARN Mensajero/metabolismo
20.
Br J Pharmacol ; 146(6): 872-81, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16158072

RESUMEN

S 21403 (mitiglinide) is a new drug for type 2 diabetes mellitus (T2DM). Its action on insulin release and biosynthesis was investigated in several experimental systems utilizing pancreas from normal and T2DM animals. At high concentrations (10 microM), S 21403, like classical sulphonylurea, induced insulin release in the absence of glucose. In contrast, at therapeutic (0.1-1.0 microM) concentrations, S 21403 amplified insulin secretion glucose dose-dependently and with similar magnitude in normal and diabetic GK rat islets. In perfused GK rat pancreas, S 21403 induced normal kinetics of insulin secretion including first-phase response. The effect of S 21403 was strongly modulated by physiological factors. Thus, 0.1 microM adrenaline inhibited S 21403-induced insulin release. There was marked synergism between S 21403 and arginine in GK rat islets, combination of the two normalizing insulin secretion. In primary islet cultures from normal rats or prediabetic Psammomys obesus, prolonged exposure to S 21403 did not induce further depletion of insulin stores under normal or 'glucotoxic' conditions. Proinsulin biosynthesis was not affected by 2-h exposure of rat or prediabetic P. obesus islets to 1 microM S 21403. Yet, 24-h exposure of rat islets to S 21403 resulted in 30% increase in proinsulin biosynthesis at 8.3 mM glucose. Amplification by S 21403 of glucose-induced insulin secretion in diabetic GK beta-cells with restoration of first-phase response, a strong synergistic interaction with arginine and marked inhibition by adrenaline, make it a prime candidate for successful oral antidiabetic agent.


Asunto(s)
Indoles/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/biosíntesis , Insulina/metabolismo , Animales , Arginina/farmacología , Células Cultivadas , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Epinefrina/farmacología , Gerbillinae , Glucosa/metabolismo , Glucosa/farmacología , Hipoglucemiantes/farmacología , Indoles/antagonistas & inhibidores , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Isoindoles , Perfusión , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...