Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Nutr ; 153(3): 723-732, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36931751

RESUMEN

BACKGROUND: High dietary iron has been linked to an increased type 2 diabetes risk. We have previously shown that intrauterine growth restriction (IUGR) and feeding a Western diet (WD) to male Sprague-Dawley rats independently, as well as together, cause pancreatic islet inflammation, fibrosis, and hemosiderosis. OBJECTIVES: To investigate whether iron has a role in the pathogenesis of this inflammatory islet injury caused by IUGR and WD intake. METHODS: Male Sprague-Dawley offspring of bilateral uterine artery ligated (IUGR) and sham-operated (Sham) dams, fostered to nonoperated dams, were fed a WD [45% sucrose, 19.4% protein and 23% fat (w/w)] containing low iron (LI, 20 mg/kg) or high iron (HI, 500 mg/kg) from weaning. Four groups were studied: Sham-LI, Sham-HI, IUGR-LI, and IUGR-HI. Serial measurements of rat body weight, blood glucose, lipids and insulin, an intraperitoneal glucose tolerance test (age 13 wk), and histological analysis of pancreas and liver (age 14 wk) were recorded. The effects of iron, IUGR, and their interaction, on these measurements have been analyzed. RESULTS: WD with HI compared with LI caused an 11% greater weight gain by age 14 wk (P < 0.001), impaired glucose tolerance [AUC for glucose (G-AUC) 17% higher; P < 0.001), acute pancreatitis (17/18, HI; 6/17, LI; P < 0.001), pancreas-associated fat necrosis and saponification (7/18, HI; 0/17 LI; P < 0.01), and a trend to islet fibrotic injury (7/18, HI; 1/17 LI; P = 0.051). Although pancreatic and hepatic steatosis was evident in almost all WD-fed rats, pancreatic and hepatic iron accumulation was prevalent only in HI-fed rats (P < 0.0001 for both), being only mild in the livers. IUGR, independent of dietary iron, also caused impairment in glucose tolerance (G-AUC: 17% higher; P < 0.05). CONCLUSIONS: A postweaning WD containing HI, independent of IUGR, causes acute pancreatitis and islet injury in Sprague-Dawley rats suggesting a role of dietary iron in the development of steatopancreatitis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , Pancreatitis , Humanos , Femenino , Ratas , Animales , Masculino , Ratas Sprague-Dawley , Hierro de la Dieta , Diabetes Mellitus Tipo 2/metabolismo , Pancreatitis/etiología , Pancreatitis/metabolismo , Dieta Occidental , Enfermedad Aguda , Glucosa/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Islotes Pancreáticos/metabolismo , Hierro/metabolismo
2.
Diabetologia ; 65(6): 984-996, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35316840

RESUMEN

AIMS/HYPOTHESIS: Pancreatic beta cell dedifferentiation, transdifferentiation into other islet cells and apoptosis have been implicated in beta cell failure in type 2 diabetes, although the mechanisms are poorly defined. The endoplasmic reticulum stress response factor X-box binding protein 1 (XBP1) is a major regulator of the unfolded protein response. XBP1 expression is reduced in islets of people with type 2 diabetes, but its role in adult differentiated beta cells is unclear. Here, we assessed the effects of Xbp1 deletion in adult beta cells and tested whether XBP1-mediated unfolded protein response makes a necessary contribution to beta cell compensation in insulin resistance states. METHODS: Mice with inducible beta cell-specific Xbp1 deletion were studied under normal (chow diet) or metabolic stress (high-fat diet or obesity) conditions. Glucose tolerance, insulin secretion, islet gene expression, alpha cell mass, beta cell mass and apoptosis were assessed. Lineage tracing was used to determine beta cell fate. RESULTS: Deletion of Xbp1 in adult mouse beta cells led to beta cell dedifferentiation, beta-to-alpha cell transdifferentiation and increased alpha cell mass. Cell lineage-specific analyses revealed that Xbp1 deletion deactivated beta cell identity genes (insulin, Pdx1, Nkx6.1, Beta2, Foxo1) and derepressed beta cell dedifferentiation (Aldh1a3) and alpha cell (glucagon, Arx, Irx2) genes. Xbp1 deletion in beta cells of obese ob/ob or high-fat diet-fed mice triggered diabetes and worsened glucose intolerance by disrupting insulin secretory capacity. Furthermore, Xbp1 deletion increased beta cell apoptosis under metabolic stress conditions by attenuating the antioxidant response. CONCLUSIONS/INTERPRETATION: These findings indicate that XBP1 maintains beta cell identity, represses beta-to-alpha cell transdifferentiation and is required for beta cell compensation and prevention of diabetes in insulin resistance states.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Células Secretoras de Insulina , Proteína 1 de Unión a la X-Box/metabolismo , Animales , Transdiferenciación Celular/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Células Secretoras de Insulina/metabolismo , Ratones , Estrés Fisiológico , Proteína 1 de Unión a la X-Box/genética
3.
Metabolites ; 11(10)2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34677380

RESUMEN

High protein feeding has been shown to accelerate the development of type 1 diabetes in female non-obese diabetic (NOD) mice. Here, we investigated whether reducing systemic amino acid availability via knockout of the Slc6a19 gene encoding the system B(0) neutral amino acid transporter AT1 would reduce the incidence or delay the onset of type 1 diabetes in female NOD mice. Slc6a19 gene deficient NOD mice were generated using the CRISPR-Cas9 system which resulted in marked aminoaciduria. The incidence of diabetes by week 30 was 59.5% (22/37) and 69.0% (20/29) in NOD.Slc6a19+/+ and NOD.Slc6a19-/- mice, respectively (hazard ratio 0.77, 95% confidence interval 0.41-1.42; Mantel-Cox log rank test: p = 0.37). The median survival time without diabetes was 28 and 25 weeks for NOD.Slc6a19+/+ and NOD.Slc6a19-/- mice, respectively (ratio 1.1, 95% confidence interval 0.6-2.0). Histological analysis did not show differences in islet number or the degree of insulitis between wild type and Slc6a19 deficient NOD mice. We conclude that Slc6a19 deficiency does not prevent or delay the development of type 1 diabetes in female NOD mice.

4.
Front Endocrinol (Lausanne) ; 12: 799081, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069446

RESUMEN

Background: Maintenance of a normal fetal nutrient supply requires major adaptations in maternal metabolic physiology, including of the islet beta-cell. The role of lipid signaling processes in the mechanisms of islet beta-cell adaptation to pregnancy has been minimally investigated. Objective: To determine the effects of pregnancy on islet fatty acid (FA) metabolic partitioning and FA augmentation of glucose-stimulated insulin secretion (GSIS). Methods: Age matched virgin, early pregnant (gestational day-11, G11) and late pregnant (G19) Sprague-Dawley rats were studied. Fasted and fed state biochemistry, oral glucose tolerance tests (OGTT), and fasted and post-OGTT liver glycogen, were determined to assess in vivo metabolic characteristics. In isolated islets, FA (BSA-bound palmitate 0.25 mmol/l) augmentation of GSIS, FA partitioning into esterification and oxidation processes using metabolic tracer techniques, lipolysis by glycerol release, triacylglycerols (TG) content, and the expression of key beta-cell genes were determined. Results: Plasma glucose in pregnancy was lower, including during the OGTT (glucose area under the curve 0-120 min (AUC0-120); 655±24 versus 849±13 mmol.l-1.min; G19 vs virgin; P<0.0001), with plasma insulin concentrations equivalent to those of virgin rats (insulin AUC0-120; 97±7 versus 83±7 ng.ml-1.min; G19 vs virgin; not significant). Liver glycogen was depleted in fasted G19 rats with full recovery after oral glucose. Serum TG increased during pregnancy (4.4±0.4, 6.7±0.5; 17.1±1.5 mmol/l; virgin, G11, G19, P<0.0001), and islet TG content decreased (147±42, 172±27, 73±13 ng/µg protein; virgin, G11, G19; P<0.01). GSIS in isolated islets was increased in G19 compared to virgin rats, and this effect was augmented in the presence of FA. FA esterification into phospholipids, monoacylglycerols and TG were increased, whereas FA oxidation was reduced, in islets of pregnant compared to virgin rats, with variable effects on lipolysis dependent on gestational age. Expression of Ppargc1a, a key regulator of mitochondrial metabolism, was reduced by 51% in G11 and 64% in G19 pregnant rat islets compared to virgin rat islets (P<0.001). Conclusion: A lowered set-point for islet and hepatic glucose homeostasis in the pregnant rat has been confirmed. Islet adaptation to pregnancy includes increased FA esterification, reduced FA oxidation, and enhanced FA augmentation of glucose-stimulated insulin secretion.


Asunto(s)
Glucemia/metabolismo , Ácidos Grasos/metabolismo , Secreción de Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Hígado/metabolismo , Embarazo/metabolismo , Animales , Esterificación , Femenino , Prueba de Tolerancia a la Glucosa , Glicerol/metabolismo , Glucógeno/metabolismo , Lipólisis , Oxidación-Reducción , Embarazo/fisiología , Ratas , Transducción de Señal/fisiología , Triglicéridos/metabolismo
5.
Cells ; 9(7)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32630256

RESUMEN

The prevalence of obesity and obesity-related metabolic comorbidities are rapidly increasing worldwide, placing a huge economic burden on health systems. Excessive nutrient supply combined with reduced physical exercise results in positive energy balance that promotes adipose tissue expansion. However, the metabolic response and pattern of fat accumulation is variable, depending on the individual's genetic and acquired susceptibility factors. Some develop metabolically healthy obesity (MHO) and are resistant to obesity-associated metabolic diseases for some time, whereas others readily develop metabolically unhealthy obesity (MUO). An unhealthy response to excess fat accumulation could be due to susceptibility intrinsic factors (e.g., increased likelihood of dedifferentiation and/or inflammation), or by pathogenic drivers extrinsic to the adipose tissue (e.g., hyperinsulinemia), or a combination of both. This review outlines the major transcriptional factors and genes associated with adipogenesis and regulation of adipose tissue homeostasis and describes which of these are disrupted in MUO compared to MHO individuals. It also examines the potential role of pathogenic insulin hypersecretion as an extrinsic factor capable of driving the changes in adipose tissue which cause transition from MHO to MUO. On this basis, therapeutic approaches currently available and emerging to prevent and reverse the transition from MHO to MUO transition are reviewed.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Insulina/metabolismo , Síndrome Metabólico/metabolismo , Obesidad Metabólica Benigna/epidemiología , Obesidad/dietoterapia , Obesidad/metabolismo , Adipocitos/citología , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Adipogénesis/fisiología , Citocinas/metabolismo , Progresión de la Enfermedad , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Resistencia a la Insulina , Síndrome Metabólico/dietoterapia , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/terapia , Obesidad/tratamiento farmacológico , Obesidad/prevención & control , Obesidad Metabólica Benigna/metabolismo
6.
Nat Genet ; 48(5): 519-27, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26998692

RESUMEN

Type 1 (T1D) and type 2 (T2D) diabetes share pathophysiological characteristics, yet mechanistic links have remained elusive. T1D results from autoimmune destruction of pancreatic beta cells, whereas beta cell failure in T2D is delayed and progressive. Here we find a new genetic component of diabetes susceptibility in T1D non-obese diabetic (NOD) mice, identifying immune-independent beta cell fragility. Genetic variation in Xrcc4 and Glis3 alters the response of NOD beta cells to unfolded protein stress, enhancing the apoptotic and senescent fates. The same transcriptional relationships were observed in human islets, demonstrating the role of beta cell fragility in genetic predisposition to diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad , Células Secretoras de Insulina/patología , Animales , Apoptosis , Senescencia Celular , Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/patología , Dieta , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Pliegue de Proteína , Proteínas Represoras/genética , Factores Sexuales , Estrés Fisiológico , Transactivadores/genética
8.
Cell Metab ; 19(6): 993-1007, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24814481

RESUMEN

Glucose metabolism in pancreatic ß cells stimulates insulin granule exocytosis, and this process requires generation of a lipid signal. However, the signals involved in lipid amplification of glucose-stimulated insulin secretion (GSIS) are unknown. Here we show that in ß cells, glucose stimulates production of lipolysis-derived long-chain saturated monoacylglycerols, which further increase upon inhibition of the membrane-bound monoacylglycerol lipase α/ß-Hydrolase Domain-6 (ABHD6). ABHD6 expression in ß cells is inversely proportional to GSIS. Exogenous monoacylglycerols stimulate ß cell insulin secretion and restore GSIS suppressed by the pan-lipase inhibitor orlistat. Whole-body and ß-cell-specific ABHD6-KO mice exhibit enhanced GSIS, and their islets show elevated monoacylglycerol production and insulin secretion in response to glucose. Inhibition of ABHD6 in diabetic mice restores GSIS and improves glucose tolerance. Monoacylglycerol binds and activates the vesicle priming protein Munc13-1, thereby inducing insulin exocytosis. We propose saturated monoacylglycerol as a signal for GSIS and ABHD6 as a negative modulator of insulin secretion.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Monoacilglicerol Lipasas/biosíntesis , Monoglicéridos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Fármacos Antiobesidad/farmacología , Compuestos de Bifenilo/farmacología , Carbamatos/farmacología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Secreción de Insulina , Células Secretoras de Insulina , Lactonas/farmacología , Lipasa/antagonistas & inhibidores , Metabolismo de los Lípidos , Lipólisis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monoacilglicerol Lipasas/antagonistas & inhibidores , Monoacilglicerol Lipasas/genética , Monoglicéridos/biosíntesis , Monoglicéridos/farmacología , Orlistat , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Ratas Wistar , Receptores de Cannabinoides/metabolismo , Transducción de Señal
10.
Am J Pathol ; 184(5): 1446-57, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24631026

RESUMEN

Prenatal and postnatal factors such as intrauterine growth restriction (IUGR) and high-fat (HF) diet contribute to type 2 diabetes. Our aim was to determine whether IUGR and HF diets interact in type 2 diabetes pathogenesis, with particular attention focused on pancreatic islet morphology including assessment for inflammation. A surgical model of IUGR (bilateral uterine artery ligation) in Sprague-Dawley rats with sham controls was used. Pups were fed either HF or chow diets after weaning. Serial measures of body weight and glucose tolerance were performed. At 25 weeks of age, rat pancreases were harvested for histologic assessment. The birth weight of IUGR pups was 13% lower than that of sham pups. HF diet caused excess weight gain, dyslipidemia, hyperinsulinemia, and mild glucose intolerance, however, this was not aggravated further by IUGR. Markedly abnormal islet morphology was evident in 0 of 6 sham-chow, 5 of 8 sham-HF, 4 of 8 IUGR-chow, and 8 of 9 IUGR-HF rats (chi-square, P = 0.007). Abnormal islets were characterized by larger size, irregular shape, inflammation with CD68-positive cells, marked fibrosis, and hemosiderosis. ß-Cell mass was not altered by IUGR. In conclusion, HF and IUGR independently contribute to islet injury characterized by inflammation, hemosiderosis, and fibrosis. This suggests that both HF and IUGR can induce islet injury via converging pathways. The potential pathogenic or permissive role of iron in this process of islet inflammation warrants further investigation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Retardo del Crecimiento Fetal/patología , Hemosiderosis/complicaciones , Inflamación/complicaciones , Inflamación/patología , Islotes Pancreáticos/patología , Animales , Glucemia/metabolismo , Peso Corporal , Dislipidemias/complicaciones , Ayuno/sangre , Fibrosis , Hemosiderosis/patología , Proteínas de Homeodominio/metabolismo , Hiperinsulinismo/complicaciones , Islotes Pancreáticos/anomalías , Masculino , Tamaño de los Órganos , Ratas Sprague-Dawley , Transactivadores/metabolismo
11.
Liver Int ; 34(7): 1084-93, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24107103

RESUMEN

BACKGROUND & AIMS: Obese Alms1 mutant (foz/foz) NOD.B10 mice develop diabetes and fibrotic NASH when fed high-fat(HF) diet. To establish whether diabetes or obesity is more closely associated with NASH fibrosis, we compared diabetic foz/foz C57BL6/J with non-diabetic foz/foz BALB/c mice. We also determined hepatic cytokines, growth factors and related profibrotic pathways. METHODS: Male and female foz/foz BALB/c and C57BL6/J mice were fed HF or chow for 24 weeks before determining metabolic indices, liver injury, cytokines, growth factors, pathology/fibrosis and matrix deposition pathways. RESULTS: All foz/foz mice were obese. Hepatomegaly, hyperinsulinemia, hyperglycaemia and hypoadiponectinaemia occurred only in foz/foz C57BL6/J mice, whereas foz/foz BALB/c formed more adipose. Serum ALT, steatosis, ballooning, liver inflammation and NAFLD activity score were worse in C57BL6/J mice. In HF-fed mice, fibrosis was severe in foz/foz C57BL6/J, appreciable in WT C57BL6/J, but absent in foz/foz BALB/c mice. Hepatic mRNA expression of TNF-α, IL-12, IL-4, IL-10 was increased (but not IFN-γ, IL-1ß, IL-17A), and IL-4:IFN-γ ratio (indicating Th-2 predominance) was higher in HF-fed foz/foz C57BL6/J than BALB/c mice. In livers of HF-fed foz/foz C57BL6/J mice, TGF-ß was unaltered but PDGFα and CTGF were increased in association with enhanced α-SMA, CD147and MMP activity. CONCLUSIONS: In mice with equivalent genetic/dietary obesity, NASH development is linked to strain differences in hyperinsulinaemia and hyperglycaemia inversely related to lipid partitioning between adipose and liver. Diabetes-mediated CTGF-regulation of MMPs as well as cytokines/growth factors (Th-2 cytokine predominant, PDGFα, not TGF-ß) mobilized in the resultant hepatic necroinflammatory change may contribute to strain differences in NASH fibrosis.


Asunto(s)
Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 2/etiología , Dieta Alta en Grasa/efectos adversos , Cirrosis Hepática/etiología , Enfermedad del Hígado Graso no Alcohólico/etiología , Análisis de Varianza , Animales , Proteínas de Ciclo Celular , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Enfermedad del Hígado Graso no Alcohólico/patología , Especificidad de la Especie
12.
PLoS One ; 7(7): e40868, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22808281

RESUMEN

Type 2 diabetes is a metabolic disorder characterized by the inability of beta-cells to secrete enough insulin to maintain glucose homeostasis. MIN6 cells secrete insulin in response to glucose and other secretagogues, but high passage (HP) MIN6 cells lose their ability to secrete insulin in response to glucose. We hypothesized that metabolism of glucose and lipids were defective in HP MIN6 cells causing impaired glucose stimulated insulin secretion (GSIS). HP MIN6 cells had no first phase and impaired second phase GSIS indicative of global functional impairment. This was coupled with a markedly reduced ATP content at basal and glucose stimulated states. Glucose uptake and oxidation were higher at basal glucose but ATP content failed to increase with glucose. HP MIN6 cells had decreased basal lipid oxidation. This was accompanied by reduced expressions of Glut1, Gck, Pfk, Srebp1c, Ucp2, Sirt3, Nampt. MIN6 cells represent an important model of beta cells which, as passage numbers increased lost first phase but retained partial second phase GSIS, similar to patients early in type 2 diabetes onset. We believe a number of gene expression changes occurred to produce this defect, with emphasis on Sirt3 and Nampt, two genes that have been implicated in maintenance of glucose homeostasis.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Glucosa/metabolismo , Insulina/metabolismo , Metabolismo de los Lípidos , Adenosina Trifosfato/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Forma de la Célula , Regulación de la Expresión Génica , Secreción de Insulina , Espacio Intracelular/metabolismo , Ácido Láctico/metabolismo , Ratones , Modelos Biológicos , Oxidación-Reducción
13.
Am J Physiol Endocrinol Metab ; 302(2): E254-64, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22045312

RESUMEN

Physical activity improves glycemic control in type 2 diabetes (T2D), but its contribution to preserving ß-cell function is uncertain. We evaluated the role of physical activity on ß-cell secretory function and glycerolipid/fatty acid (GL/FA) cycling in male Zucker diabetic fatty (ZDF) rats. Six-week-old ZDF rats engaged in voluntary running for 6 wk (ZDF-A). Inactive Zucker lean and ZDF (ZDF-I) rats served as controls. ZDF-I rats displayed progressive hyperglycemia with ß-cell failure evidenced by falling insulinemia and reduced insulin secretion to oral glucose. Isolated ZDF-I rat islets showed reduced glucose-stimulated insulin secretion expressed per islet and per islet protein. They were also characterized by loss of the glucose regulation of fatty acid oxidation and GL/FA cycling, reduced mRNA expression of key ß-cell genes, and severe reduction of insulin stores. Physical activity prevented diabetes in ZDF rats through sustaining ß-cell compensation to insulin resistance shown in vivo and in vitro. Surprisingly, ZDF-A islets had persistent defects in fatty acid oxidation, GL/FA cycling, and ß-cell gene expression. ZDF-A islets, however, had preserved islet insulin mRNA and insulin stores compared with ZDF-I rats. Physical activity did not prevent hyperphagia, dyslipidemia, or obesity in ZDF rats. In conclusion, islets of ZDF rats have a susceptibility to failure that is possibly due to altered ß-cell fatty acid metabolism. Depletion of pancreatic islet insulin stores is a major contributor to islet failure in this T2D model, preventable by physical activity.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Dislipidemias/fisiopatología , Ácidos Grasos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Condicionamiento Físico Animal/fisiología , Hormona Adrenocorticotrópica/sangre , Animales , Peso Corporal/fisiología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/genética , Dislipidemias/metabolismo , Ingestión de Alimentos/fisiología , Péptido 1 Similar al Glucagón/sangre , Resistencia a la Insulina/fisiología , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Ratas , Ratas Zucker
14.
J Clin Invest ; 121(8): 3331-42, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21747171

RESUMEN

The failure of pancreatic ß cells to adapt to an increasing demand for insulin is the major mechanism by which patients progress from insulin resistance to type 2 diabetes (T2D) and is thought to be related to dysfunctional lipid homeostasis within those cells. In multiple animal models of diabetes, females demonstrate relative protection from ß cell failure. We previously found that the hormone 17ß-estradiol (E2) in part mediates this benefit. Here, we show that treating male Zucker diabetic fatty (ZDF) rats with E2 suppressed synthesis and accumulation of fatty acids and glycerolipids in islets and protected against ß cell failure. The antilipogenic actions of E2 were recapitulated by pharmacological activation of estrogen receptor α (ERα) or ERß in a rat ß cell line and in cultured ZDF rat, mouse, and human islets. Pancreas-specific null deletion of ERα in mice (PERα-/-) prevented reduction of lipid synthesis by E2 via a direct action in islets, and PERα-/- mice were predisposed to islet lipid accumulation and ß cell dysfunction in response to feeding with a high-fat diet. ER activation inhibited ß cell lipid synthesis by suppressing the expression (and activity) of fatty acid synthase via a nonclassical pathway dependent on activated Stat3. Accordingly, pancreas-specific deletion of Stat3 in mice curtailed ER-mediated suppression of lipid synthesis. These data suggest that extranuclear ERs may be promising therapeutic targets to prevent ß cell failure in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Lípidos/química , Receptores de Estrógenos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Modelos Animales de Enfermedad , Ácido Graso Sintasas/metabolismo , Eliminación de Gen , Humanos , Masculino , Ratones , Ratas , Ratas Zucker
15.
J Diabetes ; 2(3): 157-67, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20923481

RESUMEN

BACKGROUND: Hyperinsulinemia associated with non-ketotic hypoglycemia is observed in patients with mutated ß-oxidation enzyme short-chain 3-hydroxyacyl-CoA dehydrogenase (HADHSC). In the present study, we investigated the mechanism underlying HADHSC-mediated regulation of insulin secretion. METHODS: Knockdown of HADHSC expression by RNA interference in INS832/13 ß-cells was achieved using short hairpin RNA and short interference RNA. RESULTS: Knockdown of HADHSC increased both fuel- (glucose or leucine plus glutamine) and non-fuel (high KCl)-induced insulin secretion. Enhanced glucose-stimulated insulin secretion (GSIS) induced by HADHSC knockdown was independent of changes in cytosolic Ca(2+) and also occurred in the presence of fatty acids. L-Carnitine, used in the formation of acyl-carnitine compounds, increased GSIS in control cells, but was unable to further increase the augmented GSIS in HADHSC-knockdown cells. The pan transaminase inhibitor amino-oxyacetate reversed HADHSC knockdown-mediated increases in GSIS. Oxidation of [1-(14) C]-palmitate and -octanoate was not reduced in HADHSC-knockdown cells. L-3-Hydroxybutyryl-carnitine (tested using its precursor L-3-hydroxybutyrate) and L-3-hydroxyglutarate, which accumulate in blood and urine, respectively, of HADHSC-deficient patients, did not change insulin secretion. CONCLUSIONS: Insulin secretion promoted by both fuel and non-fuel stimuli is negatively regulated by HADHSC. Enhanced secretion after HADHSC knockdown is not due to inhibition of fatty acid oxidation causing an accumulation of long-chain fatty acids or their CoA derivatives. L-3-Hydroxybutyrate and L-3-hydroxyglutarate do not mediate enhanced secretion caused by reduced HADHSC activity. Transamination reaction(s) and the formation of short-chain acylcarnitines and CoAs may be implicated in the mechanism whereby HADHSC deficiency results in enhanced insulin secretion and hyperinsulinemia.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Adenosina Trifosfato/metabolismo , Secuencia de Bases , Calcio/metabolismo , Células Cultivadas/citología , Células Cultivadas/metabolismo , Regulación hacia Abajo , Homeostasis , Humanos , Hiperinsulinismo/fisiopatología , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/enzimología , Reacción en Cadena de la Polimerasa/métodos , ARN Interferente Pequeño/genética
16.
Appl Physiol Nutr Metab ; 32(2): 241-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17486165

RESUMEN

Deteriorating islet beta-cell function is key in the progression of an impaired glucose tolerance state to overt type 2 diabetes (T2D), a transition that can be delayed by exercise. We have previously shown that trained rats are protected from heart ischemia-reperfusion injury in correlation with an increase in cardiac tissue fatty-acid oxidation. This trained metabolic phenotype, if induced in the islet, could also prevent beta-cell failure in the pathogenesis of T2D. To assess the effect of training on islet lipid metabolism and insulin secretion, female Sprague-Dawley rats were exercised on a treadmill for 90 min/d, 4 d/week, for 10 weeks. Islet fatty-acid oxidation, the expression of key lipid metabolism genes, and glucose-stimulated insulin secretion were determined in freshly isolated islets from trained and sedentary control rats after a 48 h rest period from the last exercise. Although this moderate training reduced plasma glycerol, free fatty acids, and triglyceride levels by about 40%, consistent with reduced lipolysis from adipose tissue, it did not alter islet fatty-acid oxidation, nor the islet expression of key transcription factors and enzymes of lipid metabolism. The training also had no effect on glucose-stimulated insulin secretion or its amplification by free fatty acids. In summary, chronic exercise training did not cause an intrinsic change in islet lipid metabolism. Training did, however, substantially reduce the exposure of islets to exogenous lipid, thereby providing a potential mechanism by which exercise can prevent islet beta-cell failure leading to T2D.


Asunto(s)
Secuencia de Aminoácidos/genética , Insulina/sangre , Islotes Pancreáticos/metabolismo , Metabolismo de los Lípidos/genética , Lípidos/sangre , Condicionamiento Físico Animal , Análisis de Varianza , Animales , Ácidos Grasos no Esterificados , Femenino , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley
17.
Diabetes ; 55 Suppl 2: S16-23, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17130640

RESUMEN

Fatty acids (FAs) and other lipid molecules are important for many cellular functions, including vesicle exocytosis. For the pancreatic beta-cell, while the presence of some FAs is essential for glucose-stimulated insulin secretion, FAs have enormous capacity to amplify glucose-stimulated insulin secretion, which is particularly operative in situations of beta-cell compensation for insulin resistance. In this review, we propose that FAs do this via three interdependent processes, which we have assigned to a "trident model" of beta-cell lipid signaling. The first two arms of the model implicate intracellular metabolism of FAs, whereas the third is related to membrane free fatty acid receptor (FFAR) activation. The first arm involves the AMP-activated protein kinase/malonyl-CoA/long-chain acyl-CoA (LC-CoA) signaling network in which glucose, together with other anaplerotic fuels, increases cytosolic malonyl-CoA, which inhibits FA partitioning into oxidation, thus increasing the availability of LC-CoA for signaling purposes. The second involves glucose-responsive triglyceride (TG)/free fatty acid (FFA) cycling. In this pathway, glucose promotes LC-CoA esterification to complex lipids such as TG and diacylglycerol, concomitant with glucose stimulation of lipolysis of the esterification products, with renewal of the intracellular FFA pool for reactivation to LC-CoA. The third arm involves FFA stimulation of the G-protein-coupled receptor GPR40/FFAR1, which results in enhancement of glucose-stimulated accumulation of cytosolic Ca2+ and consequently insulin secretion. It is possible that FFA released by the lipolysis arm of TG/FFA cycling is partly "secreted" and, via an autocrine/paracrine mechanism, is additive to exogenous FFAs in activating the FFAR1 pathway. Glucose-stimulated release of arachidonic acid from phospholipids by calcium-independent phospholipase A2 and/or from TG/FFA cycling may also be involved. Improved knowledge of lipid signaling in the beta-cell will allow a better understanding of the mechanisms of beta-cell compensation and failure in diabetes.


Asunto(s)
Ácidos Grasos no Esterificados/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Acilcoenzima A/fisiología , Animales , Humanos , Secreción de Insulina , Malonil Coenzima A/fisiología , Modelos Biológicos , Fosfolipasas A/fisiología , Fosfolipasas A2 , Transducción de Señal/fisiología
18.
Endocrinology ; 146(1): 375-82, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15459119

RESUMEN

The cellular response to fasting and starvation in tissues such as heart, skeletal muscle, and liver requires peroxisome proliferator-activated receptor-alpha (PPARalpha)-dependent up-regulation of energy metabolism toward fatty acid oxidation (FAO). PPARalpha null (PPARalphaKO) mice develop hyperinsulinemic hypoglycemia in the fasting state, and we previously showed that PPARalpha expression is increased in islets at low glucose. On this basis, we hypothesized that enhanced PPARalpha expression and FAO, via depletion of lipid-signaling molecule(s) for insulin exocytosis, are also involved in the normal adaptive response of the islet to fasting. Fasted PPARalphaKO mice compared with wild-type mice had supranormal ip glucose tolerance due to increased plasma insulin levels. Isolated islets from the PPARalpha null mice had a 44% reduction in FAO, normal glucose use and oxidation, and enhanced glucose-induced insulin secretion. In normal rats, fasting for 24 h increased islet PPARalpha, carnitine palmitoyltransferase 1, and uncoupling protein-2 mRNA expression by 60%, 62%, and 82%, respectively. The data are consistent with the view that PPARalpha, via transcriptionally up-regulating islet FAO, can reduce insulin secretion, and that this mechanism is involved in the normal physiological response of the pancreatic islet to fasting such that hypoglycemia is avoided.


Asunto(s)
Adaptación Fisiológica , Ayuno/fisiología , Ácidos Grasos/metabolismo , Islotes Pancreáticos/fisiología , PPAR alfa/fisiología , Transcripción Genética , Regulación hacia Arriba , Animales , Expresión Génica/fisiología , Glucosa/metabolismo , Glucosa/fisiología , Prueba de Tolerancia a la Glucosa , Hormonas/metabolismo , Insulina/sangre , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Oxidación-Reducción , PPAR alfa/deficiencia , PPAR alfa/metabolismo , Ratas , Ratas Wistar
19.
Mol Cell Endocrinol ; 220(1-2): 41-50, 2004 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-15196698

RESUMEN

The effects of prolactin (PRL) on transcript profile expression in 24h cultured pancreatic adult rat islets were investigated by cDNA expression array analysis to identify possible candidate mRNA species that encode proteins involved in the maturation and growth of the endocrine pancreas. The expression of 54 out of 588 genes was altered by treatment with PRL. The differentially expressed transcripts identified were distributed in six main categories involved in cell proliferation and differentiation, namely, cell cycle regulation, signal transduction, transcription factors and coactivators, translational machinery, Ca(2+)-mediated exocytosis, and immuno-response. Treatment with PRL also reduced the expression of genes related to apoptosis. Several genes, whose expression was previously not known to be modulated by PRL were also identified including macrophage migration inhibitory factor and Ca(2+)/calmodulin-dependent protein kinase IV. These genes have recently been shown to play a crucial role in insulin secretion and insulin gene expression, respectively. Treatment with PRL also modified the expression of AKT2 and bone morphogenetic protein receptor 1A that control glucose homeostasis and directly affect the behavior of endocrine pancreas and/or the sensitivity of target tissues to insulin. In conclusion, PRL induces several patterns of gene expression in pancreatic islet cells. The analysis of these different patterns will be useful for understanding the complex mechanism of action of PRL in the maturation and differentiation of pancreatic islets.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Prolactina/farmacología , Animales , Western Blotting , Células Cultivadas , Femenino , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/análisis , ARN Mensajero/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Diabetes ; 53(4): 1007-19, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15047616

RESUMEN

The malonyl-CoA/long-chain acyl-CoA (LC-CoA) model of glucose-induced insulin secretion (GIIS) predicts that malonyl-CoA derived from glucose metabolism inhibits fatty acid oxidation, thereby increasing the availability of LC-CoA for lipid signaling to cellular processes involved in exocytosis. For directly testing the model, INSr3 cell clones overexpressing malonyl-CoA decarboxylase in the cytosol (MCDc) in a tetracycline regulatable manner were generated, and INS(832/13) and rat islets were infected with MCDc-expressing adenoviruses. MCD activity was increased more than fivefold, and the malonyl-CoA content was markedly diminished. This was associated with enhanced fat oxidation at high glucose, a suppression of the glucose-induced increase in cellular free fatty acid (FFA) content, and reduced partitioning at elevated glucose of exogenous palmitate into lipid esterification products. MCDc overexpression, in the presence of exogenous FFAs but not in their absence, reduced GIIS in all beta-cell lines and in rat islets. It also markedly curtailed the stimulation of insulin secretion by other fuel and nonfuel secretagogues. In the absence of MCDc overexpression, the secretory responses to all types of secretagogues were amplified by the provision of exogenous fatty acids. In the presence of exogenous FFAs, the fatty acyl-CoA synthetase inhibitor triacsin C reduced secretion in response to glucose and nonfuel stimuli. The data show the existence of important links between the metabolic coupling factor malonyl-CoA, the partitioning of fatty acids, and the stimulation of insulin secretion to both fuel and nonfuel stimuli.


Asunto(s)
Acilcoenzima A/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Lípidos/fisiología , Malonil Coenzima A/metabolismo , Transducción de Señal/fisiología , Animales , Carboxiliasas/genética , Carboxiliasas/metabolismo , Células Clonales , Glucosa/farmacología , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Modelos Biológicos , Proteínas Recombinantes/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...