Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Lipid Res ; : 100587, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38950680

RESUMEN

Lipotoxicity has been considered the main cause of pancreatic beta-cell failure during type 2 diabetes development. Lipid droplets (LD) are believed to regulate the beta-cell sensitivity to free fatty acids (FFA), but the underlying molecular mechanisms are largely unclear. Accumulating evidence points, however, to an important role of intracellular sphingosine-1-phosphate (S1P) metabolism in lipotoxicity-mediated disturbances of beta-cell function. In the present study, we compared the effects of an increased irreversible S1P degradation (S1P-lyase, SPL overexpression) with those associated with an enhanced S1P recycling (overexpression of S1P phosphatase 1, SGPP1) on LD formation and lipotoxicity in rat INS1E beta-cells. Interestingly, although both approaches led to a reduced S1P concentration, they had opposite effects on the susceptibility to FFA. Overexpression of SGPP1 prevented FFA-mediated caspase-3 activation by a mechanism involving an enhanced lipid storage capacity and prevention of oxidative stress. In contrast, SPL overexpression limited lipid droplet biogenesis, content and size, while accelerating lipophagy. This was associated with FFA-induced hydrogen peroxide formation, mitochondrial fragmentation and dysfunction, as well as ER stress. These changes coincided with upregulation of proapoptotic ceramides, but were independent of lipid peroxidation rate. Also in human EndoC-ßH1 beta-cells suppression of SPL with simultaneous overexpression of SGPP1 led to a similar and even more pronounced LD phenotype as that in INS1E-SGPP1 cells. Thus, intracellular S1P turnover significantly regulates LD content and size, and influences beta-cell sensitivity to FFA.

2.
Arch Med Sci ; 19(4): 895-911, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37560741

RESUMEN

The immune system is frequently described in the context of its protective function against infections and its role in the development of autoimmunity. For more than a decade, the interactions between the immune system and metabolic processes have been reported, in effect creating a new research field, termed immunometabolism. Accumulating evidence supports the hypothesis that the development of metabolic diseases may be linked to inflammation, and reflects, in some cases, the activation of immune responses. As such, immunometabolism is defined by 1) inflammation as a driver of disease development and/or 2) metabolic processes stimulating cellular differentiation of the immune components. In this review, the main factors capable of altering the immuno-metabolic communication leading to the development and establishment of obesity and diabetes are comprehensively presented. Tissue-specific immune responses suggested to impair metabolic processes are described, with an emphasis on the adipose tissue, gut, muscle, liver, and pancreas.

3.
Diabetologia ; 65(5): 829-843, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35122482

RESUMEN

AIMS/HYPOTHESIS: The aim of this study was to examine the effects of proinflammatory cytokines on cells of different developmental stages during the generation of stem cell-derived beta cells (SC-beta cells) from human pluripotent stem cells (hPSCs). We wanted to find out to what extent human SC-beta cells are suitable as an experimental cellular model and, with regard to a possible therapeutic use, whether SC-beta cells have a comparable vulnerability to cytokines as bona fide beta cells. METHODS: hPSCs were differentiated towards pancreatic organoids (SC-organoids) using a 3D production protocol. SC-beta cells and non-insulin-producing cells were separated by FACS and differential gene expression profiles of purified human SC-beta cells, progenitor stages and the human beta cell line EndoC-ßH1, as a reference, were determined after 24 h incubation with the proinflammatory cytokines IL-1ß, TNF-α and IFN-γ via a transcriptome microarray. Furthermore, we investigated apoptosis based on caspase cleavage, the generation of reactive oxygen species and activation of mitogen-activated protein-kinase (MAPK) stress-signalling pathways. RESULTS: A 24 h exposure of SC-beta cells to proinflammatory cytokines resulted in significant activation of caspase 3/7 and apoptosis via the extrinsic and intrinsic apoptosis signalling pathways. At this time point, SC-beta cells showed a markedly higher sensitivity towards proinflammatory cytokines than non-insulin-producing cells and EndoC-ßH1 cells. Furthermore, we were able to demonstrate the generation of reactive oxygen species and rule out the involvement of NO-mediated stress. A transient activation of stress-signalling pathways p38 mitogen-activated protein kinases (p38) and c-Jun N-terminal kinase (JNK) was already observed after 10 min of cytokine exposure. The transcriptome analysis revealed that the cellular response to proinflammatory cytokines increased with the degree of differentiation of the cells. Cytokines induced the expression of multiple inflammatory mediators including IL-32, CXCL9 and CXCL10 in SC-beta cells and in non-insulin-producing cells. CONCLUSIONS/INTERPRETATION: Our results indicate that human SC-beta cells respond to proinflammatory cytokines very similarly to human islets. Due to the fast and fulminant cellular response of SC-beta cells, we conclude that SC-beta cells represent a suitable model for diabetes research. In light of the immaturity of SC-beta cells, they may be an attractive model for developmentally young beta cells as they are, for example, present in patients with early-onset type 1 diabetes. The secretion of chemotactic signals may promote communication between SC-beta cells and immune cells, and non-insulin-producing cells possibly participate in the overall immune response and are thus capable of amplifying the immune response and further stimulating inflammation. We demonstrated that cytokine-treated SC-organoids secrete IL-32, which is considered a promising candidate for type 1 diabetes onset. This underlines the need to ensure the survival of SC-beta cells in an autoimmune environment such as that found in type 1 diabetes.


Asunto(s)
Citocinas , Diabetes Mellitus Tipo 1 , Inflamación , Células Secretoras de Insulina , Células Madre Pluripotentes , Apoptosis , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Inflamación/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucinas , Óxido Nítrico/metabolismo , Células Madre Pluripotentes/metabolismo , Especies Reactivas de Oxígeno/metabolismo
4.
Int J Mol Sci ; 23(3)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35163559

RESUMEN

Sphingosine-1 phosphate (S1P) is a bioactive sphingolipid with multiple functions conveyed by the activation of cell surface receptors and/or intracellular mediators. A growing body of evidence indicates its important role in pancreatic insulin-secreting beta-cells that are necessary for maintenance of glucose homeostasis. The dysfunction and/or death of beta-cells lead to diabetes development. Diabetes is a serious public health burden with incidence growing rapidly in recent decades. The two major types of diabetes are the autoimmune-mediated type 1 diabetes (T1DM) and the metabolic stress-related type 2 diabetes (T2DM). Despite many differences in the development, both types of diabetes are characterized by chronic hyperglycemia and inflammation. The inflammatory component of diabetes remains under-characterized. Recent years have brought new insights into the possible mechanism involved in the increased inflammatory response, suggesting that environmental factors such as a westernized diet may participate in this process. Dietary lipids, particularly palmitate, are substrates for the biosynthesis of bioactive sphingolipids. Disturbed serum sphingolipid profiles were observed in both T1DM and T2DM patients. Many polymorphisms were identified in genes encoding enzymes of the sphingolipid pathway, including sphingosine kinase 2 (SK2), the S1P generating enzyme which is highly expressed in beta-cells. Proinflammatory cytokines and free fatty acids have been shown to modulate the expression and activity of S1P-generating and S1P-catabolizing enzymes. In this review, the similarities and differences in the action of extracellular and intracellular S1P in beta-cells exposed to cytokines or free fatty acids will be identified and the outlook for future research will be discussed.


Asunto(s)
Citocinas/metabolismo , Ácidos Grasos/metabolismo , Células Secretoras de Insulina/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Polimorfismo de Nucleótido Simple , Esfingosina/metabolismo
5.
Int J Mol Sci ; 22(19)2021 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-34639233

RESUMEN

Elevated levels of free fatty acids (FFAs) have been related to pancreatic beta-cell failure in type 2 diabetes (T2DM), though the underlying mechanisms are not yet fully understood. FFAs have been shown to dysregulate formation of bioactive sphingolipids, such as ceramides and sphingosine-1 phosphate (S1P) in beta-cells. The aim of this study was to analyze the role of sphingosine-1 phosphate lyase (SPL), a key enzyme of the sphingolipid pathway that catalyzes an irreversible degradation of S1P, in the sensitivity of beta-cells to lipotoxicity. To validate the role of SPL in lipotoxicity, we modulated SPL expression in rat INS1E cells and in human EndoC-ßH1 beta-cells. SPL overexpression in INS1E cells (INS1E-SPL), which are characterized by a moderate basal expression level of SPL, resulted in an acceleration of palmitate-mediated cell viability loss, proliferation inhibition and induction of oxidative stress. SPL overexpression affected the mRNA expression of ER stress markers and mitochondrial chaperones. In contrast to control cells, in INS1E-SPL cells no protective effect of oleate was detected. Moreover, Plin2 expression and lipid droplet formation were strongly reduced in OA-treated INS1E-SPL cells. Silencing of SPL in human EndoC-ßH1 beta-cells, which are characterized by a significantly higher SPL expression as compared to rodent beta-cells, resulted in prevention of FFA-mediated caspase-3/7 activation. Our findings indicate that an adequate control of S1P degradation by SPL might be crucially involved in the susceptibility of pancreatic beta-cells to lipotoxicity.


Asunto(s)
Aldehído-Liasas/metabolismo , Ácidos Grasos no Esterificados/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Lisofosfolípidos/metabolismo , Estrés Oxidativo , Esfingosina/análogos & derivados , Aldehído-Liasas/genética , Animales , Supervivencia Celular , Humanos , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Ratas , Esfingosina/metabolismo
6.
Biochim Biophys Acta Mol Basis Dis ; 1867(10): 166199, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34144091

RESUMEN

During diabetes development insulin production and glucose-stimulated insulin secretion (GSIS) are defective due to inflammation-related, yet not fully understood mechanisms. MCPIP1 (monocyte chemotactic protein-induced protein-1) is a strong regulator of inflammation, and acts predominantly as a specific RNase. The impact of MCPIP1 on insulin secretory capacity is unknown. We show that the expression of the ZC3H12A gene, which encodes MCPIP1, was induced by T1DM- and by T2DM-simulating conditions, with a stronger effect of cytokines. The number of MCPIP1-positive pancreatic islet-cells, including beta-cells, was significantly higher in diabetic compared to nondiabetic individuals. In the 3'UTR regions of mRNAs coding for Pdx1 (pancreatic and duodenal homeobox 1), FoxO1 (forkhead box protein O1), and of a novel regulator of insulin handling, Grp94 (glucose-regulated protein 94), MCPIP1-target structures were detected. Overexpression of the wild type MCPIP1wt, but not of the mutant MCPIP1D141N (lacking the RNase activity), decreased the expression of genes involved in insulin production and GSIS. Additionally INS1-E-MCPIP1wt cells exhibited a higher Ire1 (inositol-requiring enzyme 1) expression. MCPIP1wt overexpression blunted GSIS and glucose-mediated calcium influx with no deleterious effects on glucose uptake or glucokinase activity. We identify MCPIP1 as a new common link between diabetogenic conditions and beta-cell failure. MCPIP1 may serve as an interesting target for novel beta-cell protective approaches.


Asunto(s)
Diabetes Mellitus/metabolismo , Secreción de Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Regiones no Traducidas 3'/fisiología , Animales , Calcio/metabolismo , Línea Celular , Citocinas/metabolismo , Diabetes Mellitus/patología , Proteína Forkhead Box O1/metabolismo , Glucosa/metabolismo , Humanos , Células Secretoras de Insulina/patología , ARN Mensajero/metabolismo , Ratas
7.
Cells ; 9(8)2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32759843

RESUMEN

Type 1 diabetes (T1DM) is a chronic autoimmune disease, with a strong genetic background, leading to a gradual loss of pancreatic beta-cells, which secrete insulin and control glucose homeostasis. Patients with T1DM require life-long substitution with insulin and are at high risk for development of severe secondary complications. The incidence of T1DM has been continuously growing in the last decades, indicating an important contribution of environmental factors. Accumulating data indicates that sphingolipids may be crucially involved in T1DM development. The serum lipidome of T1DM patients is characterized by significantly altered sphingolipid composition compared to nondiabetic, healthy probands. Recently, several polymorphisms in the genes encoding the enzymatic machinery for sphingolipid production have been identified in T1DM individuals. Evidence gained from studies in rodent islets and beta-cells exposed to cytokines indicates dysregulation of the sphingolipid biosynthetic pathway and impaired function of several sphingolipids. Moreover, a number of glycosphingolipids have been suggested to act as beta-cell autoantigens. Studies in animal models of autoimmune diabetes, such as the Non Obese Diabetic (NOD) mouse and the LEW.1AR1-iddm (IDDM) rat, indicate a crucial role of sphingolipids in immune cell trafficking, islet infiltration and diabetes development. In this review, the up-to-date status on the findings about sphingolipids in T1DM will be provided, the under-investigated research areas will be identified and perspectives for future studies will be given.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Esfingolípidos/metabolismo , Animales , Autoantígenos/inmunología , Autoantígenos/metabolismo , Biomarcadores/metabolismo , Vías Biosintéticas , Citocinas/inmunología , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Humanos , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Esfingolípidos/inmunología
8.
Cell Death Dis ; 10(1): 29, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30631045

RESUMEN

The autoimmune-mediated beta-cell death in type 1 diabetes (T1DM) is associated with local inflammation (insulitis). We examined the role of MCPIP1 (monocyte chemotactic protein-induced protein 1), a novel cytokine-induced antiinflammatory protein, in this process. Basal MCPIP1 expression was lower in rat vs. human islets and beta-cells. Proinflammatory cytokines stimulated MCPIP1 expression in rat and human islets and in insulin-secreting cells. Moderate overexpression of MCPIP1 protected insulin-secreting INS1E cells against cytokine toxicity by a mechanism dependent on the presence of the PIN/DUB domain in MCPIP1. It also reduced cytokine-induced Chop and C/ebpß expression and maintained MCL-1 expression. The shRNA-mediated suppression of MCPIP1 led to the potentiation of cytokine-mediated NFκB activation and cytokine toxicity in human EndoC-ßH1 beta-cells. MCPIP1 expression was very high in infiltrated beta-cells before and after diabetes manifestation in the LEW.1AR1-iddm rat model of human T1DM. The extremely high expression of MCPIP1 in clonal beta-cells was associated with a failure of the regulatory feedback-loop mechanism, ER stress induction and high cytokine toxicity. In conclusion, our data indicate that the expression level of MCPIP1 affects the susceptibility of insulin-secreting cells to cytokines and regulates the mechanism of beta-cell death in T1DM.


Asunto(s)
Citocinas/toxicidad , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Ribonucleasas/genética , Ribonucleasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Expresión Génica , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Estrés Nitrosativo/efectos de los fármacos , Ratas , Ratas Endogámicas Lew , Transfección
9.
J Biol Chem ; 292(49): 20292-20304, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29070677

RESUMEN

Increasing evidence suggests a crucial role of inflammation in cytokine-mediated ß-cell dysfunction and death in type 1 diabetes mellitus, although the mechanisms are incompletely understood. Sphingosine 1-phosphate (S1P) is a multifunctional bioactive sphingolipid involved in the development of many autoimmune and inflammatory diseases. Here, we investigated the role of intracellular S1P in insulin-secreting INS1E cells by genetically manipulating the S1P-metabolizing enzyme S1P lyase (SPL). The expression of spl was down-regulated by cytokines in INS1E cells and rat islets. Overexpression of SPL protected against cytokine toxicity. Interestingly, the SPL overexpression did not suppress the cytokine-induced NFκB-iNOS-NO pathway but attenuated calcium leakage from endoplasmic reticulum (ER) stores as manifested by lower cytosolic calcium levels, higher expression of the ER protein Sec61a, decreased dephosphorylation of Bcl-2-associated death promoter (Bad) protein, and weaker caspase-3 activation in cytokine-treated (IL-1ß, TNFα, and IFNγ) cells. This coincided with reduced cytokine-mediated ER stress, indicated by measurements of CCAAT/enhancer-binding protein homologous protein (chop) and immunoglobulin heavy chain binding protein (bip) levels. Moreover, cytokine-treated SPL-overexpressing cells exhibited increased expression of prohibitin 2 (Phb2), involved in the regulation of mitochondrial assembly and respiration. SPL-overexpressing cells were partially protected against cytokine-mediated ATP reduction and inhibition of glucose-induced insulin secretion. siRNA-mediated spl suppression resulted in effects opposite to those observed for SPL overexpression. Knockdown of phb2 partially reversed beneficial effects of SPL overexpression. In conclusion, the relatively low endogenous Spl expression level in insulin-secreting cells contributes to their extraordinary vulnerability to proinflammatory cytokine toxicity and may therefore represent a promising target for ß-cell protection in type 1 diabetes mellitus.


Asunto(s)
Aldehído-Liasas/genética , Aldehído-Liasas/fisiología , Citocinas/toxicidad , Células Secretoras de Insulina/enzimología , Adenosina Trifosfato/metabolismo , Aldehído-Liasas/biosíntesis , Animales , Línea Celular , Citocinas/farmacología , Diabetes Mellitus Tipo 1/patología , Estrés del Retículo Endoplásmico , Inflamación/inducido químicamente , Inflamación/prevención & control , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/enzimología , Ratas
10.
Diabetologia ; 59(10): 2125-33, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27460666

RESUMEN

AIMS/HYPOTHESIS: The aim of this study was to perform a detailed analysis of cytokine toxicity in the new human EndoC-ßH1 beta cell line. METHODS: The expression profile of the antioxidative enzymes in the new human EndoC-ßH1 beta cells was characterised and compared with that of primary beta cells in the human pancreas. The effects of proinflammatory cytokines on reactive oxygen species formation, insulin secretory responsiveness and apoptosis of EndoC-ßH1 beta cells were determined. RESULTS: EndoC-ßH1 beta cells were sensitive to the toxic action of proinflammatory cytokines. Glucose-dependent stimulation of insulin secretion and an increase in the ATP/ADP ratio was abolished by proinflammatory cytokines without induction of IL-1ß expression. Cytokine-mediated caspase-3 activation was accompanied by reactive oxygen species formation and developed more slowly than in rodent beta cells. Cytokines transiently increased the expression of unfolded protein response genes, without inducing endoplasmic reticulum stress-marker genes. Cytokine-mediated NFκB activation was too weak to induce inducible nitric oxide synthase expression. The resultant lack of nitric oxide generation in EndoC-ßH1 cells, in contrast to rodent beta cells, makes these cells dependent on exogenously generated nitric oxide, which is released from infiltrating immune cells in human type 1 diabetes, for full expression of proinflammatory cytokine toxicity. CONCLUSIONS/INTERPRETATION: EndoC-ßH1 beta cells are characterised by an imbalance between H2O2-generating and -inactivating enzymes, and react to cytokine exposure in a similar manner to primary human beta cells. They are a suitable beta cell surrogate for cytokine-toxicity studies.


Asunto(s)
Citocinas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Western Blotting , Caspasa 3/metabolismo , Línea Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Glucosa/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Insulina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Pancrelipasa/metabolismo , Especies Reactivas de Oxígeno , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa-1/metabolismo
11.
Chem Biol Interact ; 256: 37-46, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27317948

RESUMEN

Homocysteine (HC) is considered to play an important role in the development of metabolic syndrome complications. Insulin-producing cells are prone to HC toxicity and this has been linked to oxidative stress. However, the exact mechanisms remain unknown. Therefore it was the aim of this study to determine the nature of reactive oxygen species responsible for HC toxicity. Chronic exposure of RINm5F and INS1E insulin-producing cells to HC decreased cell viability and glucose-induced insulin secretion in a concentration-dependent manner and led to a significant induction of hydrogen peroxide generation in the cytosolic, but not the mitochondrial compartment of the cell. Cytosolic overexpression of catalase, a hydrogen peroxide detoxifying enzyme, provided a significant protection against viability loss and hydrogen peroxide generation, while mitochondrial overexpression of catalase did not protect against HC toxicity. Overexpression of CuZnSOD, a cytosolic superoxide dismutating enzyme, also protected against HC toxicity. However, the best protection was achieved in the case of a combined overexpression of CuZnSOD and catalase. Incubation of cells in combination with alloxan resulted in a significant increase of HC toxicity and an increase of hydrogen peroxide generation. Overexpression of CuZnSOD or catalase protected against the toxicity of HC plus alloxan, with a superior protection achieved again by combined overexpression. The results indicate that HC induces oxidative stress in insulin-producing cells by stimulation of superoxide radical and hydrogen peroxide generation in the cytoplasm. The low antioxidative defence status makes the insulin-producing cells very vulnerable to HC toxicity.


Asunto(s)
Homocisteína/metabolismo , Células Secretoras de Insulina/metabolismo , Estrés Oxidativo , Aloxano/metabolismo , Aloxano/toxicidad , Animales , Catalasa/metabolismo , Línea Celular , Supervivencia Celular , Diabetes Mellitus Experimental/metabolismo , Peróxido de Hidrógeno/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
12.
Biochem Biophys Res Commun ; 464(1): 13-9, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26028562

RESUMEN

In the new human EndoC-ßH1 ß-cell line, a detailed analysis of the physiological characteristics was performed. This new human ß-cell line expressed all target structures on the gene and protein level, which are crucial for physiological function and insulin secretion induced by glucose and other secretagogues. Glucose influx measurements revealed an excellent uptake capacity of EndoC-ßH1 ß-cells by the Glut1 and Glut2 glucose transporters. A high expression level of glucokinase enabled efficient glucose phosphorylation, increasing the ATP/ADP ratio along with stimulation of insulin secretion in the physiological glucose concentration range. The EC50 value of glucose for insulin secretion was 10.3 mM. Mannoheptulose, a specific glucokinase inhibitor, blocked glucose-induced insulin secretion (GSIS). The nutrient insulin secretagogues l-leucine and 2-ketoisocaproate also stimulated insulin secretion, with a potentiating effect of l-glutamine. The Kir 6.2 potassium channel blocker glibenclamide and Bay K 8644, an opener of the voltage-sensitive Ca(2+) channel significantly potentiated GSIS. Potentiation of GSIS by IBMX and forskolin went along with a strong stimulation of cAMP generation. In conclusion, the new human EndoC-ßH1 ß-cell line fully mirrors the analogous physiological characteristics of primary mouse, rat and human ß-cells. Thus, this new human EndoC-ßH1 ß-cell line is very well suited for physiological ß-cell studies.


Asunto(s)
Efecto Fundador , Glucosa/metabolismo , Células Secretoras de Insulina/fisiología , Insulina/metabolismo , 1-Metil-3-Isobutilxantina/farmacología , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/biosíntesis , Transporte Biológico , Canales de Calcio/genética , Canales de Calcio/metabolismo , Línea Celular , Colforsina/farmacología , Expresión Génica , Glucoquinasa/antagonistas & inhibidores , Glucoquinasa/genética , Glucoquinasa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Glutamina/metabolismo , Glutamina/farmacología , Gliburida/farmacología , Humanos , Células Secretoras de Insulina/citología , Cetoácidos/metabolismo , Cetoácidos/farmacología , Leucina/metabolismo , Leucina/farmacología , Manoheptulosa/metabolismo , Manoheptulosa/farmacología , Fosforilación , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo
14.
Hum Mol Genet ; 24(8): 2274-86, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25552656

RESUMEN

Friedreich's ataxia (FRDA) is a neurodegenerative disorder associated with cardiomyopathy and diabetes. Effective therapies for FRDA are an urgent unmet need; there are currently no options to prevent or treat this orphan disease. FRDA is caused by reduced expression of the mitochondrial protein frataxin. We have previously demonstrated that pancreatic ß-cell dysfunction and death cause diabetes in FRDA. This is secondary to mitochondrial dysfunction and apoptosis but the underlying molecular mechanisms are not known. Here we show that ß-cell demise in frataxin deficiency is the consequence of oxidative stress-mediated activation of the intrinsic pathway of apoptosis. The pro-apoptotic Bcl-2 family members Bad, DP5 and Bim are the key mediators of frataxin deficiency-induced ß-cell death. Importantly, the intrinsic pathway of apoptosis is also activated in FRDA patients' induced pluripotent stem cell-derived neurons. Interestingly, cAMP induction normalizes mitochondrial oxidative status and fully prevents activation of the intrinsic pathway of apoptosis in frataxin-deficient ß-cells and neurons. This preclinical study suggests that incretin analogs hold potential to prevent/delay both diabetes and neurodegeneration in FRDA.


Asunto(s)
Apoptosis , Ataxia de Friedreich/fisiopatología , Células Secretoras de Insulina/citología , Neuronas/citología , Animales , Línea Celular , Diabetes Mellitus/etiología , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Femenino , Ataxia de Friedreich/complicaciones , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Masculino , Persona de Mediana Edad , Neuronas/metabolismo , Estrés Oxidativo , Ratas , Ratas Wistar , Frataxina
15.
Am J Physiol Endocrinol Metab ; 306(6): E648-57, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24425765

RESUMEN

A high-sucrose, low-copper-diet (HSD) induces inhibition of glucose-sensitive rats (CDs) but not Cohen diabetes-resistant rats (CDr). Copper-supplemented HSD increased activity of the copper-dependent mitochondrial respiratory chain enzyme cytochrome c oxidase (COX) and reversed hyperglycemia. This study examined the mechanism by which interleukin-1ß modulates GSIS and the role of COX in this process. We measured COX activity, ATP content, GSIS, iNOS expression, and nitrite production with and without IL-1ß, N(ω)-nitro-l-arginine, copper, or potassium cyanide in isolated islets of CDs and CDr fed different diets. We found reduced COX activity, ATP content, and GSIS in isolated islets of CDs rats fed a regular diet. These were severely reduced following HSD and were restored to regular diet levels on copper-supplemented HSD (P < 0.01 vs. CDr islets). Potassium cyanide chemically reduced COX activity, decreasing GSIS and thus reinforcing the link between islet COX activity and GSIS. Interleukin-1ß (2.5 U/ml) reduced GSIS and COX activity in CDs islets. Exposure to 10 U/ml interleukin-1ß decreased GSIS and COX activity in both CDs and CDr islets, inducing a similar nitrite production. Nevertheless, the effect on GSIS was more marked in CDs islets. A significant iNOS expression was detected in CDs on the HSD diet, which was reduced by copper supplementation. N(ω)-nitro-l-arginine and copper prevented the deleterious effect of interleukin-1ß on COX activity and GSIS. We conclude that reduced islet COX activity renders vulnerability to GSIS inhibition on low-copper HSD through two interrelated pathways: 1) by further reducing the activity of COX that is essential for ß-cell ATP-production and insulin secretion and 2) by inducing the expression of iNOS and nitric oxide-mediated COX inhibition. We suggest that islet COX activity must be maintained above a critical threshold to sustain adequate GSIS with exposure to low-copper HSD.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Insulina/metabolismo , Interleucina-1beta/metabolismo , Islotes Pancreáticos/metabolismo , Óxido Nítrico/metabolismo , Animales , Cobre/deficiencia , Cobre/metabolismo , Cobre/uso terapéutico , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/prevención & control , Sacarosa en la Dieta/efectos adversos , Complejo IV de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/prevención & control , Resistencia a la Insulina , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Masculino , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Concentración Osmolar , Ratas , Ratas Endogámicas , Técnicas de Cultivo de Tejidos
16.
Acta Biochim Pol ; 60(4): 595-602, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24432310

RESUMEN

Growth arrest and DNA damage-inducible (GADD) 45 proteins are regulators of cell death and survival. The proinflammatory cytokine IL-1ß strongly increases the level of the transcript encoding GADD45α in rat insulin-producing INS-1E cells. The activation of Gadd45α gene is clearly dependent on JNK and NF-κB activation and the synthesis of the secondary mediator nitric oxide (NO). Interestingly, the observed twelve-fold increase in the GADD45α-coding transcript level is not followed by increased expression of GADD45α at the protein level. An analysis of IL-1ß toxicity in INS-1E cells overexpressing GADD45α revealed no correlation between the GADD45α protein level and the sensitivity to IL-1ß toxicity. These findings suggest that the potential engagement of GADD45α in IL-1ß toxicity towards beta cells is limited to the effects induced by the basal expression level of this protein.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Interleucina-1beta/metabolismo , Proteínas Nucleares/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Muerte Celular/genética , Supervivencia Celular/genética , Regulación de la Expresión Génica , Interleucina-1beta/genética , Óxido Nítrico/metabolismo , Proteínas Nucleares/genética , Ratas
17.
Nitric Oxide ; 27(4): 235-41, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22921991

RESUMEN

Nitric oxide (NO), produced by the action of the inducible NO synthase, plays a crucial role in cytokine toxicity to pancreatic beta cells during type 1 diabetes development. It was the aim of this study to analyze the role of the neuronal NOS (nNOS) in proinflammatory cytokine-mediated beta cell toxicity. Expression of different isoforms of nitric oxide synthase in insulin-secreting INS1E cells and rat islets was analyzed by quantitative real-time PCR and Western blotting. The expression of nNOS in insulin-secreting INS1E cells was similar to that found in rat brain, while two other isoforms, namely the endothelial eNOS and inducible iNOS were not expressed in untreated cells. IL-1ß alone or in combination with TNF-α and/or IFNγ induced iNOS but not eNOS expression. In contrast, nNOS expression was strongly decreased by the mixture of the three proinflammatory cytokines (IL-1ß, TNF-α and IFNγ) both on the gene and protein level in INS1E cells and rat islet cells. The effects of cytokines on glucose-induced insulin-secretion followed the pattern of nNOS expression reduction and, on the other hand, of the iNOS induction. The data indicate that a low level of nitric oxide originating from the constitutive expression of nNOS in pancreatic beta cells is not deleterious. In particular since proinflammatory cytokines reduce this expression. This nNOS suppression can compensate for NO generation by low concentrations of IL-1ß through iNOS induction. Thus, this basal nNOS expression level in pancreatic beta cells represents a protective element against cytokine toxicity.


Asunto(s)
Citocinas/toxicidad , Células Secretoras de Insulina/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Secuencia de Bases , Citocinas/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Datos de Secuencia Molecular , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Ratas Endogámicas Lew , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
18.
Biochem J ; 445(3): 349-59, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22587331

RESUMEN

Mimitin, a novel mitochondrial protein, has been shown to act as a molecular chaperone for the mitochondrial complex I and to regulate ATP synthesis. During Type 1 diabetes development, pro-inflammatory cytokines induce mitochondrial damage in pancreatic ß-cells, inhibit ATP synthesis and reduce glucose-induced insulin secretion. Mimitin was expressed in rat pancreatic islets including ß-cells and decreased by cytokines. In the ob/ob mouse, a model of insulin resistance and obesity, mimitin expression was down-regulated in liver and brain, up-regulated in heart and kidney, but not affected in islets. To further analyse the impact of mimitin on ß-cell function, two ß-cell lines, one with a low (INS1E) and another with a higher (MIN6) mimitin expression were studied. Mimitin overexpression protected INS1E cells against cytokine-induced caspase 3 activation, mitochondrial membrane potential reduction and ATP production inhibition, independently from the NF-κB (nuclear factor κB)-iNOS (inducible NO synthase) pathway. Mimitin overexpression increased basal and glucose-induced insulin secretion and prevented cytokine-mediated suppression of insulin secretion. Mimitin knockdown in MIN6 cells had opposite effects to those observed after overexpression. Thus mimitin has the capacity to modulate pancreatic islet function and to reduce cytokine toxicity.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Células Secretoras de Insulina/fisiología , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Secuencia de Bases , Caspasas/metabolismo , Línea Celular , Proliferación Celular , Supervivencia Celular , Citocinas/farmacología , Cartilla de ADN/genética , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Técnicas In Vitro , Insulina/genética , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Obesos , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Chaperonas Moleculares/antagonistas & inhibidores , Chaperonas Moleculares/genética , Obesidad/fisiopatología , Ratas , Ratas Endogámicas Lew
19.
Endocrinology ; 153(6): 2612-22, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22495672

RESUMEN

Arachidonic acid metabolites are crucial mediators of inflammation in diabetes. Although eicosanoids are established modulators of pancreatic ß-cell function, the role of prostacyclin (prostaglandin I2) is unknown. Therefore, this study aimed to analyze the role of prostacyclin in ß-cell function. Prostacyclin synthase (PGIS) was weakly expressed in rat islet cells but nevertheless significantly increased by incubation with 30 mM glucose, especially in non-ß-cells. PGIS was overexpressed in INS1E cells, and the regulation of insulin secretion was analyzed. PGIS overexpression strongly potentiated glucose-induced insulin secretion along with increased insulin content and ATP production. Importantly, overexpression of PGIS potentiated only nutrient-induced insulin secretion. The effect of PGIS overexpression was mediated by prostacyclin released from insulin-secreting cells and dependent on prostacyclin receptor (IP receptor) activation, with concomitant cAMP production. The cAMP-mediated potentiation of glucose-induced insulin secretion by prostacyclin was independent of the protein kinase A pathway but strongly attenuated by the knockdown of the exchange protein directly activated by cAMP 2 (Epac2), pointing to a crucial role for Epac2 in this process. Thus, prostacyclin is a powerful potentiator of glucose-induced insulin secretion. It improves the secretory capacity by inducing insulin biosynthesis and probably by stimulating exocytosis. Our findings open a new therapeutical perspective for an improved treatment of type 2 diabetes.


Asunto(s)
Epoprostenol/farmacología , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , AMP Cíclico/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Sinergismo Farmacológico , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Interferencia de ARN , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Biochim Biophys Acta ; 1813(10): 1827-35, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21784110

RESUMEN

Pro-inflammatory cytokine-mediated beta cell apoptosis is activated through multiple signaling pathways involving mitochondria and endoplasmic reticulum. Activation of organelle-specific caspases has been implicated in the progression and execution of cell death. This study was therefore performed to elucidate the effects of pro-inflammatory cytokines on a possible cross-talk between the compartment-specific caspases 9 and 12 and their differential contribution to beta cell apoptosis. Moreover, the occurrence of ROS-mediated mitochondrial damage in response to beta cell toxic cytokines has been quantified. ER-specific caspase-12 was strongly activated in response to pro-inflammatory cytokines; however, its inhibition did not abolish cytokine-induced mitochondrial caspase-9 activation and loss of cell viability. In addition, there was a significant induction of oxidative mitochondrial DNA damage and elevated cardiolipin peroxidation in insulin-producing RINm5F cells and rat islet cells. Overexpression of the H(2)O(2) detoxifying enzyme catalase effectively reduced the observed cytokine-induced oxidative damage of mitochondrial structures. Taken together, the results strongly indicate that mitochondrial caspase-9 is not a downstream substrate of ER-specific caspase-12 and that pro-inflammatory cytokines cause apoptotic beta cell death through activation of caspase-9 primarily by hydroxyl radical-mediated mitochondrial damage.


Asunto(s)
Apoptosis/fisiología , Caspasa 12/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Caspasa 12/genética , Caspasa 9/metabolismo , Caspasa 9/fisiología , Catalasa/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citocinas/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Activación Enzimática/fisiología , Mediadores de Inflamación/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Ratas , Ratas Endogámicas Lew
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...