Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 25(2)2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38255875

RESUMEN

It has been proposed that antidiabetic drugs, such as metformin and imatinib, at least in part, promote improved glucose tolerance in type 2 diabetic patients via increased production of the inflammatory cytokine GDF15. This is supported by studies, performed in rodent cell lines and mouse models, in which the addition or production of GDF15 improved beta-cell function and survival. The aim of the present study was to determine whether human beta cells produce GDF15 in response to antidiabetic drugs and, if so, to further elucidate the mechanisms by which GDF15 modulates the function and survival of such cells. The effects and expression of GDF15 were analyzed in human insulin-producing EndoC-betaH1 cells and human islets. We observed that alpha and beta cells exhibit considerable heterogeneity in GDF15 immuno-positivity. The predominant form of GDF15 present in islet and EndoC-betaH1 cells was pro-GDF15. Imatinib, but not metformin, increased pro-GDF15 levels in EndoC-betaH1 cells. Under basal conditions, exogenous GDF15 increased human islet oxygen consumption rates. In EndoC-betaH1 cells and human islets, exogenous GDF15 partially ameliorated cytokine- or palmitate + high-glucose-induced loss of function and viability. GDF15-induced cell survival was paralleled by increased inosine levels, suggesting a more efficient disposal of intracellular adenosine. Knockdown of adenosine deaminase, the enzyme that converts adenosine to inosine, resulted in lowered inosine levels and loss of protection against cytokine- or palmitate + high-glucose-induced cell death. It is concluded that imatinib-induced GDF15 production may protect human beta cells partially against inflammatory and metabolic stress. Furthermore, it is possible that the GDF15-mediated activation of adenosine deaminase and the increased disposal of intracellular adenosine participate in protection against beta-cell death.


Asunto(s)
Insulinas , Metformina , Ratones , Humanos , Animales , Citocinas , Adenosina Desaminasa , Desaminación , Mesilato de Imatinib , Adenosina/farmacología , Hipoglucemiantes , Inosina , Metformina/farmacología , Palmitatos , Estrés Fisiológico , Glucosa , Factor 15 de Diferenciación de Crecimiento/genética
2.
Front Endocrinol (Lausanne) ; 14: 1060675, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36761184

RESUMEN

Introduction: High intracellular concentrations of adenosine and 2'-deoxyadenosine have been suggested to be an important mediator of cell death. The aim of the present study was to characterize adenosine-induced death in insulin-producing beta-cells, at control and high glucose + palmitate-induced stress conditions. Methods: Human insulin-producing EndoC-betaH1 cells were treated with adenosine, 2'-deoxyadenosine, inosine and high glucose + sodium palmitate, and death rates using flow cytometry were studied. Results: We observed that adenosine and the non-receptor-activating analogue 2-deoxyadenosine, but not the adenosine deamination product inosine, promoted beta-cell apoptosis at concentrations exceeding maximal adenosine-receptor stimulating concentrations. Both adenosine and inosine were efficiently taken up by EndoC-betaH1 cells, and inosine counteracted the cell death promoting effect of adenosine by competing with adenosine for uptake. Both adenosine and 2'-deoxyadenosine promptly reduced insulin-stimulated production of plasma membrane PI(3,4,5)P3, an effect that was reversed upon wash out of adenosine. In line with this, adenosine, but not inosine, rapidly diminished Akt phosphorylation. Both pharmacological Bax inhibition and Akt activation blocked adenosine-induced beta-cell apoptosis, indicating that adenosine/2'-deoxyadenosine inhibits the PI3K/Akt/BAD anti-apoptotic pathway. High glucose + palmitate-induced cell death was paralleled by increased intracellular adenosine and inosine levels. Overexpression of adenosine deaminase-1 (ADA1) in EndoC-betaH1 cells, which increased Akt phosphorylation, prevented both adenosine-induced apoptosis and high glucose + palmitate-induced necrosis. ADA2 overexpression not only failed to protect against adenosine and high glucose + palmitate-activated cell death, but instead potentiated the apoptosis-stimulating effect of adenosine. In line with this, ADA1 overexpression increased inosine production from adenosine-exposed cells, whereas ADA2 did not. Knockdown of ADA1 resulted in increased cell death rates in response to both adenosine and high glucose + palmitate. Inhibition of miR-30e-3p binding to the ADA1 mRNA 3'-UTR promoted the opposite effects on cell death rates and reduced intracellular adenosine contents. Discussion: It is concluded that intracellular adenosine/2'-deoxyadenosine regulates negatively the PI3K pathway and is therefore an important mediator of beta-cell apoptosis. Adenosine levels are controlled, at least in part, by ADA1, and strategies to upregulate ADA1 activity, during conditions of metabolic stress, could be useful in attempts to preserve beta-cell mass in diabetes.


Asunto(s)
Adenosina , Células Secretoras de Insulina , Proteínas Proto-Oncogénicas c-akt , Humanos , Adenosina/farmacología , Apoptosis , Glucosa/farmacología , Glucosa/metabolismo , Insulina/metabolismo , Palmitatos , Fosfatidilinositol 3-Quinasas , Células Secretoras de Insulina/citología
3.
Clin Sci (Lond) ; 135(19): 2243-2263, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34569605

RESUMEN

The protein tyrosine kinase inhibitor imatinib is used in the treatment of various malignancies but may also promote beneficial effects in the treatment of diabetes. The aim of the present investigation was to characterize the mechanisms by which imatinib protects insulin producing cells. Treatment of non-obese diabetic (NOD) mice with imatinib resulted in increased beta-cell AMP-activated kinase (AMPK) phosphorylation. Imatinib activated AMPK also in vitro, resulting in decreased ribosomal protein S6 phosphorylation and protection against islet amyloid polypeptide (IAPP)-aggregation, thioredoxin interacting protein (TXNIP) up-regulation and beta-cell death. 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) mimicked and compound C counteracted the effect of imatinib on beta-cell survival. Imatinib-induced AMPK activation was preceded by reduced glucose/pyruvate-dependent respiration, increased glycolysis rates, and a lowered ATP/AMP ratio. Imatinib augmented the fractional oxidation of fatty acids/malate, possibly via a direct interaction with the beta-oxidation enzyme enoyl coenzyme A hydratase, short chain, 1, mitochondrial (ECHS1). In non-beta cells, imatinib reduced respiratory chain complex I and II-mediated respiration and acyl-CoA carboxylase (ACC) phosphorylation, suggesting that mitochondrial effects of imatinib are not beta-cell specific. In conclusion, tyrosine kinase inhibitors modestly inhibit mitochondrial respiration, leading to AMPK activation and TXNIP down-regulation, which in turn protects against beta-cell death.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Hipoglucemiantes/farmacología , Mesilato de Imatinib/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Animales , Proteínas Portadoras/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Diabetes Mellitus/enzimología , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Enoil-CoA Hidratasa/metabolismo , Activación Enzimática , Humanos , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Masculino , Ratones Endogámicos NOD , Mitocondrias/enzimología , Mitocondrias/patología , Fosforilación , Ratas Sprague-Dawley , Proteína S6 Ribosómica/metabolismo
4.
Diabetologia ; 64(10): 2292-2305, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34296320

RESUMEN

AIMS/HYPOTHESIS: ZBED6 (zinc finger, BED-type containing 6) is known to regulate muscle mass by suppression of Igf2 gene transcription. In insulin-producing cell lines, ZBED6 maintains proliferative capacity at the expense of differentiation and beta cell function. The aim was to study the impact of Zbed6 knockout on beta cell function and glucose tolerance in C57BL/6 mice. METHODS: Beta cell area and proliferation were determined in Zbed6 knockout mice using immunohistochemical analysis. Muscle and fat distribution were assessed using micro-computed tomography. Islet gene expression was assessed by RNA sequencing. Effects of a high-fat diet were analysed by glucose tolerance and insulin tolerance tests. ZBED6 was overexpressed in EndoC-ßH1 cells and human islet cells using an adenoviral vector. Beta cell cell-cycle analysis, insulin release and mitochondrial function were studied in vitro using propidium iodide staining and flow cytometry, ELISA, the Seahorse technique, and the fluorescent probes JC-1 and MitoSox. RESULTS: Islets from Zbed6 knockout mice showed lowered expression of the cell cycle gene Pttg1, decreased beta cell proliferation and decreased beta cell area, which occurred independently from ZBED6 effects on Igf2 gene expression. Zbed6 knockout mice, but not wild-type mice, developed glucose intolerance when given a high-fat diet. The high-fat diet Zbed6 knockout islets displayed upregulated expression of oxidative phosphorylation genes and genes associated with beta cell differentiation. In vitro, ZBED6 overexpression resulted in increased EndoC-ßH1 cell proliferation and a reduced glucose-stimulated insulin release in human islets. ZBED6 also reduced mitochondrial JC-1 J-aggregate formation, mitochondrial oxygen consumption rates (OCR) and mitochondrial reactive oxygen species (ROS) production, both at basal and palmitate + high glucose-stimulated conditions. ZBED6-induced inhibition of OCR was not rescued by IGF2 addition. ZBED6 reduced levels of the mitochondrial regulator PPAR-γ related coactivator 1 protein (PRC) and bound its promoter/enhancer region. Knockdown of PRC resulted in a lowered OCR. CONCLUSIONS/INTERPRETATION: It is concluded that ZBED6 is required for normal beta cell replication and also limits excessive beta cell mitochondrial activation in response to an increased functional demand. ZBED6 may act, at least in part, by restricting PRC-mediated mitochondrial activation/ROS production, which may lead to protection against beta cell dysfunction and glucose intolerance in vivo.


Asunto(s)
Dieta Alta en Grasa , Intolerancia a la Glucosa/metabolismo , Células Secretoras de Insulina/fisiología , Mitocondrias/metabolismo , Proteínas Represoras/fisiología , Adenoviridae/genética , Animales , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Regulación de la Expresión Génica/fisiología , Vectores Genéticos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Consumo de Oxígeno/fisiología , Fosforilación , Especies Reactivas de Oxígeno , Reacción en Cadena en Tiempo Real de la Polimerasa , Securina/genética
5.
FASEB J ; 33(1): 88-100, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29957057

RESUMEN

Zinc finger BED domain containing protein 6 ( Zbed6) has evolved from a domesticated DNA transposon and encodes a transcription factor unique to placental mammals. The aim of the present study was to investigate further the role of ZBED6 in insulin-producing cells, using mouse MIN6 cells, and to evaluate the effects of Zbed6 knockdown on basal ß-cell functions, such as morphology, transcriptional regulation, insulin content, and release. Zbed6-silenced cells and controls were characterized with a range of methods, including RNA sequencing, chromatin immunoprecipitation sequencing, insulin content and release, subplasma membrane Ca2+ measurements, cAMP determination, and morphologic studies. More than 700 genes showed differential expression in response to Zbed6 knockdown, which was paralleled by increased capacity to generate cAMP, as well as by augmented subplasmalemmal calcium concentration and insulin secretion in response to glucose stimulation. We identified >4000 putative ZBED6-binding sites in the MIN6 genome, with an enrichment of ZBED6 sites at upregulated genes, such as the ß-cell transcription factors v-maf musculoaponeurotic fibrosarcoma oncogene homolog A and Nk6 homeobox 1. We also observed altered morphology/growth patterns, as indicated by increased cell clustering, and in the appearance of axon-like Neurofilament, medium polypeptide and tubulin ß 3, class III-positive protrusions. We conclude that ZBED6 acts as a transcriptional regulator in MIN6 cells and that its activity suppresses insulin production, cell aggregation, and neuronal-like differentiation.-Wang, X., Jiang, L., Wallerman, O., Younis, S., Yu, Q., Klaesson, A., Tengholm, A., Welsh, N., Andersson, L. ZBED6 negatively regulates insulin production, neuronal differentiation, and cell aggregation in MIN6 cells.


Asunto(s)
Diferenciación Celular , Células Secretoras de Insulina/patología , Insulina/metabolismo , Insulinoma/patología , Neuronas/patología , Neoplasias Pancreáticas/patología , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Adhesión Celular , Agregación Celular , Regulación de la Expresión Génica , Silenciador del Gen , Glucosa/administración & dosificación , Secuenciación de Nucleótidos de Alto Rendimiento , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Ratones , Neuronas/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transcripción Genética , Células Tumorales Cultivadas
6.
PLoS One ; 13(9): e0204271, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30265686

RESUMEN

It has been proposed that pancreatic beta-cell dysfunction in type 2 diabetes is promoted by oxidative stress caused by NADPH oxidase (Nox) over-activity. The aim of the present study was to evaluate the efficacy of novel Nox inhibitors as protective agents against cytokine- or high glucose + palmitate-induced human beta-cell death. The Nox2 protein was present mainly in the cytoplasm and was induced by cytokines. Nox4 protein immunoreactivity, with some nuclear accumulation, was observed in human islet cells, and was not affected by islet culture in the presence of cytokines or high glucose + palmitate. Nox inhibitors with partial or no isoform selectivity (DPI, dapsone, GLX351322, and GLX481372) all reduced ROS production of human islet cells exposed to high glucose + palmitate. This was paralleled by improved viability and reduced caspase 3 activation. The Nox1 selective inhibitor ML171 failed to reduce human islet cell death in response to both cytokines and high glucose + palmitate. The selective Nox2 inhibitor Phox-I2 also failed to protect against cytokines, but protected partially against high glucose + palmitate-induced cellular death. The highly selective Nox4 inhibitor GLX7013114 protected islet cells against both cytokines and high glucose + palmitate. However, as no osmotic control for high glucose was used, we cannot exclude the possibility that the high glucose effect was due to osmosis. It is concluded that Nox4 may participate in stress-induced islet cell death in human islets in vitro. We propose that Nox4 mediates pro-apoptotic effects in intact islets under stressful conditions and that selective Nox4-inhibition may be a therapeutic strategy in type 2 diabetes.


Asunto(s)
Muerte Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , NADPH Oxidasa 4/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Células HEK293 , Humanos , Islotes Pancreáticos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , NADPH Oxidasa 2/metabolismo , NADPH Oxidasa 4/metabolismo , Ácido Palmítico/farmacología , Superóxidos/metabolismo
7.
Proc Natl Acad Sci U S A ; 115(12): E2752-E2761, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29507232

RESUMEN

Aggregation of islet amyloid polypeptide (IAPP) into amyloid fibrils in islets of Langerhans is associated with type 2 diabetes, and formation of toxic IAPP species is believed to contribute to the loss of insulin-producing beta cells. The BRICHOS domain of integral membrane protein 2B (Bri2), a transmembrane protein expressed in several peripheral tissues and in the brain, has recently been shown to prevent fibril formation and toxicity of Aß42, an amyloid-forming peptide in Alzheimer disease. In this study, we demonstrate expression of Bri2 in human islets and in the human beta-cell line EndoC-ßH1. Bri2 colocalizes with IAPP intracellularly and is present in amyloid deposits in patients with type 2 diabetes. The BRICHOS domain of Bri2 effectively inhibits fibril formation in vitro and instead redirects IAPP into formation of amorphous aggregates. Reduction of endogenous Bri2 in EndoC-ßH1 cells with siRNA increases sensitivity to metabolic stress leading to cell death while a concomitant overexpression of Bri2 BRICHOS is protective. Also, coexpression of IAPP and Bri2 BRICHOS in lateral ventral neurons of Drosophila melanogaster results in an increased cell survival. IAPP is considered to be the most amyloidogenic peptide known, and described findings identify Bri2, or in particular its BRICHOS domain, as an important potential endogenous inhibitor of IAPP aggregation and toxicity, with the potential to be a possible target for the treatment of type 2 diabetes.


Asunto(s)
Amiloide/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Animales Modificados Genéticamente , Apoptosis/fisiología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Diabetes Mellitus Tipo 2/patología , Drosophila melanogaster/genética , Femenino , Glucosa/farmacología , Humanos , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ácido Palmítico/farmacología , Dominios Proteicos
8.
Ups J Med Sci ; 122(3): 149-159, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28980863

RESUMEN

BACKGROUND: Enhanced IAPP production may contribute to islet amyloid formation in type 2 diabetes. The objective of this study was to determine the effects of the saturated fatty acid palmitate on IAPP levels in human ß-cells. METHODS: EndoC-ßH1 cells and human islets were cultured in the presence of sodium palmitate. Effects on IAPP/insulin mRNA expression and secretion were determined using real-time qPCR/ELISA. Pharmacological activators and/or inhibitors and RNAi were used to determine the underlying mechanisms. RESULTS: We observed that EndoC-ßH1 cells exposed to palmitate for 72 h displayed decreased expression of Pdx-1 and MafA and increased expression of thioredoxin-interacting protein (TXNIP), reduced insulin mRNA expression and glucose-induced insulin secretion, as well as increased IAPP mRNA expression and secretion. Further, these effects were independent of fatty acid oxidation, but abolished in response to GPR40 inhibition/downregulation. In human islets both a high glucose concentration and palmitate promoted increased IAPP mRNA levels, resulting in an augmented IAPP/insulin mRNA ratio. This was paralleled by elevated IAPP/insulin protein secretion and content ratios. CONCLUSIONS: Addition of exogenous palmitate to human ß-cells increased the IAPP/insulin expression ratio, an effect contributed to by activation of GPR40. These findings may be pertinent to our understanding of the islet amyloid formation process.


Asunto(s)
Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Ácido Palmítico/farmacología , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Línea Celular , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Humanos , Insulina/genética , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Oxidación-Reducción , Proteína Quinasa C/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
9.
Heliyon ; 2(9): e00159, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27699280

RESUMEN

OBJECTIVES: Insulin expression is highly controlled on the posttranscriptional level. The RNA binding proteins (RBPs) responsible for this result are still largely unknown. METHODS AND RESULTS: To identify RBPs that bind to insulin mRNA we performed mass spectrometry analysis on proteins that bound synthetic oligonucloetides mimicing the 5'- and the 3'-untranslated regions (UTRs) of rat and human insulin mRNA in vitro. We observed that the RBPs heterogeneous nuclear ribonucleoprotein (hnRNP) U, polypyrimidine tract binding protein (PTB), hnRNP L and T-cell restricted intracellular antigen 1-related protein (TIA-1-related protein; TIAR) bind to insulin mRNA sequences, and that the in vitro binding affinity of these RBPs changed when INS-1 cells were exposed to glucose, 3-isobutyl-1-methylxanthine (IBMX) or nitric oxide. High glucose exposure resulted in a modest increase in PTB and TIAR binding to an insulin mRNA sequence. The inducer of nitrosative stress DETAnonoate increased markedly hnRNP U and TIAR mRNA binding. An increased PTB to TIAR binding ratio in vitro correlated with higher insulin mRNA levels and insulin biosynthesis rates in INS-1 cells. To further investigate the importance of RNA-binding proteins for insulin mRNA stability, we decreased INS-1 and EndoC-ßH1 cell levels of PTB and TIAR by RNAi. In both cell lines, decreased levels of PTB resulted in lowered insulin mRNA levels while decreased levels of TIAR resulted in increased insulin mRNA levels. Thapsigargin-induced stress granule formation was associated with a redistribution of TIAR from the cytosol to stress granules. CONCLUSIONS: These experiments indicate that alterations in insulin mRNA stability and translation correlate with differential RBP binding. We propose that the balance between PTB on one hand and TIAR on the other participates in the control of insulin mRNA stability and utilization for insulin biosynthesis.

10.
Eur J Pharmacol ; 784: 69-80, 2016 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-27178899

RESUMEN

We presently report that treatment with tyrphostin AG-126 (2-(3-hydroxy-4-nitrobenzylidene)malononitrile) and ten other aromatic malononitrile compounds (AMN) improves the resistance of insulin-producing ßTC6, RIN-5AH, and MIN6 cells to oxidative stress and pro-inflammatory cytokines. On the molecular level AMN compounds promote nuclear accumulation of the Nrf2 transcription factor and expression of the cytoprotective genes heme ogygenase 1 (HO-1) and NAD(P)H/quinone oxidoreductase 1 (NQO1), inhibit cytokine-dependent inducible nitric oxide synthase (iNOS) induction, suppress intracellular production of reactive oxygen species in ßTC6 and counteract to impairments of glucose-stimulated insulin secretion induced by pro-inflammatory cytokines in MIN6 cells. Nrf2 up-regulation and HO-1 induction by AG-126 are attenuated at the presence of siRNA against Nrf2 and brusatol, an inhibitor of the Nrf2 signaling pathway. Our present results indicate that in respect of inhibition of IL-1ß-dependent iNOS induction, ßTC6 cells are more sensitive to EMK 1071 (2-((5-methylthiophen-2-yl)methylene)malononitrile) and EMK 31 (2-(4-hydroxy-3-methoxybenzylidene)malononitrile) as compared to other analyzed AMN compounds. We suggest that the ability of AMN compounds to inhibit iNOS induction and other cytokine-induced transcriptional events might be a tool to achieve improved ß-cell survival and functionality.


Asunto(s)
Citocinas/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/efectos de los fármacos , Nitrilos/farmacología , Oxidantes/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Hemo-Oxigenasa 1/metabolismo , Peróxido de Hidrógeno/toxicidad , Inflamación/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/farmacología , Ratones , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Nitrilos/química , Estrés Oxidativo/efectos de los fármacos , Cuassinas/farmacología , Ratas , Transcripción Genética/efectos de los fármacos , Vitamina K 3/toxicidad
11.
Mol Cell Endocrinol ; 414: 53-63, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26213325

RESUMEN

The aim of the present investigation was to delineate cytokine-induced signaling and death using the EndoC-ßH1 cells as a model for primary human beta-cells. The cytokines IL-1ß and IFN-γ induced a rapid and transient activation of NF-κB, STAT-1, ERK, JNK and eIF-2α signaling. The EndoC-ßH1 cells died rapidly when exposed to IL-1ß + IFN-γ, and this occurred also in the presence of the actinomycin D. Inhibition of NF-κB and STAT-1 did not protect against cell death, nor did the cytokines activate iNOS expression. Instead, cytokines promoted a rapid decrease in EndoC-ßH1 cell respiration and ATP levels, and we observed protection by the AMPK activator AICAR against cytokine-induced cell death. It is concluded that EndoC-ßH1 cell death can be prevented by AMPK activation, which suggests a role for ATP depletion in cytokine-induced human beta-cell death.


Asunto(s)
Adenilato Quinasa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Fragmentos de Péptidos/farmacología , Adenosina Trifosfato/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Dactinomicina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/enzimología , Ratones , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos
12.
Clin Sci (Lond) ; 128(1): 17-28, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24865476

RESUMEN

Low-density lipoprotein receptor-related protein 1 (LRP1) is an endocytic and multi-functional type I cell surface membrane protein, which is known to be phosphorylated by the activated platelet-derived growth factor receptor (PDGFR). The tyrosine kinase inhibitor imatinib, which inhibits PDGFR and c-Abl, and which has previously been reported to counteract ß-cell death and diabetes, has been suggested to reduce atherosclerosis by inhibiting PDGFR-induced LRP1 phosphorylation. The aim of the present study was to study LRP1 function in ß-cells and to what extent imatinib modulates LRP1 activity. LRP1 and c-Abl gene knockdown was performed by RNAi using rat INS-1 832/13 and human EndoC1-ßH1 cells. LRP1 was also antagonized by treatment with the antagonist low-density lipoprotein receptor-related protein associated protein 1 (LRPAP1). We have used PDGF-BB, a PDGFR agonist, and apolipoprotein E (ApoE), an LRP1 agonist, to stimulate the activities of PDGFR and LRP1 respectively. Knockdown or inhibition of LRP1 resulted in increased hydrogen peroxide (H2O2)- or cytokine-induced cell death, and glucose-induced insulin release was lowered in LRP1-silenced cells. These results indicate that LRP1 function is necessary for ß-cell function and that LRP1 is adversely affected by challenges to ß-cell health. PDGF-BB, or the combination of PDGF-BB+ApoE, induced phosphorylation of extracellular-signal-regulated kinase (ERK), Akt and LRP1. LRP1 silencing blocked this event. Imatinib blocked phosphorylation of LRP1 by PDGFR activation but induced phosphorylation of ERK. LRP1 silencing blocked imatinib-induced phosphorylation of ERK. Sunitinib also blocked LRP1 phosphorylation in response to PDGF-BB and induced phosphorylation of ERK, but this latter event was not affected by LRP1 knockdown. siRNA-mediated knockdown of the imatinib target c-Abl resulted in an increased ERK phosphorylation at basal conditions, with no further increase in response to imatinib. Imatinib-induced cell survival of tunicamycin-treated cells was partially mediated by ERK activation. We have concluded that imatinib promotes LRP1-dependent ERK activation, possibly via inhibition of c-Abl, and that this could contribute to the pro-survival effects of imatinib on ß-cells.


Asunto(s)
Benzamidas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Piperazinas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Técnicas de Silenciamiento del Gen , Mesilato de Imatinib , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-abl/efectos de los fármacos , Proteínas Proto-Oncogénicas c-abl/fisiología , Ratas
13.
Ups J Med Sci ; 119(4): 306-15, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25266628

RESUMEN

BACKGROUND: Sodium palmitate causes apoptosis of ß-cells, and the anti-apoptotic protein Bcl-2 has been shown to counteract this event. However, the exact mechanisms that underlie palmitate-induced pancreatic ß-cell apoptosis and through which pathway Bcl-2 executes the protective effect are still unclear. METHODS: A stable Bcl-2-overexpressing RINm5F cell clone (BMG) and its negative control (B45) were exposed to palmitate for up to 8 h, and cell viability, mitochondrial membrane potential (Δψm), reactive oxygen species (ROS) generation, endoplasmic reticulum (ER) stress, and NF-κB activation were studied in time course experiments. RESULTS: Palmitate exposure for 8 h resulted in increased cell death rates, and this event was partially counteracted by Bcl-2. Bcl-2 overexpression promoted in parallel also a delayed induction of GADD153/CHOP and a weaker phosphorylation of BimEL in palmitate-exposed cells. At earlier time points (2-4 h) palmitate exposure resulted in increased generation of ROS, a decrease in mitochondrial membrane potential (Δψm), and a modest increase in the phosphorylation of eIF2α and IRE1α. BMG cells produced similar amounts of ROS and displayed the same eIF2α and IRE1α phosphorylation rates as B45 cells. However, the palmitate-induced dissipation of Δψm was partially counteracted by Bcl-2. In addition, basal NF-κB activity was increased in BMG cells. CONCLUSIONS: Our results indicate that Bcl-2 counteracts palmitate-induced ß-cell death by maintaining mitochondrial membrane integrity and augmenting NF-κB activity, but not by affecting ROS production and ER stress.


Asunto(s)
Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/metabolismo , Potencial de la Membrana Mitocondrial/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis , Línea Celular , Supervivencia Celular , Retículo Endoplásmico/metabolismo , Ácidos Grasos no Esterificados/química , Metilación , Ratones , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Ácido Palmítico/química , Fosforilación , Transducción de Señal
14.
Pancreas ; 43(4): 624-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24717804

RESUMEN

OBJECTIVE: It has been proposed that the histamine 1 (H(1)) receptor not only promotes allergic reactions but also modulates autoimmune diseases, such as type 1 diabetes. In line with this, it has recently been reported that the H(1)-receptor antagonist cetirizine can counteract the activation of signals/factors pertinent to the pathogenesis of type 1 diabetes and cytokine-induced ß-cell destruction. Therefore, the overall aim of this study was to determine whether H(1)-receptor antagonists affect cytokine-induced ß-cell death and signaling in vitro. METHODS: The insulin-producing cell line ß-TC6 was exposed to the proinflammatory cytokines interleukin 1ß(+) interferon γ, or hydrogen peroxide. The H(1)-receptor antagonists desloratadine and cetirizine were added to the cell cultures and cell viability; macrophage inhibitory factor levels, c-Jun N-terminal kinase phosphorylation, c-Jun expression, and ß-catenin levels were analyzed by flow cytometry, real-time polymerase chain reaction, and immunoblotting. RESULTS: Cetirizine protected partially against both cytokine- and hydrogen peroxide-induced cell death. This effect was paralleled by an inhibition of cytokine-induced c-Jun N-terminal kinase phosphorylation, c-Jun induction, and a restoration of macrophage inhibitory factor contents. Cetirizine also increased the ß-TC6 cell contents of ß-catenin at basal conditions. CONCLUSIONS: Our results indicate a protective effect of a specific H(1)-receptor antagonist.


Asunto(s)
Cetirizina/farmacología , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Interferón gamma/toxicidad , Interleucina-1beta/toxicidad , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citoprotección , Relación Dosis-Respuesta a Droga , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Oxidorreductasas Intramoleculares/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Óxido Nítrico/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/efectos de los fármacos , beta Catenina/metabolismo
15.
Genomics ; 103(4): 264-75, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24462878

RESUMEN

Type 1 Diabetes (T1D) is an autoimmune disease where local release of cytokines such as IL-1ß and IFN-γ contributes to ß-cell apoptosis. To identify relevant genes regulating this process we performed a meta-analysis of 8 datasets of ß-cell gene expression after exposure to IL-1ß and IFN-γ. Two of these datasets are novel and contain time-series expressions in human islet cells and rat INS-1E cells. Genes were ranked according to their differential expression within and after 24 h from exposure, and characterized by function and prior knowledge in the literature. A regulatory network was then inferred from the human time expression datasets, using a time-series extension of a network inference method. The two most differentially expressed genes previously unknown in T1D literature (RIPK2 and ELF3) were found to modulate cytokine-induced apoptosis. The inferred regulatory network is thus supported by the experimental validation, providing a proof-of-concept for the proposed statistical inference approach.


Asunto(s)
Citocinas/metabolismo , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Células Secretoras de Insulina/fisiología , Animales , Citocinas/farmacología , Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 1 , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Islotes Pancreáticos/fisiología , Proteínas Proto-Oncogénicas c-ets/genética , Ratas , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Reproducibilidad de los Resultados , Factores de Transcripción/genética
16.
PLoS One ; 8(4): e61828, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23613946

RESUMEN

PURPOSE: Transplantation of pancreatic islets to Type 1 diabetes patients is hampered by inflammatory reactions at the transplantation site leading to dysfunction and death of insulin producing beta-cells. Recently we have shown that co-transplantation of neural crest stem cells (NCSCs) together with the islet cells improves transplantation outcome. The aim of the present investigation was to describe in vitro interactions between NCSCs and insulin producing beta-TC6 cells that may mediate protection against cytokine-induced beta-cell death. PROCEDURES: Beta-TC6 and NCSC cells were cultured either alone or together, and either with or without cell culture inserts. The cultures were then exposed to the pro-inflammatory cytokines IL-1ß and IFN-γ for 48 hours followed by analysis of cell death rates (flow cytometry), nitrite production (Griess reagent), protein localization (immunofluorescence) and protein phosphorylation (flow cytometry). RESULTS: We observed that beta-TC6 cells co-cultured with NCSCs were protected against cytokine-induced cell death, but not when separated by cell culture inserts. This occurred in parallel with (i) augmented production of nitrite from beta-TC6 cells, indicating that increased cell survival allows a sustained production of nitric oxide; (ii) NCSC-derived laminin production; (iii) decreased phospho-FAK staining in beta-TC6 cell focal adhesions, and (iv) decreased beta-TC6 cell phosphorylation of ERK(T202/Y204), FAK(Y397) and FAK(Y576). Furthermore, co-culture also resulted in cadherin and beta-catenin accumulations at the NCSC/beta-TC6 cell junctions. Finally, the gap junction inhibitor carbenoxolone did not affect cytokine-induced beta-cell death during co-culture with NCSCs. CONCLUSION: In summary, direct contacts, but not soluble factors, promote improved beta-TC6 viability when co-cultured with NCSCs. We hypothesize that cadherin junctions between NCSC and beta-TC6 cells promote powerful signals that maintain beta-cell survival even though ERK and FAK signaling are suppressed. It may be that future strategies to improve islet transplantation outcome may benefit from attempts to increase beta-cell cadherin junctions to neighboring cells.


Asunto(s)
Cadherinas/metabolismo , Citocinas/farmacología , Citoprotección/efectos de los fármacos , Uniones Comunicantes/metabolismo , Células Secretoras de Insulina/citología , Cresta Neural/citología , Células-Madre Neurales/citología , Animales , Muerte Celular/efectos de los fármacos , Técnicas de Cocultivo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Uniones Comunicantes/efectos de los fármacos , Insulina/biosíntesis , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/enzimología , Integrina alfa6/metabolismo , Laminina/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo
17.
Expert Opin Investig Drugs ; 21(11): 1743-50, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22998750

RESUMEN

INTRODUCTION: The small tyrosine kinase inhibitor Imatinib Mesylate (Gleevec) protects against diabetes, but it is not known how. AREAS COVERED: It has been suggested that islet amyloid and fibrotic deposits promote beta-cell failure and death, leading to Type-2 diabetes. As Imatinib is known to possess anti-fibrotic/amyloid properties, in for example systemic sclerosis and mouse models for Alzheimer's disease, the present review will discuss the possibility that Imatinib acts, at least in part, by ameliorating islet hyalinization and its consequences in the pathogenesis of Type-2 diabetes. EXPERT OPINION: A better understanding of how Imatinib counteracts Type-2 diabetes will possibly help to clarify the pathogenic role of islet amyloid and fibrosis, and hopefully lead to improved treatment of the disease.


Asunto(s)
Diabetes Mellitus Tipo 2/prevención & control , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Amiloidosis/complicaciones , Amiloidosis/tratamiento farmacológico , Animales , Benzamidas , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/prevención & control , Diabetes Mellitus Tipo 2/etiología , Modelos Animales de Enfermedad , Fibrosis , Humanos , Mesilato de Imatinib , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/patología , Ratones , Enfermedades Pancreáticas/complicaciones , Enfermedades Pancreáticas/tratamiento farmacológico , Enfermedades Pancreáticas/patología
18.
PLoS One ; 6(9): e24831, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21935477

RESUMEN

PURPOSE: Imatinib Mesylate (Gleevec) is a drug that potently counteracts diabetes both in humans and in animal models for human diabetes. We have previously reported that this compound in human pancreatic islets stimulates NF-κB signaling and islet cell survival. The aim of this study was to investigate control of NF-κB post-translational modifications exerted by Imatinib and whether any such effects are associated with altered islet gene expression and chemokine production in vitro. PROCEDURES: Human islets were either left untreated or treated with Imatinib for different timepoints. IκB-α and NF-κB p65 phosphorylation and methylation were assessed by immunoblot analysis. Islet gene expression was assessed using a commercial Pathway Finder microarray kit and RT-PCR. Islet chemokine production was determined by flow cytometric bead array analysis. FINDINGS: Human islet IκB-α and Ser276-p65 phosphorylation were increased by a 20 minute Imatinib exposure. Methylation of p65 at position Lys221 was increased after 60 min of Imatinib exposure and persisted for 3 hours. Microarray analysis of islets exposed to Imatinib for 4 hours revealed increased expression of the inflammatory genes IL-4R, TCF5, DR5, I-TRAF, I-CAM, HSP27 and IL-8. The islet release of IL-8 was augmented in islets cultured over night in the presence of Imatinib. Following 30 hours of Imatinib exposure, the cytokine-induced IκB-α and STAT1 phosphorylation was abolished and diminished, respectively. The cytokine-induced release of the chemokines MIG and IP10 was lower in islets exposed to Imatinib for 30 hours. CONCLUSION: Imatinib by itself promotes a modest activation of NF-κB. However, a prolonged exposure of human islets to Imatinib is associated with a dampened response to cytokines. It is possible that Imatinib induces NF-κB preconditioning of islet cells leading to lowered cytokine sensitivity and a mitigated islet inflammation.


Asunto(s)
Piperazinas/farmacología , Pirimidinas/farmacología , Factor de Transcripción ReIA/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Benzamidas , Quimiocinas/metabolismo , Citometría de Flujo , Proteínas de Choque Térmico HSP27/genética , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Mesilato de Imatinib , Técnicas In Vitro , Interleucina-8/genética , Metilación/efectos de los fármacos , Inhibidor NF-kappaB alfa , Fosforilación/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Factor de Transcripción ReIA/genética
19.
Biochem Biophys Res Commun ; 412(4): 693-8, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21867683

RESUMEN

The aim of this study was to investigate whether cap-independent insulin mRNA translation occurs in human pancreatic islets at basal conditions, during stimulation at a high glucose concentration and at conditions of nitrosative stress. We also aimed at correlating cap-independent insulin mRNA translation with binding of the IRES trans-acting factor polypyrimidine tract binding protein (PTB) to the 5'-UTR of insulin mRNA. For this purpose, human islets were incubated for 2h in the presence of low (1.67 mM) or high glucose (16.7 mM). Nitrosative stress was induced by addition of 1 mM DETA/NO and cap-dependent mRNA translation was inhibited with hippuristanol. Insulin biosynthesis rates were determined by radioactive labeling and immunoprecipitation. PTB affinity to insulin mRNA 5'-UTR was assessed by a magnetic micro bead pull-down procedure. We observed that in the presence of 1.67 mM glucose, approximately 70% of the insulin mRNA translation was inhibited by hippuristanol. Corresponding value from islets incubated at 16.7 mM glucose was 93%. DETA/NO treatment significantly decreased the translation of insulin by 85% in high glucose incubated islets, and by 50% at a low glucose concentration. The lowered insulin biosynthesis rates of DETA/NO-exposed islets were further suppressed by hippuristanol with 55% at 16.7 mM glucose but not at 1.67 mM glucose. Thus, hippuristanol-induced inhibition of insulin biosynthesis was less pronounced in DETA/NO-treated islets as compared to control islets. We observed also that PTB bound specifically to the insulin mRNA 5'-UTR in vitro, and that this binding corresponded well with rates of cap-independent insulin biosynthesis at the different conditions. In conclusion, our studies show that insulin biosynthesis is mainly cap-dependent at a high glucose concentration, but that the cap-independent biosynthesis of insulin can constitute as much as 40-100% of all insulin biosynthesis during conditions of nitrosative stress. These data suggest that the pancreatic ß-cell is able to uphold basal insulin synthesis at conditions of starvation and stress via a cap- and eIF4A-independent mechanism, possibly mediated by the binding of PTB to the 5'-UTR of the human insulin mRNA.


Asunto(s)
Factor 4A Eucariótico de Iniciación/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , Biosíntesis de Proteínas/genética , Caperuzas de ARN/metabolismo , ARN Mensajero/genética , Regiones no Traducidas 5'/genética , Células Cultivadas , Glucosa/farmacología , Humanos , Insulina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Esteroles/farmacología , Triazenos/farmacología
20.
PLoS One ; 5(5): e10843, 2010 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-20520763

RESUMEN

BACKGROUND: Prolonged periods of high glucose exposure results in human islet dysfunction in vitro. The underlying mechanisms behind this effect of high glucose are, however, unknown. The polypyrimidine tract binding protein (PTB) is required for stabilization of insulin mRNA and the PTB mRNA 3'-UTR contains binding sites for the microRNA molecules miR-133a, miR-124a and miR-146. The aim of this study was therefore to investigate whether high glucose increased the levels of these three miRNAs in association with lower PTB levels and lower insulin biosynthesis rates. METHODOLOGY/PRINCIPAL FINDINGS: Human islets were cultured for 24 hours in the presence of low (5.6 mM) or high glucose (20 mM). Islets were also exposed to sodium palmitate or the proinflammatory cytokines IL-1beta and IFN-gamma, since saturated free fatty acids and cytokines also cause islet dysfunction. RNA was then isolated for real-time RT-PCR analysis of miR-133a, miR-124a, miR-146, insulin mRNA and PTB mRNA contents. Insulin biosynthesis rates were determined by radioactive labeling and immunoprecipitation. Synthetic miR-133a precursor and inhibitor were delivered to dispersed islet cells by lipofection, and PTB was analyzed by immunoblotting following culture at low or high glucose. Culture in high glucose resulted in increased islet contents of miR-133a and reduced contents of miR-146. Cytokines increased the contents of miR-146. The insulin and PTB mRNA contents were unaffected by high glucose. However, both PTB protein levels and insulin biosynthesis rates were decreased in response to high glucose. The miR-133a inhibitor prevented the high glucose-induced decrease in PTB and insulin biosynthesis, and the miR-133a precursor decreased PTB levels and insulin biosynthesis similarly to high glucose. CONCLUSION: Prolonged high-glucose exposure down-regulates PTB levels and insulin biosynthesis rates in human islets by increasing miR-133a levels. We propose that this mechanism contributes to hyperglycemia-induced beta-cell dysfunction.


Asunto(s)
Glucosa/farmacología , Insulina/biosíntesis , Islotes Pancreáticos/metabolismo , MicroARNs/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Citocinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Insulina/genética , Islotes Pancreáticos/efectos de los fármacos , MicroARNs/genética , Ácido Palmítico/farmacología , Proteína de Unión al Tracto de Polipirimidina/genética , Precursores del ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA