Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Diabetes ; 67(11): 2337-2348, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30348823

RESUMEN

The ß-cell has become recognized as a central player in the pathogenesis of type 1 diabetes with the generation of neoantigens as potential triggers for breaking immune tolerance. We report that posttranslationally modified glucose-regulated protein 78 (GRP78) is a novel autoantigen in human type 1 diabetes. When human islets were exposed to inflammatory stress induced by interleukin-1ß, tumor necrosis factor-α, and interferon-γ, arginine residue R510 within GRP78 was converted into citrulline, as evidenced by liquid chromatography-tandem mass spectrometry. This conversion, known as citrullination, led to the generation of neoepitopes, which effectively could be presented by HLA-DRB1*04:01 molecules. With the use of HLA-DRB1*04:01 tetramers and ELISA techniques, we demonstrate enhanced antigenicity of citrullinated GRP78 with significantly increased CD4+ T-cell responses and autoantibody titers in patients with type 1 diabetes compared with healthy control subjects. Of note, patients with type 1 diabetes had a predominantly higher percentage of central memory cells and a lower percentage of effector memory cells directed against citrullinated GRP78 compared with the native epitope. These results strongly suggest that citrullination of ß-cell proteins, exemplified here by the citrullination of GRP78, contributes to loss of self-tolerance toward ß-cells in human type 1 diabetes, indicating that ß-cells actively participate in their own demise.


Asunto(s)
Autoantígenos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Proteínas de Choque Térmico/metabolismo , Inflamación/metabolismo , Islotes Pancreáticos/metabolismo , Autoantígenos/inmunología , Citrulinación , Citocinas/farmacología , Diabetes Mellitus Tipo 1/inmunología , Chaperón BiP del Retículo Endoplásmico , Humanos , Inflamación/inmunología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/inmunología
2.
Cell Metab ; 28(6): 946-960.e6, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30078552

RESUMEN

Although CD8+ T-cell-mediated autoimmune ß cell destruction occurs in type 1 diabetes (T1D), the target epitopes processed and presented by ß cells are unknown. To identify them, we combined peptidomics and transcriptomics strategies. Inflammatory cytokines increased peptide presentation in vitro, paralleling upregulation of human leukocyte antigen (HLA) class I expression. Peptide sources featured several insulin granule proteins and all known ß cell antigens, barring islet-specific glucose-6-phosphatase catalytic subunit-related protein. Preproinsulin yielded HLA-A2-restricted epitopes previously described. Secretogranin V and its mRNA splice isoform SCG5-009, proconvertase-2, urocortin-3, the insulin gene enhancer protein ISL-1, and an islet amyloid polypeptide transpeptidation product emerged as antigens processed into HLA-A2-restricted epitopes, which, as those already described, were recognized by circulating naive CD8+ T cells in T1D and healthy donors and by pancreas-infiltrating cells in T1D donors. This peptidome opens new avenues to understand antigen processing by ß cells and for the development of T cell biomarkers and tolerogenic vaccination strategies.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/inmunología , Transcriptoma/inmunología , Animales , Biomarcadores/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Estudios de Casos y Controles , Línea Celular , Hormona Liberadora de Corticotropina/metabolismo , Citocinas/metabolismo , Antígenos HLA/metabolismo , Humanos , Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Ratones , Proteína 7B2 Secretora Neuroendocrina/metabolismo , Proproteína Convertasa 2/metabolismo , Precursores de Proteínas/metabolismo , Proteómica/métodos , Urocortinas/metabolismo
3.
Diabetes Obes Metab ; 20(12): 2800-2810, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29974637

RESUMEN

AIMS: To investigate the effect of kisspeptin on glucose-stimulated insulin secretion and appetite in humans. MATERIALS AND METHODS: In 15 healthy men (age: 25.2 ± 1.1 years; BMI: 22.3 ± 0.5 kg m-2 ), we compared the effects of 1 nmol kg-1 h-1 kisspeptin versus vehicle administration on glucose-stimulated insulin secretion, metabolites, gut hormones, appetite and food intake. In addition, we assessed the effect of kisspeptin on glucose-stimulated insulin secretion in vitro in human pancreatic islets and a human ß-cell line (EndoC-ßH1 cells). RESULTS: Kisspeptin administration to healthy men enhanced insulin secretion following an intravenous glucose load, and modulated serum metabolites. In keeping with this, kisspeptin increased glucose-stimulated insulin secretion from human islets and a human pancreatic cell line in vitro. In addition, kisspeptin administration did not alter gut hormones, appetite or food intake in healthy men. CONCLUSIONS: Collectively, these data demonstrate for the first time a beneficial role for kisspeptin in insulin secretion in humans in vivo. This has important implications for our understanding of the links between reproduction and metabolism in humans, as well as for the ongoing translational development of kisspeptin-based therapies for reproductive and potentially metabolic conditions.


Asunto(s)
Apetito/efectos de los fármacos , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Kisspeptinas/farmacología , Adolescente , Adulto , Línea Celular , Glucosa/metabolismo , Voluntarios Sanos , Humanos , Insulina/sangre , Masculino , Adulto Joven
4.
Diabetologia ; 61(8): 1780-1793, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29754287

RESUMEN

AIMS/HYPOTHESIS: Dietary n-3 polyunsaturated fatty acids, especially docosahexaenoic acid (DHA), are known to influence glucose homeostasis. We recently showed that Elovl2 expression in beta cells, which regulates synthesis of endogenous DHA, was associated with glucose tolerance and played a key role in insulin secretion. The present study aimed to examine the role of the very long chain fatty acid elongase 2 (ELOVL2)/DHA axis on the adverse effects of palmitate with high glucose, a condition defined as glucolipotoxicity, on beta cells. METHODS: We detected ELOVL2 in INS-1 beta cells and mouse and human islets using quantitative PCR and western blotting. Downregulation and adenoviral overexpression of Elovl2 was carried out in beta cells. Ceramide and diacylglycerol levels were determined by radio-enzymatic assay and lipidomics. Apoptosis was quantified using caspase-3 assays and poly (ADP-ribose) polymerase cleavage. Palmitate oxidation and esterification were determined by [U-14C]palmitate labelling. RESULTS: We found that glucolipotoxicity decreased ELOVL2 content in rodent and human beta cells. Downregulation of ELOVL2 drastically potentiated beta cell apoptosis induced by glucolipotoxicity, whereas adenoviral Elovl2 overexpression and supplementation with DHA partially inhibited glucolipotoxicity-induced cell death in rodent and human beta cells. Inhibition of beta cell apoptosis by the ELOVL2/DHA axis was associated with a decrease in ceramide accumulation. However, the ELOVL2/DHA axis was unable to directly alter ceramide synthesis or metabolism. By contrast, DHA increased palmitate oxidation but did not affect its esterification. Pharmacological inhibition of AMP-activated protein kinase and etomoxir, an inhibitor of carnitine palmitoyltransferase 1 (CPT1), the rate-limiting enzyme in fatty acid ß-oxidation, attenuated the protective effect of the ELOVL2/DHA axis during glucolipotoxicity. Downregulation of CPT1 also counteracted the anti-apoptotic action of the ELOVL2/DHA axis. By contrast, a mutated active form of Cpt1 inhibited glucolipotoxicity-induced beta cell apoptosis when ELOVL2 was downregulated. CONCLUSIONS/INTERPRETATION: Our results identify ELOVL2 as a critical pro-survival enzyme for preventing beta cell death and dysfunction induced by glucolipotoxicity, notably by favouring palmitate oxidation in mitochondria through a CPT1-dependent mechanism.


Asunto(s)
Acetiltransferasas/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Animales , Apoptosis/fisiología , Elongasas de Ácidos Grasos , Glucosa/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Oxidación-Reducción , Palmitatos/metabolismo
5.
Nat Commun ; 9(1): 1488, 2018 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-29662071

RESUMEN

Type 1 diabetes mellitus (T1DM) is due to the selective destruction of islet beta cells by immune cells. Current therapies focused on repressing the immune attack or stimulating beta cell regeneration still have limited clinical efficacy. Therefore, it is timely to identify innovative targets to dampen the immune process, while promoting beta cell survival and function. Liver receptor homologue-1 (LRH-1) is a nuclear receptor that represses inflammation in digestive organs, and protects pancreatic islets against apoptosis. Here, we show that BL001, a small LRH-1 agonist, impedes hyperglycemia progression and the immune-dependent inflammation of pancreas in murine models of T1DM, and beta cell apoptosis in islets of type 2 diabetic patients, while increasing beta cell mass and insulin secretion. Thus, we suggest that LRH-1 agonism favors a dialogue between immune and islet cells, which could be druggable to protect against diabetes mellitus.


Asunto(s)
Comunicación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/terapia , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Fenalenos/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Insulina/metabolismo , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/inmunología , Estreptozocina , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Trasplante Heterólogo
6.
Mol Cell Endocrinol ; 473: 186-193, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29409957

RESUMEN

It has been reported that the incretin system, including regulated GLP-1 secretion and locally expressed DPP-4, is present in pancreatic islets. In this study we comprehensively evaluated the expression and role of DPP-4 in islet alpha and beta cells from non-diabetic (ND) and type 2 diabetic (T2D) individuals, including the effects of its inhibition on beta cell function and survival. Isolated islets were prepared from 25 ND and 18 T2D organ donors; studies were also performed with the human insulin-producing EndoC-ßH1 cells. Morphological (including confocal microscopy), ultrastructural (electron microscopy, EM), functional (glucose-stimulated insulin secretion), survival (EM and nuclear dyes) and molecular (RNAseq, qPCR and western blot) studies were performed under several different experimental conditions. DPP-4 co-localized with glucagon and was also expressed in human islet insulin-containing cells. Furthermore, DPP-4 was expressed in EndoC-ßH1 cells. The proportions of DPP-4 positive alpha and beta cells and DPP-4 gene expression were significantly lower in T2D islets. A DPP-4 inhibitor protected ND human beta cells and EndoC-ßH1 cells against cytokine-induced toxicity, which was at least in part independent from GLP1 and associated with reduced NFKB1 expression. Finally, DPP-4 inhibition augmented glucose-stimulated insulin secretion, reduced apoptosis and improved ultrastructure in T2D beta cells. These results demonstrate the presence of DPP-4 in human islet alpha and beta cells, with reduced expression in T2D islets, and show that DPP-4 inhibition has beneficial effects on human ND and T2D beta cells. This suggests that DPP-4, besides playing a role in incretin effects, directly affects beta cell function and survival.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/patología , Dipeptidil Peptidasa 4/metabolismo , Células Secretoras de Insulina/enzimología , Anciano , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/toxicidad , Citoprotección/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/ultraestructura , Masculino , Persona de Mediana Edad
7.
Cell Death Dis ; 9(3): 279, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449530

RESUMEN

HMG20A (also known as iBRAF) is a chromatin factor involved in neuronal differentiation and maturation. Recently small nucleotide polymorphisms (SNPs) in the HMG20A gene have been linked to type 2 diabetes mellitus (T2DM) yet neither expression nor function of this T2DM candidate gene in islets is known. Herein we demonstrate that HMG20A is expressed in both human and mouse islets and that levels are decreased in islets of T2DM donors as compared to islets from non-diabetic donors. In vitro studies in mouse and human islets demonstrated that glucose transiently increased HMG20A transcript levels, a result also observed in islets of gestating mice. In contrast, HMG20A expression was not altered in islets from diet-induced obese and pre-diabetic mice. The T2DM-associated rs7119 SNP, located in the 3' UTR of the HMG20A transcript reduced the luciferase activity of a reporter construct in the human beta 1.1E7 cell line. Depletion of Hmg20a in the rat INS-1E cell line resulted in decreased expression levels of its neuronal target gene NeuroD whereas Rest and Pax4 were increased. Chromatin immunoprecipitation confirmed the interaction of HMG20A with the Pax4 gene promoter. Expression levels of Mafa, Glucokinase, and Insulin were also inhibited. Furthermore, glucose-induced insulin secretion was blunted in HMG20A-depleted islets. In summary, our data demonstrate that HMG20A expression in islet is essential for metabolism-insulin secretion coupling via the coordinated regulation of key islet-enriched genes such as NeuroD and Mafa and that depletion induces expression of genes such as Pax4 and Rest implicated in beta cell de-differentiation. More importantly we assign to the T2DM-linked rs7119 SNP the functional consequence of reducing HMG20A expression likely translating to impaired beta cell mature function.


Asunto(s)
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Células Secretoras de Insulina/metabolismo , Polimorfismo de Nucleótido Simple , Regiones no Traducidas 3' , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Glucemia/metabolismo , Línea Celular Tumoral , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Femenino , Predisposición Genética a la Enfermedad , Proteínas del Grupo de Alta Movilidad/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Secretoras de Insulina/patología , Lípidos/sangre , Masculino , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Fenotipo , Ratas
8.
Diabetologia ; 61(3): 641-657, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29185012

RESUMEN

AIMS/HYPOTHESIS: Pancreatic islet beta cell failure causes type 2 diabetes in humans. To identify transcriptomic changes in type 2 diabetic islets, the Innovative Medicines Initiative for Diabetes: Improving beta-cell function and identification of diagnostic biomarkers for treatment monitoring in Diabetes (IMIDIA) consortium ( www.imidia.org ) established a comprehensive, unique multicentre biobank of human islets and pancreas tissues from organ donors and metabolically phenotyped pancreatectomised patients (PPP). METHODS: Affymetrix microarrays were used to assess the islet transcriptome of islets isolated either by enzymatic digestion from 103 organ donors (OD), including 84 non-diabetic and 19 type 2 diabetic individuals, or by laser capture microdissection (LCM) from surgical specimens of 103 PPP, including 32 non-diabetic, 36 with type 2 diabetes, 15 with impaired glucose tolerance (IGT) and 20 with recent-onset diabetes (<1 year), conceivably secondary to the pancreatic disorder leading to surgery (type 3c diabetes). Bioinformatics tools were used to (1) compare the islet transcriptome of type 2 diabetic vs non-diabetic OD and PPP as well as vs IGT and type 3c diabetes within the PPP group; and (2) identify transcription factors driving gene co-expression modules correlated with insulin secretion ex vivo and glucose tolerance in vivo. Selected genes of interest were validated for their expression and function in beta cells. RESULTS: Comparative transcriptomic analysis identified 19 genes differentially expressed (false discovery rate ≤0.05, fold change ≥1.5) in type 2 diabetic vs non-diabetic islets from OD and PPP. Nine out of these 19 dysregulated genes were not previously reported to be dysregulated in type 2 diabetic islets. Signature genes included TMEM37, which inhibited Ca2+-influx and insulin secretion in beta cells, and ARG2 and PPP1R1A, which promoted insulin secretion. Systems biology approaches identified HNF1A, PDX1 and REST as drivers of gene co-expression modules correlated with impaired insulin secretion or glucose tolerance, and 14 out of 19 differentially expressed type 2 diabetic islet signature genes were enriched in these modules. None of these signature genes was significantly dysregulated in islets of PPP with impaired glucose tolerance or type 3c diabetes. CONCLUSIONS/INTERPRETATION: These studies enabled the stringent definition of a novel transcriptomic signature of type 2 diabetic islets, regardless of islet source and isolation procedure. Lack of this signature in islets from PPP with IGT or type 3c diabetes indicates differences possibly due to peculiarities of these hyperglycaemic conditions and/or a role for duration and severity of hyperglycaemia. Alternatively, these transcriptomic changes capture, but may not precede, beta cell failure.


Asunto(s)
Bancos de Muestras Biológicas , Diabetes Mellitus Tipo 2/metabolismo , Biología de Sistemas/métodos , Donantes de Tejidos , Transcriptoma/genética , Anciano , Anciano de 80 o más Años , Biología Computacional , Femenino , Humanos , Masculino , Pancreatectomía
9.
Diabetes ; 67(2): 278-290, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29203512

RESUMEN

Excessive glucocorticoid exposure has been shown to be deleterious for pancreatic ß-cell function and insulin release. However, glucocorticoids at physiological levels are essential for many homeostatic processes, including glycemic control. We show that corticosterone and cortisol and their less active precursors 11-dehydrocorticosterone (11-DHC) and cortisone suppress voltage-dependent Ca2+ channel function and Ca2+ fluxes in rodent as well as in human ß-cells. However, insulin secretion, maximal ATP/ADP responses to glucose, and ß-cell identity were all unaffected. Further examination revealed the upregulation of parallel amplifying cAMP signals and an increase in the number of membrane-docked insulin secretory granules. Effects of 11-DHC could be prevented by lipotoxicity and were associated with paracrine regulation of glucocorticoid activity because global deletion of 11ß-hydroxysteroid dehydrogenase type 1 normalized Ca2+ and cAMP responses. Thus, we have identified an enzymatically amplified feedback loop whereby glucocorticoids boost cAMP to maintain insulin secretion in the face of perturbed ionic signals. Failure of this protective mechanism may contribute to diabetes in states of glucocorticoid excess, such as Cushing syndrome, which are associated with frank dyslipidemia.


Asunto(s)
Señalización del Calcio , Corticosterona/metabolismo , Glucocorticoides/metabolismo , Hidrocortisona/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Biomarcadores/metabolismo , Canales de Calcio/química , Canales de Calcio/metabolismo , Diferenciación Celular , Corticosterona/análogos & derivados , Cortisona/metabolismo , AMP Cíclico/metabolismo , Glucosa/metabolismo , Humanos , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/citología , Cinética , Ratones Endogámicos , Ratones Noqueados , Técnicas de Cultivo de Tejidos
10.
Diabetes ; 67(3): 461-472, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29282201

RESUMEN

Although the mechanisms by which glucose regulates insulin secretion from pancreatic ß-cells are now well described, the way glucose modulates gene expression in such cells needs more understanding. Here, we demonstrate that MondoA, but not its paralog carbohydrate-responsive element-binding protein, is the predominant glucose-responsive transcription factor in human pancreatic ß-EndoC-ßH1 cells and in human islets. In high-glucose conditions, MondoA shuttles to the nucleus where it is required for the induction of the glucose-responsive genes arrestin domain-containing protein 4 (ARRDC4) and thioredoxin interacting protein (TXNIP), the latter being a protein strongly linked to ß-cell dysfunction and diabetes. Importantly, increasing cAMP signaling in human ß-cells, using forskolin or the glucagon-like peptide 1 mimetic Exendin-4, inhibits the shuttling of MondoA and potently inhibits TXNIP and ARRDC4 expression. Furthermore, we demonstrate that silencing MondoA expression improves glucose uptake in EndoC-ßH1 cells. These results highlight MondoA as a novel target in ß-cells that coordinates transcriptional response to elevated glucose levels.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Sistemas de Mensajero Secundario , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/antagonistas & inhibidores , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , AMP Cíclico/metabolismo , Exenatida , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Incretinas/farmacología , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Péptidos/farmacología , Interferencia de ARN , Sistemas de Mensajero Secundario/efectos de los fármacos , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Técnicas de Cultivo de Tejidos , Ponzoñas/farmacología
11.
Sci Rep ; 7(1): 11863, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28928397

RESUMEN

Statins are a class of drugs widely prescribed as frontline therapy for lowering plasma LDL-cholesterol in cardiovascular risk prevention. Several clinical reports have recently suggested an increased risk of type 2 diabetes associated with chronic use of these drugs. The pathophysiology of this effect remains to be fully elucidated but impaired ß-cell function constitutes a potential mechanism. The aim of this study was to explore the effect of a chronic treatment with lipophilic and hydrophilic statins on ß-cell function, using human pancreatic islets and rat insulin-secreting INS-1 cells; we particularly focused on the role of mitochondria and oxidative stress. The present study demonstrates, for the first time, that atorvastatin (lipophilic) but not pravastatin (hydrophilic) affected insulin release and mitochondrial metabolism due to the suppression of antioxidant defense system and induction of ROS production in pancreatic ß-cell models. Mevalonate addition and treatment with a specific antioxidant (N-AcetylCysteine) effectively reversed the observed defects. These data demonstrate that mitochondrial oxidative stress is a key element in the pathogenesis of statin-related diabetes and may have clinical relevance to design strategies for prevention or reduction of statin induced ß-cell dysfunction and diabetes in patients treated with lipophilic statins.


Asunto(s)
Atorvastatina , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mitocondrias/metabolismo , Pravastatina/farmacología , Animales , Atorvastatina/efectos adversos , Atorvastatina/farmacología , Línea Celular Tumoral , Humanos , Células Secretoras de Insulina/patología , Mitocondrias/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo
12.
Diabetes ; 66(1): 100-112, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27737950

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease leading to ß-cell destruction. MicroRNAs (miRNAs) are small noncoding RNAs that control gene expression and organ formation. They participate in the pathogenesis of several autoimmune diseases, but the nature of miRNAs contributing to ß-cell death in T1D and their target genes remain to be clarified. We performed an miRNA expression profile on human islet preparations exposed to the cytokines IL-1ß plus IFN-γ. Confirmation of miRNA and target gene modification in human ß-cells was performed by real-time quantitative PCR. Single-stranded miRNAs inhibitors were used to block selected endogenous miRNAs. Cell death was measured by Hoechst/propidium iodide staining and activation of caspase-3. Fifty-seven miRNAs were detected as modulated by cytokines. Three of them, namely miR-23a-3p, miR-23b-3p, and miR-149-5p, were downregulated by cytokines and selected for further studies. These miRNAs were found to regulate the expression of the proapoptotic Bcl-2 proteins DP5 and PUMA and consequent human ß-cell apoptosis. These results identify a novel cross talk between a key family of miRNAs and proapoptotic Bcl-2 proteins in human pancreatic ß-cells, broadening our understanding of cytokine-induced ß-cell apoptosis in early T1D.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Células Secretoras de Insulina/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Anciano , Proteínas Reguladoras de la Apoptosis/genética , Western Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Masculino , MicroARNs/genética , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN
13.
Cell Death Dis ; 7(8): e2329, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27512950

RESUMEN

Type 2 diabetes is a chronic metabolic disorder, where failure to maintain normal glucose homoeostasis is associated with, and exacerbated by, obesity and the concomitant-elevated free fatty acid concentrations typically found in these patients. Hyperglycaemia and hyperlipidaemia together contribute to a decline in insulin-producing ß-cell mass through activation of the transcription factors nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator of transcription (STAT)-1. There are however a large number of molecules potentially able to modulate NF-κB and STAT1 activity, and the mechanism(s) by which glucolipotoxicity initially induces NF-κB and STAT1 activation is currently poorly defined. Using high-density microarray analysis of the ß-cell transcritptome, we have identified those genes and proteins most sensitive to glucose and fatty acid environment. Our data show that of those potentially able to activate STAT1 or NF-κB pathways, tumour necrosis factor receptor (TNFR)-5 is the most highly upregulated by glucolipotoxicity. Importantly, our data also show that the physiological ligand for TNFR5, CD40L, elicits NF-κB activity in ß-cells, whereas selective knockdown of TNFR5 ameliorates glucolipotoxic induction of STAT1 expression and NF-κB activity. This data indicate for the first time that TNFR5 signalling has a major role in triggering glucolipotoxic islet cell death.


Asunto(s)
Antígenos CD40/metabolismo , Glucosa/toxicidad , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Lípidos/toxicidad , FN-kappa B/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
14.
J Biol Chem ; 291(23): 12040-56, 2016 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-27044747

RESUMEN

Pro-inflammatory cytokines contribute to pancreatic beta cell apoptosis in type 1 diabetes at least in part by inducing endoplasmic reticulum (ER) stress and the consequent unfolded protein response (UPR). It remains to be determined what causes the transition from "physiological" to "apoptotic" UPR, but accumulating evidence indicates that signaling by the ER transmembrane protein IRE1α is critical for this transition. IRE1α activation is regulated by both intra-ER and cytosolic cues. We evaluated the role for the presently discovered cytokine-induced and IRE1α-interacting protein ubiquitin D (UBD) on the regulation of IRE1α and its downstream targets. UBD was identified by use of a MAPPIT (mammalian protein-protein interaction trap)-based IRE1α interactome screen followed by comparison against functional genomic analysis of human and rodent beta cells exposed to pro-inflammatory cytokines. Knockdown of UBD in human and rodent beta cells and detailed signal transduction studies indicated that UBD modulates cytokine-induced UPR/IRE1α activation and apoptosis. UBD expression is induced by the pro-inflammatory cytokines interleukin (IL)-1ß and interferon (IFN)-γ in rat and human pancreatic beta cells, and it is also up-regulated in beta cells of inflamed islets from non-obese diabetic mice. UBD interacts with IRE1α in human and rodent beta cells, modulating IRE1α-dependent activation of JNK and cytokine-induced apoptosis. Our data suggest that UBD provides a negative feedback on cytokine-induced activation of the IRE1α/JNK pro-apoptotic pathway in cytokine-exposed beta cells.


Asunto(s)
Apoptosis , Endorribonucleasas/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitinas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Citocinas/farmacología , Endorribonucleasas/genética , Femenino , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Persona de Mediana Edad , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitinas/genética , Adulto Joven
15.
Diabetologia ; 58(10): 2307-16, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26099855

RESUMEN

AIMS/HYPOTHESIS: Proinflammatory cytokines contribute to beta cell damage in type 1 diabetes in part through activation of endoplasmic reticulum (ER) stress. In rat beta cells, cytokine-induced ER stress involves NO production and consequent inhibition of the ER Ca(2+) transporting ATPase sarco/endoplasmic reticulum Ca(2+) pump 2 (SERCA2B). However, the mechanisms by which cytokines induce ER stress and apoptosis in mouse and human pancreatic beta cells remain unclear. The purpose of this study is to elucidate the role of ER stress on cytokine-induced beta cell apoptosis in these three species and thus solve ongoing controversies in the field. METHODS: Rat and mouse insulin-producing cells, human pancreatic islets and human EndoC-ßH1 cells were exposed to the cytokines IL-1ß, TNF-α and IFN-γ, with or without NO inhibition. A global comparison of cytokine-modulated gene expression in human, mouse and rat beta cells was also performed. The chemical chaperone tauroursodeoxycholic acid (TUDCA) and suppression of C/EBP homologous protein (CHOP) were used to assess the role of ER stress in cytokine-induced apoptosis of human beta cells. RESULTS: NO plays a key role in cytokine-induced ER stress in rat islets, but not in mouse or human islets. Bioinformatics analysis indicated greater similarity between human and mouse than between human and rat global gene expression after cytokine exposure. The chemical chaperone TUDCA and suppression of CHOP or c-Jun N-terminal kinase (JNK) protected human beta cells against cytokine-induced apoptosis. CONCLUSIONS/INTERPRETATION: These observations clarify previous results that were discrepant owing to the use of islets from different species, and confirm that cytokine-induced ER stress contributes to human beta cell death, at least in part via JNK activation.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Ratas , Ratas Wistar , Ácido Tauroquenodesoxicólico/farmacología , Factor de Transcripción CHOP/farmacología , omega-N-Metilarginina/farmacología
16.
Diabetologia ; 58(6): 1260-71, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25810038

RESUMEN

AIMS/HYPOTHESIS: The role of the redox adaptor protein p66(Shc) as a potential mediator of saturated fatty acid (FA)-induced beta cell death was investigated. METHODS: The effects of the FA palmitate on p66(Shc) expression were evaluated in human and murine islets and in rat insulin-secreting INS-1E cells. p66(Shc) expression was also measured in islets from mice fed a high-fat diet (HFD) and from human donors with different BMIs. Cell apoptosis was quantified by two independent assays. The role of p66(Shc) was investigated using pancreatic islets from p66 (Shc-/-) mice and in INS-1E cells with knockdown of p66(Shc) or overexpression of wild-type and phosphorylation-defective p66(Shc). Production of reactive oxygen species (ROS) was evaluated by the dihydroethidium oxidation method. RESULTS: Palmitate induced a selective increase in p66(Shc) protein expression and phosphorylation on Ser(36) and augmented apoptosis in human and mouse islets and in INS-1E cells. Inhibiting the tumour suppressor protein p53 prevented both the palmitate-induced increase in p66(Shc) expression and beta cell apoptosis. Palmitate-induced apoptosis was abrogated in islets from p66 (Shc-/-) mice and following p66 (Shc) knockdown in INS-1E cells; by contrast, overexpression of p66(Shc), but not that of the phosphorylation-defective p66(Shc) mutant, enhanced palmitate-induced apoptosis. The pro-apoptotic effects of p66(Shc) were dependent upon its c-Jun N-terminal kinase-mediated phosphorylation on Ser(36) and associated with generation of ROS. p66(Shc) protein expression and function were also elevated in islets from HFD-fed mice and from obese/overweight cadaveric human donors. CONCLUSIONS/INTERPRETATION: p53-dependent augmentation of p66(Shc) expression and function represents a key signalling response contributing to beta cell apoptosis under conditions of lipotoxicity.


Asunto(s)
Apoptosis , Ácidos Grasos/metabolismo , Células Secretoras de Insulina/citología , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Adenoviridae/genética , Anciano , Animales , Índice de Masa Corporal , Dieta Alta en Grasa , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Oxidación-Reducción , Fosforilación , ARN Interferente Pequeño/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Proteína p53 Supresora de Tumor/metabolismo
17.
Mol Endocrinol ; 28(6): 860-71, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24766140

RESUMEN

Incretins such as glucagon-like peptide 1 (GLP-1) are released from the gut and potentiate insulin release in a glucose-dependent manner. Although this action is generally believed to hinge on cAMP and protein kinase A signaling, up-regulated beta cell intermediary metabolism may also play a role in incretin-stimulated insulin secretion. By employing recombinant probes to image ATP dynamically in situ within intact mouse and human islets, we sought to clarify the role of GLP-1-modulated energetics in beta cell function. Using these techniques, we show that GLP-1 engages a metabolically coupled subnetwork of beta cells to increase cytosolic ATP levels, an action independent of prevailing energy status. We further demonstrate that the effects of GLP-1 are accompanied by alterations in the mitochondrial inner membrane potential and, at elevated glucose concentration, depend upon GLP-1 receptor-directed calcium influx through voltage-dependent calcium channels. Lastly, and highlighting critical species differences, beta cells within mouse but not human islets respond coordinately to incretin stimulation. Together, these findings suggest that GLP-1 alters beta cell intermediary metabolism to influence ATP dynamics in a species-specific manner, and this may contribute to divergent regulation of the incretin-axis in rodents and man.


Asunto(s)
Péptido 1 Similar al Glucagón/fisiología , Incretinas/fisiología , Células Secretoras de Insulina/metabolismo , Adenosina Trifosfato/metabolismo , Adulto , Animales , Señalización del Calcio , Metabolismo Energético , Receptor del Péptido 1 Similar al Glucagón , Glucosa/metabolismo , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Persona de Mediana Edad , Receptores de Glucagón/metabolismo , Especificidad de la Especie , Técnicas de Cultivo de Tejidos
18.
J Clin Invest ; 123(10): 4182-94, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24018562

RESUMEN

Pancreatic ß cell dysfunction is pathognomonic of type 2 diabetes mellitus (T2DM) and is driven by environmental and genetic factors. ß cell responses to glucose and to incretins such as glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are altered in the disease state. While rodent ß cells act as a coordinated syncytium to drive insulin release, this property is unexplored in human islets. In situ imaging approaches were therefore used to monitor in real time the islet dynamics underlying hormone release. We found that GLP-1 and GIP recruit a highly coordinated subnetwork of ß cells that are targeted by lipotoxicity to suppress insulin secretion. Donor BMI was negatively correlated with subpopulation responses to GLP-1, suggesting that this action of incretin contributes to functional ß cell mass in vivo. Conversely, exposure of mice to a high-fat diet unveiled a role for incretin in maintaining coordinated islet activity, supporting the existence of species-specific strategies to maintain normoglycemia. These findings demonstrate that ß cell connectedness is an inherent property of human islets that is likely to influence incretin-potentiated insulin secretion and may be perturbed by diabetogenic insults to disrupt glucose homeostasis in humans.


Asunto(s)
Incretinas/fisiología , Células Secretoras de Insulina/metabolismo , Animales , Glucemia , Índice de Masa Corporal , Señalización del Calcio , Comunicación Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Ácidos Grasos no Esterificados/metabolismo , Ácidos Grasos no Esterificados/farmacología , Uniones Comunicantes/metabolismo , Péptido 1 Similar al Glucagón/fisiología , Glucosa/fisiología , Homeostasis , Humanos , Insulina/metabolismo , Secreción de Insulina , Ratones , Ratones Endogámicos C57BL , Especificidad de la Especie
19.
J Mol Cell Biol ; 4(4): 221-30, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22611253

RESUMEN

Elderly patients often suffer from multiple age-related diseases. Here we show that the expression of DJ-1, an antioxidant protein with reduced expression in the central nervous system of patients with Parkinson's disease, is reduced in pancreatic islets of patients with type 2 diabetes mellitus (T2DM). In contrast, under non-diabetic conditions, DJ-1 expression increases in mouse and human islets during aging. In mouse islets, we show that DJ-1 prevents an increase in reactive oxygen species levels as the mice age. This antioxidant function preserves mitochondrial integrity and physiology, prerequisites for glucose-stimulated insulin secretion. Accordingly, DJ-1-deficient mice develop glucose intolerance and reduced ß cell area as they age or gain weight. Our data suggest that DJ-1 is more generally involved in age- and lifestyle-related human diseases and show for the first time that DJ-1 plays a key role in glucose homeostasis and might serve as a novel drug target for T2DM.


Asunto(s)
Envejecimiento/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta , Glucosa/metabolismo , Homeostasis , Proteínas Oncogénicas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/efectos de los fármacos , Animales , Diabetes Mellitus Tipo 2/complicaciones , Glucosa/farmacología , Intolerancia a la Glucosa/complicaciones , Intolerancia a la Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Insulina/metabolismo , Secreción de Insulina , Insulinoma/metabolismo , Insulinoma/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/metabolismo , Peroxirredoxinas , Proteína Desglicasa DJ-1 , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/efectos de los fármacos
20.
PLoS One ; 7(5): e36188, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22563482

RESUMEN

We have investigated the in vitro effects of increased levels of glucose and free fatty acids on autophagy activation in pancreatic beta cells. INS-1E cells and isolated rat and human pancreatic islets were incubated for various times (from 2 to 24 h) at different concentrations of glucose and/or palmitic acid. Then, cell survival was evaluated and autophagy activation was explored by using various biochemical and morphological techniques. In INS-1E cells as well as in rat and human islets, 0.5 and 1.0 mM palmitate markedly increased autophagic vacuole formation, whereas high glucose was ineffective alone and caused little additional change when combined with palmitate. Furthermore, LC3-II immunofluorescence co-localized with that of cathepsin D, a lysosomal marker, showing that the autophagic flux was not hampered in PA-treated cells. These effects were maintained up to 18-24 h incubation and were associated with a significant decline of cell survival correlated with both palmitate concentration and incubation time. Ultrastructural analysis showed that autophagy activation, as evidenced by the occurrence of many autophagic vacuoles in the cytoplasm of beta cells, was associated with a diffuse and remarkable swelling of the endoplasmic reticulum. Our results indicate that among the metabolic alterations typically associated with type 2 diabetes, high free fatty acids levels could play a role in the activation of autophagy in beta cells, through a mechanism that might involve the induction of endoplasmic reticulum stress.


Asunto(s)
Autofagia/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Palmitatos/farmacología , Animales , Western Blotting , Catepsina D/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/ultraestructura , Glucosa/farmacología , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestructura , Insulinoma/metabolismo , Insulinoma/patología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA