Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Diabetes Res Clin Pract ; 211: 111655, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574895

RESUMEN

AIMS: We aimed to assess physical activity (PA) levels, adherence to PA guidelines, and fitness capacity in individuals with type 1 diabetes (T1D) and control population. METHODS: This cross-sectional study included 232 T1D and 248 controls. PA levels (IPAQ-SF questionnaire), adherence to guidelines (>150 min/week of moderate-to-vigorous PA), fitness capacity (VO2max, maximal incremental test on a cycle ergometer and 1RM test) were assessed, along with other clinical variables. RESULTS: Total PA levels (T1D 2202 ± 1839 vs. controls 2357 ± 2189 METs/min/week), adherence (T1D 53.1 % vs controls 53.2 %), and sedentariness (T1D 27.3 % vs. controls 25.1 %) were similar between groups. However, participants with T1D exhibited significantly lower levels of VO2max (29.1 ± 10.5 vs. 32.5 ± 11.5 mlO2/kg/min, p < 0.001), work capacity (2.73 ± 1.03 vs. 3 ± 10 W/kg of body weight, p = 0.004) and strength capacity (2.29 ± 0.53 vs. 2.41 ± 0.79 kg/kg body weight in 1RM, p = 0.01) than controls, after adjusting for sex and age. CONCLUSIONS: Individuals with T1D exhibit lower fitness capacity compared to a control population, regardless of age and sex, even when presenting similar levels of total physical activity and adherence to guidelines.


Asunto(s)
Diabetes Mellitus Tipo 1 , Tolerancia al Ejercicio , Ejercicio Físico , Humanos , Diabetes Mellitus Tipo 1/fisiopatología , Masculino , Femenino , Estudios Transversales , Ejercicio Físico/fisiología , Adulto , Tolerancia al Ejercicio/fisiología , Persona de Mediana Edad , Aptitud Física/fisiología , Consumo de Oxígeno/fisiología , Adulto Joven , Estudios de Casos y Controles , Encuestas y Cuestionarios
2.
Commun Biol ; 6(1): 256, 2023 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-36964318

RESUMEN

Direct lineage reprogramming of one somatic cell into another without transitioning through a progenitor stage has emerged as a strategy to generate clinically relevant cell types. One cell type of interest is the pancreatic insulin-producing ß cell whose loss and/or dysfunction leads to diabetes. To date it has been possible to create ß-like cells from related endodermal cell types by forcing the expression of developmental transcription factors, but not from more distant cell lineages like fibroblasts. In light of the therapeutic benefits of choosing an accessible cell type as the cell of origin, in this study we set out to analyze the feasibility of transforming human skin fibroblasts into ß-like cells. We describe how the timed-introduction of five developmental transcription factors (Neurog3, Pdx1, MafA, Pax4, and Nkx2-2) promotes conversion of fibroblasts toward a ß-cell fate. Reprogrammed cells exhibit ß-cell features including ß-cell gene expression and glucose-responsive intracellular calcium mobilization. Moreover, reprogrammed cells display glucose-induced insulin secretion in vitro and in vivo. This work provides proof-of-concept of the capacity to make insulin-producing cells from human fibroblasts via transcription factor-mediated direct reprogramming.


Asunto(s)
Insulina , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Insulina/metabolismo , Regulación de la Expresión Génica , Diferenciación Celular/fisiología , Fibroblastos/metabolismo
3.
Diabetes Metab ; 48(6): 101361, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35714884

RESUMEN

AIMS: to investigate if a High Intensity Interval Training (HIIT) protocol improves glycemic control and fitness capacity, compared to traditional moderate Intensity Continuous Training (MICT) exercise. METHODS: 30 sedentary individuals with type 1 diabetes (T1D) and 26 healthy controls were assigned to a 3-week HIIT or MICT protocol. Blood glucose levels by continuous glucose monitoring system and fitness status were compared before and after the study period. RESULTS: During workouts, blood glucose levels remained stable in HIIT exercise (+3.2 ± 16.2 mg/dl (p = 0.43)), while decreased in MICT (-27.1 ± 17.5 mg/dl (p < 0.0001)) exercise. In addition, out of the 9 training sessions, HIIT volunteers needed to take carbohydrate supplements to avoid hypoglycemia in 0.56 ± 0.9 sessions, compared to 1.83 ± 0.5 sessions (p < 0.04) in MICT individuals. In the analysis of blood glucose levels between rest and training days (24h-period), training significantly reduced mean glycemic levels in both groups, but the MICT exercise results in an increase in the frequency of hypoglycemic episodes. The response to exercise seems to be attenuated in individuals with T1D, especially in HIIT group. CONCLUSION: HIIT training results in a greater glycemic stability, with reduction of hypoglycemic episodes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Entrenamiento de Intervalos de Alta Intensidad , Hipoglucemia , Humanos , Entrenamiento de Intervalos de Alta Intensidad/métodos , Diabetes Mellitus Tipo 1/terapia , Glucemia , Hipoglucemiantes , Automonitorización de la Glucosa Sanguínea , Hipoglucemia/prevención & control
4.
NMR Biomed ; 35(9): e4745, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35435283

RESUMEN

Liver fibrosis staging is a key element driving the prognosis of patients with chronic liver disease. Currently, biopsy is the only technique capable of diagnosing liver fibrosis in patients with alcohol-related liver disease (ArLD) and nonalcoholic fatty liver disease (NAFLD) unequivocally. Noninvasive (e.g. plasma-based) biomarker assays are attractive tools to diagnose and stage disease, yet must prove that they are reliable and sensitive to be used clinically. Here, we demonstrate proton nuclear magnetic resonance as a method to rapidly quantify the endogenous concentration of ammonium ions from human plasma extracts and show their ability to report upon early and advanced stages of ArLD and NAFLD. We show that, irrespective of the disease etiology, ammonium concentration is a more robust and informative marker of fibrosis stage than current clinically assessed blood hepatic biomarkers. Subject to validation in larger cohorts, the study indicates that the method can provide accurate and rapid staging of ArLD and NAFLD without the need for an invasive biopsy.


Asunto(s)
Compuestos de Amonio , Diagnóstico por Imagen de Elasticidad , Enfermedad del Hígado Graso no Alcohólico , Biomarcadores , Biopsia , Diagnóstico por Imagen de Elasticidad/métodos , Humanos , Hígado/diagnóstico por imagen , Hígado/patología , Cirrosis Hepática/diagnóstico por imagen , Espectroscopía de Resonancia Magnética , Enfermedad del Hígado Graso no Alcohólico/diagnóstico por imagen , Protones
5.
Sci Rep ; 11(1): 11878, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34088954

RESUMEN

Amyloid deposits in pancreatic islets, mainly formed by human islet amyloid polypeptide (hIAPP) aggregation, have been associated with loss of ß-cell mass and function, and are a pathological hallmark of type 2 diabetes (T2D). Treatment with chaperones has been associated with a decrease in endoplasmic reticulum stress leading to improved glucose metabolism. The aim of this work was to investigate whether the chemical chaperone 4-phenylbutyrate (PBA) prevents glucose metabolism abnormalities and amyloid deposition in obese agouti viable yellow (Avy) mice that overexpress hIAPP in ß cells (Avy hIAPP mice), which exhibit overt diabetes. Oral PBA treatment started at 8 weeks of age, when Avy hIAPP mice already presented fasting hyperglycemia, glucose intolerance, and impaired insulin secretion. PBA treatment strongly reduced the severe hyperglycemia observed in obese Avy hIAPP mice in fasting and fed conditions throughout the study. This effect was paralleled by a decrease in hyperinsulinemia. Importantly, PBA treatment reduced the prevalence and the severity of islet amyloid deposition in Avy hIAPP mice. Collectively, these results show that PBA treatment elicits a marked reduction of hyperglycemia and reduces amyloid deposits in obese and diabetic mice, highlighting the potential of chaperones for T2D treatment.


Asunto(s)
Hiperglucemia/tratamiento farmacológico , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/metabolismo , Obesidad/tratamiento farmacológico , Fenilbutiratos/farmacología , Amiloide/metabolismo , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis , Hiperglucemia/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Obesidad/metabolismo
6.
Mol Metab ; 53: 101251, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34015524

RESUMEN

OBJECTIVE: Pancreatic ß-cell dysfunction is a central feature in the pathogenesis of type 2 diabetes (T2D). Accumulating evidence indicates that ß-site APP-cleaving enzyme 2 (BACE2) inhibition exerts a beneficial effect on ß-cells in different models of T2D. Thus, targeting BACE2 may represent a potential therapeutic strategy for the treatment of this disease. Here, we aimed to investigate the effects of BACE2 suppression on glucose homeostasis in a model of diet-induced obesity. METHODS: BACE2 knock-out (BKO) and wild-type (WT) mice were fed with a high-fat diet (HFD) for 2 or 16 weeks. Body weight, food intake, respiratory exchange ratio, locomotor activity, and energy expenditure were determined. Glucose homeostasis was evaluated by glucose and insulin tolerance tests. ß-cell proliferation was assessed by Ki67-positive nuclei, and ß-cell function was determined by measuring glucose-stimulated insulin secretion. Leptin sensitivity was evaluated by quantifying food intake and body weight after an intraperitoneal leptin injection. Neuropeptide gene expression and insulin signaling in the mediobasal hypothalamus were determined by qPCR and Akt phosphorylation, respectively. RESULTS: After 16 weeks of HFD feeding, BKO mice exhibited an exacerbated body weight gain and hyperphagia, in comparison to WT littermates. Glucose tolerance was similar in both groups, whereas HFD-induced hyperinsulinemia, insulin resistance, and ß-cell expansion were more pronounced in BKO mice. In turn, leptin-induced food intake inhibition and hypothalamic insulin signaling were impaired in BKO mice, regardless of the diet, in accordance with deregulation of the expression of hypothalamic neuropeptide genes. Importantly, BKO mice already showed increased ß-cell proliferation and glucose-stimulated insulin secretion with respect to WT littermates after two weeks of HFD feeding, before the onset of obesity. CONCLUSIONS: Collectively, these results reveal that BACE2 suppression in an obesogenic setting leads to exacerbated body weight gain, hyperinsulinemia, and insulin resistance. Thus, we conclude that inhibition of BACE2 may aggravate the adverse metabolic effects associated with obesity.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Obesidad/metabolismo , Animales , Dieta/efectos adversos , Masculino , Ratones , Ratones Transgénicos
7.
Mol Metab ; 37: 100984, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32229246

RESUMEN

OBJECTIVE: Pancreatic ß-cell failure is central to the development and progression of type 2 diabetes (T2D). The aggregation of human islet amyloid polypeptide (hIAPP) has been associated with pancreatic islet inflammation and dysfunction in T2D. Alpha1-antitrypsin (AAT) is a circulating protease inhibitor with anti-inflammatory properties. Here, we sought to investigate the potential therapeutic effect of AAT treatment in a mouse model characterized by hIAPP overexpression in pancreatic ß-cells. METHODS: Mice overexpressing hIAPP (hIAPP-Tg) in pancreatic ß-cells were used as a model of amyloid-induced ß-cell dysfunction. Glucose homeostasis was evaluated by glucose tolerance tests and insulin secretion assays. Apoptosis and amyloid formation was assessed in hIAPP-Tg mouse islets cultured at high glucose levels. Dissociated islet cells were cocultured with macrophages obtained from the peritoneal cavity. RESULTS: Nontreated hIAPP-Tg mice were glucose intolerant and exhibited impaired insulin secretion. Interestingly, AAT treatment improved glucose tolerance and restored the insulin secretory response to glucose in hIAPP-Tg mice. Moreover, AAT administration normalized the expression of the essential ß-cell genes MafA and Pdx1, which were downregulated in pancreatic islets from hIAPP-Tg mice. AAT prevented the formation of amyloid deposits and apoptosis in hIAPP-Tg islets cultured at high glucose concentrations. Since islet macrophages mediate hIAPP-induced ß-cell dysfunction, we investigated the effect of AAT in cocultures of macrophages and islet cells. AAT prevented hIAPP-induced ß-cell apoptosis in these cocultures without reducing the hIAPP-induced secretion of IL-1ß by macrophages. Remarkably, AAT protected ß-cells against the cytotoxic effects of conditioned medium from hIAPP-treated macrophages. Similarly, AAT also abrogated the cytotoxic effects of exogenous proinflammatory cytokines on pancreatic ß-cells. CONCLUSIONS: These results demonstrate that treatment with AAT improves glucose homeostasis in mice overexpressing hIAPP and protects pancreatic ß-cells from the cytotoxic actions of hIAPP mediated by macrophages. These results support the use of AAT-based therapies to recover pancreatic ß-cell function for the treatment of T2D.


Asunto(s)
Células Secretoras de Insulina/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , alfa 1-Antitripsina/metabolismo , Amiloide/metabolismo , Animales , Apoptosis , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/fisiología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Transgénicos , alfa 1-Antitripsina/genética
8.
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
9.
Diabetes ; 66(12): 3029-3040, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28970284

RESUMEN

The pancreatic ß-cell transcriptome is highly sensitive to external signals such as glucose oscillations and stress cues. MicroRNAs (miRNAs) have emerged as key factors in gene expression regulation. Here, we aimed to identify miRNAs that are modulated by glucose in mouse pancreatic islets. We identified miR-708 as the most upregulated miRNA in islets cultured at low glucose concentrations, a setting that triggers a strong stress response. miR-708 was also potently upregulated by triggering endoplasmic reticulum (ER) stress with thapsigargin and in islets of ob/ob mice. Low-glucose induction of miR-708 was blocked by treatment with the chemical chaperone 4-phenylbutyrate, uncovering the involvement of ER stress in this response. An integrative analysis identified neuronatin (Nnat) as a potential glucose-regulated target of miR-708. Indeed, Nnat expression was inversely correlated with miR-708 in islets cultured at different glucose concentrations and in ob/ob mouse islets and was reduced after miR-708 overexpression. Consistent with the role of Nnat in the secretory function of ß-cells, miR-708 overexpression impaired glucose-stimulated insulin secretion (GSIS), which was recovered by NNAT overexpression. Moreover, miR-708 inhibition recovered GSIS in islets cultured at low glucose. Finally, miR-708 overexpression suppressed ß-cell proliferation and induced ß-cell apoptosis. Collectively, our results provide a novel mechanism of glucose regulation of ß-cell function and growth by repressing stress-induced miR-708.


Asunto(s)
Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/fisiología , MicroARNs/fisiología , Animales , Apoptosis , Células Cultivadas , Células Secretoras de Insulina/metabolismo , Masculino , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , MicroARNs/análisis , Proteínas del Tejido Nervioso/análisis , Factor de Transcripción CHOP/genética
10.
Sci Rep ; 7(1): 11643, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28912479

RESUMEN

Jarid2 is a component of the Polycomb Repressor complex 2 (PRC2), which is responsible for genome-wide H3K27me3 deposition, in embryonic stem cells. However, Jarid2 has also been shown to exert pleiotropic PRC2-independent actions during embryogenesis. Here, we have investigated the role of Jarid2 during pancreas development. Conditional ablation of Jarid2 in pancreatic progenitors results in reduced endocrine cell area at birth due to impaired endocrine cell differentiation and reduced prenatal proliferation. Inactivation of Jarid2 in endocrine progenitors demonstrates that Jarid2 functions after endocrine specification. Furthermore, genome-wide expression analysis reveals that Jarid2 is required for the complete activation of the insulin-producing ß-cell differentiation program. Jarid2-deficient pancreases exhibit impaired deposition of RNAPII-Ser5P, the initiating form of RNAPII, but no changes in H3K27me3, at the promoters of affected endocrine genes. Thus, our study identifies Jarid2 as a fine-tuner of gene expression during late stages of pancreatic endocrine cell development. These findings are relevant for generation of transplantable stem cell-derived ß-cells.


Asunto(s)
Diferenciación Celular/genética , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Complejo Represivo Polycomb 2/genética , Células Madre/citología , Células Madre/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proliferación Celular , Células Endocrinas/citología , Células Endocrinas/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , ARN Polimerasa II/metabolismo , Transcripción Genética , Transcriptoma
11.
FASEB J ; 31(12): 5296-5306, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28821639

RESUMEN

Human islet amyloid polypeptide (hIAPP) aggregation is associated with ß-cell dysfunction and death in type 2 diabetes (T2D). we aimed to determine whether in vivo treatment with chemical chaperone 4-phenylbutyrate (PBA) ameliorates hIAPP-induced ß-cell dysfunction and islet amyloid formation. Oral administration of PBA in hIAPP transgenic (hIAPP Tg) mice expressing hIAPP in pancreatic ß cells counteracted impaired glucose homeostasis and restored glucose-stimulated insulin secretion. Moreover, PBA treatment almost completely prevented the transcriptomic alterations observed in hIAPP Tg islets, including the induction of genes related to inflammation. PBA also increased ß-cell viability and improved insulin secretion in hIAPP Tg islets cultured under glucolipotoxic conditions. Strikingly, PBA not only prevented but even reversed islet amyloid deposition, pointing to a direct effect of PBA on hIAPP. This was supported by in silico calculations uncovering potential binding sites of PBA to monomeric, dimeric, and pentameric fibrillar structures, and by in vitro assays showing inhibition of hIAPP fibril formation by PBA. Collectively, these results uncover a novel beneficial effect of PBA on glucose homeostasis by restoring ß-cell function and preventing amyloid formation in mice expressing hIAPP in ß cells, highlighting the therapeutic potential of PBA for the treatment of T2D.-Montane, J., de Pablo, S., Castaño, C., Rodríguez-Comas, J., Cadavez, L., Obach, M., Visa, M., Alcarraz-Vizán, G., Sanchez-Martinez, M., Nonell-Canals, A., Parrizas, M., Servitja, J.-M., Novials, A. Amyloid-induced ß-cell dysfunction and islet inflammation are ameliorated by 4-phenylbutyrate (PBA) treatment.


Asunto(s)
Amiloide/toxicidad , Células Secretoras de Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Fenilbutiratos/farmacología , Animales , Prueba de Tolerancia a la Glucosa , Humanos , Inmunohistoquímica , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/ultraestructura , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Cell Metab ; 25(6): 1390-1399.e6, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28591639

RESUMEN

Proopiomelanocortin (POMC) neurons are critical sensors of nutrient availability implicated in energy balance and glucose metabolism control. However, the precise mechanisms underlying nutrient sensing in POMC neurons remain incompletely understood. We show that mitochondrial dynamics mediated by Mitofusin 1 (MFN1) in POMC neurons couple nutrient sensing with systemic glucose metabolism. Mice lacking MFN1 in POMC neurons exhibited defective mitochondrial architecture remodeling and attenuated hypothalamic gene expression programs during the fast-to-fed transition. This loss of mitochondrial flexibility in POMC neurons bidirectionally altered glucose sensing, causing abnormal glucose homeostasis due to defective insulin secretion by pancreatic ß cells. Fed mice lacking MFN1 in POMC neurons displayed enhanced hypothalamic mitochondrial oxygen flux and reactive oxygen species generation. Central delivery of antioxidants was able to normalize the phenotype. Collectively, our data posit MFN1-mediated mitochondrial dynamics in POMC neurons as an intrinsic nutrient-sensing mechanism and unveil an unrecognized link between this subset of neurons and insulin release.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/trasplante , Insulina/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Neuronas/metabolismo , Proopiomelanocortina , Animales , GTP Fosfohidrolasas/genética , Glucosa/genética , Insulina/genética , Secreción de Insulina , Ratones , Ratones Noqueados , Mitocondrias/genética
13.
Cell Mol Life Sci ; 74(15): 2827-2838, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28337562

RESUMEN

BACE2 (ß-site APP-cleaving enzyme 2) is a protease expressed in the brain, but also in the pancreas, where it seems to play a physiological role. Amyloidogenic diseases, including Alzheimer's disease and type 2 diabetes (T2D), share the accumulation of abnormally folded and insoluble proteins that interfere with cell function. In T2D, islet amyloid polypeptide (IAPP) deposits have been shown to be a pathogenic key feature of the disease. The aim of the present study was to investigate the effect of BACE2 modulation on ß-cell alterations in a mouse model of T2D induced by IAPP overexpression. Heterozygous mice carrying the human transcript of IAPP (hIAPP-Tg) were used as a model to study the deleterious effects of IAPP upon ß-cell function. These animals showed glucose intolerance and impaired insulin secretion. When crossed with BACE2-deficient mice, the animals presented a significant improvement in glucose tolerance accompanied with an enhanced insulin secretion, as compared to hIAPP-Tg mice. BACE2 deficiency also partially reverted gene expression changes observed in islets from hIAPP-Tg mice, including a set of genes related to inflammation. Moreover, homozygous hIAPP mice presented a severe hyperglycemia and a high lethality rate from 8 weeks onwards due to a massive destruction of ß-cell mass. This process was significantly reduced when crossed with the BACE2-KO model, improving the survival rate of the animals. Altogether, the absence of BACE2 ameliorates glucose tolerance defects induced by IAPP overexpression in the ß-cell and promotes ß-cell survival. Thus, targeting BACE2 may represent a promising therapeutic strategy to improve ß-cell function in T2D.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/genética , Ácido Aspártico Endopeptidasas/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Regulación hacia Abajo , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Regulación hacia Arriba , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Proliferación Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transcriptoma
14.
Endocrinology ; 157(11): 4158-4171, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27623287

RESUMEN

Alterations during development of metabolic key organs such as the endocrine pancreas affect the phenotype later in life. There is evidence that in utero or perinatal exposure to bisphenol-A (BPA) leads to impaired glucose metabolism during adulthood. However, how BPA exposure during pregnancy affects pancreatic ß-cell growth and function in offspring during early life has not been explored. We exposed pregnant mice to either vehicle (control) or BPA (10 and 100 µg/kg·d, BPA10 and BPA100) and examined offspring on postnatal days (P) P0, P21, P30, and P120. BPA10 and BPA100 mice presented lower birth weight than control and subsequently gained weight until day 30. At that age, concentration of plasma insulin, C-peptide, and leptin were increased in BPA-exposed animals in the nonfasting state. Insulin secretion and content were diminished in BPA10 and maintained in BPA100 compared with control. A global gene expression analysis indicated that genes related with cell division were increased in islets from BPA-treated animals. This was associated with an increase in pancreatic ß-cell mass at P0, P21, and P30 together with increased ß-cell proliferation and decreased apoptosis. On the contrary, at P120, BPA-treated animals presented either equal or decreased ß-cell mass compared with control and altered fasting glucose levels. These data suggest that in utero exposure to environmentally relevant doses of BPA alters the expression of genes involved in ß-cell growth regulation, incrementing ß-cell mass/area, and ß-cell proliferation during early life. An excess of insulin signaling during early life may contribute to impaired glucose tolerance during adulthood.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Células Secretoras de Insulina/fisiología , Exposición Materna/efectos adversos , Fenoles/toxicidad , Animales , Apoptosis/efectos de los fármacos , Péptido C/sangre , Proliferación Celular/efectos de los fármacos , Ayuno/sangre , Femenino , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Leptina/sangre , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal , Útero/efectos de los fármacos
15.
Mol Cell Endocrinol ; 420: 57-65, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26607804

RESUMEN

Human islet amyloid polypeptide (hIAPP) is the major component of amyloid deposits in islets of type 2 diabetic patients. hIAPP misfolding and aggregation is one of the factors that may lead to ß-cell dysfunction and death. Endogenous chaperones are described to be important for the folding and functioning of proteins. Here, we examine the effect of the endoplasmic reticulum chaperone protein disulfide isomerase (PDI) on ß-cell dysfunction. Among other chaperones, PDI was found to interact with hIAPP in human islet lysates. Furthermore, intrinsically recovered PDI levels were able to restore the effect of high glucose- and palmitate-induced ß-cell dysfunction by increasing 3.9-fold the glucose-stimulated insulin secretion levels and restoring insulin content up to basal control values. Additionally, PDI transduction decreased induced apoptosis by glucolipotoxic conditions. This approach could reveal a new therapeutic target and aid in the development of strategies to improve ß-cell dysfunction in type 2 diabetic patients.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Amiloide/metabolismo , Animales , Apoptosis/efectos de los fármacos , Glucosa/farmacología , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones Transgénicos , Chaperonas Moleculares/metabolismo , Ácido Palmítico/farmacología , Unión Proteica/efectos de los fármacos , Extractos de Tejidos/metabolismo , Transducción Genética
16.
FASEB J ; 29(7): 2970-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25808537

RESUMEN

The toxic effects of human islet amyloid polypeptide (IAPP) on pancreatic islets have been widely studied. However, much less attention has been paid to the physiologic actions of IAPP on pancreatic ß cells, which secrete this peptide together with insulin upon glucose stimulation. Here, we aimed to explore the signaling pathways and mitogenic actions of IAPP on ß cells. We show that IAPP activated Erk1/2 and v-akt murine thymoma viral oncogene homolog 1 (Akt) at the picomolar range (10-100 pM) in mouse pancreatic islets and MIN6 ß cells cultured at low glucose concentrations. In contrast, IAPP decreased the induction of these pathways by high glucose levels. Consistently, IAPP induced a 1.7-fold increase of ß-cell proliferation at low-glucose conditions, whereas it reduced ß-cell proliferation at high glucose levels. Strikingly, the specific antagonist of the IAPP receptor AC187 (100 nM) decreased the activation of Erk1/2 and Akt and reduced ß-cell proliferation by 24% in glucose-stimulated ß cells, uncovering a key role of endogenously released IAPP in ß-cell responses to glucose. We conclude that exogenously added IAPP exerts a dual effect on ß-cell mitogenic signaling and proliferation, depending on the glucose concentration. Importantly, secreted IAPP contributes to the signaling and mitogenic response of ß cells to glucose through an autocrine mechanism.


Asunto(s)
Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Animales , Comunicación Autocrina/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/antagonistas & inhibidores , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/metabolismo
17.
FASEB J ; 29(1): 95-104, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25342134

RESUMEN

BACE2 (ß-site APP-cleaving enzyme 2) is a protease localized in the brain, where it appears to play a role in the development of Alzheimer disease (AD). It is also found in the pancreas, although its biologic function is not fully known. Amyloidogenic diseases, including AD and type 2 diabetes mellitus (T2D), share the accumulation of abnormally folded and insoluble proteins that interfere with cell function. Islet amyloid polypeptide (IAPP) deposits are a key pathogenic feature of T2D. Within this context, we found by global gene expression profiling that BACE2 was up-regulated in the rat pancreatic ß-cell line INS1E stably transfected with human IAPP gene (hIAPP-INS1E). Glucose-stimulated insulin secretion (GSIS) in hIAPP-INS1E cells was 30% lower than in INS1E cells. Additionally, INS1E cells transfected with a transient overexpression of BACE2 showed a 60% decrease in proliferation, a 3-fold increase in reactive oxygen species production, and a 25% reduction in GSIS compared to control cells. Remarkably, silencing of endogenous BACE2 in hIAPP-INS1E cells resulted in a significant improvement in GSIS (3-fold increase vs. untransfected cells), revealing the significant role of BACE2 expression in ß-cell function. Thus, BACE2 inhibition may be useful to recover insulin secretion in hIAPP-INS1E defective cells and may be proposed as a therapeutic target for T2D.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Línea Celular , Diabetes Mellitus Tipo 2/fisiopatología , Perfilación de la Expresión Génica , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
18.
J Clin Endocrinol Metab ; 100(3): E407-15, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25532038

RESUMEN

CONTEXT: Diabetes is frequently diagnosed late, when the development of complications is almost inevitable, decreasing the quality of life of patients. However, early detection of affected individuals would allow the implementation of timely and effective therapies. OBJECTIVE: Here we set to describe the profile of circulating microRNAs (miRNAs) in prediabetic patients with the intention of identifying novel diagnostic and therapeutic tools. DESIGN: We used real-time RT-PCR to measure the abundance of 176 miRNAs in serum of a cohort of 92 control and prediabetic individuals with either impaired fasting glucose or impaired glucose tolerance, as well as newly diagnosed diabetic patients. We validated the results in a second cohort of control and prediabetic subjects undergoing a therapeutic exercise intervention, as well as in a mouse model of glucose intolerance. RESULTS: We identified two miRNAs, miR-192 and miR-193b, whose abundance is significantly increased in the prediabetic state but not in diabetic patients. Strikingly, these miRNAs are also increased in plasma of glucose-intolerant mice. Moreover, circulating levels of miR-192 and miR-193b return to baseline in both prediabetic humans and glucose-intolerant mice undergoing a therapeutic intervention consisting in chronic exercise, which succeeded in normalizing metabolic parameters. CONCLUSIONS: Our data show that the pattern of circulating miRNAs is modified by defects in glucose metabolism in a similar manner in mice and humans. This circulating miRNA signature for prediabetes could be used as a new diagnostic tool, as well as to monitor response to intervention.


Asunto(s)
Biomarcadores/sangre , Terapia por Ejercicio , MicroARNs/sangre , Estado Prediabético/sangre , Estado Prediabético/terapia , Animales , Ejercicio Físico/fisiología , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estado Prediabético/genética , Transcriptoma
19.
PLoS One ; 9(7): e101797, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25010593

RESUMEN

In type 2 diabetes, beta-cell dysfunction is thought to be due to several causes, one being the formation of toxic protein aggregates called islet amyloid, formed by accumulations of misfolded human islet amyloid polypeptide (hIAPP). The process of hIAPP misfolding and aggregation is one of the factors that may activate the unfolded protein response (UPR), perturbing endoplasmic reticulum (ER) homeostasis. Molecular chaperones have been described to be important in regulating ER response to ER stress. In the present work, we evaluate the role of chaperones in a stressed cellular model of hIAPP overexpression. A rat pancreatic beta-cell line expressing hIAPP exposed to thapsigargin or treated with high glucose and palmitic acid, both of which are known ER stress inducers, showed an increase in ER stress genes when compared to INS1E cells expressing rat IAPP or INS1E control cells. Treatment with molecular chaperone glucose-regulated protein 78 kDa (GRP78, also known as BiP) or protein disulfite isomerase (PDI), and chemical chaperones taurine-conjugated ursodeoxycholic acid (TUDCA) or 4-phenylbutyrate (PBA), alleviated ER stress and increased insulin secretion in hIAPP-expressing cells. Our results suggest that the overexpression of hIAPP induces a stronger response of ER stress markers. Moreover, endogenous and chemical chaperones are able to ameliorate induced ER stress and increase insulin secretion, suggesting that improving chaperone capacity can play an important role in improving beta-cell function in type 2 diabetes.


Asunto(s)
Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Chaperonas Moleculares/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/farmacología , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Ácido Palmítico/farmacología , Ratas , Tapsigargina/farmacología , Factor de Transcripción CHOP/deficiencia , Factor de Transcripción CHOP/genética
20.
Exp Cell Res ; 322(2): 236-48, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24503054

RESUMEN

Diabetes is caused by loss or dysfunction of pancreatic beta cells. Generation of beta cells in vitro is a promising strategy to develop a full-scale cell therapy against diabetes, and the development of methods without gene transfer may provide safer protocols for human therapy. Here we show that thyroid hormone receptors are expressed in embryonic murine pancreas. Addition of the thyroid hormone T3 in an ex vivo culture model of embryonic (E12.5) dorsal pancreas, mimicking embryonic pancreatic development, promoted an increase of ductal cell number at expenses of the acinar compartment. Double labeled cells expressing specific markers for ductal and acinar cells were observed, suggesting cell reprogramming. Increased mRNA levels of the pro-endocrine gene Ngn3 and an increased number of beta cells were detected in cultures treated previously with T3 suggesting that ductal cells promoted by T3 can subsequently differentiate into endocrine cells. So, indirectly, T3 induced endocrine differentiation. Moreover, T3 induced the expression of the pro-endocrine gene Ngn3 in the acinar 266-6 cell line. The pro-endocrine effect of T3 in the pancreatic explants and in the acinar cell line, was abrogated by the Akt inhibitor Ly294002 indicating the involvement of Akt signaling in this process. Altogether we show numerous evidences that define T3 as a promising candidate to generate endocrine cells from exocrine tissue, using ectopically gene expression free protocols, for cell therapy against diabetes.


Asunto(s)
Células Acinares/citología , Células Secretoras de Insulina/citología , Páncreas/embriología , Conductos Pancreáticos/citología , Triyodotironina/farmacología , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Morfolinas/farmacología , Técnicas de Cultivo de Órganos , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Conductos Pancreáticos/efectos de los fármacos , Conductos Pancreáticos/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Hormona Tiroidea/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...