Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 295(45): 15245-15252, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-32855238

RESUMEN

Type-2 diabetes (T2D) is a global disease caused by the inability of pancreatic ß-cells to secrete adequate insulin. However, the molecular mechanisms underlying the failure of ß-cells to respond to glucose in T2D remains unknown. Here, we investigated the relative contribution of UDP-glucose (UDP-G), a P2Y14-specific agonist, in the regulation of insulin release using human isolated pancreatic islets and INS-1 cells. P2Y14 was expressed in both human and rodent pancreatic ß-cells. Dose-dependent activation of P2Y14 by UDP-G suppressed glucose-stimulated insulin secretion (GSIS) and knockdown of P2Y14 abolished the UDP-G effect. 12-h pretreatment of human islets with pertussis-toxin (PTX) improved GSIS and prevented the inhibitory effect of UDP-G on GSIS. UDP-G on GSIS suppression was associated with suppression of cAMP in INS-1 cells. UDP-G decreased the reductive capacity of nondiabetic human islets cultured at 5 mm glucose for 72 h and exacerbated the negative effect of 20 mm glucose on the cell viability during culture period. T2D donor islets displayed a lower reductive capacity when cultured at 5 mm glucose for 72 h that was further decreased in the presence of 20 mm glucose and UDP-G. Presence of a nonmetabolizable cAMP analog during culture period counteracted the effect of glucose and UDP-G. Islet cultures at 20 mm glucose increased apoptosis, which was further amplified when UDP-G was present. UDP-G modulated glucose-induced proliferation of INS-1 cells. The data provide intriguing evidence for P2Y14 and UDP-G's role in the regulation of pancreatic ß-cell function.


Asunto(s)
AMP Cíclico/biosíntesis , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Toxina del Pertussis/farmacología , Uridina Difosfato Glucosa/antagonistas & inhibidores , Animales , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas , Células Tumorales Cultivadas , Uridina Difosfato Glucosa/metabolismo
2.
Pflugers Arch ; 471(4): 633-645, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30767071

RESUMEN

We have recently shown that the G protein-coupled receptor 142 (GPR142) is expressed in both rodent and human pancreatic ß-cells. Herein, we investigated the cellular distribution of GPR142 within islets and the effects of selective agonists of GPR142 on glucose-stimulated insulin secretion (GSIS) in the mouse islets and INS-1832/13 cells. Double-immunostaining revealed that GPR142 immunoreactivity in islets mainly occurs in insulin-positive cells. Potentiation of GSIS by GPR142 activation was accompanied by increased cAMP content in INS-1832/13 cells. PKA/Epac inhibition markedly suppressed the effect of GPR142 activation on insulin release. Gpr142 knockdown (Gpr142-KD) in islets was accompanied by elevated release of MCP-1, IFNγ, and TNFα during culture period and abolished the modulatory effect of GPR142 activation on the GSIS. Gpr142-KD had no effect on Ffar1, Ffar2, or Ffar3 mRNA while reducing Gpr56 and increasing Tlr5 and Tlr7 mRNA expression. Gpr142-KD was associated with an increased expression of Chrebp, Txnip, RhoA, and mitochondrial Vdac1 concomitant with a reduced Pdx1, Pax6, and mitochondrial Vdac2 mRNA levels. Long-term exposure of INS-1832/13 cells to hyperglycemia reduced Gpr142 and Vdac2 while increased Chrebp, Txnip, and Vdac1 mRNA expression. GPR142 agonists or Bt2-cAMP counteracted this effect. Glucotoxicity-induced decrease of cell viability in Gpr142-KD INS-1 cells was not affected by GPR142-agonists while Bt2-cAMP prevented it. The results show the importance of Gpr142 in the maintenance of pancreatic ß-cell function in rodents and that GPR142 agonists potentiate GSIS by an action, which most likely is due to increased cellular generation of second messenger molecule cAMP.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Roedores/metabolismo , Animales , Supervivencia Celular/fisiología , AMP Cíclico/metabolismo , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina/fisiología , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
3.
Biochem Biophys Res Commun ; 520(2): 473-478, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31607476

RESUMEN

The possible implication of the gasotransmitters NO and CO for the development of diabetes remains unresolved. Our previous investigations in rodents suggested NO being inhibitory, and CO stimulatory, to glucose-stimulated insulin secretion (GSIS). Here we studied the possible role of these gasotransmitters in both murine and human type 2 diabetes (T2D) by mapping the expression pattern of neural nitric oxide synthase (nNOS), inducible NOS (iNOS), constitutive heme oxygenase (HO-2), and inducible HO (HO-1) in isolated pancreatic islets. Two variants of obese murine diabetes with distinct phenotype, the db/db and the ob/ob mouse, were studied at the initiation of the diabetic condition. Plasma glucose and plasma insulin were recorded and ß-cell expression levels of the different enzymes were measured with confocal microscopy and fluorescence intensity recordings. In human islets taken from nondiabetic controls (ND) and type 2 diabetes (T2D) the expression of the enzymes was analyzed by RNA-sequencing and qPCR. At the initiation of murine diabetes plasma glucose was slightly increased, whereas plasma insulin was extremely enhanced in both db/db and ob/ob mice. The ß-cell expression of nNOS and iNOS was markedly increased over controls in db/db mice, known to develop severe diabetes, while it was very low in ob/ob mice, known to develop mild diabetes. HO-2 expression was unaffected in db/db and modestly decreased in ob/ob mice. HO-1 expression was slightly enhanced in ob/ob, but, in contrast, extremely enhanced in db/db mice, suggesting a counteracting, antidiabetic action by CO. Moreover, the diabetic pattern of highly increased nNOS, iNOS and HO-1 expression seen in db/db mice was also fully recognized in human T2D islets. These results suggest that increased expression of the NOS-enzymes, especially an early upregulation of nNOS, could be involved in the initial development of the severe diabetes of db/db mice as well as in human T2D. Hence, nNOS, iNOS and HO-1 might be regarded as interesting targets to take into consideration in the early treatment of a diabetic condition in different variants of T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Células Secretoras de Insulina/metabolismo , Animales , Monóxido de Carbono/metabolismo , Diabetes Mellitus Experimental/enzimología , Femenino , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Células Secretoras de Insulina/enzimología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo
4.
Cell Mol Life Sci ; 75(16): 3039-3050, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29455414

RESUMEN

INTRODUCTION: Islets synthesise and secrete numerous peptides, some of which are known to be important regulators of islet function and glucose homeostasis. In this study, we quantified mRNAs encoding all peptide ligands of islet G protein-coupled receptors (GPCRs) in isolated human and mouse islets and carried out in vitro islet hormone secretion studies to provide functional confirmation for the species-specific role of peptide YY (PYY) in mouse islets. MATERIALS AND METHODS: GPCR peptide ligand mRNAs in human and mouse islets were quantified by quantitative real-time PCR relative to the reference genes ACTB, GAPDH, PPIA, TBP and TFRC. The pathways connecting GPCR peptide ligands with their receptors were identified by manual searches in the PubMed, IUPHAR and Ingenuity databases. Distribution of PYY protein in mouse and human islets was determined by immunohistochemistry. Insulin, glucagon and somatostatin secretion from islets was measured by radioimmunoassay. RESULTS: We have quantified GPCR peptide ligand mRNA expression in human and mouse islets and created specific signalomes mapping the pathways by which islet peptide ligands regulate human and mouse GPCR signalling. We also identified species-specific islet expression of several GPCR ligands. In particular, PYY mRNA levels were ~ 40,000-fold higher in mouse than human islets, suggesting a more important role of locally secreted Pyy in mouse islets. This was confirmed by IHC and functional experiments measuring insulin, glucagon and somatostatin secretion. DISCUSSION: The detailed human and mouse islet GPCR peptide ligand atlases will allow accurate translation of mouse islet functional studies for the identification of GPCR/peptide signalling pathways relevant for human physiology, which may lead to novel treatment modalities of diabetes and metabolic disease.


Asunto(s)
Islotes Pancreáticos/metabolismo , Péptido YY/metabolismo , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Expresión Génica , Humanos , Inmunohistoquímica , Ligandos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Péptido YY/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Biochem Biophys Res Commun ; 495(1): 1426-1431, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29180017

RESUMEN

Osteopontin (OPN) is involved in various physiological processes and also implicated in multiple pathological states. It has been suggested that OPN may have a role in type 2 diabetes (T2D) by protecting pancreatic islets and interaction with incretins. However, the regulation and function of OPN in islets, especially in humans, remains largely unexplored. In this study, we performed our investigations on both diabetic mouse model SUR1-E1506K+/+ and islets from human donors. We demonstrated that OPN protein, secretion and gene expression was elevated in the diabetic SUR1-E1506K+/+ islets. We also showed that high glucose and incretins simultaneously stimulated islet OPN secretion. In islets from human cadaver donors, OPN gene expression was elevated in diabetic islets, and externally added OPN significantly increased glucose-stimulated insulin secretion (GSIS) from diabetic but not normal glycemic donors. The increase in GSIS by OPN in diabetic human islets was Ca2+ dependent, which was abolished by Ca2+-channel inhibitor isradipine. Furthermore, we also confirmed that OPN promoted cell metabolic activity when challenged by high glucose. These observations provided evidence on the protective role of OPN in pancreatic islets under diabetic condition, and may point to novel therapeutic targets for islet protection in T2D.


Asunto(s)
Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Osteopontina/metabolismo , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
6.
Endocr J ; 64(3): 325-338, 2017 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-28228611

RESUMEN

Pancreatic islets express high levels of the orphan G-protein coupled receptor C5C (GPRC5C), the function of which remains to be established. Here we have examined the role of GPRC5C in the regulation of insulin secretion and ß-cell survival and proliferation using human and mouse pancreatic islets. The expression of GPRC5C was analysed by RNA-sequencing, qPCR, western blotting and confocal microscopy. Insulin secretion and cell viability were determined by RIA and MTS assays, respectively. GPRC5C mRNA expression and protein level were reduced in the islets from type-2 diabetic donors. RNA sequencing in human islets revealed GPRC5C expression correlated with the expression of genes controlling apoptosis, cell survival and proliferation. A reduction in Gprc5c mRNA and protein expression was observed in islets isolated from old mice (>46 weeks of age) compared to that in islets from newborn (<3 weeks) mice. Down-regulation of Gprc5c led to both moderately reduced glucose-stimulated insulin release and also reduced cAMP content in mouse islets. Potentiation of glucose-stimulated insulin secretion concomitant with enhanced islet cAMP level by all-trans retinoic acid (ATRA) was attenuated upon Gprc5c-KD. ATRA also increased [Ca+2]i in Huh7-cells. Gprc5c over expression in Huh7 cells was associated with increased ERK1/2 activity. Gprc5c-KD in clonal MIN6c4 cells reduced cell proliferation and in murine islets increased apoptosis and the sensitivity of primary islet cells to a cocktail of pro-apoptotic cytokines. Our results demonstrate that agents activating GPRC5C represent a novel modality for the treatment and/or prevention of diabetes by restoring and/or maintaining functional ß-cell mass.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Hipoglucemiantes/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Tretinoina/metabolismo , Envejecimiento , Animales , Animales Recién Nacidos , Apoptosis , Señalización del Calcio , Línea Celular , Diabetes Mellitus Tipo 2/patología , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Técnicas de Cultivo de Tejidos
7.
Nat Cell Biol ; 9(4): 453-60, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17369816

RESUMEN

Pancreatic islets have a central role in blood glucose homeostasis. In addition to insulin-producing beta-cells and glucagon-secreting alpha-cells, the islets contain somatostatin-releasing delta-cells. Somatostatin is a powerful inhibitor of insulin and glucagon secretion. It is normally secreted in response to glucose and there is evidence suggesting its release becomes perturbed in diabetes. Little is known about the control of somatostatin release. Closure of ATP-regulated K(+)-channels (K(ATP)-channels) and a depolarization-evoked increase in cytoplasmic free Ca(2+) concentration ([Ca(2+)](i)) have been proposed to be essential. Here, we report that somatostatin release evoked by high glucose (>or=10 mM) is unaffected by the K(ATP)-channel activator diazoxide and proceeds normally in K(ATP)-channel-deficient islets. Glucose-induced somatostatin secretion is instead primarily dependent on Ca(2+)-induced Ca(2+)-release (CICR). This constitutes a novel mechanism for K(ATP)-channel-independent metabolic control of pancreatic hormone secretion.


Asunto(s)
Canales de Calcio Tipo R/fisiología , Calcio/metabolismo , Glucosa/farmacología , Somatostatina/metabolismo , Animales , Calcio/farmacología , Canales de Calcio Tipo R/genética , Citofotometría , Diazóxido/farmacología , Relación Dosis-Respuesta a Droga , Electrofisiología , Inmunohistoquímica , Técnicas In Vitro , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Isradipino/farmacología , Compuestos Macrocíclicos/farmacología , Manoheptulosa/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Microscopía Confocal , Oxazoles/farmacología , Potasio/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/fisiología , Rianodina/farmacología , Células Secretoras de Somatostatina/efectos de los fármacos , Células Secretoras de Somatostatina/metabolismo
8.
Cell Metab ; 7(1): 57-67, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18177725

RESUMEN

CAPS1 and CAPS2 regulate dense-core vesicle release of transmitters and hormones in neuroendocrine cells, but their precise roles in the secretory process remain enigmatic. Here we show that CAPS2(-/-) and CAPS1(+/-);CAPS2(-/-) mice, despite having increased insulin sensitivity, are glucose intolerant and that this effect is attributable to a marked reduction of glucose-induced insulin secretion. This correlates with diminished Ca(2+)-dependent exocytosis, a reduction in the size of the morphologically docked pool, a decrease in the readily releasable pool of secretory vesicles, slowed granule priming, and suppression of second-phase (but not first-phase) insulin secretion. In beta cells of CAPS1(+/-);CAPS2(-/-) mice, the lowered insulin content and granule numbers were associated with an increase in lysosome numbers and lysosomal enzyme activity. We conclude that although CAPS proteins are not required for Ca(2+)-dependent exocytosis to proceed, they exert a modulatory effect on insulin granule priming, exocytosis, and stability.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/fisiología , Electrofisiología , Exocitosis , Inmunohistoquímica , Células Secretoras de Insulina/ultraestructura , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Transmisión , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Pancrelipasa/metabolismo , Pancrelipasa/ultraestructura
9.
Biochem Biophys Res Commun ; 441(3): 643-8, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24404583

RESUMEN

GPRC5B is an orphan receptor belonging to the group C family of G protein-coupled receptors (GPCRs). GPRC5B is abundantly expressed in both human and mouse pancreatic islets, and both GPRC5B mRNA and protein are up-regulated 2.5-fold in islets from organ donors with type 2 diabetes. Expression of Gprc5b is 50% lower in islets isolated from newborn (<3 weeks) than in adult (>36 weeks) mice. Lentiviral shRNA-mediated down-regulation of Gprc5b in intact islets from 12 to 16 week-old mice strongly (2.5-fold) increased basal (1 mmol/l) and moderately (40%) potentiated glucose (20 mmol/l) stimulated insulin secretion and also enhanced the potentiating effect of glutamate on insulin secretion. Downregulation of Gprc5b protected murine insulin-secreting clonal MIN6 cells against cytokine-induced apoptosis. We propose that increased expression of GPRC5B contributes to the reduced insulin secretion and b-cell viability observed in type-2 diabetes. Thus, pharmacological targeting of GPRC5B might provide a novel means therapy for the treatment and prevention of type-2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Anciano , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Femenino , Humanos , Secreción de Insulina , Islotes Pancreáticos , Masculino , Ratones , Persona de Mediana Edad , Receptores Acoplados a Proteínas G/genética , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo
10.
FEBS J ; 290(3): 620-648, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-34847289

RESUMEN

Diabetes is a complex and multifactorial disease that affects millions of people worldwide, reducing the quality of life significantly, and results in grave consequences for our health care system. In type 2 diabetes (T2D), the lack of ß-cell compensatory mechanisms overcoming peripherally developed insulin resistance is a paramount factor leading to disturbed blood glucose levels and lipid metabolism. Impaired ß-cell functions and insulin resistance have been studied extensively resulting in a good understanding of these pathways but much less is known about interorgan crosstalk, which we define as signaling between tissues by secreted factors. Besides hormones and organokines, dysregulated blood glucose and long-lasting hyperglycemia in T2D is associated with changes in metabolism with metabolites from different tissues contributing to the development of this disease. Recent data suggest that metabolites, such as lipids including free fatty acids and amino acids, play important roles in the interorgan crosstalk during the development of T2D. In general, metabolic remodeling affects physiological homeostasis and impacts the development of T2D. Hence, we highlight the importance of metabolic interorgan crosstalk in this review to gain enhanced knowledge of the pathophysiology of T2D, which may lead to new therapeutic approaches to treat this disease.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Humanos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Resistencia a la Insulina/genética , Glucemia/metabolismo , Calidad de Vida , Aminoácidos/metabolismo
11.
Biomolecules ; 13(3)2023 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-36979492

RESUMEN

The activation of G Protein-Coupled Receptor 56 (GPR56), also referred to as Adhesion G-Protein-Coupled Ceceptor G1 (ADGRG1), by Collagen Type III (Coll III) prompts cell growth, proliferation, and survival, among other attributes. We investigated the signaling cascades mediating this functional effect in relation to the mitochondrial outer membrane voltage-dependent anion Channel-1 (VDAC1) expression in pancreatic ß-cells. GPR56KD attenuated the Coll III-induced suppression of P70S6K, JNK, AKT, NFκB, STAT3, and STAT5 phosphorylation/activity in INS-1 cells cultured at 20 mM glucose (glucotoxicity) for 72 h. GPR56-KD also increased Chrebp, Txnip, and Vdac1 while decreasing Vdac2 mRNA expression. In GPR56-KD islet ß-cells, Vdac1 was co-localized with SNAP-25, demonstrating its plasma membrane translocation. This resulted in ATP loss, reduced cAMP production and impaired glucose-stimulated insulin secretion (GSIS) in INS-1 and human EndoC ßH1 cells. The latter defects were reversed by an acute inhibition of VDAC1 with an antibody or the VDAC1 inhibitor VBIT-4. We demonstrate that Coll III potentiates GSIS by increasing cAMP and preserving ß-cell functionality under glucotoxic conditions in a GPR56-dependent manner by attenuating the inflammatory response. These results emphasize GPR56 and VDAC1 as drug targets in conditions with impaired ß-cell function.


Asunto(s)
Islotes Pancreáticos , Receptores Acoplados a Proteínas G , Canal Aniónico 1 Dependiente del Voltaje , Humanos , Adenosina Trifosfato/metabolismo , Membrana Celular/metabolismo , Colágeno Tipo III/metabolismo , Glucosa/farmacología , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/genética , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
12.
Biochem Biophys Res Commun ; 417(4): 1219-23, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22227186

RESUMEN

Recent studies of isolated human islets have shown that glucose induces hormone release with repetitive pulses of insulin and somatostatin in antisynchrony with those of glucagon. Since the mouse is the most important animal model we studied the temporal relation between hormones released from mouse islets. Batches of 5-10 islets were perifused and the hormones measured with radioimmunoassay in 30s fractions. At 3mM glucose, hormone secretion was stable with no detectable pulses of glucagon, insulin or somatostatin. Increase of glucose to 20mM resulted in an early secretory phase with a glucagon peak followed by peaks of insulin and somatostatin. Subsequent hormone secretion was pulsatile with a periodicity of 5min. Cross-correlation analyses showed that the glucagon pulses were antisynchronous to those of insulin and somatostatin. In contrast to the marked stimulation of insulin and somatostatin secretion, the pulsatility resulted in inhibition of overall glucagon release. The cytoarchitecture of mouse islets differs from that of human islets, which may affect the interactions between the hormone-producing cells. Although indicating that paracrine regulation is important for the characteristic patterns of pulsatile hormone secretion, the mouse data mimic those of human islets with more than 20-fold variations of the insulin/glucagon ratio. The data indicate that the mouse serves as an appropriate animal model for studying the temporal relation between the islet hormones controlling glucose production in the liver.


Asunto(s)
Glucagón/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Somatostatina/metabolismo , Animales , Separación Celular , Células Cultivadas , Femenino , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Ratones , Modelos Animales
13.
Endocrine ; 76(3): 543-557, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35237909

RESUMEN

PURPOSE: Metformin (MF) intake associates with reduced levels of circulating low-density lipoprotein-cholesterol (LDL-C). This has been attributed to the activation of AMPK, which differentially regulates the expression of multiple genes involved in cholesterol synthesis and trafficking. However, the exact mechanism underlying the LDL-C lowering effect of MF remains ambiguous. METHODS: MF-treated Hep-G2 and HuH7 cells were evaluated for cell viability and the expression status of key lipid metabolism-related genes along with LDL-C uptake efficiency. RESULTS: MF treatment resulted in decreased expression and secretion of PCSK9, increased expression of LDLR and enhanced LDL-C uptake in hepatocytes. It also resulted in increased expression of activated AMPK (p-AMPK) and decreased expression of SREBP2 and HNF-1α proteins. Transcriptomic analysis of MF-treated Hep-G2 cells confirmed these findings and showed that other key lipid metabolism-related genes including those that encode apolipoproteins (APOB, APOC2, APOC3 and APOE), MTTP and LIPC are downregulated. Lastly, MF treatment associated with reduced HMG-CoA reductase expression and activity. CONCLUSIONS: These findings suggest that MF treatment reduces circulating LDL-C levels by suppressing PCSK9 expression and enhancing LDLR expression; hence the potential therapeutic utility of MF in hypercholesterolemia.


Asunto(s)
Metformina , Proproteína Convertasa 9 , Proteínas Quinasas Activadas por AMP/metabolismo , LDL-Colesterol , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Metformina/farmacología , Metformina/uso terapéutico , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Subtilisina/metabolismo , Subtilisina/farmacología
14.
Biochem Biophys Res Commun ; 404(1): 16-22, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21094635

RESUMEN

Many microRNAs (miRNAs) are known to be cell-type specific and are implicated in development of diseases. We investigated the global expression pattern of miRNAs in human pancreatic islets compared to liver and skeletal muscle, using bead-based technology and quantitative RT-PCR. In addition to the known islet-specific miR-375, we also found enrichment of miR-127-3p, miR-184, miR-195 and miR-493∗ in the pancreatic islets. The expression of miR-375, miR-127-3p, miR-184 and the liver-enriched miR-122 is positively correlated to insulin biosynthesis, while the expression of miR-127-3p and miR-184 is negatively correlated to glucose-stimulated insulin secretion (GSIS). These correlations were absent in islets of glucose intolerant donors (HbA1c ≥ 6.1). We suggest that the presence of an islet-specific miRNA network, which consists of at least miR-375, miR-127-3p and miR-184, potentially involved in insulin secretion. Our results provide new insight into miRNA-mediated regulation of insulin secretion in healthy and glucose intolerant subjects.


Asunto(s)
Intolerancia a la Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , MicroARNs/metabolismo , Humanos , Insulina/biosíntesis , Secreción de Insulina , Hígado/metabolismo , Músculo Esquelético/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 30(2): 218-24, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19965778

RESUMEN

OBJECTIVE: Hyperglycemia is a recognized risk factor for cardiovascular disease in diabetes. Recently, we reported that high glucose activates the Ca(2+)/calcineurin-dependent transcription factor nuclear factor of activated T cells (NFAT) in arteries ex vivo. Here, we sought to determine whether hyperglycemia activates NFAT in vivo and whether this leads to vascular complications. METHODS AND RESULTS: An intraperitoneal glucose-tolerance test in mice increased NFATc3 nuclear accumulation in vascular smooth muscle. Streptozotocin-induced diabetes resulted in increased NFATc3 transcriptional activity in arteries of NFAT-luciferase transgenic mice. Two NFAT-responsive sequences in the osteopontin (OPN) promoter were identified. This proinflammatory cytokine has been shown to exacerbate atherosclerosis and restenosis. Activation of NFAT resulted in increased OPN mRNA and protein in native arteries. Glucose-induced OPN expression was prevented by the ectonucleotidase apyrase, suggesting a mechanism involving the release of extracellular nucleotides. The calcineurin inhibitor cyclosporin A or the novel NFAT blocker A-285222 prevented glucose-induced OPN expression. Furthermore, diabetes resulted in higher OPN expression, which was significantly decreased by in vivo treatment with A-285222 for 4 weeks or prevented in arteries from NFATc3(-/-) mice. CONCLUSIONS: These results identify a glucose-sensitive transcription pathway in vivo, revealing a novel molecular mechanism that may underlie vascular complications of diabetes.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Angiopatías Diabéticas/etiología , Hiperglucemia/metabolismo , Músculo Liso Vascular/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteopontina/metabolismo , Animales , Apirasa/farmacología , Arterias/metabolismo , Sitios de Unión , Calcineurina/metabolismo , Inhibidores de la Calcineurina , Ciclosporina/farmacología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Angiopatías Diabéticas/metabolismo , Angiopatías Diabéticas/prevención & control , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/tratamiento farmacológico , Células Jurkat , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/deficiencia , Factores de Transcripción NFATC/genética , Osteopontina/deficiencia , Osteopontina/genética , Regiones Promotoras Genéticas , Pirazoles/farmacología , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Tiempo , Activación Transcripcional , Transfección , Uridina Trifosfato/metabolismo
16.
Cell Mol Life Sci ; 67(3): 445-53, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19915796

RESUMEN

Pancreatic beta-cell loss represents a key factor in the pathogenesis of diabetes. Since the influence of purinergic signaling in beta-cell apoptosis has not been much investigated, we examined the role of the ADP receptor P2Y(13) using the pancreatic insulinoma-cell line MIN6c4 as a model system. Real time-PCR revealed high expression of the ADP receptors P2Y(1) and P2Y(13). Adding the ADP analogue, 2MeSADP, to MIN6c4 cells induced calcium influx/mobilization and inhibition of cAMP production by activation of P2Y(1) and P2Y(13), respectively. 2MeSADP reduced cell proliferation and increased Caspase-3 activity; both these effects could be fully reversed by the P2Y(13) receptor antagonist MRS2211. We further discovered that blocking the P2Y(13) receptor results in enhanced ERK1/2, Akt/PKB and CREB phosphorylation mechanisms involved in beta-cell survival. These results indicate that P2Y(13) is a proapoptotic receptor in beta-cells as the P2Y(13) receptor antagonist MRS2211 is able to protect the cells from ADP induced apoptosis.


Asunto(s)
Apoptosis , Células Secretoras de Insulina/citología , Receptores Purinérgicos P2/metabolismo , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Calcio/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , AMP Cíclico/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Antagonistas del Receptor Purinérgico P2 , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y1
17.
Exp Clin Endocrinol Diabetes ; 129(12): 864-872, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31614370

RESUMEN

Silencing of Chl1 gene expression has been previously reported to reduce insulin secretion. Nevertheless, the mechanism underlying this effect remains unclear. In this study, we performed a serial of studies to investigate how Chl1 affects insulin secretion in INS-1 cells. RNA-sequencing was used to investigate the expression of CHL1 in human adipose, liver, muscle, and human islets. Silencing of Chl1 in INS-1 cells was done to assess its impact on the insulin secretion, content, cell viability, and apoptosis. In addition, gene set enrichment analysis (GSEA) was performed to identify possible molecular signatures that associate with Chl1 expression silencing.RNA sequencing data revealed a high expression of CHL1 in pancreatic islets and adipose tissues compared to liver and muscles tissues. Diabetic islets exhibited a lower expression of CHL1 as compared to non-diabetic islets. CHL1 expression was found to correlate positively with insulin secretory index, GLP1R but inversely with HbA1c and BMI. Silencing of Chl1 in INS-1 cells markedly reduced insulin content and secretion. The expression of key molecules of ß-cell function including Insulin, Pdx1, Gck, Glut2, and Insrß was down-regulated in Chl1-silenced cells at transcriptional and translational levels. Cell viability, apoptosis, and proliferation rate were not affected. GSEA showed that the insulin-signaling pathway was influenced in Chl1-silenced cells. Silencing of Chl1 impairs ß-cell function by disrupting the activity of key signaling pathways of importance for insulin biosynthesis and secretion.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Regulación de la Expresión Génica/fisiología , Secreción de Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Línea Celular , Regulación hacia Abajo , Humanos , Análisis de Secuencia de ARN , Transducción de Señal/fisiología
18.
Exp Clin Endocrinol Diabetes ; 129(6): 420-428, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31185507

RESUMEN

It well known that long-lasting hyperglycaemia disrupts neuronal function and leads to neuropathy and other neurodegenerative diseases. The α-ketoglutarate analogue (DMOG) and the caspase-inhibitor "Ac-LETD-CHO are potential neuroprotective molecules. Whether their protections may also extend glucotoxicity-induced neuropathy is not known. Herein, we evaluated the possible cell-protective effects of DMOG and Ac-LETD-CHO against hyperglycaemia-induced reactive oxygen species and apoptosis in ND7/23 neuronal cells. The impact of glucotoxicity on the expression of HIF-1α and a panel of micro-RNAs of significance in hyperglycaemia and apoptosis was also investigated.ND7/23 cells cultured under hyperglycaemic conditions showed decreased cell viability and elevated levels of ROS production in a dose- and time-dependent manner. However, presence DMOG (500 µM) and/or Ac-LETD-CHO (50 µM) counteracted this effect and increase cell viability concomitant with reduction in ROS production, DNA damage and apoptosis. AcLETD-CHO suppressed hyperglycaemia-induced caspase 3 activation in ND7/23 cells. Both DMOG and Ac-LETD-CHO increased HIF-1α expression paralleled with the suppression of miR-126-5p, miR-128-3p and miR-181 expression and upregulation of miR-26b, 106a-5p, 106b-5p, 135a-5p, 135b-5p, 138-5p, 199a-5p, 200a-3p and 200c-3p expression.We demonstrate a mechanistic link for the DMOG and Ac-LETD-CHO protection against hyperglycaemia-induced neuronal dysfunction, DNA damage and apoptosis and thereby propose that pharmacological agents mimicking these effects may represent a promising novel therapy for the hyperglycaemia-induced neuropathy.


Asunto(s)
Aminoácidos Dicarboxílicos/farmacología , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Neuropatías Diabéticas/prevención & control , Hiperglucemia/complicaciones , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Células Cultivadas , Humanos
19.
PLoS Biol ; 5(6): e143, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17503968

RESUMEN

Glucagon, secreted from pancreatic islet alpha cells, stimulates gluconeogenesis and liver glycogen breakdown. The mechanism regulating glucagon release is debated, and variously attributed to neuronal control, paracrine control by neighbouring beta cells, or to an intrinsic glucose sensing by the alpha cells themselves. We examined hormone secretion and Ca(2+) responses of alpha and beta cells within intact rodent and human islets. Glucose-dependent suppression of glucagon release persisted when paracrine GABA or Zn(2+) signalling was blocked, but was reversed by low concentrations (1-20 muM) of the ATP-sensitive K(+) (KATP) channel opener diazoxide, which had no effect on insulin release or beta cell responses. This effect was prevented by the KATP channel blocker tolbutamide (100 muM). Higher diazoxide concentrations (>/=30 muM) decreased glucagon and insulin secretion, and alpha- and beta-cell Ca(2+) responses, in parallel. In the absence of glucose, tolbutamide at low concentrations (<1 muM) stimulated glucagon secretion, whereas high concentrations (>10 muM) were inhibitory. In the presence of a maximally inhibitory concentration of tolbutamide (0.5 mM), glucose had no additional suppressive effect. Downstream of the KATP channel, inhibition of voltage-gated Na(+) (TTX) and N-type Ca(2+) channels (omega-conotoxin), but not L-type Ca(2+) channels (nifedipine), prevented glucagon secretion. Both the N-type Ca(2+) channels and alpha-cell exocytosis were inactivated at depolarised membrane potentials. Rodent and human glucagon secretion is regulated by an alpha-cell KATP channel-dependent mechanism. We propose that elevated glucose reduces electrical activity and exocytosis via depolarisation-induced inactivation of ion channels involved in action potential firing and secretion.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Glucosa/metabolismo , Canales de Potasio/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio Tipo N/metabolismo , Exocitosis/fisiología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Canales de Sodio/metabolismo
20.
J Clin Med Res ; 12(12): 758-772, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33447309

RESUMEN

The dreadful fear of the coronavirus disease 2019 (COVID-19), which is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with the deadly consequences, requires rapid development of pharmacological cures. The objective of this review is to speculate about possible pharmacological options, already available today to prevent or treat the COVID-19 in the early stage of its outbreak. A literature search across PubMed and internet was conducted. A number of studies dealing with COVID-19 were identified. The data elucidated that increased pro-inflammatory and decreased anti-inflammatory cytokines in combination with hypoxia, thromboembolism and pneumonia are involved in the pathogenesis of SARS-CoV-2 infection. Although many drugs has been tested in monotherapy regimen with varying outcome or without desirable effect, there is still hope for better results by simultaneously targeting the virus itself and its symptoms. Theoretically, a mixture of at least two available antiviral drugs in combination with other anti-pathogenic and immune system-enhancing drugs or combination of antiviral drugs with convalescent plasma seems likely to have much better effect than the monotherapy regimen of either of these drugs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA