Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomed Pharmacother ; 145: 112462, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34844105

RESUMEN

A previous study indicated that microRNA-378b (miR-378b) plays a critical role in controlling hepatic insulin resistance by targeting insulin receptor (IR) and p110α in alcoholic liver disease (ALD). Methyl ferulic acid (MFA), a bioactive ingredient in Securidaca inappendiculata Hassk rhizomes, exhibits multiple pharmacological activities. It has been reported that MFA ameliorates insulin resistance in ALD, whereas the underlying molecular mechanism remains unclear. The objective of study was to evaluate the influence of MFA on insulin sensitivity in ethanol-induced L-02 cells as well as alcohol-fed mice and illuminate the function of miR-378b-mediated phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway in system. MFA was found to remarkably down-regulate miR-378b level and increase IR and p110α expressions. Furthermore, the effect of MFA on modulating miR-378b/PI3K-AKT pathway to enhance insulin sensitivity was corroborated by overexpressing and inhibiting miR-378b. Taken together, MFA exhibited a positive effect against ALD by attenuating the inhibition of miR-378b on IR/p110α and partly activating the insulin signaling to alleviate alcohol-induced hepatic insulin resistance.


Asunto(s)
Ácidos Cafeicos/farmacología , Resistencia a la Insulina/fisiología , Hepatopatías Alcohólicas , MicroARNs/metabolismo , Securidaca , Animales , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hepatopatías Alcohólicas/tratamiento farmacológico , Hepatopatías Alcohólicas/metabolismo , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Fitoquímicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
2.
Front Pharmacol ; 11: 717, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508647

RESUMEN

Insulin resistance has been implicated in alcoholic liver disease. A previous study has shown that microRNAs (miRNAs) play a major role in the production, secretion, and function of insulin. MiRNAs are capable of repressing multiple target genes that in turn negatively regulate various physiological and pathological activities. However, current information on the biological function of miRNAs in insulin resistance is limited. The goal of the present study was to elucidate the role of miR-378b in alcohol-induced hepatic insulin resistance and its underlying mechanism. This study has observed that miR-378b is up-regulated in National Institute on Alcohol Abuse and Alcoholism (NIAAA) alcoholic mouse models as well as in ethanol-induced L-02 cells in vitro. Furthermore, miR-378b overexpression impaired the insulin signaling pathway, and inhibition of miR-378b improved insulin sensitivity in vivo and in vitro. A mechanistic study revealed that IR and p110α are direct targets of miR-378b. Together, these results suggest that miR-378b controls insulin sensitivity by targeting the insulin receptor (IR) as well as p110α and possibly play an inhibitory role in the development of insulin resistance, thereby providing insights into the development of novel diagnostic and treatment methods.

3.
Diabetes Obes Metab ; 21(10): 2219-2227, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31148332

RESUMEN

AIMS: To compare outcomes of glucagon-stimulated C-peptide tests (GSCTs) in people with latent autoimmune diabetes in adults (LADA) after a 21-month intervention with either insulin or the dipeptidyl peptidase-4 inhibitor sitagliptin. RESEARCH DESIGN AND METHODS: We included 64 glutamic acid decarboxylase (GAD) antibody-positive individuals, who were diagnosed with diabetes <3 years before the study, aged 30 to 70 years, and without clinical need for insulin treatment. We stratified participants by age and body mass index (BMI) and evaluated ß-cell function by GSCT after a 48-hour temporary withdrawal of study medication. RESULTS: Age at randomization (mean 53 years), BMI (mean 27 kg/m2 ) and metabolic markers were similar between treatment arms. Glycated haemoglobin concentrations during intervention did not differ between arms. Fasting C-peptide concentrations after the intervention were similar, as were stimulated C-peptide levels (0.82 ± 0.63 nmol/L after insulin, 0.82 ± 0.46 nmol/L after sitagliptin; nonsignificant). Autoimmunity in the study population (estimated from GAD antibody titres and positivity/no positivity for zinc transporter 8 and islet antigen 2 antibodies) affected the evolution of the GSCT results significantly, which deteriorated in participants with high but not in those with low autoimmunity. Adjustment using analysis of covariance for the degree of autoimmunity did not alter the findings of no difference between treatment arms. CONCLUSIONS: ß-cell function after intervention was similar in patients with insulin- and sitagliptin-treated LADA, regardless of the strength of autoimmunity. Further, participants with low levels of GAD antibodies did not experience progressive deterioration of ß-cell function over a 21-month period. Taken together, these findings could be useful for clinicians' choices of treatment in people with LADA.


Asunto(s)
Hipoglucemiantes/uso terapéutico , Células Secretoras de Insulina , Diabetes Autoinmune Latente del Adulto/tratamiento farmacológico , Adulto , Anciano , Autoinmunidad/fisiología , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Femenino , Humanos , Insulina/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Diabetes Autoinmune Latente del Adulto/fisiopatología , Masculino , Persona de Mediana Edad , Fosfato de Sitagliptina/uso terapéutico
4.
Int J Oncol ; 53(1): 225-236, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29749464

RESUMEN

The present study aimed to investigate the anti-apoptotic effects of methyl ferulic acid (MFA) on L-02 cell apoptosis induced by ethanol, and to elucidate the possible underlying mechanisms. L-02 cells were examined after being soaked in ethanol (400 mM) to allow the ethanol to permeate into the cells for 24 h. Cell survival was measured by MTT assay. Cell apoptosis was assessed by both flow cytometry and single-stranded DNA assays. Intracellular reactive oxygen species (ROS) production was determined using the 2',7'-dichlorofluorescein-diacetate dye. The protein expression levels of p38, p-p38, JNK, p-JNK, NADPH oxidase 4 (NOX4), p22, Bax and Bcl-2 were measured by western blot analysis. The mRNA expression levels of NOX4 and p22 were measured by RT-PCR. It was identified that MFA markedly suppressed the ethanol-induced apoptosis and necrosis of L-02 cells. In addition, MFA decreased the expression levels of superoxide dismutase, catalase and phospholipid hydroperoxide gluthione peroxidase, and downregulated the levels of Bax/Bcl-2 and the cleaved forms of caspase-3 in a dose- and time-dependent manner. This indicated that MFA attenuated the apoptosis of L-02 cells. MFA also decreased the elevated mRNA and protein expression levels of Nox4 and p22phox, and the production of intracellular ROS triggered by ethanol. Further analysis demonstrated that MFA significantly attenuated the phosphorylation of JNK and p38, which are major components of the mitogen-activated protein kinase (MAPK) pathways. On the whole, the findings of this study demonstrated that MFA attenuated the apoptotic cell death of L-02 cells by reducing the generation of ROS and inactivating the MAPK pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Cumáricos/administración & dosificación , Neoplasias Hepáticas/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , ADN de Cadena Simple/efectos de los fármacos , Etanol/toxicidad , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Exp Ther Med ; 15(3): 2228-2238, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29467841

RESUMEN

The present study aimed to investigate the hepatoprotective effects of methyl ferulic acid (MFA) against oxidative stress and apoptosis in acute liver injury induced by carbon tetrachloride (CCl4) in rats, as well as the underlying mechanisms. Sprague Dawley rats were treated with CCl4 after oral administration of MFA (25, 50, and 100 mg/kg) or dimethyl diphenyl bicarboxylate (200 mg/kg) for 7 days. The hepatoprotective effects of MFA were determined by analyzing serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities as well as changes of oxidant parameters. Histopathological analysis was performed to determine the degree of hepatic injury. The mechanisms were investigated by detecting the levels of NADPH oxidase (NOX) trans-membrane subunit NOX4, its ligand p22phox, as well as caspase3, cleaved caspase3, B-cell lymphoma (Bcl)-2, Bcl-2-associated X protein (Bax), tumor necrosis factor (TNF)-α, interleukin (IL)-1, reactive oxygen species (ROS), thiobarbituric acid-reactive substances (TBARS), total anti-oxidant capacity (TAC), phosphorylated J-Jun N-terminal kinase (p-JNK) and p-p38 mitogen-activated protein kinase (MAPK) using semi-quantitative polymerase chain reaction, western blot analysis and colorimetric assays. MFA treatment significantly decreased serum enzymatic activities of ALT and AST. MFA markedly increased activities of liver superoxide dismutase, catalase and glutathione peroxidase, and reduced the malondialdehyde concentration. Histopathological examination demonstrated that MFA reduced lipid degeneration, cytoplasmic vacuolization, necrosis and inflammatory cell infiltration in the liversof CCl4-treated rats. MFA treatment markedly inhibited the expression of inflammatory factors TNF-α and IL-1ß. Mechanistic study revealed that MFA decreased the TAC and the levels of ROS and TBARS. Furthermore, MFA treatment led to a reduction of the mRNA and protein expression of NOX4 and p22phox, as well as the protein levels of caspase3, cleaved caspase-3 and Bax, and an upregulation of p-JNK, p-p38 MAPK and Bcl-2 proteins in the liver. The present study demonstrated that MFA has hepatoprotective effects against CCl4-induced acute liver damage. MFA has anti-oxidant, anti-inflammatory and anti-apoptotic activities and was able to modulate the NOX4/p22phox/ROS-JNK/p38 MAPK signaling pathway.

6.
Int J Mol Med ; 41(4): 2306-2316, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29336467

RESUMEN

The aim of the present study was to assess the molecular mechanism of ethanol­induced oxidative stress­mediated apoptosis in L­02 liver cells in order to elucidate novel pathways associated with alcoholic liver disease. L­02 cells were treated with 400 mM ethanol with or without inhibitors. The cell viability was measured by an MTT assay. Cell apoptosis was assessed by flow cytometry and a single­stranded DNA (ssDNA) assay. Intracellular reactive oxygen species (ROS) production of L­02 cells was determined using the 2',7'­dichlorofluorescein­diacetate dye. The protein expression of c­Jun N­terminal kinase (JNK), phosphorylated (p)­JNK, P38, p­P38, NADPH oxidase (NOX)1, NOX4, p22phox, B­cell lymphoma 2 (Bcl­2) and Bcl­2­associated X protein were measured by western blot analysis. The mRNA expression of NOX1, NOX4 and p22phox was measured by reverse transcription polymerase chain reaction analysis. The results indicated that after treatment with various concentrations of ethanol for the indicated durations, L­02 cells were displayed a significant decrease in cell viability in a dose­and time­dependent manner. Ethanol­induced apoptosis and cell death of L­02 cells was accompanied by the generation of ROS, elevated expression of NOX, as well as phosphorylation of JNK and P­38. In addition, increased expression of Bcl­2 was induced by 400 mM ethanol. Furthermore, treatment with NOX inhibitor attenuated the ethanol­induced a decrease in cell viability, and an increase in apoptosis and Bcl­2 expression. In conclusion, ethanol induced apoptosis in the L­02 hepatocyte cell line via generation of ROS and elevated expression of NOX4. This indicated that activation of JNK and p38 in the mitogen­activated protein kinase pathway promotes apoptosis in L­02 cells.


Asunto(s)
Apoptosis , Etanol/efectos adversos , Hepatocitos/efectos de los fármacos , Hepatopatías Alcohólicas/metabolismo , Sistema de Señalización de MAP Quinasas , NADPH Oxidasa 4/metabolismo , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular , Etanol/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hepatopatías Alcohólicas/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
7.
J Diabetes Res ; 2017: 5131785, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29098165

RESUMEN

Pancreatic ß-cells express several ion channels of the transient receptor potential family, which play important roles in mediating the stimulus-secretion coupling. One of these channels, the TRPM4 is a Ca2+-activated monovalent cation channel. This channel is inhibited by 9-phenanthrol, which also inhibits the TMEM16a Cl- channel, and activates the Ca2+-activated K+ channel, Kca3.1. The net effects of ion-channel modulation by 9-phenantherol on the insulin secretion remain unclear. We tested the effects of 9-phenanthrol on glucose- and GLP-1-induced insulin secretion from isolated rat islets in static incubations. When applied to the islets in the presence of 3.3 mM glucose, 9-phenanthrol caused a small increase in insulin secretion (~7% of the insulin secretion stimulated by 10 mM glucose). 10 µM 9-phenanthrol did not inhibit glucose- or GLP-1-induced insulin secretion. 20 µM and 30 µM 9-phenanthrol inhibited glucose-induced insulin secretion by ~80% and ~85%, respectively. Inhibition of the GLP-1-induced insulin secretion by 20 µM and 30 µM 9-phenanthrol was 65% and 94%, respectively. Our study shows that the major effect of 9-phenanthrol on the islets is a strong inhibition of insulin secretion, and we speculate that compounds related to 9-phenanthrol may be potentially useful in treating the pancreatogenous hyperinsulinemic hypoglycemia syndromes.


Asunto(s)
Péptido 1 Similar al Glucagón/farmacología , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Fenantrenos/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
8.
Physiol Rep ; 5(19)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29038359

RESUMEN

We previously showed that hyperoxia in vitro negatively affects beta cells of the rat. Here, we tested for possible clinical significance as well as mitochondrial interactions by hyperoxia, using human islets (function and viability), INS-1 832/13 cells (mitochondrial metabolism), and mouse neonates (effects in vivo). Lastly, we assessed relevant parameters in a cohort of individuals born preterm and then exposed to hyperoxia. Human islets and INS-1 832/13 cells were exposed to 24 h of hyperoxia (90-92% oxygen). Mouse neonates were subjected to 5 days of continuous hyperoxia. Individuals born preterm were evaluated in terms of glucose homeostasis and beta cell function by HbA1c and the HOMA2 formula. In human islets, hyperoxia significantly reduced glucose-stimulated insulin secretion by 42.2 ± 5.3% and viability assessed by MTT by 22.5 ± 5.4%. Hyperoxia down-regulated mitochondrial complex II by 21 ± 5% and upregulated complex III by 26 ± 10.1% and complex IV by 37 ± 10.6%. Partly similar effects on mitochondrial complexes were found in hyperoxia-exposed INS-1 832/13 cells. Exposure to hyperoxia swiftly reduced oxygen consumption in these cells and increased mitochondrial uncoupling. Hyperoxia transiently but significantly reduced insulin release in mouse neonates. Individuals born preterm displayed higher HbA1c versus controls, as well as insulin resistance. Thus, hyperoxia exerts negative effects in vitro on human beta cells and results indicate inhibitory effects on insulin secretion in vivo in mouse neonates. Negative effects may be lessened by the demonstrated swift and profound mitochondrial adaptability. Our findings open the possibility that hyperoxia could negatively affect beta cells of preterm human neonates.


Asunto(s)
Hiperoxia/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/sangre , Mitocondrias/metabolismo , Animales , Glucemia/metabolismo , Línea Celular , Células Cultivadas , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/metabolismo , Hemoglobinas/metabolismo , Humanos , Hiperoxia/sangre , Hiperoxia/patología , Recién Nacido , Recien Nacido Prematuro/sangre , Ratones , Consumo de Oxígeno
9.
Islets ; 8(6): 165-176, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27763807

RESUMEN

We tested whether exposure of ß cells at reduced glucose leads to mitochondrial adaptions and whether such adaptions modulate effects of hypoxia. Rat islets, human islets and INS-1 832/13 cells were pre-cultured short term at half standard glucose concentrations (5.5 mM for rat islets and cells, 2.75 mM for human islets) without overtly negative effects on subsequently measured function (insulin secretion and cellular insulin contents) or on viability. Culture at half standard glucose upregulated complex I and tended to upregulate complex II in islets and INS-1 cells alike. An increased release of lactate dehydrogenase that followed exposure to hypoxia was attenuated in rat islets which had been pre-cultured at half standard glucose. In INS-1 cells exposure to half standard glucose attenuated hypoxia-induced effects on several viability parameters (MTT, cell number and incremental apoptotic DNA). Thus culture at reduced glucose of pancreatic islets and clonal ß cells leads to mitochondrial adaptions which possibly lessen the negative impact of hypoxia on ß cell viability. These findings appear relevant in the search for optimization of pre-transplant conditions in a clinical setting.


Asunto(s)
Supervivencia Celular/fisiología , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Ratas
10.
Horm Mol Biol Clin Investig ; 26(1): 67-76, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26562042

RESUMEN

BACKGROUND: Diabetes type 1 is characterized by the failure of beta cells to produce insulin. Suppressor of cytokine signaling (SOCS) proteins are important regulators of the Janus kinase/signal transducer and activator of transcription (JAK-STAT) pathway. Previous studies have shown that GH can prevent the development of type I diabetes in mice and that SOCS2 deficiency mimics a state of increased GH sensitivity. METHODOLOGY: The elevated sensitivity of SOCS2-/- mice to GH and possibly to PRL was the rationale to analyze the effects of multiple low dose streptozotocin (MLDSTZ)-induced diabetes in SOCS2-/- mice. RESULTS: We show that 6-month-old SOCS2-/- mice, but not 2-month-old mice, were less sensitive to MLDSTZ-induced diabetes, compared to controls. MLDSTZ treatment induced glucose intolerance in both SOCS2+/+ and SOCS2-/- mice, as shown by glucose tolerance tests, with SOCS2+/+ mice showing a more marked intolerance, compared to SOCS2-/- mice. Furthermore, insulin tolerance tests showed that the SOCS2-/- mice have an improved hypoglycemic response to exogenous insulin, compared to SOCS2+/+ mice. Moreover, in isolated islets, lipotoxic effects on insulin release could partly be overcome by ligands, which bind to GH or PRL receptors. CONCLUSION: Knockdown of SOCS2 makes mice less sensitive to MLDSTZ. These results are consistent with the proposal that elimination of SOCS2 in pancreatic islets creates a state of ß-cell hypersensitivity to GH/PRL that mimics events in pregnancy, and which is protective against MLDSTZ-induced type I diabetes in mice. SOCS2-dependent control of ß-cell survival may be of relevance to islet regeneration and survival in transplantation.


Asunto(s)
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Eliminación de Gen , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Glucosa/metabolismo , Hormona del Crecimiento/farmacología , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Prolactina/farmacología , Estreptozocina/toxicidad , Proteínas Supresoras de la Señalización de Citocinas/genética
11.
PLoS One ; 10(9): e0138558, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26401848

RESUMEN

OBJECTIVE: To provide novel insights on mitochondrial respiration in ß-cells and the adaptive effects of hypoxia. METHODS AND DESIGN: Insulin-producing INS-1 832/13 cells were exposed to 18 hours of hypoxia followed by 20-22 hours re-oxygenation. Mitochondrial respiration was measured by high-resolution respirometry in both intact and permeabilized cells, in the latter after establishing three functional substrate-uncoupler-inhibitor titration (SUIT) protocols. Concomitant measurements included proteins of mitochondrial complexes (Western blotting), ATP and insulin secretion. RESULTS: Intact cells exhibited a high degree of intrinsic uncoupling, comprising about 50% of oxygen consumption in the basal respiratory state. Hypoxia followed by re-oxygenation increased maximal overall respiration. Exploratory experiments in peremabilized cells could not show induction of respiration by malate or pyruvate as reducing substrates, thus glutamate and succinate were used as mitochondrial substrates in SUIT protocols. Permeabilized cells displayed a high capacity for oxidative phosphorylation for both complex I- and II-linked substrates in relation to maximum capacity of electron transfer. Previous hypoxia decreased phosphorylation control of complex I-linked respiration, but not in complex II-linked respiration. Coupling control ratios showed increased coupling efficiency for both complex I- and II-linked substrates in hypoxia-exposed cells. Respiratory rates overall were increased. Also previous hypoxia increased proteins of mitochondrial complexes I and II (Western blotting) in INS-1 cells as well as in rat and human islets. Mitochondrial effects were accompanied by unchanged levels of ATP, increased basal and preserved glucose-induced insulin secretion. CONCLUSIONS: Exposure of INS-1 832/13 cells to hypoxia, followed by a re-oxygenation period increases substrate-stimulated respiratory capacity and coupling efficiency. Such effects are accompanied by up-regulation of mitochondrial complexes also in pancreatic islets, highlighting adaptive capacities of possible importance in an islet transplantation setting. Results also indicate idiosyncrasies of ß-cells that do not respire in response to a standard inclusion of malate in SUIT protocols.


Asunto(s)
Respiración de la Célula , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Adaptación Biológica , Adenosina Trifosfato/metabolismo , Animales , Muerte Celular , Línea Celular , Proliferación Celular , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Humanos , Hipoxia/metabolismo , Insulina/metabolismo , Malatos/metabolismo , Consumo de Oxígeno , Permeabilidad , Ratas , Especificidad por Sustrato
12.
Diabetes ; 64(10): 3425-38, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25918231

RESUMEN

Toll-like receptors (TLRs) have been implicated in the pathogenesis of type 2 diabetes. We examined the function of TLR3 in glucose metabolism and type 2 diabetes-related phenotypes in animals and humans. TLR3 is highly expressed in the pancreas, suggesting that it can influence metabolism. Using a diet-induced obesity model, we show that TLR3-deficient mice had enhanced glycemic control, facilitated by elevated insulin secretion. Despite having high insulin levels, Tlr3(-/-) mice did not experience disturbances in whole-body insulin sensitivity, suggesting that they have a robust metabolic system that manages increased insulin secretion. Increase in insulin secretion was associated with upregulation of islet glucose phosphorylation as well as exocytotic protein VAMP-2 in Tlr3(-/-) islets. TLR3 deficiency also modified the plasma lipid profile, decreasing VLDL levels due to decreased triglyceride biosynthesis. Moreover, a meta-analysis of two healthy human populations showed that a missense single nucleotide polymorphism in TLR3 (encoding L412F) was linked to elevated insulin levels, consistent with our experimental findings. In conclusion, our results increase the understanding of the function of innate receptors in metabolic disorders and implicate TLR3 as a key control system in metabolic regulation.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Receptor Toll-Like 3/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Área Bajo la Curva , Glucemia/metabolismo , VLDL-Colesterol/sangre , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Glucosa/farmacocinética , Prueba de Tolerancia a la Glucosa , Homeostasis , Humanos , Insulina/sangre , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Polimorfismo Genético , Receptor Toll-Like 3/genética
13.
Pancreas ; 44(5): 773-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25822155

RESUMEN

OBJECTIVES: The use of primary human ß-cells for studying Ca signaling is limited by the scarcity of human pancreatic islets. Rodent insulinoma cell lines are widely used, but it is difficult to extrapolate results obtained from rodent cells to human. Recently, a genetically engineered human ß-cell line EndoC-BH1 has been developed. We have examined whether the EndoC-BH1 cells could be used as a model for studying Ca signaling in the ß-cells. METHODS: We used microscope-based fluorometry to measure cytoplasmic-free Ca concentration from fura-2-loaded single EndoC-BH1 cells cultured on glass cover slips. Ca responses to different agonists of insulin secretion were studied. Insulin secretion was measured by radioimmunoassay. RESULTS: EndoC-BH1 cells secreted insulin in response to glucose in a dose-dependent manner, and the secretion was enhanced by GLP-1 (glucagon-like peptide 1). Glucose, potassium chloride, carbachol, L-arginine, and tolbutamide increased cytoplasmic-free Ca concentration in the EndoC-BH1 cells. We found that GLP-1 was essential for Ca response to glucose and tolbutamide. CONCLUSIONS: We concluded that the EndoC-BH1 cells can be used as model cells to study Ca signaling and stimulus-secretion coupling in the human ß-cells.


Asunto(s)
Señalización del Calcio , Ingeniería Genética , Células Secretoras de Insulina/metabolismo , Señalización del Calcio/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes , Fluorometría , Fura-2 , Glucosa/farmacología , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Cinética , Potenciales de la Membrana
14.
Pancreas ; 43(4): 597-604, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24632551

RESUMEN

OBJECTIVE: Several studies have reported that the transient receptor potential melastatin-like subtype 5 (TRPM5) channel, a Ca(2+)-activated monovalent cation channel, is involved in the stimulus-secretion coupling in the mouse pancreatic ß-cells. We have studied the role of the TRPM5 channel in regulating insulin secretion and cytoplasmic free Ca(2+) concentration ([Ca(2+)](i)) in the rat ß-cells by using triphenylphosphine oxide, a selective inhibitor of the channel. METHODS: Insulin secretion from islets from Sprague-Dawley rats was measured in batch incubations. Cytoplasmic free Ca(2+) concentration was measured from single ß-cells by fura-2-based microfluorometry. RESULTS: Triphenylphosphine oxide did not alter insulin secretion and [Ca(2+)](i) response triggered by KCl or fructose. It inhibited insulin secretion in response to glucose, L-arginine, and glucagon-like peptide 1. It also inhibited glucose-induced insulin secretion by mechanisms that are independent of the adenosine triphosphate-sensitive potassium channels and [Ca(2+)](i) increase. CONCLUSIONS: Our results suggest that in the rat islets, TRPM5 is involved in mediating insulin secretion by glucose and l-arginine and in potentiating the glucose-induced insulin secretion by glucagon-like peptide 1.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canales Catiónicos TRPM/metabolismo , Animales , Arginina/metabolismo , Calcio/metabolismo , Células Cultivadas , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Potenciales de la Membrana , Compuestos Organofosforados/farmacología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPM/antagonistas & inhibidores , Factores de Tiempo
15.
Biochem Biophys Res Commun ; 443(1): 223-8, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24299957

RESUMEN

Isolated pancreatic islets containing the insulin-producing beta cells are devoid of circulation. They may therefore experience hypoxia with possible negative effects on beta cell function and survival. We investigated (1) whether hyperoxia in vitro would be beneficial by counteracting putative effects of lost circulation and, further, (2) whether previous hyperoxia would attenuate the impact of subsequently induced severe hypoxia. Islets from Sprague-Dawley rats were exposed to 95% O2 for 18 h. This hyperoxic exposure diminished glucose-induced insulin secretion by 47% and inhibited oxygen consumption by 39-41%. Mitochondrial complexes I-III were decreased by 29-37%. Negative effects on insulin secretion and complexes III and IV waned after a 22 h period of normoxia following hyperoxia whereas complexes I and II were still diminished, ROS production was increased and rates of apoptosis tended to be increased (P=0.07). The effects of previous hyperoxia on susceptibility to damage by subsequent hypoxia were tested after 5.5h of 0.8% O2. Previous hyperoxia did not affect hypoxia-induced enhancement of HIF-1 alpha but modestly and significantly attenuated hypoxia-induced decreases in insulin contents. We conclude that hyperoxia exerts largely negative effects on beta cells, effects which are functional and possibly also toxic. A paradoxical positive finding (attenuation of hypoxia-induced effects) could be secondary to a protective effect of the hyperoxia-induced reduction of oxidative metabolism.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mitocondrias/metabolismo , Oxígeno/metabolismo , Animales , Apoptosis , Separación Celular , ADN/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Glucosa/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Fosforilación Oxidativa , Oxígeno/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
16.
PLoS One ; 8(7): e67498, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935835

RESUMEN

OBJECTIVE: Beta cells of pancreatic islets are susceptible to functional deficits and damage by hypoxia. Here we aimed to characterize such effects and to test for and pharmacological means to alleviate a negative impact of hypoxia. METHODS AND DESIGN: Rat and human pancreatic islets were subjected to 5.5 h of hypoxia after which functional and viability parameters were measured subsequent to the hypoxic period and/or following a 22 h re-oxygenation period. Preconditioning with diazoxide or other agents was usually done during a 22 h period prior to hypoxia. RESULTS: Insulin contents decreased by 23% after 5.5 h of hypoxia and by 61% after a re-oxygenation period. Preconditioning with diazoxide time-dependently alleviated these hypoxia effects in rat and human islets. Hypoxia reduced proinsulin biosynthesis ((3)H-leucine incorporation into proinsulin) by 35%. Preconditioning counteracted this decrease by 91%. Preconditioning reduced hypoxia-induced necrosis by 40%, attenuated lowering of proteins of mitochondrial complexes I-IV and enhanced stimulation of HIF-1-alpha and phosphorylated AMPK proteins. Preconditioning by diazoxide was abolished by co-exposure to tolbutamide or elevated potassium (i.e. conditions which increase Ca(2+) inflow). Preconditioning with nifedipine, a calcium channel blocker, partly reproduced effects of diazoxide. Both diazoxide and nifedipine moderately reduced basal glucose oxidation whereas glucose-induced oxygen consumption (tested with diazoxide) was unaffected. Preconditioning with diaxoxide enhanced insulin contents in transplants of rat islets to non-diabetic rats and lowered hyperglycemia vs. non-preconditioned islets in streptozotocin-diabetic rats. Preconditioning of human islet transplants lowered hyperglycemia in streptozotocin-diabetic nude mice. CONCLUSIONS: 1) Prior blocking of Ca(2+) inflow associates with lesser hypoxia-induced damage, 2) preconditioning affects basal mitochondrial metabolism and accelerates activation of hypoxia-reactive and potentially protective factors, 3) results indicate that preconditioning by K(+)-ATP-channel openers has therapeutic potential for islet transplantations.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Calcio/metabolismo , Diabetes Mellitus Experimental/terapia , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Nifedipino/farmacología , Oxígeno/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diazóxido/farmacología , Proteínas del Complejo de Cadena de Transporte de Electrón/agonistas , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Humanos , Hipoglucemiantes/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/agonistas , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Masculino , Ratones , Fosforilación , Potasio/metabolismo , Proinsulina/metabolismo , Ratas , Estreptozocina , Tolbutamida/farmacología
17.
Islets ; 4(3): 233-42, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22854606

RESUMEN

We investigated the impact of a diabetic state with hyperglycemia on morphometry of ß cell mitochondria and modifying influence of a K (+) -ATP channel opener and we related in vivo findings with glucose effects in vitro. For in vivo experiments islets from syngeneic rats were transplanted under the kidney capsule to neonatally streptozotocin-diabetic or non-diabetic recipients. Diabetic recipients received vehicle, or tifenazoxide (NN414), intragastrically for 9 weeks. Non-diabetic rats received vehicle. Transplants were excised 7 d after cessation of treatment (wash-out) and prepared for electron microscopy. Morphological parameters were measured from approx. 25,000 mitochondria. Rat islets were cultured in vitro for 2-3 weeks at 27 or 11 (control) mmol/l glucose. Transplants to diabetic rats displayed decreased numbers of mitochondria (-31%, p < 0.05), increased mitochondrial volume and increased mitochondrial outer surface area, p < 0.001. Diabetes increased variability in mitochondrial size with frequent appearance of mega-mitochondria. Tifenazoxide partly normalized diabetes-induced effects, and mega-mitochondria disappeared. Long-term culture of islets at 27 mmol/l glucose reproduced the in vivo morphological abnormalities. High-glucose culture was also associated with reduced ATP and ADP contents, reduced oxygen consumption, reduced signaling by MitoTracker Red and reduction of mitochondrial proteins (complexes I-IV), OPA 1 and glucose-induced insulin release. We conclude that (1) a long-term diabetic state leads to a reduced number of mitochondria and to distinct morphological abnormalities which are replicated by high glucose in vitro; (2) the morphological abnormalities are coupled to dysfunction; (3) K (+) -ATP channel openers may have potential to partly reverse glucose-induced effects.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/patología , Hiperglucemia/patología , Islotes Pancreáticos/patología , Mitocondrias/patología , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Óxidos S-Cíclicos/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Hiperglucemia/metabolismo , Hipoglucemiantes/farmacología , Islotes Pancreáticos/metabolismo , Masculino , Microscopía Electrónica , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Ratas , Ratas Endogámicas WF , Ratas Sprague-Dawley
18.
Biochem Biophys Res Commun ; 423(2): 259-64, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22634308

RESUMEN

Evidence is conflicting as to the impact of elevated levels of uncoupling protein-2 (UCP-2) on insulin-producing beta cells. Here we investigated effects of a fourfold induction of UCP-2 protein primarily on mitochondrial parameters and tested for replication of positive findings at a lower level of induction. We transfected INS-1 cells to obtain a tet-on inducible cell line. A 48 h exposure to 1 µg/ml of doxycycline (dox) induced UCP-2 fourfold (424 ± 113%, mean±SEM) and 0.1 µg/ml twofold (178 ± 29%, n=3). Fourfold induced cells displayed normal viability (MTT, apoptosis), normal cellular insulin contents and, glucose-induced insulin secretion (+27 ± 11%) as well as D-[U-(14)C]-glucose oxidation (+5 ± 9% at 11 mM glucose). Oxidation of [1-(14)C]-oleate was increased from 4088 to 5797 fmol/µg prot/2h at 3.3mM glucose, p<0.03. Oxidation of L-[(14)C(U)]-glutamine was unaffected. Induction of UCP-2 did not significantly affect measures of mitochondrial membrane potential (Rhodamine 123) or mitochondrial mass (Mitotracker Green) and did not affect ATP levels. Oligomycin-inhibited oxygen consumption (a measure of mitochondrial uncoupling) was marginally increased, the effect being significant in comparison with dox-only treated cells, p<0.05. Oxygen radicals, assessed by dichlorofluorescin diacetate, were decreased by 30%, p<0.025. Testing for the lower level of UCP-2 induction did not reproduce any of the positive findings. A fourfold induction of UCP-2 was required to exert minor metabolic effects. These findings question an impact of moderately elevated UCP-2 levels in beta cells as seen in diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Canales Iónicos/biosíntesis , Mitocondrias/metabolismo , Proteínas Mitocondriales/biosíntesis , Animales , Línea Celular Tumoral , Doxiciclina/farmacología , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Canales Iónicos/genética , Potencial de la Membrana Mitocondrial , Proteínas Mitocondriales/genética , Oligomicinas/farmacología , Oxidación-Reducción , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Ratas , Transfección , Proteína Desacopladora 2
19.
PLoS One ; 6(4): e18213, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21483673

RESUMEN

BACKGROUND: Voltage-dependent K(+) channels (Kv) mediate repolarisation of ß-cell action potentials, and thereby abrogate insulin secretion. The role of the Kv1.1 K(+) channel in this process is however unclear. We tested for presence of Kv1.1 in different species and tested for a functional role of Kv1.1 by assessing pancreatic islet function in BALB/cByJ (wild-type) and megencephaly (mceph/mceph) mice, the latter having a deletion in the Kv1.1 gene. METHODOLOGY/PRINCIPAL FINDINGS: Kv1.1 expression was detected in islets from wild-type mice, SD rats and humans, and expression of truncated Kv1.1 was detected in mceph/mceph islets. Full-length Kv1.1 protein was present in islets from wild-type mice, but, as expected, not in those from mceph/mceph mice. Kv1.1 expression was localized to the ß-cell population and also to α- and δ-cells, with evidence of over-expression of truncated Kv1.1 in mceph/mceph islets. Blood glucose, insulin content, and islet morphology were normal in mceph/mceph mice, but glucose-induced insulin release from batch-incubated islets was (moderately) higher than that from wild-type islets. Reciprocal blocking of Kv1.1 by dendrotoxin-K increased insulin secretion from wild-type but not mceph/mceph islets. Glucose-induced action potential duration, as well as firing frequency, was increased in mceph/mceph mouse ß-cells. This duration effect on action potential in ß-cells from mceph/mceph mice was mimicked by dendrotoxin-K in ß-cells from wild-type mice. Observations concerning the effects of both the mceph mutation, and of dendrotoxin-K, on glucose-induced insulin release were confirmed in pancreatic islets from Kv1.1 null mice. CONCLUSION/SIGNIFICANCE: Kv1.1 channels are expressed in the ß-cells of several species, and these channels can influence glucose-stimulated insulin release.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Canal de Potasio Kv.1.1/metabolismo , Potenciales de Acción , Animales , Glucemia/metabolismo , Peso Corporal , Recolección de Datos , Femenino , Fura-2/metabolismo , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Canal de Potasio Kv.1.1/deficiencia , Canal de Potasio Kv.1.1/genética , Masculino , Ratones , Imagen Molecular , Perfusión , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Eliminación de Secuencia , Canales de Potasio Shab/metabolismo , Especificidad de la Especie
20.
Biochim Biophys Acta ; 1801(2): 191-7, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19931418

RESUMEN

Facilitation of fatty acid uptake in beta cells could potentially affect beta cell metabolism and secretory function; however such effects have not been clearly documented. CD36 facilitates uptake of fatty acids (FA) in muscle and adipose tissue and is likely to exert a similar effect in beta cells. We investigated the impact of over-expressing CD36 on fatty acid uptake and beta cell function by a Tet-on system in INS-1 cells. Doxycycline dose-dependently increased the CD36 protein with localization mainly in the cell membrane. Over-expression increased both specific uptake and efflux of oleate whereas intracellular glycerides were only marginally increased and incorporation of 14C-oleate or -palmitate into di- or triglycerides not affected. The normal potentiation of glucose-induced insulin secretion by acute addition of FA (50-100 micromol/l oleate and palmitate) was lost and the normal inhibitory effect of high glucose both on oleate oxidation and on the activity of carnitine palmitoyltransferase I was reduced. Over-expression did not induce apoptosis. We conclude that induction of the CD36 transporter increases uptake of FA, the consequences of which are blunting of the functional interplay between glucose and FA on insulin secretion and oxidative metabolism.


Asunto(s)
Antígenos CD36/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Antibacterianos/farmacología , Apoptosis/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Doxiciclina/farmacología , Técnica del Anticuerpo Fluorescente , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Desacopladora 2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...