Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 334, 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38184650

RESUMEN

Pancreatic ß-cells respond to metabolic stress by upregulating insulin secretion, however the underlying mechanisms remain unclear. Here we show, in ß-cells from overweight humans without diabetes and mice fed a high-fat diet for 2 days, insulin exocytosis and secretion are enhanced without increased Ca2+ influx. RNA-seq of sorted ß-cells suggests altered metabolic pathways early following high fat diet, where we find increased basal oxygen consumption and proton leak, but a more reduced cytosolic redox state. Increased ß-cell exocytosis after 2-day high fat diet is dependent on this reduced intracellular redox state and requires the sentrin-specific SUMO-protease-1. Mice with either pancreas- or ß-cell-specific deletion of this fail to up-regulate exocytosis and become rapidly glucose intolerant after 2-day high fat diet. Mechanistically, redox-sensing by the SUMO-protease requires a thiol group at C535 which together with Zn+-binding suppresses basal protease activity and unrestrained ß-cell exocytosis, and increases enzyme sensitivity to regulation by redox signals.


Asunto(s)
Dieta Alta en Grasa , Exocitosis , Animales , Humanos , Ratones , Cisteína Endopeptidasas/genética , Citosol , Dieta Alta en Grasa/efectos adversos , Glucosa , Péptido Hidrolasas
2.
Diabetes ; 71(8): 1706-1720, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35622000

RESUMEN

Hypoxia-induced islet cell death, caused by an insufficient revascularization of the grafts, is a major obstacle for successful pancreatic islet transplantation. Recently, it has been reported that the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome is expressed in pancreatic islets and that its loss protects against hypoxia-induced cell death. Therefore, we hypothesized that the inhibition of NLRP3 in islets improves the survival and endocrine function of the grafts. The transplantation of Nlrp3-/- islets or wild-type (WT) islets exposed to the NLRP3 inhibitor CY-09 into mouse dorsal skinfold chambers resulted in an improved revascularization compared with controls. An increased insulin release after NLRP3 inhibition caused the enhanced angiogenic response. Moreover, the inhibition of NLRP3 in hypoxic ß-cells triggered insulin gene expression by inducing the shuttling of MafA and pancreatic and duodenal homeobox-1 into the nucleus. This was mediated by a reduced interaction of NLRP3 with the thioredoxin-interacting protein (TXNIP). Transplantation of Nlrp3-/- islets or WT islets exposed to CY-09 under the kidney capsule of diabetic mice markedly improved the restoration of normoglycemia. These findings indicate that the inhibition of NLRP3 in isolated islets represents a promising therapeutic strategy to improve engraftment and function of the islets.


Asunto(s)
Diabetes Mellitus Experimental , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Animales , Diabetes Mellitus Experimental/metabolismo , Hipoxia/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
3.
Antioxidants (Basel) ; 10(8)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34439552

RESUMEN

In type 1 diabetes (T1D) development, proinflammatory cytokines (PIC) released by immune cells lead to increased reactive oxygen species (ROS) production in ß-cells. Nonetheless, the temporality of the events triggered and the role of different ROS sources remain unclear. Isolated islets from C57BL/6J wild-type (WT), NOX1 KO and NOX2 KO mice were exposed to a PIC combination. We show that cytokines increase O2•- production after 2 h in WT and NOX1 KO but not in NOX2 KO islets. Using transgenic mice constitutively expressing a genetically encoded compartment specific H2O2 sensor, we show, for the first time, a transient increase of cytosolic/nuclear H2O2 in islet cells between 4 and 5 h during cytokine exposure. The H2O2 increase coincides with the intracellular NAD(P)H decrease and is absent in NOX2 KO islets. NOX2 KO confers better glucose tolerance and protects against cytokine-induced islet secretory dysfunction and death. However, NOX2 absence does not counteract the cytokine effects in ER Ca2+ depletion, Store-Operated Calcium Entry (SOCE) increase and ER stress. Instead, the activation of ER stress precedes H2O2 production. As early NOX2-driven ROS production impacts ß-cells' function and survival during insulitis, NOX2 might be a potential target for designing therapies against early ß-cell dysfunction in the context of T1D onset.

4.
Acta Diabetol ; 58(12): 1637-1647, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34254190

RESUMEN

AIMS: The exposure of isolated pancreatic islets to pro-angiogenic factors prior to their transplantation represents a promising strategy to accelerate the revascularization of the grafts. It has been shown that erythropoietin (EPO), a glycoprotein regulating erythropoiesis, also induces angiogenesis. Therefore, we hypothesized that EPO exposure of isolated islets improves their posttransplant revascularization. METHODS: Flow cytometric, immunohistochemical and quantitative real-time (qRT)-PCR analyses were performed to study the effect of EPO on the viability, cellular composition and gene expression of isolated islets. Moreover, islets expressing a mitochondrial or cytosolic H2O2 sensor were used to determine reactive oxygen species (ROS) levels. The dorsal skinfold chamber model in combination with intravital fluorescence microscopy was used to analyze the revascularization of transplanted islets. RESULTS: We found that the exposure of isolated islets to EPO (3 units/mL) for 24 h does not affect the viability and the production of ROS when compared to vehicle-treated and freshly isolated islets. However, the exposure of islets to EPO increased the number of CD31-positive cells and enhanced the gene expression of insulin and vascular endothelial growth factor (VEGF)-A. The revascularization of the EPO-cultivated islets was accelerated within the initial phase after transplantation when compared to both controls. CONCLUSION: These findings indicate that the exposure of isolated islets to EPO may be a promising approach to improve clinical islet transplantation.


Asunto(s)
Eritropoyetina , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Eritropoyetina/farmacología , Peróxido de Hidrógeno , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/genética
5.
EMBO Mol Med ; 13(1): e12616, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33135383

RESUMEN

Pancreatic islet transplantation still represents a promising therapeutic strategy for curative treatment of type 1 diabetes mellitus. However, a limited number of organ donors and insufficient vascularization with islet engraftment failure restrict the successful transfer of this approach into clinical practice. To overcome these problems, we herein introduce a novel strategy for the generation of prevascularized islet organoids by the fusion of pancreatic islet cells with functional native microvessels. These insulin-secreting organoids exhibit a significantly higher angiogenic activity compared to freshly isolated islets, cultured islets, and non-prevascularized islet organoids. This is caused by paracrine signaling between the ß-cells and the microvessels, mediated by insulin binding to its corresponding receptor on endothelial cells. In vivo, the prevascularized islet organoids are rapidly blood-perfused after transplantation by the interconnection of their autochthonous microvasculature with surrounding blood vessels. As a consequence, a lower number of islet grafts are required to restore normoglycemia in diabetic mice. Thus, prevascularized islet organoids may be used to improve the success rates of clinical islet transplantation.


Asunto(s)
Diabetes Mellitus Experimental , Células Secretoras de Insulina , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Animales , Células Endoteliales , Insulina , Ratones
6.
EMBO Mol Med ; 12(4): e09271, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32187826

RESUMEN

The role of the endothelium is not just limited to acting as an inert barrier for facilitating blood transport. Endothelial cells (ECs), through expression of a repertoire of angiocrine molecules, regulate metabolic demands in an organ-specific manner. Insulin flux across the endothelium to muscle cells is a rate-limiting process influencing insulin-mediated lowering of blood glucose. Here, we demonstrate that Notch signaling in ECs regulates insulin transport to muscle. Notch signaling activity was higher in ECs isolated from obese mice compared to non-obese. Sustained Notch signaling in ECs lowered insulin sensitivity and increased blood glucose levels. On the contrary, EC-specific inhibition of Notch signaling increased insulin sensitivity and improved glucose tolerance and glucose uptake in muscle in a high-fat diet-induced insulin resistance model. This was associated with increased transcription of Cav1, Cav2, and Cavin1, higher number of caveolae in ECs, and insulin uptake rates, as well as increased microvessel density. These data imply that Notch signaling in the endothelium actively controls insulin sensitivity and glucose homeostasis and may therefore represent a therapeutic target for diabetes.


Asunto(s)
Células Endoteliales/metabolismo , Resistencia a la Insulina , Insulina , Músculo Esquelético/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Glucosa/metabolismo , Insulina/metabolismo , Ratones
7.
J Mol Biol ; 432(5): 1461-1493, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-31634466

RESUMEN

Insulin-secreting pancreatic ß-cells play a critical role in blood glucose homeostasis and the development of type 2 diabetes (T2D) in the context of insulin resistance. Based on data obtained at the whole cell level using poorly specific chemical probes, reactive oxygen species (ROS) such as superoxide and hydrogen peroxide have been proposed to contribute to the stimulation of insulin secretion by nutrients (positive role) and to the alterations of cell survival and secretory function in T2D (negative role). This raised the controversial hypothesis that any attempt to decrease ß-cell oxidative stress and apoptosis in T2D would further impair insulin secretion. Over the last decade, the development of genetically-encoded redox probes that can be targeted to cellular compartments of interest and are specific of redox couples allowed the evaluation of short- and long-term effects of nutrients on ß-cell redox changes at the subcellular level. The data indicated that the nutrient regulation of ß-cell redox signaling and ROS toxicity is far more complex than previously thought and that the subcellular compartmentation of these processes cannot be neglected when evaluating the mechanisms of ROS production or the efficacy of antioxidant enzymes and antioxidant drugs under glucolipotoxic conditions and in T2D. In this review, we present what is currently known about the compartmentation of redox homeostatic systems and tools to investigate it. We then review data about the effects of nutrients on ß-cell subcellular redox state under normal conditions and in the context of T2D and discuss challenges and opportunities in the field.


Asunto(s)
Compartimento Celular , Células Secretoras de Insulina , Islotes Pancreáticos , Estrés Oxidativo , Animales , Antioxidantes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Retículo Endoplásmico/metabolismo , Colorantes Fluorescentes , Glucosa/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Mitocondrias/metabolismo , Nutrientes/metabolismo , Nutrientes/toxicidad , Oxidación-Reducción , Peroxisomas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Superóxido Dismutasa/metabolismo
8.
Br J Pharmacol ; 177(7): 1651-1665, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31721150

RESUMEN

BACKGROUND AND PURPOSE: Pancreatic islet transplantation is a promising therapeutic approach for Type 1 diabetes. A major prerequisite for the survival of grafted islets is a rapid revascularization after transplantation. Erythropoietin (EPO), the primary regulator of erythropoiesis, has been shown to promote angiogenesis. Therefore, we investigated in this study whether EPO improves the revascularization of transplanted islets. EXPERIMENTAL APPROACH: Islets from FVB/N mice were transplanted into dorsal skinfold chambers of recipient animals, which were daily treated with an intraperitoneal injection of EPO (500 IU·kg-1 ) or vehicle (control) throughout an observation period of 14 days. In a second set of experiments, animals were only pretreated with EPO over a 6-day period prior to islet transplantation. The revascularization of the grafts was assessed by repetitive intravital fluorescence microscopy and immunohistochemistry. In addition, a streptozotocin-induced diabetic mouse model was used to study the effect of EPO-pretreatment on the endocrine function of the grafts. KEY RESULTS: EPO treatment slightly accelerated the revascularization of the islet grafts. This effect was markedly more pronounced in EPO-pretreated animals, resulting in significantly higher numbers of engrafted islets and an improved perfusion of endocrine tissue without affecting systemic haematocrit levels when compared with controls. Moreover, EPO-pretreatment significantly accelerated the recovery of normoglycaemia in diabetic mice after islet transplantation. CONCLUSION AND IMPLICATIONS: These findings demonstrate that, particularly, short-term EPO-pretreatment represents a promising therapeutic approach to improve the outcome of islet transplantation, without an increased risk of thromboembolic events.


Asunto(s)
Diabetes Mellitus Experimental , Eritropoyetina , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Eritropoyetina/farmacología , Ratones , Neovascularización Fisiológica , Estreptozocina
10.
Cell ; 169(5): 807-823.e19, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28479188

RESUMEN

Dormant hematopoietic stem cells (dHSCs) are atop the hematopoietic hierarchy. The molecular identity of dHSCs and the mechanisms regulating their maintenance or exit from dormancy remain uncertain. Here, we use single-cell RNA sequencing (RNA-seq) analysis to show that the transition from dormancy toward cell-cycle entry is a continuous developmental path associated with upregulation of biosynthetic processes rather than a stepwise progression. In addition, low Myc levels and high expression of a retinoic acid program are characteristic for dHSCs. To follow the behavior of dHSCs in situ, a Gprc5c-controlled reporter mouse was established. Treatment with all-trans retinoic acid antagonizes stress-induced activation of dHSCs by restricting protein translation and levels of reactive oxygen species (ROS) and Myc. Mice maintained on a vitamin A-free diet lose HSCs and show a disrupted re-entry into dormancy after exposure to inflammatory stress stimuli. Our results highlight the impact of dietary vitamin A on the regulation of cell-cycle-mediated stem cell plasticity. VIDEO ABSTRACT.


Asunto(s)
Células Madre Hematopoyéticas/citología , Transducción de Señal , Tretinoina/farmacología , Vitamina A/administración & dosificación , Animales , Vías Biosintéticas , Técnicas de Cultivo de Célula , Ciclo Celular/efectos de los fármacos , Supervivencia Celular , Dieta , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Poli I-C/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Análisis de la Célula Individual , Estrés Fisiológico , Vitamina A/farmacología , Vitaminas/administración & dosificación , Vitaminas/farmacología
11.
Mol Cell Endocrinol ; 439: 354-362, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27664519

RESUMEN

High glucose-induced oxidative stress and increased NADPH oxidase-2 (NOX2) activity may contribute to the progressive decline of the functional ß-cell mass in type 2 diabetes. To test that hypothesis, we characterized, in islets from male NOX2 knockout (NOX2-KO) and wild-type (WT) C57BL/6J mice cultured for up to 3 weeks at 10 or 30 mmol/l glucose (G10 or G30), the in vitro effects of glucose on cytosolic oxidative stress using probes sensing glutathione oxidation (GRX1-roGFP2), thiol oxidation (roGFP1) or H2O2 (roGFP2-Orp1), on ß-cell stimulus-secretion coupling events and on ß-cell apoptosis. After 1-2 days of culture in G10, the glucose stimulation of insulin secretion (GSIS) was ∼1.7-fold higher in NOX2-KO vs. WT islets at 20-30 mmol/l glucose despite similar rises in NAD(P)H and intracellular calcium concentration ([Ca2+]i) and no differences in cytosolic GRX1-roGFP2 oxidation. After long-term culture at G10, roGFP1 and roGFP2-Orp1 oxidation and ß-cell apoptosis remained low, and the glucose-induced rises in NAD(P)H, [Ca2+]i and GSIS were similarly preserved in both islet types. After prolonged culture at G30, roGFP1 and roGFP2-Orp1 oxidation increased in parallel with ß-cell apoptosis, the glucose sensitivity of the NADPH, [Ca2+]i and insulin secretion responses increased, the maximal [Ca2+]i response decreased, but maximal GSIS was preserved. These responses were almost identical in both islet types. In conclusion, NOX2 is a negative regulator of maximal GSIS in C57BL/6J mouse islets, but it does not detectably contribute to the in vitro glucotoxic induction of cytosolic oxidative stress and alterations of ß-cell survival and function.


Asunto(s)
Glucosa/toxicidad , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , NADPH Oxidasa 2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citosol/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 2/deficiencia , Oxidación-Reducción , ARN Mensajero/genética , ARN Mensajero/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Técnicas de Cultivo de Tejidos
12.
Sci Signal ; 9(419): rs1, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26980443

RESUMEN

Mapping the in vivo distribution of endogenous oxidants in animal tissues is of substantial biomedical interest. Numerous health-related factors, including diet, physical activity, infection, aging, toxins, or pharmacological intervention, may cause redox changes. Tools are needed to pinpoint redox state changes to particular organs, tissues, cell types, and subcellular organelles. We describe a procedure that preserves the in vivo redox state of genetically encoded redox biosensors within histological tissue sections, thus providing "redox maps" for any tissue and comparison of interest. We demonstrate the utility of the technique by visualizing endogenous redox differences and changes in the context of tumor growth, inflammation, embryonic development, and nutrient starvation.


Asunto(s)
Imagen Molecular/métodos , Sondas Moleculares/metabolismo , Transgenes , Animales , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Sondas Moleculares/genética , Oxidación-Reducción
13.
Am J Physiol Endocrinol Metab ; 309(7): E632-9, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26264555

RESUMEN

In rat pancreatic islets, ß-cell gene expression, survival, and subsequent acute glucose stimulation of insulin secretion (GSIS) are optimally preserved by prolonged culture at 10 mM glucose (G10) and markedly altered by culture at G5 or G30. Here, we tested whether pharmacological glucokinase (GK) activation prevents these alterations during culture or improves GSIS after culture. Rat pancreatic islets were cultured 1-7 days at G5, G10, or G30 with or without 3 µM of the GK activator Ro 28-0450 (Ro). After culture, ß-cell apoptosis and islet gene mRNA levels were measured, and the acute glucose-induced increase in NAD(P)H autofluorescence, intracellular calcium concentration, and insulin secretion were tested in the absence or presence of Ro. Prolonged culture of rat islets at G5 or G30 instead of G10 triggered ß-cell apoptosis and reduced their glucose responsiveness. Addition of Ro during culture differently affected ß-cell survival and glucose responsiveness depending on the glucose concentration during culture: it was beneficial to ß-cell survival and function at G5, detrimental at G10, and ineffective at G30. In contrast, acute GK activation with Ro increased the glucose sensitivity of islets cultured at G10 but failed at restoring ß-cell glucose responsiveness after culture at G5 or G30. We conclude that pharmacological GK activation prevents the alteration of ß-cell survival and function by long-term culture at G5 but mimics glucotoxicity when added to G10. The complex effects of glucose on the ß-cell phenotype result from changes in glucose metabolism and not from an effect of glucose per se.


Asunto(s)
Glucoquinasa/metabolismo , Glucosa/farmacología , Islotes Pancreáticos/efectos de los fármacos , Sulfonas/farmacología , Tiazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Islotes Pancreáticos/metabolismo , Masculino , Ratas , Ratas Wistar
14.
Pancreas ; 44(2): 287-95, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25426612

RESUMEN

OBJECTIVES: The aim of the study was to evaluate the potential changes induced by fish oil (FO) supplementation on the redox status of pancreatic islets from healthy rats. To test whether these effects were due to eicosapentaenoic acid and docosahexaenoic acid (ω-3), in vitro experiments were performed. METHODS: Rats were supplemented with FO, and pancreatic islets were obtained. Islets were also treated in vitro with palmitate (P) or eicosapentaenoic acid + docosahexaenoic acid (ω-3). Insulin secretion (GSIS), glucose oxidation, protein expression, and superoxide content were analyzed. RESULTS: The FO group showed a reduction in superoxide content. Moreover, FO reduced the expression of NAD(P)H oxidase subunits and increased superoxide dismutase, without altering ß-cell function. Palmitate increased ß-cell reactive oxygen species (ROS) production, apoptosis, and impaired GSIS. Under these conditions, ω-3 triggered a parallel reduction in ROS production and ß-cell apoptosis induced by P and protected against the impairment in GSIS. There was no difference in mitochondrial ROS production. CONCLUSIONS: Our results show that ω-3 protect pancreatic islets from alterations induced by P. In vivo FO supplementation modulates the redox state of pancreatic ß-cell. Considering that in vitro effects do not involve mitochondrial superoxide production, we can speculate that this protection might involve NAD(P)H oxidase activity.


Asunto(s)
Antioxidantes/administración & dosificación , Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Ácido Eicosapentaenoico/administración & dosificación , Islotes Pancreáticos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/sangre , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Glutatión/metabolismo , Insulina/sangre , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Ácido Palmítico/toxicidad , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos
15.
Biochem J ; 460(3): 411-23, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24678915

RESUMEN

The glucose stimulation of insulin secretion by pancreatic ß-cells depends on increased production of metabolic coupling factors, among which changes in NADPH and ROS (reactive oxygen species) may alter the glutathione redox state (EGSH) and signal through changes in thiol oxidation. However, whether nutrients affect EGSH in ß-cell subcellular compartments is unknown. Using redox-sensitive GFP2 fused to glutaredoxin 1 and its mitochondria-targeted form, we studied the acute nutrient regulation of EGSH in the cytosol/nucleus or the mitochondrial matrix of rat islet cells. These probes were mainly expressed in ß-cells and reacted to low concentrations of exogenous H2O2 and menadione. Under control conditions, cytosolic/nuclear EGSH was close to -300 mV and unaffected by glucose (from 0 to 30 mM). In comparison, mitochondrial EGSH was less negative and rapidly regulated by glucose and other nutrients, ranging from -280 mV in the absence of glucose to -299 mV in 30 mM glucose. These changes were largely independent from changes in intracellular Ca(2+) concentration and in mitochondrial pH. They were unaffected by overexpression of SOD2 (superoxide dismutase 2) and mitochondria-targeted catalase, but were inversely correlated with changes in NAD(P)H autofluorescence, suggesting that they indirectly resulted from increased NADPH availability rather than from changes in ROS concentration. Interestingly, the opposite regulation of mitochondrial EGSH and NAD(P)H autofluorescence by glucose was also observed in human islets isolated from two donors. In conclusion, the present study demonstrates that glucose and other nutrients acutely reduce mitochondrial, but not cytosolic/nuclear, EGSH in pancreatic ß-cells under control conditions.


Asunto(s)
Glucosa/farmacología , Glutatión/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocondrias/efectos de los fármacos , Animales , Calcio/metabolismo , Catalasa/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Células HEK293 , Humanos , Peróxido de Hidrógeno/metabolismo , Concentración de Iones de Hidrógeno , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Mitocondrias/fisiología , NADP/metabolismo , Oxidación-Reducción , Ratas , Especies Reactivas de Oxígeno/metabolismo , Vitamina K 3/metabolismo
16.
PLoS One ; 7(10): e46831, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23056475

RESUMEN

AIM/HYPOTHESIS: Rat pancreatic islet cell apoptosis is minimal after prolonged culture in 10 mmol/l glucose (G10), largely increased in 5 mmol/l glucose (G5) and moderately increased in 30 mmol/l glucose (G30). This glucose-dependent asymmetric V-shaped profile is preceded by parallel changes in the mRNA levels of oxidative stress-response genes like Metallothionein 1a (Mt1a). In this study, we tested the effect of ZnCl(2), a potent inducer of Mt1a, on apoptosis, mitochondrial oxidative stress and alterations of glucose-induced insulin secretion (GSIS) induced by prolonged exposure to low and high vs. intermediate glucose concentrations. METHODS: Male Wistar rat islets were cultured in RPMI medium. Islet gene mRNA levels were measured by RTq-PCR. Apoptosis was quantified by measuring islet cytosolic histone-associated DNA fragments and the percentage of TUNEL-positive ß-cells. Mitochondrial thiol oxidation was measured in rat islet cell clusters expressing "redox sensitive GFP" targeted to the mitochondria (mt-roGFP1). Insulin secretion was measured by RIA. RESULTS: As observed for Mt1a mRNA levels, ß-cell apoptosis and loss of GSIS, culture in either G5 or G30 vs. G10 significantly increased mt-roGFP1 oxidation. While TPEN decreased Mt1a/2a mRNA induction by G5, addition of 50-100 µM ZnCl(2) to the culture medium strongly increased Mt1a/2a mRNA and protein levels, reduced early mt-roGFP oxidation and significantly decreased late ß-cell apoptosis after prolonged culture in G5 or G30 vs. G10. It did not, however, prevent the loss of GSIS under these culture conditions. CONCLUSION: ZnCl(2) reduces mitochondrial oxidative stress and improves rat ß-cell survival during culture in the presence of low and high vs. intermediate glucose concentrations without improving their acute GSIS.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Cloruros/farmacología , Citoprotección/efectos de los fármacos , Glucosa/farmacología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Compuestos de Zinc/farmacología , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Técnicas de Cultivo de Célula , Quelantes/farmacología , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Metalotioneína/genética , Metalotioneína/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Compuestos de Sulfhidrilo/metabolismo , Factores de Tiempo , Transportador 8 de Zinc
17.
Gen Physiol Biophys ; 31(1): 65-76, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22447832

RESUMEN

Chronic administration of glucocorticoids (GC) leads to characteristic features of type 2 diabetes in mammals. The main action of dexamethasone in target cells occurs through modulation of gene expression, although the exact mechanisms are still unknown. We therefore investigated the gene expression profile of pancreatic islets from rats treated with dexamethasone using a cDNA array screening analysis. The expression of selected genes and proteins involved in mitochondrial apoptosis was further analyzed by PCR and immunoblotting. Insulin, triglyceride and free fatty acid plasma levels, as well as glucose-induced insulin secretion, were significantly higher in dexamethasone-treated rats compared with controls. Out of 1176 genes, 60 were up-regulated and 28 were down-regulated by dexamethasone treatment. Some of the modulated genes are involved in apoptosis, stress response, and proliferation pathways. RT-PCR confirmed the cDNA array results for 6 selected genes. Bax α protein expression was increased, while Bcl-2 was decreased. In vivo dexamethasone treatment decreased the mitochondrial production of NAD(P)H, and increased ROS production. Concluding, our data indicate that dexamethasone modulates the expression of genes and proteins involved in several pathways of pancreatic-islet cells, and mitochondria dysfunction might be involved in the deleterious effects after long-term GC treatment.


Asunto(s)
Dexametasona/farmacología , Regulación de la Expresión Génica/fisiología , Islotes Pancreáticos/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Islotes Pancreáticos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
18.
Biochem J ; 441(3): 971-8, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22050124

RESUMEN

Using the ROS (reactive oxygen species)-sensitive fluorescent dyes dichlorodihydrofluorescein and dihydroethidine, previous studies yielded opposite results about the glucose regulation of oxidative stress in insulin-secreting pancreatic ß-cells. In the present paper, we used the ratiometric fluorescent proteins HyPer and roGFP1 (redox-sensitive green fluorescent protein 1) targeted to mitochondria [mt-HyPer (mitochondrial HyPer)/mt-roGFP1 (mitochondrial roGFP1)] to monitor glucose-induced changes in mitochondrial hydrogen peroxide concentration and glutathione redox state in adenovirus-infected rat islet cell clusters. Because of the reported pH sensitivity of HyPer, the results were compared with those obtained with the mitochondrial pH sensors mt-AlpHi and mt-SypHer. The fluorescence ratio of the mitochondrial probes slowly decreased (mt-HyPer) or increased (mt-roGFP1) in the presence of 10 mmol/l glucose. Besides its expected sensitivity to H2O2, mt-HyPer was also highly pH sensitive. In agreement, changes in mitochondrial metabolism similarly affected mt-HyPer, mt-AlpHi and mt-SypHer fluorescence signals. In contrast, the mt-roGFP1 fluorescence ratio was only slightly affected by pH and reversibly increased when glucose was lowered from 10 to 2 mmol/l. This increase was abrogated by the catalytic antioxidant Mn(III) tetrakis (4-benzoic acid) porphyrin but not by N-acetyl-L-cysteine. In conclusion, due to its pH sensitivity, mt-HyPer is not a reliable indicator of mitochondrial H2O2 in ß-cells. In contrast, the mt-roGFP1 fluorescence ratio monitors changes in ß-cell mitochondrial glutathione redox state with little interference from pH changes. Our results also show that glucose acutely decreases rather than increases mitochondrial thiol oxidation in rat ß-cells.


Asunto(s)
Glutatión/análisis , Proteínas Fluorescentes Verdes/análisis , Peróxido de Hidrógeno/análisis , Células Secretoras de Insulina/química , Mediciones Luminiscentes/métodos , Mitocondrias/química , Animales , Glutatión/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Peróxido de Hidrógeno/metabolismo , Concentración de Iones de Hidrógeno , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestructura , Cinética , Masculino , Mitocondrias/metabolismo , Concentración Osmolar , Oxidación-Reducción , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Sensibilidad y Especificidad
19.
Redox Rep ; 16(4): 173-80, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21888768

RESUMEN

OBJECTIVES: Reactive oxygen species (ROS) are involved in many physiological and pathological processes. In the present study, we analysed whether the synthetic glucocorticoid dexamethasone induces oxidative stress in cultured pancreatic islets and whether the effects of dexamethasone on insulin secretion, gene expression, and viability can be counteracted by concomitant incubation with N-acetylcysteine (NAC). METHODS: ROS production was measured by dichlorofluorescein (DCFH-DA) assay, insulin secretion by radioimmunoassay, intracellular calcium dynamics by fura-2-based fluorescence, gene expression by real-time polymerase chain reaction analyses and cell viability by the MTS assay. RESULTS: Dexamethasone (Dexa) increased ROS production and decreased glucose-stimulated insulin secretion after 72 hours incubation. Intracellular ROS levels were decreased and the insulin secretion capacity was recovered by concomitant treatment with Dexa+NAC. The total insulin content and intracellular Ca2+ levels were not modulated in either Dexa or Dexa+NAC groups. There was a decrease in the NAD(P)H production, used as an indicator of viability, after dexamethasone treatment. Concomitant incubation with NAC returned viability to control levels. Dexa also decreased synaptotagmin VII (SYT VII) gene expression. In contrast, the Dexa+NAC group demonstrated an increased expression of SYT VII compared to controls. Surprisingly, treatment with NAC decreased the gene expression of the antioxidant enzyme copper zinc superoxide dismutase soluble. DISCUSSION: Our results indicate that dexamethasone increases ROS production, decreases viability, and impairs insulin secretion in pancreatic rat islets. These effects can be counteracted by NAC, which not only decreases ROS levels but also modulates the expression of genes involved in the secretory pathway and those coding for antioxidant enzymes.


Asunto(s)
Acetilcisteína/farmacología , Dexametasona/antagonistas & inhibidores , Glucocorticoides/antagonistas & inhibidores , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Sustancias Protectoras/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dexametasona/toxicidad , Depuradores de Radicales Libres/farmacología , Radicales Libres/metabolismo , Glucocorticoides/toxicidad , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Sinaptotagminas/efectos de los fármacos , Sinaptotagminas/metabolismo
20.
Free Radic Biol Med ; 47(10): 1386-93, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19698781

RESUMEN

Pancreatic beta cells are very sensitive to reactive oxygen species (ROS) and this might play an important role in beta cell death in diabetes. Dexamethasone is a synthetic diabetogenic glucocorticoid, which impairs pancreatic beta cell function. Therefore we investigated the toxicity of dexamethasone in RINm5F insulin-producing cells and its dependence on the expression level of the antioxidant enzyme catalase, which inactivates hydrogen peroxide. This was correlated with oxidative stress and cell death. An increased generation of ROS was observed in dexamethasone-treated cells together with an increase in caspase-3 activity and apoptosis rate. Interestingly, exposure to dexamethasone increased the cytosolic superoxide dismutase Cu/ZnSOD protein expression and activity, whereas the mitochondrial MnSOD isoform was not affected by the glucocorticoid. Catalase overexpression in insulin-producing cells prevented all the cytotoxic effects of dexamethasone. In conclusion, dexamethasone-induced cell death in insulin-producing cells is ROS mediated. Increased levels of expression and activity of the Cu/ZnSOD might favor the generation of hydrogen peroxide in dexamethasone-treated cells. Increased ROS scavenging capacity in insulin-producing cells, through overexpression of catalase, prevents a deleterious increase in hydrogen peroxide generation and thus prevents dexamethasone-induced apoptosis.


Asunto(s)
Antioxidantes/metabolismo , Catalasa/metabolismo , Dexametasona/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , Animales , Caspasa 3/metabolismo , Catalasa/biosíntesis , Catalasa/genética , Muerte Celular , Células Cultivadas , Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Células Secretoras de Insulina/metabolismo , Estrés Oxidativo , Ratas , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...