Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 14: 1182428, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37284303

RESUMEN

Introduction: The high prevalence of neurodegenerative diseases in our population and the lack of effective treatments encourage the search for new therapeutic targets for these pathologies. We have recently described that submaximal inhibition of the Sarco-Endoplasmic Reticulum Ca2+ ATPase (SERCA), the main responsible for ER calcium storage, is able to increase lifespan in Caenorhabditis elegans worms by mechanisms involving mitochondrial metabolism and nutrient-sensitive pathways. Methods: We have studied here the effects of submaximal SERCA inhibition in a chemical model of Parkinson's disease (PD) induced in C. elegans worms by treatment with the mitochondrial complex I inhibitor rotenone. For specific SERCA inhibition, we treated worms with RNAi against sca-1, the sole orthologue of SERCA in C. elegans. Results and Discussion: Our results show that rotenone produces alterations in worms that include decreased lifespan, smaller size, reduced fertility, decreased motility, defecation and pumping rate, increased mitochondrial ROS production, reduced mitochondrial membrane potential and oxygen consumption rate, altered mitochondrial structure, and altered ethanol preference in behavioral studies. Most of these alterations were either fully or partially reversed in worms treated with sca-1 RNAi, suggesting that SERCA inhibition could be a novel pharmacological target in the prevention or treatment of neurodegeneration.

2.
Redox Biol ; 64: 102759, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37302345

RESUMEN

Regulation of mitochondrial redox balance is emerging as a key event for cell signaling in both physiological and pathological conditions. However, the link between the mitochondrial redox state and the modulation of these conditions remains poorly defined. Here, we discovered that activation of the evolutionary conserved mitochondrial calcium uniporter (MCU) modulates mitochondrial redox state. By using mitochondria-targeted redox and calcium sensors and genetic MCU-ablated models, we provide evidence of the causality between MCU activation and net reduction of mitochondrial (but not cytosolic) redox state. Redox modulation of redox-sensitive groups via MCU stimulation is required for maintaining respiratory capacity in primary human myotubes and C. elegans, and boosts mobility in worms. The same benefits are obtained bypassing MCU via direct pharmacological reduction of mitochondrial proteins. Collectively, our results demonstrate that MCU regulates mitochondria redox balance and that this process is required to promote the MCU-dependent effects on mitochondrial respiration and mobility.


Asunto(s)
Caenorhabditis elegans , Mitocondrias , Animales , Humanos , Caenorhabditis elegans/metabolismo , Calcio/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Respiración
3.
J Cell Sci ; 136(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36942724

RESUMEN

Glucose sensing in pancreatic ß-cells depends on oxidative phosphorylation and mitochondria-derived signals that promote insulin secretion. Using mass spectrometry-based phosphoproteomics to search for downstream effectors of glucose-dependent signal transduction in INS-1E insulinoma cells, we identified the outer mitochondrial membrane protein SLC25A46. Under resting glucose concentrations, SLC25A46 was phosphorylated on a pair of threonine residues (T44/T45) and was dephosphorylated in response to glucose-induced Ca2+ signals. Overexpression of SLC25A46 in INS-1E cells caused complete mitochondrial fragmentation, resulting in a mild mitochondrial defect associated with lowered glucose-induced insulin secretion. In contrast, inactivation of the Slc25a46 gene resulted in dramatic mitochondrial hyperfusion, without affecting respiratory activity or insulin secretion. Consequently, SLC25A46 is not essential for metabolism-secretion coupling under normal nutrient conditions. Importantly, insulin-secreting cells lacking SLC25A46 had an exacerbated sensitivity to lipotoxic conditions, undergoing massive apoptosis when exposed to palmitate. Therefore, in addition to its role in mitochondrial dynamics, SLC25A46 plays a role in preventing mitochondria-induced apoptosis in INS-E cells exposed to nutrient stress. By protecting mitochondria, SLC25A46 might help to maintain ß-cell mass essential for blood glucose control.


Asunto(s)
Células Secretoras de Insulina , Neoplasias Pancreáticas , Animales , Ratas , Glucosa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Dinámicas Mitocondriales , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas de Transporte de Fosfato/metabolismo
4.
Methods Mol Biol ; 2565: 153-177, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36205893

RESUMEN

The molecular mechanisms that mediate and regulate calcium (Ca2+) fluxes through the membranes of intracellular organelles play a key role in the generation and shaping of the local and global cytosolic Ca2+ signals triggering the process of regulated exocytosis in chromaffin cells. Beyond that role, intraorganellar Ca2+ homeostasis also regulates organelle-specific processes such as oxidative phosphorylation in mitochondria, maturation of secretory granules, or stress in the endoplasmic reticulum. In this chapter, we describe current methods to study mitochondrial, endoplasmic reticulum, and secretory vesicle calcium homeostasis in living chromaffin cells using engineered targeted aequorins.


Asunto(s)
Aequorina , Células Cromafines , Aequorina/metabolismo , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cromafines/metabolismo , Retículo Endoplásmico/metabolismo , Orgánulos/metabolismo
5.
J Biol Chem ; 298(3): 101652, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35101444

RESUMEN

Mitochondrial dysfunction induces a strong adaptive retrograde signaling response; however, many of the downstream effectors of this response remain to be discovered. Here, we studied the shared transcriptional responses to three different mitochondrial respiratory chain inhibitors in human primary skin fibroblasts using QuantSeq 3'-RNA-sequencing. We found that genes involved in the mevalonate pathway were concurrently downregulated, irrespective of the respiratory chain complex affected. Targeted metabolomics demonstrated that impaired mitochondrial respiration at any of the three affected complexes also had functional consequences on the mevalonate pathway, reducing levels of cholesterol precursor metabolites. A deeper study of complex I inhibition showed a reduced activity of endoplasmic reticulum-bound sterol-sensing enzymes through impaired processing of the transcription factor Sterol Regulatory Element-Binding Protein 2 and accelerated degradation of the endoplasmic reticulum cholesterol-sensors squalene epoxidase and HMG-CoA reductase. These adaptations of mevalonate pathway activity affected neither total intracellular cholesterol levels nor the cellular free (nonesterified) cholesterol pool. Finally, measurement of intracellular cholesterol using the fluorescent cholesterol binding dye filipin revealed that complex I inhibition elevated cholesterol on intracellular compartments. Taken together, our study shows that mitochondrial respiratory chain dysfunction elevates intracellular free cholesterol levels and therefore attenuates the expression of mevalonate pathway enzymes, which lowers endogenous cholesterol biosynthesis, disrupting the metabolic output of the mevalonate pathway. We conclude that intracellular disturbances in cholesterol homeostasis may alter systemic cholesterol management in diseases associated with declining mitochondrial function.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón , Ácido Mevalónico , Mitocondrias , Proteína 2 de Unión a Elementos Reguladores de Esteroles , Esteroles , Colesterol/metabolismo , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Ácido Mevalónico/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Transducción de Señal , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Esteroles/metabolismo
6.
Biomedicines ; 10(2)2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35203497

RESUMEN

Alzheimer's disease (AD) is the most frequent cause of dementia. After decades of research, we know the importance of the accumulation of protein aggregates such as ß-amyloid peptide and phosphorylated tau. We also know that mutations in certain proteins generate early-onset Alzheimer's disease (EOAD), and many other genes modulate the disease in its sporadic form. However, the precise molecular mechanisms underlying AD pathology are still unclear. Because of ethical limitations, we need to use animal models to investigate these processes. The nematode Caenorhabditis elegans has received considerable attention in the last 25 years, since the first AD models overexpressing Aß peptide were described. We review here the main results obtained using this model to study AD. We include works studying the basic molecular mechanisms of the disease, as well as those searching for new therapeutic targets. Although this model also has important limitations, the ability of this nematode to generate knock-out or overexpression models of any gene, single or combined, and to carry out toxicity, recovery or survival studies in short timeframes with many individuals and at low cost is difficult to overcome. We can predict that its use as a model for various diseases will certainly continue to increase.

7.
Br J Pharmacol ; 178(10): 2077-2095, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32056196

RESUMEN

Pancreatic beta-cells are central regulators of glucose homeostasis. By tightly coupling nutrient sensing and granule exocytosis, beta-cells adjust the secretion of insulin to the circulating blood glucose levels. Failure of beta-cells to augment insulin secretion in insulin-resistant individuals leads progressively to impaired glucose tolerance, Type 2 diabetes, and diabetes-related diseases. Mitochondria play a crucial role in ß-cells during nutrient stimulation, linking the metabolism of glucose and other secretagogues to the generation of signals that promote insulin secretion. Mitochondria are double-membrane organelles containing numerous channels allowing the transport of ions across both membranes. These channels regulate mitochondrial energy production, signalling, and cell death. The mitochondria of ß-cells express ion channels whose physio/pathological role is underappreciated. Here, we describe the mitochondrial ion channels identified in pancreatic ß-cells, we further discuss the possibility of targeting specific ß-cell mitochondrial channels for the treatment of Type 2 diabetes, and we finally highlight the evidence from clinical studies. LINKED ARTICLES: This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Canales Iónicos/metabolismo , Iones/metabolismo , Mitocondrias/metabolismo
8.
Diabetologia ; 63(12): 2628-2640, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32960311

RESUMEN

AIMS/HYPOTHESIS: In islets from individuals with type 2 diabetes and in islets exposed to chronic elevated glucose, mitochondrial energy metabolism is impaired. Here, we studied early metabolic changes and mitochondrial adaptations in human beta cells during chronic glucose stress. METHODS: Respiration and cytosolic ATP changes were measured in human islet cell clusters after culture for 4 days in 11.1 mmol/l glucose. Metabolomics was applied to analyse intracellular metabolite changes as a result of glucose stress conditions. Alterations in beta cell function were followed using insulin secretion assays or cytosolic calcium signalling after expression of the calcium probe YC3.6 specifically in beta cells of islet clusters. RESULTS: At early stages of glucose stress, mitochondrial energy metabolism was augmented in contrast to the previously described mitochondrial dysfunction in beta cells from islets of diabetic donors. Following chronic glucose stress, mitochondrial respiration increased (by 52.4%, p < 0.001) and, as a consequence, the cytosolic ATP/ADP ratio in resting human pancreatic islet cells was elevated (by 27.8%, p < 0.05). Because of mitochondrial overactivation in the resting state, nutrient-induced beta cell activation was reduced. In addition, chronic glucose stress caused metabolic adaptations that resulted in the accumulation of intermediates of the glycolytic pathway, the pentose phosphate pathway and the TCA cycle; the most strongly augmented metabolite was glycerol 3-phosphate. The changes in metabolites observed are likely to be due to the inability of mitochondria to cope with continuous nutrient oversupply. To protect beta cells from chronic glucose stress, we inhibited mitochondrial pyruvate transport. Metabolite concentrations were partially normalised and the mitochondrial respiratory response to nutrients was markedly improved. Furthermore, stimulus-secretion coupling as assessed by cytosolic calcium signalling, was restored. CONCLUSION/INTERPRETATION: We propose that metabolic changes and associated mitochondrial overactivation are early adaptations to glucose stress, and may reflect what happens as a result of poor blood glucose control. Inhibition of mitochondrial pyruvate transport reduces mitochondrial nutrient overload and allows beta cells to recover from chronic glucose stress. Graphical abstract.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Metabolismo Energético/fisiología , Glucosa/metabolismo , Humanos , Metabolómica/métodos
10.
Nutrients ; 12(2)2020 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-32093050

RESUMEN

Pancreatic ß-cells secrete insulin to lower blood glucose, following a meal. Maintenance of ß-cell function is essential to preventing type 2 diabetes. In pancreatic ß-cells, mitochondrial matrix calcium is an activating signal for insulin secretion. Recently, the molecular identity of the mitochondrial calcium uniporter (MCU), the transporter that mediates mitochondrial calcium uptake, was revealed. Its role in pancreatic ß-cell signal transduction modulation was clarified, opening new perspectives for intervention. Here, we investigated the effects of a mitochondrial Ca2+-targeted nutritional intervention strategy on metabolism/secretion coupling, in a model of pancreatic insulin-secreting cells (INS-1E). Acute treatment of INS-1E cells with the natural plant flavonoid and MCU activator kaempferol, at a low micromolar range, increased mitochondrial calcium rise during glucose stimulation, without affecting the expression level of the MCU and with no cytotoxicity. Enhanced mitochondrial calcium rises potentiated glucose-induced insulin secretion. Conversely, the MCU inhibitor mitoxantrone inhibited mitochondrial Ca2+ uptake and prevented both glucose-induced insulin secretion and kaempferol-potentiated effects. The kaempferol-dependent potentiation of insulin secretion was finally validated in a model of a standardized pancreatic human islet. We conclude that the plant product kaempferol activates metabolism/secretion coupling in insulin-secreting cells by modulating mitochondrial calcium uptake.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Quempferoles/farmacología , Animales , Técnicas de Cultivo de Célula , Humanos , Mitocondrias/metabolismo
11.
J Mol Biol ; 432(5): 1446-1460, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-31628953

RESUMEN

Mitochondria carry out many essential functions in metabolism. A central task is the oxidation of nutrients and the generation of ATP by oxidative phosphorylation. Mitochondrial metabolism needs to be tightly regulated for the cell to respond to changes in ATP demand and nutrient supply. Here, we review how protein lysine acetylation contributes to the regulation of mitochondrial metabolism in insulin target tissues and the insulin-secreting pancreatic ß-cell. We summarize recent evidence showing that in pancreatic ß-cells, lysine acetylation occurs on a large number of proteins involved in metabolism. Furthermore, we give a brief overview of the molecular mechanism that controls lysine acetylation dynamics. We propose that protein lysine acetylation is an important mechanism for the fine-tuning of mitochondrial activity in ß-cells during normal physiology. In contrast, nutrient oversupply, oxidative stress, or inhibition of the mitochondrial deacetylase SIRT3 leads to protein lysine hyperacetylation, which impairs mitochondrial function. By perturbing mitochondrial activity in ß-cells and insulin target tissues, protein lysine hyperacetylation may contribute to the development of type 2 diabetes.


Asunto(s)
Acetilación , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina/metabolismo , Lisina/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Mitocondrias/metabolismo , NAD/metabolismo , Fosforilación Oxidativa , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Sirtuina 3/metabolismo
12.
Br J Pharmacol ; 176(17): 3250-3263, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31166006

RESUMEN

BACKGROUND AND PURPOSE: Quinic acid (QA) is an abundant natural compound from plant sources which may improve metabolic health. However, little attention has been paid to its effects on pancreatic beta-cell functions, which contribute to the control of metabolic health by lowering blood glucose. Strategies targeting beta-cell signal transduction are a new approach for diabetes treatment. This study investigated the efficacy of QA to stimulate beta-cell function by targeting the basic molecular machinery of metabolism-secretion coupling. EXPERIMENTAL APPROACH: We measured bioenergetic parameters and insulin exocytosis in a model of insulin-secreting beta-cells (INS-1E), together with Ca2+ homeostasis, using genetically encoded sensors, targeted to different subcellular compartments. Islets from mice chronically infused with QA were also assessed. KEY RESULTS: QA triggered transient cytosolic Ca2+ increases in insulin-secreting cells by mobilizing Ca2+ from intracellular stores, such as endoplasmic reticulum. Following glucose stimulation, QA increased glucose-induced mitochondrial Ca2+ transients. We also observed a QA-induced rise of the NAD(P)H/NAD(P)+ ratio, augmented ATP synthase-dependent respiration, and enhanced glucose-stimulated insulin secretion. QA promoted beta-cell function in vivo as islets from mice infused with QA displayed improved glucose-induced insulin secretion. A diet containing QA improved glucose tolerance in mice. CONCLUSIONS AND IMPLICATIONS: QA modulated intracellular Ca2+ homeostasis, enhancing glucose-stimulated insulin secretion in both INS-1E cells and mouse islets. By increasing mitochondrial Ca2+ , QA activated the coordinated stimulation of oxidative metabolism, mitochondrial ATP synthase-dependent respiration, and therefore insulin secretion. Bioactive agents raising mitochondrial Ca2+ in pancreatic beta-cells could be used to treat diabetes.


Asunto(s)
Productos Biológicos/farmacología , Calcio/metabolismo , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Ácido Quínico/farmacología , Actinidia/química , Animales , Productos Biológicos/química , Productos Biológicos/aislamiento & purificación , Células Cultivadas , Café/química , Relación Dosis-Respuesta a Droga , Hippophae/química , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Prunus/química , Ácido Quínico/química , Ácido Quínico/aislamiento & purificación , Ratas , Relación Estructura-Actividad , Vaccinium macrocarpon/química , Vaccinium myrtillus/química
13.
Cell Commun Signal ; 17(1): 14, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30786936

RESUMEN

BACKGROUND: Glucose is the main secretagogue of pancreatic beta-cells. Uptake and metabolism of the nutrient stimulates the beta-cell to release the blood glucose lowering hormone insulin. This metabolic activation is associated with a pronounced increase in mitochondrial respiration. Glucose stimulation also initiates a number of signal transduction pathways for the coordinated regulation of multiple biological processes required for insulin secretion. METHODS: Shotgun proteomics including TiO2 enrichment of phosphorylated peptides followed by liquid chromatography tandem mass spectrometry on lysates from glucose-stimulated INS-1E cells was used to identify glucose regulated phosphorylated proteins and signal transduction pathways. Kinase substrate enrichment analysis (KSEA) was applied to identify key regulated kinases and phosphatases. Glucose-induced oxygen consumption was measured using a XF96 Seahorse instrument to reveal cross talk between glucose-regulated kinases and mitochondrial activation. RESULTS: Our kinetic analysis of substrate phosphorylation reveal the molecular mechanism leading to rapid activation of insulin biogenesis, vesicle trafficking, insulin granule exocytosis and cytoskeleton remodeling. Kinase-substrate enrichment identified upstream kinases and phosphatases and time-dependent activity changes during glucose stimulation. Activity trajectories of well-known glucose-regulated kinases and phosphatases are described. In addition, we predict activity changes in a number of kinases including NUAK1, not or only poorly studied in the context of the pancreatic beta-cell. Furthermore, we pharmacologically tested whether signaling pathways predicted by kinase-substrate enrichment analysis affected glucose-dependent acceleration of mitochondrial respiration. We find that phosphoinositide 3-kinase, Ca2+/calmodulin dependent protein kinase and protein kinase C contribute to short-term regulation of energy metabolism. CONCLUSIONS: Our results provide a global view into the regulation of kinases and phosphatases in insulin secreting cells and suggest cross talk between glucose-induced signal transduction and mitochondrial activation.


Asunto(s)
Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Transducción de Señal , Animales , Línea Celular , Respiración de la Célula/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Cinética , Ratones , Mitocondrias/efectos de los fármacos , Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteómica , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato/efectos de los fármacos , Factores de Tiempo
14.
Mol Cell Endocrinol ; 481: 71-83, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30476561

RESUMEN

Changes in mitochondrial and cytosolic pH alter the chemical gradient across the inner mitochondrial membrane. The proton chemical gradient contributes to mitochondrial ATP synthesis as well as the uptake and release of metabolites and ions from the organelle. Here mitochondrial pH and ΔpH were studied for the first time in human pancreatic ß-cells. Adenoviruses were used for rat insulin promoter dependent expression of the pH sensor SypHer targeted to either the mitochondrial matrix or the cytosol. The matrix pH in resting human ß-cells is low (pH = 7.50 ±â€¯SD 0.17) compared to published values in other cell types. Consequently, the ΔpH of ß-cells mitochondria is small. Glucose stimulation consistently resulted in acidification of the matrix pH in INS-1E insulinoma cells and ß-cells in intact human islets or islet monolayer cultures. We registered acidification with similar kinetics but of slightly smaller amplitude in the cytosol of ß-cells, thus glucose stimulation further reduced the ΔpH. Infection of human islets with high levels of adenoviruses caused the mitochondrial pH to increase. The apoptosis inducer and broad-spectrum kinase inhibitor staurosporine had similar effects on pH homeostasis. Although staurosporine alone does not affect the mitochondrial pH, glucose slightly increases the matrix pH of staurosporine treated cells. These two cellular stressors alter the normal mitochondrial pH response to glucose in pancreatic ß-cells.


Asunto(s)
Glucosa/farmacología , Células Secretoras de Insulina/citología , Proteínas Luminiscentes/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Adenoviridae/genética , Animales , Células Cultivadas , Genes Reporteros , Humanos , Concentración de Iones de Hidrógeno , Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Proteínas Luminiscentes/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Membranas Mitocondriales/química , Regiones Promotoras Genéticas , Ratas , Transfección
15.
Cell Calcium ; 77: 68-76, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30557841

RESUMEN

The 5' AMP-activated protein kinase (AMPK) is a nutrient-sensitive kinase that plays a key role in the control of cellular energy metabolism. We have explored here the relationship between AMPK and Ca2+ signaling by looking at the effect of an AMPK activator (A769662) and an AMPK inhibitor (dorsomorphin) on histamine-induced Ca2+-release from the endoplasmic reticulum (ER) in HeLa cells. Our data show that incubation with A769662 (EC50 = 29 µM) inhibited histamine-induced Ca2+-release from the ER in intact cells, as well as inositol-1,4,5-trisphosphate (IP3)-induced Ca2+ release in permeabilized cells. On the contrary, dorsomorphin (EC50 = 0.4 µM) activated both histamine and IP3-induced Ca2+-release and reversed the effect of A769662. These results suggest a direct effect of AMPK regulation on IP3 receptor (IP3R) function. A phosphoproteomic study did not reveal changes in IP3R phosphorylation, but showed significant changes in phosphorylation of proteins placed upstream in the IP3R interactome and in several proteins related with Ca2+ metabolism, which could be candidates to mediate the effects observed. In conclusion, our data suggest that AMPK negatively regulates IP3R. This effect constitutes a novel and very important link between Ca2+ signaling and the AMPK pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Pironas/farmacología , Tiofenos/farmacología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Compuestos de Bifenilo , Células HeLa , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo
16.
Exp Cell Res ; 357(2): 170-180, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28527697

RESUMEN

Chloramphenicol and several other antibiotics targeting bacterial ribosomes inhibit mitochondrial protein translation. Inhibition of mitochondrial protein synthesis leads to mitonuclear protein imbalance and reduced respiratory rates as confirmed here in HeLa and PC12 cells. Unexpectedly, respiration in INS-1E insulinoma cells and primary human islets was unaltered in the presence of chloramphenicol. Resting respiratory rates and glucose stimulated acceleration of respiration were also not lowered when a range of antibiotics including, thiamphenicol, streptomycin, gentamycin and doxycycline known to interfere with bacterial protein synthesis were tested. However, chloramphenicol efficiently reduced mitochondrial protein synthesis in INS-1E cells, lowering expression of the mtDNA encoded COX1 subunit of the respiratory chain but not the nuclear encoded ATP-synthase subunit ATP5A. Despite a marked reduction of the essential respiratory chain subunit COX1, normal respiratory rates were maintained in INS-1E cells. ATP-synthase dependent respiration was even elevated in chloramphenicol treated INS-1E cells. Consistent with these findings, glucose-dependent calcium signaling reflecting metabolism-secretion coupling in beta-cells, was augmented. We conclude that antibiotics targeting mitochondria are able to cause mitonuclear protein imbalance in insulin secreting cells. We hypothesize that in contrast to other cell types, compensatory mechanisms are sufficiently strong to maintain normal respiratory rates and surprisingly even result in augmented ATP-synthase dependent respiration and calcium signaling following glucose stimulation. The result suggests that in insulin secreting cells only lowering COX1 below a threshold level may result in a measurable impairment of respiration. When focusing on mitochondrial function, care should be taken when including antibiotics targeting translation for long-term cell culture as depending on the sensitivity of the cell type analyzed, respiration, mitonuclear protein imbalance or down-stream signaling may be altered.


Asunto(s)
Antibacterianos/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/tratamiento farmacológico , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Consumo de Oxígeno/fisiología , Células PC12 , Neoplasias Pancreáticas/metabolismo , Ratas , Respiración/efectos de los fármacos
18.
Sci Rep ; 7: 45961, 2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28401888

RESUMEN

A robust and reproducible method for culturing monolayers of adherent and well-spread primary islet cells on glass coverslips is required for detailed imaging studies by super-resolution and live-cell microscopy. Guided by an observation that dispersed islet cells spread and adhere well on glass surfaces in neuronal co-culture and form a monolayer of connected cells, we demonstrate that in the absence of neurons, well-defined surface coatings combined with components of neuronal culture media collectively support robust attachment and growth of primary human or rat islet cells as monolayers on glass surfaces. The islet cell monolayer cultures on glass stably maintain distinct mono-hormonal insulin+, glucagon+, somatostatin+ and PP+ cells and glucose-responsive synchronized calcium signaling as well as expression of the transcription factors Pdx-1 and NKX-6.1 in beta cells. This technical advance enabled detailed observation of sub-cellular processes in primary human and rat beta cells by super-resolution microscopy. The protocol is envisaged to have broad applicability to sophisticated analyses of pancreatic islet cells that reveal new biological insights, as demonstrated by the identification of an in vitro protocol that markedly increases proliferation of primary beta cells and is associated with a reduction in ciliated, ostensibly proliferation-suppressed beta cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Cilios/metabolismo , Vidrio/química , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Microscopía/métodos , Organogénesis , Actinas/metabolismo , Adolescente , Adulto , Animales , Calcio/metabolismo , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Glucosa/metabolismo , Hipocampo/citología , Humanos , Insulina/metabolismo , Masculino , Microtúbulos/metabolismo , Persona de Mediana Edad , Neuronas/citología , Neuronas/metabolismo , Fenotipo , Ratas Sprague-Dawley , Adulto Joven
19.
J Cell Sci ; 130(11): 1929-1939, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28404787

RESUMEN

Pancreatic ß-cells sense glucose, promoting insulin secretion. Glucose sensing requires the sequential stimulation of glycolysis, mitochondrial metabolism and Ca2+ entry. To elucidate how mitochondrial activation in ß-cells contributes to insulin secretion, we compared the effects of glucose and the mitochondrial substrate methylsuccinate in the INS-1E insulin-secreting cell line at the respective concentrations at which they maximally activate mitochondrial respiration. Both substrates induced insulin secretion with distinct respiratory profiles, mitochondrial hyperpolarization, NADH production and ATP-to-ADP ratios. In contrast to glucose, methylsuccinate failed to induce large [Ca2+] rises and exocytosis proceeded largely independently of mitochondrial ATP synthesis. Both glucose- and methylsuccinate-induced secretion was blocked by diazoxide, indicating that Ca2+ is required for exocytosis. Dynamic assessment of the redox state of mitochondrial thiols revealed a less marked reduction in response to methylsuccinate than with glucose. Our results demonstrate that insulin exocytosis can be promoted by two distinct mechanisms one of which is dependent on mitochondrial ATP synthesis and large Ca2+ transients, and one of which is independent of mitochondrial ATP synthesis and relies on small Ca2+ signals. We propose that the combined effects of Ca2+ and redox reactions can trigger insulin secretion by these two mechanisms.


Asunto(s)
Calcio/metabolismo , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mitocondrias/metabolismo , Succinatos/farmacología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Línea Celular Tumoral , Diazóxido/farmacología , Exocitosis/efectos de los fármacos , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Glucólisis/fisiología , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Imagen Molecular , Consumo de Oxígeno/efectos de los fármacos , Ratas , Análisis de la Célula Individual , Succinatos/metabolismo
20.
EMBO Rep ; 18(3): 451-463, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28174208

RESUMEN

Mitochondrial flashes mediated by optic atrophy 1 (OPA1) fusion protein are bioenergetic responses to stochastic drops in mitochondrial membrane potential (Δψm) whose origin is unclear. Using structurally distinct genetically encoded pH-sensitive probes, we confirm that flashes are matrix alkalinization transients, thereby establishing the pH nature of these events, which we renamed "mitopHlashes". Probes located in cristae or intermembrane space as verified by electron microscopy do not report pH changes during Δψm drops or respiratory chain inhibition. Opa1 ablation does not alter Δψm fluctuations but drastically decreases the efficiency of mitopHlash/Δψm coupling, which is restored by re-expressing fusion-deficient OPA1K301A and preserved in cells lacking the outer-membrane fusion proteins MFN1/2 or the OPA1 proteases OMA1 and YME1L, indicating that mitochondrial membrane fusion and OPA1 proteolytic processing are dispensable. pH/Δψm uncoupling occurs early during staurosporine-induced apoptosis and is mitigated by OPA1 overexpression, suggesting that OPA1 maintains mitopHlash competence during stress conditions. We propose that OPA1 stabilizes respiratory chain supercomplexes in a conformation that enables respiring mitochondria to compensate a drop in Δψm by an explosive matrix pH flash.


Asunto(s)
GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Fusión de Membrana , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Membranas Mitocondriales/metabolismo , Apoptosis , Técnicas Biosensibles , Dinaminas/genética , Dinaminas/metabolismo , Etopósido/farmacología , Expresión Génica , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Concentración de Iones de Hidrógeno , Potencial de la Membrana Mitocondrial , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/ultraestructura , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...