Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 33(3): 4154-4165, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30550357

RESUMEN

Chronic exposure of pancreatic ß cells to high concentrations of free fatty acids leads to lipotoxicity (LT)-mediated suppression of glucose-stimulated insulin secretion. This effect is in part caused by a decline in mitochondrial function as well as by a reduction in lysosomal acidification. Because both mitochondria and lysosomes can alter one another's function, it remains unclear which initiating dysfunction sets off the detrimental cascade of LT, ultimately leading to ß-cell failure. Here, we investigated the effects of restoring lysosomal acidity on mitochondrial function under LT. Our results show that LT induces a dose-dependent lysosomal alkalization accompanied by an increase in mitochondrial mass. This increase is due to a reduction in mitochondrial turnover as analyzed by MitoTimer, a fluorescent protein for which the emission is regulated by mitochondrial clearance rate. Mitochondrial oxygen consumption rate, citrate synthase activity, and ATP content are all reduced by LT. Restoration of lysosomal acidity using lysosome-targeted nanoparticles is accompanied by stimulation of mitochondrial turnover as revealed by mitophagy measurements and the recovery of mitochondrial mass. Remarkably, re-acidification restores citrate synthase activity and ATP content in an insulin secreting ß-cell line (INS-1). Furthermore, nanoparticle-mediated lysosomal reacidification rescues mitochondrial maximal respiratory capacity in both INS-1 cells and primary mouse islets. Therefore, our results indicate that mitochondrial dysfunction is downstream of lysosomal alkalization under lipotoxic conditions and that recovery of lysosomal acidity is sufficient to restore the bioenergetic defects.-Assali, E. A., Shlomo, D., Zeng, J., Taddeo, E. P., Trudeau, K. M., Erion, K. A., Colby, A. H., Grinstaff, M. W., Liesa, M., Las, G., Shirihai, O. S. Nanoparticle-mediated lysosomal reacidification restores mitochondrial turnover and function in ß cells under lipotoxicity.


Asunto(s)
Células Secretoras de Insulina/efectos de los fármacos , Lisosomas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Nanopartículas/administración & dosificación , Animales , Células Cultivadas , Ácidos Grasos no Esterificados/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitofagia/efectos de los fármacos
2.
EMBO Rep ; 18(7): 1123-1138, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28539390

RESUMEN

BAT-controlled thermogenic activity is thought to be required for its capacity to prevent the development of insulin resistance. This hypothesis predicts that mediators of thermogenesis may help prevent diet-induced insulin resistance. We report that the mitochondrial fusion protein Mitofusin 2 (Mfn2) in BAT is essential for cold-stimulated thermogenesis, but promotes insulin resistance in obese mice. Mfn2 deletion in mice through Ucp1-cre (BAT-Mfn2-KO) causes BAT lipohypertrophy and cold intolerance. Surprisingly however, deletion of Mfn2 in mice fed a high fat diet (HFD) results in improved insulin sensitivity and resistance to obesity, while impaired cold-stimulated thermogenesis is maintained. Improvement in insulin sensitivity is associated with a gender-specific remodeling of BAT mitochondrial function. In females, BAT mitochondria increase their efficiency for ATP-synthesizing fat oxidation, whereas in BAT from males, complex I-driven respiration is decreased and glycolytic capacity is increased. Thus, BAT adaptation to obesity is regulated by Mfn2 and with BAT-Mfn2 absent, BAT contribution to prevention of insulin resistance is independent and inversely correlated to whole-body cold-stimulated thermogenesis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/genética , Resistencia a la Insulina , Termogénesis/genética , Animales , Dieta Alta en Grasa , Metabolismo Energético , Femenino , Glucólisis , Masculino , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Obesidad
3.
J Mol Biol ; 430(24): 4823-4833, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30389435

RESUMEN

Elamipretide is a tetrapeptide that restores defects in mitochondrial function, binds to cardiolipin, and is being tested in clinical trials for mitochondria-related diseases. However, whether elamipretide modulates mitochondrial quality control and dynamics, processes essential to preserve mitochondrial function, is unclear. Thus, we tested the effects of elamipretide on mitochondrial morphology, mitophagosome formation, and their early disruption induced by excess nutrients in INS1 ß-cells. Elamipretide treatment was sufficient to increase engulfment of mitochondria into autophagosomes in control INS1 ß-cells, without inducing widespread changes in mitochondrial morphology or membrane potential. In an early pathogenic context mimicked by short-term exposure to nutrient excess, elamipretide treatment prevented both mitochondrial fragmentation and defects in the engulfment of mitochondria into autophagosomes. On the other hand, elamipretide did not prevent lysosomal defects induced by nutrient excess. Accordingly, elamipretide treatment did not entail benefits on pathogenic p62 and LC3II accumulation or on insulin secretory function. In conclusion, our data show that elamipretide selectively stimulates the engulfment of mitochondria into autophagosomes and prevents its defects induced by nutrient excess. Thus, we propose that improved selectivity of mitochondrial quality control processes might contribute to the benefits stemming from elamipretide treatments in other disease models.


Asunto(s)
Autofagosomas/metabolismo , Células Secretoras de Insulina/citología , Mitocondrias/efectos de los fármacos , Nutrientes/farmacología , Oligopéptidos/farmacología , Línea Celular , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Lisosomas/metabolismo , Potencial de la Membrana Mitocondrial , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Recambio Mitocondrial/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo
4.
EMBO Mol Med ; 9(3): 353-370, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28130275

RESUMEN

Mechanistic target of rapamycin (mTOR) coordinates biosynthetic and catabolic processes in response to multiple extracellular and intracellular signals including growth factors and nutrients. This serine/threonine kinase has long been known as a critical regulator of muscle mass. The recent finding that the decision regarding its activation/inactivation takes place at the lysosome undeniably brings mTOR into the field of lysosomal storage diseases. In this study, we have examined the involvement of the mTOR pathway in the pathophysiology of a severe muscle wasting condition, Pompe disease, caused by excessive accumulation of lysosomal glycogen. Here, we report the dysregulation of mTOR signaling in the diseased muscle cells, and we focus on potential sites for therapeutic intervention. Reactivation of mTOR in the whole muscle of Pompe mice by TSC knockdown resulted in the reversal of atrophy and a striking removal of autophagic buildup. Of particular interest, we found that the aberrant mTOR signaling can be reversed by arginine. This finding can be translated into the clinic and may become a paradigm for targeted therapy in lysosomal, metabolic, and neuromuscular diseases.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Arginina/administración & dosificación , Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Ratones , Ratones Noqueados , Músculos/patología , Resultado del Tratamiento
5.
J Cell Biol ; 214(1): 25-34, 2016 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-27377248

RESUMEN

In pancreatic ß-cells, liver hepatocytes, and cardiomyocytes, chronic exposure to high levels of fatty acids (lipotoxicity) inhibits autophagic flux and concomitantly decreases lysosomal acidity. Whether impaired lysosomal acidification is causally inhibiting autophagic flux and cellular functions could not, up to the present, be determined because of the lack of an approach to modify lysosomal acidity. To address this question, lysosome-localizing nanoparticles are described that, upon UV photoactivation, enable controlled acidification of impaired lysosomes. The photoactivatable, acidifying nanoparticles (paNPs) demonstrate lysosomal uptake in INS1 and mouse ß-cells. Photoactivation of paNPs in fatty acid-treated INS1 cells enhances lysosomal acidity and function while decreasing p62 and LC3-II levels, indicating rescue of autophagic flux upon acute lysosomal acidification. Furthermore, paNPs improve glucose-stimulated insulin secretion that is reduced under lipotoxicity in INS1 cells and mouse islets. These results establish a causative role for impaired lysosomal acidification in the deregulation of autophagy and ß-cell function under lipotoxicity.


Asunto(s)
Ácidos/metabolismo , Autofagia/efectos de los fármacos , Luz , Lípidos/toxicidad , Lisosomas/metabolismo , Nanopartículas/efectos de la radiación , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Endocitosis/efectos de los fármacos , Glucosa/farmacología , Concentración de Iones de Hidrógeno , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Lisosomas/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Nanopartículas/química , Nanopartículas/ultraestructura , Ácido Palmítico/toxicidad , Rayos Ultravioleta
6.
Methods Enzymol ; 547: 21-38, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25416350

RESUMEN

Current methodologies available to quantify changes in mitochondrial turnover are limited to pulse-chase assays or specific assays that quantify mitophagy. Accordingly, new tools that can assess mitochondrial turnover are needed for the study of cellular, subcellular, and spatial parameters of mitochondrial turnover and quality control. Recently, a group of studies described the use of the MitoTimer fluorescent probe to investigate various aspects of mitochondrial turnover, including changes to protein import, interorganelle protein sharing, and autophagy-mediated turnover. MitoTimer provides a fluorescent readout which directly relates to the mitochondrial turnover rate and allows quantification of relative changes to turnover. Importantly, MitoTimer can be used to investigate mitochondrial turnover on the subcellular level. Due to the fact that MitoTimer is a dual-emission probe and a number of factors can affect MitoTimer readout, certain considerations must be taken into account when using this tool both in experimental design and data interpretation. When used and interpreted appropriately, MitoTimer serves as a unique tool to understand pivotal aspects of mitochondrial turnover.


Asunto(s)
Proteínas Luminiscentes/metabolismo , Mitocondrias/metabolismo , Biología Molecular/métodos , Proteínas Recombinantes de Fusión/metabolismo , Animales , Células COS/efectos de los fármacos , Chlorocebus aethiops , Doxiciclina/farmacología , Fibroblastos , Citometría de Flujo , Colorantes Fluorescentes/metabolismo , Ratones Noqueados , Biología Molecular/instrumentación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA