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












Base de datos
Intervalo de año de publicación
1.
PLoS One ; 14(5): e0216385, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31048932

RESUMEN

FINDINGS: Here, we demonstrate that OP2113 (5-(4-Methoxyphenyl)-3H-1,2-dithiole-3-thione, CAS 532-11-6), synthesized and used as a drug since 1696, does not act as an unspecific antioxidant molecule (i.e., as a radical scavenger) but unexpectedly decreases mitochondrial reactive oxygen species (ROS/H2O2) production by acting as a specific inhibitor of ROS production at the IQ site of complex I of the mitochondrial respiratory chain. Studies performed on isolated rat heart mitochondria also showed that OP2113 does not affect oxidative phosphorylation driven by complex I or complex II substrates. We assessed the effect of OP2113 on an infarct model of ex vivo rat heart in which mitochondrial ROS production is highly involved and showed that OP2113 protects heart tissue as well as the recovery of heart contractile activity. CONCLUSION / SIGNIFICANCE: This work represents the first demonstration of a drug authorized for use in humans that can prevent mitochondria from producing ROS/H2O2. OP2113 therefore appears to be a member of the new class of mitochondrial ROS blockers (S1QELs) and could protect mitochondrial function in numerous diseases in which ROS-induced mitochondrial dysfunction occurs. These applications include but are not limited to aging, Parkinson's and Alzheimer's diseases, cardiac atrial fibrillation, and ischemia-reperfusion injury.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Depuradores de Radicales Libres/farmacología , Mitocondrias Cardíacas/enzimología , Infarto del Miocardio/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Mitocondrias Cardíacas/patología , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Fosforilación Oxidativa/efectos de los fármacos , Ratas , Ratas Wistar
2.
Artículo en Inglés | MEDLINE | ID: mdl-31133988

RESUMEN

Metformin is a drug from the biguanide family that is used for decades as the first-line therapeutic choice for the treatment of type 2 diabetes. Despite its worldwide democratization, owing to its clinical efficacy, high safety profile and cheap cost, the exact mechanism(s) of action of this anti-hyperglycemic molecule with pleiotropic properties still remains to be fully elucidated. The concept that metformin would exert some of its actions though modulation of the mitochondrial bioenergetics was initially forged in the 50s but undeniably revived at the beginning of the twenty-first century when it was shown to induce a weak but specific inhibition of the mitochondrial respiratory-chain complex 1. Furthermore, metformin has been reported to reduce generation of reactive oxygen species at the complex 1 and to prevent mitochondrial-mediated apoptosis, suggesting that it can protect against oxidative stress-induced cell death. Nevertheless, despite some recent progress and the demonstration of its key role in the inhibition of hepatic gluconeogenesis, the exact nature of the mitochondrial interaction between the drug and the complex 1 is still poorly characterized. Recent studies reported that metformin may also have anti-neoplastic properties by inhibiting cancer cell growth and proliferation, at least partly through its mitochondrial action. As such, many trials are currently conducted for exploring the repositioning of metformin as a potential drug for cancer therapy. In this mini-review, we discuss both historical and more recent findings on the central role played by the interaction between metformin and the mitochondria in its cellular mechanism of action.

3.
Circ Arrhythm Electrophysiol ; 11(8): e005913, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30354313

RESUMEN

Background Papillary muscles are an important source of ventricular tachycardia (VT). Yet little is known about the role of the right ventricular (RV) endocavity structure, the moderator band (MB). The aim of this study was to determine the characteristics of the MB that may predispose to arrhythmia substrates. Methods Ventricular wedge preparations with intact MBs were studied from humans (n=2) and sheep (n=15; 40-50 kg). RV endocardium was optically mapped, and electrical recordings were measured along the MB and septum. S1S2 pacing of the RV free wall, MB, or combined S1-RV S2-MB sites were assessed. Human (n=2) and sheep (n=4) MB tissue constituents were assessed histologically. Results The MB structure was remarkably organized as 2 excitable, yet uncoupled compartments of myocardium and Purkinje. In humans, action potential duration heterogeneity between MB and RV myocardium was found (324.6±12.0 versus 364.0±8.4 ms; P<0.0001). S1S2-MB pacing induced unidirectional propagation via MB myocardium, permitting sustained macroreentrant VT. In sheep, the incidence of VT for RV, MB, and S1-RV S2-MB pacing was 1.3%, 5.1%, and 10.3%. Severing the MB led to VT termination, confirming a primary arrhythmic role. Inducible preparations had shorter action potential duration in the MB than RV (259.3±45.2 versus 300.7±38.5 ms; P<0.05), whereas noninducible preparations showed no difference (312.0±30.3 versus 310.0±24.6 ms, respectively). Conclusions The MB presents anatomic and electrical compartmentalization between myocardium and Purkinje fibers, providing a substrate for macroreentry. The vulnerability to sustain VT via this mechanism is dependent on MB structure and action potential duration gradients between the RV free wall and MB.


Asunto(s)
Potenciales de Acción , Frecuencia Cardíaca , Músculos Papilares/fisiopatología , Taquicardia Ventricular/etiología , Animales , Estimulación Cardíaca Artificial , Simulación por Computador , Técnicas Electrofisiológicas Cardíacas , Humanos , Técnicas In Vitro , Modelos Cardiovasculares , Miocardio/patología , Músculos Papilares/patología , Ramos Subendocárdicos/fisiopatología , Oveja Doméstica , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Imagen de Colorante Sensible al Voltaje
4.
PLoS One ; 12(2): e0172053, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28222147

RESUMEN

INTRODUCTION: While metformin (MET) is the most widely prescribed antidiabetic drug worldwide, its beneficial effects in Psammomys obesus (P. obesus), a rodent model that mimics most of the metabolic features of human diabetes, have not been explored thoroughly. Here, we sought to investigate whether MET might improve insulin sensitivity, glucose homeostasis, lipid profile as well as cellular redox and energy balance in P. obesus maintained on a high energy diet (HED). MATERIALS AND METHODS: P. obesus gerbils were randomly assigned to receive either a natural diet (ND) consisting of halophytic plants (control group) or a HED (diabetic group) for a period of 24 weeks. MET (50 mg/kg per os) was administered in both animal groups after 12 weeks of feeding, i.e., the time required for the manifestation of insulin resistance in P. obesus fed a HED. Parallel in vitro experiments were conducted on isolated hepatocytes that were shortly incubated (30 min) with MET and energetic substrates (lactate + pyruvate or alanine, in the presence of octanoate). RESULTS: In vivo, MET lowered glycemia, glycosylated haemoglobin, circulating insulin and fatty acid levels in diabetic P. obesus. It also largely reversed HED-induced hepatic lipid alterations. In vitro, MET increased glycolysis but decreased both gluconeogenesis and ketogenesis in the presence of glucogenic precursors and medium-chain fatty acid. Importantly, these changes were associated with an increase in cytosolic and mitochondrial redox states along with a decline in respiration capacity. CONCLUSIONS: MET prevents the progression of insulin resistance in diabetes-prone P. obesus, possibly through a tight control of gluconeogenesis and fatty acid ß-oxidation depending upon mitochondrial function. While the latter is increasingly becoming a therapeutic issue in diabetes, the gut microbiota is another promising target that would need to be considered as well.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Hígado/efectos de los fármacos , Metformina/uso terapéutico , Mitocondrias Hepáticas/efectos de los fármacos , Animales , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Gerbillinae , Resistencia a la Insulina , Hígado/metabolismo , Masculino , Mitocondrias Hepáticas/metabolismo , Oxidación-Reducción/efectos de los fármacos
5.
J Am Soc Echocardiogr ; 29(12): 1217-1228, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27751650

RESUMEN

BACKGROUND: Left ventricular (LV) afterload could be associated with reduced myocardial contractility. The aim of this study was to evaluate the relative impact of increased afterload on LV myocardial deformation indices in chronic aortic constriction, with regard to hypertrophy, myocardial fibrosis, and mitochondrial function, and to differentiate acute versus chronic afterload effect. METHODS: Young pigs underwent aortic banding (n = 11) or sham (n = 7) operations. Nineteen weeks later, LV morphology and systolic function, including myocardial deformation, were assessed by echocardiography before and after banding release or acute aortic constriction (in the sham group). After the animals were euthanized, mitochondrial function and LV interstitial fibrosis were assessed. RESULTS: The chronic banding group (n = 8) presented with significant LV hypertrophy compared with the sham group (n = 7), and longitudinal strain (LS) was significantly altered (16.9 ± 0.7% vs 20.3 ± 0.7%, P = .001) while circumferential, radial strain, and ejection fraction were not. LS abnormalities were situated mostly on the basal and mid segments and on the septal wall. There was also significantly more myocardial fibrosis in the chronic banding group compared with the sham group, while mitochondrial function was preserved. The relative contributions of hypertrophic and fibrotic remodeling and of afterload to alter global LS were 62%, and 38%, respectively. Acute aortic banding also significantly altered LS. The ratio of LS to septal wall thickness enabled differentiation between chronic and acute afterload increase (1.9 ± 0.2 in the chronic group vs 2.9 ± 0.3 in the acute group, P = .001). CONCLUSIONS: LS is susceptible to both hypertrophic and fibrotic remodeling and afterload increase, particularly on the basal and mid LV segments of the septum. The ratio of LS to septal wall thickness enables differentiation of acute from chronic afterload LS alteration.


Asunto(s)
Estenosis de la Válvula Aórtica/diagnóstico por imagen , Estenosis de la Válvula Aórtica/fisiopatología , Presión Sanguínea , Contracción Miocárdica , Volumen Sistólico , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/fisiopatología , Animales , Estenosis de la Válvula Aórtica/complicaciones , Ecocardiografía/métodos , Módulo de Elasticidad , Diagnóstico por Imagen de Elasticidad/métodos , Masculino , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Estrés Mecánico , Porcinos , Disfunción Ventricular Izquierda/etiología
6.
Physiol Rep ; 4(3)2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26847727

RESUMEN

The aim of this study was to characterize the early alterations of the liver mitochondrial function in ZDF (fa/fa) rats that develop diabetes compared to that of their lean counterparts ZDF (fa/+). Liver mitochondrial function was examined in 11- and 14-week-old ZDF (fa/fa) and ZDF lean (fa/+) rats. Oxygen consumption, H2O2 release, calcium retention capacity (CRC), membrane potential, membrane fluidity, and fatty acid composition were analyzed. State 3 oxygen consumption with palmitoyl-carnitine increases between 11 and 14 weeks of age in lean but not in diabetic animals. This response was not seen with other substrates, suggesting that the use of fatty acids is impaired in diabetic rats. H2O2 release was lower in 14-week-old ZDF (fa/fa) rats as compared to ZDF lean (fa/+). These changes were not associated with differences in enzymatic activities of the respiratory complexes, suggesting regulatory mechanisms independent of their expression levels. Membrane fluidity and composition analyses show only slight effects linked to diabetes progression. The most salient feature was a reduction in CRC in the presence of CsA, an effect reflecting PTP dysregulation. Our data suggest few changes of mitochondrial function in ZDF fa/fa rats. At the age of 11 weeks, liver mitochondria have mainly a reduced effect of CsA on CRC.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hígado/metabolismo , Mitocondrias/metabolismo , Animales , Western Blotting , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Espectroscopía de Resonancia por Spin del Electrón , Citometría de Flujo , Hígado/patología , Espectroscopía de Resonancia Magnética , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/patología , Estrés Oxidativo/fisiología , Consumo de Oxígeno/fisiología , Ratas , Especies Reactivas de Oxígeno/metabolismo
7.
Front Physiol ; 6: 369, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26733871

RESUMEN

Mitochondrial membrane potential is the major regulator of mitochondrial functions, including coupling efficiency and production of reactive oxygen species (ROS). Both functions are crucial for cell bioenergetics. We previously presented evidences for a specific modulation of adenine nucleotide translocase (ANT) appearing during aging that results in a decrease in membrane potential - and therefore ROS production-but surprisingly increases coupling efficiency under conditions of low ATP turnover. Careful study of the bioenergetic parameters (oxidation and phosphorylation rates, membrane potential) of isolated mitochondria from skeletal muscles (gastrocnemius) of aged and young rats revealed a remodeling at the level of the phosphorylation system, in the absence of alteration of the inner mitochondrial membrane (uncoupling) or respiratory chain complexes regulation. We further observed a decrease in mitochondrial affinity for ADP in aged isolated mitochondria, and higher sensitivity of ANT to its specific inhibitor atractyloside. This age-induced modification of ANT results in an increase in the ADP concentration required to sustain the same ATP turnover as compared to young muscle, and therefore in a lower membrane potential under phosphorylating-in vivo-conditions. Thus, for equivalent ATP turnover (cellular ATP demand), coupling efficiency is even higher in aged muscle mitochondria, due to the down-regulation of inner membrane proton leak caused by the decrease in membrane potential. In the framework of the radical theory of aging, these modifications in ANT function may be the result of oxidative damage caused by intra mitochondrial ROS and may appear like a virtuous circle where ROS induce a mechanism that reduces their production, without causing uncoupling, and even leading in improved efficiency. Because of the importance of ROS as therapeutic targets, this new mechanism deserves further studies.

8.
J Diabetes ; 6(2): 184-92, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23953934

RESUMEN

BACKGROUND: Insulin resistance and oxidative stress are major pathogenic mechanisms leading to chronic liver diseases in diabetic subjects. The gerbil Psammomys obesus is a unique model of nutritional diabetes resembling the disease in humans. This study investigated whether the natural ingredient silibinin, known as hepatoprotective, could decrease oxidative stress and reduce liver damage in obese gerbils. METHODS: Control animals were fed their vegetable-based low caloric diet while two other rat groups ingested a high calorie diet for 14 weeks. Silibinin, or its vehicle, was administrated by gastric intubation (100 mg/kg per day) from the 7th week of treatment, which corresponds to an established insulin resistance state. At the end of the experiments, the hepatic biochemical profile, markers of oxidative stress in either plasma or liver tissue, and histological alterations were examined. RESULTS: Diabetic P. obesus displayed many metabolic disturbances (hyperinsulinemia, hyperglycemia, dyslipidemia), which were aggravated for the last 8 weeks. These events were coupled with greater oxidative stress (decline in glutathione, rise in lipoperoxidation). In addition, glutathione peroxidase activity was reduced while the level of superoxide dismutase was elevated. Interestingly, treatment with silibinin alleviated most of the metabolic defects, especially high triglyceride levels, reduced insulin resistance and largely restored antioxidant status. Also, Masson's trichrome staining revealed distinct steatosis, yet silibinin partially reversed this manifestation. CONCLUSION: Silibinin affords substantial protection against the progression of insulin resistance in Type 2 diabetes mellitus for P. obesus by hampering the oxidative process and improving hepatic metabolism.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hígado Graso/tratamiento farmacológico , Síndrome Metabólico/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Silimarina/farmacología , Animales , Antioxidantes/administración & dosificación , Antioxidantes/metabolismo , Antioxidantes/farmacología , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hígado Graso/sangre , Hígado Graso/metabolismo , Gerbillinae , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Insulina/sangre , Lípidos/análisis , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Síndrome Metabólico/sangre , Síndrome Metabólico/metabolismo , Obesidad/sangre , Obesidad/metabolismo , Silibina , Silimarina/administración & dosificación , Superóxido Dismutasa/metabolismo
9.
Aging Cell ; 13(1): 39-48, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23919652

RESUMEN

With aging, most skeletal muscles undergo a progressive loss of mass and strength, a process termed sarcopenia. Aging-related defects in mitochondrial energetics have been proposed to be causally involved in sarcopenia. However, changes in muscle mitochondrial oxidative phosphorylation with aging remain a highly controversial issue, creating a pressing need for integrative approaches to determine whether mitochondrial bioenergetics are impaired in aged skeletal muscle. To address this issue, mitochondrial bioenergetics was first investigated in vivo in the gastrocnemius muscle of adult (6 months) and aged (21 months) male Wistar rats by combining a modular control analysis approach with (31) P magnetic resonance spectroscopy measurements of energetic metabolites. Using this innovative approach, we revealed that the in vivo responsiveness ('elasticity') of mitochondrial oxidative phosphorylation to contraction-induced increase in ATP demand is significantly reduced in aged skeletal muscle, a reduction especially pronounced under low contractile activities. In line with this in vivo aging-related defect in mitochondrial energetics, we found that the mitochondrial affinity for ADP is significantly decreased in mitochondria isolated from aged skeletal muscle. Collectively, the results of this study demonstrate that mitochondrial bioenergetics are effectively altered in vivo in aged skeletal muscle and provide a novel cellular basis for this phenomenon.


Asunto(s)
Envejecimiento/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Translocador 1 del Nucleótido Adenina/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Concentración de Iones de Hidrógeno , Masculino , Contracción Muscular/fisiología , Oxidación-Reducción , Fosforilación Oxidativa , Fosfatos/metabolismo , Fosfocreatina/metabolismo , Ratas , Ratas Wistar
10.
Exp Diabetes Res ; 2012: 430176, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22675340

RESUMEN

Mitochondrial dysfunction is considered to be a pivotal component of insulin resistance and associated metabolic diseases. Psammomys obesus is a relevant model of nutritional diabetes since these adult animals exhibit a state of insulin resistance when fed a standard laboratory chow, hypercaloric for them as compared to their natural food. In this context, alterations in bioenergetics were studied. Using liver mitochondria isolated from these rats fed such a diet for 18 weeks, oxygen consumption rates, activities of respiratory complexes, and content in cytochromes were examined. Levels of malondialdehyde (MDA) and gluthatione (GSH) were measured in tissue homogenates. Diabetic Psammomys showed a serious liver deterioration (hepatic mass accretion, lipids accumulation), accompanied by an enhanced oxidative stress (MDA increased, GSH depleted). On the other hand, both ADP-dependent and uncoupled respirations greatly diminished below control values, and the respiratory flux to cytochrome oxydase was mildly lowered. Furthermore, an inhibition of complexes I and III together with an activation of complex II were found. With emergence of oxidative stress, possibly related to a defect in oxidative phosphorylation, some molecular adjustments could contribute to alleviate, at least in part, the deleterious outcomes of insulin resistance in this gerbil species.


Asunto(s)
Diabetes Mellitus/patología , Gerbillinae/metabolismo , Hígado/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Adenosina Difosfato/química , Animales , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/metabolismo , Electrones , Glutatión/metabolismo , Malondialdehído/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa , Consumo de Oxígeno , Fosforilación , Ratas , Temperatura
11.
Cell Physiol Biochem ; 28(5): 899-910, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22178942

RESUMEN

BACKGROUND/AIMS: Glitazones are synthetic insulin-sensitizing drugs which act as agonists of peroxisome proliferator-activated receptor gamma (PPARγ). However, TZDs action does not exclude independent PPARγ-activation effects. Remarkably, direct mitochondrial action of these agents has not been fully studied yet. METHODS: Oxygen consumption rates (JO(2)) were measured using a Clark-type oxygen electrode in intact hepatocytes and isolated liver mitochondria. Mitochondrial reactive oxygen species (ROS) production was quantified by fluorescence assay. Moreover, activities of mitochondrial respiratory chain complex I, II and III were spectrometrically determined. RESULTS: Pioglitazone and rosiglitazone inhibited JO(2) in liver cells and mitochondria. This inhibition affected the state 3 of respiration (in the presence of ADP) and the uncoupled state (after addition of dinitrophenol). Moreover, these agents dramatically reduced mitochondrial ROS production in all situations tested. We also demonstrated that both glitazones specifically inhibited the activities of complex I and complex III, by 50% and 35% respectively. Additionally, they do not modify neither the oxidative phosphorylation yield nor the permeability transition pore opening. CONCLUSIONS: Pioglitazone and rosiglitazone reduce both respiration intensity and ROS production, acutely and by a probable PPARγ-independent way, through inhibition of complex I and III activities. This new finding could positively contribute to their anti-diabetic properties.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hipoglucemiantes/farmacología , Mitocondrias Hepáticas/efectos de los fármacos , Tiazolidinedionas/farmacología , Animales , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Hepatocitos/fisiología , Masculino , Mitocondrias Hepáticas/metabolismo , Fosforilación Oxidativa , Consumo de Oxígeno , PPAR gamma/agonistas , PPAR gamma/metabolismo , Pioglitazona , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Rosiglitazona
12.
Kidney Int ; 77(10): 861-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20164825

RESUMEN

The antidiabetic drug metformin can diminish apoptosis induced by oxidative stress in endothelial cells and prevent vascular dysfunction even in nondiabetic patients. Here we tested whether it has a beneficial effect in a rat model of gentamicin toxicity. Mitochondrial analysis, respiration intensity, levels of reactive oxygen species, permeability transition, and cytochrome c release were assessed 3 and 6 days after gentamicin administration. Metformin treatment fully blocked gentamicin-mediated acute renal failure. This was accompanied by a lower activity of N-acetyl-beta-D-glucosaminidase, together with a decrease of lipid peroxidation and increase of antioxidant systems. Metformin also protected the kidney from histological damage 6 days after gentamicin administration. These in vivo markers of kidney dysfunction and their correction by metformin were complemented by in vitro studies of mitochondrial function. We found that gentamicin treatment depleted respiratory components (cytochrome c, NADH), probably due to the opening of mitochondrial transition pores. These injuries, partly mediated by a rise in reactive oxygen species from the electron transfer chain, were significantly decreased by metformin. Thus, our study suggests that pleiotropic effects of metformin can lessen gentamicin nephrotoxicity and improve mitochondrial homeostasis.


Asunto(s)
Gentamicinas/farmacología , Hipoglucemiantes/farmacología , Metformina/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Acetilglucosaminidasa/metabolismo , Acetilglucosaminidasa/farmacología , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Grupo Citocromo c , Citocromos c/metabolismo , Citocromos c/farmacología , Transporte de Electrón/efectos de los fármacos , Gentamicinas/metabolismo , Hipoglucemiantes/metabolismo , Riñón/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Metformina/metabolismo , Mitocondrias/fisiología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Permeabilidad , Ratas , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología
13.
J Biol Chem ; 284(22): 15117-25, 2009 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-19346250

RESUMEN

To study the effect of mitochondrial permeability transition pore (PTP) opening on NAD(P)H localization, intact cells were exposed to the Ca(2+) ionophore A23187. PTP opening, mitochondrial membrane potential, mitochondrial volume, and NAD(P)H localization were assessed by time-lapse laser confocal microscopy using the calcein-cobalt technique, tetramethylrhodamine methyl ester, MitoTracker, and NAD(P)H autofluorescence, respectively. Concomitant with PTP opening, NAD(P)H fluorescence increased outside mitochondria. These events occurred in all cells and were prevented by cyclosporin A. Mitochondrial membrane potential was not systematically collapsed, whereas mitochondrial volume did not change, confirming that A23187 induced transient PTP opening in a subpopulation of cells and suggesting that mitochondrial swelling did not immediately occur after PTP opening in intact cells. NAD(P)H autofluorescence remained elevated after PTP opening, particularly after membrane potential had been collapsed by an uncoupler. Extraction of nucleotide for NAD(P)H quantification confirmed that PTP opening led to an increase in NAD(P)H content. Because the oxygen consumption rate decreased, whereas the lactate/pyruvate ratio increased after PTP opening in intact cells, we conclude that PTP opening inhibits respiration and dramatically affects the cytosolic redox potential in intact cells.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial/metabolismo , NADP/metabolismo , Transporte Biológico/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Separación Celular , Fluorescencia , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Imagenología Tridimensional , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , NAD/farmacología , Tamaño de los Orgánulos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos
14.
Life Sci ; 82(21-22): 1070-6, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18448125

RESUMEN

Silibinin, the most biologically active component of the polyphenolic extract from milk thistle seeds, is widely used to prevent many types of hepatobiliary disorders. Recent evidence suggests new applications for this ancient medication, notably for the treatment of type 2 diabetes owing to its antihyperglycemic properties. As we have lately demonstrated that silibinin lowered glucose production from various gluconeogenic substrates in perifused rat hepatocytes, the aim of this study was to examine the effect of silibinin on both oxidative glucose utilization and reactive oxygen species (ROS) generation since the release of ROS secondary to an increased mitochondrial metabolism may contribute to diabetic damage. We found that silibinin dose-dependently reduced glycolysis from carbohydrates in a cell perifusion system via an inhibitory effect targeted on pyruvate kinase activity. Furthermore, a dramatic effect upon oxidative phosphorylation was shown, as evidenced by a fall in ATP-to-ADP ratio, together with an increase in lactate-to-pyruvate ratio. The most attractive finding was that silibinin, at a concentration as low as 10 microM, fully mitigated the rise in metabolic flow-driven ROS formation. In addition, studies on isolated liver mitochondria revealed that this low dose of silibinin depressed ROS production linked to the electron transfer chain activity. From these results, one may tentatively suggest that interesting activities for silibinin, beyond its general antioxidant status, could be expected from its potential clinical use, especially in pathological conditions when mitochondrial ROS formation is severely enhanced.


Asunto(s)
Glucólisis/efectos de los fármacos , Hepatocitos/metabolismo , Mitocondrias Hepáticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Nucleótidos de Adenina/metabolismo , Animales , Separación Celular , Dihidroxiacetona/farmacología , Transporte de Electrón/efectos de los fármacos , Fluoresceínas , Colorantes Fluorescentes , Glucosa/metabolismo , Hepatocitos/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Técnicas In Vitro , Ácido Láctico/metabolismo , Masculino , Mitocondrias Hepáticas/efectos de los fármacos , Oxidantes/farmacología , Perfusión , Proteínas Quinasas/metabolismo , Piruvato Quinasa/metabolismo , Ácido Pirúvico/metabolismo , Ratas , Silibina , Silimarina/farmacología
15.
J Mol Neurosci ; 34(1): 77-87, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18040888

RESUMEN

Oxidative damage has been reported to be involved in the pathogenesis of diabetic neuropathy and neurodegenerative diseases. Recent evidence suggests that the antidiabetic drug metformin prevents oxidative stress-related cellular death in non-neuronal cell lines. In this report, we point to the direct neuroprotective effect of metformin, using the etoposide-induced cell death model. The exposure of intact primary neurons to this cytotoxic insult induced permeability transition pore (PTP) opening, the dissipation of mitochondrial membrane potential (DeltaPsim), cytochrome c release, and subsequent death. More importantly, metformin, together with the PTP classical inhibitor cyclosporin A (CsA), strongly mitigated the activation of this apoptotic cascade. Furthermore, the general antioxidant N-acetyl-L: -cysteine also prevented etoposide-promoted neuronal death. In addition, metformin was shown to delay CsA-sensitive PTP opening in permeabilized neurons, as triggered by a calcium overload, probably through its mild inhibitory effect on the respiratory chain complex I. We conclude that (1) etoposide-induced neuronal death is partly attributable to PTP opening and the disruption of DeltaPsim, in association with the emergence of oxidative stress, and (2) metformin inhibits this PTP opening-driven commitment to death. We thus propose that metformin, beyond its antihyperglycemic role, can also function as a new therapeutic tool for diabetes-associated neurodegenerative disorders.


Asunto(s)
Apoptosis/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Metformina/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/fisiología , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Ciclosporina/farmacología , Citocromos c/metabolismo , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/fisiopatología , Inhibidores Enzimáticos/farmacología , Etopósido/farmacología , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Metformina/uso terapéutico , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/metabolismo , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar
16.
Biochem J ; 404(3): 499-507, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17324122

RESUMEN

AICA riboside (5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside) has been extensively used in cells to activate the AMPK (AMP-activated protein kinase), a metabolic sensor involved in cell energy homoeostasis. In the present study, we investigated the effects of AICA riboside on mitochondrial oxidative; phosphorylation. AICA riboside was found to dose-dependently inhibit the oligomycin-sensitive JO2 (oxygen consumption rate) of isolated rat hepatocytes. A decrease in P(i) (inorganic phosphate), ATP, AMP and total adenine nucleotide contents was also observed with AICA riboside concentrations >0.1 mM. Interestingly, in hepatocytes from mice lacking both alpha1 and alpha2 AMPK catalytic subunits, basal JO2 and expression of several mitochondrial proteins were significantly reduced compared with wild-type mice, suggesting that mitochondrial biogenesis was perturbed. However, inhibition of JO2 by AICA riboside was still present in the mutant mice and thus was clearly not mediated by AMPK. In permeabilized hepatocytes, this inhibition was no longer evident, suggesting that it could be due to intracellular accumulation of Z nucleotides and/or loss of adenine nucleotides and P(i). ZMP did indeed inhibit respiration in isolated rat mitochondria through a direct effect on the respiratory-chain complex I. In addition, inhibition of JO2 by AICA riboside was also potentiated in cells incubated with fructose to deplete adenine nucleotides and P(i). We conclude that AICA riboside inhibits cellular respiration by an AMPK-independent mechanism that likely results from the combined intracellular P(i) depletion and ZMP accumulation. Our data also demonstrate that the cellular effects of AICA riboside are not necessarily caused by AMPK activation and that their interpretation should be taken with caution.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Hipoglucemiantes/farmacología , Hígado/metabolismo , Mitocondrias Hepáticas , Complejos Multienzimáticos/metabolismo , Fosforilación Oxidativa , Proteínas Serina-Treonina Quinasas/metabolismo , Ribonucleósidos/farmacología , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/farmacología , Animales , Células Cultivadas , Coformicina/metabolismo , Relación Dosis-Respuesta a Droga , Complejo I de Transporte de Electrón/fisiología , Activación Enzimática , Inhibidores Enzimáticos/metabolismo , Fructosa/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Complejos Multienzimáticos/genética , Oxígeno/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Wistar
17.
J Bioenerg Biomembr ; 38(1): 33-42, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16732470

RESUMEN

Mitochondrial reactive oxygen species (ROS) production was investigated in mitochondria extracted from liver of rats treated with or without metformin, a mild inhibitor of respiratory chain complex 1 used in type 2 diabetes. A high rate of ROS production, fully suppressed by rotenone, was evidenced in non-phosphorylating mitochondria in the presence of succinate as a single complex 2 substrate. This ROS production was substantially lowered by metformin pretreatment and by any decrease in membrane potential (Delta Phi(m)), redox potential (NADH/NAD), or phosphate potential, as induced by malonate, 2,4-dinitrophenol, or ATP synthesis, respectively. ROS production in the presence of glutamate-malate plus succinate was lower than in the presence of succinate alone, but higher than in the presence of glutamate-malate. Moreover, while rotenone both increased and decreased ROS production at complex 1 depending on forward (glutamate-malate) or reverse (succinate) electron flux, no ROS overproduction was evidenced in the forward direction with metformin. Therefore, we propose that reverse electron flux through complex 1 is an alternative pathway, which leads to a specific metformin-sensitive ROS production.


Asunto(s)
Complejo I de Transporte de Electrón/fisiología , Hipoglucemiantes/farmacología , Metformina/farmacología , Mitocondrias Hepáticas/fisiología , Especies Reactivas de Oxígeno/metabolismo , 2,4-Dinitrofenol/farmacología , Adenosina Trifosfato/biosíntesis , Animales , Transporte de Electrón , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Ácido Glutámico/farmacología , Técnicas In Vitro , Malatos/farmacología , Malonatos/farmacología , Potenciales de la Membrana , Mitocondrias Hepáticas/efectos de los fármacos , Oxidación-Reducción , Fosforilación , Ratas , Ratas Wistar , Rotenona/farmacología , Ácido Succínico/farmacología
18.
Diabetes ; 54(7): 2179-87, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15983220

RESUMEN

Hyperglycemia-induced oxidative stress is detrimental for endothelial cells, contributing to the vascular complications of diabetes. The mitochondrial permeability transition pore (PTP) is an oxidative stress-sensitive channel involved in cell death; therefore, we have examined its potential role in endothelial cells exposed to oxidative stress or high glucose level. Metformin, an antihyperglycemic agent used in type 2 diabetes, was also investigated because it inhibits PTP opening in transformed cell lines. Cyclosporin A (CsA), the reference PTP inhibitor, and a therapeutic dose of metformin (100 micromol/l) led to PTP inhibition in permeabilized human microvascular endothelial cells (HMEC-1). Furthermore, exposure of intact HMEC-1 or primary endothelial cells from either human umbilical vein or bovine aorta to the oxidizing agent tert-butylhydroperoxide or to 30 mmol/l glucose triggered PTP opening, cytochrome c decompartmentalization, and cell death. CsA or metformin prevented all of these effects. The antioxidant N-acetyl-l-cysteine also prevented hyperglycemia-induced apoptosis. We conclude that 1) elevated glucose concentration leads to an oxidative stress that favors PTP opening and subsequent cell death in several endothelial cell types and 2) metformin prevents this PTP opening-related cell death. We propose that metformin improves diabetes-associated vascular disease both by lowering blood glucose and by its effect on PTP regulation.


Asunto(s)
Muerte Celular/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Glucosa/farmacología , Metformina/farmacología , Mitocondrias/fisiología , Línea Celular , Endotelio Vascular/citología , Glucosa/antagonistas & inhibidores , Humanos , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Mitocondrias/efectos de los fármacos , Permeabilidad , Piel/irrigación sanguínea
19.
Biochem J ; 382(Pt 3): 877-84, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15175014

RESUMEN

Metformin, a drug widely used in the treatment of Type II diabetes, has recently received attention owing to new findings regarding its mitochondrial and cellular effects. In the present study, the effects of metformin on respiration, complex 1 activity, mitochondrial permeability transition, cytochrome c release and cell death were investigated in cultured cells from a human carcinoma-derived cell line (KB cells). Metformin significantly decreased respiration both in intact cells and after permeabilization. This was due to a mild and specific inhibition of the respiratory chain complex 1. In addition, metformin prevented to a significant extent mitochondrial permeability transition both in permeabilized cells, as induced by calcium, and in intact cells, as induced by the glutathione-oxidizing agent t-butyl hydroperoxide. This effect was equivalent to that of cyclosporin A, the reference inhibitor. Finally, metformin impaired the t-butyl hydroperoxide-induced cell death, as judged by Trypan Blue exclusion, propidium iodide staining and cytochrome c release. We propose that metformin prevents the permeability transition-related commitment to cell death in relation to its mild inhibitory effect on complex 1, which is responsible for a decreased probability of mitochondrial permeability transition.


Asunto(s)
Muerte Celular/efectos de los fármacos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Canales Iónicos/efectos de los fármacos , Metformina/farmacología , Mitocondrias/efectos de los fármacos , Citrato (si)-Sintasa/metabolismo , Citocromos c/metabolismo , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Canales Iónicos/metabolismo , Células KB , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial , Poro de Transición de la Permeabilidad Mitocondrial , Consumo de Oxígeno/efectos de los fármacos , Permeabilidad
20.
Biochem Pharmacol ; 63(7): 1259-72, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11960602

RESUMEN

From recent findings about the indirect effect of metformin (MET) targeted on the respiratory chain complex I, we reconsidered this question and tried to determine the causality of any alteration at this enzymatic level using Xenopus laevis oocytes. Addition of MET (50 microM) reduced by 40% the rotenone-sensitive activity of complex I only in incubating intact oocytes but not in mitochondria isolated by differential centrifugation. The drug prior injected inside these cells had also no measurable effect. In spite of this and the weak binding of MET to the mitochondrial fraction, there was a fairly good correlation between the marked inhibitory action of MET on complex I and its progressive appearance within the oocyte cytoplasm. The intriguing observation that MET as a liposomal form was again able to exert its role when added directly to isolated mitochondria is in accordance with a membrane-mediated uptake and vesicular routing of MET. Furthermore, a temperature-dependent effect was clearly shown. At 4 degrees, oocytes failed to take up efficiently MET and accordingly its subsequent action on respiration was therefore lost. Likewise, MET transport was hindered and inhibition of complex I totally disappeared when a structural analog, asymmetrical dimethylarginine (ADMA), was placed together with MET either at an identical concentration or in excess. These data strongly support the view that MET may recognise some specific membranous sites, likely belonging to effector systems, before penetrating the cell in a bound state via an obscure endocytotic event which still has to be identified.


Asunto(s)
Hipoglucemiantes/farmacología , Metformina/farmacología , Mitocondrias/efectos de los fármacos , Oocitos/efectos de los fármacos , Animales , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/fisiología , Liposomas , Mitocondrias/enzimología , Oocitos/fisiología , Respiración/efectos de los fármacos , Fracciones Subcelulares , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...