Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Int J Mol Sci ; 22(7)2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807195

RESUMEN

(1) Background: The exact mechanism(s) underlying pathological changes in a heart in transition to hypertrophy and failure are not yet fully understood. However, alterations in cardiac energy metabolism seem to be an important contributor. We characterized an in vitro model of adrenergic stimulation-induced cardiac hypertrophy for studying metabolic, structural, and functional changes over time. Accordingly, we investigated whether metabolic interventions prevent cardiac structural and functional changes; (2) Methods: Primary rat cardiomyocytes were treated with phenylephrine (PE) for 16 h, 24 h, or 48 h, whereafter hypertrophic marker expression, protein synthesis rate, glucose uptake, and contractile function were assessed; (3) Results: 24 h PE treatment increased expression of hypertrophic markers, phosphorylation of hypertrophy-related signaling kinases, protein synthesis, and glucose uptake. Importantly, the increased glucose uptake preceded structural and functional changes, suggesting a causal role for metabolism in the onset of PE-induced hypertrophy. Indeed, PE treatment in the presence of a PAN-Akt inhibitor or of a GLUT4 inhibitor dipyridamole prevented PE-induced increases in cellular glucose uptake and ameliorated PE-induced contractile alterations; (4) Conclusions: Pharmacological interventions, forcing substrate metabolism away from glucose utilization, improved contractile properties in PE-treated cardiomyocytes, suggesting that targeting glucose uptake, independent from protein synthesis, forms a promising strategy to prevent hypertrophy and hypertrophy-induced cardiac dysfunction.


Asunto(s)
Cardiomegalia/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Metabolismo Energético , Glucosa/metabolismo , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Miocitos Cardíacos/efectos de los fármacos , Fenilefrina/farmacología , Fosforilación , Ratas , Transducción de Señal/efectos de los fármacos
2.
Methods Mol Biol ; 1732: 343-361, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29480486

RESUMEN

Here we describe an assay for simultaneous measurement of cellular uptake rates of long-chain fatty acids (LCFA) and glucose that can be applied to cells in suspension. The uptake assay includes the use of radiolabeled substrates at such concentrations and incubation periods that exact information is provided about unidirectional uptakes rates. Cellular uptake of both substrates is under regulation of AMPK. The underlying mechanism includes the translocation of LCFA and glucose transporters from intracellular membrane compartments to the cell surface, leading to an increase in substrate uptake. In this chapter, we explain the principles of the uptake assay before detailing the exact procedure. We also provide information of the specific LCFA and glucose transporters subject to AMPK-mediated subcellular translocation. Finally, we discuss the application of AMPK inhibitors and activators in combination with cellular substrate uptake assays.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Pruebas de Enzimas/métodos , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Animales , Células Cultivadas , Pruebas de Enzimas/instrumentación , Transportador de Glucosa de Tipo 4/metabolismo , Membranas Intracelulares/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Cultivo Primario de Células , Ratas
3.
Cardiovasc Res ; 113(10): 1148-1160, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28549111

RESUMEN

AIMS: Heart failure is associated with altered myocardial substrate metabolism and impaired cardiac energetics. Comorbidities like diabetes may influence the metabolic adaptations during heart failure development. We quantified to what extent changes in substrate preference, lipid accumulation, and energy status predict the longitudinal development of hypertrophy and failure in the non-diabetic and the diabetic heart. METHODS AND RESULTS: Transverse aortic constriction (TAC) was performed in non-diabetic (db/+) and diabetic (db/db) mice to induce pressure overload. Magnetic resonance imaging, 31P magnetic resonance spectroscopy (MRS), 1H MRS, and 18F-fluorodeoxyglucose-positron emission tomography (PET) were applied to measure cardiac function, energy status, lipid content, and glucose uptake, respectively. In vivo measurements were complemented with ex vivo techniques of high-resolution respirometry, proteomics, and western blotting to elucidate the underlying molecular pathways. In non-diabetic mice, TAC induced progressive cardiac hypertrophy and dysfunction, which correlated with increased protein kinase D-1 (PKD1) phosphorylation and increased glucose uptake. These changes in glucose utilization preceded a reduction in cardiac energy status. At baseline, compared with non-diabetic mice, diabetic mice showed normal cardiac function, higher lipid content and mitochondrial capacity for fatty acid oxidation, and lower PKD1 phosphorylation, glucose uptake, and energetics. Interestingly, TAC affected cardiac function only mildly in diabetic mice, which was accompanied by normalization of phosphorylated PKD1, glucose uptake, and cardiac energy status. CONCLUSION: The cardiac metabolic adaptations in diabetic mice seem to prevent the heart from failing upon pressure overload, suggesting that restoring the balance between glucose and fatty acid utilization is beneficial for cardiac function.


Asunto(s)
Aorta/cirugía , Glucemia/metabolismo , Complicaciones de la Diabetes/prevención & control , Diabetes Mellitus/sangre , Metabolismo Energético , Insuficiencia Cardíaca/prevención & control , Imagen por Resonancia Magnética , Miocardio/metabolismo , Tomografía de Emisión de Positrones , Espectroscopía de Protones por Resonancia Magnética , Adaptación Fisiológica , Animales , Aorta/fisiopatología , Presión Arterial , Constricción , Complicaciones de la Diabetes/diagnóstico por imagen , Complicaciones de la Diabetes/metabolismo , Complicaciones de la Diabetes/fisiopatología , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/fisiopatología , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Fluorodesoxiglucosa F18/administración & dosificación , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratones Endogámicos C57BL , Fosforilación , Valor Predictivo de las Pruebas , Proteína Quinasa C/metabolismo , Radiofármacos/administración & dosificación , Factores de Tiempo , Función Ventricular Izquierda , Remodelación Ventricular
4.
Cardiovasc Res ; 113(7): 737-748, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28419197

RESUMEN

AIMS: The type 2 diabetic heart oxidizes more fat and less glucose, which can impair metabolic flexibility and function. Increased sarcolemmal fatty acid translocase (FAT/CD36) imports more fatty acid into the diabetic myocardium, feeding increased fatty acid oxidation and elevated lipid deposition. Unlike other metabolic modulators that target mitochondrial fatty acid oxidation, we proposed that pharmacologically inhibiting fatty acid uptake, as the primary step in the pathway, would provide an alternative mechanism to rebalance metabolism and prevent lipid accumulation following hypoxic stress. METHODS AND RESULTS: Hearts from type 2 diabetic and control male Wistar rats were perfused in normoxia, hypoxia and reoxygenation, with the FAT/CD36 inhibitor sulfo-N-succinimidyl oleate (SSO) infused 4 min before hypoxia. SSO infusion into diabetic hearts decreased the fatty acid oxidation rate by 29% and myocardial triglyceride concentration by 48% compared with untreated diabetic hearts, restoring fatty acid metabolism to control levels following hypoxia-reoxygenation. SSO infusion increased the glycolytic rate by 46% in diabetic hearts during hypoxia, increased pyruvate dehydrogenase activity by 53% and decreased lactate efflux rate by 56% compared with untreated diabetic hearts during reoxygenation. In addition, SSO treatment of diabetic hearts increased intermediates within the second span of the Krebs cycle, namely fumarate, oxaloacetate, and the FAD total pool. The cardiac dysfunction in diabetic hearts following decreased oxygen availability was prevented by SSO-infusion prior to the hypoxic stress. Infusing SSO into diabetic hearts increased rate pressure product by 60% during hypoxia and by 32% following reoxygenation, restoring function to control levels. CONCLUSIONS: Diabetic hearts have limited metabolic flexibility and cardiac dysfunction when stressed, which can be rapidly rectified by reducing fatty acid uptake with the FAT/CD36 inhibitor, SSO. This novel therapeutic approach not only reduces fat oxidation but also lipotoxicity, by targeting the primary step in the fatty acid metabolism pathway.


Asunto(s)
Antígenos CD36/antagonistas & inhibidores , Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocardio/metabolismo , Ácidos Oléicos/farmacología , Sarcolema/efectos de los fármacos , Succinimidas/farmacología , Animales , Antígenos CD36/metabolismo , Hipoxia de la Célula , Ciclo del Ácido Cítrico/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Ácidos Grasos/metabolismo , Preparación de Corazón Aislado , Masculino , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Sarcolema/metabolismo , Factores de Tiempo , Triglicéridos/metabolismo , Función Ventricular Izquierda/efectos de los fármacos , Presión Ventricular/efectos de los fármacos
5.
J Biol Chem ; 292(17): 7105-7114, 2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28320859

RESUMEN

Several studies have linked impaired glucose uptake and insulin resistance (IR) to functional impairment of the heart. Recently, endocannabinoids have been implicated in cardiovascular disease. However, the mechanisms involving endocannabinoid signaling, glucose uptake, and IR in cardiomyocytes are understudied. Here we report that the endocannabinoid 2-arachidonoylglycerol (2-AG), via stimulation of cannabinoid type 1 (CB1) receptor and Ca2+/calmodulin-dependent protein kinase ß, activates AMP-activated kinase (AMPK), leading to increased glucose uptake. Interestingly, we have observed that the mRNA expression of CB1 and CB2 receptors was decreased in diabetic mice, indicating reduced endocannabinoid signaling in the diabetic heart. We further establish that TNFα induces IR in cardiomyocytes. Treatment with 2-AG suppresses TNFα-induced proinflammatory markers and improves IR and glucose uptake. Conversely, pharmacological inhibition or knockdown of AMPK attenuates the anti-inflammatory effect and reversal of IR elicited by 2-AG. Additionally, in human embryonic stem cell-derived cardiomyocytes challenged with TNFα or FFA, we demonstrate that 2-AG improves insulin sensitivity and glucose uptake. In conclusion, 2-AG abates inflammatory responses, increases glucose uptake, and overcomes IR in an AMPK-dependent manner in cardiomyocytes.


Asunto(s)
Ácidos Araquidónicos/química , Endocannabinoides/química , Glicéridos/química , Resistencia a la Insulina , Miocitos Cardíacos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Antiinflamatorios/química , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Diferenciación Celular , Diabetes Mellitus Experimental/metabolismo , Células Madre Embrionarias/citología , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Glucógeno/metabolismo , Humanos , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/citología , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Lew , Receptor Cannabinoide CB1/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
6.
Diabetes ; 66(6): 1521-1534, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28302654

RESUMEN

Dietary fat overconsumption leads to myocardial lipid accumulation through mechanisms that are incompletely resolved. Previously, we identified increased translocation of the fatty acid transporter CD36 from its endosomal storage compartment to the sarcolemma as the primary mechanism of excessive myocellular lipid import. Here, we show that increased CD36 translocation is caused by alkalinization of endosomes resulting from inhibition of proton pumping activity of vacuolar-type H+-ATPase (v-ATPase). Endosomal alkalinization was observed in hearts from rats fed a lard-based high-fat diet and in rodent and human cardiomyocytes upon palmitate overexposure, and appeared as an early lipid-induced event preceding the onset of insulin resistance. Either genetic or pharmacological inhibition of v-ATPase in cardiomyocytes exposed to low palmitate concentrations reduced insulin sensitivity and cardiomyocyte contractility, which was rescued by CD36 silencing. The mechanism of palmitate-induced v-ATPase inhibition involved its dissociation into two parts: the cytosolic V1 and the integral membrane V0 subcomplex. Interestingly, oleate also inhibits v-ATPase function, yielding triacylglycerol accumulation but not insulin resistance. In conclusion, lipid oversupply increases CD36-mediated lipid uptake that directly impairs v-ATPase function. This feeds forward to enhanced CD36 translocation and further increased lipid uptake. In the case of palmitate, its accelerated uptake ultimately precipitates into cardiac insulin resistance and contractile dysfunction.


Asunto(s)
Antígenos CD36/metabolismo , Endosomas/efectos de los fármacos , Glucosa/metabolismo , Corazón/efectos de los fármacos , Resistencia a la Insulina , Contracción Miocárdica/efectos de los fármacos , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Palmitatos/farmacología , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores , Animales , Western Blotting , Radioisótopos de Carbono , Células Cultivadas , Desoxiglucosa/metabolismo , Dieta Alta en Grasa , Endosomas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células Madre Pluripotentes Inducidas , Masculino , Miocitos Cardíacos/metabolismo , Cadenas Pesadas de Miosina/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Lew , Triglicéridos/metabolismo , Tritio , Troponina T/genética
7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(5): 541-551, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28214558

RESUMEN

Small heterodimer partner (SHP) is an atypical nuclear receptor expressed in heart that has been shown to inhibit the hypertrophic response. Here, we assessed the role of SHP in cardiac metabolism and inflammation. Mice fed a high-fat diet (HFD) displayed glucose intolerance accompanied by increased cardiac mRNA levels of Shp. In HL-1 cardiomyocytes, SHP overexpression inhibited both basal and insulin-stimulated glucose uptake and impaired the insulin signalling pathway (evidenced by reduced AKT and AS160 phosphorylation), similar to insulin resistant cells generated by high palmitate/high insulin treatment (HP/HI; 500µM/100nM). In addition, SHP overexpression increased Socs3 mRNA and reduced IRS-1 protein levels. SHP overexpression also induced Cd36 expression (~6.2 fold; p<0.001) linking to the observed intramyocellular lipid accumulation. SHP overexpressing cells further showed altered expression of genes involved in lipid metabolism, i.e., Acaca, Acadvl or Ucp3, augmented NF-κB DNA-binding activity and induced transcripts of inflammatory genes, i.e., Il6 and Tnf mRNA (~4-fold induction, p<0.01). Alterations in metabolism and inflammation found in SHP overexpressing cells were associated with changes in the mRNA levels of Ppara (79% reduction, p<0.001) and Pparg (~58-fold induction, p<0.001). Finally, co-immunoprecipitation studies showed that SHP overexpression strongly reduced the physical interaction between PPARα and the p65 subunit of NF-κB, suggesting that dissociation of these two proteins is one of the mechanisms by which SHP initiates the inflammatory response in cardiac cells. Overall, our results suggest that SHP upregulation upon high-fat feeding leads to lipid accumulation, insulin resistance and inflammation in cardiomyocytes.


Asunto(s)
Inflamación/metabolismo , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/genética , Miocardio/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Animales , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Inflamación/patología , Insulina/metabolismo , Ratones , Miocardio/patología , Miocitos Cardíacos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal
8.
Biochem J ; 473(7): 937-47, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26831516

RESUMEN

AMP-activated protein kinase (AMPK) is a metabolic stress-sensing kinase. We previously showed that glucose deprivation induces autophosphorylation of AMPKß at Thr-148, which prevents the binding of AMPK to glycogen. Furthermore, in MIN6 cells, AMPKß1 binds to R6 (PPP1R3D), a glycogen-targeting subunit of protein phosphatase type 1 (PP1), thereby regulating the glucose-induced inactivation of AMPK. In the present study, we further investigated the interaction of R6 with AMPKß and the possible dependency on Thr-148 phosphorylation status. Yeast two-hybrid (Y2H) analyses and co-immunoprecipitation (IP) of the overexpressed proteins in human embryonic kidney (HEK) 293T) cells revealed that both AMPKß1 and AMPK-ß2 wild-type (WT) isoforms bind to R6. The AMPKß-R6 interaction was stronger with the muscle-specific AMPKß2-WT and required association with the substrate-binding motif of R6. When HEK293T cells or C2C12 myotubes were cultured in high-glucose medium, AMPKß2-WT and R6 weakly interacted. In contrast, glycogen depletion significantly enhanced this protein interaction. Mutation of AMPKß2 Thr-148 prevented the interaction with R6 irrespective of the intracellular glycogen content. Treatment with the AMPK activator oligomycin enhanced the AMPKß2-R6 interaction in conjunction with increased Thr-148 phosphorylation in cells grown in low-glucose medium. These data are in accordance with R6 binding directly to AMPKß2 when both proteins detach from the diminishing glycogen particle, which is simultaneous with increased AMPKß2 Thr-148 autophosphorylation. Such a model points to a possible control of AMPK by PP1-R6 upon glycogen depletion in muscle.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucógeno/metabolismo , Mutación Missense , Proteína Fosfatasa 1/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Sustitución de Aminoácidos , Glucógeno/genética , Células HEK293 , Humanos , Fosforilación , Unión Proteica , Proteína Fosfatasa 1/genética
9.
Am J Physiol Cell Physiol ; 308(4): C297-307, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25472960

RESUMEN

Insulin resistance is an important risk factor for the development of several cardiac pathologies, thus advocating strategies for restoring insulin sensitivity of the heart in these conditions. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs), mainly eicosapentaenoic acid (EPA, C20:5n-3) and docosahexaenoic acid (DHA, C22:6n-3), have been shown to improve insulin sensitivity in insulin-sensitive tissues, but their direct effect on insulin signaling and metabolic parameters in the myocardium has not been reported previously. The aim of this study was therefore to examine the ability of EPA and DHA to prevent insulin resistance in isolated rat cardiomyocytes. Primary rat cardiomyocytes were made insulin resistant by 48 h incubation in high insulin (HI) medium. Parallel incubations were supplemented by 200 µM EPA or DHA. Addition of EPA or DHA to the medium prevented the induction of insulin resistance in cardiomyocytes by preserving the phosphorylation state of key proteins in the insulin signaling cascade and by preventing persistent relocation of fatty acid transporter CD36 to the sarcolemma. Only cardiomyocytes incubated in the presence of EPA, however, exhibited improvements in glucose and fatty acid uptake and cell shortening. We conclude that ω-3 PUFAs protect metabolic and functional properties of cardiomyocytes subjected to insulin resistance-evoking conditions.


Asunto(s)
Cardiotónicos/farmacología , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Metabolismo Energético/efectos de los fármacos , Resistencia a la Insulina , Insulina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Animales , Antígenos CD36/metabolismo , Células Cultivadas , Ácidos Docosahexaenoicos/metabolismo , Ácido Eicosapentaenoico/metabolismo , Glucosa/metabolismo , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación , Transporte de Proteínas , Ratas Endogámicas Lew , Sarcolema/efectos de los fármacos , Sarcolema/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
10.
J Mol Cell Cardiol ; 76: 208-17, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25173922

RESUMEN

In the insulin resistant heart, energy fuel selection shifts away from glucose utilization towards almost complete dependence on long-chain fatty acids (LCFA). This shift results in excessive cardiac lipid accumulation and eventually heart failure. Lipid-induced cardiomyopathy may be averted by strategies that increase glucose uptake without elevating LCFA uptake. Protein kinase-D1 (PKD1) is involved in contraction-induced glucose, but not LCFA, uptake allowing to hypothesize that this kinase is an attractive target to treat lipid-induced cardiomyopathy. For this, cardiospecific constitutively active PKD1 overexpression (caPKD1)-mice were subjected to an insulin resistance-inducing high fat-diet for 20-weeks. Substrate utilization was assessed by microPET and cardiac function by echocardiography. Cardiomyocytes were isolated for measurement of substrate uptake, lipid accumulation and insulin sensitivity. Wild-type mice on a high fat-diet displayed increased basal myocellular LCFA uptake, increased lipid deposition, greatly impaired insulin signaling, and loss of insulin-stimulated glucose and LCFA uptake, which was associated with concentric hypertrophic remodeling. The caPKD1 mice on high-fat diet showed none of these characteristics, whereas on low-fat diet a shift towards cardiac glucose utilization in combination with hypertrophy and ventricular dilation was observed. In conclusion, these data suggest that PKD pathway activation may be an attractive therapeutic strategy to mitigate lipid accumulation, insulin resistance and maladaptive remodeling in the lipid-overloaded heart, but this requires further investigation.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Resistencia a la Insulina , Proteína Quinasa C/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Expresión Génica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Histona Desacetilasas/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/enzimología , Miocardio/patología , Miocitos Cardíacos/enzimología , Fosforilación , Proteína Quinasa C/genética , Procesamiento Proteico-Postraduccional
11.
Am J Physiol Endocrinol Metab ; 307(2): E225-36, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24895286

RESUMEN

Activation of AMP-activated protein kinase (AMPK) in cardiomyocytes induces translocation of glucose transporter GLUT4 and long-chain fatty acid (LCFA) transporter CD36 from endosomal stores to the sarcolemma to enhance glucose and LCFA uptake, respectively. Ca(2+)/calmodulin-activated kinase kinase-ß (CaMKKß) has been positioned directly upstream of AMPK. However, it is unknown whether acute increases in [Ca(2+)]i stimulate translocation of GLUT4 and CD36 and uptake of glucose and LCFA or whether Ca(2+) signaling converges with AMPK signaling to exert these actions. Therefore, we studied the interplay between Ca(2+) and AMPK signaling in regulation of cardiomyocyte substrate uptake. Exposure of primary cardiomyocytes to inhibitors or activators of Ca(2+) signaling affected neither AMPK-Thr(172) phosphorylation nor basal and AMPK-mediated glucose and LCFA uptake. Despite their lack of an effect on substrate uptake, Ca(2+) signaling activators induced GLUT4 and CD36 translocation. In contrast, AMPK activators stimulated GLUT4/CD36 translocation as well as glucose/LCFA uptake. When cardiomyocytes were cotreated with Ca(2+) signaling and AMPK activators, Ca(2+) signaling activators further enhanced AMPK-induced glucose/LCFA uptake. In conclusion, Ca(2+) signaling shows no involvement in AMPK-induced GLUT4/CD36 translocation and substrate uptake but elicits transporter translocation via a separate pathway requiring CaMKKß/CaMKs. Ca(2+)-induced transporter translocation by itself appears to be ineffective to increase substrate uptake but requires additional AMPK activation to effectuate transporter translocation into increased substrate uptake. Ca(2+)-induced transporter translocation might be crucial under excessive cardiac stress conditions that require supraphysiological energy demands. Alternatively, Ca(2+) signaling might prepare the heart for substrate uptake during physiological contraction by inducing transporter translocation.


Asunto(s)
Antígenos CD36/metabolismo , Señalización del Calcio/fisiología , Ácidos Grasos/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Glucosa/metabolismo , Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Animales , Calcimicina/farmacología , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Miocitos Cardíacos/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Endogámicas Lew , Sarcolema/efectos de los fármacos , Tapsigargina/farmacología
12.
Front Physiol ; 3: 361, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22973240

RESUMEN

AIM: The signaling pathways involved in the regulation of cardiac GLUT4 translocation/glucose uptake and CD36 translocation/long-chain fatty acid uptake are not fully understood. We compared in heart/muscle-specific PKC-λ knockout mice the roles of atypical PKCs (PKC-ζ and PKC-λ) in regulating cardiac glucose and fatty acid uptake. RESULTS: Neither insulin-stimulated nor AMPK-mediated glucose and fatty acid uptake were inhibited upon genetic PKC-λ ablation in cardiomyocytes. In contrast, myristoylated PKC-ζ pseudosubstrate inhibited both insulin-stimulated and AMPK-mediated glucose and fatty acid uptake by >80% in both wild-type and PKC-λ-knockout cardiomyocytes. In PKC-λ knockout cardiomyocytes, PKC-ζ is the sole remaining atypical PKC isoform, and its expression level is not different from wild-type cardiomyocytes, in which it contributes to 29% and 17% of total atypical PKC expression and phosphorylation, respectively. CONCLUSION: Taken together, atypical PKCs are necessary for insulin-stimulated and AMPK-mediated glucose uptake into the heart, as well as for insulin-stimulated and AMPK-mediated fatty acid uptake. However, the residual PKC-ζ activity in PKC-λ-knockout cardiomyocytes is sufficient to allow optimal stimulation of glucose and fatty acid uptake, indicating that atypical PKCs are necessary but not rate-limiting in the regulation of cardiac substrate uptake and that PKC-λ and PKC-ζ have interchangeable functions in these processes.

13.
J Biol Chem ; 287(44): 37530-9, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22936810

RESUMEN

Cardiac glucose utilization is regulated by reversible translocation of the glucose transporter GLUT4 from intracellular stores to the plasma membrane. During the onset of diet-induced insulin resistance, elevated lipid levels in the circulation interfere with insulin-stimulated GLUT4 translocation, leading to impaired glucose utilization. Recently, we identified vesicle-associated membrane protein (VAMP) 2 and 3 to be required for insulin- and contraction-stimulated GLUT4 translocation, respectively, in cardiomyocytes. Here, we investigated whether overexpression of VAMP2 and/or VAMP3 could protect insulin-stimulated GLUT4 translocation under conditions of insulin resistance. HL-1 atrial cardiomyocytes transiently overexpressing either VAMP2 or VAMP3 were cultured for 16 h with elevated concentrations of palmitate and insulin. Upon subsequent acute stimulation with insulin, we measured GLUT4 translocation, plasmalemmal presence of the fatty acid transporter CD36, and myocellular lipid accumulation. Overexpression of VAMP3, but not VAMP2, completely prevented lipid-induced inhibition of insulin-stimulated GLUT4 translocation. Furthermore, the plasmalemmal presence of CD36 and intracellular lipid levels remained normal in cells overexpressing VAMP3. However, insulin signaling was not retained, indicating an effect of VAMP3 overexpression downstream of PKB/Akt. Furthermore, we revealed that endogenous VAMP3 is bound by the contraction-activated protein kinase D (PKD), and contraction and VAMP3 overexpression protect insulin-stimulated GLUT4 translocation via a common mechanism. These observations indicate that PKD activates GLUT4 translocation via a VAMP3-dependent trafficking step, which pathway might be valuable to rescue constrained glucose utilization in the insulin-resistant heart.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Resistencia a la Insulina , Miocitos Cardíacos/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/metabolismo , Animales , Antígenos CD36/metabolismo , Línea Celular , Grasas de la Dieta/farmacología , Expresión Génica , Cardiopatías/metabolismo , Cardiopatías/patología , Insulina/farmacología , Insulina/fisiología , Metabolismo de los Lípidos , Masculino , Ratones , Contracción Miocárdica , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Palmitatos/farmacología , Proteína Quinasa C/metabolismo , Transporte de Proteínas , Ratas , Ratas Endogámicas Lew , Transducción de Señal , Proteína 2 de Membrana Asociada a Vesículas/genética , Proteína 3 de Membrana Asociada a Vesículas/genética
14.
Biochem J ; 448(1): 43-53, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22780108

RESUMEN

An increased cardiac fatty acid supply and increased sarcolemmal presence of the long-chain fatty acid transporter CD36 are associated with and contribute to impaired cardiac insulin sensitivity and function. In the present study we aimed at preventing the development of insulin resistance and contractile dysfunction in cardiomyocytes by blocking CD36-mediated palmitate uptake. Insulin resistance and contractile dysfunction were induced in primary cardiomyocytes by 48 h incubation in media containing either 100 nM insulin (high insulin; HI) or 200 µM palmitate (high palmitate; HP). Under both culture conditions, insulin-stimulated glucose uptake and Akt phosphorylation were abrogated or markedly reduced. Furthermore, cardiomyocytes cultured in each medium displayed elevated sarcolemmal CD36 content, increased basal palmitate uptake, lipid accumulation and decreased sarcomere shortening. Immunochemical CD36 inhibition enhanced basal glucose uptake and prevented elevated basal palmitate uptake, triacylglycerol accumulation and contractile dysfunction in cardiomyocytes cultured in either medium. Additionally, CD36 inhibition prevented loss of insulin signalling in cells cultured in HP, but not in HI medium. In conclusion, CD36 inhibition prevents lipid accumulation and lipid-induced contractile dysfunction in cardiomyocytes, but probably independently of effects on insulin signalling. Nonetheless, pharmacological CD36 inhibition may be considered as a treatment strategy to counteract impaired functioning of the lipid-loaded heart.


Asunto(s)
Antígenos CD36/fisiología , Resistencia a la Insulina/fisiología , Miocitos Cardíacos/metabolismo , Palmitatos/metabolismo , Animales , Transporte Biológico , Señalización del Calcio/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/prevención & control , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Insulina/farmacología , Masculino , Mitocondrias Cardíacas/metabolismo , Contracción Miocárdica , Miocitos Cardíacos/efectos de los fármacos , Palmitatos/farmacología , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Endogámicas Lew , Sarcolema/metabolismo , Sarcómeros/ultraestructura , Transducción de Señal/efectos de los fármacos , Triglicéridos/metabolismo
15.
J Biol Chem ; 287(8): 5871-81, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22158620

RESUMEN

Increased contraction enhances substrate uptake into cardiomyocytes via translocation of the glucose transporter GLUT4 and the long chain fatty acid (LCFA) transporter CD36 from intracellular stores to the sarcolemma. Additionally, contraction activates the signaling enzymes AMP-activated protein kinase (AMPK) and protein kinase D1 (PKD1). Although AMPK has been implicated in contraction-induced GLUT4 and CD36 translocation in cardiomyocytes, the precise role of PKD1 in these processes is not known. To study this, we triggered contractions in cardiomyocytes by electric field stimulation (EFS). First, the role of PKD1 in GLUT4 and CD36 translocation was defined. In PKD1 siRNA-treated cardiomyocytes as well as cardiomyocytes from PKD1 knock-out mice, EFS-induced translocation of GLUT4, but not CD36, was abolished. In AMPK siRNA-treated cardiomyocytes and cardiomyocytes from AMPKα2 knock-out mice, both GLUT4 and CD36 translocation were abrogated. Hence, unlike AMPK, PKD1 is selectively involved in glucose uptake. Second, we analyzed upstream factors in PKD1 activation. Cardiomyocyte contractions enhanced reactive oxygen species (ROS) production. Using ROS scavengers, we found that PKD1 signaling and glucose uptake are more sensitive to changes in intracellular ROS than AMPK signaling or LCFA uptake. Furthermore, silencing of death-activated protein kinase (DAPK) abrogated EFS-induced GLUT4 but not CD36 translocation. Finally, possible links between PKD1 and AMPK signaling were investigated. PKD1 silencing did not affect AMPK activation. Reciprocally, AMPK silencing did not alter PKD1 activation. In conclusion, we present a novel contraction-induced ROS-DAPK-PKD1 pathway in cardiomyocytes. This pathway is activated separately from AMPK and mediates GLUT4 translocation/glucose uptake, but not CD36 translocation/LCFA uptake.


Asunto(s)
Ácidos Grasos/metabolismo , Glucosa/metabolismo , Contracción Muscular , Miocitos Cardíacos/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Antígenos CD36/metabolismo , Línea Celular , Femenino , Técnicas de Silenciamiento del Gen , Transportador de Glucosa de Tipo 4/metabolismo , Masculino , Ratones , Miocitos Cardíacos/citología , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Transporte de Proteínas , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
16.
Am J Physiol Endocrinol Metab ; 301(4): E618-27, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21712535

RESUMEN

Cardiac patients often are obese and have hypertension, but in most studies these conditions are investigated separately. Here, we aimed at 1) elucidating the interaction of metabolic and mechanophysical stress in the development of cardiac dysfunction in mice and 2) preventing this interaction by ablation of the fatty acid transporter CD36. Male wild-type (WT) C57Bl/6 mice and CD36(-/-) mice received chow or Western-type diet (WTD) for 10 wk and then underwent a sham surgery or transverse aortic constriction (TAC) under anesthesia. After a 6-wk continuation of the diet, cardiac function, morphology, lipid profiles, and molecular parameters were assessed. WTD administration affected body and organ weights of WT and CD36(-/-) mice, but it affected only plasma glucose and insulin concentrations in WT mice. Cardiac lipid concentrations increased in WT mice receiving WTD, decreased in CD36(-/-) on chow, and remained unchanged in CD36(-/-) receiving WTD. TAC induced cardiac hypertrophy in WT mice on chow but did not affect cardiac function and cardiac lipid concentrations. WTD or CD36 ablation worsened the outcome of TAC. Ablation of CD36 protected against the WTD-related aggravation of cardiac functional and structural changes induced by TAC. In conclusion, cardiac dysfunction and remodeling worsen when the heart is exposed to two stresses, metabolic and mechanophysical, at the same time. CD36 ablation prevents the metabolic stress resulting from a WTD. Thus, metabolic conditions are a critical factor for the compromised heart and provide new targets for metabolic manipulation in cardioprotection.


Asunto(s)
Antígenos CD36/genética , Cardiomegalia/metabolismo , Miocardio/metabolismo , Animales , Estenosis de la Válvula Aórtica/complicaciones , Glucemia/metabolismo , Antígenos CD36/metabolismo , Cardiomegalia/etiología , Cardiomegalia/fisiopatología , Dieta , Corazón/fisiopatología , Masculino , Ratones , Ratones Noqueados , Miocardio/patología , Obesidad/complicaciones
17.
Am J Physiol Cell Physiol ; 298(6): C1549-59, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20375272

RESUMEN

Insulin and contraction stimulate both cardiac glucose and long-chain fatty acid (LCFA) uptake via translocation of the substrate transporters GLUT4 and CD36, respectively, from intracellular compartments to the sarcolemma. Little is known about the role of vesicular trafficking elements in insulin- and contraction-stimulated glucose and LCFA uptake in the heart, especially whether certain trafficking elements are specifically involved in GLUT4 versus CD36 translocation. Therefore, we studied the role of coat proteins, actin- and microtubule-filaments and endosomal pH on glucose and LCFA uptake into primary cardiomyocytes under basal conditions and during stimulation with insulin or oligomycin (contraction-like AMP-activated protein kinase activator). Inhibition of coat protein targeting to Golgi/endosomes decreased insulin/oligomycin-stimulated glucose (-42%/-51%) and LCFA (-39%/-68%) uptake. Actin disruption decreased insulin/oligomycin-stimulated glucose uptake (-41%/-75%), while not affecting LCFA uptake. Microtubule disruption did not affect substrate uptake under any condition. Endosomal alkalinization increased basal sarcolemmal CD36 (2-fold), but not GLUT4, content, and concomitantly decreased basal intracellular membrane GLUT4 and CD36 content (-60% and -62%, respectively), indicating successful CD36 translocation and incomplete GLUT4 translocation. Additionally, endosomal alkalinization elevated basal LCFA uptake (1.4-fold) in a nonadditive manner to insulin/oligomycin, and decreased insulin/oligomycin-stimulated glucose uptake (-32%/-68%). In conclusion, 1) CD36 translocation, just like GLUT4 translocation, is a vesicle-mediated process depending on coat proteins, and 2) GLUT4 and CD36 trafficking are differentially dependent on endosomal pH and actin filaments. The latter conclusion suggests novel strategies to alter cardiac substrate preference as part of metabolic modulation therapy.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Antígenos CD36/metabolismo , Desoxiglucosa/metabolismo , Endosomas/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Miocitos Cardíacos/metabolismo , Ácido Palmítico/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/ultraestructura , Animales , Transporte Biológico , Brefeldino A/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteína Coatómero/metabolismo , Colchicina/farmacología , Endosomas/efectos de los fármacos , Endosomas/ultraestructura , Activación Enzimática , Activadores de Enzimas/farmacología , Concentración de Iones de Hidrógeno , Insulina/metabolismo , Masculino , Contracción Miocárdica , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/ultraestructura , Oligomicinas/farmacología , Transporte de Proteínas , Ratas , Ratas Endogámicas Lew , Tiazolidinas/farmacología , Moduladores de Tubulina/farmacología
18.
Endocrinology ; 151(1): 422-31, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19940039

RESUMEN

Thyroid hormones can modify cardiac metabolism via multiple molecular mechanisms, yet their integrated effect on overall substrate metabolism is poorly understood. Here we determined the effect of hyperthyroidism on substrate metabolism in the isolated, perfused, contracting rat heart. Male Wistar rats were injected for 7 d with T(3) (0.2 mg/kg x d ip). Plasma free fatty acids increased by 97%, heart weights increased by 33%, and cardiac rate pressure product, an indicator of contractile function, increased by 33% in hyperthyroid rats. Insulin-stimulated glycolytic rates and lactate efflux rates were increased by 33% in hyperthyroid rat hearts, mediated by an increased insulin-stimulated translocation of the glucose transporter GLUT4 to the sarcolemma. This was accompanied by a 70% increase in phosphorylated AMP-activated protein kinase (AMPK) and a 100% increase in phosphorylated acetyl CoA carboxylase, confirming downstream signaling from AMPK. Fatty acid oxidation rates increased in direct proportion to the increased heart weight and rate pressure product in the hyperthyroid heart, mediated by synchronized changes in mitochondrial enzymes and respiration. Protein levels of the fatty acid transporter, fatty acid translocase (FAT/CD36), were reduced by 24% but were accompanied by a 19% increase in the sarcolemmal content of fatty acid transport protein 1 (FATP1). Thus, the relationship between fatty acid metabolism, cardiac mass, and contractile function was maintained in the hyperthyroid heart, associated with a sarcolemmal reorganization of fatty acid transporters. The combined effects of T(3)-induced AMPK activation and insulin stimulation were associated with increased sarcolemmal GLUT4 localization and glycolytic flux in the hyperthyroid heart.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hipertiroidismo/metabolismo , Contracción Miocárdica/fisiología , Animales , Activación Enzimática/fisiología , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Corazón/fisiología , Hipertiroidismo/fisiopatología , Insulina/metabolismo , Metabolismo de los Lípidos/fisiología , Masculino , Miocardio/metabolismo , Oxidación-Reducción , Transporte de Proteínas , Ratas , Ratas Wistar , Triyodotironina/metabolismo , Triyodotironina/fisiología
19.
J Endocrinol ; 201(2): 199-209, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19273501

RESUMEN

Insulin stimulates cardiac long-chain fatty acid (LCFA) and glucose uptake via translocation of human homolog of rat fatty acid translocase (CD36) and GLUT4 respectively, from intracellular membrane compartments to the sarcolemma, a process dependent on the activation of phosphatidylinositol-3 kinase. To identify downstream kinases of insulin signaling involved in translocation of CD36 and GLUT4 in the heart, we tested i) which cardiac protein kinase C (PKC) isoforms (alpha, delta, epsilon or zeta) are activated by insulin, and ii) whether PKC isoform-specific inhibition affects insulin-stimulated substrate uptake in the heart. Insulin-stimulated LCFA and glucose uptake were completely blunted by inhibition of PKC-zeta, but not by inhibition of conventional or novel PKCs. Concomitantly, translocation of CD36 and GLUT4 to the sarcolemma was completely blunted upon inhibition of PKC-zeta. However, insulin, in contrast to the diacylglycerol-analog phorbol-12-myristate-13-acetate (PMA), did not induce membrane-attachment of the conventional and novel PKCs-alpha, -delta, and -epsilon. PKC-zeta was already entirely membrane-bound in non-stimulated cells, and neither insulin nor PMA treatment had any effect on the subcellular localization of PKC-zeta. Furthermore, insulin treatment did not change phosphorylation of PKC-alpha, -delta, and -zeta or enzymatic activity of PKC-zeta towards a PKC-zeta substrate peptide. It is concluded that PKC-zeta, but not any other PKC isoform, is necessary for insulin-induced translocation of GLUT4 and CD36. However, PKC-zeta is already fully active under basal conditions and not further activated by insulin, indicating that its role in insulin-stimulated uptake of both glucose and LCFA is permissive rather than regulatory.


Asunto(s)
Antígenos CD36/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Proteína Quinasa C/fisiología , Animales , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Ácidos Grasos/metabolismo , Masculino , Modelos Biológicos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miocitos Cardíacos/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Endogámicas Lew , Acetato de Tetradecanoilforbol/farmacología
20.
Biochem J ; 419(2): 447-55, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19138173

RESUMEN

Although CPT-I (carnitine palmitoyltransferase-I) is generally regarded to present a major rate-controlling site in mitochondrial beta-oxidation, it is incompletely understood whether CPT-I is rate-limiting in the overall LCFA (long-chain fatty acid) flux in the heart. Another important site of regulation of the LCFA flux in the heart is trans-sarcolemmal LCFA transport facilitated by CD36 and FABPpm (plasma membrane fatty acid-binding protein). Therefore, we explored to what extent a chronic pharmacological blockade of the LCFA flux at the level of mitochondrial entry of LCFA-CoA would affect sarcolemmal LCFA uptake. Rats were injected daily with saline or etomoxir, a specific CPT-I inhibitor, for 8 days at 20 mg/kg of body mass. Etomoxir-treated rats displayed a 44% reduced cardiac CPT-I activity. Sarcolemmal contents of CD36 and FABPpm, as well as the LCFA transport capacity, were not altered in the hearts of etomoxir-treated versus control rats. Furthermore, rates of LCFA uptake and oxidation, and glucose uptake by cardiac myocytes from etomoxir-treated rats were not different from control rats, neither under basal nor under acutely induced maximal metabolic demands. Finally, hearts from etomoxir-treated rats did not display triacylglycerol accumulation. Therefore CPT-I appears not to present a major rate-controlling site in total cardiac LCFA flux. It is likely that sarcolemmal LCFA entry rather than mitochondrial LCFA-CoA entry is a promising target for normalizing LCFA flux in cardiac metabolic diseases.


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Compuestos Epoxi/farmacología , Ácidos Grasos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Western Blotting , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Activación Enzimática/efectos de los fármacos , Masculino , Oxidación-Reducción/efectos de los fármacos , Ratas , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA