Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circ Heart Fail ; 15(3): e009195, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34743528

RESUMEN

BACKGROUND: Current heart failure therapies unload the failing heart without targeting the underlying problem of reduced cardiac contractility. Traditional inotropes (ie, calcitropes) stimulate contractility via energetically costly augmentation of calcium cycling and worsen patient survival. A new class of agents-myotropes-activates the sarcomere directly, independent of calcium. We hypothesize that a novel myotrope TA1 increases contractility without the deleterious myocardial energetic impact of a calcitrope dobutamine. METHODS: We determined the effect of TA1 in bovine cardiac myofibrils and human cardiac microtissues, ex vivo in mouse cardiac fibers and in vivo in anesthetized normal rats. Effects of increasing concentrations of TA1 or dobutamine on contractile function, phosphocreatine and ATP concentrations, and ATP production were assessed by 31P nuclear magnetic resonance spectroscopy on isolated perfused rat hearts. RESULTS: TA1 increased the rate of myosin ATPase activity in isolated bovine myofibrils and calcium sensitivity in intact mouse papillary fibers. Contractility increased dose dependently in human cardiac microtissues and in vivo in rats as assessed by echocardiography. In isolated rat hearts, TA1 and dobutamine similarly increased the rate-pressure product. Dobutamine increased both developed pressure and heart rate accompanied by decreased phosphocreatine-to-ATP ratio and decreased free energy of ATP hydrolysis (ΔG~ATP) and elevated left ventricular end diastolic pressure. In contrast, the TA1 increased developed pressure without any effect on heart rate, left ventricular end diastolic pressure, phosphocreatine/ATP ratio, or ΔG~ATP. CONCLUSIONS: Novel myotrope TA1 increased myocardial contractility by sensitizing the sarcomere to calcium without impairing diastolic function or depleting the cardiac energy reserve. Since energetic depletion negatively correlates with long-term survival, myotropes may represent a superior alternative to traditional inotropes in heart failure management.


Asunto(s)
Dobutamina , Insuficiencia Cardíaca , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Bovinos , Dobutamina/farmacología , Metabolismo Energético , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones , Contracción Miocárdica , Miocardio/metabolismo , Fosfocreatina/metabolismo , Ratas , Troponina/metabolismo
2.
EMBO J ; 39(13): e104073, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32432379

RESUMEN

Respirometry is the gold standard measurement of mitochondrial oxidative function, as it reflects the activity of the electron transport chain complexes working together. However, the requirement for freshly isolated mitochondria hinders the feasibility of respirometry in multi-site clinical studies and retrospective studies. Here, we describe a novel respirometry approach suited for frozen samples by restoring electron transfer components lost during freeze/thaw and correcting for variable permeabilization of mitochondrial membranes. This approach preserves 90-95% of the maximal respiratory capacity in frozen samples and can be applied to isolated mitochondria, permeabilized cells, and tissue homogenates with high sensitivity. We find that primary changes in mitochondrial function, detected in fresh tissue, are preserved in frozen samples years after collection. This approach will enable analysis of the integrated function of mitochondrial Complexes I to IV in one measurement, collected at remote sites or retrospectively in samples residing in tissue biobanks.


Asunto(s)
Criopreservación , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Consumo de Oxígeno , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Masculino , Ratones
3.
Emerg Microbes Infect ; 8(1): 624-636, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30999821

RESUMEN

Flaviviruses are (re)-emerging RNA viruses strictly dependent on lipid metabolism for infection. In the search for host targeting antivirals, we explored the effect of pharmacological modulation of fatty acid metabolism during flavivirus infection. Considering the central role of acetyl-Coenzyme A carboxylase (ACC) on fatty acid metabolism, we analyzed the effect of three small-molecule ACC inhibitors (PF-05175157, PF-05206574, and PF-06256254) on the infection of medically relevant flaviviruses, namely West Nile virus (WNV), dengue virus, and Zika virus. Treatment with these compounds inhibited the multiplication of the three viruses in cultured cells. PF-05175157 induced a reduction of the viral load in serum and kidney in WNV-infected mice, unveiling its therapeutic potential for the treatment of chronic kidney disease associated with persistent WNV infection. This study constitutes a proof of concept of the reliability of ACC inhibitors to become viable antiviral candidates. These results support the repositioning of metabolic inhibitors as broad-spectrum antivirals.


Asunto(s)
Acetil-CoA Carboxilasa/antagonistas & inhibidores , Virus del Dengue/fisiología , Dengue/enzimología , Inhibidores Enzimáticos/administración & dosificación , Fiebre del Nilo Occidental/enzimología , Virus del Nilo Occidental/fisiología , Infección por el Virus Zika/enzimología , Virus Zika/fisiología , Acetil-CoA Carboxilasa/metabolismo , Animales , Antivirales/administración & dosificación , Dengue/tratamiento farmacológico , Dengue/virología , Virus del Dengue/efectos de los fármacos , Virus del Dengue/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Replicación Viral/efectos de los fármacos , Fiebre del Nilo Occidental/tratamiento farmacológico , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/genética , Virus Zika/efectos de los fármacos , Virus Zika/genética , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virología
4.
Sci Transl Med ; 8(326): 326rv3, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26888432

RESUMEN

Mitochondria-classically viewed as the powerhouses of the cell-have taken center stage in disease pathogenesis and resolution. Mitochondrial dysfunction, which originates from primary defects within the organelle or is induced by environmental stresses, plays a critical role in human disease. Despite their central role in human health and disease, there are no approved drugs that directly target mitochondria. We present possible new druggable targets in mitochondrial biology, including protein modification, calcium ion (Ca(2+)) transport, and dynamics, as we move into a new era of mitochondrial medicine.


Asunto(s)
Medicina , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Calcio/metabolismo , Humanos , Dinámicas Mitocondriales , Proteínas Mitocondriales/metabolismo
5.
Curr Vasc Pharmacol ; 14(1): 5-13, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463990

RESUMEN

Several cellular and molecular mechanisms have been implicated in the development of myocardial dysfunction and low cardiac output in pediatric patients undergoing heart surgery. Ischemia- reperfusion injury with alterations in calcium homeostasis as well as mitochondrial function has been strongly related to myocyte damage and heart failure in this population. In this article, we will review the main mechanisms of postoperative cardiac dysfunction at cellular and molecular levels and the subsequent protective strategies. In addition, we will describe cellular features of the neonatal or immature myocardium and will suggest possible protective management strategies. This article addresses the first of eight topics comprising the special issue entitled "Pharmacologic strategies with afterload reduction in low cardiac output syndrome after pediatric cardiac surgery".


Asunto(s)
Gasto Cardíaco Bajo/fisiopatología , Procedimientos Quirúrgicos Cardíacos/métodos , Daño por Reperfusión Miocárdica/complicaciones , Animales , Calcio/metabolismo , Gasto Cardíaco Bajo/etiología , Gasto Cardíaco Bajo/prevención & control , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Niño , Humanos , Recién Nacido , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/fisiopatología
6.
PLoS Comput Biol ; 11(6): e1004332, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26083688

RESUMEN

Development of heart diseases is driven by dynamic changes in both the activity and connectivity of gene pathways. Understanding these dynamic events is critical for understanding pathogenic mechanisms and development of effective treatment. Currently, there is a lack of computational methods that enable analysis of multiple gene networks, each of which exhibits differential activity compared to the network of the baseline/healthy condition. We describe the iMDM algorithm to identify both unique and shared gene modules across multiple differential co-expression networks, termed M-DMs (multiple differential modules). We applied iMDM to a time-course RNA-Seq dataset generated using a murine heart failure model generated on two genotypes. We showed that iMDM achieves higher accuracy in inferring gene modules compared to using single or multiple co-expression networks. We found that condition-specific M-DMs exhibit differential activities, mediate different biological processes, and are enriched for genes with known cardiovascular phenotypes. By analyzing M-DMs that are present in multiple conditions, we revealed dynamic changes in pathway activity and connectivity across heart failure conditions. We further showed that module dynamics were correlated with the dynamics of disease phenotypes during the development of heart failure. Thus, pathway dynamics is a powerful measure for understanding pathogenesis. iMDM provides a principled way to dissect the dynamics of gene pathways and its relationship to the dynamics of disease phenotype. With the exponential growth of omics data, our method can aid in generating systems-level insights into disease progression.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Redes Reguladoras de Genes/genética , Insuficiencia Cardíaca/genética , Animales , Perfilación de la Expresión Génica/métodos , Insuficiencia Cardíaca/metabolismo , Ratones , Ratones Transgénicos , Biología de Sistemas , Transcriptoma/genética
7.
Am J Physiol Heart Circ Physiol ; 307(9): H1307-16, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25172896

RESUMEN

Mitochondrial dysfunction in animal models of heart failure is associated with downregulation of the peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α pathway. To test whether PGC-1α is an appropriate therapeutic target for increasing mitochondrial biogenesis and improving function in heart failure, we used a transgenic (TG) mouse model of moderate overexpression of PGC-1α (∼3-fold) in the heart. TG mice had small increases in citrate synthase activity and mitochondria size in the heart without alterations in myocardial energetics or cardiac function at baseline. In vivo dobutamine stress increased fractional shortening in wild-type mice, but this increase was attenuated in TG mice, whereas ex vivo isolated perfused TG hearts demonstrated normal functional and energetic response to high workload challenge. When subjected to pressure overload by transverse aortic constriction (TAC), TG mice displayed a significantly greater acute mortality for both male and female mice; however, long-term survival up to 8 wk was similar between the two groups. TG mice also showed a greater decrease in fractional shortening and a greater increase in left ventricular chamber dimension in response to TAC. Mitochondrial gene expression and citrate synthase activity were mildly increased in TG mice compared with wild-type mice, and this difference was also maintained after TAC. Our data suggest that a moderate level of PGC-1α overexpression in the heart compromises acute survival and does not improve cardiac function during chronic pressure overload in mice.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Mitocondrias Cardíacas/metabolismo , Recambio Mitocondrial , Factores de Transcripción/metabolismo , Animales , Citrato (si)-Sintasa/genética , Citrato (si)-Sintasa/metabolismo , Femenino , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratones , Mitocondrias Cardíacas/ultraestructura , Contracción Miocárdica , Factores de Transcripción/genética , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
8.
J Am Heart Assoc ; 3(1): e000555, 2014 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-24470522

RESUMEN

BACKGROUND: The NADPH oxidase family (Nox) produces reactive oxygen species by adding the electron donated by NADPH to oxygen. Excessive reactive oxygen species production under a variety of pathological conditions has been attributed to increased Nox activity. Here, we aimed at investigating the role of Nox in cardiac ischemic injury through gain- and loss-of-function approaches. METHODS AND RESULTS: We modulated Nox activity in the heart by cardiac-specific expression of Nox4 and dominant negative Nox4. Modulation of Nox activity drastically changes the cellular redox status. Increasing Nox activity by cardiac-specific overexpression of Nox4 imposed oxidative stress on the myocardium [increased NAD(P)(+)/NAD(P)H and decreased glutathione/glutathione disulfide ratio] and worsened cardiac energetics and contractile function after ischemia-reperfusion. Overexpression of the dominant negative Nox4 (DN), which abolished the Nox function, led to a markedly reduced state [decreased NAD(P)(+)/NAD(P)H and increased glutathione/glutathione disulfide ratio] at baseline and paradoxically promoted mitochondrial reactive oxygen species production during ischemia resulting in no recovery of heart function after reperfusion. Limiting the generation of reducing equivalent through modulating carbon substrates availability partially restored the NAD(+)/NADH ratio and protected dominant negative Nox4 hearts from ischemic injury. CONCLUSIONS: This study reveals an important role of Nox in cardiac redox regulation and highlights the complexity of developing therapies that affect the intricately connected redox states.


Asunto(s)
Infarto del Miocardio/enzimología , Daño por Reperfusión Miocárdica/enzimología , Miocardio/metabolismo , NADPH Oxidasas/deficiencia , Estrés Oxidativo , Animales , Modelos Animales de Enfermedad , Metabolismo Energético , Glutatión/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/metabolismo , Contracción Miocárdica , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , NAD/metabolismo , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
9.
Cell Metab ; 18(2): 239-50, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23931755

RESUMEN

Mitochondrial respiratory dysfunction is linked to the pathogenesis of multiple diseases, including heart failure, but the specific mechanisms for this link remain largely elusive. We modeled the impairment of mitochondrial respiration by the inactivation of the Ndufs4 gene, a protein critical for complex I assembly, in the mouse heart (cKO). Although complex I-supported respiration decreased by >40%, the cKO mice maintained normal cardiac function in vivo and high-energy phosphate content in isolated perfused hearts. However, the cKO mice developed accelerated heart failure after pressure overload or repeated pregnancy. Decreased NAD(+)/NADH ratio by complex I deficiency inhibited Sirt3 activity, leading to an increase in protein acetylation and sensitization of the permeability transition in mitochondria (mPTP). NAD(+) precursor supplementation to cKO mice partially normalized the NAD(+)/NADH ratio, protein acetylation, and mPTP sensitivity. These findings describe a mechanism connecting mitochondrial dysfunction to the susceptibility to diseases and propose a potential therapeutic target.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Insuficiencia Cardíaca/metabolismo , Mitocondrias Cardíacas/metabolismo , Enfermedades Mitocondriales/metabolismo , NAD/metabolismo , Acetilación , Animales , Cardiotónicos/farmacología , Dobutamina/farmacología , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Estrés Oxidativo , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 305(3): H397-402, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23709599

RESUMEN

The C57BL/6 mouse strain is one of the most commonly used in experimental research. It is known to differ from other strains in baseline cardiovascular phenotypes as well as in response to pressure overload induced by aortic constriction. Since the generation of the C57BL/6 mouse line over a century ago, multiple substrains have been generated from the original. To identify potential substrain specific differences in response to pressure overload, we evaluated the effects of transverse aortic constriction (TAC) on survival, cardiac function, and expression of hypertrophic markers in three commonly used C57BL/6 substrains: C57BL/6J (JL), C57BL/6NCrl (CL), and C57BL/6NTac (TF). Survival and cardiac function were significantly lower in the CL and TF substrains compared with JL mice after TAC. Furthermore, the heart weight and lung weight as well as the expression of the hypertrophic marker Bnp were significantly greater in the CL mice compared with the JL. Histological assessment revealed marked left ventricular dilatation of CL and TF hearts while JL hearts showed increased wall thickness without dilatation. Our data demonstrate that cardiac response to pressure overload is distinct among the three commonly used C57BL/6 substrains of mice, which raises a cautionary note in study design and data interpretation.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/fisiopatología , Disfunción Ventricular Izquierda/fisiopatología , Adaptación Fisiológica , Animales , Aorta/fisiopatología , Aorta/cirugía , Biomarcadores/metabolismo , Miosinas Cardíacas/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Genotipo , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/metabolismo , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica , Miocardio/metabolismo , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Péptido Natriurético Encefálico/genética , Fenotipo , ARN Mensajero/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Especificidad de la Especie , Factores de Tiempo , Ultrasonografía , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/metabolismo , Función Ventricular Izquierda
11.
J Nutr Biochem ; 24(4): 656-63, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22819557

RESUMEN

The increasing incidence of insulin resistance has been linked to both increased intake of saturated fatty acids and disruption of the hypothalamic-pituitary-adrenal (HPA) axis. We tested the hypothesis that adding saturated fat/cholesterol to the diet of growing pigs would both disrupt HPA function and cause insulin resistance. Three-month-old pigs were fed either a control (13% energy from fat) or a high saturated fatty acid cholesterol (HSFC) diet (44% energy from fat; 2% cholesterol). After 10 weeks on the diets, intravenous ACTH, insulin and glucose challenges were performed, and after 12 weeks, tissue samples were taken for measurement of mRNA and for lipid-rich aortic lesions. Plasma total, HDL- and LDL-cholesterol were significantly increased in pigs fed the HSFC diet. Cortisol release during the ACTH challenge was suppressed in HSFC-fed pigs which were also more insulin resistant and glucose intolerant than controls. The HSFC diet decreased the expression of insulin receptor (IR) and insulin receptor substrate-1 in muscle and adipose tissue as well as adiponectin and adiponectin receptor 2 expression in fat. The HSFC diet decreased PGC-1α and PPARα expression in muscle but increased PPARα expression in liver. There was a trend for an increase in lipid-stained lesion frequency around the abdominal branches of the aorta in HSFC-fed pigs. We conclude that feeding increased saturated fat to pigs causes disruption in the HPA axis, insulin resistance and decreased muscle and adipose expression of genes controlling insulin signalling and mitochondrial oxidative capacity.


Asunto(s)
Glándulas Suprarrenales/fisiología , Colesterol en la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Sistema Hipotálamo-Hipofisario/fisiología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , PPAR alfa/metabolismo , Transactivadores/metabolismo , Tejido Adiposo/metabolismo , Animales , Aorta/patología , Proteínas Sustrato del Receptor de Insulina/genética , Hígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , PPAR alfa/genética , Porcinos , Transactivadores/genética
12.
J Vis Exp ; (67)2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22987105

RESUMEN

The mitochondrial permeability transition pore (mtPTP) is a non specific channel that forms in the inner mitochondrial membrane to transport solutes with a molecular mass smaller than 1.5 kDa. Although the definitive molecular identity of the pore is still under debate, proteins such as cyclophilin D, VDAC and ANT contribute to mtPTP formation. While the involvement of mtPTP opening in cell death is well established(1), accumulating evidence indicates that the mtPTP serves a physiologic role during mitochondrial Ca(2+) homeostasis(2), bioenergetics and redox signaling( 3). mtPTP opening is triggered by matrix Ca(2+) but its activity can be modulated by several other factors such as oxidative stress, adenine nucleotide depletion, high concentrations of Pi, mitochondrial membrane depolarization or uncoupling, and long chain fatty acids(4). In vitro, mtPTP opening can be achieved by increasing Ca(2+) concentration inside the mitochondrial matrix through exogenous additions of Ca(2+) (calcium retention capacity). When Ca(2+) levels inside mitochondria reach a certain threshold, the mtPTP opens and facilitates Ca(2+) release, dissipation of the proton motive force, membrane potential collapse and an increase in mitochondrial matrix volume (swelling) that ultimately leads to the rupture of the outer mitochondrial membrane and irreversible loss of organelle function. Here we describe a fluorometric assay that allows for a comprehensive characterization of mtPTP opening in isolated mouse heart mitochondria. The assay involves the simultaneous measurement of 3 mitochondrial parameters that are altered when mtPTP opening occurs: mitochondrial Ca(2+) handling (uptake and release, as measured by Ca(2+) concentration in the assay medium), mitochondrial membrane potential, and mitochondrial volume. The dyes employed for Ca(2+) measurement in the assay medium and mitochondrial membrane potential are Fura FF, a membrane impermeant, ratiometric indicator which undergoes a shift in the excitation wavelength in the presence of Ca(2+), and JC-1, a cationic, ratiometric indicator which forms green monomers or red aggregates at low and high membrane potential, respectively. Changes in mitochondrial volume are measured by recording light scattering by the mitochondrial suspension. Since high-quality, functional mitochondria are required for the mtPTP opening assay, we also describe the steps necessary to obtain intact, highly coupled and functional isolated heart mitochondria.


Asunto(s)
Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/química , Membranas Mitocondriales/metabolismo , Animales , Bencimidazoles/química , Calcio/análisis , Calcio/metabolismo , Carbocianinas/química , Colorantes Fluorescentes/química , Fluorometría/métodos , Ratones , Mitocondrias Cardíacas/química , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Membranas Mitocondriales/química , Poro de Transición de la Permeabilidad Mitocondrial
13.
J Mol Cell Cardiol ; 52(5): 1066-73, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22314372

RESUMEN

AMP-activated protein kinase (AMPK) is a master metabolic switch that plays an important role in energy homeostasis at the cellular and whole body level, hence a promising drug target. AMPK is a heterotrimeric complex composed of catalytic α-subunit and regulatory ß- and γ-subunits with multiple isoforms for each subunit. It has been shown that AMPK activity is increased in cardiac hypertrophy and failure but it is unknown whether changes in subunit composition of AMPK contribute to the altered AMPK activity. In this study, we determined the protein expression pattern of AMPK subunit isoforms during cardiac development as well as during cardiac hypertrophy and heart failure in mouse heart. We also compared the findings in failing mouse heart to that of the human failing hearts in order to determine whether the mouse heart is a good model of AMPK in human diseases. In mouse developmental hearts, AMPK was highly expressed in the fetal stages and fell back to the adult level after birth. In the failing mouse heart, there was a significant increase in α2, ß2, and γ2 subunits both at the mRNA and protein levels. In contrary, we found significant increases in the protein level of α1, ß1 and γ2c subunits in human failing hearts with no change in the mRNA level. We also compared isoform-specific AMPK activity in the mouse and human failing hearts. Consistent with the literature, in the failing mouse heart, the α2 complexes accounted for ~2/3 of total AMPK activity while the α1 complexes accounted for the remaining 30-35%. In the human hearts, however, the contribution of α1-AMPK activity was significantly higher (>40%) in the non-failing hearts, and it further increased to 50% in the failing hearts. Thus, the human hearts have a greater amount of α1-AMPK activity compared to the rodent hearts. In summary, the protein level and the isoform distribution of AMPK in the heart change significantly during normal development as well as in heart failure. These observations provide a basis for future development of therapeutic strategies for targeting AMPK.


Asunto(s)
Adenilato Quinasa/metabolismo , Expresión Génica , Insuficiencia Cardíaca/enzimología , Adenilato Quinasa/genética , Adulto , Anciano , Animales , Cardiomiopatía Dilatada/enzimología , Estudios de Casos y Controles , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Isquemia Miocárdica/enzimología , Miocardio/enzimología , Adulto Joven
14.
Circ Heart Fail ; 4(6): 707-13, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21840936

RESUMEN

BACKGROUND: The outcome of the surgical repair in congenital heart disease correlates with the degree of myocardial damage. In this study, we determined whether mitochondrial DNA depletion is a sensitive marker of right ventricular (RV) damage and whether impaired mitochondrial DNA (mtDNA) replication contributes to the transition from compensated hypertrophy to failure. METHODS AND RESULTS: RV samples obtained from 31 patients undergoing cardiac surgery were compared with 5 RV samples from nonfailing hearts (control). Patients were divided into compensated hypertrophy and failure groups, based on preoperative echocardiography, catheterization, and/or MRI data. Mitochondrial enzyme activities (citrate synthase and succinate dehydrogenase) were maintained during hypertrophy and decreased by ≈40% (P<0.05 versus control) at the stage of failure. In contrast, mtDNA content was progressively decreased in the hypertrophied RV through failure (by 28±8% and 67±11%, respectively, P<0.05 for both), whereas mtDNA-encoded gene expression was sustained by increased transcriptional activity during compensated hypertrophy but not in failure. Mitochondrial DNA depletion was attributed to reduced mtDNA replication in both hypertrophied and failing RV, and it was independent of PGC-1 downregulation but was accompanied by reduced expression of proteins constituting the mtDNA replication fork. Decreased mtDNA content in compensated hypertrophy was also associated with pathological changes of mitochondria ultrastructure. CONCLUSIONS: Impaired mtDNA replication causes early and progressive depletion of mtDNA in the RV of the patients with congenital heart disease during the transition from hypertrophy to failure. Decreased mtDNA content probably is a sensitive marker of mitochondrial injury in this patient population.


Asunto(s)
Progresión de la Enfermedad , Cardiopatías/congénito , Cardiopatías/complicaciones , Insuficiencia Cardíaca/etiología , Hipertrofia Ventricular Derecha/etiología , Mitocondrias Cardíacas/patología , Adolescente , Biomarcadores , Estudios de Casos y Controles , Niño , Preescolar , Citrato (si)-Sintasa/metabolismo , Replicación del ADN/genética , ADN Mitocondrial/genética , Femenino , Cardiopatías/patología , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/patología , Humanos , Hipertrofia Ventricular Derecha/diagnóstico , Hipertrofia Ventricular Derecha/patología , Lactante , Masculino , Mitocondrias Cardíacas/enzimología , Mitocondrias Cardíacas/ultraestructura , Pronóstico , Succinato Deshidrogenasa/metabolismo , Adulto Joven
15.
Circ Res ; 106(9): 1541-8, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20339121

RESUMEN

RATIONALE: Mitochondrial dysfunction plays a pivotal role in the development of heart failure. Animal studies suggest that impaired mitochondrial biogenesis attributable to downregulation of the peroxisome proliferator-activated receptor gamma coactivator (PGC)-1 transcriptional pathway is integral of mitochondrial dysfunction in heart failure. OBJECTIVE: The study sought to define mechanisms underlying the impaired mitochondrial biogenesis and function in human heart failure. METHODS AND RESULTS: We collected left ventricular tissue from end-stage heart failure patients and from nonfailing hearts (n=23, and 19, respectively). The mitochondrial DNA (mtDNA) content was decreased by >40% in the failing hearts, after normalization for a moderate decrease in citrate synthase activity (P<0.05). This was accompanied by reductions in mtDNA-encoded proteins (by 25% to 80%) at both mRNA and protein level (P<0.05). The mRNA levels of PGC-1alpha/beta and PRC (PGC-1-related coactivator) were unchanged, whereas PGC-1alpha protein increased by 58% in the failing hearts. Among the PGC-1 coactivating targets, the expression of estrogen-related receptor alpha and its downstream genes decreased by up to 50% (P<0.05), whereas peroxisome proliferator-activated receptor alpha and its downstream gene expression were unchanged in the failing hearts. The formation of D-loop in the mtDNA was normal but D-loop extension, which dictates the replication process of mtDNA, was decreased by 75% in the failing hearts. Furthermore, DNA oxidative damage was increased by 50% in the failing hearts. CONCLUSIONS: Mitochondrial biogenesis is severely impaired as evidenced by reduced mtDNA replication and depletion of mtDNA in the human failing heart. These defects are independent of the downregulation of the PGC-1 expression suggesting novel mechanisms for mitochondrial dysfunction in heart failure.


Asunto(s)
Replicación del ADN , ADN Mitocondrial/biosíntesis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Mitocondrias/genética , Mitocondrias/patología , Adulto , Anciano , ADN Mitocondrial/genética , Regulación hacia Abajo , Femenino , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Adulto Joven
16.
J Biol Chem ; 282(34): 24660-9, 2007 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-17584738

RESUMEN

cAMP-dependent protein kinase induction of PPARgamma coactivator-1alpha (PGC-1alpha) and uncoupling protein 1 (UCP1) expression is an essential step in the commitment of preadipocytes to the brown adipose tissue (BAT) lineage. We studied the molecular mechanisms responsible for differential expression of PGC-1alpha in HIB1B (BAT) and 3T3-L1 white adipose tissue (WAT) precursor cell lines. In HIB1B cells PGC-1alpha and UCP1 expression is cAMP-inducible, but in 3T3-L1 cells, expression is reduced and is cAMP-insensitive. A proximal 264-bp PGC-1alpha reporter construct was cAMP-inducible only in HIB1B cells and was suppressed by site-directed mutagenesis of the proximal cAMP response element (CRE). In electrophoretic mobility shift assays, the transcription factors CREB and C/EBPbeta, but not C/EBPalpha and C/EBPdelta, bound to the CRE on the PGC-1alpha promoter region in HIB1B and 3T3-L1 cells. Chromatin immunoprecipitation studies demonstrated that C/EBPbeta and CREB bound to the CRE region in HIB1B and 3T3-L1 cell lysates. C/EBPbeta expression was induced by cAMP only in HIB1B cells, and overexpression of C/EBPbeta rescued cAMP-inducible PGC-1alpha and UCP1 expression in 3T3-L1 cells. These data demonstrate that differentiation of preadipocytes toward the BAT rather than the WAT phenotype is controlled in part by the action of C/EBPbeta on the CRE in PGC-1alpha proximal promoter.


Asunto(s)
Adipocitos/citología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica , Células 3T3-L1 , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Diferenciación Celular , AMP Cíclico/metabolismo , Ratones , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Regiones Promotoras Genéticas , Unión Proteica , Transactivadores/genética , Factores de Transcripción
17.
Endocrinology ; 148(1): 461-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17023522

RESUMEN

In ruminants and other large animals, expression of uncoupling protein-1 (UCP1) in brown adipose tissue (BAT) is confined to the perinatal period when it plays a key role in nonshivering thermogenesis. This study determined whether loss of expression of the BAT phenotype was due to reduced response to a beta-agonist, isoprenaline, and expression of the peroxisome proliferator-activated receptor (PPAR) family [PPARalpha, PPARgamma, PPAR coactivator 1alpha (PGC-1alpha)], which regulates UCP1 gene expression. Perirenal adipose tissue (PAT) was sampled from ovine fetuses, newborn lambs, and lambs on d 1, 5, 7, and 21 of life. UCP1 mRNA and protein in PAT increased from d 123 of fetal life to reach a maximum at birth followed by a rapid decrease over the first 5 d of life. Expression of the coactivator, PGC-1alpha and PPAR alpha, peaked between fetal day 123 and birth, and then declined to undetectable levels in the first days of life. In vivo administration of isoprenaline was able to induce expression of UCP1, PGC-1alpha, and PPARalpha in BAT up to 5 d of age but thereafter was ineffective. In vitro addition of beta-receptor, PPARalpha, and PPARgamma agonists were unable to overcome the suppression of UCP1, PPARalpha, and PPARgamma expression observed in differentiated adipocytes prepared from 30-d-old compared with 1-d-old lambs. These data are consistent with a model in which postnatal loss of UCP1 expression and beta-adrenergic induction of the brown adipocyte phenotype is due to loss of expression of PGC-1alpha and PPARalpha.


Asunto(s)
Tejido Adiposo Pardo , Agonistas Adrenérgicos beta/farmacología , Animales Recién Nacidos/fisiología , Isoproterenol/farmacología , Adipocitos/fisiología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Pardo/fisiología , Animales , Regulación de la Temperatura Corporal/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Núcleo Celular/metabolismo , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Canales Iónicos/genética , Masculino , Proteínas Mitocondriales/genética , PPAR alfa/agonistas , PPAR alfa/genética , PPAR gamma/agonistas , PPAR gamma/genética , Embarazo , ARN Mensajero/metabolismo , Receptores Adrenérgicos beta/metabolismo , Ovinos , Transactivadores/genética , Transactivadores/metabolismo , Proteína Desacopladora 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...