Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Mol Cell Cardiol ; 130: 49-58, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30910669

RESUMEN

Adenosine exerts numerous protective actions in the heart, including attenuation of cardiac hypertrophy. Adenosine kinase (ADK) converts adenosine to adenosine monophosphate (AMP) and is the major route of myocardial adenosine metabolism, however, the impact of ADK activity on cardiac structure and function is unknown. To examine the role of ADK in cardiac homeostasis and adaptation to stress, conditional cardiomyocyte specific ADK knockout mice (cADK-/-) were produced using the MerCreMer-lox-P system. Within 4 weeks of ADK disruption, cADK-/- mice developed spontaneous hypertrophy and increased ß-Myosin Heavy Chain expression without observable LV dysfunction. In response to 6 weeks moderate left ventricular pressure overload (transverse aortic constriction;TAC), wild type mice (WT) exhibited ~60% increase in ventricular ADK expression and developed LV hypertrophy with preserved LV function. In contrast, cADK-/- mice exhibited significantly greater LV hypertrophy and cardiac stress marker expression (atrial natrurietic peptide and ß-Myosin Heavy Chain), LV dilation, reduced LV ejection fraction and increased pulmonary congestion. ADK disruption did not decrease protein methylation, inhibit AMPK, or worsen fibrosis, but was associated with persistently elevated mTORC1 and p44/42 ERK MAP kinase signaling and a striking increase in microtubule (MT) stabilization/detyrosination. In neonatal cardiomyocytes exposed to hypertrophic stress, 2-chloroadenosine (CADO) or adenosine treatment suppressed MT detyrosination, which was reversed by ADK inhibition with iodotubercidin or ABT-702. Conversely, adenoviral over-expression of ADK augmented CADO destabilization of MTs and potentiated CADO attenuation of cardiomyocyte hypertrophy. Together, these findings indicate a novel adenosine receptor-independent role for ADK-mediated adenosine metabolism in cardiomyocyte microtubule dynamics and protection against maladaptive hypertrophy.


Asunto(s)
Adenosina Quinasa/metabolismo , Cardiomegalia/metabolismo , Sistema de Señalización de MAP Quinasas , Microtúbulos/metabolismo , Miocitos Cardíacos/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Quinasa/genética , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Ratones , Ratones Noqueados , Microtúbulos/genética , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley , Volumen Sistólico/genética , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
2.
J Mol Cell Cardiol ; 128: 105-116, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30707992

RESUMEN

While most mitochondrial proteins are encoded in the nucleus and translated on cytosolic/endoplasmic reticulum ribosomes, proteins encoded by mitochondrial DNA are translated on mitochondrial ribosomes. Mitochondrial GTPases 1 (MTG1) regulates mitochondrial ribosome assembly and translation, but its impact on cardiac adaptation to stress is unknown. Here, we found that MTG1 is dramatically elevated in hearts of dilated cardiomyopathy patients and in mice exposed to left ventricular pressure overload (AB). To examine the role of MTG1 in cardiac hypertrophy and heart failure, MTG1 loss/gain of function studies were performed in cultured cardiomyocytes and mice exposed to hypertrophic stress. MTG1 shRNA and adenoviral overexpression studies indicated that MTG1 expression attenuates angiotensin II-induced hypertrophy in cultured cardiomyocytes, while MTG1 KO mice exhibited no observable cardiac phenotype under basal conditions. MTG1 deficiency significantly exacerbated AB-induced cardiac hypertrophy, expression of hypertrophic stress markers, fibrosis, and LV dysfunction in comparison to WT mice. Conversely, transgenic cardiac MTG1 expression attenuated AB-induced hypertrophy and LV dysfunction. Mechanistically, MTG1 preserved mitochondrial respiratory chain complex activity during pressure overload, which further attenuated ROS generation. Moreover, we demonstrated that TAK1, P38 and JNK1/2 activity is downregulated in the MTG1 overexpression group. Importantly, dampening oxidative stress with N-acetylcysteine (NAC) lowered hypertrophy in MTG1 KO to WT levels. Collectively, our data indicate that MTG1 protects against pressure overload-induced cardiac hypertrophy and dysfunction by preserving mitochondrial function and reducing oxidative stress and downstream TAK1 stress signaling.


Asunto(s)
Cardiomiopatía Dilatada/genética , GTP Fosfohidrolasas/genética , Insuficiencia Cardíaca/genética , Quinasas Quinasa Quinasa PAM/genética , Angiotensina II/genética , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomiopatía Dilatada/patología , Insuficiencia Cardíaca/patología , Humanos , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/genética
3.
Analyst ; 144(14): 4219-4232, 2019 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-31199423

RESUMEN

We present the ability to conduct single micrometer-sized uranium particle age-dating measurements on particles that are younger, smaller, and less enriched in 235U content than previously reported. Specifically, we use large geometry secondary ion mass spectrometry (LG-SIMS) to precisely measure the 230Th/234U radiochronometer, combined with a systematic treatment of relevant parameters such as particle size, enrichment, and age, to achieve this development. We describe the necessary requirements for instrument background, interference rejection, abundance sensitivity, and other instrumental conditions that allow for this advance in single-particle uranium age-dating. We introduce the use of statistics developed by Feldman and Cousins to generate 95% confidence intervals in particle age, even when 230Th daughter ions are not detected. For particles where counts are limited and are of identical isotopic signatures, we provide an option for aggregating individual measurements of single particles to reduce measurement uncertainty, as if the measurement had been performed on one larger particle. The methodology is validated on a range of certified reference materials and 'real-world' samples, ranging in age from 15 to 60 years, and on individual particles ranging in equivalent size from 0.6 to 6.8 micrometers. Additionally, we provide model age calculations for particles ranging in size from 1.0 to 3.0 micrometers across enrichments ranging from natural uranium to highly-enriched uranium and on ages ranging from 0 to 60 years. Experimental results compare well with the predicted model ages, providing realistic guidance for expectations of single micrometer-sized uranium particle age-dating measurements. The age-dating capabilities described herein are directly relevant to the International Atomic Energy Agency (IAEA) and its mission to safeguard nuclear materials and monitor member state nuclear programs.

4.
Mol Pharmacol ; 93(4): 335-343, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29358221

RESUMEN

According to current views, oxidation of aldehyde dehydrogenase-2 (ALDH2) during glyceryltrinitrate (GTN) biotransformation is essentially involved in vascular nitrate tolerance and explains the dependence of this reaction on added thiols. Using a novel fluorescent intracellular nitric oxide (NO) probe expressed in vascular smooth muscle cells (VSMCs), we observed ALDH2-catalyzed formation of NO from GTN in the presence of exogenously added dithiothreitol (DTT), whereas only a short burst of NO, corresponding to a single turnover of ALDH2, occurred in the absence of DTT. This short burst of NO associated with oxidation of the reactive C302 residue in the active site was followed by formation of low-nanomolar NO, even without added DTT, indicating slow recovery of ALDH2 activity by an endogenous reductant. In addition to the thiol-reversible oxidation of ALDH2, thiol-refractive inactivation was observed, particularly under high-turnover conditions. Organ bath experiments with rat aortas showed that relaxation by GTN lasted longer than that caused by the NO donor diethylamine/NONOate, in line with the long-lasting nanomolar NO generation from GTN observed in VSMCs. Our results suggest that an endogenous reductant with low efficiency allows sustained generation of GTN-derived NO in the low-nanomolar range that is sufficient for vascular relaxation. On a longer time scale, mechanism-based, thiol-refractive irreversible inactivation of ALDH2, and possibly depletion of the endogenous reductant, will render blood vessels tolerant to GTN. Accordingly, full reactivation of oxidized ALDH2 may not occur in vivo and may not be necessary to explain GTN-induced vasodilation.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/metabolismo , Tolerancia a Medicamentos/fisiología , Músculo Liso Vascular/metabolismo , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Nitroglicerina/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Ditiotreitol/farmacología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Nitratos/farmacología , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
5.
J Biol Chem ; 291(46): 24076-24084, 2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27679490

RESUMEN

Aldehyde dehydrogenase-2 (ALDH2) catalyzes vascular bioactivation of the antianginal drug nitroglycerin (GTN), resulting in activation of soluble guanylate cyclase (sGC) and cGMP-mediated vasodilation. We have previously shown that a minor reaction of ALDH2-catalyzed GTN bioconversion, accounting for about 5% of the main clearance-based turnover yielding inorganic nitrite, results in direct NO formation and concluded that this minor pathway could provide the link between vascular GTN metabolism and activation of sGC. However, lack of detectable NO at therapeutically relevant GTN concentrations (≤1 µm) in vascular tissue called into question the biological significance of NO formation by purified ALDH2. We addressed this issue and used a novel, highly sensitive genetically encoded fluorescent NO probe (geNOp) to visualize intracellular NO formation at low GTN concentrations (≤1 µm) in cultured vascular smooth muscle cells (VSMC) expressing an ALDH2 mutant that reduces GTN to NO but lacks clearance-based GTN denitration activity. NO formation was compared with GTN-induced activation of sGC. The addition of 1 µm GTN to VSMC expressing either wild-type or C301S/C303S ALDH2 resulted in pronounced intracellular NO elevation, with maximal concentrations of 7 and 17 nm, respectively. Formation of GTN-derived NO correlated well with activation of purified sGC in VSMC lysates and cGMP accumulation in intact porcine aortic endothelial cells infected with wild-type or mutant ALDH2. Formation of NO and cGMP accumulation were inhibited by ALDH inhibitors chloral hydrate and daidzin. The present study demonstrates that ALDH2-catalyzed NO formation is necessary and sufficient for GTN bioactivation in VSMC.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Óxido Nítrico/metabolismo , Nitroglicerina/farmacocinética , Aldehído Deshidrogenasa Mitocondrial/antagonistas & inhibidores , Aldehído Deshidrogenasa Mitocondrial/genética , Sustitución de Aminoácidos , Animales , Bovinos , Hidrato de Cloral/farmacología , Humanos , Isoflavonas/farmacología , Ratones , Ratones Noqueados , Mutación Missense , Nitroglicerina/farmacología , Porcinos
6.
Circulation ; 134(18): 1353-1368, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27678262

RESUMEN

BACKGROUND: The recruitment of leukocytes to the vascular wall is a key step in hypertension development. Chemokine receptor CXCR2 mediates inflammatory cell chemotaxis in several diseases. However, the role of CXCR2 in hypertension development and the underlying mechanisms remain unknown. METHODS: Angiotensin II (490 ng·kg-1·min-1) or deoxycorticosterone acetate (DOCA) salt-induced mouse hypertensive models in genetic ablation, pharmacologic inhibition of CXCR2, and adoptive bone marrow transfer mice were used to determine the role of CXCR2 in hypertension (measured by radiotelemetry and tail-cuff system), inflammation (verified by flow cytometry and quantitative real-time polymerase chain reaction [PCR] analysis), vascular remodeling (studied by haematoxylin and eosin and Masson's trichrome staining), vascular dysfunction (assessed by aortic ring), and oxidative stress (indicated by nicotinamide adenine dinucleotide phosphate [NADPH] oxidase activity, dihydroethidium staining, and quantitative real-time PCR analysis). Moreover, the blood CXCR2+ cells in normotensive controls and hypertension patients were analyzed by flow cytometry. RESULTS: Angiotensin II significantly upregulated the expression of CXCR2 mRNA and protein and increased the number of CD45+ CXCR2+ cells in mouse aorta (n=8 per group). Selective CXCR2 knockout (CXCR2-/-) or pharmacological inhibition of CXCR2 markedly reduced angiotensin II- or DOCA-salt-induced blood pressure elevation, aortic thickness and collagen deposition, accumulation of proinflammatory cells into the vascular wall, and expression of cytokines (n=8 per group). CXCR2 inhibition also ameliorated angiotensin II-induced vascular dysfunction and reduced vascular superoxide formation, NADPH activity, and expression of NADPH oxidase subunits (n=6 per group). Bone marrow reconstitution of wild-type mice with CXCR2-/- bone marrow cells also significantly abolished angiotensin II-induced responses (n=6 per group). It is important to note that CXCR2 blockade reversed established hypertension induced by angiotensin II or DOCA-salt challenge (n=10 per group). Furthermore, we demonstrated that CXCR2+ proinflammatory cells were higher in hypertensive patients (n=30) compared with normotensive individuals (n=20). CONCLUSIONS: Infiltration of CXCR2+ cells plays a pathogenic role in arterial hypertension and vascular dysfunction. Inhibition of CXCR2 pathway may represent a novel therapeutic approach to treat hypertension.


Asunto(s)
Angiotensina II/farmacología , Hipertensión/prevención & control , Receptores de Interleucina-8B/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Hipertensión/genética , Hipertensión/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores de Interleucina-8B/antagonistas & inhibidores , Receptores de Interleucina-8B/genética , Regulación hacia Arriba/genética , Remodelación Vascular/genética
7.
Basic Res Cardiol ; 112(3): 25, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28349258

RESUMEN

Inflammatory responses play an important role in the development of left ventricular (LV) hypertrophy and dysfunction. Recent studies demonstrated that increased T-cell infiltration and T-cell activation contribute to LV hypertrophy and dysfunction. Dendritic cells (DCs) are professional antigen-presenting cells that orchestrate immune responses, especially by modulating T-cell function. In this study, we investigated the role of bone marrow-derived CD11c+ DCs in transverse aortic constriction (TAC)-induced LV fibrosis and hypertrophy in mice. We observed that TAC increased the number of CD11c+ cells and the percentage of CD11c+ MHCII+ (major histocompatibility complex class II molecule positive) DCs in the LV, spleen and peripheral blood in mice. Using bone marrow chimeras and an inducible CD11c+ DC ablation model, we found that depletion of bone marrow-derived CD11c+ DCs significantly attenuated LV fibrosis and hypertrophy in mice exposed to 24 weeks of moderate TAC. CD11c+ DC ablation significantly reduced TAC-induced myocardial inflammation as indicated by reduced myocardial CD45+ cells, CD11b+ cells, CD8+ T cells and activated effector CD8+CD44+ T cells in LV tissues. Moreover, pulsing of autologous DCs with LV homogenates from TAC mice promoted T-cell proliferation. These data indicate that bone marrow-derived CD11c+ DCs play a maladaptive role in hemodynamic overload-induced cardiac inflammation, hypertrophy and fibrosis through the presentation of cardiac self-antigens to T cells.


Asunto(s)
Células Dendríticas/inmunología , Hipertrofia Ventricular Izquierda/inmunología , Activación de Linfocitos/inmunología , Remodelación Ventricular/inmunología , Animales , Presentación de Antígeno/inmunología , Células de la Médula Ósea/inmunología , Antígeno CD11c/inmunología , Linfocitos T CD8-positivos/inmunología , Cardiomegalia/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Miocarditis/inmunología
8.
Basic Res Cardiol ; 112(5): 55, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819685

RESUMEN

Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthases that limits nitric oxide bioavailability. Dimethylarginine dimethylaminohydrolase-1 (DDAH1) exerts a critical role for ADMA degradation and plays an important role in NO signaling. In the heart, DDAH1 is observed in endothelial cells and in the sarcolemma of cardiomyocytes. While NO signaling is important for cardiac adaptation to stress, DDAH1 impact on cardiomyocyte homeostasis is not clear. Here we used the MerCreMer-LoxP model to specifically disrupt cardiomyocyte DDAH1 expression in adult mice to determine the physiological impact of cardiomyocyte DDAH1 under basal conditions and during hypertrophic stress imposed by transverse aortic constriction (TAC). Under control conditions, cardiomyocyte-specific DDAH1 knockout (cDDAH KO) had no detectable effect on plasma ADMA and left ventricular (LV) hypertrophy or function in adult or aging mice. In response to TAC, DDAH1 levels were elevated 2.5-fold in WT mice, which exhibited no change in LV or plasma ADMA content and moderate LV hypertrophy and LV dysfunction. In contrast, cDDAH1 KO mice exposed to TAC showed no increase in LV DDAH1 expression, slightly increased LV tissue ADMA levels, no increase in plasma ADMA, but significantly exacerbated LV hypertrophy, fibrosis, nitrotyrosine production, and LV dysfunction. These findings indicate cardiomyocyte DDAH1 activity is dispensable for cardiac function under basal conditions, but plays an important role in attenuating cardiac hypertrophy and ventricular remodeling under stress conditions, possibly through locally confined regulation of subcellular ADMA and NO signaling.


Asunto(s)
Amidohidrolasas/metabolismo , Hipertrofia Ventricular Izquierda/prevención & control , Miocitos Cardíacos/enzimología , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda , Remodelación Ventricular , Amidohidrolasas/deficiencia , Amidohidrolasas/genética , Animales , Arginina/análogos & derivados , Arginina/sangre , Factor Natriurético Atrial/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Predisposición Genética a la Enfermedad , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones Noqueados , Miocitos Cardíacos/patología , Óxido Nítrico/metabolismo , Fenotipo , Transducción de Señal , Tirosina/análogos & derivados , Tirosina/metabolismo , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología
9.
J Anal At Spectrom ; 32(2): 393-401, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28626281

RESUMEN

The determination of the relative isotopic abundance by secondary ion mass spectrometry of 236U in uranium-containing material is complicated by the presence of 235U1H+ ions at the same nominal mass as the uranium isotopic peak. The net intensity of the 236U signal is usually determined by a peak-stripping procedure, whereby the 235U1H+ contribution is obtained by applying the 238U1H+/238U+ ratio to the 235U+ signal. The subtraction of one signal from another has consequences for the uncertainty of the final 236U abundance determination that may be especially significant when the amount of sample is limited, as is the case with small uranium particles that are of great interest for nuclear safeguards. We have developed a model based on Poisson counting statistics to determine the effects of various parameters on the uncertainty of the 236U abundance, including uranium enrichment level, hydride-to-parent ratio, uranium mass consumed during analysis, single versus multichannel counting, and sample substrate composition. The model predictions have been successfully tested against experimental measurements of uranium oxide particles of both 3% and 90% enrichment in 235U.

10.
Vasc Med ; 22(3): 179-188, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28145161

RESUMEN

Pharmacologic inhibition of nitric oxide production inhibits growth of coronary collateral vessels. Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is the major enzyme that degrades asymmetric dimethylarginine (ADMA), a potent inhibitor of nitric oxide synthase. Here we examined regulation of the ADMA-DDAH1 pathway in a canine model of recurrent myocardial ischemia during the time when coronary collateral growth is known to occur. Under basal conditions, DDAH1 expression was non-uniform across the left ventricular (LV) wall, with expression strongest in the subepicardium. In response to ischemia, DDAH1 expression was up-regulated in the midmyocardium of the ischemic zone, and this was associated with a significant reduction in myocardial interstitial fluid (MIF) ADMA. The decrease in MIF ADMA during ischemia was likely due to increased DDAH1 because myocardial protein arginine N-methyl transferase 1 (PRMT1) and the methylated arginine protein content (the source of ADMA) were unchanged or increased, respectively, at this time. The inflammatory mediators interleukin (IL-1ß) and tumor necrosis factor (TNF-α) were also elevated in the midmyocardium where DDAH1 expression was increased. Both of these factors significantly up-regulated DDAH1 expression in cultured human coronary artery endothelial cells. Taken together, these results suggest that inflammatory factors expressed in response to myocardial ischemia contributed to up-regulation of DDAH1, which was responsible for the decrease in MIF ADMA.


Asunto(s)
Amidohidrolasas/metabolismo , Vasos Coronarios/enzimología , Isquemia Miocárdica/enzimología , Miocardio/enzimología , Neovascularización Fisiológica , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Hipoxia de la Célula , Células Cultivadas , Circulación Colateral , Circulación Coronaria , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Perros , Células Endoteliales/enzimología , Humanos , Interleucina-1beta/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Miocardio/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Transducción de Señal , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Nitric Oxide ; 54: 8-14, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26805578

RESUMEN

Scavenging of nitric oxide (NO) often interferes with studies on NO signaling in cell-free preparations. We observed that formation of cGMP by NO-stimulated purified soluble guanylate cyclase (sGC) was virtually abolished in the presence of cytosolic preparations of porcine coronary arteries, with the scavenging activity localized in the tunica media (smooth muscle layer). Electrochemical measurement of NO release from a donor compound and light absorbance spectroscopy showed that cytosolic preparations contained a reduced heme protein that scavenged NO. This protein, which reacted with anti-human hemoglobin antibodies, was efficiently removed from the preparations by haptoglobin affinity chromatography. The cleared cytosols showed only minor scavenging of NO according to electrochemical measurements and did not decrease cGMP formation by NO-stimulated sGC. In contrast, the column flow-through caused a nearly 2-fold increase of maximal sGC activity (from 33.1 ± 1.6 to 54.9 ± 2.2 µmol × min(-1) × mg(-1)). The proteins retained on the affinity column were identified as hemoglobin α and ß subunits. The results indicate that hemoglobin, presumably derived from vasa vasorum erythrocytes, is present and scavenges NO in preparations of porcine coronary artery smooth muscle. Selective removal of hemoglobin-mediated scavenging unmasked stimulation of maximal NO-stimulated sGC activity by a soluble factor expressed in vascular tissue.


Asunto(s)
Vasos Coronarios/metabolismo , Hemoglobinas/metabolismo , Óxido Nítrico/metabolismo , Túnica Media/metabolismo , Animales , Bovinos , GMP Cíclico/metabolismo , Citoglobina , Globinas/metabolismo , Haptoglobinas/metabolismo , Humanos , Técnicas In Vitro , Guanilil Ciclasa Soluble/metabolismo , Porcinos
12.
Nitric Oxide ; 54: 73-81, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26923818

RESUMEN

Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthases that limits nitric oxide bioavailability and can increase production of NOS derived reactive oxidative species. Increased plasma ADMA is a one of the strongest predictors of mortality in patients who have had a myocardial infarction or suffer from chronic left heart failure, and is also an independent risk factor for several other conditions that contribute to heart failure development, including hypertension, coronary artery disease/atherosclerosis, diabetes, and renal dysfunction. The enzyme responsible for ADMA degradation is dimethylarginine dimethylaminohydrolase-1 (DDAH1). DDAH1 plays an important role in maintaining nitric oxide bioavailability and preserving cardiovascular function in the failing heart. Here, we examine mechanisms of abnormal NO production in heart failure, with particular focus on the role of ADMA and DDAH1.


Asunto(s)
Arginina/análogos & derivados , Insuficiencia Cardíaca/metabolismo , Óxido Nítrico/biosíntesis , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Arginina/metabolismo , Insuficiencia Cardíaca/etiología , Humanos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Ácido Peroxinitroso/metabolismo , Transducción de Señal , Superóxidos/metabolismo , omega-N-Metilarginina/metabolismo
13.
Circulation ; 129(13): 1397-406, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24463368

RESUMEN

BACKGROUND: Double-stranded RNA-dependent protein kinase (PKR) is a eukaryotic initiation factor 2α kinase that inhibits mRNA translation under stress conditions. PKR also mediates inflammatory and apoptotic signaling independently of translational regulation. Congestive heart failure is associated with cardiomyocyte hypertrophy, inflammation, and apoptosis, but the role of PKR in left ventricular hypertrophy and the development of congestive heart failure has not been examined. METHODS AND RESULTS: We observed increased myocardial PKR expression and translocation of PKR into the nucleus in humans and mice with congestive heart failure. To determine the impact of PKR on the development of congestive heart failure, PKR knockout and wild-type mice were exposed to pressure overload produced by transverse aortic constriction. Although heart size increased similarly in wild-type and PKR knockout mice after transverse aortic constriction, PKR knockout mice exhibited very little pulmonary congestion, well-preserved left ventricular ejection fraction and contractility, and significantly less myocardial fibrosis compared with wild-type mice. Bone marrow-derived cells from wild-type mice did not abolish the cardiac protective effect observed in PKR knockout mice, whereas bone marrow-derived cells from PKR knockout mice had no cardiac protective effect in wild-type mice. Mechanistically, PKR knockout attenuated transverse aortic constriction-induced tumor necrosis factor-α expression and leukocyte infiltration and lowered cardiac expression of proapoptotic factors (Bax and caspase-3), so that PKR knockout hearts were more resistant to transverse aortic constriction-induced cardiomyocyte apoptosis. PKR depletion in isolated cardiomyocytes also conferred protection against tumor necrosis factor-α- or lipopolysaccharide-induced apoptosis. CONCLUSION: PKR is a maladaptive factor upregulated in hemodynamic overload that contributes to myocardial inflammation, cardiomyocyte apoptosis, and the development of congestive heart failure.


Asunto(s)
Presión Sanguínea/fisiología , Insuficiencia Cardíaca/prevención & control , Insuficiencia Cardíaca/fisiopatología , Hemodinámica/fisiología , Disfunción Ventricular Izquierda/prevención & control , eIF-2 Quinasa/deficiencia , Adulto , Anciano , Animales , Aorta/fisiopatología , Apoptosis/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Insuficiencia Cardíaca/metabolismo , Humanos , Hipertrofia/fisiopatología , Hipertrofia/prevención & control , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Regulación hacia Arriba/fisiología , eIF-2 Quinasa/genética , eIF-2 Quinasa/fisiología
14.
Am J Physiol Heart Circ Physiol ; 304(5): H749-58, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23316058

RESUMEN

Cell hypertrophy requires increased protein synthesis and expansion of the cytoskeletal networks that support cell enlargement. AMPK limits anabolic processes, such as protein synthesis, when energy supply is insufficient, but its role in cytoskeletal remodeling is not known. Here, we examined the influence of AMPK in cytoskeletal remodeling during cardiomyocyte hypertrophy, a clinically relevant condition in which cardiomyocytes enlarge but do not divide. In neonatal cardiomyocytes, activation of AMPK with 5-aminoimidazole carboxamide ribonucleotide (AICAR) or expression of constitutively active AMPK (CA-AMPK) attenuated cell area increase by hypertrophic stimuli (phenylephrine). AMPK activation had little effect on intermediate filaments or myofilaments but dramatically reduced microtubule stability, as measured by detyrosinated tubulin levels and cytoskeletal tubulin accumulation. Importantly, low-level AMPK activation limited cell expansion and microtubule growth independent of mTORC1 or protein synthesis repression, identifying a new mechanism by which AMPK regulates cell growth. Mechanistically, AICAR treatment increased Ser-915 phosphorylation of microtubule-associated protein 4 (MAP4), which reduces affinity for tubulin and prevents stabilization of microtubules (MTs). RNAi knockdown of MAP4 confirmed its critical role in cardiomyocyte MT stabilization. In support of a pathophysiological role for AMPK regulation of cardiac microtubules, AMPK α2 KO mice exposed to pressure overload (transverse aortic constriction; TAC) demonstrated reduced MAP4 phosphorylation and increased microtubule accumulation that correlated with the severity of contractile dysfunction. Together, our data identify the microtubule cytoskeleton as a sensitive target of AMPK activity, and the data suggest a novel role for AMPK in limiting accumulation and densification of microtubules that occurs in response to hypertrophic stress.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patología , Microtúbulos/metabolismo , Miocitos Cardíacos/enzimología , Proteínas Quinasas Activadas por AMP/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/citología , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Tubulina (Proteína)/metabolismo , Presión Ventricular/fisiología
15.
Redox Biol ; 67: 102884, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37725888

RESUMEN

Adenosine kinase (ADK) plays the major role in cardiac adenosine metabolism, so that inhibition of ADK increases myocardial adenosine levels. While the cardioprotective actions of extracellular adenosine against ischemia/reperfusion (I/R) are well-established, the role of cellular adenosine in protection against I/R remains unknown. Here we investigated the role of cellular adenosine in epigenetic regulation on cardiomyocyte gene expression, glucose metabolism and tolerance to I/R. Evans blue/TTC staining and echocardiography were used to assess the extent of I/R injury in mice. Glucose metabolism was evaluated by positron emission tomography and computed tomography (PET/CT). Methylated DNA immunoprecipitation (MeDIP) and bisulfite sequencing PCR (BSP) were used to evaluate DNA methylation. Lentiviral/adenovirus transduction was used to overexpress DNMT1, and the OSI-906 was administered to inhibit IGF-1. Cardiomyocyte-specific ADK/IGF-1-knockout mice were used for mechanistic experiments.Cardiomyocyte-specific ADK knockout enhanced glucose metabolism and ameliorated myocardial I/R injury in vivo. Mechanistically, ADK deletion caused cellular adenosine accumulation, decreased DNA methyltransferase 1 (DNMT1) expression and caused hypomethylation of multiple metabolic genes, including insulin growth factor 1 (IGF-1). DNMT1 overexpression abrogated these beneficial effects by enhancing apoptosis and decreasing IGF-1 expression. Inhibition of IGF-1 signaling with OSI-906 or genetic knocking down of IGF-1 also abrogated the cardioprotective effects of ADK knockout, revealing the therapeutic potential of increasing IGF-1 expression in attenuating myocardial I/R injury. In conclusion, the present study demonstrated that cardiomyocyte ADK deletion ameliorates myocardial I/R injury via epigenetic upregulation of IGF-1 expression via the cardiomyocyte adenosine/DNMT1/IGF-1 axis.


Asunto(s)
Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Ratones , Animales , Miocitos Cardíacos/metabolismo , Epigénesis Genética , Adenosina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Isquemia/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Ratones Noqueados , Apoptosis , Reperfusión , ADN/metabolismo , Glucosa/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 31(7): 1540-6, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21493890

RESUMEN

OBJECTIVE: The objective of this study was to identify the role of dimethylarginine dimethylaminohydrolase-1 (DDAH1) in degrading the endogenous nitric oxide synthase inhibitors asymmetrical dimethylarginine (ADMA) and N(g)-monomethyl-L-arginine (L-NMMA). METHODS AND RESULTS: We generated a global-DDAH1 gene-deficient (DDAH1(-/-)) mouse strain to examine the role of DDAH1 in ADMA and l-NMMA degradation and the physiological consequences of loss of DDAH1. Plasma and tissue ADMA and L-NMMA levels in DDAH1(-/-) mice were several folds higher than in wild-type mice, but growth and development of these DDAH1(-/-) mice were similar to those of their wild-type littermates. Although the expression of DDAH2 was unaffected, DDAH activity was undetectable in all tissues tested. These findings indicate that DDAH1 is the critical enzyme for ADMA and L-NMMA degradation. Blood pressure was ≈ 20 mm Hg higher in the DDAH1(-/-) mice than in wild-type mice, but no other cardiovascular phenotype was found under unstressed conditions. Crossing DDAH1(+/-) male with DDAH1(+/-) female mice yielded DDAH1(+/+), DDAH1(+/-), and DDAH1(-/-) mice at the anticipated ratio of 1:2:1, indicating that DDAH1 is not required for embryonic development in this strain. CONCLUSIONS: Our findings indicate that DDAH1 is required for metabolizing ADMA and L-NMMA in vivo, whereas DDAH2 had no detectable role for degrading ADMA and l-NMMA.


Asunto(s)
Amidohidrolasas/metabolismo , Arginina/análogos & derivados , Enfermedades Cardiovasculares/etiología , Células Endoteliales/enzimología , Amidohidrolasas/deficiencia , Amidohidrolasas/genética , Animales , Arginina/sangre , Arginina/metabolismo , Presión Sanguínea , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/genética , Células Cultivadas , Inhibidores Enzimáticos/administración & dosificación , Femenino , Genotipo , Hipertensión/enzimología , Hipertensión/fisiopatología , Bombas de Infusión Implantables , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , NG-Nitroarginina Metil Éster/administración & dosificación , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Fenotipo , Interferencia de ARN , Factores de Riesgo , Especificidad por Sustrato , Factores de Tiempo , Transfección , omega-N-Metilarginina/metabolismo
17.
Eur J Pharmacol ; 927: 175050, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35618039

RESUMEN

Pharmacological inhibition of adenosine kinase (ADK), the major route of myocardial adenosine metabolism, can elicit acute cardioprotection against ischemia-reperfusion (IR) by increasing adenosine signaling. Here, we identified a novel, extended effect of the ADK inhibitor, ABT-702, on cardiac ADK protein longevity and investigated its impact on sustained adenosinergic cardioprotection. We found that ABT-702 treatment significantly reduced cardiac ADK protein content in mice 24-72 h after administration (IP or oral). ABT-702 did not alter ADK mRNA levels, but strongly diminished (ADK-L) isoform protein content through a proteasome-dependent mechanism. Langendorff perfusion experiments revealed that hearts from ABT-702-treated mice maintain higher adenosine release long after ABT-702 tissue elimination, accompanied by increased basal coronary flow (CF) and robust tolerance to IR. Sustained cardioprotection by ABT-702 did not involve increased nitric oxide synthase expression, but was completely dependent upon increased adenosine release in the delayed phase (24 h), as indicated by the loss of cardioprotection and CF increase upon perfusion of adenosine deaminase or adenosine receptor antagonist, 8-phenyltheophylline. Importantly, blocking adenosine receptor activity with theophylline during ABT-702 administration prevented ADK degradation, preserved late cardiac ADK activity, diminished CF increase and abolished delayed cardioprotection, indicating that early adenosine receptor signaling induces late ADK degradation to elicit sustained adenosine release. Together, these results indicate that ABT-702 induces a distinct form of delayed cardioprotection mediated by adenosine receptor-dependent, proteasomal degradation of cardiac ADK and enhanced adenosine signaling in the late phase. These findings suggest ADK protein stability may be pharmacologically targeted to achieve sustained adenosinergic cardioprotection.


Asunto(s)
Adenosina Quinasa , Morfolinas , Pirimidinas , Adenosina Quinasa/antagonistas & inhibidores , Adenosina Quinasa/metabolismo , Animales , Cardiotónicos/farmacología , Corazón/diagnóstico por imagen , Ratones , Morfolinas/farmacología , Miocardio/enzimología , Proteolisis/efectos de los fármacos , Pirimidinas/farmacología , Receptores Purinérgicos P1/metabolismo
18.
Circulation ; 121(13): 1474-83, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20308615

RESUMEN

BACKGROUND: Phosphodiesterase type 5 (PDE5) inhibition has been shown to exert profound beneficial effects in the failing heart, suggesting a significant role for PDE5 in the development of congestive heart failure (CHF). The purpose of this study is to test the hypothesis that oxidative stress causes increased PDE5 expression in cardiac myocytes and that increased PDE5 contributes to the development of CHF. METHODS AND RESULTS: Myocardial PDE5 expression and cellular distribution were determined in left ventricular samples from patients with end-stage CHF and normal donors and from mice after transverse aortic constriction (TAC)-induced CHF. Compared with donor human hearts, myocardial PDE5 protein was increased approximately equal 4.5-fold in CHF samples, and the increase of myocardial PDE5 expression was significantly correlated with myocardial oxidative stress markers 3'-nitrotyrosine or 4-hydroxynonenal expression (P<0.05). Histological examination demonstrated that PDE5 was mainly expressed in vascular smooth muscle in normal donor hearts, but its expression was increased in both cardiac myocytes and vascular smooth muscle of CHF hearts. Myocardial PDE5 protein content and activity also increased in mice after TAC-induced CHF (P<0.05). When the superoxide dismutase (SOD) mimetic M40401 was administered to attenuate oxidative stress, the increased PDE5 protein and activity caused by TAC was blunted, and the hearts were protected against left ventricular hypertrophy and CHF. Conversely, increased myocardial oxidative stress in superoxide dismutase 3 knockout mice caused a greater increase of PDE5 expression and CHF after TAC. In addition, administration of sildenafil to inhibit PDE5 attenuated TAC-induced myocardial oxidative stress, PDE5 expression, and CHF. CONCLUSIONS: Myocardial oxidative stress increases PDE5 expression in the failing heart. Reducing oxidative stress by treatment with M40401 attenuated cardiomyocyte PDE5 expression. This and selective inhibition of PDE5 protected the heart against pressure overload-induced left ventricular hypertrophy and CHF.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/enzimología , Estrés Oxidativo/fisiología , Animales , Antioxidantes/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Compuestos Organometálicos/farmacología , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5 , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Citrato de Sildenafil , Sulfonas/farmacología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 300(5): H1722-32, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21335462

RESUMEN

There is evidence that extracellular adenosine can attenuate cardiac hypertrophy, but the mechanism by which this occurs is not clear. Here we investigated the role of adenosine receptors and adenosine metabolism in attenuation of cardiomyocyte hypertrophy. Phenylephrine (PE) caused hypertrophy of neonatal rat cardiomyocytes with increases of cell surface area, protein synthesis, and atrial natriuretic peptide (ANP) expression. These responses were attenuated by 5 µM 2-chloroadenosine (CADO; adenosine deaminase resistant adenosine analog) or 10 µM adenosine. While antagonism of adenosine receptors partially blocked the reduction of ANP expression produced by CADO, it did not restore cell size or protein synthesis. In support of a role for intracellular adenosine metabolism in regulating hypertrophy, the adenosine kinase (AK) inhibitors iodotubercidin and ABT-702 completely reversed the attenuation of cell size, protein synthesis, and expression of ANP by CADO or ADO. Examination of PE-induced phosphosignaling pathways revealed that CADO treatment did not reduce AKT(Ser47³) phosphorylation but did attenuate sustained phosphorylation of Raf(Ser³³8) (24-48 h), mTOR(Ser²448) (24-48 h), p70S6k(Thr³89) (2.5-48 h), and ERK(Thr²°²/Tyr²°4) (48 h). Inhibition of AK restored activation of these enzymes in the presence of CADO. Using dominant negative and constitutively active Raf adenoviruses, we found that Raf activation is necessary and sufficient for PE-induced mTORC1 signaling and cardiomyocyte hypertrophy. CADO treatment still blocked p70S6k(Thr³89) phosphorylation and hypertrophy downstream of constitutively active Raf, however, despite a high level phosphorylation of ERK(Thr202/Tyr204) and AKT(Ser47³). Reduction of Raf-induced p70S6k(Thr³89) phosphorylation and hypertrophy by CADO was reversed by inhibiting AK. Together, these results identify AK as an important mediator of adenosine attenuation of cardiomyocyte hypertrophy, which acts, at least in part, through inhibition of Raf signaling to mTOR/p70S6k.


Asunto(s)
Adenosina Quinasa/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Transducción de Señal/fisiología , Quinasas de la Proteína-Quinasa Activada por el AMP , Adenosina/metabolismo , Adenosina Quinasa/antagonistas & inhibidores , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Hipertrofia/metabolismo , Hipertrofia/patología , Hipertrofia/prevención & control , Modelos Animales , Morfolinas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Proteínas Quinasas/metabolismo , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P1/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Quinasas raf/metabolismo
20.
Circ Res ; 103(9): 1009-17, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18802029

RESUMEN

Sarcolemmal ATP-sensitive potassium channels (K(ATP)) act as metabolic sensors that facilitate adaptation of the left ventricle to changes in energy requirements. This study examined the mechanism by which K(ATP) dysfunction impairs the left ventricular response to stress using transgenic mouse strains with cardiac-specific disruption of K(ATP) activity (SUR1-tg mice) or Kir6.2 gene deficiency (Kir6.2 KO). Both SUR1-tg and Kir6.2 KO mice had normal left ventricular mass and function under unstressed conditions. Following chronic transverse aortic constriction, both SUR1-tg and Kir6.2 KO mice developed more severe left ventricular hypertrophy and dysfunction as compared with their corresponding WT controls. Both SUR1-tg and Kir6.2 KO mice had significantly decreased expression of peroxisome proliferator-activated receptor gamma coactivator (PGC)-1alpha and a group of energy metabolism related genes at both protein and mRNA levels. Furthermore, disruption of K(ATP) repressed expression and promoter activity of PGC-1alpha in cultured rat neonatal cardiac myocytes in response to hypoxia, indicating that K(ATP) activity is required to maintain PGC-1alpha expression under stress conditions. PGC-1alpha gene deficiency also exacerbated chronic transverse aortic constriction-induced ventricular hypertrophy and dysfunction, suggesting that depletion of PGC-1alpha can worsen systolic overload induced ventricular dysfunction. Both SUR1-tg and Kir6.2 KO mice had decreased FOXO1 after transverse aortic constriction, in agreement with the reports that a decrease of FOXO1 can repress PGC-1alpha expression. Furthermore, inhibition of K(ATP) caused a decrease of FOXO1 associated with PGC-1alpha promoter. These data indicate that K(ATP) channels facilitate the cardiac response to stress by regulating PGC-1alpha and its target genes, at least partially through the FOXO1 pathway.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Hemodinámica , Hipertrofia Ventricular Izquierda/metabolismo , Canales KATP/metabolismo , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Droga/metabolismo , Sarcolema/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/genética , Animales , Animales Recién Nacidos , Aorta/cirugía , Secuencia de Bases , Hipoxia de la Célula , Células Cultivadas , Constricción , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Canales KATP/deficiencia , Canales KATP/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/deficiencia , Canales de Potasio de Rectificación Interna/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Receptores de Droga/antagonistas & inhibidores , Receptores de Droga/genética , Sarcolema/efectos de los fármacos , Índice de Severidad de la Enfermedad , Receptores de Sulfonilureas , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción , Transfección , Disfunción Ventricular Izquierda/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA