Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 377(2): 293-304, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33602875

RESUMEN

In failing hearts, Na+/Ca2+ exchanger (NCX) overactivity contributes to Ca2+ depletion, leading to contractile dysfunction. Inhibition of NCX is expected to normalize Ca2+ mishandling, to limit afterdepolarization-related arrhythmias, and to improve cardiac function in heart failure (HF). SAR340835/SAR296968 is a selective NCX inhibitor for all NCX isoforms across species, including human, with no effect on the native voltage-dependent calcium and sodium currents in vitro. Additionally, it showed in vitro and in vivo antiarrhythmic properties in several models of early and delayed afterdepolarization-related arrhythmias. Its effect on cardiac function was studied under intravenous infusion at 250,750 or 1500 µg/kg per hour in dogs, which were either normal or submitted to chronic ventricular pacing at 240 bpm (HF dogs). HF dogs were infused with the reference inotrope dobutamine (10 µg/kg per minute, i.v.). In normal dogs, NCX inhibitor increased cardiac contractility (dP/dtmax) and stroke volume (SV) and tended to reduce heart rate (HR). In HF dogs, NCX inhibitor significantly and dose-dependently increased SV from the first dose (+28.5%, +48.8%, and +62% at 250, 750, and 1500 µg/kg per hour, respectively) while significantly increasing dP/dtmax only at 1500 (+33%). Furthermore, NCX inhibitor significantly restored sympathovagal balance and spontaneous baroreflex sensitivity (BRS) from the first dose and reduced HR at the highest dose. In HF dogs, dobutamine significantly increased dP/dtmax and SV (+68.8%) but did not change HR, sympathovagal balance, or BRS. Overall, SAR340835, a selective potent NCX inhibitor, displayed a unique therapeutic profile, combining antiarrhythmic properties, capacity to restore systolic function, sympathovagal balance, and BRS in HF dogs. NCX inhibitors may offer new therapeutic options for acute HF treatment. SIGNIFICANCE STATEMENT: HF is facing growing health and economic burden. Moreover, patients hospitalized for acute heart failure are at high risk of decompensation recurrence, and no current acute decompensated HF therapy definitively improved outcomes. A new potent, Na+/Ca2+ exchanger inhibitor SAR340835 with antiarrhythmic properties improved systolic function of failing hearts without creating hypotension, while reducing heart rate and restoring sympathovagal balance. SAR340835 may offer a unique and attractive pharmacological profile for patients with acute heart failure as compared with current inotrope, such as dobutamine.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Moduladores del Transporte de Membrana/uso terapéutico , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Nervio Vago/efectos de los fármacos , Animales , Barorreflejo , Perros , Corazón/efectos de los fármacos , Frecuencia Cardíaca , Moduladores del Transporte de Membrana/administración & dosificación , Moduladores del Transporte de Membrana/farmacología , Contracción Miocárdica , Miocardio/metabolismo , Porcinos
2.
Hepatol Commun ; 4(7): 1056-1072, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32626837

RESUMEN

The worldwide obesity and type 2 diabetes epidemics have led to an increase in nonalcoholic fatty liver disease (NAFLD). NAFLD covers a spectrum of hepatic pathologies ranging from simple steatosis to nonalcoholic steatohepatitis, characterized by fibrosis and hepatic inflammation. Nonalcoholic steatohepatitis predisposes to the onset of hepatocellular carcinoma (HCC). Here, we characterized the effect of a pharmacological activator of the intracellular energy sensor adenosine monophosphate-activated protein kinase (AMPK) on NAFLD progression in a mouse model. The compound stimulated fat oxidation by activating AMPK in both liver and skeletal muscle, as revealed by indirect calorimetry. This translated into an ameliorated hepatic steatosis and reduced fibrosis progression in mice fed a diet high in fat, cholesterol, and fructose for 20 weeks. Feeding mice this diet for 80 weeks caused the onset of HCC. The administration of the AMPK activator for 12 weeks significantly reduced tumor incidence and size. Conclusion: Pharmacological activation of AMPK reduces NAFLD progression to HCC in preclinical models.

4.
Europace ; 17(10): 1457-66, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26364316

RESUMEN

Atrial fibrillation (AF) is the most common sustained arrhythmia in the general population. As an age-related arrhythmia AF is becoming a huge socio-economic burden for European healthcare systems. Despite significant progress in our understanding of the pathophysiology of AF, therapeutic strategies for AF have not changed substantially and the major challenges in the management of AF are still unmet. This lack of progress may be related to the multifactorial pathogenesis of atrial remodelling and AF that hampers the identification of causative pathophysiological alterations in individual patients. Also, again new mechanisms have been identified and the relative contribution of these mechanisms still has to be established. In November 2010, the European Union launched the large collaborative project EUTRAF (European Network of Translational Research in Atrial Fibrillation) to address these challenges. The main aims of EUTRAF are to study the main mechanisms of initiation and perpetuation of AF, to identify the molecular alterations underlying atrial remodelling, to develop markers allowing to monitor this processes, and suggest strategies to treat AF based on insights in newly defined disease mechanisms. This article reports on the objectives, the structure, and initial results of this network.


Asunto(s)
Fibrilación Atrial/diagnóstico , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Remodelación Atrial , Investigación Biomédica Traslacional/tendencias , Conducta Cooperativa , Electrocardiografía , Europa (Continente) , Humanos
5.
Nat Chem Biol ; 9(8): 507-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23728494

RESUMEN

Most known small-molecule inhibitors of voltage-gated ion channels have poor subtype specificity because they interact with a highly conserved binding site in the central cavity. Using alanine-scanning mutagenesis, electrophysiological recordings and molecular modeling, we have identified a new drug-binding site in Kv1.x channels. We report that Psora-4 can discriminate between related Kv channel subtypes because, in addition to binding the central pore cavity, it binds a second, less conserved site located in side pockets formed by the backsides of S5 and S6, the S4-S5 linker, part of the voltage sensor and the pore helix. Simultaneous drug occupation of both binding sites results in an extremely stable nonconducting state that confers high affinity, cooperativity, use-dependence and selectivity to Psora-4 inhibition of Kv1.x channels. This new mechanism of inhibition represents a molecular basis for the development of a new class of allosteric and selective voltage-gated channel inhibitors.


Asunto(s)
Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/química , Ficusina/química , Ficusina/farmacología , Canal de Potasio Kv1.5/metabolismo , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Especificidad por Sustrato
6.
Sleep ; 36(5): 699-708, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23633752

RESUMEN

STUDY OBJECTIVES: Drug treatment for obstructive sleep apnea (OSA) is desirable because at least 30% of patients do not tolerate continuous positive airway pressure (CPAP) treatment. The negative pressure reflex (NPR) involving superficially located mechanoreceptors in the upper airway (UA) is an important mechanism for UA patency inhibitable by topical UA anesthesia (lidocaine). The NPR may serve as a target for pharmacological intervention for a topical treatment of OSA. The objective was to determine the effect of pharmacological augmentation of the NPR on UA collapsibility. DESIGN: We developed a model of UA collapsibility in which application of negative pressures caused UA collapses in spontaneously breathing α-chloralose-urethane anesthetized pigs as indicated by characteristic tracheal pressure and air flow changes. SETTING: N/A. PATIENTS OR PARTICIPANTS: N/A. INTERVENTIONS: N/A. MEASUREMENTS AND RESULTS: The potassium channel blocker AVE0118 administered topically to the UA in doses of 1, 3, and 10 mg per nostril sensitized the NPR, shifting the mechanoreceptor response threshold for the genioglossus muscle to more positive pressures (P < 0.001; n = 6 per group) and dose-dependently inhibited UA collapsibility. Ten mg of AVE0118 prevented UA collapses against negative pressures of -150 mbar (P < 0.01) for > 4 h in all pigs, while in control pigs the UA collapsed at -50 mbar or less negative pressures. The effect of AVE0118 was abolished by UA lidocaine anesthesia. Acute intravenous administration of naloxone or acetazolamide was ineffective; paroxetine and mirtazepine were weakly effective and fluoxetine was moderately effective in line with reported clinical efficacy. CONCLUSION: Topical administration of AVE0118 to the UA is a promising pharmacologic approach for the treatment of OSA.


Asunto(s)
Resistencia de las Vías Respiratorias/fisiología , Compuestos de Bifenilo/administración & dosificación , Mecanorreceptores/fisiología , Bloqueadores de los Canales de Potasio/administración & dosificación , Apnea Obstructiva del Sueño/tratamiento farmacológico , Administración Intranasal , Anestésicos Intravenosos , Animales , Cloralosa , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Masculino , Mecanorreceptores/efectos de los fármacos , Apnea Obstructiva del Sueño/etiología , Porcinos
7.
FASEB J ; 26(2): 513-22, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22002906

RESUMEN

Inward rectifier potassium channels of the Kir2 subfamily are important determinants of the electrical activity of brain and muscle cells. Genetic mutations in Kir2.1 associate with Andersen-Tawil syndrome (ATS), a familial disorder leading to stress-triggered periodic paralysis and ventricular arrhythmia. To identify the molecular mechanisms of this stress trigger, we analyze Kir channel function and localization electrophysiologically and by time-resolved confocal microscopy. Furthermore, we employ a mathematical model of muscular membrane potential. We identify a novel corticoid signaling pathway that, when activated by glucocorticoids, leads to enrichment of Kir2 channels in the plasma membranes of mammalian cell lines and isolated cardiac and skeletal muscle cells. We further demonstrate that activation of this pathway can either partly restore (40% of cases) or further impair (20% of cases) the function of mutant ATS channels, depending on the particular Kir2.1 mutation. This means that glucocorticoid treatment might either alleviate or deteriorate symptoms of ATS depending on the patient's individual Kir2.1 genotype. Thus, our findings provide a possible explanation for the contradictory effects of glucocorticoid treatment on symptoms in patients with ATS and may open new pathways for the design of personalized medicines in ATS therapy.


Asunto(s)
Síndrome de Andersen/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Síndrome de Andersen/tratamiento farmacológico , Síndrome de Andersen/genética , Animales , Femenino , Glucocorticoides/uso terapéutico , Cobayas , Células HEK293 , Células HeLa , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Técnicas In Vitro , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Miocitos Cardíacos/metabolismo , Oocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Estrés Fisiológico , Xenopus laevis
8.
Biochem Biophys Res Commun ; 384(1): 28-31, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19371729

RESUMEN

Scaffolding growth factor receptor-bound (Grb) adaptor proteins are components of macromolecular signaling complexes at the plasma membrane and thus are putative regulators of ion channel activity. The present study aimed to define the impact of Grb adaptor proteins on the function of cardiac K(+) channels. To this end channel proteins were coinjected with the adaptor proteins in Xenopus oocytes and channel activity analyzed with two-electrode voltage-clamp. It is shown that coexpression of Grb adaptor proteins can reduce current amplitudes of coexpressed channels. Grb7 and 10 significantly inhibited functional currents generated by hERG, Kv1.5 and Kv4.3 channels. Only Grb10 significantly inhibited KCNQ1/KCNE1 K(+) channels, and only Grb7 reduced Kir2.3 activity, whereas neither Grb protein significantly affected the closely related Kir2.1 and Kir2.2 channels. The present observations for the first time provide evidence for a selective and modulatory role of Grb adaptor proteins in the functional expression of cardiac K(+) channels.


Asunto(s)
Proteína Adaptadora GRB10/metabolismo , Proteína Adaptadora GRB7/metabolismo , Canales de Potasio de Rectificación Interna/biosíntesis , Canales de Potasio con Entrada de Voltaje/biosíntesis , Animales , Proteína Adaptadora GRB10/genética , Proteína Adaptadora GRB7/genética , Humanos , Miocardio/metabolismo , Oocitos , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio con Entrada de Voltaje/genética , Transfección , Xenopus laevis
9.
Cell Physiol Biochem ; 20(6): 791-800, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17982261

RESUMEN

In this study, we analysed the inhibitory potency, blocking characteristics and putative binding sites of three structurally distinct Kv1.5 channel inhibitors on cloned human Kv1.5 channels. Obtained IC(50) values for S9947, MSD-D and ICAGEN-4 were 0.7 microM, 0.5 microM, and 1.6 microM, respectively. The Hill-coefficients were close to 1 for S9947 and approximately 2 for MSD-D and ICAGEN-4. All three compounds inhibited Kv1.5 channels preferentially in the open state, with Kv1.5 block displaying positive frequency dependence, but no clear voltage and potassium dependence. In contrast to slow on- and off-rates of apparent binding of MSD-D and ICAGEN-4, S9947 had fast on- and off-rates resulting in faster adaptation to changes in pulse frequency. Utilizing Alanine-scanning and in silico modeling we suggest binding of the compounds to the central cavity with crucial residues Ile508 and Val512 in the S6-segment. Residue Thr480 located at the base of the selectivity filter is important for ICAGEN-4 and S9947 inhibition, but less so for MSD-D binding. Our docking models suggest that the innermost potassium ion in the selectivity filter may form a tertiary complex with oxygens of S9947 and ICAGEN-4 and residue Thr480. This binding component is absent in the MSD-D block. As S9947 and ICAGEN-4 show faster block with proceeding channel openings, formation of this tertiary complex may increasingly stabilise binding of S9947 and ICAGEN-4, thereby explaining open channel block kinetics of these compounds.


Asunto(s)
Canal de Potasio Kv1.5/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Sitios de Unión , Compuestos de Bifenilo/química , Compuestos de Bifenilo/metabolismo , Compuestos de Bifenilo/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Activación del Canal Iónico/efectos de los fármacos , Cinética , Potasio/farmacología , Bloqueadores de los Canales de Potasio/química , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacología , Factores de Tiempo
10.
J Cardiovasc Pharmacol ; 49(4): 197-206, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17438404

RESUMEN

The novel compound AVE1231 was investigated in order to elucidate its potential against atrial fibrillation. In CHO cells, the current generated by hKv1.5 or hKv4.3 + KChIP2.2b channels was blocked with IC50 values of 3.6 microM and 5.9 microM, respectively. In pig left atrial myocytes, a voltage-dependent outward current was blocked with an IC50 of 1.1 microM, mainly by accelerating the time constant of decay. Carbachol-activated IKACh was blocked by AVE1231 with an IC50 of 8.4 microM. Other ionic currents, like the IKr, IKs, IKATP, ICa, and INa were only mildly affected by 10 microM AVE1231. In guinea pig papillary muscle the APD90 and the upstroke velocity were not significantly altered by 30 microM AVE1231. In anesthetized pigs, oral doses of 0.3, 1, and 3 mg/kg AVE1231 caused a dose-dependent increase in left atrial refractoriness (LAERP), associated by inhibition of left atrial vulnerability to arrhythmia. There were no effects on the ECG intervals, ventricular monophasic action potentials, or ventricular refractory periods at 3 mg/kg AVE1231 applied intravenously. In conscious goats, both AVE1231 (3 mg/kg/h iv) and dofetilide (10 microg/kg/h iv) significantly prolonged LAERP. After 72 hours of tachypacing, when LAERP was shortened significantly (electrical remodelling), the prolongation of LAERP induced by AVE1231 was even more pronounced than in sinus rhythm. In contrast, the effect of dofetilide was strongly decreased. The present data demonstrate that AVE1231 blocks early atrial K channels and prolongs atrial refractoriness with no effects on ECG intervals and ventricular repolarisation, suggesting that it is suited for the prevention of atrial fibrillation in patients.


Asunto(s)
Antiarrítmicos/farmacología , Arritmias Cardíacas/tratamiento farmacológico , Potenciales de Acción/efectos de los fármacos , Análisis de Varianza , Animales , Arritmias Cardíacas/fisiopatología , Función Atrial/efectos de los fármacos , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Células CHO , Carbacol/farmacología , Cardiotónicos/farmacología , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Técnicas Electrofisiológicas Cardíacas , Femenino , Cabras , Cobayas , Atrios Cardíacos/citología , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Humanos , Concentración 50 Inhibidora , Canales Iónicos/efectos de los fármacos , Canales Iónicos/metabolismo , Masculino , Miocitos Cardíacos/efectos de los fármacos , Músculos Papilares/efectos de los fármacos , Músculos Papilares/fisiopatología , Técnicas de Placa-Clamp , Fenetilaminas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Periodo Refractario Electrofisiológico/efectos de los fármacos , Sulfonamidas/farmacología , Porcinos , Factores de Tiempo
11.
Mol Pharmacol ; 71(6): 1503-11, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17347319

RESUMEN

The chromanol 293B (293B, trans-6-cyano-4-(N-ethylsulfonyl-N-methylamino)-3-hydroxy-2,2-dimethyl-chroman) is a lead compound of potential class III antiarrhythmics that inhibit cardiac I(Ks) potassium channels. These channels are formed by the coassembly of KCNQ1 (Kv7.1, KvLQT1) and KCNE1 subunits. Although homomeric KCNQ1 channels are the principal molecular targets, entry of KCNE1 to the channel complex enhances the chromanol block. Because closely related neuronal KCNQ2 potassium channels are insensitive to the drug, we used KCNQ1/KCNQ2 chimeras to identify the binding site of the inhibitor. We localized the putative drug receptor to the H5 selectivity filter and the S6 transmembrane segment. Single residues affecting 293B inhibition were subsequently identified through systematic exchange of amino acids that were either different in KCNQ1 and KCNQ2 or predicted by a docking model of 293B in the open and closed conformation of KCNQ1. Mutant channel proteins T312S, I337V, and F340Y displayed dramatically lowered sensitivity to chromanol block. The predicted drug binding receptor lies in the inner pore vestibule containing the lower part of the selectivity filter, and the S6 transmembrane domain also reported to be important for binding of benzodiazepines. We propose that the block of the ion permeation pathway involves hydrophobic interactions with the S6 transmembrane residues Ile337 and Phe340, and stabilization of chromanol 293B binding through electrostatic interactions of its oxygen atoms with the most internal potassium ion within the selectivity filter.


Asunto(s)
Cromanos/farmacología , Canal de Potasio KCNQ1/metabolismo , Sulfonamidas/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Canal de Potasio KCNQ1/efectos de los fármacos , Modelos Moleculares , Datos de Secuencia Molecular , Canales de Potasio con Entrada de Voltaje/metabolismo , Homología de Secuencia de Aminoácido , Xenopus laevis
12.
Cell Physiol Biochem ; 17(1-2): 1-12, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16543716

RESUMEN

The weak inward rectifier potassium channel ROMK is important for water and salt reabsorption in the kidney. Here we identified Golgin-160 as a novel interacting partner of the ROMK channel. By using yeast two-hybrid assays and co-immunoprecipitations from transfected cells, we demonstrate that Golgin-160 associates with the ROMK C-terminus. Immunofluorescence microscopy confirmed that both proteins are co-localized in the Golgi region. The interaction was further confirmed by the enhancement of ROMK currents by the co-expressed Golgin-160 in Xenopus oocytes. The increase in ROMK current amplitude was due to an increase in cell surface density of ROMK protein. Golgin-160 also stimulated current amplitudes of the related Kir2.1, and of voltage-gated Kv1.5 and Kv4.3 channels, but not the current amplitude of co-expressed HERG channel. These results demonstrate that the Golgi-associated Golgin-160 recognizes the cytoplasmic C-terminus of ROMK, thereby facilitating the transport of ROMK to the cell surface. However, the stimulatory effect on the activity of more distantly-related potassium channels suggests a more general role of Golgin-160 in the trafficking of plasma membrane proteins.


Asunto(s)
Autoantígenos/metabolismo , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Secuencia de Aminoácidos , Animales , Autoantígenos/genética , Sitios de Unión , Transporte Biológico Activo , Células COS , Línea Celular , Chlorocebus aethiops , Femenino , Biblioteca de Genes , Proteínas de la Matriz de Golgi , Humanos , Técnicas In Vitro , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Complejos Multiproteicos , Oocitos/metabolismo , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Técnicas del Sistema de Dos Híbridos , Xenopus laevis
13.
Pflugers Arch ; 450(4): 201-8, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15877233

RESUMEN

The purpose of our investigation was to identify the transcriptional basis for ultrastructural and functional specialization of human atria and ventricles. Using exploratory microarray analysis (Affymetrix U133A+B), we detected 11,740 transcripts expressed in human heart, representing the most comprehensive report of the human myocardial transcriptome to date. Variation in gene expression between atria and ventricles accounted for the largest differences in this data set, as 3.300 and 2.974 transcripts showed higher expression in atria and ventricles, respectively. Functional classification based on Gene Ontology identified chamber-specific patterns of gene expression and provided molecular insights into the regional specialization of cardiomyocytes, correlating important functional pathways to transcriptional activity: Ventricular myocytes preferentially express genes satisfying contractile and energetic requirements, while atrial myocytes exhibit specific transcriptional activities related to neurohumoral function. In addition, several pro-fibrotic and apoptotic pathways were concentrated in atrial myocardium, substantiating the higher susceptibility of atria to programmed cell death and extracellular matrix remodelling observed in human and experimental animal models of heart failure. Differences in transcriptional profiles of atrial and ventricular myocardium thus provide molecular insights into myocardial cell diversity and distinct region-specific adaptations to physiological and pathophysiological conditions. Moreover, as major functional classes of atrial- and ventricular-specific transcripts were common to human and murine myocardium, an evolutionarily conserved chamber-specific expression pattern in mammalian myocardium is suggested.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Animales , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
14.
Circ Res ; 96(9): 1022-9, 2005 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-15817885

RESUMEN

Atrial fibrillation is associated with increased expression of ventricular myosin isoforms in atrial myocardium, regarded as part of a dedifferentiation process. Whether reexpression of ventricular isoforms in atrial fibrillation is restricted to transcripts encoding for contractile proteins is unknown. Therefore, this study compares atrial mRNA expression in patients with permanent atrial fibrillation to atrial mRNA expression in patients with sinus rhythm and to ventricular gene expression using Affymetrix U133 arrays. In atrial myocardium, we identified 1434 genes deregulated in atrial fibrillation, the majority of which, including key elements of calcium-dependent signaling pathways, displayed downregulation. Functional classification based on Gene Ontology provided the specific gene sets of the interdependent processes of structural, contractile, and electrophysiological remodeling. In addition, we demonstrate for the first time a prominent upregulation of transcripts involved in metabolic activities, suggesting an adaptive response to increased metabolic demand in fibrillating atrial myocardium. Ventricular-predominant genes were 5 times more likely to be upregulated in atrial fibrillation (174 genes upregulated, 35 genes downregulated), whereas atrial-specific transcripts were predominantly downregulated (56 genes upregulated, 564 genes downregulated). Overall, in fibrillating atrial myocardium, functional classes of genes characteristic of ventricular myocardium were found to be upregulated (eg, metabolic processes), whereas functional classes predominantly expressed in atrial myocardium were downregulated (eg, signal transduction and cell communication). Therefore, dedifferentiation with adoption of a ventricular-like signature is a general feature of the fibrillating atrium.


Asunto(s)
Fibrilación Atrial/genética , Regulación hacia Abajo , Atrios Cardíacos/metabolismo , Transcripción Genética , Fibrilación Atrial/metabolismo , Calcio/metabolismo , Perfilación de la Expresión Génica , Genoma Humano , Ventrículos Cardíacos/metabolismo , Humanos , ARN Mensajero/metabolismo
15.
J Physiol ; 557(Pt 3): 761-72, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15107477

RESUMEN

Kv4.3 channels conduct transient outward K(+) currents in the human heart and brain where they mediate the early phase of action potential repolarization. KChIP2 proteins are members of a new class of calcium sensors that modulate the surface expression and biophysical properties of Kv4 K(+) channels. Here we describe three novel isoforms of KChIP2 with an alternatively spliced C-terminus (KChIP2e, KChIP2f) or N-terminus (KChIP2g). KChIP2e and KChIP2f are expressed in the human atrium, whereas KChIP2g is predominantly expressed in the brain. The KChIP2 isoforms were coexpressed with Kv4.3 channels in Xenopus oocytes and currents recorded with two-microelectrode voltage-clamp techniques. KChIP2e caused a reduction in current amplitude, an acceleration of inactivation and a slowing of the recovery from inactivation of Kv4.3 currents. KChIP2f increased the current amplitude and slowed the rate of inactivation, but did not alter the recovery from inactivation or the voltage of half-maximal inactivation of Kv4.3 channels. KChIP2g increased current amplitudes, slowed the rate of inactivation and shifted the voltage of half-maximal inactivation to more negative potentials. The biophysical changes induced by these alternatively spliced KChIP2 proteins differ markedly from previously described KChIP2 proteins and would be expected to increase the diversity of native transient outward K(+) currents.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Química Encefálica , Proteínas de Unión al Calcio/antagonistas & inhibidores , Clonación Molecular , ADN Complementario/biosíntesis , ADN Complementario/genética , Estimulación Eléctrica , Electrofisiología , Humanos , Activación del Canal Iónico , Isomerismo , Proteínas de Interacción con los Canales Kv , Mediciones Luminiscentes , Potenciales de la Membrana/fisiología , Datos de Secuencia Molecular , Miocardio/metabolismo , Oocitos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/química , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Potasio Shal , Xenopus laevis
16.
Eur J Pharmacol ; 489(1-2): 13-9, 2004 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-15063150

RESUMEN

We have investigated the effects of the ethacrynic acid derivative 4-(2-butyl-6,7-dichloro-2-cyclopentyl-indan-1-on-5-yl) oxobutyric acid (DCPIB), an inhibitor of the volume-sensitive anion channel (VSAC), on electrical activity and insulin secretion in rat pancreatic beta-cells. DCPIB inhibited whole-cell VSAC currents in beta-cells with IC50 values of 2.2 and 1.7 microM for inhibition of outward and inward currents, respectively. DCPIB also inhibited the VSAC at the single channel level in cells activated by glucose. In intact cells, DCPIB caused a net increase in beta-cell input conductance and evoked an outward current that was sensitive to inhibition by tolbutamide, suggesting KATP channel activation. However, no KATP channel activation was evident under conventional whole-cell conditions, suggesting that the drug might activate the channel in intact cells via an indirect mechanism, possibly involving nutrient metabolism. DCPIB suppressed glucose-induced electrical activity in beta-cells, hyperpolarised the cell membrane potential at a substimulatory glucose concentration and prevented depolarisation when the glucose concentration was raised to stimulatory levels. The suppression of electrical activity by DCPIB was associated with a marked inhibition of glucose-stimulated insulin release from intact islets. It is concluded that DCPIB inhibits electrical and secretory activity in the beta-cell as a combined result of a reciprocal inhibition of VSAC and activation of KATP channel activities, thus producing a marked hyperpolarisation of the beta-cell membrane potential.


Asunto(s)
Ciclopentanos/farmacología , Glucosa/antagonistas & inhibidores , Glucosa/farmacología , Indanos/farmacología , Canales Iónicos/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Electrofisiología , Hipoglucemiantes/farmacología , Técnicas In Vitro , Insulina/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Tolbutamida/farmacología
17.
J Mol Med (Berl) ; 82(5): 308-16, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15103417

RESUMEN

To obtain region- and disease-specific transcription profiles of human myocardial tissue, we explored mRNA expression from all four chambers of eight explanted failing [idiopathic dilated cardiomyopathy (DCM), n=5; ischemic cardiomyopathy (ICM), n=3], and five non-failing hearts using high-density oligonucleotide arrays (Affymetrix U95Av2). We performed pair-wise comparisons of gene expression in the categories (1) atria versus ventricles, (2) disease-regulated genes in atria and (3) disease-regulated genes in ventricles. In the 51 heart samples examined, 549 genes showed divergent distribution between atria and ventricles (272 genes with higher expression in atria, 277 genes with higher expression in ventricles). Two hundred and eighty-eight genes were differentially expressed in failing myocardium compared to non-failing hearts (19 genes regulated in atria and ventricles, 172 regulated in atria only, 97 genes regulated in ventricles only). For disease-regulated genes, down-regulation was 4.5-times more common than up-regulation. Functional classification according to Gene Ontology identified specific biological patterns for differentially expressed genes. Eleven genes were validated by RT-PCR showing a good correlation with the microarray data. Our goal was to determine a gene expression fingerprint of the heart, accounting for region- and disease-specific aspects. Recognizing common gene expression patterns in heart failure will significantly contribute to the understanding of heart failure and may eventually lead to the development of pathway-specific therapies.


Asunto(s)
Gasto Cardíaco Bajo/genética , Cardiomiopatía Dilatada/genética , Perfilación de la Expresión Génica , Isquemia Miocárdica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Adulto , Gasto Cardíaco Bajo/metabolismo , Cardiomiopatía Dilatada/metabolismo , Regulación hacia Abajo , Femenino , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Isquemia Miocárdica/metabolismo , Péptido Natriurético Encefálico , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/genética , Transcripción Genética , Regulación hacia Arriba
18.
J Biol Chem ; 279(1): 394-400, 2004 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-14578345

RESUMEN

Kv1.5 channels conduct the ultrarapid delayed rectifier current (IKur) that contributes to action potential repolarization of human atrial myocytes. Block of these channels has been proposed as a treatment for atrial arrhythmias. Here we report a novel and potent inhibitor of Kv1.5 potassium channels, N-benzyl-N-pyridin-3-yl-methyl-2-(toluene-4-sulfonylamino)-benzamide hydrochloride (S0100176), which exhibits features consistent with preferential block of the open state. The IC50 of S0100176 for Kv1.5 expressed in Xenopus oocytes was 0.7 microm. Ala-scanning mutagenesis within the pore helix and the S6 segment, regions that form the walls of the central cavity, was combined with voltage clamp analysis to identify point mutations that altered drug affinity. This approach identified Thr-479, Thr-480, Val-505, Ile-508, and Val-512 as the most important residues for block by S0100176. Mutations of these key residues to Ala or other amino acids caused marked changes in the IC50 of S0100176 (p<0.01). For example, the IC50 of S0100176 increased 362-fold for T480A, 26-fold for V505A, 150-fold for I508A, and 99-fold for V512A. We used modeling to dock S0100176 into the inner cavity of a Kv1.5 pore homology model that was generated based on the crystal structure of KcsA. The docking predicted that the five residues identified by the Ala scan were positioned less than 4.5 A from the compound. Based on the homology models, the positions of the five amino acids identified to interact with S0100176 face toward the central cavity and overlap with putative binding sites for other blockers and voltage-gated potassium channels.


Asunto(s)
Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/fisiología , Sulfonamidas/farmacología , ortoaminobenzoatos/farmacología , Sustitución de Aminoácidos , Animales , Sitios de Unión , Humanos , Canal de Potasio Kv1.5 , Potenciales de la Membrana/efectos de los fármacos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Oocitos , Canales de Potasio/química , Canales de Potasio/efectos de los fármacos , Canales de Potasio/fisiología , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/genética , Conformación Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Xenopus laevis
19.
Pflugers Arch ; 446(6): 633-40, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12856183

RESUMEN

The HCN4 gene encodes a hyperpolarization-activated cation current contributing to the slow components of the pacemaking currents I(f) in the sinoatrial node and I(h) or I(q) in the thalamus. Heterologous expression studies of individual HCN channels have, however, failed to reproduce fully the diversity of native I(f/h/q) currents, suggesting the presence of modulating auxiliary subunits. Consistent with this is the recent description of KCNE2, which is highly expressed in the sinoatrial node, as a beta-subunit of rapidly activating HCN1 and HCN2 channels. To determine whether KCNE2 can also modulate the slow component of native I(f/h/q) currents, we co-expressed KCNE2 with HCN4 in Xenopus oocytes and in Chinese hamster ovary (CHO) cells and analysed the resulting currents using two-electrode voltage-clamp and patch-clamp techniques, respectively. In both cell types, co-expressed KCNE2 enhanced HCN4-generated current amplitudes, slowed the activation kinetics and shifted the voltage for half-maximal activation of currents to more negative voltages. In contrast, the related family members KCNE1, KCNE3 and KCNE4 did not change current characteristics of HCN4. Consistent with these electrophysiological results, the carboxy-terminal tail of KCNE2, but not of other KCNE subunits, interacted with the carboxy-terminal tail of HCN4 in yeast two-hybrid assays. KCNE2, by modulating I(f) or I(h) currents, might thus contribute to the electrophysiological diversity of known pacemaking currents in the heart and brain.


Asunto(s)
Canales Iónicos/fisiología , Proteínas Musculares/fisiología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/fisiología , Animales , Biotransformación/fisiología , Células CHO , Clonación Molecular , Cricetinae , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Electrofisiología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Cinética , Potenciales de la Membrana/fisiología , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Oocitos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Xenopus laevis
20.
J Am Coll Cardiol ; 41(5): 858-69, 2003 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-12628735

RESUMEN

OBJECTIVES: The purpose of the study was to characterize the ionic and molecular mechanisms in the very early phases of electrical remodeling in a rabbit model of rapid atrial pacing (RAP). BACKGROUND: Long-term atrial fibrillation reduces L-type Ca(2+) (I(Ca,L)) and transient outward K(+) (I(to)) currents by transcriptional downregulation of the underlying ionic channels. However, electrical remodeling starts early after the onset of rapid atrial rates. The time course of ion current and channel modulation in these early phases of remodeling is currently unknown. METHODS: Rapid (600 beats/min) right atrial pacing was performed in rabbits. Animals were divided into five groups with pacing durations between 0 and 96 h. Ionic currents were measured by patch clamp techniques; messenger ribonucleic acid (mRNA) and protein expression were measured by reverse transcription-polymerase chain reaction and Western blot, respectively. RESULTS: L-type calcium current started to be reduced (by 47%) after 12 h of RAP and continued to decline as pacing continued. Current changes were preceded or paralleled by decreased mRNA expression of the Ca(2+) channel beta subunits CaB2a, CaB2b, and CaB3, whereas significant reductions in the alpha(1) subunit mRNA and protein expression began 24 h after pacing onset. Transient outward potassium current densities were not altered within the first 12 h, but after 24 h, currents were reduced by 48%. Longer pacing periods did not further decrease I(to). Current changes were paralleled by reduced Kv4.3 mRNA expression. Kv4.2, Kv1.4, and the auxiliary subunit KChIP2 were not affected. CONCLUSIONS: L-type calcium current and I(to) are reduced in early phases of electrical remodeling. A major mechanism appears to be transcriptional downregulation of underlying ion channels, which partially preceded ion current changes.


Asunto(s)
Fibrilación Atrial/terapia , Canales de Calcio Tipo L/metabolismo , Transporte Iónico/fisiología , Canales de Potasio/metabolismo , ARN Mensajero/análisis , Análisis de Varianza , Animales , Fibrilación Atrial/patología , Secuencia de Bases , Western Blotting , Canales de Calcio Tipo L/análisis , Estimulación Cardíaca Artificial , Modelos Animales de Enfermedad , Regulación hacia Abajo , Conductividad Eléctrica , Electrofisiología , Femenino , Masculino , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Canales de Potasio/análisis , Probabilidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...