Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 318(6): H1357-H1370, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32196358

RESUMEN

Synapse-associated protein 97 (SAP97) is a scaffolding protein crucial for the functional expression of several cardiac ion channels and therefore proper cardiac excitability. Alterations in the functional expression of SAP97 can modify the ionic currents underlying the cardiac action potential and consequently confer susceptibility for arrhythmogenesis. In this study, we generated a murine model for inducible, cardiac-targeted Sap97 ablation to investigate arrhythmia susceptibility and the underlying molecular mechanisms. Furthermore, we sought to identify human SAP97 (DLG1) variants that were associated with inherited arrhythmogenic disease. The murine model of cardiac-specific Sap97 ablation demonstrated several ECG abnormalities, pronounced action potential prolongation subject to high incidence of arrhythmogenic afterdepolarizations and notable alterations in the activity of the main cardiac ion channels. However, no DLG1 mutations were found in 40 unrelated cases of genetically elusive long QT syndrome (LQTS). Instead, we provide the first evidence implicating a gain of function in human DLG1 mutation resulting in an increase in Kv4.3 current (Ito) as a novel, potentially pathogenic substrate for Brugada syndrome (BrS). In conclusion, DLG1 joins a growing list of genes encoding ion channel interacting proteins (ChIPs) identified as potential channelopathy-susceptibility genes because of their ability to regulate the trafficking, targeting, and modulation of ion channels that are critical for the generation and propagation of the cardiac electrical impulse. Dysfunction in these critical components of cardiac excitability can potentially result in fatal cardiac disease.NEW & NOTEWORTHY The gene encoding SAP97 (DLG1) joins a growing list of genes encoding ion channel-interacting proteins (ChIPs) identified as potential channelopathy-susceptibility genes because of their ability to regulate the trafficking, targeting, and modulation of ion channels that are critical for the generation and propagation of the cardiac electrical impulse. In this study we provide the first data supporting DLG1-encoded SAP97's candidacy as a minor Brugada syndrome susceptibility gene.


Asunto(s)
Arritmias Cardíacas/metabolismo , Homólogo 1 de la Proteína Discs Large/metabolismo , Corazón/fisiopatología , Miocardio/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Homólogo 1 de la Proteína Discs Large/genética , Humanos , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo
2.
Heart Fail Clin ; 12(2): 157-66, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26968662

RESUMEN

Atrial fibrillation (AF) is by far the most common sustained tachyarrhythmia, affecting 1% to 2% of the general population. AF prevalence and the total annual cost for treatment are alarming, emphasizing the need for an urgent attention to the problem. Thus, having up-to-date information on AF risk factors and appreciating how they promote maintenance of AF maintenance are essential. This article presents a simplified examination of AF risk factors, including emerging genetic risks.

3.
Proc Natl Acad Sci U S A ; 109(31): E2134-43, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22509027

RESUMEN

The cardiac electrical impulse depends on an orchestrated interplay of transmembrane ionic currents in myocardial cells. Two critical ionic current mechanisms are the inwardly rectifying potassium current (I(K1)), which is important for maintenance of the cell resting membrane potential, and the sodium current (I(Na)), which provides a rapid depolarizing current during the upstroke of the action potential. By controlling the resting membrane potential, I(K1) modifies sodium channel availability and therefore, cell excitability, action potential duration, and velocity of impulse propagation. Additionally, I(K1)-I(Na) interactions are key determinants of electrical rotor frequency responsible for abnormal, often lethal, cardiac reentrant activity. Here, we have used a multidisciplinary approach based on molecular and biochemical techniques, acute gene transfer or silencing, and electrophysiology to show that I(K1)-I(Na) interactions involve a reciprocal modulation of expression of their respective channel proteins (Kir2.1 and Na(V)1.5) within a macromolecular complex. Thus, an increase in functional expression of one channel reciprocally modulates the other to enhance cardiac excitability. The modulation is model-independent; it is demonstrable in myocytes isolated from mouse and rat hearts and with transgenic and adenoviral-mediated overexpression/silencing. We also show that the post synaptic density, discs large, and zonula occludens-1 (PDZ) domain protein SAP97 is a component of this macromolecular complex. We show that the interplay between Na(v)1.5 and Kir2.1 has electrophysiological consequences on the myocardium and that SAP97 may affect the integrity of this complex or the nature of Na(v)1.5-Kir2.1 interactions. The reciprocal modulation between Na(v)1.5 and Kir2.1 and the respective ionic currents should be important in the ability of the heart to undergo self-sustaining cardiac rhythm disturbances.


Asunto(s)
Potenciales de Acción , Arritmias Cardíacas/mortalidad , Regulación de la Expresión Génica , Potenciales de la Membrana , Proteínas Musculares/biosíntesis , Miocitos Cardíacos/metabolismo , Canales de Potasio de Rectificación Interna/biosíntesis , Canales de Sodio/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Homólogo 1 de la Proteína Discs Large , Silenciador del Gen , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Miocitos Cardíacos/patología , Canal de Sodio Activado por Voltaje NAV1.5 , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Canales de Potasio de Rectificación Interna/genética , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Canales de Sodio/genética , Proteína de la Zonula Occludens-1
4.
Circ Res ; 111(7): 842-53, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22843785

RESUMEN

RATIONALE: Kv1.5 (KCNA5) is expressed in the heart, where it underlies the I(Kur) current that controls atrial repolarization, and in the pulmonary vasculature, where it regulates vessel contractility in response to changes in oxygen tension. Atrial fibrillation and hypoxic pulmonary hypertension are characterized by downregulation of Kv1.5 protein expression, as well as with oxidative stress. Formation of sulfenic acid on cysteine residues of proteins is an important, dynamic mechanism for protein regulation under oxidative stress. Kv1.5 is widely reported to be redox-sensitive, and the channel possesses 6 potentially redox-sensitive intracellular cysteines. We therefore hypothesized that sulfenic acid modification of the channel itself may regulate Kv1.5 in response to oxidative stress. OBJECTIVE: To investigate how oxidative stress, via redox-sensitive modification of the channel with sulfenic acid, regulates trafficking and expression of Kv1.5. METHODS AND RESULTS: Labeling studies with the sulfenic acid-specific probe DAz and horseradish peroxidase-streptavidin Western blotting demonstrated a global increase in sulfenic acid-modified proteins in human patients with atrial fibrillation, as well as sulfenic acid modification to Kv1.5 in the heart. Further studies showed that Kv1.5 is modified with sulfenic acid on a single COOH-terminal cysteine (C581), and the level of sulfenic acid increases in response to oxidant exposure. Using live-cell immunofluorescence and whole-cell voltage-clamping, we found that modification of this cysteine is necessary and sufficient to reduce channel surface expression, promote its internalization, and block channel recycling back to the cell surface. Moreover, Western blotting demonstrated that sulfenic acid modification is a trigger for channel degradation under prolonged oxidative stress. CONCLUSIONS: Sulfenic acid modification to proteins, which is elevated in diseased human heart, regulates Kv1.5 channel surface expression and stability under oxidative stress and diverts channel from a recycling pathway to degradation. This provides a molecular mechanism linking oxidative stress and downregulation of channel expression observed in cardiovascular diseases.


Asunto(s)
Fibrilación Atrial/metabolismo , Canal de Potasio Kv1.5/química , Canal de Potasio Kv1.5/metabolismo , Miocardio/metabolismo , Ácidos Sulfénicos/metabolismo , Secuencia de Aminoácidos , Animales , Fibrilación Atrial/patología , Estudios de Casos y Controles , Línea Celular , Células Cultivadas , Humanos , Ratones , Modelos Animales , Datos de Secuencia Molecular , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxidación-Reducción , Estrés Oxidativo/fisiología , Ratas , Especies Reactivas de Oxígeno , Transducción de Señal/fisiología
6.
J Biol Chem ; 285(36): 28000-9, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20530486

RESUMEN

Synapse-associated protein-97 (SAP97) is a membrane-associated guanylate kinase scaffolding protein expressed in cardiomyocytes. SAP97 has been shown to associate and modulate voltage-gated potassium (Kv) channel function. In contrast to Kv channels, little information is available on interactions involving SAP97 and inward rectifier potassium (Kir2.x) channels that underlie the classical inward rectifier current, I(K1). To investigate the functional effects of silencing SAP97 on I(K1) in adult rat ventricular myocytes, SAP97 was silenced using an adenoviral short hairpin RNA vector. Western blot analysis showed that SAP97 was silenced by approximately 85% on day 3 post-infection. Immunostaining showed that Kir2.1 and Kir2.2 co-localize with SAP97. Co-immunoprecipitation (co-IP) results demonstrated that Kir2.x channels associate with SAP97. Voltage clamp experiments showed that silencing SAP97 reduced I(K1) whole cell density by approximately 55%. I(K1) density at -100 mV was -1.45 +/- 0.15 pA/picofarads (n = 6) in SAP97-silenced cells as compared with -3.03 +/- 0.37 pA/picofarads (n = 5) in control cells. Unitary conductance properties of I(K1) were unaffected by SAP97 silencing. The major mechanism for the reduction of I(K1) density appears to be a decrease in Kir2.x channel abundance. Furthermore, SAP97 silencing impaired I(K1) regulation by beta(1)-adrenergic receptor (beta1-AR) stimulation. In control, isoproterenol reduced I(K1) amplitude by approximately 75%, an effect that was blunted following SAP97 silencing. Our co-IP data show that beta1-AR associates with SAP97 and Kir2.1 and also that Kir2.1 co-IPs with protein kinase A and beta1-AR. SAP97 immunolocalizes with protein kinase A and beta1-AR in the cardiac myocytes. Our results suggest that in cardiac myocytes SAP97 regulates surface expression of channels underlying I(K1), as well as assembles a signaling complex involved in beta1-AR regulation of I(K1).


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Conductividad Eléctrica , Proteínas de la Membrana/metabolismo , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Inmunoprecipitación , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Células Musculares/metabolismo , Transporte de Proteínas , Ratas , Receptores Adrenérgicos beta 1/metabolismo
8.
J Mol Cell Cardiol ; 48(1): 45-54, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19703462

RESUMEN

Cardiac I(K1) and I(KACh) are the major potassium currents displaying classical strong inward rectification, a unique property that is critical for their roles in cardiac excitability. In the last 15 years, research on I(K1) and I(KACh) has been propelled by the cloning of the underlying inwardly rectifying potassium (Kir) channels, the discovery of the molecular mechanism of strong rectification and the linking of a number of disorders of cardiac excitability to defects in genes encoding Kir channels. Disease-causing mutations in Kir genes have been shown experimentally to affect one or more of the following channel properties: structure, assembly, trafficking, and regulation, with the ultimate effect of a gain- or a loss-of-function of the channel. It is now established that I(K1) and I(KACh) channels are heterotetramers of Kir2 and Kir3 subunits, respectively. Each homomeric Kir channel has distinct biophysical and regulatory properties, and individual Kir subunits often display different patterns of regional, cellular, and membrane distribution. These differences are thought to underlie important variations in the physiological properties of I(K1) and I(KACh). It has become increasingly clear that the contribution of I(K1) and I(KACh) channels to cardiac electrical activity goes beyond their long recognized role in the stabilization of resting membrane potential and shaping the late phase of action potential repolarization in individual myocytes but extends to being critical elements determining the overall electrical stability of the heart.


Asunto(s)
Corazón/fisiología , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Animales , Corazón/fisiopatología , Humanos , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Modelos Biológicos , Miocardio/patología , Canales de Potasio de Rectificación Interna/genética
9.
Biophys J ; 96(7): 2961-76, 2009 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-19348777

RESUMEN

The mechanisms controlling the rotation frequency of functional reentry in ventricular fibrillation (VF) are poorly understood. It has been previously shown that Ba2+ at concentrations up to 50 mumol/L slows the rotation frequency in the intact guinea pig (GP) heart, suggesting a role of the inward rectifier current (I(K1)) in the mechanism governing the VF response to Ba2+. Given that other biological (e.g., sinoatrial node) and artificial systems display phase-locking behavior, we hypothesized that the mechanism for controlling the rotation frequency of a rotor by I(K1) blockade is phase-driven, i.e., the phase shift between transmembrane current and voltage remains constant at varying levels of I(K1) blockade. We measured whole-cell admittance in isolated GP myocytes and in transfected human embryonic kidney (HEK) cells stably expressing Kir 2.1 and 2.3 channels. The admittance phase, i.e., the phase difference between current and voltage, was plotted versus the frequency in control conditions and at 10 or 50 micromol/L Ba2+ (in GP heart cells) or 1 mM Ba2+ (in HEK cells). The horizontal distance between plots was called the "frequency shift in a single cell" and analyzed. The frequency shift in a single cell was -14.14 +/- 5.71 Hz (n = 14) at 10 microM Ba2+ and -18.51 +/- 4.00 Hz (n = 10) at 50 microM Ba2+, p < 0.05. The values perfectly matched the Ba2+-induced reduction of VF frequency observed previously in GP heart. A similar relationship was found in the computer simulations. The phase of Ba2+-sensitive admittance in GP cells was -2.65 +/- 0.32 rad at 10 Hz and -2.79 +/- 0.26 rad at 30 Hz. In HEK cells, the phase of Ba2+-sensitive admittance was 3.09 +/- 0.03 rad at 10 Hz and 3.00 +/- 0.17 rad at 30 Hz. We have developed a biological single-cell model of rotation-frequency control. The results show that although rotation frequency changes as a result of I(K1) blockade, the phase difference between transmembrane current and transmembrane voltage remains constant, enabling us to quantitatively predict the change of VF frequency resulting from I(K1) blockade, based on single-cell measurement.


Asunto(s)
Modelos Biológicos , Fibrilación Ventricular/patología , Animales , Bario/farmacología , Línea Celular , Fenómenos Fisiológicos Celulares/efectos de los fármacos , Simulación por Computador , Conductividad Eléctrica , Cobayas , Humanos , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Células Musculares/patología , Sensibilidad y Especificidad , Fibrilación Ventricular/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 297(4): H1387-97, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19633205

RESUMEN

We examined the impact of coexpressing the inwardly rectifying potassium channel, Kir2.3, with the scaffolding protein, synapse-associated protein (SAP) 97, and determined that coexpression of these proteins caused an approximately twofold increase in current density. A combination of techniques was used to determine if the SAP97-induced increase in Kir2.3 whole cell currents resulted from changes in the number of channels in the cell membrane, unitary channel conductance, or channel open probability. In the absence of SAP97, Kir2.3 was found predominantly in a cytoplasmic, vesicular compartment with relatively little Kir2.3 localized to the plasma membrane. The introduction of SAP97 caused a redistribution of Kir2.3, leading to prominent colocalization of Kir2.3 and SAP97 and a modest increase in cell surface Kir2.3. The median Kir2.3 single channel conductance in the absence of SAP97 was approximately 13 pS, whereas coexpression of SAP97 led to a wide distribution of channel events with three distinct peaks centered at 16, 29, and 42 pS. These changes occurred without altering channel open probability, current rectification properties, or pH sensitivity. Thus association of Kir2.3 with SAP97 in HEK293 cells increased channel cell surface expression and unitary channel conductance. However, changes in single channel conductance play the major role in determining whole cell currents in this model system. We further suggest that the SAP97 effect results from SAP97 binding to the Kir2.3 COOH-terminal domain and altering channel conformation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Activación del Canal Iónico , Proteínas de la Membrana/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Sitios de Unión , Línea Celular , Membrana Celular/metabolismo , Vesículas Citoplasmáticas/metabolismo , Cobayas , Atrios Cardíacos/metabolismo , Humanos , Potenciales de la Membrana , Proteínas de la Membrana/genética , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/genética , Conformación Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Ovinos , Transfección
11.
Front Physiol ; 9: 2, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29403390

RESUMEN

Anatomical evidence in several species shows highly heterogeneous fat distribution in the atrial and ventricular myocardium. Atrial appendages have fat deposits, and more so on the posterior left atrium. Although such fat distributions are considered normal, fatty infiltration is regarded arrhythmogenic, and various cardiac pathophysiological conditions show excess myocardial fat deposits, especially in the epicardium. Hypotheses have been presented for the physiological and pathophysiological roles of epicardial fat, however this issue is poorly understood. Therefore, this mini-review will focus on epicardial fat distribution and the (patho)-physiological implications of this distribution. Potential molecular mechanisms that may drive structural and electrical myocardial remodeling attendant to fatty infiltration of the heart are also reviewed.

12.
Circ Arrhythm Electrophysiol ; 11(3): e005659, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29540372

RESUMEN

BACKGROUND: The mechanisms underlying spontaneous atrial fibrillation (AF) associated with atrial ischemia/infarction are incompletely elucidated. Here, we investigate the mechanisms underlying spontaneous AF in an ovine model of left atrial myocardial infarction (LAMI). METHODS AND RESULTS: LAMI was created by ligating the atrial branch of the left anterior descending coronary artery. ECG loop recorders were implanted to monitor AF episodes. In 7 sheep, dantrolene-a ryanodine receptor blocker-was administered in vivo during the 8-day observation period (LAMI-D, 2.5 mg/kg, IV, BID). LAMI animals experienced numerous spontaneous AF episodes during the 8-day monitoring period that were suppressed by dantrolene (LAMI, 26.1±5.1; sham, 4.3±1.1; LAMI-D, 2.8±0.8; mean±SEM episodes per sheep, P<0.01). Optical mapping showed spontaneous focal discharges (SFDs) originating from the ischemic/normal-zone border. SFDs were calcium driven, rate dependent, and enhanced by isoproterenol (0.03 µmol/L, from 210±87 to 3816±1450, SFDs per sheep) but suppressed by dantrolene (to 55.8±32.8, SFDs per sheep, mean±SEM). SFDs initiated AF-maintaining reentrant rotors anchored by marked conduction delays at the ischemic/normal-zone border. NOS1 (NO synthase-1) protein expression decreased in ischemic zone myocytes, whereas NADPH (nicotinamide adenine dinucleotide phosphate, reduced form) oxidase and xanthine oxidase enzyme activities and reactive oxygen species (DCF [6-carboxy-2',7'-dichlorodihydrofluorescein diacetate]-fluorescence) increased. CaM (calmodulin) aberrantly increased [3H]ryanodine binding to cardiac RyR2 (ryanodine receptors) in the ischemic zone. Dantrolene restored the physiological binding of CaM to RyR2. CONCLUSIONS: Atrial ischemia causes spontaneous AF episodes in sheep, caused by SFDs that initiate reentry. Nitroso-redox imbalance in the ischemic zone is associated with intense reactive oxygen species production and altered RyR2 responses to CaM. Dantrolene administration normalizes the CaM response, prevents LAMI-related SFDs, and AF initiation. These findings provide novel insights into the mechanisms underlying ischemia-related atrial arrhythmias.


Asunto(s)
Fibrilación Atrial/complicaciones , Dantroleno/farmacología , Isquemia Miocárdica/etiología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/terapia , Western Blotting , Señalización del Calcio , Modelos Animales de Enfermedad , Atrios Cardíacos , Masculino , Relajantes Musculares Centrales/farmacología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Ovinos
13.
Heart Rhythm ; 4(4): 487-96, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17399639

RESUMEN

BACKGROUND: Data on pH regulation of the cardiac potassium current I(K1) suggest species-dependent differences in the molecular composition of the underlying Kir2 channel proteins. OBJECTIVE: The purpose of this study was to test the hypothesis that the presence of the Kir2.3 isoform in heterotetrameric channels modifies channel sensitivity to pH. METHODS: Voltage clamp was performed on HEK293 cells stably expressing guinea pig Kir2.1 and/or Kir2.3 isoforms and on sheep cardiac ventricular myocytes at varying extracellular pH (pH(o)) and in the presence of CO(2) to determine the sensitivity of macroscopic currents to pH. Single-channel activity was recorded from the HEK293 stables to determine the mechanisms of the changes in whole cell current. RESULTS: Biophysical characteristics of whole-cell and single-channel currents in Kir2.1/Kir2.3 double stables displayed properties attributable to isoform heteromerization. Whole-cell Kir2.1/Kir2.3 currents rectified in a manner reminiscent of Kir2.1 but were significantly inhibited by extracellular acidification in the physiologic range (pK(a) approximately 7.4). Whole-cell currents were more sensitive to a combined extracellular and intracellular acidification produced by CO(2). At pH(o) = 6.0, unitary conductances of heteromeric channels were reduced. Ovine cardiac ventricular cell I(K1) was pH(o) and CO(2) sensitive, consistent with the expression of Kir2.1 and Kir2.3 in this species. CONCLUSION: Kir2.1 and Kir2.3 isoforms form heteromeric channels in HEK293. The presence of Kir2.3 subunit(s) in heteromeric channels confers pH sensitivity to the channels. The single and double stable cells presented in this study are useful models for studying physiologic regulation of heteromeric Kir2 channels in mammalian cells.


Asunto(s)
Miocitos Cardíacos/fisiología , Canales de Potasio de Rectificación Interna/fisiología , Análisis de Varianza , Animales , Western Blotting , Línea Celular , Electrofisiología , Cobayas , Ventrículos Cardíacos/citología , Concentración de Iones de Hidrógeno , Modelos Animales , Oocitos/fisiología , Técnicas de Placa-Clamp , Isoformas de Proteínas , Proyectos de Investigación , Ovinos , Xenopus
14.
Circ Res ; 94(10): 1332-9, 2004 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-15087421

RESUMEN

The inwardly rectifying potassium (Kir) 2.x channels mediate the cardiac inward rectifier potassium current (I(K1)). In addition to differences in current density, atrial and ventricular I(K1) have differences in outward current profiles and in extracellular potassium ([K+]o) dependence. The whole-cell patch-clamp technique was used to study these properties in heterologously expressed Kir2.x channels and atrial and ventricular I(K1) in guinea pig and sheep hearts. Kir2.x channels showed distinct rectification profiles: Kir2.1 and Kir2.2 rectified completely at potentials more depolarized than -30 mV (I approximately 0 pA). In contrast, rectification was incomplete for Kir2.3 channels. In guinea pig atria, which expressed mainly Kir2.1, I(K1) rectified completely. In sheep atria, which predominantly expressed Kir2.3 channels, I(K1) did not rectify completely. Single-channel analysis of sheep Kir2.3 channels showed a mean unitary conductance of 13.1+/-0.1 pS in 15 cells, which corresponded with I(K1) in sheep atria (9.9+/-0.1 pS in 32 cells). Outward Kir2.1 currents were increased in 10 mmol/L [K+]o, whereas Kir2.3 currents did not increase. Correspondingly, guinea pig (but not sheep) atrial I(K1) showed an increase in outward currents in 10 mmol/L [K+]o. Although the ventricles of both species expressed Kir2.1 and Kir2.3, outward I(K1) currents rectified completely and increased in high [K+]o-displaying Kir2.1-like properties. Likewise, outward current properties of heterologously expressed Kir2.1-Kir2.3 complexes in normal and 10 mmol/L [K+]o were similar to Kir2.1 but not Kir2.3. Thus, unique properties of individual Kir2.x isoforms, as well as heteromeric Kir2.x complexes, determine regional and species differences of I(K1) in the heart.


Asunto(s)
Función Atrial , Canales de Potasio de Rectificación Interna/metabolismo , Función Ventricular , Animales , Línea Celular , Conductividad Eléctrica , Cobayas , Atrios Cardíacos/citología , Ventrículos Cardíacos/citología , Humanos , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Isoformas de Proteínas/metabolismo , Ovinos , Especificidad de la Especie
15.
Cardiovasc Res ; 59(4): 863-73, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14553826

RESUMEN

OBJECTIVE: We tested the hypothesis that left atrial (LA) myocytes are more sensitive to acetylcholine (ACh) than right atrial (RA) myocytes, which results in a greater dose-dependent increase in LA than RA rotor frequency, increased LA-to-RA frequency gradient and increased incidence of wavelet formation during atrial fibrillation (AF). METHODS AND RESULTS: AF was induced in seven Langendorff-perfused sheep hearts in the presence of ACh (0.1-4.0 microM) and studied using optical mapping and bipolar recordings. Dominant frequencies (DFs) were determined in optical and electrical signals and phase movies were used to identify rotors and quantify their dynamics. DFs in both atria increased monotonically with ACh concentration until saturation, but the LA frequency predominated at all concentrations. Rotors were also seen more often in the LA, and although their life span decreased, their frequency and number of rotations increased. Patch-clamp studies demonstrated that ACh-activated potassium current (I(K,ACh)) density was greater in LA than RA sheep myocytes. Additionally, ribonuclease protection assay demonstrated that Kir3.4 and Kir3.1 mRNAs were more abundant in LA than in RA. CONCLUSIONS: A greater abundance of Kir3.x channels and higher I(K,ACh) density in LA than RA myocytes result in greater ACh-induced speeding-up of rotors in the LA than in the RA, which explains the ACh dose-dependent changes in overall AF frequency and wavelet formation.


Asunto(s)
Acetilcolina/farmacología , Fibrilación Atrial/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Electrocardiografía , Atrios Cardíacos , Perfusión , Canales de Potasio/efectos de los fármacos , Ovinos , Procesamiento de Señales Asistido por Computador
16.
Trends Cardiovasc Med ; 25(6): 487-96, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25701094

RESUMEN

The understanding of ionic mechanisms underlying cardiac rhythm disturbances (arrhythmias) is an issue of significance in the medical science community. Several advances in molecular, cellular, and optical techniques in the past few decades have substantially increased our knowledge of ionic mechanisms that are thought to underlie arrhythmias. The application of these techniques in the study of ion channel biophysics and regulatory properties has provided a wealth of information, with some important therapeutic implications for dealing with the disease. In this review, we briefly consider the cellular and tissue manifestations of a number of cardiac rhythm disturbances, while focusing on our current understanding of the ionic current mechanisms that have been implicated in such rhythm disturbances.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Electrofisiología Cardíaca , Canales Iónicos/fisiología , Arritmias Cardíacas/diagnóstico , Automatización , Canales de Calcio/fisiología , Electrocardiografía , Femenino , Humanos , Masculino , Sensibilidad y Especificidad , Canales de Sodio/fisiología
17.
PLoS One ; 10(8): e0133052, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26274906

RESUMEN

BACKGROUND: Epicardial adiposity and plasma levels of free fatty acids (FFAs) are elevated in atrial fibrillation, heart failure and obesity, with potentially detrimental effects on myocardial function. As major components of epicardial fat, FFAs may be abnormally regulated, with a potential to detrimentally modulate electro-mechanical function. The cellular mechanisms underlying such effects of FFAs are unknown. OBJECTIVE: To determine the mechanisms underlying electrophysiological effects of palmitic (PA), stearic (SA) and oleic (OA) FFAs on sheep atrial myocytes. METHODS: We used electrophysiological techniques, numerical simulations, biochemistry and optical imaging to examine the effects of acutely (≤ 15 min), short-term (4-6 hour) or 24-hour application of individual FFAs (10 µM) on isolated ovine left atrial myocytes (LAMs). RESULTS: Acute and short-term incubation in FFAs resulted in no differences in passive or active properties of isolated left atrial myocytes (LAMs). 24-hour application had differential effects depending on the FFA. PA did not affect cellular passive properties but shortened (p<0.05) action potential duration at 30% repolarization (APD30). APD50 and APD80 were unchanged. SA had no effect on resting membrane potential but reduced membrane capacitance by 15% (p<0.05), and abbreviated APD at all values measured (p≤0.001). OA did not significantly affect passive or active properties of LAMs. Measurement of the major voltage-gated ion channels in SA treated LAMs showed a ~60% reduction (p<0.01) of the L-type calcium current (ICa-L) and ~30% reduction (p<0.05) in the transient outward potassium current (ITO). A human atrial cell model recapitulated SA effects on APD. Optical imaging showed that SA incubated for 24 hours altered t-tubular structure in isolated cells (p<0.0001). CONCLUSIONS: SA disrupts t-tubular architecture and remodels properties of membrane ionic currents in sheep atrial myocytes, with potential implications in arrhythmogenesis.


Asunto(s)
Ácidos Grasos no Esterificados/farmacología , Atrios Cardíacos/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Miocardio/citología , Miocardio/metabolismo , Animales , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Electrofisiología , Immunoblotting , Masculino , Ovinos
18.
J Interv Card Electrophysiol ; 9(2): 119-29, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14574022

RESUMEN

A major goal of basic research in cardiac electrophysiology is to understand the mechanisms responsible for ventricular fibrillation (VF). Here we review recent experimental and numerical results, from the ion channel to the organ level, which might lead to a better understanding of the cellular and molecular mechanisms of VF. The discussion centers on data derived from a model of stable VF in the Langendorff-perfused guinea pig heart that demonstrate distinct patterns of organization in the left (LV) and right (RV) ventricles. Analysis of optical mapping data reveals that VF excitation frequencies are distributed throughout the ventricles in clearly demarcated domains. The highest frequency domains are usually found on the anterior wall of the LV, demonstrating that a high frequency reentrant source (a rotor) that remains stationary in the LV is the mechanism that sustains VF in this model. Computer simulations predict that the inward rectifying potassium current (IK1) is an essential determinant of rotor stability and rotation frequency, and patch-clamp results strongly suggest that the outward component of the background current (presumably IK1) of cells in the LV is significantly larger in the LV than in the RV. These data have opened a new and potentially exciting avenue of research on the possible role played by inward rectifier channels in the mechanism of VF and may lead us toward an understanding of its molecular basis and hopefully lead to new preventative approaches.


Asunto(s)
Fibrilación Ventricular/etiología , Potenciales de Acción/fisiología , Animales , Electrofisiología , Sistema de Conducción Cardíaco/patología , Sistema de Conducción Cardíaco/fisiopatología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Miocardio/patología , Canales de Potasio/fisiología , Estados Unidos/epidemiología , Fibrilación Ventricular/fisiopatología
19.
Cardiol Clin ; 32(4): 485-94, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25443231

RESUMEN

Atrial fibrillation (AF) is by far the most common sustained tachyarrhythmia, affecting 1% to 2% of the general population. AF prevalence and the total annual cost for treatment are alarming, emphasizing the need for an urgent attention to the problem. Thus, having up-to-date information on AF risk factors and appreciating how they promote maintenance of AF maintenance are essential. This article presents a simplified examination of AF risk factors, including emerging genetic risks.


Asunto(s)
Fibrilación Atrial , Factores de Edad , Fibrilación Atrial/epidemiología , Fibrilación Atrial/etiología , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/fisiopatología , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipertensión/complicaciones , Hipertensión/fisiopatología , Prevalencia , Factores de Riesgo
20.
Heart Rhythm ; 10(1): 80-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23041576

RESUMEN

BACKGROUND: Collecting electrophysiological and molecular data from the murine conduction system presents technical challenges. Thus, only little advantage has been taken of numerous genetically engineered murine models to study excitation through the cardiac conduction system of the mouse. OBJECTIVE: To develop an approach for isolating murine cardiac Purkinje cells (PCs), to characterize major ionic currents and to use the data to simulate action potentials (APs) recorded from PCs. METHODS: Light microscopy was used to isolate and identify PCs from apical and septal cells. Current and voltage clamp techniques were used to record APs and whole cell currents. We then simulated a PC AP on the basis of our experimental data. RESULTS: APs recorded from PCs were significantly longer than those recorded from ventricular cells. The prominent plateau phase of the PC AP was very negative (≈-40 mV). Spontaneous activity was observed only in PCs. The inward rectifier current demonstrated no significant differences compared to ventricular myocytes (VMs). However, sodium current density was larger, and the voltage-gated potassium current density was significantly less in PCs compared with myocytes. T-type Ca(2+) currents (I(Ca,T)) were present in PCs but not VMs. Computer simulations suggest that I(Ca,T) and cytosolic calcium diffusion significantly modulate AP profile recorded in PCs, as compared to VMs. CONCLUSIONS: Our study provides the first comprehensive ionic profile of murine PCs. The data show unique features of PC ionic mechanisms that govern its excitation process. Experimental data and numerical modeling results suggest that a smaller voltage-gated potassium current and the presence of I(Ca,T) are important determinants of the longer and relatively negative plateau phase of the APs.


Asunto(s)
Potenciales de Acción/fisiología , Ventrículos Cardíacos/citología , Células de Purkinje/fisiología , Animales , Calcio/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Células de Purkinje/metabolismo , Sodio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA