Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Front Oncol ; 11: 747022, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34765551

RESUMEN

A large number of studies have revealed that epigenetics plays an important role in cancer development. However, the currently-developed epigenetic drugs cannot achieve a stable curative effect. Thus, it may be necessary to redefine the role of epigenetics in cancer development. It has been shown that embryonic development and tumor development share significant similarities in terms of biological behavior and molecular expression patterns, and epigenetics may be the link between them. Cell differentiation is likely a manifestation of epigenetic homeostasis at the cellular level. In this article, we introduced the importance of epigenetic homeostasis in cancer development and analyzed the shortcomings of current epigenetic treatment regimens. Understanding the dynamic process of epigenetic homeostasis in organ development can help us characterize cancer according to its differentiation stages, explore new targets for cancer treatment, and improve the clinical prognosis of patients with cancer.

2.
Zhongguo Yi Liao Qi Xie Za Zhi ; 41(1): 13-6, 2017 Jan.
Artículo en Chino | MEDLINE | ID: mdl-29792645

RESUMEN

This paper presents a new, simple and fast algorithm of automated P wave detection in multi-lead ECG Signals. Range of QRS-T complex is detected firstly. Then QRS-T complex is eliminated. Final y, P wave's onset and offset are detected by using the average of low-pass difference method and tangent method. Tangent method is always used to locate the offset of T Wave but it wil firstly be used to locate the P wave onset and offset in this paper. The proposed algorithm is tested by the annotated CSE database. Result shows that algorithm test result has a good consistency with BIS CSE annotation. Compared with the mean and standard deviation of P wave onset and offset, our algorithm and CSE annotation is-2.01 ms, 3.94 ms and 4.96 ms, 5.86 ms.


Asunto(s)
Algoritmos , Arritmias Cardíacas/diagnóstico , Electrocardiografía , Bases de Datos Factuales , Humanos , Procesamiento de Señales Asistido por Computador
3.
Heart Rhythm ; 12(7): 1584-94, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25998140

RESUMEN

BACKGROUND: Atrioventricular nodal reentrant tachycardia (AVNRT) may coexist with Brugada syndrome (BrS). OBJECTIVES: The present study was designed to determine the prevalence of drug-induced type 1 Brugada ECG pattern (concealed BrS) in patients presenting with clinical spontaneous AVNRT and to investigate their electrocardiographic, electrophysiological, and genetic characteristics. METHODS: Ninety-six consecutive patients without any sign of BrS on baseline electrocardiogram undergoing electrophysiological study and ablation for symptomatic, drug-resistant AVNRT and 66 control subjects underwent an ajmaline challenge to unmask BrS. Genetic screening was performed in 17 patients displaying both AVNRT and BrS. RESULTS: A concealed BrS electrocardiogram was uncovered in 26 of 96 patients with AVNRT (27.1%) and in 3 of 66 control subjects (4.5%) (P ≤ .001). Patients with concealed BrS were predominantly female patients (n=23 [88.5%] vs n=44 [62.9%], P = .015), had higher prevalence of chest pain (n=10 [38.5%] vs n=13 [18.6%], p=0.042), migraine headaches (n=10 [38.5%] vs n=10 [14.2%], p=0.008), and drug-induced initiation and/or worsening of duration and/or frequency of AVNRT (n=4 [15.4%] vs n=1 [1.4%], p=0.006) as compared to patients with AVNRT without BrS. Genetic screening identified 19 mutations or rare variants in 13 genes in 13 of 17 patients with both AVNRT and BrS (yield = 76.5%). Ten of these 13 genotype-positive patients (76.9%) harbored genetic variants known or suspected to cause a loss of function of cardiac sodium channel current (SCN5A, SCN10A, SCN1B, GPD1L, PKP2, and HEY2). CONCLUSION: Our results suggest that spontaneous AVNRT and concealed BrS co-occur, particularly in female patients, and that genetic variants that reduce sodium channel current may provide a mechanistic link between AVNRT and BrS and predispose to expression of both phenotypes.


Asunto(s)
Ajmalina/farmacología , Síndrome de Brugada , Ablación por Catéter/métodos , Taquicardia por Reentrada en el Nodo Atrioventricular , Adulto , Síndrome de Brugada/inducido químicamente , Síndrome de Brugada/diagnóstico , Síndrome de Brugada/epidemiología , Síndrome de Brugada/genética , Síndrome de Brugada/fisiopatología , Electrocardiografía/métodos , Técnicas Electrofisiológicas Cardíacas/métodos , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.8/genética , Prevalencia , Taquicardia por Reentrada en el Nodo Atrioventricular/diagnóstico , Taquicardia por Reentrada en el Nodo Atrioventricular/epidemiología , Taquicardia por Reentrada en el Nodo Atrioventricular/genética , Taquicardia por Reentrada en el Nodo Atrioventricular/fisiopatología , Taquicardia por Reentrada en el Nodo Atrioventricular/cirugía , Estados Unidos/epidemiología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética
4.
Int J Cardiol ; 171(3): 431-42, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24439875

RESUMEN

BACKGROUND: Genetic defects in KCNJ8, encoding the Kir6.1 subunit of the ATP-sensitive K(+) channel (I(K-ATP)), have previously been associated with early repolarization (ERS) and Brugada (BrS) syndromes. Here we test the hypothesis that genetic variants in ABCC9, encoding the ATP-binding cassette transporter of IK-ATP (SUR2A), are also associated with both BrS and ERS. METHODS AND RESULTS: Direct sequencing of all ERS/BrS susceptibility genes was performed on 150 probands and family members. Whole-cell and inside-out patch-clamp methods were used to characterize mutant channels expressed in TSA201-cells. Eight ABCC9 mutations were uncovered in 11 male BrS probands. Four probands, diagnosed with ERS, carried a highly-conserved mutation, V734I-ABCC9. Functional expression of the V734I variant yielded a Mg-ATP IC50 that was 5-fold that of wild-type (WT). An 18-y/o male with global ERS inherited an SCN5A-E1784K mutation from his mother, who displayed long QT intervals, and S1402C-ABCC9 mutation from his father, who displayed an ER pattern. ABCC9-S1402C likewise caused a gain of function of IK-ATP with a shift of ATP IC50 from 8.5 ± 2 mM to 13.4 ± 5 µM (p<0.05). The SCN5A mutation reduced peak INa to 39% of WT (p<0.01), shifted steady-state inactivation by -18.0 mV (p<0.01) and increased late I(Na) from 0.14% to 2.01% of peak I(Na) (p<0.01). CONCLUSION: Our study is the first to identify ABCC9 as a susceptibility gene for ERS and BrS. Our findings also suggest that a gain-of-function in I(K-ATP) when coupled with a loss-of-function in SCN5A may underlie type 3 ERS, which is associated with a severe arrhythmic phenotype.


Asunto(s)
Síndrome de Brugada/epidemiología , Síndrome de Brugada/genética , Predisposición Genética a la Enfermedad/epidemiología , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Receptores de Sulfonilureas/genética , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Síndrome de Brugada/diagnóstico , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Conejos , Ratas , Receptores de Sulfonilureas/química , Adulto Joven
5.
ScientificWorldJournal ; 2013: 834159, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24307882

RESUMEN

The authors find some new inequalities of Jordan type for the sine function. These newly established inequalities are of new form and are applied to deduce some known results.


Asunto(s)
Conceptos Matemáticos
6.
J Mol Cell Cardiol ; 60: 36-46, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23542310

RESUMEN

BACKGROUND: The ability to recapitulate mature adult phenotypes is critical to the development of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) as models of disease. The present study examines the characteristics of the transient outward current (Ito) and its contribution to the hiPSC-CM action potential (AP). METHOD: Embryoid bodies were made from a hiPS cell line reprogrammed with Oct4, Nanog, Lin28 and Sox2. Sharp microelectrodes were used to record APs from beating-clusters (BC) and patch-clamp techniques were used to record Ito in single hiPSC-CM. mRNA levels of Kv1.4, KChIP2 and Kv4.3 were quantified from BCs. RESULTS: BCs exhibited spontaneous beating (60.5±2.6 bpm) and maximum-diastolic-potential (MDP) of 67.8±0.8 mV (n=155). A small 4-aminopyridine-sensitive phase-1-repolarization was observed in only 6/155 BCs. A robust Ito was recorded in the majority of cells (13.7±1.9 pA/pF at +40 mV; n=14). Recovery of Ito from inactivation (at -80 mV) showed slow kinetics (τ1=200±110 ms (12%) and τ2=2380±240 ms (80%)) accounting for its minimal contribution to the AP. Transcript data revealed relatively high expression of Kv1.4 and low expression of KChIP2 compared to human native ventricular tissues. Mathematical modeling predicted that restoration of IK1 to normal levels would result in a more negative MDP and a prominent phase-1-repolarization. CONCLUSION: The slow recovery kinetics of Ito coupled with a depolarized MDP account for the lack of an AP notch in the majority of hiPSC-CM. These characteristics reveal a deficiency for the development of in vitro models of inherited cardiac arrhythmia syndromes in which Ito-induced AP notch is central to the disease phenotype.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Potenciales de la Membrana/fisiología , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Proteínas de Interacción con los Canales Kv/metabolismo , Canal de Potasio Kv1.4/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Miocitos Cardíacos/citología , Canales de Potasio Shal/metabolismo
7.
Am J Physiol Heart Circ Physiol ; 304(11): H1471-82, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23542916

RESUMEN

The voltage-sensitive dye bis-(1,3-dibutylbarbituric acid)trimethine oxonol [DiBAC4(3)] has been reported as a novel large-conductance Ca²âº-activated K⁺ (BK) channel activator with selectivity for its ß1- or ß4-subunits. In arterial smooth muscle, BK channels are formed by a pore-forming α-subunit and a smooth muscle-abundant regulatory ß1-subunit. This tissue specificity has driven extensive pharmacological research aimed at regulating arterial tone. Using animals with a disruption of the gene for the ß1-subunit, we explored the effects of DiBAC4(3) in native channels from arterial smooth muscle. We tested the hypothesis that, in native BK channels, activation by DiBAC4(3) relies mostly on its α-subunit. We studied BK channels from wild-type and transgenic ß1-knockout mice in excised patches. BK channels from brain arteries, with or without the ß1-subunit, were similarly activated by DiBAC4(3). In addition, we found that saturating concentrations of DiBAC4(3) (~30 µM) promote an unprecedented persistent activation of the channel that negatively shifts its voltage dependence by as much as -300 mV. This "sweet spot" for persistent activation is independent of Ca²âº and/or the ß1₋4-subunits and is fully achieved when DiBAC4(3) is applied to the intracellular side of the channel. Arterial BK channel response to DiBAC4(3) varies across species and/or vascular beds. DiBAC4(3) unique effects can reveal details of BK channel gating mechanisms and help in the rational design of BK channel activators.


Asunto(s)
Barbitúricos/farmacología , Isoxazoles/farmacología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Animales , Arterias/efectos de los fármacos , Arterias/metabolismo , Barbitúricos/metabolismo , Interpretación Estadística de Datos , Perros , Isoxazoles/metabolismo , Cinética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Técnicas de Placa-Clamp , Reacción en Cadena en Tiempo Real de la Polimerasa , Canales de Sodio/efectos de los fármacos
8.
PLoS One ; 7(7): e40288, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815737

RESUMEN

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) hold promise for therapeutic applications. To serve these functions, the hiPSC-CM must recapitulate the electrophysiologic properties of native adult cardiomyocytes. This study examines the electrophysiologic characteristics of hiPSC-CM between 11 and 121 days of maturity. Embryoid bodies (EBs) were generated from hiPS cell line reprogrammed with Oct4, Nanog, Lin28 and Sox2. Sharp microelectrodes were used to record action potentials (AP) from spontaneously beating clusters (BC) micro-dissected from the EBs (n = 103; 37°C) and to examine the response to 5 µM E-4031 (n = 21) or BaCl(2) (n = 22). Patch-clamp techniques were used to record I(Kr) and I(K1) from cells enzymatically dissociated from BC (n = 49; 36°C). Spontaneous cycle length (CL) and AP characteristics varied widely among the 103 preparations. E-4031 (5 µM; n = 21) increased Bazett-corrected AP duration from 291.8±81.2 to 426.4±120.2 msec (p<0.001) and generated early afterdepolarizations in 8/21 preparations. In 13/21 BC, E-4031 rapidly depolarized the clusters leading to inexcitability. BaCl(2), at concentrations that selectively block I(K1) (50-100 µM), failed to depolarize the majority of clusters (13/22). Patch-clamp experiments revealed very low or negligible I(K1) in 53% (20/38) of the cells studied, but presence of I(Kr) in all (11/11). Consistent with the electrophysiological data, RT-PCR and immunohistochemistry studies showed relatively poor mRNA and protein expression of I(K1) in the majority of cells, but robust expression of I(Kr.) In contrast to recently reported studies, our data point to major deficiencies of hiPSC-CM, with remarkable diversity of electrophysiologic phenotypes as well as pharmacologic responsiveness among beating clusters and cells up to 121 days post-differentiation (dpd). The vast majority have a maximum diastolic potential that depends critically on I(Kr) due to the absence of I(K1). Thus, efforts should be directed at producing more specialized and mature hiPSC-CM for future therapeutic applications.


Asunto(s)
Potenciales de Acción , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Potenciales de Acción/efectos de los fármacos , Compuestos de Bario/farmacología , Diferenciación Celular/efectos de los fármacos , Cloruros/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Piperidinas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/genética , Piridinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo
9.
Heart Rhythm ; 9(4): 548-55, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22056721

RESUMEN

BACKGROUND: Adenosine triphosphate (ATP)-sensitive potassium cardiac channels consist of inward-rectifying channel subunits Kir6.1 or Kir6.2 (encoded by KCNJ8 or KCNJ11) and the sulfonylurea receptor subunits SUR2A (encoded by ABCC9). OBJECTIVE: To examine the association of mutations in KCNJ8 with Brugada syndrome (BrS) and early repolarization syndrome (ERS) and to elucidate the mechanism underlying the gain of function of ATP-sensitive potassium channel current. METHODS: Direct sequencing of KCNJ8 and other candidate genes was performed on 204 BrS and ERS probands and family members. Whole-cell and inside-out patch-clamp methods were used to study mutated channels expressed in TSA201 cells. RESULTS: The same missense mutation, p.Ser422Leu (c.1265C>T) in KCNJ8, was identified in 3 BrS and 1 ERS probands but was absent in 430 alleles from ethnically matched healthy controls. Additional genetic variants included CACNB2b-D601E. Whole-cell patch-clamp studies showed a 2-fold gain of function of glibenclamide-sensitive ATP-sensitive potassium channel current when KCNJ8-S422L was coexpressed with SUR2A-wild type. Inside-out patch-clamp evaluation yielded a significantly greater half maximal inhibitory concentration for ATP in the mutant channels (785.5 ± 2 vs 38.4 ± 3 µM; n = 5; P <.01), pointing to incomplete closing of the ATP-sensitive potassium channels under normoxic conditions. Patients with a CACNB2b-D601E polymorphism displayed longer QT/corrected QT intervals, likely owing to their effect to induce an increase in L-type calcium channel current (I(Ca-L)). CONCLUSIONS: Our results support the hypothesis that KCNJ8 is a susceptibility gene for BrS and ERS and point to S422L as a possible hotspot mutation. Our findings suggest that the S422L-induced gain of function in ATP-sensitive potassium channel current is due to reduced sensitivity to intracellular ATP.


Asunto(s)
Síndrome de Brugada/genética , Muerte Súbita Cardíaca/epidemiología , Canales KATP/genética , Biología Molecular , Mutación Missense/genética , Taquicardia Ventricular/genética , Adulto , Anciano , Femenino , Francia/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Estadística como Asunto , Síndrome , Taquicardia Ventricular/epidemiología , Adulto Joven
10.
Pacing Clin Electrophysiol ; 33(3): 274-85, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20025708

RESUMEN

BACKGROUND: Inherited loss of function mutations in SCN5A have been linked to overlapping syndromes including cardiac conduction disease and Brugada syndrome (BrS). The mechanisms responsible for the development of one without the other are poorly understood. METHODS: Direct sequencing was performed in a family with cardiac conduction disease. Wild-type (WT) and mutant channels were expressed in TSA201 cells for electrophysiological study. Green fluorescent protein (GFP)-fused WT or mutant genes were used to assess channel trafficking. RESULTS: A novel SCN5A mutation, P1008S, was identified in all family members displaying first-degree atrioventricular block, but not in unaffected family members nor in 430 reference alleles. Peak P1008S current was 11.77% of WT (P < 0.001). Confocal microscopy showed that WT channels tagged with GFP were localized on the cell surface, whereas GFP-tagged P1008S channels remained trapped in intracellular organelles. Trafficking could be rescued by incubation at room temperature, but not by incubation with mexiletine (300 muM) at 37 degrees C. We also identified a novel polymorphism (D601E) in CACNB2b that slowed inactivation of L-type calcium current (I(Ca,L)), significantly increased total charge. Using the Luo-Rudy action potential (AP) model, we show that the reduction in sodium current (I(Na)) can cause loss of the right ventricular epicardial AP dome in the absence but not in the presence of the slowed inactivation of I(Ca,L). Slowed conduction was present in both cases. CONCLUSIONS: Our results suggest genetic variations leading to a loss-of-function in I(Na) coupled with a gain of function in I(Ca,L) may underlie the development of cardiac conduction disease without BrS.


Asunto(s)
Bradicardia/genética , Canales de Calcio Tipo L/genética , Bloqueo Cardíaco/genética , Sistema de Conducción Cardíaco/fisiopatología , Proteínas Musculares/genética , Mutación , Polimorfismo de Nucleótido Simple , Canales de Sodio/genética , Adolescente , Alelos , Análisis de Varianza , Bradicardia/fisiopatología , Síndrome de Brugada/genética , Síndrome de Brugada/fisiopatología , Técnicas Electrofisiológicas Cardíacas , Femenino , Bloqueo Cardíaco/fisiopatología , Humanos , Masculino , Microscopía Confocal , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5 , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa
11.
Circ Cardiovasc Genet ; 2(3): 270-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20031595

RESUMEN

BACKGROUND: Brugada syndrome, characterized by ST-segment elevation in the right precordial ECG leads and the development of life-threatening ventricular arrhythmias, has been associated with mutations in 6 different genes. We identify and characterize a mutation in a new gene. METHODS AND RESULTS: A 64-year-old white male displayed a type 1 ST-segment elevation in V1 and V2 during procainamide challenge. Polymerase chain reaction-based direct sequencing was performed using a candidate gene approach. A missense mutation (L10P) was detected in exon 1 of SCN3B, the beta 3 subunit of the cardiac sodium channel, but not in any other gene known to be associated with Brugada syndrome or in 296 controls. Wild-type (WT) and mutant genes were expressed in TSA201 cells and studied using whole-cell patch-clamp techniques. Coexpression of SCN5A/WT+SCN1B/WT+SCN3B/L10P resulted in an 82.6% decrease in peak sodium current density, accelerated inactivation, slowed reactivation, and a -9.6-mV shift of half-inactivation voltage compared with SCN5A/WT+SCN1B/WT+SCN3B/WT. Confocal microscopy revealed that SCN5A/WT channels tagged with green fluorescent protein are localized to the cell surface when coexpressed with WT SCN1B and SCN3B but remain trapped in intracellular organelles when coexpressed with SCN1B/WT and SCN3B/L10P. Western blot analysis confirmed the presence of Na(V)beta 3 in human ventricular myocardium. CONCLUSIONS: Our results provide support for the hypothesis that mutations in SCN3B can lead to loss of transport and functional expression of the hNa(v)1.5 protein, leading to reduction in sodium channel current and clinical manifestation of a Brugada phenotype.


Asunto(s)
Síndrome de Brugada/genética , Canales de Sodio/genética , Secuencia de Aminoácidos , Síndrome de Brugada/diagnóstico , Síndrome de Brugada/diagnóstico por imagen , Línea Celular , Electrocardiografía , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Técnicas de Placa-Clamp , Fenotipo , Radiografía , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Canales de Sodio/metabolismo , Subunidad beta-3 de Canal de Sodio Activado por Voltaje
12.
Circ Res ; 103(4): 396-404, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18599870

RESUMEN

Brugada syndrome has been linked to mutations in SCN5A. Agents that dissociate slowly from the sodium channel such as flecainide and ajmaline unmask the Brugada syndrome electrocardiogram and precipitate ventricular tachycardia/fibrillation. Lidocaine, an agent with rapid dissociation kinetics, has previously been shown to exert no effect in patients with Brugada syndrome. We characterized a novel double mutation of SCN5A (V232I in DI-S4+L1308F in DIII-S4) identified in a rare case of lidocaine (1 mg/kg)-induced Brugada syndrome. We studied lidocaine blockade of I(Na) generated by wild-type and V232I+L1308F mutant cardiac sodium channels expressed in mammalian TSA201 cells using patch clamp techniques. Despite no significant difference in steady-state gating parameters between V232I+L1308F and wild-type sodium currents at baseline, use-dependent inhibition of I(Na) by lidocaine was more pronounced in V232I+L1308F versus wild-type (73.0+/-0.1% versus 18.23+/-0.04% at 10 micromol/L measured at 10 Hz, respectively). A dose of 10 micromol/L lidocaine also caused a more negative shift of steady-state inactivation in V232I+L1308F versus wild-type (-14.1+/-0.3 mV and -4.8+/-0.3 mV, respectively). The individual mutations produced a much less accentuated effect. We report the first case of lidocaine-induced Brugada electrocardiogram phenotype. The double mutation in SCN5A, V232I, and L1308F alters the affinity of the cardiac sodium channel for lidocaine such that the drug assumes Class IC characteristics with potent use-dependent block of the sodium channel. Our results demonstrate an additive effect of the 2 missense mutations to sensitize the sodium channel to lidocaine. These findings suggest caution when treating patients carrying such genetic variations with Class I antiarrhythmic drugs.


Asunto(s)
Anestésicos Locales/efectos adversos , Síndrome de Brugada/genética , Lidocaína/efectos adversos , Proteínas Musculares/genética , Mutación Missense/genética , Canales de Sodio/genética , Síndrome de Brugada/fisiopatología , Electrocardiografía , Humanos , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5 , Fenotipo
13.
Heart Rhythm ; 5(3): 427-35, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18313602

RESUMEN

BACKGROUND: Atrial fibrillation (AF) is the most common clinical arrhythmia and a major cause of cardiovascular morbidity and mortality. Among the gene defects previously associated with AF is a gain of function of the slowly activating delayed rectifier potassium current IKs, secondary to mutations in KCNQ1. Coexpression of KCNE5, the gene encoding the MiRP4 beta-subunit, has been shown to reduce IKs. OBJECTIVE: The purpose of this study was to test the hypothesis that mutations in KCNE5 are associated with AF in a large cohort of patients with AF. METHODS: One-hundred fifty-eight patients with AF were screened for mutations in the coding region of KCNE5. RESULTS: A missense mutation involving substitution of a phenylalanine for leucine at position 65 (L65F) was identified in one patient. This patient did not have a history of familial AF, and neither KCNQ1 nor KCNE2 mutations were found. Transient transfection of Chinese hamster ovary (CHO) cells expressing IKs(KCNQ1+KCNE1) with KCNE5 suppressed the developing and tail currents of IKs in a concentration-dependent manner. Transient transfection with KCNE5-L65F failed to suppress IKs, yielding a current indistinguishable from that recorded in the absence of KCNE5. Developing currents recorded during a test pulse to +60 mV and tail currents recorded upon repolarization to -40 mV both showed a significant concentration-dependent gain of function in IKs with expression of KCNE5-L65F vs KCNE5-WT. CONCLUSION: The results of this study suggest that a missense mutation in KCNE5 may be associated with nonfamilial or acquired forms of AF. The arrhythmogenic mechanism most likely is a gain of function of IKs.


Asunto(s)
Fibrilación Atrial/genética , Mutación Missense , Canales de Potasio con Entrada de Voltaje/genética , Adulto , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Fibrilación Atrial/fisiopatología , Dinamarca , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Humanos , Masculino , Persona de Mediana Edad
14.
Circ Arrhythm Electrophysiol ; 1(3): 209-18, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19122847

RESUMEN

INTRODUCTION: The Brugada Syndrome (BrS), an inherited syndrome associated with a high incidence of sudden cardiac arrest, has been linked to mutations in four different genes leading to a loss of function in sodium and calcium channel activity. Although the transient outward current (I(to)) is thought to play a prominent role in the expression of the syndrome, mutations in I(to)-related genes have not been identified as yet. METHODS AND RESULTS: One hundred and five probands with BrS were screened for ion channel gene mutations using single strand conformation polymorphism (SSCP) electrophoresis and direct sequencing. A missense mutation (R99H) in KCNE3 (MiRP2) was detected in one proband. The R99H mutation was found 4/4 phenotype positive and 0/3 phenotype-negative family members. Chinese hamster ovary (CHO)-K1 cells were co-transfected using wild-type (WT) or mutant KCNE3 and either WT KCND3 or KCNQ1. Whole-cell patch clamp studies were performed after 48 hours. Interactions between Kv4.3 and KCNE3 were analyzed in co-immunoprecipitation experiments in human atrial samples. Co-transfection of R99H-KCNE3 with KCNQ1 produced no alteration in current magnitude or kinetics. However, co-transfection of R99H KCNE3 with KCND3 resulted in a significant increase in the I(to) intensity compared to WT KCNE3+KCND3. Using tissues isolated from left atrial appendages of human hearts, we also demonstrate that K(v)4.3 and KCNE3 can be co-immunoprecipitated. CONCLUSIONS: These results provide definitive evidence for a functional role of KCNE3 in the modulation of I(to) in the human heart and suggest that mutations in KCNE3 can underlie the development of BrS.


Asunto(s)
Síndrome de Brugada/genética , ADN/genética , Predisposición Genética a la Enfermedad , Mutación Missense , Canales de Potasio con Entrada de Voltaje/genética , Potenciales de Acción , Adolescente , Adulto , Anciano , Síndrome de Brugada/metabolismo , Síndrome de Brugada/fisiopatología , Células Cultivadas , Niño , Análisis Mutacional de ADN , Femenino , Estudios de Seguimiento , Humanos , Inmunoprecipitación , Masculino , Persona de Mediana Edad , Miocardio/metabolismo , Miocardio/patología , Técnicas de Placa-Clamp , Linaje , Canales de Potasio con Entrada de Voltaje/metabolismo , Adulto Joven
15.
Heart Rhythm ; 4(8): 1072-80, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17675083

RESUMEN

BACKGROUND: Ventricular tachycardia (VT) and ventricular fibrillation (VF) complicating Brugada syndrome, a genetic disorder linked to SCN5A mutations, and VF complicating acute myocardial infarction (AMI) both have been linked to phase 2 reentry. OBJECTIVE: Given the mechanistic similarities in arrhythmogenesis, the purpose of this study was to examine the contribution of SCN5A mutations to VT/VF complicating AMI. METHODS: Nineteen consecutive patients developing VF during AMI were enrolled in the study. Wild-type (WT) and mutant SCN5A genes were coexpressed with SCN1B in TSA201 cells and studied using whole-cell patch clamp techniques. RESULTS: Among the cohort of 19 patients, one missense mutation (G400A) in SCN5A was detected in a conserved region. An H558R polymorphism was detected on the same allele. Unlike the other 18 patients, who each developed 1-2 VF episodes during AMI, the mutation carrier developed six episodes of VT/VF within the first 12 hours. All VT/VF episodes were associated with ST-segment changes and were initiated by short-coupled extrasystoles. Flecainide and adenosine challenge performed to unmask Brugada and long QT syndromes both were negative. Peak G400A and G400A+H558R current were 70.7% and 88.4% less than WT current at -35 mV (P

Asunto(s)
Predisposición Genética a la Enfermedad/genética , Proteínas Musculares/genética , Mutación Missense , Infarto del Miocardio/genética , Canales de Sodio/genética , Taquicardia Ventricular/genética , Fibrilación Ventricular/genética , Potenciales de Acción , Adulto , Anciano , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/complicaciones , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Taquicardia Ventricular/etiología , Transfección , Fibrilación Ventricular/etiología
16.
J Cardiovasc Electrophysiol ; 18(9): 972-7, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17655673

RESUMEN

INTRODUCTION: Long QT Syndrome (LQTS) is an inherited disorder characterized by prolonged QT intervals and life-threatening polymorphic ventricular tachyarrhythmias. LQT1 caused by KCNQ1 mutations is the most common form of LQTS. METHODS AND RESULTS: Patients diagnosed with LQTS were screened for disease-associated mutations in KCNQ1, KCNH2, KCNE1, KCNE2, KCNJ2, and SCN5A. A novel mutation was identified in KCNQ1 caused by a three-base deletion at the position 824-826, predicting a deletion of phenylalanine at codon 275 in segment 5 of KCNQ1 (DeltaF275). Wild-type (WT) and DeltaF275-KCNQ1 constructs were generated and transiently transfected together with a KCNE1 construct in CHO-K1 cells to characterize the properties of the slowly activating delayed rectifier current (IKs) using conventional whole-cell patch-clamp techniques. Cells transfected with WT-KCNQ1 and KCNE1 (1:1.3 molar ratio) produced slowly activating outward current with the characteristics of IKs. Tail current density measured at -40 mV following a two-second step to +60 mV was 381.3 +/- 62.6 pA/pF (n = 11). Cells transfected with DeltaF275-KCNQ1 and KCNE1 exhibited essentially no current. (Tail current density: 0.8 +/- 2.1 pA/pF, n = 11, P = 0.00001 vs WT). Cotransfection of WT- and DeltaF275- KCNQ1 (50/50), along with KCNE1, produced little to no current (tail current density: 10.3 +/- 3.5 pA/pF, n = 11, P = 0.00001 vs WT alone), suggesting a potent dominant negative effect. Immunohistochemistry showed normal membrane trafficking of DeltaF275-KCNQ1. CONCLUSION: Our data suggest that a DeltaF275 mutation in KCNQ1 is associated with a very potent dominant negative effect leading to an almost complete loss of function of IKs and that this defect underlies a LQT1 form of LQTS.


Asunto(s)
Genes Dominantes/genética , Predisposición Genética a la Enfermedad/genética , Sistema de Conducción Cardíaco/fisiopatología , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/clasificación , Síndrome de QT Prolongado/fisiopatología , Adolescente , Animales , Células CHO , Cricetinae , Cricetulus , Femenino , Humanos , Síndrome de QT Prolongado/genética
17.
Circulation ; 115(4): 442-9, 2007 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-17224476

RESUMEN

BACKGROUND: Cardiac ion channelopathies are responsible for an ever-increasing number and diversity of familial cardiac arrhythmia syndromes. We describe a new clinical entity that consists of an ST-segment elevation in the right precordial ECG leads, a shorter-than-normal QT interval, and a history of sudden cardiac death. METHODS AND RESULTS: Eighty-two consecutive probands with Brugada syndrome were screened for ion channel gene mutations with direct sequencing. Site-directed mutagenesis was performed, and CHO-K1 cells were cotransfected with cDNAs encoding wild-type or mutant CACNB2b (Ca(v beta2b)), CACNA2D1 (Ca(v alpha2delta1)), and CACNA1C tagged with enhanced yellow fluorescent protein (Ca(v)1.2). Whole-cell patch-clamp studies were performed after 48 to 72 hours. Three probands displaying ST-segment elevation and corrected QT intervals < or = 360 ms had mutations in genes encoding the cardiac L-type calcium channel. Corrected QT ranged from 330 to 370 ms among probands and clinically affected family members. Rate adaptation of QT interval was reduced. Quinidine normalized the QT interval and prevented stimulation-induced ventricular tachycardia. Genetic and heterologous expression studies revealed loss-of-function missense mutations in CACNA1C (A39V and G490R) and CACNB2 (S481L) encoding the alpha1- and beta2b-subunits of the L-type calcium channel. Confocal microscopy revealed a defect in trafficking of A39V Ca(v)1.2 channels but normal trafficking of channels containing G490R Ca(v)1.2 or S481L Ca(v beta2b)-subunits. CONCLUSIONS: This is the first report of loss-of-function mutations in genes encoding the cardiac L-type calcium channel to be associated with a familial sudden cardiac death syndrome in which a Brugada syndrome phenotype is combined with shorter-than-normal QT intervals.


Asunto(s)
Canales de Calcio Tipo L/genética , Muerte Súbita Cardíaca , Electrocardiografía , Taquicardia Ventricular/genética , Fibrilación Ventricular/genética , Adulto , Animales , Células CHO , Canales de Calcio/genética , Canales de Calcio/fisiología , Canales de Calcio Tipo L/fisiología , Cricetinae , Cricetulus , Salud de la Familia , Femenino , Ligamiento Genético , Humanos , Masculino , Mutagénesis Sitio-Dirigida , Mutación Missense , Técnicas de Placa-Clamp , Fenotipo , Sistema de Registros , Taquicardia Ventricular/etnología , Taquicardia Ventricular/fisiopatología , Fibrilación Ventricular/etnología , Fibrilación Ventricular/fisiopatología , Población Blanca/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA