Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(3)2021 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-33498651

RESUMO

KCNQ1 encodes the voltage-gated potassium (Kv) channel KCNQ1, also known as KvLQT1 or Kv7.1. Together with its ß-subunit KCNE1, also denoted as minK, this channel generates the slowly activating cardiac delayed rectifier current IKs, which is a key regulator of the heart rate dependent adaptation of the cardiac action potential duration (APD). Loss-of-function mutations in KCNQ1 cause congenital long QT1 (LQT1) syndrome, characterized by a delayed cardiac repolarization and a prolonged QT interval in the surface electrocardiogram. Autosomal dominant loss-of-function mutations in KCNQ1 result in long QT syndrome, called Romano-Ward Syndrome (RWS), while autosomal recessive mutations lead to Jervell and Lange-Nielsen syndrome (JLNS), associated with deafness. Here, we identified a homozygous KCNQ1 mutation, c.1892_1893insC (p.P631fs*20), in a patient with an isolated LQT syndrome (LQTS) without hearing loss. Nevertheless, the inheritance trait is autosomal recessive, with heterozygous family members being asymptomatic. The results of the electrophysiological characterization of the mutant, using voltage-clamp recordings in Xenopus laevis oocytes, are in agreement with an autosomal recessive disorder, since the IKs reduction was only observed in homomeric mutants, but not in heteromeric IKs channel complexes containing wild-type channel subunits. We found that KCNE1 rescues the KCNQ1 loss-of-function in mutant IKs channel complexes when they contain wild-type KCNQ1 subunits, as found in the heterozygous state. Action potential modellings confirmed that the recessive c.1892_1893insC LQT1 mutation only affects the APD of homozygous mutation carriers. Thus, our study provides the molecular mechanism for an atypical autosomal recessive LQT trait that lacks hearing impairment.


Assuntos
Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Síndrome de Romano-Ward/genética , Potenciais de Ação , Animais , Surdez/genética , Feminino , Genes Recessivos , Heterozigoto , Homozigoto , Humanos , Masculino , Mutação , Oócitos/fisiologia , Técnicas de Patch-Clamp , Linhagem , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Síndrome de Romano-Ward/etiologia , Xenopus laevis
2.
Am J Case Rep ; 19: 1515-1518, 2018 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-30568157

RESUMO

BACKGROUND QT prolongation is a common, easily overlooked clinical problem with potentially dire consequences. Drug-induced and congenital forms are not mutually exclusive, but are treated differently. Here, we present a case of cryptogenic underlying congenital long QT syndrome (cLQTS) successfully treated with isoproterenol, a drug contraindicated in most congenital forms of this condition. CASE REPORT We present the case of a 54-year-old man who experienced severe QT prolongation after drug administration followed by recurrent episodes of torsade de pointes (TdP) with subsequent ventricular fibrillation (VF) arrest unresponsive to typical therapy. After failing electrolyte repletion, magnesium, amiodarone, and lidocaine, the patient was started on an isoproterenol drip to achieve a heart rate of at least 90 beats per minute (bpm). Isoproterenol resulted in an immediate near-normalization of his QT interval and cessation of his recurrent TdP. The patient was subsequently found to have a mutation of undetermined significance in the KCNQ1 gene, which is implicated in long QT syndrome type 1 (LQT1). Although isoproterenol is contraindicated in LQT1, our patient had an astonishingly therapeutic benefit. CONCLUSIONS After reviewing the electrophysiology of the delayed rectifier potassium current as it relates to long QT syndrome, we propose a mechanism by which our patient's specific mutation may have allowed him to derive benefit from isoproterenol treatment. We believe that there are patients with variants of LQT1 who can be safely treated with isoproterenol.


Assuntos
Eletrocardiografia , Síndrome do QT Longo/diagnóstico , Torsades de Pointes/diagnóstico , Cardiotônicos/uso terapêutico , Humanos , Isoproterenol/uso terapêutico , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/genética , Masculino , Pessoa de Meia-Idade , Mutação , Síndrome de Romano-Ward/tratamento farmacológico , Síndrome de Romano-Ward/etiologia , Torsades de Pointes/complicações
4.
J Cardiol ; 64(2): 121-6, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24373870

RESUMO

BACKGROUND: Several KCNQ1 splicing mutations have been identified in patients with type-1 long QT syndrome (LQT1). It was suggested that the clinical severity may differ according to the aberrant splicing products. There may be precipitating factors that cause cardiac events in those with a mild clinical phenotype (forme fruste LQT1). METHODS AND RESULTS: We analyzed the KCNQ1, KCNH2, SCN5A, KCNE1, and KCNE2 genes in 31 consecutive LQTS patients. A novel KCNQ1 1251+1G>A (IVS9+1G>A) mutation was identified in three probands and their two relatives. The QT interval in all of the five individuals with mutation was not much prolonged in the absence of precipitating factors (mean QTc was 461±30ms.). Two of the five individuals with mutation were symptomatic. One patient (a 38-year-old female) had experienced recurrent episodes of syncope due to ventricular tachyarrhythmias (VTAs) accompanied by QT prolongation (QTc: 750ms) when the serum potassium concentration ([K(+)]) was 2.7mEq/L. After correction of [K(+)], the QTc interval was shortened to 515ms, and the occurrence of VTAs ceased. Another patient (a 22-year-old female) was resuscitated from cardio-pulmonary arrest due to VTAs. Just after resuscitation, the QTc interval was 629ms, and [K(+)] was 2.9mEq/L. After correction of [K(+)], the QTc interval was dramatically shortened to 440ms. In order to identify abnormal splicing products of the responsible mutation, we analyzed the reverse transcription-polymerase chain reaction products from peripheral bloods of the mutation carrier, and identified exon 9-skipping (Δ9) and cryptic sequential exons 8 and 9-skipping (Δ8-9) products, as well as a no exon-skipping product. CONCLUSIONS: We identified a novel KCNQ splicing mutation 1251+1G>A in forme fruste LQT1, which induces cryptic splicing. Two of the five individuals with mutation experienced VTAs in the setting of hypokalemia, emphasizing the need to increase awareness of the significance of hypokalemia in this subgroup of LQT1 patients.


Assuntos
Hipopotassemia/complicações , Hipopotassemia/genética , Canal de Potássio KCNQ1/genética , Mutação , Síndrome de Romano-Ward/etiologia , Síndrome de Romano-Ward/genética , Adolescente , Adulto , Éxons/genética , Feminino , Humanos , Masculino , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taquicardia Ventricular/etiologia , Adulto Jovem
5.
J Cardiovasc Electrophysiol ; 20(8): 859-65, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19490272

RESUMO

BACKGROUND: Type-1 long-QT syndrome (LQT1) is caused by mutations in the KCNQ1 gene. The purpose of this study was to investigate whether KCNQ1 mutations in highly conserved amino acid residues within the voltage-gated potassium channel family are associated with an increased risk of cardiac events. METHODS AND RESULTS: The study population involved 492 LQT1 patients with 54 missense mutations in the transmembrane region of the KCNQ1 channel. The amino acid sequences of the transmembrane region of 38 human voltage-gated potassium channels were aligned. An adjusted Shannon entropy score for each amino acid residue was calculated ranging from 0 (no conservation) to 1.0 (full conservation). Cox analysis was used to identify independent factors associated with the first cardiac event (syncope, aborted cardiac arrest, or death). Patients were subcategorized into tertiles by their adjusted Shannon entropy scores. The lowest tertile (score 0-0.469; n = 146) was used as a reference group; patients with intermediate tertile scores (0.470-0.665; n = 150) had no increased risk of cardiac events (HR = 1.19, P = 0.42) or aborted cardiac arrest/sudden cardiac death (HR = 1.58, P = 0.26), and those with the highest tertile scores (>0.665; n = 196) showed significantly increased risk of cardiac events (HR = 3.32, P <0.001) and aborted cardiac arrest/sudden cardiac death (HR = 2.62, P = 0.04). The increased risk in patients with the highest conservation scores was independent of QTc, gender, age, and beta-blocker therapy. CONCLUSIONS: Mutations in highly conserved amino acid residues in the KCNQ1 gene are associated with a significant risk of cardiac events independent of QTc, gender, and beta-blocker therapy.


Assuntos
Sequência Conservada , Canal de Potássio KCNQ1/genética , Mutação de Sentido Incorreto/genética , Síndrome de Romano-Ward/genética , Adolescente , Adulto , Sequência de Aminoácidos , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/genética , Criança , Pré-Escolar , Biologia Computacional/métodos , Feminino , Seguimentos , Humanos , Lactente , Recém-Nascido , Masculino , Dados de Sequência Molecular , Fatores de Risco , Síndrome de Romano-Ward/etiologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...