Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Sci Rep ; 12(1): 2904, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35190635

RESUMO

Long QT syndrome (LQTS) is commonly presented with life-threatening ventricular arrhythmias (VA). Renal artery denervation (RDN) is an alternative antiadrenergic treatment that attenuates sympathetic activity. We aimed to evaluate the efficacy of RDN on preventing VAs in LQTS rabbits induced by drugs. The subtypes of LQTS were induced by infusion of HMR-1556 for LQTS type 1 (LQT1), erythromycin for LQTS type 2 (LQT2), and veratridine for LQTS type 3 (LQT3). Forty-four rabbits were randomized into the LQT1, LQT2, LQT3, LQT1-RDN, LQT2-RDN, and LQT3-RDN groups. All rabbits underwent cardiac electrophysiology studies. The QTc interval of the LQT2-RDN group was significantly shorter than those in the LQT2 group (650.08 ± 472.67 vs. 401.78 ± 42.91 ms, p = 0.011). The QTc interval of the LQT3-RDN group was significantly shorter than those in the LQT3 group (372.00 ± 22.41 vs. 335.70 ± 28.21 ms, p = 0.035). The VA inducibility in all subtypes of the LQT-RDN groups was significantly lower than those in the LQT-RDN groups, respectively (LQT1: 9.00 ± 3.30 vs. 47.44 ± 4.21%, p < 0.001; LQT2: 11.43 ± 6.37 vs. 45.38 ± 5.29%, p = 0.026; LQT3: 10.00 ± 6.32 vs. 32.40 ± 7.19%, p = 0.006). This study demonstrated the neuromodulation of RDN leading to electrical remodeling and reduced VA inducibility of the ventricular substrate in LQT models.


Assuntos
Arritmias Cardíacas/prevenção & controle , Denervação/métodos , Síndrome do QT Longo/complicações , Artéria Renal/inervação , Animais , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/etiologia , Remodelamento Atrial , Modelos Animais de Doenças , Eletrocardiografia , Ventrículos do Coração/fisiopatologia , Síndrome do QT Longo/classificação , Coelhos
2.
Circ Genom Precis Med ; 13(4): e002922, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32469608

RESUMO

BACKGROUND: Long-QT syndrome (LQTS) is characterized by a prolonged heart rate-corrected QT interval (QTc). Genome-wide association studies identified common genetic variants that collectively explain ≈8% to 10% of QTc variation in the general population. METHODS: Overall, 423 patients with LQT1, LQT2, or LQT3 were genotyped for 61 QTc-associated genetic variants used in a prototype QTc-polygenic risk score (QTc-PRS). A weighted QTc-PRS (range, 0-154.8 ms) was calculated for each patient, and the FHS (Framingham Heart Study) population-based reference cohort (n=853). RESULTS: The average QTc-PRS in LQTS was 88.0±7.2 and explained only ≈2.0% of the QTc variability. The QTc-PRS in LQTS probands (n=137; 89.3±6.8) was significantly greater than both FHS controls (87.2±7.4, difference-in-means±SE: 2.1±0.7, P<0.002) and LQTS genotype-positive family members (87.5±7.4, difference-in-mean, 1.8±.7, P<0.009). There was no difference in QTc-PRS between symptomatic (n=156, 88.6±7.3) and asymptomatic patients (n=267; 87.7±7.2, difference-in-mean, 0.9±0.7, P=0.15). LQTS patients with a QTc≥480 ms (n=120) had a significantly higher QTc-PRS (89.3±6.7) than patients with a QTc<480 ms (n=303, 87.6±7.4, difference-in-mean, 1.7±0.8, P<0.05). There was no difference in QTc-PRS or QTc between genotypes. CONCLUSIONS: The QTc-PRS explained <2% of the QTc variability in our LQT1, LQT2, and LQT3 cohort, contributing 5× less to their QTc value than in the general population. This prototype QTc-PRS does not distinguish/predict the clinical outcomes of individuals with LQTS.


Assuntos
Síndrome do QT Longo/genética , Polimorfismo de Nucleotídeo Único , Adulto , Estudos de Coortes , Eletrocardiografia , Família , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Frequência Cardíaca , Humanos , Modelos Lineares , Síndrome do QT Longo/classificação , Síndrome do QT Longo/patologia , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Adulto Jovem
3.
Mol Genet Genomic Med ; 8(9): e1300, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32383558

RESUMO

BACKGROUND: Genes encoding cardiac ion channels or regulating proteins have been associated with the inherited form of long QT syndrome (LQTS). Complex pathophysiology and missing functional studies, however, often bedevil variant interpretation and classification. We aimed to evaluate the rate of change in variant classification based on current interpretation standards and dependent on clinical findings. METHODS: Medical charts of children with a molecular genetic diagnosis of LQTS presenting at our centers were retrospectively reviewed. Reinterpretation of originally reported variants in genes associated with LQTS was performed based on current knowledge (March 2019) and according to the "Standards and Guidelines for the Interpretation of Sequence Variants" by the ACMG 2015. RESULTS: About 84 distinct (likely) pathogenic variants identified in 127 patients were reinterpreted. In 12 variants (12/84, 14.3%), classification changed from (likely) pathogenic to variant of unknown significance (VUS). One of these variants was a hypomorphic allele escaping the standard variant classification. Individuals with variants that downgraded to VUS after reevaluation showed significantly lower Schwartz scores and QTc intervals compared to individuals with unchanged variant characterization. CONCLUSION: This finding confirms genetic variant interpretation as a dynamic process and underlines the importance of ongoing genetic counseling, especially in LQTS patients with minor clinical criteria.


Assuntos
Síndrome do QT Longo/genética , Mutação , Adolescente , Calmodulina/genética , Criança , Pré-Escolar , Canal de Potássio ERG1/genética , Feminino , Frequência do Gene , Humanos , Lactente , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/classificação , Síndrome do QT Longo/patologia , Masculino , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Fenótipo , Canais de Potássio Corretores do Fluxo de Internalização/genética
4.
Stem Cell Reports ; 13(2): 394-404, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31378668

RESUMO

For long QT syndrome (LQTS), recent progress in genome-sequencing technologies enabled the identification of rare genomic variants with diagnostic, prognostic, and therapeutic implications. However, pathogenic stratification of the identified variants remains challenging, especially in variants of uncertain significance. This study aimed to propose a phenotypic cell-based diagnostic assay for identifying LQTS to recognize pathogenic variants in a high-throughput manner suitable for screening. We investigated the response of LQT2-induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iPSC-CMs) following IKr blockade using a multi-electrode array, finding that the response to IKr blockade was significantly smaller than in Control-iPSC-CMs. Furthermore, we found that LQT1-iPSC-CMs and LQT3-iPSC-CMs could be distinguished from Control-iPSC-CMs by IKs blockade and INa blockade, respectively. This strategy might be helpful in compensating for the shortcomings of genetic testing of LQTS patients.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Síndrome do QT Longo/diagnóstico , Miócitos Cardíacos/fisiologia , Potenciais de Ação/efeitos dos fármacos , Adolescente , Adulto , Estudos de Casos e Controles , Diferenciação Celular , Criança , Feminino , Ensaios de Triagem em Larga Escala/métodos , Humanos , Síndrome do QT Longo/classificação , Masculino , Pessoa de Meia-Idade , Técnicas de Patch-Clamp , Fenótipo , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Adulto Jovem
5.
Curr Probl Cardiol ; 44(3): 92-106, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29784533

RESUMO

Long QT syndrome (LQT) represents a heterogeneous family of cardiac electrophysiologic disorders characterized by QT prolongation and T-wave abnormalities on the electrocardiogram. It is commonly associated with syncope, however, sudden cardiac death can occur due to torsades de pointes. LQT is a clinical diagnosis and should be suspected in individuals on the basis of clinical presentation, family history and ECG characteristics. Management is focused on the prevention of syncope and ultimately sudden death. Complete cessation of symptoms is the goal. Life-style modification, beta blockers and ICD implantation are the most important therapeutic modalities in proper management of patients with LQT. Awareness should be raised regarding possible circumstances that could increase the risk of QT prolongation. Advanced age, hypokalemia, a history of heart failure, and structural heart disease are often mentioned in this context. Prudent consideration is needed before making a decision to recommend an ICD implantation in a young, active patient. Medical and/or device therapy still represent important therapeutic modalities in the management of patients with LQT with careful clinical judgement for the substrate of patients who will benefit. Insights from benchside to bedside have facilitated progress toward better therapeutic strategies, there also remains a need for tailoring management toward individuals in a mechanism-specific manner to optimize care. In addition, continued progress toward fundamental understanding of mechanisms of ion channel function and drug-channel interaction will guide the development of more effective, mechanism-based molecular agents in the treatment of LQT.


Assuntos
Morte Súbita Cardíaca/prevenção & controle , Gerenciamento Clínico , Técnicas Eletrofisiológicas Cardíacas/métodos , Síndrome do QT Longo , Morte Súbita Cardíaca/etiologia , Humanos , Síndrome do QT Longo/classificação , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/fisiopatologia , Síndrome do QT Longo/terapia
6.
Circulation ; 137(6): 619-630, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29431662

RESUMO

The acquired and congenital forms of long QT syndrome represent 2 distinct but clinically and genetically intertwined disorders of cardiac repolarization characterized by the shared final common pathway of QT interval prolongation and risk of potentially life-threatening arrhythmias. Over the past 2 decades, our understanding of the spectrum of genetic variation that (1) perturbs the function of cardiac ion channel macromolecular complexes and intracellular calcium-handling proteins, (2) underlies acquired/congenital long QT syndrome susceptibility, and (3) serves as a determinant of QT interval duration in the general population has grown exponentially. In turn, these molecular insights led to the development and increased utilization of clinically impactful genetic testing for congenital long QT syndrome. However, the widespread adoption and potential misinterpretation of the 2015 American College of Medical Genetics and Genomics variant classification and reporting guidelines may have contributed unintentionally to the reduced reporting of common genetic variants, with compelling epidemiological and functional evidence to support a potentially proarrhythmic role in patients with congenital and acquired long QT syndrome. As a result, some genetic testing reports may fail to convey the full extent of a patient's genetic susceptibility for a potentially life-threatening arrhythmia to the ordering healthcare professional. In this white paper, we examine the current classification and reporting (or lack thereof) of potentially proarrhythmic common genetic variants and investigate potential mechanisms to facilitate the reporting of these genetic variants without increasing the risk of diagnostic miscues.


Assuntos
Potenciais de Ação , Testes Genéticos/métodos , Variação Genética , Sistema de Condução Cardíaco/fisiopatologia , Frequência Cardíaca , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Erros de Diagnóstico , Predisposição Genética para Doença , Humanos , Síndrome do QT Longo/classificação , Síndrome do QT Longo/fisiopatologia , Fenótipo , Valor Preditivo dos Testes , Prognóstico , Reprodutibilidade dos Testes , Medição de Risco , Fatores de Risco
7.
Artigo em Inglês | MEDLINE | ID: mdl-28794082

RESUMO

BACKGROUND: Insight into type 6 long-QT syndrome (LQT6), stemming from mutations in the KCNE2-encoded voltage-gated channel ß-subunit, is limited. We sought to further characterize its clinical phenotype. METHODS AND RESULTS: Individuals with reported pathogenic KCNE2 mutations identified during arrhythmia evaluation were collected from inherited arrhythmia clinics and the Rochester long-QT syndrome (LQTS) registry. Previously reported LQT6 cases were identified through a search of the MEDLINE database. Clinical features were assessed, while reported KCNE2 mutations were evaluated for genotype-phenotype segregation and classified according to the contemporary American College of Medical Genetics guidelines. Twenty-seven probands possessed reported pathogenic KCNE2 mutations, while a MEDLINE search identified 17 additional LQT6 cases providing clinical and genetic data. Sixteen probands had normal resting QTc values and only developed QT prolongation and malignant arrhythmias after exposure to QT-prolonging stressors, 10 had other LQTS pathogenic mutations, and 10 did not have an LQTS phenotype. Although the remaining 8 subjects had an LQTS phenotype, evidence suggested that the KCNE2 variant was not the underlying culprit. The collective frequency of KCNE2 variants implicated in LQT6 in the Exome Aggregation Consortium database was 1.4%, in comparison with a 0.0005% estimated clinical prevalence for LQT6. CONCLUSIONS: On the basis of clinical phenotype, the high allelic frequencies of LQT6 mutations in the Exome Aggregation Consortium database, and absence of previous documentation of genotype-phenotype segregation, our findings suggest that many KCNE2 variants, and perhaps all, have been erroneously designated as LQTS-causative mutations. Instead, KCNE2 variants may confer proarrhythmic susceptibility when provoked by additional environmental/acquired or genetic factors, or both.


Assuntos
Síndrome do QT Longo/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Criança , Pré-Escolar , Feminino , Genótipo , Humanos , Síndrome do QT Longo/classificação , Masculino , Pessoa de Meia-Idade , Mutação , Fenótipo
8.
Cardiol Young ; 27(S1): S43-S48, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28084960

RESUMO

Untreated congenital long QT syndrome may result in potentially lethal ventricular tachycardia. In the most common type, risk of such an event has been linked to exercise. This originally resulted in very restrictive guidelines for sports participation in affected individuals. Although the complex interactions of a specific genotype, modifying cofactors, and risk are only now being explored, scientific evidence based on clinical experience now suggests that in many instances such restrictive guidelines are unwarranted. In particular, patients with this condition who are compliant with ß-blocker therapy and who have never had symptoms during exertion are now enjoying the benefits of athletic activity.


Assuntos
Antagonistas Adrenérgicos beta/uso terapêutico , Morte Súbita Cardíaca/etiologia , Sistema de Condução Cardíaco/fisiopatologia , Síndrome do QT Longo/classificação , Esportes , Taquicardia Ventricular/diagnóstico , Eletrocardiografia , Exercício Físico/fisiologia , Guias como Assunto , Humanos , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/genética
9.
Artigo em Inglês | MEDLINE | ID: mdl-27406603

RESUMO

BACKGROUND: Congenital long QT syndrome (LQTS) is characterized by QT prolongation. However, the QT interval itself is insufficient for diagnosis, unless the corrected QT interval is repeatedly ≥500 ms without an acquired explanation. Further, the majority of LQTS patients have a corrected QT interval below this threshold, and a significant minority has normal resting corrected QT interval values. Here, we aimed to develop and validate a novel, quantitative T wave morphological analysis program to differentiate LQTS patients from healthy controls. METHODS AND RESULTS: We analyzed a genotyped cohort of 420 patients (22±16 years, 43% male) with either LQT1 (61%) or LQT2 (39%). ECG analysis was conducted using a novel, proprietary T wave analysis program that quantitates subtle changes in T wave morphology. The top 3 discriminating features in each ECG lead were determined and the lead with the best discrimination selected. Classification was performed using a linear discriminant classifier and validated on an untouched cohort. The top 3 features were Tpeak-Tend interval, T wave left slope, and T wave center of gravity x axis (last 25% of the T wave). Lead V6 had the best discrimination. It could distinguish 86.8% of LQTS patients from healthy controls. Moreover, it distinguished 83.33% of patients with concealed LQTS from controls, despite having essentially identical resting corrected QT interval values. CONCLUSIONS: T wave quantitative analysis on the 12-lead surface ECG provides an effective, novel tool to distinguish patients with either LQT1/LQT2 from healthy matched controls. It can provide guidance while mutation-specific genetic testing is in motion for family members.


Assuntos
Eletrocardiografia/métodos , Síndrome do QT Longo/diagnóstico , Feminino , Genótipo , Humanos , Síndrome do QT Longo/classificação , Síndrome do QT Longo/genética , Masculino , Sensibilidade e Especificidade , Processamento de Sinais Assistido por Computador , Adulto Jovem
10.
Circ Cardiovasc Genet ; 8(4): 582-95, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25904541

RESUMO

BACKGROUND: A 2% to 5% background rate of rare SCN5A nonsynonymous single nucleotide variants (nsSNVs) among healthy individuals confounds clinical genetic testing. Therefore, the purpose of this study was to enhance interpretation of SCN5A nsSNVs for clinical genetic testing using estimated predictive values derived from protein-topology and 7 in silico tools. METHODS AND RESULTS: Seven in silico tools were used to assign pathogenic/benign status to nsSNVs from 2888 long-QT syndrome cases, 2111 Brugada syndrome cases, and 8975 controls. Estimated predictive values were determined for each tool across the entire SCN5A-encoded Na(v)1.5 channel as well as for specific topographical regions. In addition, the in silico tools were assessed for their ability to correlate with cellular electrophysiology studies. In long-QT syndrome, transmembrane segments S3-S5+S6 and the DIII/DIV linker region were associated with high probability of pathogenicity. For Brugada syndrome, only the transmembrane spanning domains had a high probability of pathogenicity. Although individual tools distinguished case- and control-derived SCN5A nsSNVs, the composite use of multiple tools resulted in the greatest enhancement of interpretation. The use of the composite score allowed for enhanced interpretation for nsSNVs outside of the topological regions that intrinsically had a high probability of pathogenicity, as well as within the transmembrane spanning domains for Brugada syndrome nsSNVs. CONCLUSIONS: We have used a large case/control study to identify regions of Na(v)1.5 associated with a high probability of pathogenicity. Although topology alone would leave the variants outside these identified regions in genetic purgatory, the synergistic use of multiple in silico tools may help promote or demote a variant's pathogenic status.


Assuntos
Síndrome de Brugada/genética , Predisposição Genética para Doença/genética , Síndrome do QT Longo/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Polimorfismo de Nucleotídeo Único , Sequência de Aminoácidos , Síndrome de Brugada/classificação , Síndrome de Brugada/fisiopatologia , Estudos de Casos e Controles , Biologia Computacional/métodos , Simulação por Computador , Eletrofisiologia , Frequência do Gene , Humanos , Síndrome do QT Longo/classificação , Síndrome do QT Longo/fisiopatologia , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Fenótipo , Estrutura Secundária de Proteína
13.
Circ J ; 78(12): 2827-33, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25274057

RESUMO

Congenital long QT syndrome (LQTS) is an inherited arrhythmia syndrome characterized by a prolonged QT interval on the 12-lead ECG, torsades de pointes and a higher chance of sudden cardiac death. LQTS segregates in a Mendelian fashion, which includes Romano-Ward syndrome with an autosomal dominant pattern as well as a rare autosomal recessive pattern (Jervell and Lange-Nielsen syndrome). Since 1957 when Jervell and Lange-Nielsen reported the first familial LQTS with congenital deafness, progress in understanding the genetic and electrophysiological mechanisms of LQTS has tremendously improved diagnostic methods and treatments. In the meantime, it has become evident that LQTS may not always be explained by a single gene mutation, but seems to follow a more complex genetic model intertwined with genetic common polymorphisms that have a mild to moderate effect on disease expression. In this review, we summarize the characteristics of LQTS (mainly LQT1-3) and briefly describe the most recent advances in LQTS clinical diagnostics as well as genetics.


Assuntos
Síndrome do QT Longo/genética , Antagonistas Adrenérgicos beta/uso terapêutico , Denervação Autônoma , Terapia Combinada , Morte Súbita Cardíaca/etiologia , Desfibriladores Implantáveis , Eletrocardiografia , Genes Dominantes , Genes Recessivos , Heterogeneidade Genética , Genótipo , Humanos , Canais Iônicos/genética , Síndrome do QT Longo/classificação , Síndrome do QT Longo/congênito , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/epidemiologia , Síndrome do QT Longo/fisiopatologia , Síndrome do QT Longo/terapia , Herança Multifatorial , Fenótipo , Potássio/uso terapêutico , Síndrome de Romano-Ward/genética , Síncope/etiologia
14.
Yonsei Med J ; 54(2): 529-33, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23364992

RESUMO

Long QT syndrome (LQTs) is an uncommon genetic disease causing sudden cardiac death with Torsade de Pointes (TdP). The first line drug treatment has been known to be ß-blocker. We encountered a 15-year-old female student with LQTs who had prolonged QTc and multiple episodes of syncope or agonal respiration during sleep. Although her T wave morphology in surface electrocardiography resembled LQTs type 1, her clinical presentation was unusual. During the epinephrine test, TdP was aggravated during ß-blocker medication, but alleviated by sodium channel blocker (mexiletine). Therefore, she underwent implantable cardioverter defibrillator implantation.


Assuntos
Antagonistas Adrenérgicos beta/efeitos adversos , Epinefrina , Síndrome do QT Longo/diagnóstico , Mexiletina/uso terapêutico , Síncope , Adolescente , Antagonistas Adrenérgicos beta/uso terapêutico , Desfibriladores Implantáveis , Diagnóstico Diferencial , Técnicas de Diagnóstico Cardiovascular , Feminino , Humanos , Síndrome do QT Longo/classificação , Síndrome do QT Longo/genética , Síndrome do QT Longo/terapia , Linhagem
15.
Yonsei Medical Journal ; : 529-533, 2013.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-149915

RESUMO

Long QT syndrome (LQTs) is an uncommon genetic disease causing sudden cardiac death with Torsade de Pointes (TdP). The first line drug treatment has been known to be beta-blocker. We encountered a 15-year-old female student with LQTs who had prolonged QTc and multiple episodes of syncope or agonal respiration during sleep. Although her T wave morphology in surface electrocardiography resembled LQTs type 1, her clinical presentation was unusual. During the epinephrine test, TdP was aggravated during beta-blocker medication, but alleviated by sodium channel blocker (mexiletine). Therefore, she underwent implantable cardioverter defibrillator implantation.


Assuntos
Adolescente , Feminino , Humanos , Antagonistas Adrenérgicos beta/efeitos adversos , Desfibriladores Implantáveis , Diagnóstico Diferencial , Técnicas de Diagnóstico Cardiovascular , Epinefrina , Síndrome do QT Longo/classificação , Mexiletina/uso terapêutico , Linhagem , Síncope
16.
Circ Cardiovasc Genet ; 5(5): 519-28, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22949429

RESUMO

BACKGROUND: Hundreds of nonsynonymous single nucleotide variants (nsSNVs) have been identified in the 2 most common long-QT syndrome-susceptibility genes (KCNQ1 and KCNH2). Unfortunately, an ≈3% BACKGROUND: and KCNH2 nsSNVs amongst healthy individuals complicates the ability to distinguish rare pathogenic mutations from similarly rare yet presumably innocuous variants. METHODS AND RESULTS: In this study, 4 tools [(1) conservation across species, (2) Grantham values, (3) sorting intolerant from tolerant, and (4) polymorphism phenotyping] were used to predict pathogenic or benign status for nsSNVs identified across 388 clinically definite long-QT syndrome cases and 1344 ostensibly healthy controls. From these data, estimated predictive values were determined for each tool independently, in concert with previously published protein topology-derived estimated predictive values, and synergistically when ≥3 tools were in agreement. Overall, all 4 tools displayed a statistically significant ability to distinguish between case-derived and control-derived nsSNVs in KCNQ1, whereas each tool, except Grantham values, displayed a similar ability to differentiate KCNH2 nsSNVs. Collectively, when at least 3 of the 4 tools agreed on the pathogenic status of C-terminal nsSNVs located outside the KCNH2/Kv11.1 cyclic nucleotide-binding domain, the topology-specific estimated predictive value improved from 56% to 91%. CONCLUSIONS: Although in silico prediction tools should not be used to predict independently the pathogenicity of a novel, rare nSNV, our results support the potential clinical use of the synergistic utility of these tools to enhance the classification of nsSNVs, particularly for Kv11.1's difficult to interpret C-terminal region.


Assuntos
Síndrome do QT Longo/genética , Síndrome de Romano-Ward/genética , Algoritmos , Estudos de Casos e Controles , Biologia Computacional , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Humanos , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Síndrome do QT Longo/classificação , Síndrome do QT Longo/patologia , Fenótipo , Filogenia , Polimorfismo de Nucleotídeo Único , Estrutura Terciária de Proteína , Síndrome de Romano-Ward/classificação , Síndrome de Romano-Ward/patologia
17.
J Electrocardiol ; 45(4): 368-372, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22560601

RESUMO

BACKGROUND: Long QT syndrome (LQTS) gene mutation carriers with indeterminate electrocardiogram frequently escape clinical diagnosis. We assessed the use of epinephrine bolus injection in revealing T-wave abnormalities. METHODS: We recruited 30 genotyped asymptomatic LQTS gene carriers with nondiagnostic QT interval and 15 controls. Electrocardiogram was recorded with body surface potential mapping after an intravenous epinephrine bolus. T-wave morphology was determined as normal, biphasic, inverted, bifid, or combined pattern. RESULTS: Long QT syndrome carriers and healthy controls had different T-wave profiles (P = .027). Of controls, 12 (80%) of 15 had no change or biphasic appearance, whereas only 10 (33%) of 30 of LQTS carriers had so. Bifid or combined pattern occurred in 15 (50%) of 30 in LQTS and in 6 (60%) of 10 in the LQT3 subgroup but only in 1 (7%) of 15 of healthy. CONCLUSIONS: Modification of ventricular repolarization with low-dose epinephrine injection helps to distinguish silent LQTS mutation carriers. This concerns also the LQT3 subtype, which may escape tests.


Assuntos
Mapeamento Potencial de Superfície Corporal , Epinefrina , Canais de Potássio Éter-A-Go-Go/genética , Triagem de Portadores Genéticos , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/genética , Mutação , Canais de Sódio/genética , Adulto , Doenças Assintomáticas , Canal de Potássio ERG1 , Epinefrina/administração & dosagem , Feminino , Genótipo , Frequência Cardíaca/efeitos dos fármacos , Humanos , Injeções Intravenosas , Síndrome do QT Longo/classificação , Masculino , Canal de Sódio Disparado por Voltagem NAV1.5
18.
Congenit Heart Dis ; 7(4): E42-5, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22129298

RESUMO

Congenital Long QT syndrome (LQTS) is a cardiac channelopathy, which leads to prolongation of the QT interval. This prolongation can lead to ventricular tachyarrhythmias, syncope, and sudden cardiac death. There are various types of LQTS associated with the mutations, which affect the genes coding the ion channels in the myocardial cells. Because of the differences in the ion channel physiology, clinical presentation of the subtypes can show significant differences. The most common types of LQTS are LQT1, LQT2, and LQT3. In LQT1 and LQT2, mutations are in the genes encoding Potassium channels (KCNQ1 and KCNH2 genes). These two subtypes show sensitivity to adrenergic stimuli. In LQT1, episodes are usually exercise-triggered, and in LQT2, episodes are more likely to be related to sudden arousal and emotional stress. Therefore treatment of LQT1 and LQT2 is mainly based on antiadrenergic therapy. LQT3 on the other hand, is a result of a mutation of SCN5A gene, which encodes the Sodium channels. In this type, patients are sensitive to vagal stimuli and episodes tend to occur at rest. Therefore, treatment choices between different subtypes of LQTS vary. In this case report, we report a patient with congenital LQT3 (Y1795C mutation) presenting with implantable cardioverter defibrillator (ICD) storm. Patient's arrhythmia burden was eliminated following successful treatment with flecainide.


Assuntos
Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/etiologia , Flecainida/uso terapêutico , Síndrome do QT Longo/complicações , Adolescente , Humanos , Síndrome do QT Longo/classificação , Síndrome do QT Longo/congênito , Masculino , Indução de Remissão
19.
Dis Model Mech ; 5(2): 220-30, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22052944

RESUMO

Long QT syndrome (LQTS) is caused by functional alterations in cardiac ion channels and is associated with prolonged cardiac repolarization time and increased risk of ventricular arrhythmias. Inherited type 2 LQTS (LQT2) and drug-induced LQTS both result from altered function of the hERG channel. We investigated whether the electrophysiological characteristics of LQT2 can be recapitulated in vitro using induced pluripotent stem cell (iPSC) technology. Spontaneously beating cardiomyocytes were differentiated from two iPSC lines derived from an individual with LQT2 carrying the R176W mutation in the KCNH2 (HERG) gene. The individual had been asymptomatic except for occasional palpitations, but his sister and father had died suddenly at an early age. Electrophysiological properties of LQT2-specific cardiomyocytes were studied using microelectrode array and patch-clamp, and were compared with those of cardiomyocytes derived from control cells. The action potential duration of LQT2-specific cardiomyocytes was significantly longer than that of control cardiomyocytes, and the rapid delayed potassium channel (I(Kr)) density of the LQT2 cardiomyocytes was significantly reduced. Additionally, LQT2-derived cardiac cells were more sensitive than controls to potentially arrhythmogenic drugs, including sotalol, and demonstrated arrhythmogenic electrical activity. Consistent with clinical observations, the LQT2 cardiomyocytes demonstrated a more pronounced inverse correlation between the beating rate and repolarization time compared with control cells. Prolonged action potential is present in LQT2-specific cardiomyocytes derived from a mutation carrier and arrhythmias can be triggered by a commonly used drug. Thus, the iPSC-derived, disease-specific cardiomyocytes could serve as an important platform to study pathophysiological mechanisms and drug sensitivity in LQT2.


Assuntos
Células-Tronco Pluripotentes Induzidas/fisiologia , Síndrome do QT Longo/etiologia , Síndrome do QT Longo/fisiopatologia , Modelos Cardiovasculares , Potenciais de Ação , Substituição de Aminoácidos , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/fisiopatologia , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Primers do DNA/genética , Canal de Potássio ERG1 , Fenômenos Eletrofisiológicos , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Síndrome do QT Longo/classificação , Síndrome do QT Longo/genética , Mutação de Sentido Incorreto , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp
20.
Europace ; 13(10): 1478-83, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21712262

RESUMO

AIMS: KCNE1 encodes an auxiliary subunit of cardiac potassium channels. Loss-of-function variations in this gene have been associated with the LQT5 form of the long QT syndrome (LQTS), secondary to reduction of I(Ks) current. We present a case in which a D85N rare polymorphism in KCNE1 is associated with an LQT2 phenotype. METHODS AND RESULTS: An 11-year old competitive athlete presented with mild bradycardia and a QTc interval of 470 ms. An LQT2 phenotype, consisting of low-voltage bifid T waves, was evident in the right precordial electrocardiogram leads. During the tachycardia phase following adenosine, QTc increased to 620 ms. Genetic analysis revealed a rare heterozygous polymorphism in KCNE1 predicting the substitution of asparagine for aspartic acid at position 85 of minK (D85N). Patch clamp experiments showed that KCNE1-D85N, when co-expressed with KCNH2 in TSA201 cells, significantly reduced I(Kr). Homozygous co-expression of the mutant with KCNH2 reduced I(Kr) tail current by 85%, whereas heterozygous co-expression reduced the current by 52%, demonstrating for the first time a dominant-negative effect of D85N to reduce I(Kr). Co-expression of the mutant with KCNQ1, either homozygously or heterozygously, produced no change in I(Ks). CONCLUSIONS: Our results suggest that a rare polymorphism KCNE1-D85N underlies the development of an LQT2 phenotype in this young athlete by interacting with KCNH2 to cause a dominant-negative effect to reduce I(Kr). Our data provide further evidence in support of the promiscuity of potassium channel ß subunits in modulating the function of multiple potassium channels leading to a diversity of clinical phenotypes.


Assuntos
Eletrocardiografia , Canais de Potássio Éter-A-Go-Go/genética , Síndrome do QT Longo/classificação , Síndrome do QT Longo/diagnóstico , Polimorfismo de Nucleotídeo Único/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Atletas , Bradicardia/diagnóstico , Bradicardia/etiologia , Bradicardia/fisiopatologia , Criança , Diagnóstico Diferencial , Canal de Potássio ERG1 , Feminino , Humanos , Síndrome do QT Longo/fisiopatologia , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...