Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Europace ; 26(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38825991

RESUMEN

AIMS: KCNQ1 mutations cause QTc prolongation increasing life-threatening arrhythmias risks. Heterozygous mutations [type 1 long QT syndrome (LQT1)] are common. Homozygous KCNQ1 mutations cause type 1 Jervell and Lange-Nielsen syndrome (JLNS) with deafness and higher sudden cardiac death risk. KCNQ1 variants causing JLNS or LQT1 might have distinct phenotypic expressions in heterozygous patients. The aim of this study is to evaluate QTc duration and incidence of long QT syndrome-related cardiac events according to genetic presentation. METHODS AND RESULTS: We enrolled LQT1 or JLNS patients with class IV/V KCNQ1 variants from our inherited arrhythmia clinic (September 1993 to January 2023). Medical history, ECG, and follow-up were collected. Additionally, we conducted a thorough literature review for JLNS variants. Survival curves were compared between groups, and multivariate Cox regression models identified genetic and clinical risk factors. Among the 789 KCNQ1 variant carriers, 3 groups were identified: 30 JLNS, 161 heterozygous carriers of JLNS variants (HTZ-JLNS), and 550 LQT1 heterozygous carriers of non-JLNS variants (HTZ-Non-JLNS). At diagnosis, mean age was 3.4 ± 4.7 years for JLNS, 26.7 ± 21 years for HTZ-JLNS, and 26 ± 21 years for HTZ-non-JLNS; 55.3% were female; and the mean QTc was 551 ± 54 ms for JLNS, 441 ± 32 ms for HTZ-JLNS, and 467 ± 36 ms for HTZ-Non-JLNS. Patients with heterozygous JLNS mutations (HTZ-JLNS) represented 22% of heterozygous KCNQ1 variant carriers and had a lower risk of cardiac events than heterozygous non-JLNS variant carriers (HTZ-Non-JLNS) [hazard ratio (HR) = 0.34 (0.22-0.54); P < 0.01]. After multivariate analysis, four genetic parameters were independently associated with events: haploinsufficiency [HR = 0.60 (0.37-0.97); P = 0.04], pore localization [HR = 1.61 (1.14-1.2.26); P < 0.01], C-terminal localization [HR = 0.67 (0.46-0.98); P = 0.04], and group [HR = 0.43 (0.27-0.69); P < 0.01]. CONCLUSION: Heterozygous carriers of JLNS variants have a lower risk of cardiac arrhythmic events than other LQT1 patients.


Asunto(s)
Canal de Potasio KCNQ1 , Síndrome de Romano-Ward , Humanos , Canal de Potasio KCNQ1/genética , Femenino , Masculino , Medición de Riesgo , Síndrome de Romano-Ward/genética , Síndrome de Romano-Ward/fisiopatología , Síndrome de Romano-Ward/diagnóstico , Factores de Riesgo , Niño , Electrocardiografía , Preescolar , Heterocigoto , Mutación , Síndrome de Jervell-Lange Nielsen/genética , Síndrome de Jervell-Lange Nielsen/fisiopatología , Predisposición Genética a la Enfermedad , Lactante , Adulto , Adolescente , Fenotipo , Estudios Retrospectivos , Muerte Súbita Cardíaca/etiología , Adulto Joven , Incidencia
2.
Hum Mutat ; 41(4): 850-859, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31930659

RESUMEN

Recently, four SCN5A mutations have been associated with Multifocal Ectopic Purkinje-related Premature Contractions (MEPPC), a rare cardiac syndrome combining polymorphic ventricular arrhythmia with dilated cardiomyopathy (DCM). Here, we identified a novel heterozygous mutation in SCN5A (c.611C>A, pAla204Glu) in a young woman presenting with polymorphic premature ventricular contractions (PVCs) and DCM. After failure of antiarrhythmic drugs and an attempt of radiofrequency catheter ablation showing three exit-sites of PVCs, all with presystolic Purkinje potentials, a treatment by hydroquinidine was tried, leading to an immediate and spectacular disappearance of all PVCs and normalization of cardiac function. Electrophysiological studies showed that Nav 1.5-A204E mutant channels exhibited a significant leftward shift of 8 mV of the activation curve, leading to a larger hyperpolarized window current when compared to wild-type. Action potential modeling using Purkinje fiber and ventricular cell models predicted an arrhythmogenic effect predominant in Purkinje fibers for the A204E mutation. Comparison with other MEPPC-associated Nav 1.5 mutations revealed a common electrophysiological pattern of abnormal voltage-dependence of activation leading to a larger hyperpolarized window current as a shared biophysical mechanism of this syndrome. These features of the mutant sodium channels are likely to be responsible for the hyperexcitability of the fascicular-Purkinje system observed in patients with MEPPC.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Ramos Subendocárdicos/metabolismo , Ramos Subendocárdicos/fisiopatología , Complejos Prematuros Ventriculares/diagnóstico , Complejos Prematuros Ventriculares/etiología , Adolescente , Alelos , Secuencia de Bases , Análisis Mutacional de ADN , Electrocardiografía , Femenino , Mutación con Ganancia de Función , Estudios de Asociación Genética/métodos , Pruebas Genéticas , Genotipo , Humanos , Imagen por Resonancia Magnética , Canal de Sodio Activado por Voltaje NAV1.5 , Fenotipo , Complejos Prematuros Ventriculares/tratamiento farmacológico
3.
Europace ; 22(2): 320-329, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31845994

RESUMEN

AIMS: Desmoglein-2 (DSG2) mutations, which encode a heart-specific cadherin crucial for desmosomal adhesion, are frequent in arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D). DSG2 mutations have been associated with higher risk of biventricular involvement. Among DSG2 mutations, mutations of the inhibitory propeptide consensus cleavage-site (Arg-X-Arg/Lys-Arg), are particularly frequent. In the present work, we explored the functional consequences of DSG2 propeptide cleavage site mutations p.Arg49His, p.Arg46Trp, and p.Arg46Gln on localization, adhesive properties, and desmosome incorporation of DSG2. METHODS AND RESULTS: We studied the expression of mutant-DSG2 in human heart and in epithelial and cardiac cellular models expressing wild-type or mutant (p.Arg49His, p.Arg46Trp, and p.Arg46Gln) proDSG2-GFP fusion proteins. The consequences of the p.Arg46Trp mutation on DSG2 adhesiveness were studied by surface plasmon resonance. Incorporation of mutant p.Arg46Trp DSG2 into desmosomes was studied under low-calcium culture conditions and cyclic mechanical stress. We demonstrated in human heart and cellular models that all three mutations prevented N-terminal propeptide cleavage, but did not modify intercellular junction targeting. Surface plasmon resonance experiments showed a propeptide-dependent loss of interaction between the cadherin N-terminal extracellular 1 (EC1) domains. Additionally, proDSG2 mutant proteins were abnormally incorporated into desmosomes under low-calcium culture conditions or following mechanical stress. This was accompanied by an epidermal growth factor receptor-dependent internalization of proDSG2, suggesting increased turnover of unprocessed proDSG2. CONCLUSION: Our results strongly suggest weakened desmosomal adhesiveness due to abnormal incorporation of uncleaved mutant proDSG2 in cellular stress conditions. These results provide new insights into desmosomal cadherin regulation and ARVC/D pathophysiology, in particular, the potential role of mechanical stress on desmosomal dysfunction.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Desmogleína 2 , Displasia Ventricular Derecha Arritmogénica/genética , Desmogleína 2/genética , Corazón , Humanos , Mutación
4.
Eur Heart J ; 39(31): 2879-2887, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30059973

RESUMEN

Aims: To clarify the clinical characteristics and outcomes of children with SCN5A-mediated disease and to improve their risk stratification. Methods and results: A multicentre, international, retrospective cohort study was conducted in 25 tertiary hospitals in 13 countries between 1990 and 2015. All patients ≤16 years of age diagnosed with a genetically confirmed SCN5A mutation were included in the analysis. There was no restriction made based on their clinical diagnosis. A total of 442 children {55.7% boys, 40.3% probands, median age: 8.0 [interquartile range (IQR) 9.5] years} from 350 families were included; 67.9% were asymptomatic at diagnosis. Four main phenotypes were identified: isolated progressive cardiac conduction disorders (25.6%), overlap phenotype (15.6%), isolated long QT syndrome type 3 (10.6%), and isolated Brugada syndrome type 1 (1.8%); 44.3% had a negative electrocardiogram phenotype. During a median follow-up of 5.9 (IQR 5.9) years, 272 cardiac events (CEs) occurred in 139 (31.5%) patients. Patients whose mutation localized in the C-terminus had a lower risk. Compound genotype, both gain- and loss-of-function SCN5A mutation, age ≤1 year at diagnosis in probands and age ≤1 year at diagnosis in non-probands were independent predictors of CE. Conclusion: In this large paediatric cohort of SCN5A mutation-positive subjects, cardiac conduction disorders were the most prevalent phenotype; CEs occurred in about one-third of genotype-positive children, and several independent risk factors were identified, including age ≤1 year at diagnosis, compound mutation, and mutation with both gain- and loss-of-function.


Asunto(s)
Trastorno del Sistema de Conducción Cardíaco/genética , Estudios de Asociación Genética , Canal de Sodio Activado por Voltaje NAV1.5/genética , Factores de Edad , Enfermedades Asintomáticas , Síndrome de Brugada/genética , Niño , Preescolar , Electrocardiografía , Femenino , Estudios de Seguimiento , Mutación con Ganancia de Función , Humanos , Lactante , Recién Nacido , Síndrome de QT Prolongado/genética , Mutación con Pérdida de Función , Masculino , Estudios Retrospectivos , Factores de Riesgo
5.
Eur Heart J ; 37(18): 1456-64, 2016 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-26715165

RESUMEN

AIMS: Acquired long QT syndrome (aLQTS) exhibits QT prolongation and Torsades de Pointes ventricular tachycardia triggered by drugs, hypokalaemia, or bradycardia. Sometimes, QTc remains prolonged despite elimination of triggers, suggesting the presence of an underlying genetic substrate. In aLQTS subjects, we assessed the prevalence of mutations in major LQTS genes and their probability of being carriers of a disease-causing genetic variant based on clinical factors. METHODS AND RESULTS: We screened for the five major LQTS genes among 188 aLQTS probands (55 ± 20 years, 140 females) from Japan, France, and Italy. Based on control QTc (without triggers), subjects were designated 'true aLQTS' (QTc within normal limits) or 'unmasked cLQTS' (all others) and compared for QTc and genetics with 2379 members of 1010 genotyped congenital long QT syndrome (cLQTS) families. Cardiac symptoms were present in 86% of aLQTS subjects. Control QTc of aLQTS was 453 ± 39 ms, shorter than in cLQTS (478 ± 46 ms, P < 0.001) and longer than in non-carriers (406 ± 26 ms, P < 0.001). In 53 (28%) aLQTS subjects, 47 disease-causing mutations were identified. Compared with cLQTS, in 'true aLQTS', KCNQ1 mutations were much less frequent than KCNH2 (20% [95% CI 7-41%] vs. 64% [95% CI 43-82%], P < 0.01). A clinical score based on control QTc, age, and symptoms allowed identification of patients more likely to carry LQTS mutations. CONCLUSION: A third of aLQTS patients carry cLQTS mutations, those on KCNH2 being more common. The probability of being a carrier of cLQTS disease-causing mutations can be predicted by simple clinical parameters, thus allowing possibly cost-effective genetic testing leading to cascade screening for identification of additional at-risk family members.


Asunto(s)
Síndrome de QT Prolongado , Electrocardiografía , Femenino , Francia , Pruebas Genéticas , Humanos , Italia , Japón , Masculino , Persona de Mediana Edad , Mutación
6.
Scand J Clin Lab Invest ; 75(2): 162-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25608598

RESUMEN

OBJECTIVES: To determine the hemolysis interference on biochemical tests and immunoassays performed on Roche Diagnostics analyzers, according to different maximum allowable limits. DESIGN AND METHODS: Heparinized plasma and serum pools, free of interferences, were overloaded by increasing amounts of a hemoglobin-titrated hemolysate. This interference was evaluated for 45 analytes using Modular(®) and Cobas(®) analyzers. For each parameter, the hemolysis index (HI) corresponding to the traditional ± 10% change of concentrations from baseline (± 10%Δ) was determined, as well as those corresponding to the analytical change limit (ACL), and to the reference change value (RCV). Then, the relative frequencies distribution (% RFD) of hemolyzed tests performed in a hospital laboratory over a 25-day period were established for each HI as allowable limit. RESULTS: Considering the ± 10%Δ, the analyte concentrations enhanced by hemolysis were: Lactate dehydrogenase (LDH), aspartate aminotransferase (AST), folate, potassium, creatine kinase, phosphorus, iron, alanine aminotransferase, lipase, magnesium and triglycerides, decreasingly. The analyte concentrations decreased by hemolysis were: Haptoglobin, high-sensitive troponin T and alkaline phosphatase. Over the 25-day period, the % RFD of tests impacted more than 10%Δ by hemolysis were < 7% for LDH; < 5% for AST, folates and iron; and < 1% for the other analytes. Considering the ACL, HI were lower, giving % RFD substantially increased for many analytes, whereas only four analytes remain sensitive to hemolysis when considering RCV. CONCLUSION: This study proposes new HI based on different allowable limits, and can therefore serve as a starting point for future harmonization of hemolysis interference evaluation needed in routine laboratory practice.


Asunto(s)
Análisis Químico de la Sangre/normas , Hemólisis , Inmunoensayo/instrumentación , Artefactos , Aspartato Aminotransferasas/sangre , Análisis Químico de la Sangre/instrumentación , Análisis Químico de la Sangre/métodos , Haptoglobinas/análisis , Humanos , Inmunoensayo/métodos , Inmunoensayo/normas , L-Lactato Deshidrogenasa/sangre , Valores de Referencia , Troponina T/sangre
8.
Arch Cardiovasc Dis ; 117(6-7): 382-391, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670870

RESUMEN

BACKGROUND: With the development of advanced sequencing techniques, genetic testing has emerged as a valuable tool for the work-up of non-ischaemic sudden cardiac arrest (SCA). AIMS: To evaluate the effectiveness of genetic testing in patients with unexplained SCA, according to clinical phenotype. METHODS: All patients who underwent molecular genetic testing for non-ischaemic SCA with no left ventricular cardiomyopathy between 2012 and 2021 in two French university hospitals were included. RESULTS: Of 66 patients (mean age 36.7±11.9years, 54.5% men), 21 (31.8%; 95% confidence interval 22.4-45.3%) carried a genetic variant: eight (12.1%) had a pathogenic or likely pathogenic (P/LP) variant and 13 (19.7%) had a variant of uncertain significance (VUS). Among 37 patients (56.1%) with no phenotypic clues, genetic testing identified a P/LP variant in five (13.5%), mainly in RYR2 (n=3) and SCN5A (n=2), and a VUS in nine (24.3%). None of the nine patients with phenotypic evidence of channelopathies had P/LP variants, but two had VUS in RYR2 and NKX2.5. Among the 20 patients with suspected arrhythmogenic cardiomyopathy, three P/LP variants (15.0%) and two VUS (10.0%) were found in DSC2, PKP2, SCN5A and DSG2, TRPM4, respectively. Genetic testing was performed sooner after cardiac arrest (P<0.001) and results were obtained more rapidly (P=0.02) after versus before 2016. CONCLUSION: This study highlights the utility of molecular genetic testing with a genetic variant of interest identified in one-third of patients with unexplained SCA. Genetic testing was beneficial even in patients without phenotypic clues, with one-fourth of patients carrying a P/LP variant that could have direct implications.


Asunto(s)
Muerte Súbita Cardíaca , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Fenotipo , Valor Predictivo de las Pruebas , Humanos , Masculino , Femenino , Muerte Súbita Cardíaca/etiología , Muerte Súbita Cardíaca/prevención & control , Adulto , Persona de Mediana Edad , Francia , Adulto Joven , Factores de Riesgo , Hospitales Universitarios , Estudios Retrospectivos , Mutación , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal de Sodio Activado por Voltaje NAV1.5/genética , Marcadores Genéticos
9.
Heart Rhythm ; 21(3): 313-320, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37956775

RESUMEN

BACKGROUND: The efficacy of beta-blocker treatment in type 3 long QT syndrome (LQT3) remains debated. OBJECTIVES: The purpose of this study was to test the hypothesis that beta-blocker use is associated with cardiac events (CEs) in a French cohort of LQT3 patients. METHODS: All patients with a likely pathogenic/pathogenic variant in the SCN5A gene (linked to LQT3) were included and followed-up. Documented ventricular tachycardia/ventricular fibrillation, torsades de pointes, aborted cardiac arrest, sudden death, and appropriate shocks were considered as severe cardiac events (SCEs). CEs also included syncope. RESULTS: We included 147 patients from 54 families carrying 23 variants. Six of the patients developed symptoms before the age of 1 year and were analyzed separately. The 141 remaining patients (52.5% male; median age at diagnosis 24.0 years) were followed-up for a median of 11 years. The probabilities of a CE and an SCE from birth to the age of 40 were 20.5% and 9.9%, respectively. QTc prolongation (hazard ratio [HR] 1.12 [1.0-1.2]; P = .005]) and proband status (HR 4.07 [1.9-8.9]; P <.001) were independently associated with the occurrence of CEs. Proband status (HR 8.13 [1.7-38.8]; P = .009) was found to be independently associated with SCEs, whereas QTc prolongation (HR 1.11 [1.0-1.3]; P = .108) did not reach statistical significance. The cumulative probability of the age at first CE/SCE was not lower in patients treated with a beta-blocker. CONCLUSION: In agreement with the literature, proband status and lengthened QTc were associated with a higher risk of CEs. Our data do not show a protective effect of beta-blocker treatment.


Asunto(s)
Paro Cardíaco , Síndrome de QT Prolongado , Humanos , Masculino , Adulto Joven , Adulto , Femenino , Electrocardiografía , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/diagnóstico , Síncope , Paro Cardíaco/complicaciones , Antagonistas Adrenérgicos beta/uso terapéutico
10.
Europace ; 15(10): 1522-5, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23858024

RESUMEN

AIMS: Arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D) is an inherited cardiomyopathy characterized by fibro-fatty replacement of the right ventricle and ventricular arrhythmias. The major disease-causing genes encode cardiac desmosomal components but are involved in only ∼50% of patients. To identify the missing genetic determinants, we used a candidate gene approach, focusing on the 3'-untranslated region (UTR) of the main ARVC/D gene PKP2 and on additional genes involved in desmosomal structure or function. METHODS AND RESULTS: We screened a population of 64 ARVC/D probands with no identified mutations in any of the five known desmosomal genes (PKP2, DSG2, DSP, DSC2, and JUP). No putative mutation was identified in the 3'-UTR of PKP2 or in PNN, CTNNA3, CAV1, or PLN coding sequences. In a single proband, we identified two rare heterozygous missense variants affecting evolutionary conserved residues: c.175G>A (p.Gly59Arg) in PERP and c.1811A>G (p.Asp604Gly) in PKP4 (minor allele frequency <0.5% in control population). CONCLUSION: Our study suggests that mutations in the candidate genes studied and regulation of PKP2 mRNA via 3'-UTR dependent mechanisms are unlikely to be major causes of ARVC/D in the studied population. Additional studies are needed to investigate the putative effects of rare PKP4 and PERP variants in this disease.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/genética , Pruebas Genéticas , Secuencia de Aminoácidos , Análisis Mutacional de ADN , Frecuencia de los Genes , Genes Supresores de Tumor , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Heterocigoto , Humanos , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Mutación Missense , Placofilinas/genética
11.
Europace ; 15(12): 1805-11, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23867365

RESUMEN

AIMS: Andersen-Tawil syndrome (ATS) is an uncommon form of channelopathy linked to mutations in the KCNJ2 gene. Currently, little is known about the long-term arrhythmic prognosis of this disease. METHODS AND RESULTS: We conducted a retrospective multicentre study in nine French hospitals. Patients were recruited only if they were KCNJ2 mutation carriers. Thirty-six patients (female n = 22, 61%) from 20 unrelated kindred were included with a mean follow-up of 9.5 ± 8.2 years. We found 12 distinct KCNJ2 mutations in the 20 probands. Three of them were novel. Thirteen patients (36%) experienced syncope and one patient was resuscitated from cardiac arrest before diagnosis. The mean QTc interval was 439 ± 57 ms and QUc was 642 ± 64 ms. All patients had normal ejection fraction. Holter recordings in 33 patients found 11 272 premature ventricular complexes (PVCs) per day on average, 25 patients had episodes of bigeminy, and 25 patients had polymorphic PVCs. Twenty-three patients (70%) had non-sustained polymorphic ventricular tachycardia (VT), and six sustained polymorphic VT. Only one patient presented with torsades de pointes. Patients were treated with beta-blocker (n = 20), beta-blocker and amiodarone (n = 2), beta-blocker and flecainide (n = 6), or acetazolamide (n = 6). Radiofrequency ablation was attempted in five patients without clinical success. An implantable cardiac defibrillator was implanted in three patients. During follow-up, none of the patients died, four patients experienced syncope under treatment, and one patient had non-fatal cardiac arrest. CONCLUSION: Despite a severe clinical presentation with a very high rate of ventricular arrhythmias, the arrhythmic prognosis of the ATS patients is relatively good under treatment.


Asunto(s)
Síndrome de Andersen/genética , Mutación , Canales de Potasio de Rectificación Interna/genética , Adolescente , Adulto , Anciano , Síndrome de Andersen/complicaciones , Síndrome de Andersen/diagnóstico , Síndrome de Andersen/fisiopatología , Síndrome de Andersen/terapia , Niño , Preescolar , Análisis Mutacional de ADN , Electrocardiografía , Femenino , Francia , Predisposición Genética a la Enfermedad , Paro Cardíaco/genética , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Pronóstico , Estudios Retrospectivos , Síncope/genética , Factores de Tiempo , Adulto Joven
12.
Arch Cardiovasc Dis ; 116(1): 9-17, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36609000

RESUMEN

BACKGROUND: Few data exist on the characteristics and outcomes of patients with arrhythmogenic right ventricular cardiomyopathy and advanced heart failure who undergo heart transplantation. AIM: To explore the pretransplant course and outcomes of patients with arrhythmogenic right ventricular cardiomyopathy after heart transplantation. METHODS: This observational retrospective monocentric study included all consecutive patients with arrhythmogenic right ventricular cardiomyopathy who underwent heart transplantation during a 13-year period (2006-2019) at Pitié-Salpêtrière University Hospital (Paris). RESULTS: A total of 23 patients with arrhythmogenic right ventricular cardiomyopathy underwent heart transplantation between 2006 and 2019. The median time from diagnosis to heart transplantation was 9 years, and the median age at transplantation was 50 years. At diagnosis, half of the patients had left ventricular dysfunction, 59% had extensive T-wave inversion and 43% had a history of sustained ventricular tachycardia. Only five patients were involved in intensive sport activity. Indications for heart transplantation were end-stage biventricular dysfunction in 13 patients, end-stage right ventricular heart failure in seven and electrical storm in three. Only three patients had pulmonary hypertension, and half of the patients had atrial arrhythmias. The survival rate 1 year after heart transplantation was 74% (95% confidence interval 53-88%). Eight patients experienced primary graft dysfunction needing extracorporeal membrane oxygenation. CONCLUSIONS: Patients with arrhythmogenic right ventricular cardiomyopathy who eventually needed heart transplantation mostly exhibited extended disease with biventricular dysfunction at diagnosis. Intensive sport activity did not seem to be a major determinant. Advanced heart failure usually occurred late in the course of the disease. Primary graft dysfunction after heart transplantation was frequent, and should be anticipated. Additional data are needed to identify the optimal timing for heart transplantation and predictors of end-stage heart failure in patients with arrhythmogenic right ventricular cardiomyopathy.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Insuficiencia Cardíaca , Trasplante de Corazón , Disfunción Primaria del Injerto , Humanos , Persona de Mediana Edad , Displasia Ventricular Derecha Arritmogénica/complicaciones , Displasia Ventricular Derecha Arritmogénica/diagnóstico , Displasia Ventricular Derecha Arritmogénica/cirugía , Estudios Retrospectivos , Trasplante de Corazón/efectos adversos , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/cirugía , Progresión de la Enfermedad
13.
Am J Cardiol ; 190: 113-120, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36621286

RESUMEN

The relations between endocardial voltage mapping and the genetic background of patients with arrhythmogenic right ventricular cardiomyopathy (ARVC) have not been investigated so far. A total of 97 patients with proved or suspected ARVC who underwent 3-dimensional endocardial mapping and genetic testing have been retrospectively included. Presence, localization, and size of scar areas were correlated to ARVC diagnosis and the presence of a pathogenic variant. A total of 78 patients (80%) presented with some bipolar or unipolar scar on endocardial voltage mapping, whereas 43 carried pathogenic variants (44%). Significant associations were observed between presence of endocardial scars on voltage mapping and previous or inducible ventricular tachycardia, right ventricular function and dimensions, or electrocardiogram features of ARVC. A total of 60 of the 78 patients (77%) with an endocardial scar fulfilled the criteria for a definitive arrhythmogenic right ventricular dysplasia diagnosis versus 8 of 19 patients (42%) without scar (p = 0.003). Patients with a definitive diagnosis of ARVC had more scars from any location and the scars were larger in patients with ARVC. In the 68 patients with a definitive diagnosis of ARVC, the presence of any endocardial scar was similar whether an ARVC-causal mutation was present or not. Only scar extent was significantly greater in patients with pathogenic variants. There was no difference in the presence and characteristics of scars in PKP2 mutated versus other mutated patients. The 3-dimensional endocardial mapping could have an important role for refining ARVC diagnosis and may be able to detect minor forms with otherwise insufficient criteria for diagnosis. The trend for larger scar extent were observed in mutated patients, without any difference according to the mutated genes.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Ablación por Catéter , Taquicardia Ventricular , Humanos , Displasia Ventricular Derecha Arritmogénica/diagnóstico , Displasia Ventricular Derecha Arritmogénica/genética , Cicatriz/complicaciones , Estudios Retrospectivos , Técnicas Electrofisiológicas Cardíacas/métodos , Endocardio/patología , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/genética , Ablación por Catéter/efectos adversos
14.
J Cardiovasc Transl Res ; 16(6): 1276-1286, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37418234

RESUMEN

The presence of multiple pathogenic variants in desmosomal genes (DSC2, DSG2, DSP, JUP, and PKP2) in patients with arrhythmogenic right ventricular cardiomyopathy (ARVC) has been linked to a severe phenotype. However, the pathogenicity of variants is reclassified frequently, which may result in a changed clinical risk prediction. Here, we present the collection, reclassification, and clinical outcome correlation for the largest series of ARVC patients carrying multiple desmosomal pathogenic variants to date (n = 331). After reclassification, only 29% of patients remained carriers of two (likely) pathogenic variants. They reached the composite endpoint (ventricular arrhythmias, heart failure, and death) significantly earlier than patients with one or no remaining reclassified variant (hazard ratios of 1.9 and 1.8, respectively). Periodic reclassification of variants contributes to more accurate risk stratification and subsequent clinical management strategy. Graphical Abstract.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Humanos , Displasia Ventricular Derecha Arritmogénica/diagnóstico , Displasia Ventricular Derecha Arritmogénica/genética , Placofilinas/genética , Fenotipo , Arritmias Cardíacas , Mutación
15.
Biochemistry ; 51(45): 9076-85, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23092362

RESUMEN

Type 1 long QT syndrome (LQT1) is caused by loss-of-function mutations in the KCNQ1 gene, which encodes the K(+) channel (Kv7.1) that underlies the slowly activating delayed rectifier K(+) current in the heart. Intragenic risk stratification suggests LQT1 mutations that disrupt conserved amino acid residues in the pore are an independent risk factor for LQT1-related cardiac events. The purpose of this study is to determine possible molecular mechanisms that underlie the loss of function for these high-risk mutations. Extensive genotype-phenotype analyses of LQT1 patients showed that T322M-, T322A-, or G325R-Kv7.1 confers a high risk for LQT1-related cardiac events. Heterologous expression of these mutations with KCNE1 revealed they generated nonfunctional channels and caused dominant negative suppression of WT-Kv7.1 current. Molecular dynamics simulations of analogous mutations in KcsA (T85M-, T85A-, and G88R-KcsA) demonstrated that they disrupted the symmetrical distribution of the carbonyl oxygen atoms in the selectivity filter, which upset the balance between the strong attractive and K(+)-K(+) repulsive forces required for rapid K(+) permeation. We conclude high-risk LQT1 mutations in the pore likely disrupt the architectural and physical properties of the K(+) channel selectivity filter.


Asunto(s)
Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/fisiología , Potasio/metabolismo , Adolescente , Adulto , Secuencia de Aminoácidos , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Simulación de Dinámica Molecular , Mutación Missense , Canales de Potasio con Entrada de Voltaje/genética , Riesgo , Síndrome de Romano-Ward/genética , Alineación de Secuencia
16.
Br J Sports Med ; 46 Suppl 1: i59-68, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23097481

RESUMEN

The sudden cardiac death (SCD) of an athlete is a rare and tragic event, often caused by a number of inherited heart muscle disorders, namely the cardiomyopathies and primary arrhythmia syndromes (also known as cardiac ion channelopathies). Recent advances in the understanding of the molecular genetics of these heritable cardiovascular diseases present new challenges for clinicians who manage athletes with these types of heart muscle conditions. Unfortunately, the clinical heterogeneity of many of these SCD diseases are also matched by the genotypic heterogeneity associated with the pathogenesis of the disease. A particularly challenging situation arises when the sports physician and attending cardiologist are presented with an athlete who demonstrates a familial context of inherited cardiac disease or presents mild cardiac abnormalities suggestive of inherited cardiac disease. Alongside the complete cardiac evaluation, genetic testing may be proposed as an additional diagnostic tool in this clinical conundrum. However, debate still remains on how extensive the screening should be, in particular the use and interpretation of genetic testing in that setting. The aim of this review is to examine the role of genetic testing within the diagnostic algorithm of preparticipation screening of athletes. This will be achieved by providing the sports medicine physician with simple current cardiac genetic knowledge for the main inherited cardiac conditions known to cause SCD. Furthermore, it will examine current knowledge for the role of genetic testing upon the prediction of SCD, concluding with its impact on the sport eligibility and disqualification conundrum using case examples from our genetic testing laboratory.


Asunto(s)
Arritmias Cardíacas/diagnóstico , Cardiomiopatías/diagnóstico , Muerte Súbita Cardíaca/prevención & control , Pruebas Genéticas/métodos , Deportes/fisiología , Adolescente , Adulto , Arritmias Cardíacas/genética , Arritmias Cardíacas/terapia , Atletas , Cardiomiopatías/genética , Cardiomiopatías/terapia , Diagnóstico Precoz , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Masculino , Mutación/genética , Penetrancia , Fenotipo , Factores de Riesgo
17.
Circ Arrhythm Electrophysiol ; 15(3): e010572, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35225649

RESUMEN

BACKGROUND: CaM (calmodulin), encoded by 3 separate genes (CALM1, CALM2, and CALM3), is a multifunctional Ca2+-binding protein involved in many signal transduction events including ion channel regulation. CaM variants may present with early-onset long QT syndrome (LQTS), catecholaminergic polymorphic ventricular tachycardia, or sudden cardiac death. Most reported variants occurred de novo. We identified a novel CALM3 variant, p.Asn138Lys (N138K), in a 4-generation family segregating with LQTS. The aim of this study was to elucidate its pathogenicity and to compare it with that of p.D130G-CaM-a variant associated with a severe LQTS phenotype. METHODS: We performed whole exome sequencing for a large, 4-generation family affected by LQTS. To assess the effect of the detected CALM3 variant, the intrinsic Ca2+-binding affinity was measured by stoichiometric Ca2+ titrations and equilibrium titrations. L-type Ca2+ and slow delayed rectifier potassium currents (ICaL and IKs) were recorded by whole-cell patch-clamp. Cav1.2 and Kv7.1 membrane expression were determined by optical fluorescence assays. RESULTS: We identified 14 p.N138K-CaM carriers in a family where 2 sudden deaths occurred in children. Several members were only mildly affected compared with CaM-LQTS patients to date described in literature. The intrinsic Ca2+-binding affinity of the CaM C-terminal domain was 10-fold lower for p.N138K-CaM compared with wild-type-CaM. ICaL inactivation was slowed in cells expressing p.N138K-CaM but less than in p.D130G-CaM cells. Unexpectedly, a larger IKs current density was observed in cells expressing p.N138K-CaM, but not for p.D130G-CaM, compared with wild-type-CaM. CONCLUSIONS: The p.N138K CALM3 variant impairs Ca2+-binding affinity of CaM and ICaL inactivation but potentiates IKs. The variably expressed phenotype of this variant compared with previously published de novo LQTS-CaM variants is likely explained by a milder impairment of ICaL inactivation combined with IKs augmentation.


Asunto(s)
Calmodulina/genética , Síndrome de QT Prolongado , Taquicardia Ventricular , Calmodulina/metabolismo , Humanos , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/genética , Mutación , Miocitos Cardíacos/metabolismo , Fenotipo , Taquicardia Ventricular/etiología
18.
Eur Heart J Case Rep ; 5(10): ytab393, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34729453

RESUMEN

BACKGROUND: Calmodulinopathy is an emerging group of primary electrical disease with various, severe, and early onset phenotype. Sudden cardiac arrest (SCA)/death can be the first symptom and current medical management seems insufficient to prevent recurrences. Implantable cardioverter-defibrillator (ICD) in the young is challenging and can be harmful. CASE SUMMARY: We report the management of two very young boys (aged 3.5 and 5.5 years old) who survived an SCA due to calmodulin mutation responsible of a catecholaminergic polymorphic ventricular tachycardia phenotype. In both case, SCA had an adrenergic trigger. Despite SCA, ICD implantation was denied by the parents. After thorough discussion with the family, the patients were managed with solely betablocker treatment and loop recorder implantation. At last follow-up of 30 and 23 months, respectively, there were no recurrence of any cardiac event. DISCUSSION: The benefits of ICD implantation at a very young age must be weighed against the risk complication. In the youngest, whom recreative activities are under constant supervision, the decision, jointly made with the parents, could be to postpone ICD.

19.
Ann Biol Clin (Paris) ; 79(1): 28-40, 2021 Feb 01.
Artículo en Francés | MEDLINE | ID: mdl-33586649

RESUMEN

The molecular study of mitochondrial diseases, essential for diagnosis, is special due to the dual genetic origin of these pathologies: mitochondrial DNA and nuclear DNA. Complete mtDNA sequencing still remains the first line diagnostic test followed if negative, by resequencing panels of several hundred mitochondrially-encoded nuclear genes. This strategy, with an initial entire mtDNA sequencing, is currently justified by the presence of nuclear mitochondrial DNA sequences (NUMTs) in the nuclear genome. We designed a resequencing panel combining the mtDNA and 135 nuclear genes which was evaluated compared to the performances of the standard mtDNA sequencing. Method validation was performed on the reading depth and reproducibility of the results. Thirty patients were analyzed by both methods. We were able to demonstrate that NUMTs did not impact the mtDNA sequencing quality, as the identified variants and mutant loads were identical with the reference mtDNA sequencing method. Reading depths were higher than the recommendations of the MitoDiag French diagnostic network, for the entire mtDNA for muscle and for 70% of the mtDNA for blood. These results highlight the usefulness of combining both mtDNA and mitochondrially nuclear-encoded genes and thus obtain more complete results and faster turnaround time for mitochondrial disease patients.


Asunto(s)
Genoma Mitocondrial , Enfermedades Mitocondriales , ADN Mitocondrial/genética , Genoma Mitocondrial/genética , Humanos , Mitocondrias , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/genética , Reproducibilidad de los Resultados
20.
CJC Open ; 3(3): 256-266, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33778442

RESUMEN

BACKGROUND: Nav1.5, which is encoded by the SCN5A gene, is the predominant voltage-gated Na+ channel in the heart. Several mutations of this gene have been identified and reported to be involved in several cardiac rhythm disorders, including type 3 long QT interval syndrome, that can cause sudden cardiac death. We analyzed the biophysical properties of 2 novel variants of the Nav1.5 channel (Q1491H and G1481V) detected in 5- and 12-week-old infants diagnosed with a prolonged QT interval. METHODS: The Nav1.5 wild-type and the Q1491H and G1481V mutant channels were reproduced in vi tr o. Wild-type or mutant channels were cotransfected in human embryonic kidney (HEK) 293 cells with the beta 1 regulatory subunit. Na+ currents were recorded using the whole-cell configuration of the patch-clamp technique. RESULTS: The Q1491H mutant channel exhibited a lower current density, a persistent Na+ current, an enhanced window current due to a +20-mV shift of steady-state inactivation, a +10-mV shift of steady-state activation, a faster onset of slow inactivation, and a recovery from fast inactivation with fast and slow time constants of recovery. The G1481V mutant channel exhibited an increase in current density and a +7-mV shift of steady-state inactivation. The observed defects are characteristic of gain-of-function mutations typical of type 3 long QT interval syndrome. CONCLUSIONS: The 5- and 12-week-old infants displayed prolonged QT intervals. Our analyses of the Q1491H and G1481V mutations correlated with the clinical diagnosis. The observed biophysical dysfunctions associated with both mutations were most likely responsible for the sudden deaths of the 2 infants.


INTRODUCTION: Le canal Nav1.5, codé par le gène SCN5A, est le canal Na+ dépendant du voltage prédominant dans le cœur. Plusieurs mutations de ce gène sont impliquées dans plusieurs anomalies du rythme cardiaque, dont le syndrome du QT long de type 3, qui peut provoquer la mort subite d'origine cardiaque. Nous avons analysé les propriétés biophysiques de deux nouveaux variants du canal Nav1.5 (Q1491H et G1481V) détectés chez deux bébés âgés respectivement de 5 et 12 semaines qui avaient une prolongation de l'intervalle QT. MÉTHODES: Le canal Nav1.5 de type sauvage et les canaux mutants Q1491H et G1481V ont été reproduits in vi tr o. Les canaux de type sauvage ou mutants ont été co-transfectés dans les cellules des reins embryonnaires humains (REH) 293 avec la sous-unité régulatrice bêta 1. Les courants Na+ ont été enregistrés à partir de la configuration en cellule entière via la technique de patch-clamp. RÉSULTATS: Le canal mutant Q1491H montre une densité de courant plus faible, un courant Na+ persistant, un courant fenêtre augmenté en raison d'un changement dép de +20 mV de l'inactivation à l'état stable, un changement de +10 mV de l'activation à l'état stable, une entrée plus rapide de l'inactivation lente et une récupération de l'inactivation rapide avec des constantes de temps rapides et lentes. Le canal mutant G1481V montre une augmentation de la densité de courant et un changement de +7 mV de l'inactivation à l'état stable. Les anomalies observées sont caractéristiques des mutations avec gain de fonction typiques du syndrome du QT long de type 3. CONCLUSIONS: Les deux bébés âgés respectivement de cinq 5 et 12 semaines montraient une prolongation des intervalles QT. Nos analyses des mutations Q1491H et G1481V montrent une corrélation avec le diagnostic clinique. Les dysfonctions biophysiques observées qui sont associées aux deux mutations étaient fort probablement responsables des morts subites des deux bébés.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA