Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Commun ; 15(1): 3380, 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38643172

RESUMEN

While 3D chromatin organization in topologically associating domains (TADs) and loops mediating regulatory element-promoter interactions is crucial for tissue-specific gene regulation, the extent of their involvement in human Mendelian disease is largely unknown. Here, we identify 7 families presenting a new cardiac entity associated with a heterozygous deletion of 2 CTCF binding sites on 4q25, inducing TAD fusion and chromatin conformation remodeling. The CTCF binding sites are located in a gene desert at 1 Mb from the Paired-like homeodomain transcription factor 2 gene (PITX2). By introducing the ortholog of the human deletion in the mouse genome, we recapitulate the patient phenotype and characterize an opposite dysregulation of PITX2 expression in the sinoatrial node (ectopic activation) and ventricle (reduction), respectively. Chromatin conformation assay performed in human induced pluripotent stem cell-derived cardiomyocytes harboring the minimal deletion identified in family#1 reveals a conformation remodeling and fusion of TADs. We conclude that TAD remodeling mediated by deletion of CTCF binding sites causes a new autosomal dominant Mendelian cardiac disorder.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Animales , Ratones , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Cromatina/genética , Proteínas de Unión al ADN/metabolismo , Genoma
2.
Stem Cell Res ; 77: 103396, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38522388

RESUMEN

Mutations in the DES gene, which encodes the intermediate filament desmin, lead to desminopathy, a rare disease characterized by skeletal muscle weakness and different forms of cardiomyopathies associated with cardiac conduction defects and arrhythmias. We generated human induced pluripotent stem cells (hiPSC) from a patient carrying the DES p.R406W mutation, and employed CRISPR/Cas9 to rectify the mutation in the patient's hiPSC line and introduced the mutation in an hiPSC line from a control individual unrelated to the patient. These hiPSC lines represent useful models for delving into the mechanisms of desminopathy and developing new therapeutic approaches.


Asunto(s)
Desmina , Células Madre Pluripotentes Inducidas , Mutación , Células Madre Pluripotentes Inducidas/metabolismo , Humanos , Desmina/metabolismo , Desmina/genética , Línea Celular , Sistemas CRISPR-Cas , Técnicas de Sustitución del Gen , Diferenciación Celular
3.
Cardiovasc Res ; 119(3): 759-771, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36001550

RESUMEN

AIMS: Degenerative mitral valve dystrophy (MVD) leading to mitral valve prolapse is the most frequent form of MV disease, and there is currently no pharmacological treatment available. The limited understanding of the pathophysiological mechanisms leading to MVD limits our ability to identify therapeutic targets. This study aimed to reveal the main pathophysiological pathways involved in MVD via the multimodality imaging and transcriptomic analysis of the new and unique knock-in (KI) rat model for the FilaminA-P637Q (FlnA-P637Q) mutation associated-MVD. METHODS AND RESULTS: Wild-type (WT) and KI rats were evaluated morphologically, functionally, and histologically between 3-week-old and 3-to-6-month-old based on Doppler echocardiography, 3D micro-computed tomography (microCT), and standard histology. RNA-sequencing and Assay for Transposase-Accessible Chromatin (ATAC-seq) were performed on 3-week-old WT and KI mitral valves and valvular cells, respectively, to highlight the main signalling pathways associated with MVD. Echocardiographic exploration confirmed MV elongation (2.0 ± 0.1 mm vs. 1.8 ± 0.1, P = 0.001), as well as MV thickening and prolapse in KI animals compared to WT at 3 weeks. 3D MV volume quantified by microCT was significantly increased in KI animals (+58% vs. WT, P = 0.02). Histological analyses revealed a myxomatous remodelling in KI MV characterized by proteoglycans accumulation. A persistent phenotype was observed in adult KI rats. Signalling pathways related to extracellular matrix homeostasis, response to molecular stress, epithelial cell migration, endothelial to mesenchymal transition, chemotaxis and immune cell migration, were identified based on RNA-seq analysis. ATAC-seq analysis points to the critical role of transforming growth factor-ß and inflammation in the disease. CONCLUSION: The KI FlnA-P637Q rat model mimics human myxomatous MVD, offering a unique opportunity to decipher pathophysiological mechanisms related to this disease. Extracellular matrix organization, epithelial cell migration, response to mechanical stress, and a central contribution of immune cells are highlighted as the main signalling pathways leading to myxomatous MVD. Our findings pave the road to decipher underlying molecular mechanisms and the specific role of distinct cell populations in this context.


Asunto(s)
Prolapso de la Válvula Mitral , Válvula Mitral , Adulto , Humanos , Ratas , Animales , Lactante , Válvula Mitral/metabolismo , Filaminas/genética , Filaminas/metabolismo , Transcriptoma , Microtomografía por Rayos X , Prolapso de la Válvula Mitral/patología , Fenotipo
4.
Hum Mutat ; 43(9): 1333-1342, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35819174

RESUMEN

Arrhythmogenic cardiomyopathy with right dominant form (ACR) is a rare heritable cardiac cardiomyopathy disorder associated with sudden cardiac death. Pathogenic variants (PVs) in desmosomal genes have been causally related to ACR in 40% of cases. Other genes encoding nondesmosomal proteins have been described in ACR, but their contribution in this pathology is still debated. A panel of 71 genes associated with inherited cardiopathies was screened in an ACR population of 172 probands and 856 individuals from the general population. PVs and uncertain significance variants (VUS) have been identified in 36% and 18.6% of patients, respectively. Among the cardiopathy-associated genes, burden tests show a significant enrichment in PV and VUS only for desmosomal genes PKP2 (plakophilin-2), DSP (desmoplakin), DSC2 (desmocollin-2), and DSG2 (desmoglein-2). Importantly, VUS may account for 15% of ACR cases and should then be considered for molecular diagnosis. Among the other genes, no evidence of enrichment was detected, suggesting an extreme caution in the interpretation of these genetic variations without associated functional or segregation data. Genotype-phenotype correlation points to (1) a more severe and earlier onset of the disease in PV and VUS carriers, underlying the importance to carry out presymptomatic diagnosis in relatives and (2) to a more prevalent left ventricular dysfunction in DSP variant carriers.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Displasia Ventricular Derecha Arritmogénica/diagnóstico , Displasia Ventricular Derecha Arritmogénica/genética , Displasia Ventricular Derecha Arritmogénica/metabolismo , Desmosomas/genética , Desmosomas/metabolismo , Estudios de Asociación Genética , Heterocigoto , Humanos , Placofilinas/genética , Placofilinas/metabolismo
5.
Circ Genom Precis Med ; 12(10): e002617, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-32141789

RESUMEN

BACKGROUND: Calcific aortic valve stenosis (CAVS) is a frequent and life-threatening cardiovascular disease for which there is currently no medical treatment available. To date, only 2 genes, LPA and PALMD, have been identified as causal for CAVS. We aimed to identify additional susceptibility genes for CAVS. METHODS: A GWAS (genome-wide association study) meta-analysis of 4 cohorts, totaling 5115 cases and 354 072 controls of European descent, was performed. A TWAS (transcriptome-wide association study) was completed to integrate transcriptomic data from 233 human aortic valves. A series of post-GWAS analyses were performed, including fine-mapping, colocalization, phenome-wide association studies, pathway, and tissue enrichment as well as genetic correlation with cardiovascular traits. RESULTS: In the GWAS meta-analysis, 4 loci achieved genome-wide significance, including 2 new loci: IL6 (interleukin 6) on 7p15.3 and ALPL (alkaline phosphatase) on 1p36.12. A TWAS integrating gene expression from 233 human aortic valves identified NAV1 (neuron navigator 1) on 1q32.1 as a new candidate causal gene. The CAVS risk alleles were associated with higher mRNA expression of NAV1 in valve tissues. Fine-mapping identified rs1800795 as the most likely causal variant in the IL6 locus. The signal identified colocalizes with the expression of the IL6 RNA antisense in various tissues. Phenome-wide association analyses in the UK Biobank showed colocalized associations between the risk allele at the IL6 lead variant and higher eosinophil count, pulse pressure, systolic blood pressure, and carotid artery procedures, implicating modulation of the IL6 pathways. The risk allele at the NAV1 lead variant colocalized with higher pulse pressure and higher prevalence of carotid artery stenosis. Association results at the genome-wide scale indicated genetic correlation between CAVS, coronary artery disease, and cardiovascular risk factors. CONCLUSIONS: Our study implicates 3 new genetic loci in CAVS pathogenesis, which constitute novel targets for the development of therapeutic agents.


Asunto(s)
Fosfatasa Alcalina/genética , Estenosis de la Válvula Aórtica/genética , Válvula Aórtica/patología , Calcinosis/genética , Interleucina-6/genética , Proteínas Asociadas a Microtúbulos/genética , Estudios de Cohortes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Polimorfismo de Nucleótido Simple
6.
J Am Heart Assoc ; 5(6)2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27287695

RESUMEN

BACKGROUND: The Brugada syndrome is an inherited cardiac arrhythmia associated with high risk of sudden death. Although 20% of patients with Brugada syndrome carry mutations in SCN5A, the molecular mechanisms underlying this condition are still largely unknown. METHODS AND RESULTS: We combined whole-exome sequencing and linkage analysis to identify the genetic variant likely causing Brugada syndrome in a pedigree for which SCN5A mutations had been excluded. This approach identified 6 genetic variants cosegregating with the Brugada electrocardiographic pattern within the pedigree. In silico gene prioritization pointed to 1 variant residing in KCNAB2, which encodes the voltage-gated K(+) channel ß2-subunit (Kvß2-R12Q). Kvß2 is widely expressed in the human heart and has been shown to interact with the fast transient outward K(+) channel subunit Kv4.3, increasing its current density. By targeted sequencing of the KCNAB2 gene in 167 unrelated patients with Brugada syndrome, we found 2 additional rare missense variants (L13F and V114I). We then investigated the physiological effects of the 3 KCNAB2 variants by using cellular electrophysiology and biochemistry. Patch-clamp experiments performed in COS-7 cells expressing both Kv4.3 and Kvß2 revealed a significant increase in the current density in presence of the R12Q and L13F Kvß2 mutants. Although biotinylation assays showed no differences in the expression of Kv4.3, the total and submembrane expression of Kvß2-R12Q were significantly increased in comparison with wild-type Kvß2. CONCLUSIONS: Altogether, our results indicate that Kvß2 dysfunction can contribute to the Brugada electrocardiographic pattern.


Asunto(s)
Síndrome de Brugada/genética , Mutación con Ganancia de Función/genética , Canales de Potasio con Entrada de Voltaje/genética , Electrocardiografía , Femenino , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Masculino , Canal de Sodio Activado por Voltaje NAV1.5/genética , Linaje , Polimorfismo de Nucleótido Simple/genética , Canales de Potasio de la Superfamilia Shaker , Canales de Potasio Shal/genética , Secuenciación del Exoma
7.
J Am Heart Assoc ; 5(5)2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27207958

RESUMEN

BACKGROUND: Transient receptor potential melastatin member 4 (TRPM4) is a nonselective cation channel. TRPM4 mutations have been linked to cardiac conduction disease and Brugada syndrome. The mechanisms underlying TRPM4-dependent conduction slowing are not fully understood. The aim of this study was to characterize TRPM4 genetic variants found in patients with congenital or childhood atrioventricular block. METHODS AND RESULTS: Ninety-one patients with congenital or childhood atrioventricular block were screened for candidate genes. Five rare TRPM4 genetic variants were identified and investigated. The variants were expressed heterologously in HEK293 cells. Two of the variants, A432T and A432T/G582S, showed decreased expression of the protein at the cell membrane; inversely, the G582S variant showed increased expression. Further functional characterization of these variants using whole-cell patch-clamp configuration showed a loss of function and a gain of function, respectively. We hypothesized that the observed decrease in expression was caused by a folding and trafficking defect. This was supported by the observation that incubation of these variants at lower temperature partially rescued their expression and function. Previous studies have suggested that altered SUMOylation of TRPM4 may cause a gain of function; however, we did not find any evidence that supports SUMOylation as being directly involved for the gain-of-function variant. CONCLUSIONS: This study underpins the role of TRPM4 in the cardiac conduction system. The loss-of-function variants A432T/G582S found in 2 unrelated patients with atrioventricular block are most likely caused by misfolding-dependent altered trafficking. The ability to rescue this variant with lower temperature may provide a novel use of pharmacological chaperones in treatment strategies.


Asunto(s)
Bloqueo Atrioventricular/genética , Pliegue de Proteína , Transporte de Proteínas/genética , Deficiencias en la Proteostasis/genética , Canales Catiónicos TRPM/genética , Bloqueo Atrioventricular/fisiopatología , Bloqueo Atrioventricular/terapia , Estimulación Cardíaca Artificial , Membrana Celular/metabolismo , Niño , Preescolar , Femenino , Variación Genética , Células HEK293 , Humanos , Lactante , Recién Nacido , Masculino , Marcapaso Artificial , Técnicas de Placa-Clamp , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales Catiónicos TRPM/metabolismo , Temperatura
8.
Int J Cardiol ; 207: 349-58, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26820365

RESUMEN

BACKGROUND: Progressive cardiac conduction disease (PCCD) is one of the most common cardiac conduction disturbances. It has been causally related to rare mutations in several genes including SCN5A, SCN1B, TRPM4, LMNA and GJA5. METHODS AND RESULTS: In this study, by applying targeted next-generation sequencing (NGS) in 95 unrelated patients with PCCD, we have identified 13 rare variants in the TRPM4 gene, two of which are currently absent from public databases. This gene encodes a cardiac calcium-activated cationic channel which precise role and importance in cardiac conduction and disease is still debated. One novel variant, TRPM4-p.I376T, is carried by the proband of a large French 4-generation pedigree. Systematic familial screening showed that a total of 13 family members carry the mutation, including 10 out of the 11 tested affected individuals versus only 1 out of the 21 unaffected ones. Functional and biochemical analyses were performed using HEK293 cells, in whole-cell patch-clamp configuration and Western blotting. TRPM4-p.I376T results in an increased current density concomitant to an augmented TRPM4 channel expression at the cell surface. CONCLUSIONS: This study is the first extensive NGS-based screening of TRPM4 coding variants in patients with PCCD. It reports the third largest pedigree diagnosed with isolated Progressive Familial Heart Block type I and confirms that this subtype of PCCD is caused by mutation-induced gain-of-expression and function of the TRPM4 ion channel.


Asunto(s)
Progresión de la Enfermedad , Marcación de Gen/métodos , Predisposición Genética a la Enfermedad/genética , Bloqueo Cardíaco/genética , Análisis de Secuencia de ADN/métodos , Canales Catiónicos TRPM/genética , Adulto , Anciano , Anciano de 80 o más Años , Trastorno del Sistema de Conducción Cardíaco , Niño , Femenino , Variación Genética/genética , Células HEK293 , Bloqueo Cardíaco/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Linaje
9.
Hum Mol Genet ; 24(10): 2757-63, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25650408

RESUMEN

The Brugada syndrome (BrS) is a rare heritable cardiac arrhythmia disorder associated with ventricular fibrillation and sudden cardiac death. Mutations in the SCN5A gene have been causally related to BrS in 20-30% of cases. Twenty other genes have been described as involved in BrS, but their overall contribution to disease prevalence is still unclear. This study aims to estimate the burden of rare coding variation in arrhythmia-susceptibility genes among a large group of patients with BrS. We have developed a custom kit to capture and sequence the coding regions of 45 previously reported arrhythmia-susceptibility genes and applied this kit to 167 index cases presenting with a Brugada pattern on the electrocardiogram as well as 167 individuals aged over 65-year old and showing no history of cardiac arrhythmia. By applying burden tests, a significant enrichment in rare coding variation (with a minor allele frequency below 0.1%) was observed only for SCN5A, with rare coding variants carried by 20.4% of cases with BrS versus 2.4% of control individuals (P = 1.4 × 10(-7)). No significant enrichment was observed for any other arrhythmia-susceptibility gene, including SCN10A and CACNA1C. These results indicate that, except for SCN5A, rare coding variation in previously reported arrhythmia-susceptibility genes do not contribute significantly to the occurrence of BrS in a population with European ancestry. Extreme caution should thus be taken when interpreting genetic variation in molecular diagnostic setting, since rare coding variants were observed in a similar extent among cases versus controls, for most previously reported BrS-susceptibility genes.


Asunto(s)
Síndrome de Brugada/genética , Predisposición Genética a la Enfermedad , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética , Adulto , Arritmias Cardíacas/genética , Síndrome de Brugada/diagnóstico , Femenino , Frecuencia de los Genes , Genes , Estudios de Asociación Genética , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia de ADN , Población Blanca
10.
J Am Coll Cardiol ; 60(2): 144-56, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22766342

RESUMEN

OBJECTIVES: The aim of this study was to describe a new familial cardiac phenotype and to elucidate the electrophysiological mechanism responsible for the disease. BACKGROUND: Mutations in several genes encoding ion channels, especially SCN5A, have emerged as the basis for a variety of inherited cardiac arrhythmias. METHODS: Three unrelated families comprising 21 individuals affected by multifocal ectopic Purkinje-related premature contractions (MEPPC) characterized by narrow junctional and rare sinus beats competing with numerous premature ventricular contractions with right and/or left bundle branch block patterns were identified. RESULTS: Dilated cardiomyopathy was identified in 6 patients, atrial arrhythmias were detected in 9 patients, and sudden death was reported in 5 individuals. Invasive electrophysiological studies demonstrated that premature ventricular complexes originated from the Purkinje tissue. Hydroquinidine treatment dramatically decreased the number of premature ventricular complexes. It normalized the contractile function in 2 patients. All the affected subjects carried the c.665G>A transition in the SCN5A gene. Patch-clamp studies of resulting p.Arg222Gln (R222Q) Nav1.5 revealed a net gain of function of the sodium channel, leading, in silico, to incomplete repolarization in Purkinje cells responsible for premature ventricular action potentials. In vitro and in silico studies recapitulated the normalization of the ventricular action potentials in the presence of quinidine. CONCLUSIONS: A new SCN5A-related cardiac syndrome, MEPPC, was identified. The SCN5A mutation leads to a gain of function of the sodium channel responsible for hyperexcitability of the fascicular-Purkinje system. The MEPPC syndrome is responsive to hydroquinidine.


Asunto(s)
Ramos Subendocárdicos/fisiopatología , Canales de Sodio/genética , Complejos Prematuros Ventriculares/genética , Adolescente , Adulto , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/genética , Cardiomiopatía Dilatada/genética , Niño , Análisis Mutacional de ADN , Muerte Súbita Cardíaca , Técnicas Electrofisiológicas Cardíacas , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Mutación , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/genética , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Linaje , Fenotipo , Quinidina/análogos & derivados , Quinidina/uso terapéutico , Canales de Sodio/fisiología , Síndrome , Complejos Prematuros Ventriculares/tratamiento farmacológico , Complejos Prematuros Ventriculares/fisiopatología , Adulto Joven
11.
Heart ; 98(17): 1305-10, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22717692

RESUMEN

INTRODUCTION: Progressive cardiac conduction defect (PCCD) is a frequent disease attributed to degeneration and fibrosis of the His bundle. Over the past years, gene defects have been identified demonstrating that PCCD could be a genetic disease. The aim of this study was to show a familial aggregation for PCCD using a genetic epidemiological approach to improve in fine genetic knowledge of the transmission of the disease. METHODS AND RESULTS: Using the French social security number, the authors have been able to determine the city of birth of the 6667 patients implanted with a pacemaker (PM) for PCCD between 1995 and 2005 in the western part of France. The authors then mapped the frequency of PM implantations for PCCD. A large heterogeneity of the frequency of the disease has been observed, with a frequency of 0.21% in the major city (Nantes) ranging up to 2.28% in specific parishes. Familial studies performed in the parishes with the highest frequency of the disease allowed the authors to identify five large families with PCCD. Clinical investigations demonstrated phenotype heterogeneity between families. Three patterns have been differentiated. CONCLUSIONS: This study demonstrates a disparate geographical repartition of the frequency of PM implantation in the area of the authors at least in part related to a hereditary factor. The identification of five large families affected by PCCD using epidemiological approach underlines the existence of a major genetic background in PCCD.


Asunto(s)
Bloqueo Cardíaco/epidemiología , Bloqueo Cardíaco/genética , Sistema de Conducción Cardíaco/fisiopatología , Adulto , Anciano , Francia/epidemiología , Genética de Población , Bloqueo Cardíaco/fisiopatología , Humanos , Persona de Mediana Edad , Epidemiología Molecular , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética , Marcapaso Artificial , Linaje
12.
Circ Arrhythm Electrophysiol ; 5(1): 163-72, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22247482

RESUMEN

BACKGROUND: Progressive familial heart block type I (PFHBI) is a hereditary arrhythmia characterized by progressive conduction disturbances in the His-Purkinje system. PFHBI has been linked to genes such as SCN5A that influence cardiac excitability but not to genes that influence cell-to-cell communication. Our goal was to explore whether nucleotide substitutions in genes coding for connexin proteins would associate with clinical cases of PFHBI and if so, to establish a genotype-cell phenotype correlation for that mutation. METHODS AND RESULTS: We screened 156 probands with PFHBI. In addition to 12 sodium channel mutations, we found a germ line GJA5 (connexin40 [Cx40]) mutation (Q58L) in 1 family. Heterologous expression of Cx40-Q58L in connexin-deficient neuroblastoma cells resulted in marked reduction of junctional conductance (Cx40-wild type [WT], 22.2±1.7 nS, n=14; Cx40-Q58L, 0.56±0.34 nS, n=14; P<0.001) and diffuse localization of immunoreactive proteins in the vicinity of the plasma membrane without formation of gap junctions. Heteromeric cotransfection of Cx40-WT and Cx40-Q58L resulted in homogenous distribution of proteins in the plasma membrane rather than in membrane plaques in ≈50% of cells; well-defined gap junctions were observed in other cells. Junctional conductance values correlated with the distribution of gap junction plaques. CONCLUSIONS: Mutation Cx40-Q58L impairs gap junction formation at cell-cell interfaces. This is the first demonstration of a germ line mutation in a connexin gene that associates with inherited ventricular arrhythmias and emphasizes the importance of Cx40 in normal propagation in the specialized conduction system.


Asunto(s)
Fascículo Atrioventricular/metabolismo , Conexinas/genética , ADN/genética , Bloqueo Cardíaco/genética , Mutación , Biomarcadores/metabolismo , Western Blotting , Fascículo Atrioventricular/fisiopatología , Trastorno del Sistema de Conducción Cardíaco , Niño , Conexinas/metabolismo , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Predisposición Genética a la Enfermedad , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/fisiopatología , Frecuencia Cardíaca , Humanos , Inmunohistoquímica , Masculino , Linaje , Reacción en Cadena de la Polimerasa , Pronóstico , Proteína alfa-5 de Unión Comunicante
13.
J Am Coll Cardiol ; 57(1): 40-7, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21185499

RESUMEN

OBJECTIVES: The aim of this study was to investigate, in a set of 93 mutation-negative long QT syndrome (LQTS) probands, the frequency of copy number variants (CNVs) in LQTS genes. BACKGROUND: LQTS is an inherited cardiac arrhythmia characterized by a prolonged heart rate-corrected QT (QTc) interval associated with sudden cardiac death. Recent studies suggested the involvement of duplications or deletions in the occurrence of LQTS. However, their frequency remains unknown in LQTS patients. METHODS: Point mutations in KCNQ1, KCNH2, and SCN5A genes were excluded by denaturing high-performance liquid chromatography or direct sequencing. We applied Multiplex Ligation-dependent Probe Amplification (MLPA) to detect CNVs in exons of these 3 genes. Abnormal exon copy numbers were confirmed by quantitative multiplex PCR of short fluorescent fragment (QMPSF). Array-based comparative genomic hybridization (array CGH) analysis was performed using Agilent Human Genome 244K Microarrays to further map the genomic rearrangements. RESULTS: We identified 3 different deletions in 3 unrelated families: 1 in KCNQ1 and 2 involving KCNH2. We showed in the largest family that the deletion involving KCNH2 is fully penetrant and segregates with the long QT phenotype in 7 affected members. CONCLUSIONS: Our study demonstrates that CNVs in KCNQ1 and KCNH2 explain around 3% of LQTS in patients with no point mutation in these genes. This percentage is likely higher than the frequency of point mutations in ANKB, KCNE1, KCNE2, KCNJ2, CACNA1C, CAV3, SCN4B, AKAP9, and SNTA1 together. Thus, we propose that CNV screening in KCNQ1 and KCNH2 may be performed routinely in LQTS patients.


Asunto(s)
ADN/genética , Canales de Potasio Éter-A-Go-Go/genética , Variación Genética/genética , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Proteínas Musculares/genética , Mutación Puntual , Canales de Sodio/genética , Adolescente , Adulto , Anciano , Niño , Canal de Potasio ERG1 , Electrocardiografía , Femenino , Ligamiento Genético , Pruebas Genéticas , Humanos , Síndrome de QT Prolongado/fisiopatología , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5 , Linaje , Reacción en Cadena de la Polimerasa , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...