Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 102(1): 133-141, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29304371

RESUMO

Intracranial aneurysms (IAs) are acquired cerebrovascular abnormalities characterized by localized dilation and wall thinning in intracranial arteries, possibly leading to subarachnoid hemorrhage and severe outcome in case of rupture. Here, we identified one rare nonsense variant (c.1378A>T) in the last exon of ANGPTL6 (Angiopoietin-Like 6)-which encodes a circulating pro-angiogenic factor mainly secreted from the liver-shared by the four tested affected members of a large pedigree with multiple IA-affected case subjects. We showed a 50% reduction of ANGPTL6 serum concentration in individuals heterozygous for the c.1378A>T allele (p.Lys460Ter) compared to relatives homozygous for the normal allele, probably due to the non-secretion of the truncated protein produced by the c.1378A>T transcripts. Sequencing ANGPTL6 in a series of 94 additional index case subjects with familial IA identified three other rare coding variants in five case subjects. Overall, we detected a significant enrichment (p = 0.023) in rare coding variants within this gene among the 95 index case subjects with familial IA, compared to a reference population of 404 individuals with French ancestry. Among the 6 recruited families, 12 out of 13 (92%) individuals carrying IA also carry such variants in ANGPTL6, versus 15 out of 41 (37%) unaffected ones. We observed a higher rate of individuals with a history of high blood pressure among affected versus healthy individuals carrying ANGPTL6 variants, suggesting that ANGPTL6 could trigger cerebrovascular lesions when combined with other risk factors such as hypertension. Altogether, our results indicate that rare coding variants in ANGPTL6 are causally related to familial forms of IA.


Assuntos
Proteínas Semelhantes a Angiopoietina/genética , Predisposição Genética para Doença , Aneurisma Intracraniano/genética , Mutação/genética , Fases de Leitura Aberta/genética , Proteína 6 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina/sangue , Células Cultivadas , Códon sem Sentido/genética , Família , Feminino , Células HEK293 , Humanos , Aneurisma Intracraniano/sangue , Masculino , Pessoa de Meia-Idade , Linhagem , Fatores de Risco
2.
Pharmacol Res ; 159: 104922, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32464326

RESUMO

Down-regulation of Connexin43 (Cx43) has often been associated with the development of cardiac fibrosis. We showed previously that Scn5a heterozygous knockout mice (Scn5a+/-), which mimic familial progressive cardiac conduction defect, exhibit an age-dependent decrease of Cx43 expression and phosphorylation concomitantly with activation of TGF-ß pathway and fibrosis development in the myocardium between 45 and 60 weeks of age. The aim of this study was to investigate whether Gap-134 prevents Cx43 down-regulation with age and fibrosis development in Scn5a+/- mice. We observed in 60-week-old Scn5a+/- mouse heart a Cx43 expression and localization remodeling correlated with fibrosis. Chronic administration of a potent and selective gap junction modifier, Gap-134 (danegaptide), between 45 and 60 weeks, increased Cx43 expression and phosphorylation on serine 368 and prevented Cx43 delocalization. Furthermore, we found that Gap-134 prevented fibrosis despite the persistence of the conduction defects and the TGF-ß canonical pathway activation. In conclusion, the present study demonstrates that the age-dependent decrease of Cx43 expression is involved in the ventricular fibrotic process occurring in Scn5a+/- mice. Finally, our study suggests that gap junction modifier, such as Gap-134, could be an effective anti-fibrotic agent in the context of age-dependent fibrosis in progressive cardiac conduction disease.


Assuntos
Benzamidas/farmacologia , Cardiomiopatias/prevenção & controle , Conexina 43/metabolismo , Fibroblastos/efeitos dos fármacos , Miocárdio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/deficiência , Prolina/análogos & derivados , Animais , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Camundongos da Linhagem 129 , Camundongos Knockout , Miocárdio/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Fosforilação , Prolina/farmacologia , Pirazóis/farmacologia , Transdução de Sinais , Regulação para Cima , Remodelação Ventricular/efeitos dos fármacos
3.
Eur Heart J ; 39(15): 1269-1277, 2018 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-29020406

RESUMO

Aims: Filamin-A (FLNA) was identified as the first gene of non-syndromic mitral valve dystrophy (FLNA-MVD). We aimed to assess the phenotype of FLNA-MVD and its impact on prognosis. Methods and results: We investigated the disease in 246 subjects (72 mutated) from four FLNA-MVD families harbouring three different FLNA mutations. Phenotype was characterized by a comprehensive echocardiography focusing on mitral valve apparatus in comparison with control relatives. In this X-linked disease valves lesions were severe in men and moderate in women. Most men had classical features of mitral valve prolapse (MVP), but without chordal rupture. By contrast to regular MVP, mitral leaflet motion was clearly restricted in diastole and papillary muscles position was closer to mitral annulus. Valvular abnormalities were similar in the four families, in adults and young patients from early childhood suggestive of a developmental disease. In addition, mitral valve lesions worsened over time as encountered in degenerative conditions. Polyvalvular involvement was frequent in males and non-diagnostic forms frequent in females. Overall survival was moderately impaired in men (P = 0.011). Cardiac surgery rate (mainly valvular) was increased (33.3 ± 9.8 vs. 5.0 ± 4.9%, P < 0.0001; hazard ratio 10.5 [95% confidence interval: 2.9-37.9]) owing mainly to a lifetime increased risk in men (76.8 ± 14.1 vs. 9.1 ± 8.7%, P < 0.0001). Conclusion: FLNA-MVD is a developmental and degenerative disease with complex phenotypic expression which can influence patient management. FLNA-MVD has unique features with both MVP and paradoxical restricted motion in diastole, sub-valvular mitral apparatus impairment and polyvalvular lesions in males. FLNA-MVD conveys a substantial lifetime risk of valve surgery in men.


Assuntos
Filaminas/genética , Prolapso da Valva Mitral/genética , Prolapso da Valva Mitral/patologia , Valva Mitral/patologia , Adolescente , Adulto , Ecocardiografia , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Valva Mitral/diagnóstico por imagem , Mutação/genética , Fenótipo , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Adulto Jovem
4.
Eur Heart J ; 39(31): 2879-2887, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30059973

RESUMO

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.


Assuntos
Doença do Sistema de Condução Cardíaco/genética , Estudos de Associação Genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Fatores Etários , Doenças Assintomáticas , Síndrome de Brugada/genética , Criança , Pré-Escolar , Eletrocardiografia , Feminino , Seguimentos , Mutação com Ganho de Função , Humanos , Lactente , Recém-Nascido , Síndrome do QT Longo/genética , Mutação com Perda de Função , Masculino , Estudos Retrospectivos , Fatores de Risco
5.
Europace ; 20(12): 2014-2020, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29688407

RESUMO

Aims: QT prolongation during mental stress test (MST) has been associated with familial idiopathic ventricular fibrillation. In long QT syndrome (LQTS), up to 30% of mutation carriers have normal QT duration. Our aim was to assess the QT response during MST, and its accuracy in the diagnosis of concealed LQTS. Methods and results: All patients who are carrier of a KCNQ1 or KCNH2 mutations without QT prolongation were enrolled. A control group was constituted of patients with negative exercise and epinephrine tests. Electrocardiogram were recorded at rest and at the maximum heart rate during MST and reviewed by two physicians. Among the 70 patients enrolled (median age 41±2.1 years, 46% male), 36 were mutation carrier for LQTS (20 KCNQ1 and 16 KCNH2), and 34 were controls. KCNQ1 and KCNH2 mutation carriers presented a longer QT interval at baseline [405(389; 416) and 421 (394; 434) ms, respectively] compared with the controls [361(338; 375)ms; P < 0.0001]. QT duration during MST varied by 9 (4; 18) ms in KCNQ1, 3 (-6; 16) ms in KCNH2, and by -22 (-29; -17) ms in controls (P < 0.0001). These QT variations were independent of heart rate (P < 0.3751). Receiver operating characteristic curve analysis identified a cut-off value of QT variation superior to -11 ms as best predictor of LQTS. It provided 97% sensitivity and 97% specificity of QT prolongation in the diagnosis of LQTS. Conclusion: We identified a paradoxical response of the QT interval during MST in LQTS. Easy to assess, MST may be efficient to unmask concealed LQTS in patients at risk of this pathology.


Assuntos
Eletrocardiografia , Frequência Cardíaca/genética , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ2/genética , Síndrome do QT Longo/diagnóstico , Mutação , Estresse Psicológico/fisiopatologia , Fibrilação Ventricular/diagnóstico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Predisposição Genética para Doença , Humanos , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Masculino , Conceitos Matemáticos , Pessoa de Meia-Idade , Fenótipo , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Fatores de Risco , Estresse Psicológico/diagnóstico , Estresse Psicológico/psicologia , Fibrilação Ventricular/genética , Fibrilação Ventricular/fisiopatologia , Adulto Jovem
6.
Hum Mol Genet ; 24(10): 2757-63, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25650408

RESUMO

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.


Assuntos
Síndrome de Brugada/genética , Predisposição Genética para Doença , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Adulto , Arritmias Cardíacas/genética , Síndrome de Brugada/diagnóstico , Feminino , Frequência do Gene , Genes , Estudos de Associação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência de DNA , População Branca
7.
Hum Mol Genet ; 21(12): 2759-67, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22422768

RESUMO

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an inherited arrhythmogenic disease so far related to mutations in the cardiac ryanodine receptor (RYR2) or the cardiac calsequestrin (CASQ2) genes. Because mutations in RYR2 or in CASQ2 are not retrieved in all CPVT cases, we searched for mutations in the physiological protein partners of RyR2 and CSQ2 in a large cohort of CPVT patients with no detected mutation in these two genes. Based on a candidate gene approach, we focused our investigations on triadin and junctin, two proteins that link RyR2 and CSQ2. Mutations in the triadin (TRDN) and in the junctin (ASPH) genes were searched in a cohort of 97 CPVT patients. We identified three mutations in triadin which cosegregated with the disease on a recessive mode of transmission in two families, but no mutation was found in junctin. Two TRDN mutations, a 4 bp deletion and a nonsense mutation, resulted in premature stop codons; the third mutation, a p.T59R missense mutation, was further studied. Expression of the p.T59R mutant in COS-7 cells resulted in intracellular retention and degradation of the mutant protein. This was confirmed after in vivo expression of the mutant triadin in triadin knock-out mice by viral transduction. In this work, we identified TRDN as a new gene responsible for an autosomal recessive form of CPVT. The mutations identified in the two families lead to the absence of the protein, thereby demonstrating the importance of triadin for the normal function of the cardiac calcium release complex in humans.


Assuntos
Arritmias Cardíacas/genética , Proteínas de Transporte/genética , Morte Súbita Cardíaca , Proteínas Musculares/genética , Taquicardia Ventricular/genética , Animais , Arritmias Cardíacas/metabolismo , Western Blotting , Células COS , Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Chlorocebus aethiops , Retículo Endoplasmático/metabolismo , Saúde da Família , Feminino , Genes Recessivos , Predisposição Genética para Doença/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas Musculares/metabolismo , Mutação , Miócitos Cardíacos/metabolismo , Linhagem , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patologia
8.
Basic Res Cardiol ; 109(6): 446, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25341504

RESUMO

Brugada syndrome (BrS) is characterized by ST-segment elevation in the right precordial leads and is associated with increased risk of sudden cardiac death. We have recently reported families with BrS and SCN5A mutations where some affected members do not carry the familial mutation. We evaluated the involvement of additional genetic determinants for BrS in an affected family. We identified three distinct gene variants within a family presenting BrS (5 individuals), cardiac conduction defects (CCD, 3 individuals) and shortened QT interval (4 individuals). The first mutation is nonsense, p.Q1695*, lying within the SCN5A gene, which encodes for NaV1.5, the α-subunit of the cardiac Na(+) channel. The second mutation is missense, p.N300D, and alters the CACNA1C gene, which encodes the α-subunit CaV1.2 of the L-type cardiac Ca(2+) channel. The SCN5A mutation strictly segregates with CCD. Four out of the 5 BrS patients carry the CACNA1C variant, and three of them present shortened QT interval. One of the BrS patients carries none of these mutations but a rare variant located in the ABCC9 gene as well as his asymptomatic mother. Patch-clamp studies identified a loss-of-function of the mutated CaV1.2 channel. Western-blot experiments showed a global expression defect while increased mobility of CaV1.2 channels on cell surface was revealed by FRAP experiments. Finally, computer simulations of the two mutations recapitulated patient phenotypes. We report a rare CACNA1C mutation as causing BrS and/or shortened QT interval in a family also carrying a SCN5A stop mutation, but which does not segregate with BrS. This study underlies the complexity of BrS inheritance and its pre-symptomatic genetic screening interpretation.


Assuntos
Síndrome de Brugada/genética , Canais de Cálcio Tipo L/genética , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Adulto , Idoso de 80 Anos ou mais , Animais , Células COS , Chlorocebus aethiops , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Adulto Jovem
9.
Europace ; 16(6): 908-13, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24068444

RESUMO

AIMS: The objective of this study was to correlate the electrocardiogram (ECG) modification during an Ajmaline challenge in patients affected by the Brugada syndrome and implanted with an implantable cardioverter-defibrillator (ICD) with the morphological changes of their ICD's intracardiac electrogram (IEGM). METHODS AND RESULTS: Sixteen type 1 Brugada syndrome patients implanted with a St Jude Medical AnalyST(®) ICD were enrolled and underwent ajmaline challenge. Intracardiac electrograms and 12 lead ECG signals were collected over the duration of the study and analysed off-line. The right precordial ECG leads were in both the third and fourth intercostal space by putting V5 and V6 in V1 and V2 at the third intercostal space. Two patients were excluded from the analysis due to signal noise issues. Of the remaining 14 patients, 12 and 2 patients were adjudicated to have positive and negative ajmaline challenges, respectively, based on standard ECG criteria. In the ajmaline positive patients, the IEGM T wave amplitude changes were more prominent than those of the IEGM ST segment (-898 ± 463 vs. -55 ± 381 µV, P < 0.05). Furthermore, all of these T wave amplitude changes were in the negative polarity, whereas the change in polarity of the ST segment was mixed. The changes in the IEGM T wave amplitude and ST segment were significantly smaller in the ajmaline negative patients compared with those in the ajmaline positive patients [211 ± 158 (P < 0.05) and 107 ± 54 (P < 0.05) µV, respectively). Over all 14 analysable patients, the change in the ECG ST segment over the timecourse of the ajmaline challenge correlated better with the IEGM T wave amplitude change (R = 0.72 ± 0.33) than the IEGM ST segment change (R = 0.63 ± 0.33). Applying an IEGM T wave amplitude change cut-off of 400 µV for predicting the outcome of the ajmaline challenge yielded 92% sensitivity (11/12) and 100% specificity (2/2). CONCLUSION: In Brugada patients, ajmaline challenge elicits significant T wave amplitude changes within the ICD IEGM, greater than those of the IEGM ST segment. This study is the first step to provide new tools able to continuously monitor the type I Brugada aspect in patients affected by the Brugada syndrome.


Assuntos
Ajmalina , Mapeamento Potencial de Superfície Corporal/métodos , Síndrome de Brugada/diagnóstico , Eletrocardiografia/métodos , Antiarrítmicos , Mapeamento Potencial de Superfície Corporal/efeitos dos fármacos , Eletrocardiografia/efeitos dos fármacos , Feminino , França , Humanos , Masculino , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
10.
Heart ; 110(9): 666-674, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38148157

RESUMO

OBJECTIVE: Variants in the FLNA gene have been associated with mitral valve dystrophy (MVD), and even polyvalvular disease has been reported. This study aimed to analyse the aortic valve and root involvement in FLNA-MVD families and its impact on outcomes. METHODS: 262 subjects (37 (18-53) years, 140 male, 79 carriers: FLNA+) from 4 FLNA-MVD families were included. Echocardiography was performed in 185 patients and histological analysis in 3 explanted aortic valves. The outcomes were defined as aortic valve surgery or all-cause mortality. RESULTS: Aortic valve alterations were found in 58% of FLNA+ compared with 6% of FLNA- (p<0.001). 9 (13.4%) FLNA+ had bicuspid aortic valve compared with 4 (3.4%) FLNA- (p=0.03). Overall, the transvalvular mean gradient was slightly increased in FLNA+ (4.8 (4.1-6.1) vs 4.0 (2.9-4.9) mm Hg, p=0.02). The sinuses of Valsalva and sinotubular junction diameters were enlarged in FLNA+ subjects (all p<0.05). 8 FLNA+ patients underwent aortic valve surgery (0 in relatives; p<0.001). Myxomatous remodelling with an infiltration of immune cells was observed. Overall survival was similar between FLNA+ versus FLNA- subjects (86±5% vs 85±6%, p=0.36). There was no statistical evidence for an interaction between genetic status and sex (p=0.15), but the survival tended to be impaired in FLNA+ men (p=0.06) whereas not in women (p=0.71). CONCLUSION: The patients with FLNA variants present frequent aortic valve disease and worse outcomes. Bicuspid aortic valve is more frequent in patients carrying the FLNA-MVD variants. These unique features should be factored into the management of patients with dystrophic and/or bicuspid aortic valve.


Assuntos
Doença da Válvula Aórtica Bicúspide , Doenças das Valvas Cardíacas , Cardiopatia Reumática , Feminino , Humanos , Masculino , Valva Aórtica/diagnóstico por imagem , Valva Aórtica/cirurgia , Valva Aórtica/patologia , Filaminas/genética , Doenças das Valvas Cardíacas/diagnóstico por imagem , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/cirurgia
11.
Nat Commun ; 15(1): 3380, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38643172

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Animais , Camundongos , Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Cromatina/genética , Proteínas de Ligação a DNA/metabolismo , Genoma
12.
Circulation ; 126(12): 1469-77, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22899775

RESUMO

BACKGROUND: The origin of congenital or childhood nonimmune isolated atrioventricular (AV) block remains unknown. We hypothesized that this conduction abnormality in the young may be a heritable disease. METHODS AND RESULTS: A multicenter retrospective study (13 French referral centers, from 1980-2009) included 141 children with AV block diagnosed in utero, at birth, or before 15 years of age without structural heart abnormalities and without maternal antibodies. Parents and matched control subjects were investigated for family history and for ECG screening. In parents, a family history of sudden death or progressive cardiac conduction defect was found in 1.4% and 11.1%, respectively. Screening ECGs from 130 parents (mean age 42.0 ± 6.8 years, 57 couples) were compared with those of 130 matched healthy control subjects. All parents were asymptomatic and in sinus rhythm, except for 1 with undetected complete AV block. Conduction abnormalities were more frequent in parents than in control subjects, found in 50.8% versus 4.6%, respectively (P<0.001). A long PR interval was found in 18.5% of the parents but never in control subjects (P<0.0001). Complete or incomplete right bundle-branch block was observed in 39.2% of the parents and 1.5% of the control subjects (P<0.0001). Complete or incomplete left bundle-branch block was found in 15.4% of the parents and 3.1% of the control subjects (P<0.0006). Estimated heritability for isolated conduction disturbances was 91% (95% confidence interval, 80%-100%). SCN5A mutation screening identified 2 mutations in 2 patients among 97 children. CONCLUSIONS: ECG screening in parents of children affected by idiopathic AV block revealed a high prevalence of conduction abnormalities. These results support the hypothesis of an inheritable trait in congenital and childhood nonimmune isolated AV block.


Assuntos
Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/genética , Eletrocardiografia/métodos , Programas de Rastreamento/métodos , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Pais , Adolescente , Adulto , Idoso , Bloqueio Atrioventricular/congênito , Bloqueio Atrioventricular/epidemiologia , Criança , Pré-Escolar , Eletrocardiografia/estatística & dados numéricos , Feminino , Predisposição Genética para Doença/epidemiologia , Predisposição Genética para Doença/genética , Testes Genéticos/métodos , Testes Genéticos/estatística & dados numéricos , Humanos , Lactente , Recém-Nascido , Masculino , Programas de Rastreamento/estatística & dados numéricos , Pessoa de Meia-Idade , Fenótipo , Gravidez , Diagnóstico Pré-Natal , Prevalência , Estudos Retrospectivos , Adulto Jovem
13.
Europace ; 15(12): 1805-11, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23867365

RESUMO

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.


Assuntos
Síndrome de Andersen/genética , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Adolescente , Adulto , Idoso , Síndrome de Andersen/complicações , Síndrome de Andersen/diagnóstico , Síndrome de Andersen/fisiopatologia , Síndrome de Andersen/terapia , Criança , Pré-Escolar , Análise Mutacional de DNA , Eletrocardiografia , Feminino , França , Predisposição Genética para Doença , Parada Cardíaca/genética , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Prognóstico , Estudos Retrospectivos , Síncope/genética , Fatores de Tempo , Adulto Jovem
14.
Eur Heart J ; 33(5): 622-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21920962

RESUMO

AIMS: The natural history of congenital or childhood non-immune, isolated atrioventricular (AV) block is poorly defined. METHODS AND RESULTS: We retrospectively studied 141 children with isolated, non-immune AV block diagnosed in utero, or up to 15 years of age, at 13 French medical centres, between 1980 and 2009. Patients with structural heart disease or maternal antibodies were excluded. Atrioventricular block was asymptomatic in 119 (84.4%) and complete in 100 (70.9%) patients. There was progression to complete AV block in 29/41 (70.7%) patients with incomplete AV block over 2.8 ± 3.4 years (1-155 months), but all patients with incomplete AV block may not have been included in the study. Narrow QRS complex was present in 18 of 26 patients (69.2%) with congenital, and 106 of 115 (92.2%) with childhood AV block. Pacemakers were implanted in 112 children (79.4%), during the first year of life in 18 (16.1%) and before 10 years of age in 90 (80.4%). The mean interval between diagnosis of AV block and pacemaker implants was 2.6 ± 3.9 years (0-300 months). The pacing indication was prophylactic in 70 children (62.5%). During a mean follow-up of 11.6 ± 6.7 years (1-32 years), no patient died or developed dilated cardiomyopathy (DCM). The long-term follow-up was uncomplicated in 127 children (90.1%). CONCLUSION: In this large multicentre study, the long-term outcome of congenital or childhood non-immune, isolated AV block was favourable, regardless of the patient's age at the time of diagnosis. No patient died or developed DCM, and pacemaker-related complications were few.


Assuntos
Bloqueio Atrioventricular/terapia , Estimulação Cardíaca Artificial/métodos , Adolescente , Adulto , Idade de Início , Bloqueio Atrioventricular/congênito , Bloqueio Atrioventricular/diagnóstico , Bloqueio de Ramo/diagnóstico , Bloqueio de Ramo/etiologia , Criança , Pré-Escolar , Progressão da Doença , Intervalo Livre de Doença , Eletrocardiografia , Feminino , Humanos , Lactente , Masculino , Marca-Passo Artificial , Gravidez , Diagnóstico Pré-Natal , Estudos Retrospectivos , Fatores de Risco , Resultado do Tratamento , Adulto Jovem
15.
Cardiovasc Res ; 119(3): 759-771, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-36001550

RESUMO

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.


Assuntos
Prolapso da Valva Mitral , Valva Mitral , Adulto , Humanos , Ratos , Animais , Lactente , Valva Mitral/metabolismo , Filaminas/genética , Filaminas/metabolismo , Transcriptoma , Microtomografia por Raio-X , Prolapso da Valva Mitral/patologia , Fenótipo
16.
Circulation ; 124(11): 1212-22, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21859974

RESUMO

BACKGROUND: Atrial fibrillation (AF) is the most common cardiac arrhythmia, affecting >2 million patients in the United States alone. Despite decades of research, surprisingly little is known regarding the molecular pathways underlying the pathogenesis of AF. ANK2 encodes ankyrin-B, a multifunctional adapter molecule implicated in membrane targeting of ion channels, transporters, and signaling molecules in excitable cells. METHODS AND RESULTS: In the present study, we report early-onset AF in patients harboring loss-of-function mutations in ANK2. In mice, we show that ankyrin-B deficiency results in atrial electrophysiological dysfunction and increased susceptibility to AF. Moreover, ankyrin-B(+/-) atrial myocytes display shortened action potentials, consistent with human AF. Ankyrin-B is expressed in atrial myocytes, and we demonstrate its requirement for the membrane targeting and function of a subgroup of voltage-gated Ca(2+) channels (Ca(v)1.3) responsible for low voltage-activated L-type Ca(2+) current. Ankyrin-B is associated directly with Ca(v)1.3, and this interaction is regulated by a short, highly conserved motif specific to Ca(v)1.3. Moreover, loss of ankyrin-B in atrial myocytes results in decreased Ca(v)1.3 expression, membrane localization, and function sufficient to produce shortened atrial action potentials and arrhythmias. Finally, we demonstrate reduced ankyrin-B expression in atrial samples of patients with documented AF, further supporting an association between ankyrin-B and AF. CONCLUSIONS: These findings support that reduced ankyrin-B expression or mutations in ANK2 are associated with AF. Additionally, our data demonstrate a novel pathway for ankyrin-B-dependent regulation of Ca(v)1.3 channel membrane targeting and regulation in atrial myocytes.


Assuntos
Anquirinas/deficiência , Fibrilação Atrial/genética , Canais de Cálcio/deficiência , Transdução de Sinais/fisiologia , Adulto , Idoso , Sequência de Aminoácidos , Animais , Anquirinas/biossíntese , Anquirinas/genética , Fibrilação Atrial/metabolismo , Canais de Cálcio/genética , Criança , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Dados de Sequência Molecular , Miócitos Cardíacos/metabolismo , Transporte Proteico/fisiologia , Ratos , Adulto Jovem
17.
Stem Cell Res ; 60: 102688, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35101670

RESUMO

Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT) is an exercise and emotional stress-induced life-threatening inherited heart rhythm disorder, characterized by an abnormal cellular calcium homeostasis. Most reported cases have been linked to mutations in the gene encoding the type 2 ryanodine receptor gene, RYR2. We generated induced pluripotent stem cells (hiPSCs) from peripheral blood mononuclear cells (PBMC) from three CPVT-affected patients, two of them carrying p.R4959Q mutation and one carrying p.Y2476D mutation. These generated hiPSC lines are a useful model to study pathophysiological consequences of RYR2 dysfunction in humans and the molecular basis of CPVT.


Assuntos
Células-Tronco Pluripotentes Induzidas , Cálcio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Mutação/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Taquicardia Ventricular
18.
Europace ; 13(7): 1034-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21427091

RESUMO

AIMS: Brugada syndrome (BrS) is a hereditary arrhythmic disease, responsible for sudden death in patients without known heart disease. An implantable cardioverter defibrillator (ICD) is recommended in patients at high risk of sudden death, but the resulting psychological impact has never been studied. The aim of our study was to assess the impact on quality of life of BrS and ICD implantation. METHODS AND RESULTS: Patients were selected from the reference centre for hereditary arrhythmic disease database in Nantes. This population was divided into three groups: Group 1 (G1), symptomatic implanted patients; Group 2 (G2), asymptomatic implanted patients; and Group 3 (G3), asymptomatic patients without ICD. One hundred and ninety questionnaires [36-item short-form health survey (SF-36) and subsidiary questions] were analysed (60 in G1, 78 in G2, and 52 in G3). We failed to identify any difference in the evaluation of the SF-36 between the three groups and the SF-36 score was similar to the French population score. However, specific questions regarding tolerance of the ICD showed that ICD implantation resulted in significant negative impact, especially for professional careers and purchasing insurance, even though the patient considered ICD implantation as reassuring. CONCLUSION: Whatever the group, BrS patients have a good quality of life with no difference between implanted and non-implanted patients. However, ICD implantation is accompanied by difficulties in their social and professional life. This work emphasizes the need to propose specific recommendations applicable to insurance to reduce the complications experienced by these patients.


Assuntos
Síndrome de Brugada/psicologia , Síndrome de Brugada/terapia , Desfibriladores Implantáveis/psicologia , Adulto , Idoso , Síndrome de Brugada/prevenção & controle , Feminino , França , Inquéritos Epidemiológicos , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida/psicologia , Fatores de Risco , Inquéritos e Questionários
19.
Proc Natl Acad Sci U S A ; 105(40): 15617-22, 2008 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-18832177

RESUMO

The identification of nearly a dozen ion channel genes involved in the genesis of human atrial and ventricular arrhythmias has been critical for the diagnosis and treatment of fatal cardiovascular diseases. In contrast, very little is known about the genetic and molecular mechanisms underlying human sinus node dysfunction (SND). Here, we report a genetic and molecular mechanism for human SND. We mapped two families with highly penetrant and severe SND to the human ANK2 (ankyrin-B/AnkB) locus. Mice heterozygous for AnkB phenocopy human SND displayed severe bradycardia and rate variability. AnkB is essential for normal membrane organization of sinoatrial node cell channels and transporters, and AnkB is required for physiological cardiac pacing. Finally, dysfunction in AnkB-based trafficking pathways causes abnormal sinoatrial node (SAN) electrical activity and SND. Together, our findings associate abnormal channel targeting with human SND and highlight the critical role of local membrane organization for sinoatrial node excitability.


Assuntos
Anquirinas/genética , Anquirinas/fisiologia , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Proteínas de Membrana Transportadoras/metabolismo , Nó Sinoatrial/fisiopatologia , Adulto , Animais , Arritmias Cardíacas/metabolismo , Cálcio/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Canais Iônicos/metabolismo , Camundongos , Mutação , Nó Sinoatrial/metabolismo
20.
Europace ; 12(3): 424-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20139118

RESUMO

Implantable cardioverter-defibrillator (ICD) therapy is effective in primary and secondary prevention for patients who are at high risk of sudden cardiac death. However, the current risk stratification of patients who may benefit from this therapy is unsatisfactory. Single nucleotide polymorphisms (SNPs) are DNA sequence variations occurring when a single nucleotide in the genome differs among members of a species. A novel concept has emerged being that these common genetic variations might modify the susceptibility of a certain population to specific diseases. Thus, genetic factors may also modulate the risk for arrhythmias and sudden cardiac death, and identification of common variants could help to better identify patients at risk. The DISCOVERY study is an interventional, longitudinal, prospective, multi-centre diagnostic study that will enrol 1287 patients in approximately 80 European centres. In the genetic part of the DISCOVERY study, candidate gene polymorphisms involved in coding of the G-protein subunits will be correlated with the occurrence of ventricular arrhythmias in patients receiving an ICD for primary prevention. Furthermore, in order to search for additional sequence variants contributing to ventricular arrhythmias, a genome-wide association study will be conducted if sufficient a priori evidence can be gathered. In the second part of the study, associations of SNPs with ventricular arrhythmias will be sought and a search for potential new biological arrhythmic pathways will be investigated. As it is a diagnostic study, DISCOVERY will also investigate the impact of long-term device diagnostic data on the management of patients suffering from chronic cardiac disease as well as medical decisions made regarding their treatment.


Assuntos
Desfibriladores Implantáveis , Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/genética , Cromograninas , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Marcadores Genéticos , Predisposição Genética para Doença/epidemiologia , Testes Genéticos , Proteínas Heterotriméricas de Ligação ao GTP/genética , Humanos , Estudos Longitudinais , Estudos Multicêntricos como Assunto , Fatores de Risco , Taquicardia Ventricular/epidemiologia , Taquicardia Ventricular/terapia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa