Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Am J Med Genet A ; 185(8): 2532-2540, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34089229

RESUMO

The RNA exosome is a multi-subunit complex involved in the processing, degradation, and regulated turnover of RNA. Several subunits are linked to Mendelian disorders, including pontocerebellar hypoplasia (EXOSC3, MIM #614678; EXOSC8, MIM #616081: and EXOSC9, MIM #618065) and short stature, hearing loss, retinitis pigmentosa, and distinctive facies (EXOSC2, MIM #617763). More recently, EXOSC5 (MIM *606492) was found to underlie an autosomal recessive neurodevelopmental disorder characterized by developmental delay, hypotonia, cerebellar abnormalities, and dysmorphic facies. An unusual feature of EXOSC5-related disease is the occurrence of complete heart block requiring a pacemaker in a subset of affected individuals. Here, we provide a detailed clinical and molecular characterization of two siblings with microcephaly, developmental delay, cerebellar volume loss, hypomyelination, with cardiac conduction and rhythm abnormalities including sinus node dysfunction, intraventricular conduction delay, atrioventricular block, and ventricular tachycardia (VT) due to compound heterozygous variants in EXOSC5: (1) NM_020158.4:c.341C > T (p.Thr114Ile; pathogenic, previously reported) and (2) NM_020158.4:c.302C > A (p.Thr101Lys; novel variant). A review of the literature revealed an additional family with biallelic EXOSC5 variants and cardiac conduction abnormalities. These clinical and molecular data provide compelling evidence that cardiac conduction abnormalities and arrhythmias are part of the EXOSC5-related disease spectrum and argue for proactive screening due to potential risk of sudden cardiac death.


Assuntos
Antígenos de Neoplasias/genética , Morte Súbita Cardíaca/etiologia , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Mutação , Fenótipo , Proteínas de Ligação a RNA/genética , Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/genética , Criança , Ecocardiografia , Eletrocardiografia , Fácies , Feminino , Estudos de Associação Genética/métodos , Humanos , Masculino , Linhagem , Análise de Sequência de DNA , Adulto Jovem
2.
BMC Pediatr ; 20(1): 211, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398054

RESUMO

BACKGROUND: Vasovagal syncope (VVS) is common in children and significantly affects their quality of life. To our knowledge, this the first case report of SCN5A gene mutation associated with VVS and third-degree atrioventricular block (atrioventricular block, AVB), which could help pediatricians aware that VVS is not always a benign condition and help to identify VVS children at the risk of sudden death. CASE PRESENTATION: A twelve-year-old male child was admitted to Beijing Children's Hospital of Capital Medical University for chest tightness for 9 days and syncope in July 2018. The child was diagnosed as VVS with third-degree AVB after complete investagations. A heterozygous mutation in the exon coding region of the SCN5A gene, C. 5851G > T (coding region 5551 nucleotide changed from G to T), was detected in the peripheral blood of the child. Electrophysiological examination and modified vagal ganglion radiofrequency ablation were performed in the child. The ECG playback was normal on the second day after operation. Holter showed no abnormality and no chest tightness or syncope occurred after 3 months and 1 year follow-up. CONCLUSIONS: Our case report firstly reported that SCN5A mutation contributed to the pathogenesis of VVS with third-degree AVB. Vagal ganglion modified ablation have obtained good therapeutic effect. Gene analysis was of great value to the accurate diagnosis and treatment of VVS children.


Assuntos
Bloqueio Atrioventricular , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Síncope Vasovagal , Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/genética , Criança , Humanos , Masculino , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/química , Qualidade de Vida , Síncope Vasovagal/diagnóstico , Síncope Vasovagal/genética
3.
Ann Intern Med ; 171(7): 458-463, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31476771

RESUMO

Background: Mutations in the LMNA (lamin A/C) gene have been associated with neuromuscular and cardiac manifestations, but the clinical implications of these signs are not well understood. Objective: To learn more about the natural history of LMNA-related disease. Design: Observational study. Setting: 13 clinical centers in Italy from 2000 through 2018. Patients: 164 carriers of an LMNA mutation. Measurements: Detailed cardiologic and neurologic evaluation at study enrollment and for a median of 10 years of follow-up. Results: The median age at enrollment was 38 years, and 51% of participants were female. Neuromuscular manifestations preceded cardiac signs by a median of 11 years, but by the end of follow-up, 90% of the patients had electrical heart disease followed by structural heart disease. Overall, 10 patients (6%) died, 14 (9%) received a heart transplant, and 32 (20%) had malignant ventricular arrhythmias. Fifteen patients had gait loss, and 6 had respiratory failure. Atrial fibrillation and second- and third-degree atrioventricular block were observed, respectively, in 56% and 51% of patients with combined cardiac and neuromuscular manifestations and 37% and 33% of those with heart disease only. Limitations: Some of the data were collected retrospectively. Neuromuscular manifestations were more frequent in this analysis than in previous studies. Conclusion: Many patients with an LMNA mutation have neurologic symptoms by their 30s and develop progressive cardiac manifestations during the next decade. A substantial proportion of these patients will have life-threatening neurologic or cardiologic conditions. Primary Funding Source: None.


Assuntos
Cardiomiopatias/epidemiologia , Cardiomiopatias/genética , Lamina Tipo A/genética , Distrofias Musculares/epidemiologia , Mutação , Adulto , Arritmias Cardíacas/epidemiologia , Arritmias Cardíacas/genética , Fibrilação Atrial/epidemiologia , Fibrilação Atrial/genética , Bloqueio Atrioventricular/epidemiologia , Bloqueio Atrioventricular/genética , Progressão da Doença , Feminino , Seguimentos , Transtornos Neurológicos da Marcha/epidemiologia , Transtornos Neurológicos da Marcha/genética , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/mortalidade , Transplante de Coração/estatística & dados numéricos , Humanos , Itália/epidemiologia , Masculino , Pessoa de Meia-Idade , Distrofias Musculares/genética , Estudos Prospectivos , Insuficiência Respiratória/epidemiologia , Insuficiência Respiratória/genética
4.
J Mol Cell Cardiol ; 120: 42-52, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29750993

RESUMO

The genetic underpinnings that orchestrate the vertebrate heart rate are not fully understood yet, but of high clinical importance, since diseases of cardiac impulse formation and propagation are common and severe human arrhythmias. To identify novel regulators of the vertebrate heart rate, we deciphered the pathogenesis of the bradycardia in the homozygous zebrafish mutant hiphop (hip) and identified a missense-mutation (N851K) in Na+/K+-ATPase α1-subunit (atp1a1a.1). N851K affects zebrafish Na+/K+-ATPase ion transport capacity, as revealed by in vitro pump current measurements. Inhibition of the Na+/K+-ATPase in vivo indicates that hip rather acts as a hypomorph than being a null allele. Consequently, reduced Na+/K+-ATPase function leads to prolonged QT interval and refractoriness in the hip mutant heart, as shown by electrocardiogram and in vivo electrical stimulation experiments. We here demonstrate for the first time that Na+/K+-ATPase plays an essential role in heart rate regulation by prolonging myocardial repolarization.


Assuntos
Bradicardia/genética , Frequência Cardíaca/genética , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Potenciais de Ação , Alelos , Animais , Bloqueio Atrioventricular/genética , Estimulação Elétrica , Eletrocardiografia , Genes Modificadores , Células HEK293 , Humanos , Bombas de Íon , Transporte de Íons , Mutação de Sentido Incorreto , Miócitos Cardíacos/metabolismo , Polimorfismo de Nucleotídeo Único , Estatísticas não Paramétricas
5.
Mol Genet Genomic Med ; 6(2): 288-293, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29274115

RESUMO

BACKGROUND: DES mutations cause different cardiac and skeletal myopathies. Most of them are missense mutations. METHODS: Using a next-generation sequencing cardiac 174 gene panel, we identified a novel heterozygous in-frame indel mutation (DES-c.493_520del28insGCGT, p.Q165_A174delinsAS) in a Caucasian patient with cardiomyopathy in combination with atrioventricular block and skeletal myopathy. This indel mutation is located in the coding region of the first exon. Family anamnesis revealed a history of sudden cardiac death. We performed cell transfection experiments and in vitro assembly experiments to prove the pathogenicity of this novel DES indel mutation. RESULTS: These experiments revealed a severe filament formation defect of mutant desmin supporting the pathogenicity. In addition, we labeled a skeletal muscle biopsy from the mutation carrier revealing cytoplasmic desmin positive protein aggregates. In summary, we identified and functionally characterized a pathogenic DES indel mutation causing cardiac and skeletal myopathy. CONCLUSION: Our study has relevance for the clinical and genetic interpretation of further DES indel mutations causing cardiac or skeletal myopathies and might be helpful for risk stratification.


Assuntos
Cardiomiopatias/genética , Desmina/genética , Adulto , Bloqueio Atrioventricular/genética , Sequência de Bases/genética , Desmina/metabolismo , Humanos , Mutação INDEL/genética , Filamentos Intermediários/genética , Masculino , Músculo Esquelético/metabolismo , Doenças Musculares/genética , Linhagem
6.
J Am Coll Cardiol ; 70(3): 358-370, 2017 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-28705318

RESUMO

BACKGROUND: Inherited cardiac conduction disease is a rare bradyarrhythmia associated with mutations in various genes that affect action potential propagation. It is often characterized by isolated conduction disturbance of the His-Purkinje system, but it is rarely described as a syndromic form. OBJECTIVES: The authors sought to identify the genetic defect in families with a novel bradyarrhythmia syndrome associated with bone malformation. METHODS: The authors genetically screened 15 European cases with genotype-negative de novo atrioventricular (AV) block and their parents by trio whole-exome sequencing, plus 31 Japanese cases with genotype-negative familial AV block or sick sinus syndrome by targeted exon sequencing of 457 susceptibility genes. Functional consequences of the mutation were evaluated using an in vitro cell expression system and in vivo knockout mice. RESULTS: The authors identified a connexin-45 (Cx45) mutation (p.R75H) in 2 unrelated families (a de novo French case and a 3-generation Japanese family) who presented with progressive AV block, which resulted in atrial standstill without ventricular conduction abnormalities. Affected individuals shared a common extracardiac phenotype: a brachyfacial pattern, finger deformity, and dental dysplasia. Mutant Cx45 expressed in Neuro-2a cells showed normal hemichannel assembly and plaque formation. However, Lucifer yellow dye transfer and gap junction conductance between cell pairs were severely impaired, which suggested that mutant Cx45 impedes gap junction communication in a dominant-negative manner. Tamoxifen-induced, cardiac-specific Cx45 knockout mice showed sinus node dysfunction and atrial arrhythmia, recapitulating the intra-atrial disturbance. CONCLUSIONS: Altogether, the authors showed that Cx45 mutant p.R75H is responsible for a novel disease entity of progressive atrial conduction system defects associated with craniofacial and dentodigital malformation.


Assuntos
Bloqueio Atrioventricular/etiologia , Conexinas/genética , DNA/genética , Deformidades Dentofaciais/complicações , Mutação , Adolescente , Adulto , Animais , Bloqueio Atrioventricular/genética , Bloqueio Atrioventricular/fisiopatologia , Criança , Pré-Escolar , Conexinas/metabolismo , Análise Mutacional de DNA , Deformidades Dentofaciais/genética , Deformidades Dentofaciais/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Eletrocardiografia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Linhagem , Fenótipo , Adulto Jovem
7.
Heart Rhythm ; 14(3): 402-409, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27826129

RESUMO

BACKGROUND: Complete heart block (CHB) is a major complication that occurs after congenital heart surgery. We hypothesized that genetic and clinical factors are associated with the development of postoperative CHB and recovery of atrioventricular (AV) conduction. OBJECTIVE: The purpose of this study was to identify predictors of CHB and recovery after congenital heart surgery. METHODS: Patients undergoing congenital heart surgery at our institution from September 2007 through June 2015 were prospectively enrolled in a parent study of postoperative arrhythmias. Patients with onset of CHB within 48 hours postoperatively were included in the study. Daily rhythm assessment was performed until demonstration of 1:1 conduction or pacemaker implantation. RESULTS: Of 1199 subjects enrolled, 56 (4.7%) developed postoperative CHB. In multivariate analysis, preoperative digoxin exposure (odds ratio [OR] 2.4, 95% confidence interval [CI] 1.3-4.4), aortic cross-clamp time (OR 1.08, 95% CI 1.04-1.11), ventricular septal defect closure (OR 2.2, 95% CI 1.2-4.1), and a common polymorphism in the gene encoding connexin-40 (GJA5 rs10465885 TT genotype; OR 2.1, 95% CI 1.2-3.8) were independently associated with postoperative CHB. Junctional acceleration (JA) (OR 4.0, 95% CI 1.1-15.1) and intermittent conduction noted during complete AV block (OR 9.1, 95% CI 1.0-80) were independently associated with 1:1 AV conduction recovery. Use of a multivariate model including both JA and intermittent conduction demonstrated good discrimination with a positive predictive value of 86% (95% CI 67%-96%) in predicting 1:1 conduction recovery. CONCLUSION: Preoperative factors, including a missense polymorphism in GJA5, are independently associated with increased risk for CHB. JA and intermittent conduction may prove useful in predicting recovery of AV conduction among patients with CHB after congenital heart surgery.


Assuntos
Bloqueio Atrioventricular , Procedimentos Cirúrgicos Cardíacos , Conexinas/genética , Cardiopatias Congênitas , Complicações Pós-Operatórias , Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/etiologia , Bloqueio Atrioventricular/genética , Bloqueio Atrioventricular/prevenção & controle , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Procedimentos Cirúrgicos Cardíacos/métodos , Pré-Escolar , Feminino , Sistema de Condução Cardíaco/fisiopatologia , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/fisiopatologia , Cardiopatias Congênitas/cirurgia , Humanos , Lactente , Masculino , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Complicações Pós-Operatórias/diagnóstico , Complicações Pós-Operatórias/genética , Complicações Pós-Operatórias/prevenção & controle , Valor Preditivo dos Testes , Prognóstico , Recuperação de Função Fisiológica , Proteína alfa-5 de Junções Comunicantes
8.
Circ Heart Fail ; 9(8)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27496753
9.
Heart Rhythm ; 12(11): 2316-24, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26142302

RESUMO

BACKGROUND: Gain-of-function (GOF) mutations in the KATP channel subunits Kir6.1 and SUR2 cause Cantu syndrome (CS), a disease characterized by multiple cardiovascular abnormalities. OBJECTIVE: The purpose of this study was to better determine the electrophysiologic consequences of such GOF mutations in the heart. METHODS: We generated transgenic mice (Kir6.1-GOF) expressing ATP-insensitive Kir6.1[G343D] subunits under α-myosin heavy chain (α-MHC) promoter control, to target gene expression specifically in cardiomyocytes, and performed patch-clamp experiments on isolated ventricular myocytes and invasive electrophysiology on anesthetized mice. RESULTS: In Kir6.1-GOF ventricular myocytes, KATP channels showed decreased ATP sensitivity but no significant change in current density. Ambulatory ECG recordings on Kir6.1-GOF mice revealed AV nodal conduction abnormalities and junctional rhythm. Invasive electrophysiologic analyses revealed slowing of conduction and conduction failure through the AV node but no increase in susceptibility to atrial or ventricular ectopic activity. Surface ECGs recorded from CS patients also demonstrated first-degree AV block and fascicular block. CONCLUSION: The primary electrophysiologic consequence of cardiac KATP GOF is on the conduction system, particularly the AV node, resulting in conduction abnormalities in CS patients who carry KATP GOF mutations.


Assuntos
Bloqueio Atrioventricular/genética , Cardiomegalia/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Regulação da Expressão Gênica no Desenvolvimento , Hipertricose/genética , Canais KATP/genética , Osteocondrodisplasias/genética , Animais , Síndrome de Brugada/genética , Doença do Sistema de Condução Cardíaco , Cardiomegalia/diagnóstico por imagem , Células Cultivadas , Pré-Escolar , Modelos Animais de Doenças , Ecocardiografia Doppler , Eletrocardiografia , Fenômenos Eletrofisiológicos/genética , Humanos , Hipertricose/diagnóstico por imagem , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Osteocondrodisplasias/diagnóstico por imagem , Distribuição Aleatória , Doenças Raras , Estudos de Amostragem
10.
J Am Heart Assoc ; 4(3): e001733, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25773299

RESUMO

BACKGROUND: Atrial fibrillation (AF) is a complex disease process, and the molecular mechanisms underlying initiation and progression of the disease are unclear. Consequently, AF has been difficult to model. In this study, we have presented a novel transgenic mouse model of AF that mimics human disease and characterized the mechanisms of atrial electroanatomical remodeling in the genesis of AF. METHODS AND RESULTS: Cardiac-specific liver kinase B1 (LKB1) knockout (KO) mice were generated, and 47% aged 4 weeks and 95% aged 12 weeks developed spontaneous AF from sinus rhythm by demonstrating paroxysmal and persistent stages of the disease. Electrocardiographic characteristics of sinus rhythm were similar in KO and wild-type mice. Atrioventricular block and atrial flutter were common in KO mice. Heart rate was slower with persistent AF. In parallel with AF, KO mice developed progressive biatrial enlargement with inflammation, heterogeneous fibrosis, and loss of cardiomyocyte population with apoptosis and necrosis. Atrial tissue was infiltrated with inflammatory cells. C-reactive protein, interleukin 6, and tumor necrosis factor α were significantly elevated in serum. KO atria demonstrated elevated reactive oxygen species and decreased AMP-activated protein kinase activity. Cardiomyocyte and myofibrillar ultrastructure were disrupted. Intercellular matrix and gap junction were interrupted. Connexins 40 and 43 were reduced. Persistent AF caused left ventricular dysfunction and heart failure. Survival and exercise capacity were worse in KO mice. CONCLUSIONS: LKB1 KO mice develop spontaneous AF from sinus rhythm and progress into persistent AF by replicating the human AF disease process. Progressive inflammatory atrial cardiomyopathy is the genesis of AF, through mechanistic electrical and structural remodeling.


Assuntos
Fibrilação Atrial/enzimologia , Miócitos Cardíacos/enzimologia , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Quinases Ativadas por AMP/metabolismo , Fatores Etários , Animais , Apoptose , Fibrilação Atrial/genética , Fibrilação Atrial/patologia , Fibrilação Atrial/fisiopatologia , Flutter Atrial/enzimologia , Flutter Atrial/genética , Flutter Atrial/fisiopatologia , Bloqueio Atrioventricular/enzimologia , Bloqueio Atrioventricular/genética , Bloqueio Atrioventricular/fisiopatologia , Cardiomiopatias/enzimologia , Cardiomiopatias/genética , Cardiomiopatias/fisiopatologia , Conexina 43/metabolismo , Conexinas/metabolismo , Progressão da Doença , Tolerância ao Exercício , Fibrose , Genótipo , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca , Humanos , Mediadores da Inflamação/sangue , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/patologia , Necrose , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Espécies Reativas de Oxigênio/metabolismo , Disfunção Ventricular Esquerda/enzimologia , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda , Proteína alfa-5 de Junções Comunicantes
11.
Acta Myol ; 34(2-3): 116-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27199538

RESUMO

Mutations in the lamin A/C gene (LMNA) have been associated with several phenotypes ranging from systemic to prevalent of muscle, heart, skin, nerve etc. More recently they have been associated with dilated cardiomyopathy (DCM) and severe forms of arrhythmogenic right ventricular cardiomyopathy (ARVC). We report four novel mutations - 3 missense and 1 deletion - in 4 unrelated patients showing different phenotypes, ranging from the early onset congenital form of laminopathy to classical LGMD phenotype, to LGMD and heart involvement. All these newly identified variants were not found in 300 ethnicallymatched control subjects. The variant c.103-105del CTG was described post-mortem in a young patient with congenital muscular dystrophy who presented at the age of 9 a first degree A-V block and subsequently several episodes of supraventricular parossystic tachycardia. Two patients presented as onset symptom lower limbs muscle weakness, and developed heart conduction defects requiring pacemaker implantation at the age of 26 and 38 years, respectively. One of them who carried the mutation c.1339G>C died at the age of 40 by intractable heart failure; the second one carrying the mutation 265C>T died at the age of 30, for a trmboembolic event. A classical LGMD phenotype without heart involvement was observed in the patient with the mutation 1579C>T, who died at the age of 68 years for respiratory insufficiency.


Assuntos
Lamina Tipo A/genética , Distrofias Musculares/genética , Adulto , Idoso , Bloqueio Atrioventricular/genética , Biópsia , Síndrome de Brugada/genética , Doença do Sistema de Condução Cardíaco , Criança , Evolução Fatal , Deleção de Genes , Insuficiência Cardíaca/genética , Humanos , Mutação de Sentido Incorreto , Fenótipo , Insuficiência Respiratória/genética , Taquicardia Supraventricular/genética
12.
J Biol Chem ; 289(49): 33730-40, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25281747

RESUMO

Two recent studies (Newton-Cheh, C. et al. (2009) Common variants at ten loci influence QT interval duration in the QTGEN Study. Nat. Genet. 41, 399-406 and Pfeufer, A. et al. (2009) Common variants at ten loci modulate the QT interval duration in the QTSCD Study. Nat. Genet. 41, 407-414) identified an association, with genome-wide significance, between a single nucleotide polymorphism within the gene encoding RING finger protein 207 (RNF207) and the QT interval. We sought to determine the role of RNF207 in cardiac electrophysiology. Morpholino knockdown of RNF207 in zebrafish embryos resulted in action potential duration prolongation, occasionally a 2:1 atrioventricular block, and slowing of conduction velocity. Conversely, neonatal rabbit cardiomyocytes infected with RNF207-expressing adenovirus exhibited shortened action potential duration. Using transfections of U-2 OS and HEK293 cells, Western blot analysis and immunocytochemistry data demonstrate that RNF207 and the human ether-a-go-go-related gene (HERG) potassium channel interact and colocalize. Furthermore, RNF207 overexpression significantly elevated total and membrane HERG protein and HERG-encoded current density by ∼30-50%, which was dependent on the intact N-terminal RING domain of RNF207. Finally, coexpression of RNF207 and HSP70 increased HERG expression compared with HSP70 alone. This effect was dependent on the C terminus of RNF207. Taken together, the evidence is strong that RNF207 is an important regulator of action potential duration, likely via effects on HERG trafficking and localization in a heat shock protein-dependent manner.


Assuntos
Bloqueio Atrioventricular/genética , Canais de Potássio Éter-A-Go-Go/genética , Proteínas de Choque Térmico HSP70/genética , Coração/fisiologia , Miócitos Cardíacos/metabolismo , Ubiquitina-Proteína Ligases/genética , Potenciais de Ação/genética , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Bloqueio Atrioventricular/metabolismo , Bloqueio Atrioventricular/fisiopatologia , Canal de Potássio ERG1 , Embrião não Mamífero , Canais de Potássio Éter-A-Go-Go/metabolismo , Acoplamento Excitação-Contração , Regulação da Expressão Gênica , Vetores Genéticos , Células HEK293 , Proteínas de Choque Térmico HSP70/metabolismo , Coração/embriologia , Coração/fisiopatologia , Humanos , Dados de Sequência Molecular , Morfolinos , Miócitos Cardíacos/patologia , Estrutura Terciária de Proteína , Coelhos , Ubiquitina-Proteína Ligases/metabolismo , Peixe-Zebra
13.
Circ J ; 78(4): 938-42, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24492017

RESUMO

BACKGROUND: Loss-of-function mutations in the HCN4 gene have been shown to be associated with sinus dysfunction, but there are no reports on HCN4-mediated atrioventricular (AV) block. A novel missense HCN4 mutation G1097W was identified in a 69 year-old Japanese male with AV block, and we characterized the functional consequences of If-like channels reconstituted with the heterozygous HCN4 mutation. METHODS AND RESULTS: Wild-type (WT) HCN4 or/and HCN4-G1097W were expressed in a heterologous cell expression system. A functional assay using a whole-cell patch-clamp demonstrated that the mutant If-like currents were activated at more negative voltages compared to WT currents, while they retained the sensitivity to changes in intracellular cyclic adenosine monophosphate (cAMP) levels. Co-expression of G1097W with WT channels showed dominant-negative effects, including a reduction in peak currents and a negative voltage shifting on reconstituted currents. CONCLUSIONS: The HCN4-G1097W mutant channels displayed a loss-of-function type modulation on cardiac If channels and thus could predispose them to AV nodal dysfunction. These data provide a novel insight into the genetic basis for the AV block.


Assuntos
Bloqueio Atrioventricular , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Proteínas Musculares , Canais de Potássio , Idoso , Substituição de Aminoácidos , Bloqueio Atrioventricular/genética , Bloqueio Atrioventricular/mortalidade , Bloqueio Atrioventricular/patologia , Bloqueio Atrioventricular/fisiopatologia , Feminino , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação de Sentido Incorreto , Canais de Potássio/genética , Canais de Potássio/metabolismo
14.
Circ Arrhythm Electrophysiol ; 6(5): 946-51, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23995044

RESUMO

BACKGROUND: Fetal arrhythmias characteristic of long QT syndrome (LQTS) include torsades de pointes (TdP) and/or 2° atrioventricular block, but sinus bradycardia, defined as fetal heart rate<3% for gestational age, is most common. We hypothesized that prenatal rhythm phenotype might predict LQTS genotype and facilitate improved risk stratification and management. METHOD AND RESULTS: Records of subjects exhibiting fetal LQTS arrhythmias were reviewed. Fetal echocardiograms, neonatal ECG, and genetic testing were evaluated. We studied 43 subjects exhibiting fetal LQTS arrhythmias: TdP±2° atrioventricular block (group 1, n=7), isolated 2° atrioventricular block (group 2, n=4), and sinus bradycardia (group 3, n=32). Mutations in known LQTS genes were found in 95% of subjects tested. SCN5A mutations occurred in 71% of group 1, whereas 91% of subjects with KCNQ1 mutations were in group 3. Small numbers of subjects with KCNH2 mutations (n=4) were scattered in all 3 groups. Age at presentation did not differ among groups, and most subjects (n=42) were live-born with gestational ages of 37.5±2.8 weeks (mean±SD). However, those with TdP were typically delivered earlier. Prenatal treatment in group 1 terminated (n=2) or improved (n=4) TdP. The neonatal heart rate-corrected QT interval (mean±SE) of group 1 (664.7±24.9) was longer than neonatal heart rate-corrected QT interval in both group 2 (491.2±27.6; P=0.004) and group 3 (483.1±13.7; P<0.001). Despite medical and pacemaker therapy, postnatal cardiac arrest (n=4) or sudden death (n=1) was common among subjects with fetal/neonatal TdP. CONCLUSIONS: Rhythm phenotypes of fetal LQTS have genotype-suggestive features that, along with heart rate-corrected QT interval duration, may risk stratify perinatal management.


Assuntos
Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/genética , Bradicardia/diagnóstico , Bradicardia/genética , Canais de Potássio Éter-A-Go-Go/genética , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Diagnóstico Pré-Natal , Bloqueio Atrioventricular/fisiopatologia , Bradicardia/fisiopatologia , Canal de Potássio ERG1 , Ecocardiografia , Eletrocardiografia , Feminino , Testes Genéticos , Genótipo , Frequência Cardíaca , Humanos , Recém-Nascido , Síndrome do QT Longo/fisiopatologia , Mutação , Fenótipo , Gravidez , Resultado da Gravidez , Medição de Risco
15.
Circ Arrhythm Electrophysiol ; 6(4): 799-808, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23884198

RESUMO

BACKGROUND: When complete atrioventricular block (AVB) occurs, infranodal escape rhythms are essential to prevent bradycardic death. The role of T-type Ca(2+) channels in pacemaking outside the sinus node is unknown. We investigated the role of T-type Ca(2+) channels in escape rhythms and bradycardia-related ventricular tachyarrhythmias after AVB in mice. METHODS AND RESULTS: Adult male mice lacking the main T-type Ca(2+) channel subunit Cav3.1 (Cav3.1(-/-)) and wild-type (WT) controls implanted with ECG telemetry devices underwent radiofrequency atrioventricular node ablation to produce AVB. Before ablation, Cav3.1(-/-) mice showed sinus bradycardia (mean±SEM; RR intervals, 148±3 versus 128±2 ms WT; P<0.001). Immediately after AVB, Cav3.1(-/-) mice had slower escape rhythms (RR intervals, 650±75 versus 402±26 ms in WT; P<0.01) but a preserved heart-rate response to isoproterenol. Over the next 24 hours, mortality was markedly greater in Cav3.1(-/-) mice (19/31; 61%) versus WT (8/26; 31%; P<0.05), and Torsades de Pointes occurred more frequently (73% Cav3.1(-/-) versus 35% WT; P<0.05). Escape rhythms improved in both groups during the next 4 weeks but remained significantly slower in Cav3.1(-/-). At 4 weeks after AVB, ventricular tachycardia was more frequent in Cav3.1(-/-) than in WT mice (746±116 versus 214±78 episodes/24 hours; P<0.01). Ventricular function remodeling was similar in Cav3.1(-/-) and WT, except for smaller post-AVB fractional-shortening increase in Cav3.1(-/-). Expression changes were seen post-AVB for a variety of genes; these tended to be greater in Cav3.1(-/-) mice, and overexpression of fetal and profibrotic genes occurred only in Cav3.1(-/-). CONCLUSIONS: This study suggests that T-type Ca(2+) channels play an important role in infranodal escape automaticity. Loss of T-type Ca(2+) channels worsens bradycardia-related mortality, increases bradycardia-associated adverse remodeling, and enhances the risk of malignant ventricular tachyarrhythmias complicating AVB.


Assuntos
Bloqueio Atrioventricular/metabolismo , Bradicardia/metabolismo , Canais de Cálcio Tipo T/metabolismo , Sinalização do Cálcio , Sistema de Condução Cardíaco/metabolismo , Frequência Cardíaca , Periodicidade , Torsades de Pointes/metabolismo , Potenciais de Ação , Animais , Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/genética , Bloqueio Atrioventricular/fisiopatologia , Bradicardia/diagnóstico , Bradicardia/genética , Bradicardia/fisiopatologia , Bradicardia/prevenção & controle , Canais de Cálcio Tipo T/deficiência , Canais de Cálcio Tipo T/genética , Modelos Animais de Doenças , Eletrocardiografia Ambulatorial , Técnicas Eletrofisiológicas Cardíacas , Regulação da Expressão Gênica , Sistema de Condução Cardíaco/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Telemetria , Fatores de Tempo , Torsades de Pointes/diagnóstico , Torsades de Pointes/genética , Torsades de Pointes/fisiopatologia , Torsades de Pointes/prevenção & controle , Remodelação Ventricular
16.
Congenit Heart Dis ; 8(6): E165-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23075154

RESUMO

Long QT syndrome in the infant with 2:1 atrioventricular block is a malignant form of disease associated with frequent torsade de pointes in some cases. Those patients that do not respond to antiarrhythmic therapy are particularly challenging to manage. Ventricular pacing in this patient population has been shown to reduce arrhythmic events. We report a case of a newborn with frequent torsade de pointes requiring defibrillation and cardiopulmonary resuscitation with immediate shortening of the QTc interval with ventricular pacing and subsequent resolution of torsade de pointes.


Assuntos
Bloqueio Atrioventricular/terapia , Estimulação Cardíaca Artificial/métodos , Síndrome do QT Longo/terapia , Torsades de Pointes/terapia , Antiarrítmicos/uso terapêutico , Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/genética , Bloqueio Atrioventricular/fisiopatologia , Análise Mutacional de DNA , Canal de Potássio ERG1 , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go/genética , Predisposição Genética para Doença , Frequência Cardíaca , Humanos , Recém-Nascido , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Mutação , Linhagem , Fenótipo , Torsades de Pointes/diagnóstico , Torsades de Pointes/genética , Torsades de Pointes/fisiopatologia , Resultado do Tratamento , Função Ventricular
18.
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
19.
PLoS One ; 6(6): e20203, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21674029

RESUMO

To determine the normal function of the Coxsackievirus and Adenovirus Receptor (CAR), a protein found in tight junctions and other intercellular complexes, we constructed a mouse line in which the CAR gene could be disrupted at any chosen time point in a broad spectrum of cell types and tissues. All knockouts examined displayed a dilated intestinal tract and atrophy of the exocrine pancreas with appearance of tubular complexes characteristic of acinar-to-ductal metaplasia. The mice also exhibited a complete atrio-ventricular block and abnormal thymopoiesis. These results demonstrate that CAR exerts important functions in the physiology of several organs in vivo.


Assuntos
Inativação Gênica , Fenótipo , Receptores Virais/deficiência , Receptores Virais/genética , Animais , Bloqueio Atrioventricular/genética , Atrofia/genética , Comportamento Animal/efeitos dos fármacos , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Feminino , Fertilidade/efeitos dos fármacos , Fertilidade/genética , Técnicas de Inativação de Genes , Inativação Gênica/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Pâncreas Exócrino/efeitos dos fármacos , Pâncreas Exócrino/metabolismo , Pâncreas Exócrino/patologia , Receptores Virais/metabolismo , Tamoxifeno/farmacologia , Timo/citologia , Timo/efeitos dos fármacos , Timo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA