Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 324
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell Physiol Biochem ; 54(4): 696-706, 2020 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-32706220

RESUMO

BACKGROUND/AIMS: Mutations of desmosomal genes are known to cause arrhythmogenic cardiomyopathy characterized by arrhythmias and sudden cardiac death. Previously, we described a novel genetic variant H1684R in desmoplakin gene (DSP), associated with a progressive cardiac conduction disease (PCCD). In the present study, we aimed to investigate an effect of the DSP-H1684R genetic variant on the activity of ion channels. METHODS: We used cardiomyocytes derived from induced pluripotent stem cells (iPSC cardiomyocytes) from a patient with DSP-H1684R genetic variant and from two healthy donors. Immunofluorescent staining and western blot analyses were used to characterize patient-specific cardiomyocytes. By the whole-cell voltage-clamp technique we estimated the activity of voltage-gated sodium, calcium, and potassium channels that are responsible for action potential generation and its shape. Action potentials' parameters were measured using whole-cell current-clamp technique. RESULTS: In patient-specific cardiomyocytes we observed both lower amplitudes of currents through sodium Nav1.5 channels and L-type calcium channels, but higher amplitude of current through transient-outward potassium channels in comparison to donor cardiomyocytes. Current-clamp measurements revealed shortening of action-potential in DSP-H1684R-carrying iPSC cardiomyocytes. Therefore, observed alterations in the channels activity might have a great impact on the properties of action potential and development of PCCD. CONCLUSION: Our results show that desmoplakin genetic variants, besides conduction slowing caused by structural heart remodeling, could affect multiple ion channel activity aggravating arrhythmia manifestation in PCCD.


Assuntos
Doença do Sistema de Condução Cardíaco/genética , Desmoplaquinas/genética , Bloqueio Cardíaco/genética , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Canais Iônicos/fisiologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Potenciais de Ação/fisiologia , Canais de Cálcio/fisiologia , Doença do Sistema de Condução Cardíaco/metabolismo , Desmoplaquinas/metabolismo , Imunofluorescência , Bloqueio Cardíaco/metabolismo , Humanos , Canais Iônicos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Canais de Sódio Disparados por Voltagem/fisiologia
2.
Europace ; 20(10): 1692-1698, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29579189

RESUMO

Aims: Cardiac atrial arrhythmias are the most common type of heart rhythm disorders. Its genetic elucidation remains challenging with poor understanding of cellular and molecular processes. These arrhythmias usually affect elderly population but in rare cases, young children may also suffer from such electrical diseases. Severe complications, including stroke, are commonly age related. This study aims to identify a genetic link between electro-mechanic atrial dysfunction and stroke in children. Methods and results: In two unrelated boys of 11 and 14 years with both stroke and atrial arrhythmias, the clinical phenotype was determined through a complete physical examination, electrocardiogram (ECG), Holter ECG, and computed tomography. The genetic testing was performed on a large 95 genes panel implicated in myocardial electrical imbalance, using the next generation sequencing method. The panel also includes the genes usually associated with the development of cardiomyopathies. In one child, a left atrial dilation was observed. The 2nd boy suffered from atrial standstill. Both suffered from atrial bradycardia, flutter, and fibrillation. The complete genetic testing revealed the SCN5A c.3823G>A (p.D1275N) mutation in the first family, c.1141-2A>G and c.3157G>A (p.E1053K) mutations in the second family. Conclusion: Our results strengthen the association between Nav1.5 mutations and the occurrence of stroke in young patients. It emphasizes the need to look for atrial myopathy in the decision process for anticoagulation in young patients with atrial arrhythmic events.


Assuntos
Fibrilação Atrial/complicações , Flutter Atrial/complicações , Função do Átrio Esquerdo , Bradicardia/complicações , Cardiomiopatias/complicações , Doenças Genéticas Inatas/complicações , Átrios do Coração/anormalidades , Bloqueio Cardíaco/complicações , Acidente Vascular Cerebral/etiologia , Adolescente , Arritmias Cardíacas/complicações , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Fibrilação Atrial/genética , Fibrilação Atrial/fisiopatologia , Flutter Atrial/genética , Flutter Atrial/fisiopatologia , Bradicardia/genética , Bradicardia/fisiopatologia , Cardiomiopatias/genética , Cardiomiopatias/fisiopatologia , Criança , Eletrocardiografia , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/fisiopatologia , Átrios do Coração/fisiopatologia , Bloqueio Cardíaco/genética , Bloqueio Cardíaco/fisiopatologia , Humanos , Masculino , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Fenótipo
3.
Am J Physiol Heart Circ Physiol ; 313(3): H631-H640, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28626076

RESUMO

The signature lesion of SSA/Ro autoantibody-associated congenital heart block (CHB) is fibrosis and a macrophage infiltrate, supporting an experimental focus on cues influencing the fibroblast component. The transcriptomes of human fetal cardiac fibroblasts were analyzed using two complementary approaches. Cardiac injury conditions were simulated in vitro by incubating human fetal cardiac fibroblasts with supernatants from macrophages transfected with the SSA/Ro-associated noncoding Y ssRNA. The top 10 upregulated transcripts in the stimulated fibroblasts reflected a type I interferon (IFN) response [e.g., IFN-induced protein 44-like (IFI44L), of MX dynamin-like GTPase (MX)1, MX2, and radical S-adenosyl methionine domain containing 2 (Rsad2)]. Within the fibrotic pathway, transcript levels of endothelin-1 (EDN1), phosphodiesterase (PDE)4D, chemokine (C-X-C motif) ligand (CXCL)2, and CXCL3 were upregulated, while others, including adenomedullin, RAP guanine nucleotide exchange factor 3 (RAPGEF3), tissue inhibitor of metalloproteinase (TIMP)1, TIMP3, and dual specificity phosphatase 1, were downregulated. Agnostic Database for Annotation, Visualization and Integrated Discovery analysis revealed a significant increase in inflammatory genes, including complement C3A receptor 1 (C3AR1), F2R-like thrombin/trypsin receptor 3, and neutrophil cytosolic factor 2. In addition, stimulated fibroblasts expressed high levels of phospho-MADS box transcription enhancer factor 2 [a substrate of MAPK5 (ERK5)], which was inhibited by BIX-02189, a specific inhibitor of ERK5. Translation to human disease leveraged an unprecedented opportunity to interrogate the transcriptome of fibroblasts freshly isolated and cell sorted without stimulation from a fetal heart with CHB and a matched healthy heart. Consistent with the in vitro data, five IFN response genes were among the top 10 most highly expressed transcripts in CHB fibroblasts. In addition, the expression of matrix-related genes reflected fibrosis. These data support the novel finding that cardiac injury in CHB may occur secondary to abnormal remodeling due in part to upregulation of type 1 IFN response genes.NEW & NOTEWORTHY Congenital heart block is a rare disease of the fetal heart associated with maternal anti-Ro autoantibodies which can result in death and for survivors, lifelong pacing. This study provides in vivo and in vitro transcriptome-support that injury may be mediated by an effect of Type I Interferon on fetal fibroblasts.


Assuntos
Anticorpos Antinucleares/metabolismo , Coração Fetal/metabolismo , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Bloqueio Cardíaco/congênito , Mediadores da Inflamação/metabolismo , Interferon Tipo I/metabolismo , Transcriptoma , Adulto , Anticorpos Antinucleares/genética , Anticorpos Antinucleares/imunologia , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Feminino , Coração Fetal/imunologia , Coração Fetal/patologia , Fibroblastos/patologia , Fibrose , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Bloqueio Cardíaco/genética , Bloqueio Cardíaco/imunologia , Bloqueio Cardíaco/metabolismo , Bloqueio Cardíaco/patologia , Humanos , Mediadores da Inflamação/imunologia , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interferon Tipo I/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Miocárdio , Comunicação Parácrina , Gravidez , Transfecção
4.
Biochem Biophys Res Commun ; 494(1-2): 346-351, 2017 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-28989025

RESUMO

Malfunction of nodal pacemaker (Pm) cardiomyocytes (CMs) due to diseases or aging leads to rhythm generation disorders, necessitating electronic Pm implantation. We functionally reprogrammed human pluripotent stem cell (hPSC) derived-ventricular (V) CMs into -PmCMs via recombinant adeno-associated virus serotype 9 (rAAV9)-mediated overexpression of engineered HCN1 channel (HCN1ΔΔΔ) whose S3-S4 linker has been strategically deleted by design to promote cardiac pacemaking. rAAV9-HCN1ΔΔΔ-reprogrammed hPSC-PmCMs converted from -VCMs showed automaticity and action potential parameters typical of native nodal PmCMs. Implantation of rAAV9-HCN1ΔΔΔ-based BPm in a preclinical porcine model of complete heart block significantly reduced the dependence on device-supported pacing and generated spontaneous heart rhythms from the BPm. Collectively, these results have further laid the groundwork on BPm for future translation.


Assuntos
Dependovirus/metabolismo , Bloqueio Cardíaco/terapia , Ventrículos do Coração/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/metabolismo , Canais de Potássio/metabolismo , Potenciais de Ação/fisiologia , Animais , Diferenciação Celular , Reprogramação Celular , Dependovirus/genética , Modelos Animais de Doenças , Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Bloqueio Cardíaco/genética , Bloqueio Cardíaco/metabolismo , Bloqueio Cardíaco/fisiopatologia , Frequência Cardíaca/fisiologia , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Miócitos Cardíacos/citologia , Marca-Passo Artificial , Células-Tronco Pluripotentes/citologia , Canais de Potássio/genética , Suínos
5.
Mol Pharmacol ; 89(1): 176-86, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26494861

RESUMO

The molecular pharmacology of the G protein-coupled receptors for sphingosine 1-phosphate (S1P) provides important insight into established and new therapeutic targets. A new, potent bitopic S1P3 antagonist, SPM-354, with in vivo activity, has been used, together with S1P3-knockin and S1P3-knockout mice to define the spatial and functional properties of S1P3 in regulating cardiac conduction. We show that S1P3 is a key direct regulator of cardiac rhythm both in vivo and in isolated perfused hearts. 2-Amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol in vivo and S1P in isolated hearts induced a spectrum of cardiac effects, ranging from sinus bradycardia to complete heart block, as measured by a surface electrocardiogram in anesthetized mice and in volume-conducted Langendorff preparations. The agonist effects on complete heart block are absent in S1P3-knockout mice and are reversed in wild-type mice with SPM-354, as characterized and described here. Homologous knockin of S1P3-mCherry is fully functional pharmacologically and is strongly expressed by immunohistochemistry confocal microscopy in Hyperpolarization Activated Cyclic Nucleotide Gated Potassium Channel 4 (HCN4)-positive atrioventricular node and His-Purkinje fibers, with relative less expression in the HCN4-positive sinoatrial node. In Langendorff studies, at constant pressure, SPM-354 restored sinus rhythm in S1P-induced complete heart block and fully reversed S1P-mediated bradycardia. S1P3 distribution and function in the mouse ventricular cardiac conduction system suggest a direct mechanism for heart block risk that should be further studied in humans. A richer understanding of receptor and ligand usage in the pacemaker cells of the cardiac system is likely to be useful in understanding ventricular conduction in health, disease, and pharmacology.


Assuntos
Bloqueio Cardíaco/tratamento farmacológico , Bloqueio Cardíaco/genética , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Animais , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Bloqueio Cardíaco/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Esfingosina-1-Fosfato
6.
Zhonghua Xin Xue Guan Bing Za Zhi ; 44(5): 411-5, 2016 May 24.
Artigo em Zh | MEDLINE | ID: mdl-27220576

RESUMO

OBJECTIVE: To define the potential causative gene mutation in a Chinese pedigree with progressive cardiac conduction defect (PCCD). METHODS: Sanger sequencing was performed to define potential causative gene mutation in a four-generation family with 68 members including seven PCCD patients (5 male) from 2010 to 2015.No causative gene was detected by screening known candidate genes related to PCCD including SCN5A, NKX2.5 and LMNA.High-throughput sequencing technology on exon-enriched DNA was then used to search the causative genes in 2 patients and one normal family member. RESULTS: Eight new non-synonymous single nucleotide variants including AQP7 gene (exon5: c.T343C: p.Y115H), CACNA1B gene (NM_001243812: exon19: c.A2986G: p.T996A), CATSPERB gene (exon27: c.C3254G: p.P1085R), CLCA2 gene (exon11: c.G1725T: p.W575C), CLCA3P gene (ncRNA_intronic), MYLK-AS1 gene (ncRNA_intronic), TTN gene (ncRNA_UTR3), LMNA gene (LMNA: NM_170708: exon5: c.C922T: p.Q308X) were identified by comparing and filtering the results with known public databases.Then, more detailed biological analysis on these 8 genes was conducted.Traditional Sanger sequencing validated the exome sequencing results, and found that the mutation c. 1725Gï¹¥T in gene CLCA2 segregated with the phenotype of this PCCD pedigree.The mutation c. 1725Gï¹¥T in gene CLCA2 was thus be considered as the causative PCCD gene in this pedigree from the perspective of genetics and genomics. CONCLUSION: The heterozygote mutation c. 1725Gï¹¥T in gene CLCA2 might be causative gene in this PCCD pedigree.This finding adds new gene mutation variant responsible for PCCD.


Assuntos
Canais de Cloreto/genética , Bloqueio Cardíaco/genética , Povo Asiático , Doença do Sistema de Condução Cardíaco , China , Análise Mutacional de DNA , Éxons , Feminino , Humanos , Masculino , Mutação , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único
7.
Hum Mol Genet ; 22(18): 3654-66, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23686279

RESUMO

Focal segmental glomerulosclerosis (FSGS) is the consequence of a disease process that attacks the kidney's filtering system, causing serious scarring. More than half of FSGS patients develop chronic kidney failure within 10 years, ultimately requiring dialysis or renal transplantation. There are currently several genes known to cause the hereditary forms of FSGS (ACTN4, TRPC6, CD2AP, INF2, MYO1E and NPHS2). This study involves a large, unique, multigenerational Australian pedigree in which FSGS co-segregates with progressive heart block with apparent X-linked recessive inheritance. Through a classical combined approach of linkage and haplotype analysis, we identified a 21.19 cM interval implicated on the X chromosome. We then used a whole exome sequencing approach to identify two mutated genes, NXF5 and ALG13, which are located within this linkage interval. The two mutations NXF5-R113W and ALG13-T141L segregated perfectly with the disease phenotype in the pedigree and were not found in a large healthy control cohort. Analysis using bioinformatics tools predicted the R113W mutation in the NXF5 gene to be deleterious and cellular studies support a role in the stability and localization of the protein suggesting a causative role of this mutation in these co-morbid disorders. Further studies are now required to determine the functional consequence of these novel mutations to development of FSGS and heart block in this pedigree and to determine whether these mutations have implications for more common forms of these diseases in the general population.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X/genética , Glomerulosclerose Segmentar e Focal/genética , Bloqueio Cardíaco/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Ligação a RNA/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Austrália , Criança , Pré-Escolar , Exoma , Feminino , Genes Ligados ao Cromossomo X , Ligação Genética , Células HEK293 , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Mutação , N-Acetilglucosaminiltransferases/genética , Especificidade de Órgãos , Linhagem , Análise de Sequência de DNA , Adulto Jovem
8.
Int Heart J ; 56(4): 421-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26104176

RESUMO

Arrhythmias are associated with reduced quality of life and poor prognosis in patients with hypertrophic cardiomyopathy (HCM). Recent genome-wide association studies revealed that a nonsynonymous single nucleotide polymorphism, rs6795970, in the SCN10A gene was associated with the PR interval. We examined whether the PR prolonging allele (A allele) in the SCN10A gene may be associated with cardiac conduction abnormalities in HCM patients.We genotyped the polymorphism in 149 HCM patients. Conduction abnormalities were defined as first-degree heart block, bundle-branch block, and bifascicular heart block. Patients were divided into two groups: group A consisted of 122 patients (82%) without a conduction abnormality; and group B consisted of 27 patients (18%) with one or more cardiac conduction abnormalities. The frequency distribution of the SCN10A genotypes (G/G, G/A, and A/A) among the patients with HCM was 71%, 26%, and 3%, respectively. A cardiac conduction abnormality was documented in 9% with G/G and 40% with G/A or A/A. There was a significant difference in the genotype distribution between the two groups (P = 0.0002). In the dominant A allele model, there was a significant difference in genotypes between the two groups (P < 0.0001). In addition, the A allele remained significant after adjusting for other covariates in a multivariate model (odds ratio = 6.30 [95% confidence interval: 2.24 to 19.09], P = 0.0005).The rs6795970 in the SCN10A gene, which is reported to carry a high risk of heart block, might be associated with cardiac conduction abnormalities in HCM patients.


Assuntos
Cardiomiopatia Hipertrófica/genética , Bloqueio Cardíaco/genética , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Qualidade de Vida , Idoso , Cardiomiopatia Hipertrófica/complicações , Cardiomiopatia Hipertrófica/diagnóstico , Cardiomiopatia Hipertrófica/fisiopatologia , Eletrocardiografia/métodos , Feminino , Predisposição Genética para Doença , Bloqueio Cardíaco/diagnóstico , Bloqueio Cardíaco/etiologia , Bloqueio Cardíaco/psicologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Japão , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Prognóstico
9.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(5): 661-4, 2015 Oct.
Artigo em Zh | MEDLINE | ID: mdl-26418986

RESUMO

OBJECTIVE To explore the genetic mechanism for a family affected with cardiac conduction block. METHODS Affected family members were screened for potential mutations of known candidate genes. As no pathogenic mutation was found, two patients and one healthy member from the family were further analyzed by exomic sequencing followed by Sanger sequencing. The pathogenicity of suspected mutation was analyzed using bioinformatics software. RESULTS Sequencing of the full exome has identified a c.G1725T mutation in the CLCA2 gene. Sanger sequencing has detected the same mutation in all five patients, but not in the normal member from the family. Bioinformatics analysis indicated that the mutation has resulted in substitution of the 575th amino acid cysteine (C) by tryptophan (W). The site is highly conserved and becomes pathogenic with the mutation. CONCLUSION The heterozygous c.G1725T mutation in exon 11 of the CLCA2 gene probably underlies the disease and fit the autosomal dominant pattern of inheritance.


Assuntos
Canais de Cloreto/genética , Bloqueio Cardíaco/genética , Mutação , Sequência de Aminoácidos , Biologia Computacional , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular
11.
J Intern Med ; 275(6): 640-51, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24354957

RESUMO

OBJECTIVE: The main aim of this study was to identify foetal susceptibility genes on chromosome six for Ro/SSA autoantibody-mediated congenital heart block. SUBJECTS AND DESIGN: Single nucleotide polymorphism (SNP) genotyping of individuals in the Swedish Congenital Heart Block (CHB) study population was performed. Low-resolution HLA-A, -Cw and -DRB1 allele typing was carried out in 86 families comprising 339 individuals (86 Ro/SSA autoantibody-positive mothers, 71 fathers, 87 CHB index cases and 95 unaffected siblings). RESULTS: A case-control comparison between index cases and population-based out-of-study controls (n = 1710) revealed association of CHB with 15 SNPs in the 6p21.3 MHC locus at a chromosome-wide significance of P < 2.59 × 10(-6) (OR 2.21-3.12). In a family-based analysis of association of SNP markers as well as distinct MHC class I and II alleles with CHB, HLA-DRB1*04 and HLA-Cw*05 variants were significantly more frequently transmitted to affected individuals (P < 0.03 and P < 0.05, respectively), whilst HLA-DRB1*13 and HLA-Cw*06 variants were significantly less often transmitted to affected children (P < 0.04 and P < 0.03). We further observed marked association of increased paternal (but not maternal) HLA-DRB1*04 transmission to affected offspring (P < 0.02). CONCLUSIONS: HLA-DRB1*04 and HLA-Cw*05 were identified as novel foetal HLA allele variants that confer susceptibility to CHB in response to Ro/SSA autoantibody exposure, whilst DRB1*13 and Cw*06 emerged as protective alleles. Additionally, we demonstrated a paternal contribution to foetal susceptibility to CHB for the first time.


Assuntos
Antígenos HLA-C/genética , Cadeias HLA-DRB1/genética , Bloqueio Cardíaco/congênito , Adulto , Anticorpos Antinucleares , Criança , Saúde da Família , Pai , Feminino , Frequência do Gene , Genes MHC da Classe II/genética , Predisposição Genética para Doença , Bloqueio Cardíaco/epidemiologia , Bloqueio Cardíaco/genética , Humanos , Masculino , Polimorfismo de Nucleotídeo Único , Suécia/epidemiologia
12.
J Pediatr ; 165(5): 1050-2, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25171853

RESUMO

An 11-year-old girl on evaluation for syncope was found to have progressive sinus node dysfunction and His-Purkinje system disease with atrial standstill. Genetic analysis revealed compound heterozygous mutations of the SCN5A gene in a novel combination.


Assuntos
Arritmias Cardíacas/genética , Cardiomiopatias/genética , Doenças Genéticas Inatas/genética , Átrios do Coração/anormalidades , Bloqueio Cardíaco/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Criança , Eletrocardiografia , Feminino , Sistema de Condução Cardíaco/fisiopatologia , Heterozigoto , Humanos , Mutação
13.
Proc Natl Acad Sci U S A ; 108(4): 1705-10, 2011 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-21220308

RESUMO

Cardiac pacemaking generation and modulation rely on the coordinated activity of several processes. Although a wealth of evidence indicates a relevant role of the I(f) ("funny," or pacemaker) current, whose molecular constituents are the hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels and particularly HCN4, work with mice where Hcn genes were knocked out, or functionally modified, has challenged this view. However, no previous studies used a cardiac-specific promoter to induce HCN4 ablation in adult mice. We report here that, in an inducible and cardiac-specific HCN4 knockout (ciHCN4-KO) mouse model, ablation of HCN4 consistently leads to progressive development of severe bradycardia (∼50% reduction of original rate) and AV block, eventually leading to heart arrest and death in about 5 d. In vitro analysis of sinoatrial node (SAN) myocytes isolated from ciHCN4-KO mice at the mean time of death revealed a strong reduction of both the I(f) current (by ∼70%) and of the spontaneous rate (by ∼60%). In agreement with functional results, immunofluorescence and Western blot analysis showed reduced expression of HCN4 protein in SAN tissue and cells. In ciHCN4-KO animals, the residual I(f) was normally sensitive to ß-adrenergic receptor (ß-AR) modulation, and the permanence of rate response to ß-AR stimulation was observed both in vivo and in vitro. Our data show that cardiac HCN4 channels are essential for normal heart impulse generation and conduction in adult mice and support the notion that dysfunctional HCN4 channels can be a direct cause of rhythm disorders. This work contributes to identifying the molecular mechanism responsible for cardiac pacemaking.


Assuntos
Bradicardia/fisiopatologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Bloqueio Cardíaco/fisiopatologia , Coração/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Animais , Western Blotting , Conservadores da Densidade Óssea/farmacologia , Bradicardia/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Eletrocardiografia , Feminino , Imunofluorescência , Coração/efeitos dos fármacos , Bloqueio Cardíaco/genética , Frequência Cardíaca/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos beta/metabolismo , Nó Sinoatrial/efeitos dos fármacos , Nó Sinoatrial/metabolismo , Nó Sinoatrial/fisiologia , Tamoxifeno/farmacologia
14.
Nat Biomed Eng ; 8(9): 1124-1141, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38698155

RESUMO

The adenovirus-mediated somatic transfer of the embryonic T-box transcription factor 18 (TBX18) gene can convert chamber cardiomyocytes into induced pacemaker cells. However, the translation of therapeutic TBX18-induced cardiac pacing faces safety challenges. Here we show that the myocardial expression of synthetic TBX18 mRNA in animals generates de novo pacing and limits innate and inflammatory immune responses. In rats, intramyocardially injected mRNA remained localized, whereas direct myocardial injection of an adenovirus carrying a reporter gene resulted in diffuse expression and in substantial spillover to the liver, spleen and lungs. Transient expression of TBX18 mRNA in rats led to de novo automaticity and pacemaker properties and, compared with the injection of adenovirus, to substantial reductions in the expression of inflammatory genes and in activated macrophage populations. In rodent and clinically relevant porcine models of complete heart block, intramyocardially injected TBX18 mRNA provided rate-adaptive cardiac pacing for one month that strongly correlated with the animal's sinus rhythm and physical activity. TBX18 mRNA may aid the development of biological pacemakers.


Assuntos
Miocárdio , RNA Mensageiro , Proteínas com Domínio T , Animais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Suínos , Miocárdio/metabolismo , Bloqueio Cardíaco/terapia , Bloqueio Cardíaco/genética , Adenoviridae/genética , Masculino , Estimulação Cardíaca Artificial/métodos , Miócitos Cardíacos/metabolismo , Feminino
16.
Nat Genet ; 21(1): 133-7, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9916807

RESUMO

Mutations of mitochondrial DNA (mtDNA) cause several well-recognized human genetic syndromes with deficient oxidative phosphorylation and may also have a role in ageing and acquired diseases of old age. We report here that hallmarks of mtDNA mutation disorders can be reproduced in the mouse using a conditional mutation strategy to manipulate the expression of the gene encoding mitochondrial transcription factor A (Tfam, previously named mtTFA), which regulates transcription and replication of mtDNA. Using a loxP-flanked Tfam allele (TfamloxP) in combination with a cre-recombinase transgene under control of the muscle creatinine kinase promoter, we have disrupted Tfam in heart and muscle. Mutant animals develop a mosaic cardiac-specific progressive respiratory chain deficiency, dilated cardiomyopathy, atrioventricular heart conduction blocks and die at 2-4 weeks of age. This animal model reproduces biochemical, morphological and physiological features of the dilated cardiomyopathy of Kearns-Sayre syndrome. Furthermore, our findings provide genetic evidence that the respiratory chain is critical for normal heart function.


Assuntos
Cardiomiopatia Dilatada/genética , DNA Mitocondrial , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Bloqueio Cardíaco/genética , Coração/fisiopatologia , Proteínas de Grupo de Alta Mobilidade , Proteínas Mitocondriais , Proteínas Nucleares , Transativadores , Fatores de Transcrição/biossíntese , Proteínas Virais , Proteínas de Xenopus , Animais , Cardiomiopatia Dilatada/fisiopatologia , Creatina Quinase/genética , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Bloqueio Cardíaco/fisiopatologia , Humanos , Integrases/genética , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético , Miocárdio , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fatores de Transcrição/genética
17.
Circ Res ; 107(3): 374-87, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20558828

RESUMO

RATIONALE: Binding of maternal anti-Ro/La antibodies to cognate antigen expressed on apoptotic cardiocytes decreases clearance by healthy cardiocytes, which may contribute to the development of autoimmune associated congenital heart block and fatal cardiomyopathy. OBJECTIVE: Given recent evidence implicating the urokinase plasminogen activator receptor (uPAR) as a "don't eat me" signal during efferocytosis, experiments addressed whether surface bound anti-Ro antibodies inhibit apoptotic cell removal via an effect on the expression/function of the urokinase-type plasminogen activator protease uPA/uPAR system. METHODS AND RESULTS: As assessed by flow cytometry and confocal microscopy, uPAR colocalizes and interacts with Ro60 on the surface of apoptotic human fetal cardiocytes. Blocking of uPAR enhances phagocytosis of apoptotic cardiocytes by healthy cardiocytes and reverses the anti-Ro60-dependent impaired clearance of apoptotic cardiocytes. Binding of anti-Ro60 antibodies to apoptotic cardiocytes results in increased uPAR expression, as well as enhanced uPA activity. The binding of anti-Ro60 did not alter other surface molecules involved in cell recognition (calreticulin, CD31, or CD47). CONCLUSIONS: These data suggest that increased uPAR expression and uPA activity induced by anti-Ro60 binding to the apoptotic fetal cardiocyte provide a molecular basis by which these antibodies inhibit efferocytosis and ultimately lead to scar of the fetal conduction system and working myocardium.


Assuntos
Bloqueio Cardíaco/congênito , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Anticorpos Monoclonais , Apoptose , Antígeno CD47/metabolismo , Calreticulina/metabolismo , Cardiomiopatias/imunologia , Galinhas/imunologia , Feminino , Doenças Fetais/genética , Feto/fisiologia , Citometria de Fluxo , Bloqueio Cardíaco/etiologia , Bloqueio Cardíaco/genética , Sistema de Condução Cardíaco/patologia , Humanos , Imunoglobulina G , Camundongos , Microscopia Confocal , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
18.
J Cardiovasc Electrophysiol ; 22(8): 922-30, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21352396

RESUMO

INTRODUCTION: Congenital heart block (CHB) is a passively acquired autoimmune disease considered to be due to the transfer of maternal autoantibodies, anti-SSA/Ro -SSB/La, to the fetus resulting in atrioventricular (AV) block and sinus bradycardia. We previously established a murine model for CHB where pups born to immunized wild-type (WT) mothers exhibited electrocardiographic abnormalities similar to those seen in CHB and demonstrated inhibition of L-type Ca channels (LTCCs) by maternal antibodies. Here, we hypothesize that overexpression of LTCC should rescue, whereas knockout of LTCC should worsen the electrocardiographic abnormalities in mice. METHODS AND RESULTS: Transgenic (TG) mice were immunized with SSA/Ro and SSB/La antigens. Pups born to immunized WT mothers had significantly greater sinus bradycardia and AV block compared to pups from nonimmunized WT. TG pups overexpressing LTCC had significantly less sinus bradycardia and AV block compared to their non-TG littermates and to pups born to immunized WT mothers. All LTCC knockout pups born to immunized mothers had sinus bradycardia, advanced degree of AV block, and decreased fetal parity. No sinus bradycardia or AV block were manifested in pups from control nonimmunized WT mothers. IgG from mothers with CHB children, but not normal IgG, completely inhibited intracellular Ca transient ([Ca](i)T) amplitude. CONCLUSIONS: Cardiac-specific overexpression of LTCC significantly reduced the incidence of AV block and sinus bradycardia in pups exposed to anti-SSA/Ro -SSB/La autoantibodies, whereas exposure of LTCC knockout pups to these autoantibodies significantly worsened the electrocardiographic abnormalities. These findings support the hypothesis that maternal antibodies inhibit LTCC and [Ca](i)T thus contributing to the development of CHB. Altogether, the results are relevant to the development of novel therapies for CHB.


Assuntos
Canais de Cálcio Tipo L/deficiência , Eletrocardiografia , Bloqueio Cardíaco/congênito , Animais , Autoantígenos/imunologia , Canais de Cálcio Tipo L/genética , Eletrocardiografia/métodos , Feminino , Bloqueio Cardíaco/genética , Bloqueio Cardíaco/imunologia , Bloqueio Cardíaco/fisiopatologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Gravidez , Ribonucleoproteínas/imunologia , Antígeno SS-B
19.
Pacing Clin Electrophysiol ; 33(3): 274-85, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20025708

RESUMO

BACKGROUND: Inherited loss of function mutations in SCN5A have been linked to overlapping syndromes including cardiac conduction disease and Brugada syndrome (BrS). The mechanisms responsible for the development of one without the other are poorly understood. METHODS: Direct sequencing was performed in a family with cardiac conduction disease. Wild-type (WT) and mutant channels were expressed in TSA201 cells for electrophysiological study. Green fluorescent protein (GFP)-fused WT or mutant genes were used to assess channel trafficking. RESULTS: A novel SCN5A mutation, P1008S, was identified in all family members displaying first-degree atrioventricular block, but not in unaffected family members nor in 430 reference alleles. Peak P1008S current was 11.77% of WT (P < 0.001). Confocal microscopy showed that WT channels tagged with GFP were localized on the cell surface, whereas GFP-tagged P1008S channels remained trapped in intracellular organelles. Trafficking could be rescued by incubation at room temperature, but not by incubation with mexiletine (300 muM) at 37 degrees C. We also identified a novel polymorphism (D601E) in CACNB2b that slowed inactivation of L-type calcium current (I(Ca,L)), significantly increased total charge. Using the Luo-Rudy action potential (AP) model, we show that the reduction in sodium current (I(Na)) can cause loss of the right ventricular epicardial AP dome in the absence but not in the presence of the slowed inactivation of I(Ca,L). Slowed conduction was present in both cases. CONCLUSIONS: Our results suggest genetic variations leading to a loss-of-function in I(Na) coupled with a gain of function in I(Ca,L) may underlie the development of cardiac conduction disease without BrS.


Assuntos
Bradicardia/genética , Canais de Cálcio Tipo L/genética , Bloqueio Cardíaco/genética , Sistema de Condução Cardíaco/fisiopatologia , Proteínas Musculares/genética , Mutação , Polimorfismo de Nucleotídeo Único , Canais de Sódio/genética , Adolescente , Alelos , Análise de Variância , Bradicardia/fisiopatologia , Síndrome de Brugada/genética , Síndrome de Brugada/fisiopatologia , Técnicas Eletrofisiológicas Cardíacas , Feminino , Bloqueio Cardíaco/fisiopatologia , Humanos , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.5 , Linhagem , Fenótipo , Reação em Cadeia da Polimerase
20.
Pediatr Cardiol ; 31(3): 422-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20033147

RESUMO

Pediatric heart disease comprises many forms of cardiovascular disease in the young including cardiovascular malformations (CVM), cardiomyopathies, vasculopathies, e.g., Marfan syndrome, and cardiac arrhythmias. CVM are an important component of pediatric heart disease and constitute a major portion of clinically significant birth defects. In the past decade, the complementary nature of genetic, developmental, and biochemical approaches have contributed to extraordinary advances in understanding the origins of pediatric heart disease. Results of the studies of the cardiac transcription factor, NKX2.5, illustrate these accomplishments and at the same time provide a forecast of the nature of future genetic studies to better understand the origins of pediatric heart disease.


Assuntos
Bloqueio Cardíaco/genética , Cardiopatias Congênitas/genética , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Bloqueio Cardíaco/embriologia , Cardiopatias Congênitas/embriologia , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Humanos , Síndrome de Marfan/embriologia , Síndrome de Marfan/genética , Mutação , Fenótipo , Fatores de Risco , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA