Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Brain Dev ; 46(1): 57-61, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37778966

RESUMO

BACKGROUND: Some patients with ATP1A3 variant-associated polymicrogyria have recurrent transient heart failure. However, effective treatment for the transient cardiac condition remains to be elucidated. CASE REPORT: The patient started experiencing focal motor onset seizures in 12 h after birth, revealing bilateral diffuse polymicrogyria. The patient also experienced transient bradycardia (sinus bradycardia) attacks from 15 days old. Echocardiography revealed a reduced ejection fraction; however, no obvious electrocorticogram or electroencephalogram abnormalities were observed during the attacks. Initially, the attacks occurred in clusters daily. They later decreased in frequency, occurring at monthly intervals. Repeated episodes of transient bradycardia attacks and polymicrogyria indicated possible ATP1A3 gene abnormality and genetic testing revealed a novel heterozygous ATP1A3 variant (NM_152296: exon22:c.2977_2982del:p.(Glu993_Ile994del)), which was not found in the patient's parents. Cilostazol was administered at 3 months old for recurrent transient bradycardia attacks. Cilostazol significantly shortened the duration of bradycardia episodes and prolonged the interval between attacks. Cilostazol also effectively treats transient symptomatic bradycardia. CONCLUSION: Cilostazol could be a treatment option for recurrent transient bradycardia attacks associated with ATP1A3 gene abnormalities and polymicrogyria.


Assuntos
Insuficiência Cardíaca , Polimicrogiria , Humanos , Lactente , Cilostazol , Bradicardia/tratamento farmacológico , Bradicardia/genética , Polimicrogiria/tratamento farmacológico , Polimicrogiria/genética , Polimicrogiria/complicações , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/complicações , Convulsões/complicações , ATPase Trocadora de Sódio-Potássio/genética
2.
J Am Heart Assoc ; 10(17): e019887, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34459253

RESUMO

Background Pathogenic variation in the ATP1A3-encoded sodium-potassium ATPase, ATP1A3, is responsible for alternating hemiplegia of childhood (AHC). Although these patients experience a high rate of sudden unexpected death in epilepsy, the pathophysiologic basis for this risk remains unknown. The objective was to determine the role of ATP1A3 genetic variants on cardiac outcomes as determined by QT and corrected QT (QTc) measurements. Methods and Results We analyzed 12-lead ECG recordings from 62 patients (male subjects=31, female subjects=31) referred for AHC evaluation. Patients were grouped according to AHC presentation (typical versus atypical), ATP1A3 variant status (positive versus negative), and ATP1A3 variant (D801N versus other variants). Manual remeasurements of QT intervals and QTc calculations were performed by 2 pediatric electrophysiologists. QTc measurements were significantly shorter in patients with positive ATP1A3 variant status (P<0.001) than in patients with genotype-negative status, and significantly shorter in patients with the ATP1A3-D801N variant than patients with other variants (P<0.001). The mean QTc for ATP1A3-D801N was 344.9 milliseconds, which varied little with age, and remained <370 milliseconds throughout adulthood. ATP1A3 genotype status was significantly associated with shortened QTc by multivariant regression analysis. Two patients with the ATP1A3-D801N variant experienced ventricular fibrillation, resulting in death in 1 patient. Rare variants in ATP1A3 were identified in a large cohort of genotype-negative patients referred for arrhythmia and sudden unexplained death. Conclusions Patients with AHC who carry the ATP1A3-D801N variant have significantly shorter QTc intervals and an increased likelihood of experiencing bradycardia associated with life-threatening arrhythmias. ATP1A3 variants may represent an independent cause of sudden unexplained death. Patients with AHC should be evaluated to identify risk of sudden death.


Assuntos
Bradicardia , Hemiplegia , ATPase Trocadora de Sódio-Potássio , Fibrilação Ventricular , Arritmias Cardíacas , Bradicardia/genética , Pré-Escolar , Suscetibilidade a Doenças , Feminino , Genótipo , Hemiplegia/genética , Humanos , Masculino , Mutação , ATPase Trocadora de Sódio-Potássio/genética , Fibrilação Ventricular/genética
3.
PLoS One ; 16(4): e0249997, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33914752

RESUMO

Second-generation antipsychotics (SGAs) play a critical role in current treatment of schizophrenia (SCZ). It has been observed that sinus bradycardia, rare but in certain situations life threatening adverse drug reaction, can be induced by SGAs across different schizophrenia populations. However, the roles of genetic factors in this phenomenon have not been studied yet. In the present study, a genome-wide association study of single nucleotide polymorphisms (SNPs) was performed on Chinese Han SCZ patients to identify susceptibility loci that were associated with sinus bradycardia induced by SGAs. This study applied microarray to obtain genotype profiles of 88 Han Chinese SCZ patients. Our results found that there were no SNPs had genome-wide significant association with sinus bradycardia induced by SGAs. The top GWAS hit located in gene KIAA0247, which mainly regulated by the tumor suppressor P53 and thus plays a role in carcinogenesis based on resent research and it should not be a susceptibility locus to sinus bradycardia induced by SGAs. Using gene-set functional analysis, we tested that if top 500 SNPs mapped genes were relevant to sinus bradycardia. The result of gene prioritization analysis showed CTNNA3 was strongly correlated with sinus bradycardia, hinting it was a susceptibility gene of this ADR. Our study provides a preliminary study of genetic variants associated with sinus bradycardia induced by SGAs in Han Chinese SCZ patients. The discovery of a possible susceptibility gene shed light on further study of this adverse drug reaction in Han Chinese SCZ patients.


Assuntos
Antipsicóticos/efeitos adversos , Bradicardia/etiologia , Estudo de Associação Genômica Ampla , Esquizofrenia/genética , Adulto , Antipsicóticos/uso terapêutico , Bradicardia/genética , China , Feminino , Genótipo , Humanos , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Mapas de Interação de Proteínas/genética , Esquizofrenia/tratamento farmacológico , Esquizofrenia/patologia , Proteína Supressora de Tumor p53/metabolismo , alfa Catenina/genética
4.
Thyroid ; 31(2): 315-326, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32762296

RESUMO

Background: Mutations of thyroid hormone receptor α1 (TRα1) cause resistance to thyroid hormone (RTHα). Patients exhibit growth retardation, delayed bone development, anemia, and bradycardia. By using mouse models of RTHα, much has been learned about the molecular actions of TRα1 mutants that underlie these abnormalities in adults. Using zebrafish models of RTHα that we have recently created, we aimed to understand how TRα1 mutants affect the heart function during this period. Methods: In contrast to human and mice, the thra gene is duplicated, thraa and thrab, in zebrafish. Using CRISPR/Cas9-mediated targeted mutagenesis, we created C-terminal mutations in each of two duplicated thra genes in zebrafish (thraa 8-bp insertion or thrab 1-bp insertion mutations). We recently showed that these mutant fish faithfully recapitulated growth retardation as found in patients and thra mutant mice. In the present study, we used histological analysis, gene expression profiles, confocal fluorescence, and transmission electron microscopy (TEM) to comprehensively analyze the phenotypic characteristics of mutant fish heart during development. Results: We found both a dilated atrium and an abnormally shaped ventricle in adult mutant fish. The retention of red blood cells in the two abnormal heart chambers, and the decreased circulating blood speed and reduced expression of contractile genes indicated weakened contractility in the heart of mutant fish. These abnormalities were detected in mutant fish as early as 35 days postfertilization (juveniles). Furthermore, the expression of genes associated with the sarcomere assembly was suppressed in the heart of mutant fish, resulting in abnormalities of sarcomere organization as revealed by TEM, suggesting that the abnormal sarcomere organization could underlie the bradycardia exhibited in mutant fish. Conclusions: Using a zebrafish model of RTHα, the present study demonstrated for the first time that TRα1 mutants could act to cause abnormal heart structure, weaken contractility, and disrupt sarcomere organization that affect heart functions. These findings provide new insights into the bradycardia found in RTHα patients.


Assuntos
Bradicardia/genética , Cardiopatias Congênitas/genética , Mutação , Receptores alfa dos Hormônios Tireóideos/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Bradicardia/metabolismo , Bradicardia/patologia , Bradicardia/fisiopatologia , Predisposição Genética para Doença , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/patologia , Cardiopatias Congênitas/fisiopatologia , Contração Miocárdica , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Receptores alfa dos Hormônios Tireóideos/metabolismo , Receptores beta dos Hormônios Tireóideos/genética , Receptores beta dos Hormônios Tireóideos/metabolismo , Função Ventricular , Peixe-Zebra/anormalidades , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
5.
Pflugers Arch ; 472(12): 1733-1742, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33095298

RESUMO

The hyperpolarization-activated cation current If is a key determinant for cardiac pacemaker activity. It is conducted by subunits of the hyperpolarization-activated cyclic nucleotide-gated (HCN) channel family, of which HCN4 is predominant in mammalian heart. Both loss-of-function and gain-of-function mutations of the HCN4 gene are associated with sinus node dysfunction in humans; however, their functional impact is not fully understood yet. Here, we sought to characterize a HCN4 V759I variant detected in a patient with a family history of sick sinus syndrome. The genomic analysis yielded a mono-allelic HCN4 V759I variant in a 49-year-old woman presenting with a family history of sick sinus syndrome. This HCN4 variant was previously classified as putatively pathogenic because genetically linked to sudden infant death syndrome and malignant epilepsy. However, detailed electrophysiological and cell biological characterization of HCN4 V759I in Xenopus laevis oocytes and embryonic rat cardiomyocytes, respectively, did not reveal any obvious abnormality. Voltage dependence and kinetics of mutant channel activation, modulation of cAMP-gating by the neuronal HCN channel auxiliary subunit PEX5R, and cell surface expression were indistinguishable from wild-type HCN4. In good agreement, the clinically likewise affected mother of the patient does not exhibit the reported HCN4 variance. HCN4 V759I resembles an innocuous genetic HCN channel variant, which is not sufficient to disturb cardiac pacemaking. Once more, our work emphasizes the importance of careful functional interpretation of genetic findings not only in the context of hereditary cardiac arrhythmias.


Assuntos
Bradicardia/genética , Frequência Cardíaca , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Proteínas Musculares/genética , Mutação de Sentido Incorreto , Canais de Potássio/genética , Potenciais de Ação , Animais , Bradicardia/diagnóstico , Bradicardia/fisiopatologia , Células Cultivadas , Feminino , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Pessoa de Meia-Idade , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Canais de Potássio/metabolismo , Transporte Proteico , Ratos , Ratos Wistar , Xenopus
6.
J Toxicol Sci ; 45(9): 549-558, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32879254

RESUMO

Trimethyltin chloride (TMT) is a stabilizer by-product in the process of manufacturing plastic, which is a kind of very strong toxic substance, and has acute, cumulative and chronic toxicity. TMT may cause bradycardia in patients with occupational poisoning, the mechanism of which has not been reported. This study explored the mechanism of TMT resulting in bradycardia of C57BL/6 mice. TMT was administered to mice to measure heart rate, serum succinate dehydrogenase (SDH) level, and myocardial Na+/K+-ATPase activity and expression. The effects of TMT on myocardial apoptosis were observed by changing the expressions of caspase-3, Bax and Bcl-2 in myocardium. It was found that the heart rate and SDH activity in serum of mice gradually decreased with the increase of TMT dose compared with the control group. The activity and the expression of Na+/K+-ATPase in the heart tissue of mice exposed to TMT was measured and gradually decreased with the increase of dose and time. We measured the expression of Bcl-2, Bax, caspase-3 and cleaved caspase-3 in the heart tissues of TMT exposed mice and found that the expressions of Bax, caspase-3 and cleaved caspase-3 increased and the expressions of Bcl-2 decreased in the heart tissues of the TMT-exposed mice at different doses. With the extension of TMT exposure time, the expression of Bax and caspase-3 increased and the expression of Bcl-2 decreased in the heart tissues of TMT exposed mice. Our findings suggest the mechanisms of TMT resulting in bradycardia may be associated with the inhibited activity and decreased content of Na+/K+-ATPase, thus further leading to the changes of Bcl-2, Bax, caspase-3 and cleaved caspase-3 in the mice's ventricular tissues.


Assuntos
Apoptose/efeitos dos fármacos , Bradicardia/etiologia , Miocárdio/metabolismo , Miocárdio/patologia , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Compostos de Trimetilestanho/toxicidade , Animais , Apoptose/genética , Bradicardia/genética , Caspase 3/genética , Caspase 3/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
7.
FASEB J ; 34(9): 11624-11640, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32683751

RESUMO

Cardiac sympathetic innervation is critically involved in the regulation of circulatory dynamics. However, the molecular mechanism for the innervation patterning has remained elusive. Here, we demonstrate that nardilysin (NRDC, Nrdc), an enhancer of ectodomain shedding, regulates cardiac sympathetic innervation. Nardilysin-deficient (Nrdc-/- ) mice show hypoplastic hearts, hypotension, bradycardia, and abnormal sympathetic innervation patterning. While the innervation of left ventricle (LV) of wild-type mice is denser in the subepicardium than in the subendocardium, Nrdc-/- LV lacks such a polarity and is uniformly and more abundantly innervated. At the molecular level, the full-length form of p75 neurotrophin receptor (p75NTR , Ngfr) is increased in Nrdc-/- LV due to the reduced ectodomain shedding of p75NTR . Importantly, the reduction of p75NTR rescued the abnormal innervation phenotype of Nrdc-/- mice. Moreover, sympathetic neuron-specific, but not cardiomyocyte-specific deletion of Nrdc recapitulated the abnormal innervation patterning of Nrdc-/- mice. In conclusion, neuronal nardilysin critically regulates cardiac sympathetic innervation and circulatory dynamics via modulation of p75NTR .


Assuntos
Coração/inervação , Metaloendopeptidases/genética , Receptor de Fator de Crescimento Neural/genética , Sistema Nervoso Simpático/metabolismo , Animais , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Bradicardia/genética , Bradicardia/fisiopatologia , Células Cultivadas , Ecocardiografia , Coração/fisiopatologia , Frequência Cardíaca/genética , Frequência Cardíaca/fisiologia , Metaloendopeptidases/deficiência , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Células PC12 , Ratos , Receptor de Fator de Crescimento Neural/deficiência , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/fisiopatologia
9.
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
10.
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
11.
Mol Med Rep ; 17(5): 7073-7080, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29568937

RESUMO

Sick sinus syndrome (SSS) is a sinus node dysfunction characterized by severe sinus bradycardia. SSS results in insufficient blood supply to the brain, heart, kidneys, and other organs and is associated with the increased risk of sudden cardiac death. Bradyarrhythmia appears in the absence of any associated cardiac pathology and displays a genetic legacy. The present study identified a family with primary manifestation of sinus bradycardia (five individuals) along with early repolarization (four individuals) and atrial fibrillation (one individual). Targeted exome sequencing was used to screen exons and adjacent splice sites of 61 inherited arrhythmia­associated genes, to detect pathogenic genes and variant sites in the proband. Family members were sequenced by Sanger sequencing and protein functions predicted by Polyphen­2 software. A total of three rare variants were identified in the family, including two missense variants in calcium voltage­gated channel subunit alpha1 C (CACNA1C) (gi:193788541, NM_001129843), c.1786G>A (p.V596M) and c.5344G>A (p.A1782T), and one missense variant in titin (TTN) c.49415G>A (p.R16472H) (gi:291045222, NM_003319). The variants p.V596M and p.R16472H were predicted to be deleterious and resulted in alterations in the amino acid type and sequence of the polypeptide chain, which may partially or completely inactivate the encoded protein. The comparison of literature, gene database, and pedigree phenotype analysis suggests that p.V596M or p.R16472H variants are pathogenic. The complex overlapping variants at three loci lead to a more severe phenotype in the proband, and may increase the susceptibility of individuals to atrial fibrillation. The simultaneous occurrence of V596M and R16472H may increase the severity of early repolarization. Various family members may have carried heterozygous mutants of p.A1782T and p.R16472H due to genetic heterogeneity, however did not exhibit clinical signs of cardiac electrophysiological alterations, potentially attributable to the low vagal tone. To the best of the author's knowledge, this is the first study to suggest the involvement of the novel missense CACNA1C c.1786G>A and TTN c.49415G>A variants in the inheritance of symptomatic bradycardia and development of SSS.


Assuntos
Canais de Cálcio Tipo L/genética , Conectina/genética , Mutação de Sentido Incorreto , Mutação Puntual , Síndrome do Nó Sinusal/genética , Adulto , Idoso , Sequência de Aminoácidos , Bradicardia/genética , Canais de Cálcio Tipo L/química , Conectina/química , Feminino , Predisposição Genética para Doença , Testes Genéticos , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Alinhamento de Sequência
13.
Intern Med ; 55(3): 259-62, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26831020

RESUMO

Congenital long QT syndrome (LQTS) is an important cause of sudden cardiac death in young people without any other structural disease. Mutations in the genes encoding the cardiac ion channels or associated proteins have been shown to result in ion channel dysfunction and thereby causing LQTS. We investigated a Japanese family with LQTS for four generations, with the female family members showing severe symptoms. We performed genetic tests for LQTS-related genes and identified a heterozygous KCNH2 mutation (p.K638del). In the family, the KCNH2 mutation had a very high multigenerational inheritance, and female genotype positives showed more severe phenotypes.


Assuntos
Povo Asiático/genética , Morte Súbita Cardíaca/etiologia , Canais de Potássio Éter-A-Go-Go/genética , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/genética , Mutação/genética , Bradicardia/etiologia , Bradicardia/genética , Bradicardia/fisiopatologia , Análise Mutacional de DNA , Proteínas de Ligação a DNA , Canal de Potássio ERG1 , Eletrocardiografia , Feminino , Testes Genéticos , Genótipo , Humanos , Síndrome do QT Longo/complicações , Linhagem , Síncope/etiologia , Síncope/genética , Síncope/fisiopatologia
14.
Presse Med ; 44(11): 1103-12, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26585273

RESUMO

Since 2012, eight different abnormalities have been described in the THRA gene (encoding the TRα1 thyroid hormone receptor) of 14 patients from 9 families. These mutations induce a clinical phenotype (resistance to thyroid hormone type α) associating symptoms of untreated mild congenital hypothyroidism and a near-normal range of free and total thyroid hormones and TSH (the T4/T3 ratio is nevertheless usually low). The phenotype can diversely include short stature (due to growth retardation), dysmorphic syndrome (face and limb extremities), psychoneuromotor disorders, constipation and bradycardia. The identified genetic abnormalities are located within the ligand-binding domain and result in defective T3 binding, an abnormally strong interaction with corepressors and a dominant negative activity against still functional receptors. The identification of patients with consistent phenotypes and the underlying mutations are warranted to better delineate the spectrum of the syndromes of reduced sensitivity to thyroid hormone.


Assuntos
Receptores alfa dos Hormônios Tireóideos/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Anormalidades Múltiplas/genética , Adolescente , Adulto , Idade de Início , Bradicardia/genética , Criança , Códon sem Sentido , Nanismo/genética , Feminino , Mutação da Fase de Leitura , Genes Dominantes , Genótipo , Humanos , Hiperlipoproteinemia Tipo II/genética , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Fenótipo , Mutação Puntual , Transtornos Psicomotores/genética , Receptores alfa dos Hormônios Tireóideos/deficiência , Síndrome da Resistência aos Hormônios Tireóideos/patologia , Hormônios Tireóideos/sangue , Tireotropina/sangue , Tri-Iodotironina/metabolismo , Adulto Jovem
15.
J Mol Cell Cardiol ; 83: 88-100, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25668431

RESUMO

Sick sinus syndrome remains a highly relevant clinical entity, being responsible for the implantation of the majority of electronic pacemakers worldwide. It is an infinitely more complex disease than it was believed when first described in the mid part of the 20th century. It not only involves the innate leading pacemaker region of the heart, the sinoatrial node, but also the atrial myocardium, predisposing to atrial tachydysrhythmias. It remains controversial as to whether the dysfunction of the sinoatrial node directly causes the dysfunction of the atrial myocardium, or vice versa, or indeed whether these two aspects of the condition arise through some related underlying pathological mechanism, such as extracellular matrix remodeling, i.e., fibrosis. This review aims to shed new light on the myriad possible contributing factors in the development of sick sinus syndrome, with a particular focus on the sinoatrial nodal myocyte. This article is part of a Special Issue entitled CV Aging.


Assuntos
Envelhecimento/metabolismo , Fibrilação Atrial/metabolismo , Bradicardia/metabolismo , Átrios do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Nó Sinoatrial/metabolismo , Idoso , Envelhecimento/patologia , Animais , Fibrilação Atrial/genética , Fibrilação Atrial/patologia , Bradicardia/genética , Bradicardia/patologia , Conexinas/genética , Conexinas/metabolismo , Regulação da Expressão Gênica , Átrios do Coração/patologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Transporte de Íons , MicroRNAs/genética , MicroRNAs/metabolismo , Miócitos Cardíacos/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Sistema Renina-Angiotensina/genética , Nó Sinoatrial/patologia
16.
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
17.
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
18.
Hum Mol Genet ; 19(20): 3895-905, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20639395

RESUMO

Proximal spinal muscular atrophy (SMA) is a debilitating neurological disease marked by isolated lower motor neuron death and subsequent atrophy of skeletal muscle. Historically, SMA pathology was thought to be limited to lower motor neurons and the skeletal muscles they control, yet there are several reports describing the coincidence of cardiovascular abnormalities in SMA patients. As new therapies for SMA emerge, it is necessary to determine whether these non-neuromuscular systems need to be targeted. Therefore, we have characterized left ventricular (LV) function of SMA mice (SMN2+/+; SMNΔ7+/+; Smn-/-) and compared it with that of their unaffected littermates at 7 and 14 days of age. Anatomical and physiological measurements made by electrocardiogram and echocardiography show that affected mouse pups have a dramatic decrease in cardiac function. At 14 days of age, SMA mice have bradycardia and develop a marked dilated cardiomyopathy with a concomitant decrease in contractility. Signs of decreased cardiac function are also apparent as early as 7 days of age in SMA animals. Delivery of a survival motor neuron-1 transgene using a self-complementary adeno-associated virus serotype 9 abolished the symptom of bradycardia and significantly decreased the severity of the heart defect. We conclude that severe SMA animals have compromised cardiac function resulting at least partially from early bradycardia, which is likely attributable to aberrant autonomic signaling. Further cardiographic studies of human SMA patients are needed to clarify the clinical relevance of these findings from this SMA mouse.


Assuntos
Bradicardia , Dependovirus/genética , Técnicas de Transferência de Genes , Insuficiência Cardíaca/fisiopatologia , Atrofia Muscular Espinal/fisiopatologia , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Animais , Bradicardia/genética , Bradicardia/fisiopatologia , Bradicardia/terapia , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Modelos Animais de Doenças , Ecocardiografia , Eletrocardiografia , Terapia Genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/terapia , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/complicações , Contração Miocárdica , Proteínas do Tecido Nervoso , Proteínas do Complexo SMN , Função Ventricular Esquerda
19.
Blood Cells Mol Dis ; 42(3): 211-5, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19272819

RESUMO

The classical function of 4.1R in red blood cells is to contribute to the mechanochemical properties of the membrane by promoting the interaction between spectrin and actin. More recently, it has been recognized that 4.1R is required for the stable cell surface accumulation of a number of erythrocyte membrane proteins. 4.1R is one member of the mammalian 4.1 family - the others being 4.1N, 4.1G and 4.1B - and is expressed in many cell types other than erythrocytes. Recently we have examined the phenotype of hearts from 4.1R knockout mice. Although they had a generally normal morphology, these hearts exhibited bradycardia, and prolongation of both action potentials and QT intervals. Electrophysiological analysis revealed anomalies in a range of ion channel activities. In addition, the immunoreactivity of voltage-gated Na(+) channel NaV1.5 was reduced, indicating a role for 4.1R in the cellular accumulation of this ion channel. 4.1 proteins also have roles in the accumulation of at least two other classes of ion channel. In epithelia, 4.1 interacts with the store-operated channel TRPC4. In neurons, the ligand-gated channels GluR1 and GluR4 require 4.1 proteins for cell surface accumulation. The spectrum of transmembrane proteins that bind to 4.1 proteins overlaps with that of ankyrin. A hypothesis to investigate in the future is that differential regulation of 4.1 and ankyrins (e.g. by PIP(2)) allows highly selective control of cell surface accumulation and transport activity of a specific range of ion channels.


Assuntos
Proteínas Sanguíneas/fisiologia , Proteínas do Citoesqueleto/fisiologia , Canais Iônicos/fisiologia , Proteínas de Membrana/fisiologia , Animais , Arritmias Cardíacas/genética , Proteínas Sanguíneas/química , Proteínas Sanguíneas/deficiência , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Bradicardia/genética , Bradicardia/fisiopatologia , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Eliptocitose Hereditária/sangue , Eliptocitose Hereditária/genética , Eliptocitose Hereditária/fisiopatologia , Células Epiteliais/metabolismo , Eritrócitos/metabolismo , Coração/fisiopatologia , Humanos , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Proteínas dos Microfilamentos , Família Multigênica , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Neurônios/metabolismo , Estrutura Terciária de Proteína , Canais de Sódio/metabolismo
20.
Rev Esp Anestesiol Reanim ; 54(9): 566-9, 2007 Nov.
Artigo em Espanhol | MEDLINE | ID: mdl-18085111

RESUMO

We describe the perioperative treatment of a patient diagnosed with Angelman syndrome, which is usually caused by a deletion in chromosome 15. The patient showed the characteristic signs of psychomotor retardation, epilepsy, lack of speech, frequent laughter and happy demeanor, light skin, blue eyes, and blond hair, hyperactivity, and sleep disturbance. He was scheduled for multiple tooth extractions under general anesthesia. Intravenous anesthesia was induced using ketamine, propofol, and rocuronium, and was maintained with low concentrations of sevoflurane. There were no incidents during or after surgery. The chromosomal abnormality that causes Angelman syndrome is located on the same genes that control the production of gamma-aminobutyric acid-A receptors, which are activated by most intravenous and inhaled anesthetic agents. The effect of the condition on the response of these agents is unknown. The combined use of propofol, ketamine, and sevoflurane at low doses provided adequate anesthesia for this patient. Other characteristics of the syndrome that may affect the use of anesthesia in these patients are discussed.


Assuntos
Anestesia Intravenosa , Síndrome de Angelman , Extração Dentária , Adolescente , Androstanóis/administração & dosagem , Androstanóis/farmacologia , Anestesia por Inalação , Síndrome de Angelman/complicações , Síndrome de Angelman/genética , Síndrome de Angelman/patologia , Bradicardia/genética , Bloqueio de Ramo/genética , Bupivacaína , Contraindicações , Agonistas GABAérgicos , Agonistas de Receptores de GABA-A , Humanos , Ketamina/administração & dosagem , Masculino , Éteres Metílicos/administração & dosagem , Fármacos Neuromusculares não Despolarizantes/administração & dosagem , Fármacos Neuromusculares não Despolarizantes/farmacologia , Propofol/administração & dosagem , Receptores de GABA-A/genética , Rocurônio , Sevoflurano
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA