Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Elife ; 122023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37404133

RESUMO

Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease (CHD) with a likely oligogenic etiology, but our understanding of the genetic complexities and pathogenic mechanisms leading to HLHS is limited. We performed whole genome sequencing (WGS) on 183 HLHS patient-parent trios to identify candidate genes, which were functionally tested in the Drosophila heart model. Bioinformatic analysis of WGS data from an index family of a HLHS proband born to consanguineous parents prioritized 9 candidate genes with rare, predicted damaging homozygous variants. Of them, cardiac-specific knockdown (KD) of mitochondrial MICOS complex subunit dCHCHD3/6 resulted in drastically compromised heart contractility, diminished levels of sarcomeric actin and myosin, reduced cardiac ATP levels, and mitochondrial fission-fusion defects. These defects were similar to those inflicted by cardiac KD of ATP synthase subunits of the electron transport chain (ETC), consistent with the MICOS complex's role in maintaining cristae morphology and ETC assembly. Five additional HLHS probands harbored rare, predicted damaging variants in CHCHD3 or CHCHD6. Hypothesizing an oligogenic basis for HLHS, we tested 60 additional prioritized candidate genes from these patients for genetic interactions with CHCHD3/6 in sensitized fly hearts. Moderate KD of CHCHD3/6 in combination with Cdk12 (activator of RNA polymerase II), RNF149 (goliath, E3 ubiquitin ligase), or SPTBN1 (ß-Spectrin, scaffolding protein) caused synergistic heart defects, suggesting the likely involvement of diverse pathways in HLHS. Further elucidation of novel candidate genes and genetic interactions of potentially disease-contributing pathways is expected to lead to a better understanding of HLHS and other CHDs.


Assuntos
Cardiopatias Congênitas , Síndrome do Coração Esquerdo Hipoplásico , Humanos , Síndrome do Coração Esquerdo Hipoplásico/genética , Actomiosina , Biologia Computacional , Trifosfato de Adenosina , Proteínas Mitocondriais
2.
Ann Thorac Surg ; 113(3): 890-895, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-33539782

RESUMO

BACKGROUND: The objective of this study is to assess the safety and early impact of intramyocardial delivery of autologous bone marrow-derived mononuclear cells (BM-MNC) at time of surgical Ebstein repair. METHODS: Patients with Ebstein anomaly (ages 6 months to 30 years) scheduled to undergo repair of the tricuspid valve were eligible to participate in this open-label, non-randomized phase I clinical trial. BM-MNC target dose was 1-3 million cells/kg. Ten patients have undergone surgical intervention and cell delivery to the right ventricle (RV) and completed 6-month follow-up. RESULTS: All patients underwent surgical tricuspid valve repair and uneventful BM-MNC delivery; there were no ventricular arrhythmias and no adverse events related to study product or delivery. Echocardiographic RV myocardial performance index improved and RV fractional area change showed an initial decline and then through study follow-up. There was no evidence of delayed myocardial enhancement or regional wall motion abnormalities at injection sites on 6-month follow-up magnetic resonance imaging. CONCLUSIONS: Intramyocardial delivery of BM-MNC after surgical repair in Ebstein anomaly can be performed safely. Echocardiography variables suggest a positive impact of cell delivery on the RV myocardium with improvements in both RV size and wall motion over time. Additional follow-up and comparison to control groups are required to better characterize the impact of cell therapy on the myopathic RV in Ebstein anomaly.


Assuntos
Anomalia de Ebstein , Anomalia de Ebstein/diagnóstico , Anomalia de Ebstein/cirurgia , Ecocardiografia , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/cirurgia , Humanos , Resultado do Tratamento , Valva Tricúspide/anormalidades , Valva Tricúspide/diagnóstico por imagem , Valva Tricúspide/cirurgia
3.
Int J Mol Sci ; 22(11)2021 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-34071043

RESUMO

A de novo missense variant in Rag GTPase protein C (RagCS75Y) was recently identified in a syndromic dilated cardiomyopathy (DCM) patient. However, its pathogenicity and the related therapeutic strategy remain unclear. We generated a zebrafish RragcS56Y (corresponding to human RagCS75Y) knock-in (KI) line via TALEN technology. The KI fish manifested cardiomyopathy-like phenotypes and poor survival. Overexpression of RagCS75Y via adenovirus infection also led to increased cell size and fetal gene reprogramming in neonatal rat ventricle cardiomyocytes (NRVCMs), indicating a conserved mechanism. Further characterization identified aberrant mammalian target of rapamycin complex 1 (mTORC1) and transcription factor EB (TFEB) signaling, as well as metabolic abnormalities including dysregulated autophagy. However, mTOR inhibition failed to ameliorate cardiac phenotypes in the RagCS75Y cardiomyopathy models, concomitant with a failure to promote TFEB nuclear translocation. This observation was at least partially explained by increased and mTOR-independent physical interaction between RagCS75Y and TFEB in the cytosol. Importantly, TFEB overexpression resulted in more nuclear TFEB and rescued cardiomyopathy phenotypes. These findings suggest that S75Y is a pathogenic gain-of-function mutation in RagC that leads to cardiomyopathy. A primary pathological step of RagCS75Y cardiomyopathy is defective mTOR-TFEB signaling, which can be corrected by TFEB overexpression, but not mTOR inhibition.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Cardiomiopatia Dilatada/genética , Mutação com Ganho de Função , Proteínas Monoméricas de Ligação ao GTP/genética , Mutação de Sentido Incorreto , Mutação Puntual , Serina-Treonina Quinases TOR/antagonistas & inibidores , Transporte Ativo do Núcleo Celular , Substituição de Aminoácidos , Animais , Autofagia , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/biossíntese , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Cardiomiopatia Dilatada/terapia , Células Cultivadas , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Ventrículos do Coração/citologia , Humanos , Camundongos , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Miócitos Cardíacos/metabolismo , Fenótipo , Ratos Wistar , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
4.
Hum Mol Genet ; 28(23): 3954-3969, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31625562

RESUMO

Genetics is a significant factor contributing to congenital heart disease (CHD), but our understanding of the genetic players and networks involved in CHD pathogenesis is limited. Here, we searched for de novo copy number variations (CNVs) in a cohort of 167 CHD patients to identify DNA segments containing potential pathogenic genes. Our search focused on new candidate disease genes within 19 deleted de novo CNVs, which did not cover known CHD genes. For this study, we developed an integrated high-throughput phenotypical platform to probe for defects in cardiogenesis and cardiac output in human induced pluripotent stem cell (iPSC)-derived multipotent cardiac progenitor (MCPs) cells and, in parallel, in the Drosophila in vivo heart model. Notably, knockdown (KD) in MCPs of RPL13, a ribosomal gene and SON, an RNA splicing cofactor, reduced proliferation and differentiation of cardiomyocytes, while increasing fibroblasts. In the fly, heart-specific RpL13 KD, predominantly at embryonic stages, resulted in a striking 'no heart' phenotype. KD of Son and Pdss2, among others, caused structural and functional defects, including reduced or abolished contractility, respectively. In summary, using a combination of human genetics and cardiac model systems, we identified new genes as candidates for causing human CHD, with particular emphasis on ribosomal genes, such as RPL13. This powerful, novel approach of combining cardiac phenotyping in human MCPs and in the in vivo Drosophila heart at high throughput will allow for testing large numbers of CHD candidates, based on patient genomic data, and for building upon existing genetic networks involved in heart development and disease.


Assuntos
Variações do Número de Cópias de DNA , Cardiopatias Congênitas/genética , Miocárdio/citologia , Proteínas de Neoplasias/genética , Proteínas Ribossômicas/genética , Animais , Células Cultivadas , Estudos de Coortes , Modelos Animais de Doenças , Drosophila , Feminino , Redes Reguladoras de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/química , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/química , Miócitos Cardíacos/citologia , Miócitos Cardíacos/patologia , Estudos Retrospectivos
5.
Pediatr Rev ; 40(7): 344-353, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31263042

RESUMO

Hypoplastic left heart syndrome is one of the most complex congenital heart diseases and requires several cardiac surgeries for survival. The diagnosis is usually established prenatally or shortly after birth. Each stage of surgery poses a unique hemodynamic situation that requires deeper understanding to manage common pediatric problems such as dehydration and respiratory infections. Careful multidisciplinary involvement in the care of these complex patients is improving their outcome; however, morbidity and mortality are still substantial. In this review, we focus on the hemodynamic aspects of various surgical stages that a primary care provider should know to manage these challenging patients.


Assuntos
Procedimentos Cirúrgicos Cardíacos/métodos , Síndrome do Coração Esquerdo Hipoplásico , Desidratação/etiologia , Deficiências do Desenvolvimento/etiologia , Técnica de Fontan , Humanos , Síndrome do Coração Esquerdo Hipoplásico/complicações , Síndrome do Coração Esquerdo Hipoplásico/diagnóstico , Síndrome do Coração Esquerdo Hipoplásico/fisiopatologia , Síndrome do Coração Esquerdo Hipoplásico/cirurgia , Lactente , Cuidados Paliativos/métodos , Atenção Primária à Saúde , Infecções Respiratórias/etiologia
6.
J Am Coll Cardiol ; 73(1): 58-66, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30621952

RESUMO

BACKGROUND: Spontaneous coronary artery dissection (SCAD) is an increasingly recognized cause of acute coronary syndromes (ACS) afflicting predominantly younger to middle-aged women. Observational studies have reported a high prevalence of extracoronary vascular anomalies, especially fibromuscular dysplasia (FMD) and a low prevalence of coincidental cases of atherosclerosis. PHACTR1/EDN1 is a genetic risk locus for several vascular diseases, including FMD and coronary artery disease, with the putative causal noncoding variant at the rs9349379 locus acting as a potential enhancer for the endothelin-1 (EDN1) gene. OBJECTIVES: This study sought to test the association between the rs9349379 genotype and SCAD. METHODS: Results from case control studies from France, United Kingdom, United States, and Australia were analyzed to test the association with SCAD risk, including age at first event, pregnancy-associated SCAD (P-SCAD), and recurrent SCAD. RESULTS: The previously reported risk allele for FMD (rs9349379-A) was associated with a higher risk of SCAD in all studies. In a meta-analysis of 1,055 SCAD patients and 7,190 controls, the odds ratio (OR) was 1.67 (95% confidence interval [CI]: 1.50 to 1.86) per copy of rs9349379-A. In a subset of 491 SCAD patients, the OR estimate was found to be higher for the association with SCAD in patients without FMD (OR: 1.89; 95% CI: 1.53 to 2.33) than in SCAD cases with FMD (OR: 1.60; 95% CI: 1.28 to 1.99). There was no effect of genotype on age at first event, P-SCAD, or recurrence. CONCLUSIONS: The first genetic risk factor for SCAD was identified in the largest study conducted to date for this condition. This genetic link may contribute to the clinical overlap between SCAD and FMD.


Assuntos
Anomalias dos Vasos Coronários/epidemiologia , Anomalias dos Vasos Coronários/genética , Endotelina-1/genética , Displasia Fibromuscular/complicações , Loci Gênicos/genética , Proteínas dos Microfilamentos/genética , Doenças Vasculares/congênito , Adulto , Idoso , Austrália , Estudos de Casos e Controles , Anomalias dos Vasos Coronários/complicações , Feminino , Displasia Fibromuscular/genética , França , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , Reino Unido , Estados Unidos , Doenças Vasculares/complicações , Doenças Vasculares/epidemiologia , Doenças Vasculares/genética
7.
Genet Epidemiol ; 41(4): 297-308, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28211093

RESUMO

Next-generation sequencing technologies have afforded unprecedented characterization of low-frequency and rare genetic variation. Due to low power for single-variant testing, aggregative methods are commonly used to combine observed rare variation within a single gene. Causal variation may also aggregate across multiple genes within relevant biomolecular pathways. Kernel-machine regression and adaptive testing methods for aggregative rare-variant association testing have been demonstrated to be powerful approaches for pathway-level analysis, although these methods tend to be computationally intensive at high-variant dimensionality and require access to complete data. An additional analytical issue in scans of large pathway definition sets is multiple testing correction. Gene set definitions may exhibit substantial genic overlap, and the impact of the resultant correlation in test statistics on Type I error rate control for large agnostic gene set scans has not been fully explored. Herein, we first outline a statistical strategy for aggregative rare-variant analysis using component gene-level linear kernel score test summary statistics as well as derive simple estimators of the effective number of tests for family-wise error rate control. We then conduct extensive simulation studies to characterize the behavior of our approach relative to direct application of kernel and adaptive methods under a variety of conditions. We also apply our method to two case-control studies, respectively, evaluating rare variation in hereditary prostate cancer and schizophrenia. Finally, we provide open-source R code for public use to facilitate easy application of our methods to existing rare-variant analysis results.


Assuntos
Algoritmos , Estudos de Associação Genética/métodos , Variação Genética , Simulação por Computador , Humanos , Modelos Genéticos , Tamanho da Amostra , Estatísticas não Paramétricas
9.
Genet Epidemiol ; 40(6): 461-9, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27312771

RESUMO

Rare variants (RVs) have been shown to be significant contributors to complex disease risk. By definition, these variants have very low minor allele frequencies and traditional single-marker methods for statistical analysis are underpowered for typical sequencing study sample sizes. Multimarker burden-type approaches attempt to identify aggregation of RVs across case-control status by analyzing relatively small partitions of the genome, such as genes. However, it is generally the case that the aggregative measure would be a mixture of causal and neutral variants, and these omnibus tests do not directly provide any indication of which RVs may be driving a given association. Recently, Bayesian variable selection approaches have been proposed to identify RV associations from a large set of RVs under consideration. Although these approaches have been shown to be powerful at detecting associations at the RV level, there are often computational limitations on the total quantity of RVs under consideration and compromises are necessary for large-scale application. Here, we propose a computationally efficient alternative formulation of this method using a probit regression approach specifically capable of simultaneously analyzing hundreds to thousands of RVs. We evaluate our approach to detect causal variation on simulated data and examine sensitivity and specificity in instances of high RV dimensionality as well as apply it to pathway-level RV analysis results from a prostate cancer (PC) risk case-control sequencing study. Finally, we discuss potential extensions and future directions of this work.


Assuntos
Modelos Genéticos , Teorema de Bayes , Estudos de Casos e Controles , Variação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Análise de Sequência de DNA
10.
Heart ; 102(11): 876-81, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26864667

RESUMO

OBJECTIVE: Spontaneous coronary artery dissection (SCAD) is an under-recognised but important cause of myocardial infarction and sudden cardiac death. We sought to determine the role of medical and molecular genetic screening for connective tissue disorders in patients with SCAD. METHODS: We performed a single-centre retrospective descriptive analysis of patients with spontaneous coronary artery disease who had undergone medical genetics evaluation 1984-2014 (n=116). The presence or absence of traits suggestive of heritable connective tissue disease was extracted. Genetic testing for connective tissue disorders and/or aortopathies, if performed, is also reported. RESULTS: Of the 116 patients (mean age 44.2 years, 94.8% women and 41.4% with non-coronary fibromuscular dysplasia (FMD)), 59 patients underwent genetic testing, of whom 3 (5.1%) received a diagnosis of connective tissue disorder: a 50-year-old man with Marfan syndrome; a 43-year-old woman with vascular Ehlers-Danlos syndrome and FMD; and a 45-year-old woman with vascular Ehlers-Danlos syndrome. An additional 12 patients (20.3%) had variants of unknown significance, none of which was thought to be a definite disease-causing mutation based on in silico analyses. CONCLUSIONS: Only a minority of patients with SCAD who undergo genetic evaluation have a likely pathogenic mutation identified on gene panel testing. Even fewer exhibit clinical features of connective tissue disorder. These findings underscore the need for further studies to elucidate the molecular mechanisms of SCAD.


Assuntos
Doenças do Tecido Conjuntivo/genética , Anomalias dos Vasos Coronários/genética , Mutação , Doenças Vasculares/congênito , Adulto , Doenças do Tecido Conjuntivo/complicações , Doenças do Tecido Conjuntivo/diagnóstico , Anomalias dos Vasos Coronários/diagnóstico por imagem , Síndrome de Ehlers-Danlos/diagnóstico , Síndrome de Ehlers-Danlos/genética , Feminino , Displasia Fibromuscular/diagnóstico , Displasia Fibromuscular/genética , Marcadores Genéticos , Predisposição Genética para Doença , Testes Genéticos , Hereditariedade , Humanos , Masculino , Síndrome de Marfan/diagnóstico , Síndrome de Marfan/genética , Pessoa de Meia-Idade , Minnesota , Fenótipo , Valor Preditivo dos Testes , Sistema de Registros , Estudos Retrospectivos , Fatores de Risco , Doenças Vasculares/diagnóstico por imagem , Doenças Vasculares/genética
11.
Hum Mol Genet ; 25(2): 254-65, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26604136

RESUMO

Dilated cardiomyopathy (DCM) is a leading cause of heart failure. In families with autosomal-dominant DCM, heterozygous missense mutations were identified in RNA-binding motif protein 20 (RBM20), a spliceosome protein induced during early cardiogenesis. Dermal fibroblasts from two unrelated patients harboring an RBM20 R636S missense mutation were reprogrammed to human induced pluripotent stem cells (hiPSCs) and differentiated to beating cardiomyocytes (CMs). Stage-specific transcriptome profiling identified differentially expressed genes ranging from angiogenesis regulator to embryonic heart transcription factor as initial molecular aberrations. Furthermore, gene expression analysis for RBM20-dependent splice variants affected sarcomeric (TTN and LDB3) and calcium (Ca(2+)) handling (CAMK2D and CACNA1C) genes. Indeed, RBM20 hiPSC-CMs exhibited increased sarcomeric length (RBM20: 1.747 ± 0.238 µm versus control: 1.404 ± 0.194 µm; P < 0.0001) and decreased sarcomeric width (RBM20: 0.791 ± 0.609 µm versus control: 0.943 ± 0.166 µm; P < 0.0001). Additionally, CMs showed defective Ca(2+) handling machinery with prolonged Ca(2+) levels in the cytoplasm as measured by greater area under the curve (RBM20: 814.718 ± 94.343 AU versus control: 206.941 ± 22.417 AU; P < 0.05) and higher Ca(2+) spike amplitude (RBM20: 35.281 ± 4.060 AU versus control:18.484 ± 1.518 AU; P < 0.05). ß-adrenergic stress induced with 10 µm norepinephrine demonstrated increased susceptibility to sarcomeric disorganization (RBM20: 86 ± 10.5% versus control: 40 ± 7%; P < 0.001). This study features the first hiPSC model of RBM20 familial DCM. By monitoring human cardiac disease according to stage-specific cardiogenesis, this study demonstrates RBM20 familial DCM is a developmental disorder initiated by molecular defects that pattern maladaptive cellular mechanisms of pathological cardiac remodeling. Indeed, hiPSC-CMs recapitulate RBM20 familial DCM phenotype in a dish and establish a tool to dissect disease-relevant defects in RBM20 splicing as a global regulator of heart function.


Assuntos
Cardiomiopatia Dilatada/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas de Ligação a RNA/genética , Adulto , Animais , Cálcio/metabolismo , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/fisiopatologia , Diferenciação Celular , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Masculino , Camundongos , Modelos Biológicos , Mutação de Sentido Incorreto , Linhagem , Splicing de RNA/genética , Transcriptoma , Adulto Jovem
12.
JAMA Intern Med ; 175(5): 821-6, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25798899

RESUMO

IMPORTANCE: Spontaneous coronary artery dissection (SCAD) is a major cause of acute coronary syndrome in young women, especially among those without traditional cardiovascular risk factors. Prior efforts to study SCAD have been hampered by underrecognition and lack of registry-based studies. Risk factors and pathogenesis remain largely undefined, and inheritability has not been reported. OBSERVATIONS: Using novel research methods, patient champions, and social media, the Mayo Clinic SCAD Registry has been able to better characterize this condition, which was previously considered rare. Of 412 patient enrollees, we identified 5 familial cases of SCAD comprising affected mother-daughter, identical twin sister, sister, aunt-niece, and first-cousin pairs, implicating both recessive and dominant modes of inheritance. The mother-daughter pair also reported fatal myocardial infarction in 3 maternal relatives. None of the participants had other potential risk factors for SCAD, including connective tissue disorders or peripartum status. CONCLUSIONS AND RELEVANCE: To our knowledge, this series is the first to identify a familial association in SCAD suggesting a genetic predisposition. Recognition of SCAD as a heritable disorder has implications for at-risk family members and furthers our understanding of the pathogenesis of this complex disease. Whole-exome sequencing provides a unique opportunity to identify the molecular underpinnings of SCAD susceptibility.


Assuntos
Angioplastia Coronária com Balão/métodos , Ponte de Artéria Coronária/métodos , Anomalias dos Vasos Coronários , Doenças Vasculares/congênito , Síndrome Coronariana Aguda/etiologia , Adulto , Angiografia Coronária/métodos , Anomalias dos Vasos Coronários/sangue , Anomalias dos Vasos Coronários/complicações , Anomalias dos Vasos Coronários/diagnóstico , Anomalias dos Vasos Coronários/fisiopatologia , Anomalias dos Vasos Coronários/terapia , Eletrocardiografia , Família , Feminino , Predisposição Genética para Doença , Humanos , Pessoa de Meia-Idade , Infarto do Miocárdio/etiologia , Linhagem , Fatores de Risco , Troponina/sangue , Doenças Vasculares/sangue , Doenças Vasculares/complicações , Doenças Vasculares/diagnóstico , Doenças Vasculares/fisiopatologia , Doenças Vasculares/terapia
13.
Am J Med Genet A ; 167A(4): 886-90, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25706677

RESUMO

Idiopathic dilated cardiomyopathy is a heritable, genetically heterogeneous disorder characterized by progressive heart failure. Dilated cardiomyopathy typically exhibits autosomal dominant inheritance, yet frequently remains clinically silent until adulthood. We sought to discover the molecular basis of idiopathic, non-syndromic dilated cardiomyopathy in a one-month-old male presenting with severe heart failure. Previous comprehensive testing of blood, urine, and skin biopsy specimen was negative for metabolic, mitochondrial, storage, and infectious etiologies. Ophthalmologic examination was normal. Chromosomal microarray and commercial dilated cardiomyopathy gene panel testing failed to identify a causative mutation. Parental screening echocardiograms revealed no evidence of clinically silent dilated cardiomyopathy. Whole exome sequencing was carried out on the family trio on a research basis, filtering for rare, deleterious, recessive and de novo genetic variants. Pathogenic compound heterozygous truncating mutations were identified in ALMS1, diagnostic of Alström syndrome and prompting disclosure of genetic findings. Alström syndrome is a known cause for dilated cardiomyopathy in children yet delayed and mis-diagnosis are common owing to its rarity and age-dependent emergence of multisystem clinical manifestations. At six months of age the patient ultimately developed bilateral nystagmus and hyperopia, features characteristic of the syndrome. Early diagnosis is guiding clinical monitoring of other organ systems and allowing for presymptomatic intervention. Furthermore, recognition of recessive inheritance as the mechanism for sporadic disease has informed family planning. This case highlights a limitation of standard gene testing panels for pediatric dilated cardiomyopathy and exemplifies the potential for whole exome sequencing to solve a diagnostic dilemma and enable personalized care.


Assuntos
Síndrome de Alstrom/diagnóstico , Cardiomiopatia Dilatada/diagnóstico , Síndrome de Alstrom/genética , Cardiomiopatia Dilatada/genética , Proteínas de Ciclo Celular , Códon sem Sentido , Análise Mutacional de DNA , Exoma , Humanos , Lactente , Masculino , Técnicas de Diagnóstico Molecular , Proteínas/genética
15.
Hum Mol Genet ; 23(21): 5793-804, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24925317

RESUMO

Locus mapping has uncovered diverse etiologies for familial atrial fibrillation (AF), dilated cardiomyopathy (DCM), and mixed cardiac phenotype syndromes, yet the molecular basis for these disorders remains idiopathic in most cases. Whole-exome sequencing (WES) provides a powerful new tool for familial disease gene discovery. Here, synergistic application of these genomic strategies identified the pathogenic mutation in a familial syndrome of atrial tachyarrhythmia, conduction system disease (CSD), and DCM vulnerability. Seven members of a three-generation family exhibited the variably expressed phenotype, three of whom manifested CSD and clinically significant arrhythmia in childhood. Genome-wide linkage analysis mapped two equally plausible loci to chromosomes 1p3 and 13q12. Variants from WES of two affected cousins were filtered for rare, predicted-deleterious, positional variants, revealing an unreported heterozygous missense mutation disrupting the highly conserved kinase domain in TNNI3K. The G526D substitution in troponin I interacting kinase, with the most deleterious SIFT and Polyphen2 scores possible, resulted in abnormal peptide aggregation in vitro and in silico docking models predicted altered yet energetically favorable wild-type mutant dimerization. Ventricular tissue from a mutation carrier displayed histopathological hallmarks of DCM and reduced TNNI3K protein staining with unique amorphous nuclear and sarcoplasmic inclusions. In conclusion, mutation of TNNI3K, encoding a heart-specific kinase previously shown to modulate cardiac conduction and myocardial function in mice, underlies a familial syndrome of electrical and myopathic heart disease. The identified substitution causes a TNNI3K aggregation defect and protein deficiency, implicating a dominant-negative loss of function disease mechanism.


Assuntos
Arritmias Cardíacas/genética , Cardiomiopatia Dilatada/genética , Estudos de Associação Genética , Sistema de Condução Cardíaco/anormalidades , MAP Quinase Quinase Quinases/genética , Mutação , Taquicardia Atrial Ectópica/genética , Adulto , Sequência de Aminoácidos , Arritmias Cardíacas/diagnóstico , Síndrome de Brugada , Doença do Sistema de Condução Cardíaco , Cardiomiopatia Dilatada/diagnóstico , Cardiomiopatia Dilatada/metabolismo , Criança , Mapeamento Cromossômico , Cromossomos Humanos Par 1 , Sequência Conservada , Exoma , Feminino , Loci Gênicos , Variação Genética , Haplótipos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/ultraestrutura , Compostos Orgânicos , Linhagem , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases , Alinhamento de Sequência , Síndrome , Taquicardia Atrial Ectópica/diagnóstico
16.
Hum Mol Genet ; 23(14): 3779-91, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24584570

RESUMO

Dilated cardiomyopathy (DCM) due to mutations in RBM20, a gene encoding an RNA-binding protein, is associated with high familial penetrance, risk of progressive heart failure and sudden death. Although genetic investigations and physiological models have established the linkage of RBM20 with early-onset DCM, the underlying basis of cellular and molecular dysfunction is undetermined. Modeling human genetics using a high-throughput pluripotent stem cell platform was herein designed to pinpoint the initial transcriptome dysfunction and mechanistic corruption in disease pathogenesis. Tnnt2-pGreenZeo pluripotent stem cells were engineered to knockdown Rbm20 (shRbm20) to determine the cardiac-pathogenic phenotype during cardiac differentiation. Intracellular Ca(2+) transients revealed Rbm20-dependent alteration in Ca(2+) handling, coinciding with known pathological splice variants of Titin and Camk2d genes by Day 24 of cardiogenesis. Ultrastructural analysis demonstrated elongated and thinner sarcomeres in the absence of Rbm20 that is consistent with human cardiac biopsy samples. Furthermore, Rbm20-depleted transcriptional profiling at Day 12 identified Rbm20-dependent dysregulation with 76% of differentially expressed genes linked to known cardiac pathology ranging from primordial Nkx2.5 to mature cardiac Tnnt2 as the initial molecular aberrations. Notably, downstream consequences of Rbm20-depletion at Day 24 of differentiation demonstrated significant dysregulation of extracellular matrix components such as the anomalous overexpression of the Vtn gene. By using the pluripotent stem cell platform to model human cardiac disease according to a stage-specific cardiogenic roadmap, we established a new paradigm of familial DCM pathogenesis as a developmental disorder that is patterned during early cardiogenesis and propagated with cellular mechanisms of pathological cardiac remodeling.


Assuntos
Cálcio/metabolismo , Cardiomiopatia Dilatada/etiologia , Coração/crescimento & desenvolvimento , Proteínas de Ligação a RNA/metabolismo , Sarcômeros/patologia , Animais , Cardiomiopatia Dilatada/patologia , Diferenciação Celular , Linhagem Celular , Corpos Embrioides/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Modelos Biológicos , Células-Tronco Pluripotentes/metabolismo , Splicing de RNA , Sarcômeros/ultraestrutura
17.
Ann Thorac Surg ; 94(3): 854-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22727249

RESUMO

BACKGROUND: We reviewed our experience with the surgical management of supravalvar aortic stenosis (SVAS) to determine long-term outcomes and factors related to late reoperation. METHODS: Between August 1956 and May 2009, 78 patients (50 males) underwent surgical correction of SVAS. Median age was 10.4 years (range, 16 days to 55.2 years). Mean preoperative gradient was 57.2±21.9 mm Hg with a mean peak gradient of 99.5±34.8 mm Hg. Supravalvar aortic stenosis was discrete in 51 patients (64%) and diffuse in 27 patients (35%). Aortic valve stenosis was present in 22 patients (29%). Williams-Beuren syndrome was present in 32 patients (41%). RESULTS: Surgery was either a diamond-shaped patch in 67 patients (85.9%) or a pantaloons-shaped patch in 11 patients (14.1%). Aortic valve intervention was required in 20 patients (25.64%). Mean gradient immediately after repair was 25±25 mm Hg, with 13 patients (16.7%) having a residual gradient. A high residual gradient was more likely in the diffuse group (odds ratio, 3.73; 95% confidence interval, 1.07 to 12.98). There were 2 (2.6%) early deaths, both with diffuse SVAS. Median follow-up was 19.8 years; maximum was 48.5 years. The mean gradient across the left ventricular outflow tract at late follow-up was 8.8 mm Hg (95% confidence interval, 3.7 to 14.01). Overall survival was estimated at 90%±7%, 84%±9%, and 8%2±10% at 5, 10, and 20 years, respectively. Predictors of mortality were age younger than 2 years (p=0.021), diffuse SVAS (p=0.045), aortic valve stenosis (p=0.032), and high postoperative gradient (p=0.023). Presence of Williams-Beuren syndrome did not affect survival (p=0.305). Freedom from late reoperation was 97%±4%, 93%±7%, and 86%±10% at 5, 10, and 20 years, respectively. Significant aortic valve disease (p<0.001) and diffuse SVAS (p=0.009) were risk factors for late reoperation. CONCLUSIONS: Surgical repair for SVAS can be performed with a single-patch technique with good long-term outcome. Late mortality and need for reoperation are more likely with diffuse SVAS or the presence of aortic valve stenosis.


Assuntos
Estenose Aórtica Supravalvular/mortalidade , Estenose Aórtica Supravalvular/cirurgia , Valva Aórtica/cirurgia , Procedimentos Cirúrgicos Cardíacos/mortalidade , Procedimentos Cirúrgicos Cardíacos/métodos , Adolescente , Adulto , Estenose Aórtica Supravalvular/congênito , Estenose Aórtica Supravalvular/diagnóstico por imagem , Anuloplastia da Valva Cardíaca/métodos , Criança , Pré-Escolar , Estudos de Coortes , Intervalos de Confiança , Intervalo Livre de Doença , Ecocardiografia , Feminino , Seguimentos , Humanos , Lactente , Recém-Nascido , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Monitorização Fisiológica , Razão de Chances , Cuidados Pós-Operatórios , Estudos Retrospectivos , Análise de Sobrevida , Técnicas de Sutura , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
18.
Pflugers Arch ; 460(2): 295-306, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20033705

RESUMO

Assembly of an inward rectifier K+ channel pore (Kir6.1/Kir6.2) and an adenosine triphosphate (ATP)-binding regulatory subunit (SUR1/SUR2A/SUR2B) forms ATP-sensitive K+ (KATP) channel heteromultimers, widely distributed in metabolically active tissues throughout the body. KATP channels are metabolism-gated biosensors functioning as molecular rheostats that adjust membrane potential-dependent functions to match cellular energetic demands. Vital in the adaptive response to (patho)physiological stress, KATP channels serve a homeostatic role ranging from glucose regulation to cardioprotection. Accordingly, genetic variation in KATP channel subunits has been linked to the etiology of life-threatening human diseases. In particular, pathogenic mutations in KATP channels have been identified in insulin secretion disorders, namely, congenital hyperinsulinism and neonatal diabetes. Moreover, KATP channel defects underlie the triad of developmental delay, epilepsy, and neonatal diabetes (DEND syndrome). KATP channelopathies implicated in patients with mechanical and/or electrical heart disease include dilated cardiomyopathy (with ventricular arrhythmia; CMD1O) and adrenergic atrial fibrillation. A common Kir6.2 E23K polymorphism has been associated with late-onset diabetes and as a risk factor for maladaptive cardiac remodeling in the community-at-large and abnormal cardiopulmonary exercise stress performance in patients with heart failure. The overall mutation frequency within KATP channel genes and the spectrum of genotype-phenotype relationships remain to be established, while predicting consequences of a deficit in channel function is becoming increasingly feasible through systems biology approaches. Thus, advances in molecular medicine in the emerging field of human KATP channelopathies offer new opportunities for targeted individualized screening, early diagnosis, and tailored therapy.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Trifosfato de Adenosina/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Droga/fisiologia , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Canalopatias/genética , Homeostase/genética , Humanos , Canais KATP , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Receptores de Sulfonilureias
19.
Acta Paediatr Suppl ; 95(452): 17-21, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16801160

RESUMO

UNLABELLED: Dilated cardiomyopathy (DCM) is an idiopathic, genetically heterogeneous disorder characterized by heart failure and arrhythmia. Over the past decade, the molecular basis for DCM has been partially uncovered by discovery of mutation in genes encoding cystoskeletal, sarcomeric, nuclear membrane, and sarcoplasmic reticulum proteins. These findings have implicated pathogenic mechanisms whereby structural integrity, contractile force dynamics, and calcium regulation within the cardiac myocyte are perturbed. Recognition of dilated and hypertrophic cardiomyopathies as allelic disorders has provided the opportunity to identify genotype-phenotype relationships and to gain new insight into pathways leading to cardiac failure and hypertrophy. CONCLUSION: Collectively, family-based studies of DCM provide the rationale for clinical screening in first-degree relatives, regardless of family history or age of the index case.


Assuntos
Cardiomiopatia Dilatada/genética , Actinas/genética , Cardiomiopatia Dilatada/fisiopatologia , Proteínas do Citoesqueleto/genética , Análise Mutacional de DNA , Ligação Genética , Humanos , Mutação de Sentido Incorreto , Contração Miocárdica/genética , Miócitos Cardíacos/fisiologia
20.
J Mol Cell Cardiol ; 38(6): 937-43, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15910878

RESUMO

ATP-sensitive potassium (K(ATP)) channels are evolutionarily conserved plasma-membrane protein complexes, widely represented in tissue beds with high metabolic activity. There, they are formed through physical association of the inwardly rectifying potassium channel pore, most typically Kir6.2, and the regulatory sulfonylurea receptor subunit, an ATP-binding cassette protein. Energetic signals, received via tight integration with cellular metabolic pathways, are processed by the sulfonylurea receptor subunit that in turn gates the nucleotide sensitivity of the channel pore thereby controlling membrane potential dependent cellular functions. Recent findings, elicited from genetic disruption of channel proteins, have established in vivo the requirement of intact K(ATP) channels in the proper function of cardiac muscle under stress. In the heart, where K(ATP) channels were originally discovered, channel ablation compromises cardioprotection under ischemic insult. New data implicate the requirement of intact K(ATP) channels for the cardiac adaptive response to acute stress. K(ATP) channels have been further implicated in the adaptive cardiac response to chronic (patho)physiologic hemodynamic load, with K(ATP) channel deficiency affecting structural remodeling, rendering the heart vulnerable to calcium-dependent maladaptation and predisposing to heart failure. These findings are underscored by the identification in humans that defective K(ATP) channels induced by mutations in ABCC9, the gene encoding the cardiac sulfonylurea receptor subunit, confer susceptibility to dilated cardiomyopathy. Thus, in parallel with the developed understanding of the molecular identity and mode of action of K(ATP) channels since their discovery, there is now an expanded understanding of their critical significance in the cardiac stress response in health and disease.


Assuntos
Miocárdio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Canais de Potássio/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/química , Animais , Animais Geneticamente Modificados , Cálcio/metabolismo , Cardiomiopatias , Cardiomiopatia Dilatada/metabolismo , Membrana Celular/metabolismo , Insuficiência Cardíaca/metabolismo , Humanos , Isquemia , Camundongos , Mutação , Isquemia Miocárdica , Canais de Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ligação Proteica , Receptores de Droga/metabolismo , Receptores de Sulfonilureias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA