Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 144(17): 1409-1428, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34694888

RESUMEN

BACKGROUND: Complex molecular programs in specific cell lineages govern human heart development. Hypoplastic left heart syndrome (HLHS) is the most common and severe manifestation within the spectrum of left ventricular outflow tract obstruction defects occurring in association with ventricular hypoplasia. The pathogenesis of HLHS is unknown, but hemodynamic disturbances are assumed to play a prominent role. METHODS: To identify perturbations in gene programs controlling ventricular muscle lineage development in HLHS, we performed whole-exome sequencing of 87 HLHS parent-offspring trios, nuclear transcriptomics of cardiomyocytes from ventricles of 4 patients with HLHS and 15 controls at different stages of heart development, single cell RNA sequencing, and 3D modeling in induced pluripotent stem cells from 3 patients with HLHS and 3 controls. RESULTS: Gene set enrichment and protein network analyses of damaging de novo mutations and dysregulated genes from ventricles of patients with HLHS suggested alterations in specific gene programs and cellular processes critical during fetal ventricular cardiogenesis, including cell cycle and cardiomyocyte maturation. Single-cell and 3D modeling with induced pluripotent stem cells demonstrated intrinsic defects in the cell cycle/unfolded protein response/autophagy hub resulting in disrupted differentiation of early cardiac progenitor lineages leading to defective cardiomyocyte subtype differentiation/maturation in HLHS. Premature cell cycle exit of ventricular cardiomyocytes from patients with HLHS prevented normal tissue responses to developmental signals for growth, leading to multinucleation/polyploidy, accumulation of DNA damage, and exacerbated apoptosis, all potential drivers of left ventricular hypoplasia in absence of hemodynamic cues. CONCLUSIONS: Our results highlight that despite genetic heterogeneity in HLHS, many mutations converge on sequential cellular processes primarily driving cardiac myogenesis, suggesting novel therapeutic approaches.


Asunto(s)
Síndrome del Corazón Izquierdo Hipoplásico/genética , Organogénesis/genética , Heterogeneidad Genética , Humanos
2.
Pediatr Res ; 84(5): 733-742, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30188508

RESUMEN

BACKGROUND: Left ventricular noncompaction (LVNC) is a primary cardiomyopathy with heterogeneous genetic origins. The aim of this study was to elucidate the role of sarcomere gene variants in the pathogenesis and prognosis of LVNC. METHODS AND RESULTS: We screened 82 Japanese patients (0-35 years old), with a diagnosis of LVNC, for mutations in seven genes encoding sarcomere proteins, by direct DNA sequencing. We identified variants in a significant proportion of cases (27%), which were associated with poor prognosis (p = 0.012), particularly variants in TPM1, TNNC1, and ACTC1 (p = 0.012). To elucidate the pathological role, we developed and studied human-induced pluripotent stem cells (hiPSCs) from a patient carrying a TPM1 p.Arg178His mutation, who underwent heart transplantation. These cells displayed pathological changes, with mislocalization of tropomyosin 1, causing disruption of the sarcomere structure in cardiomyocytes, and impaired calcium handling. Microarray analysis indicated that the TPM1 mutation resulted in the down-regulation of the expression of numerous genes involved in heart development, and positive regulation of cellular process, especially the calcium signaling pathway. CONCLUSIONS: Sarcomere genes are implicated as genetic triggers in the development of LVNC, regulating the expression of numerous genes involved in heart development, or modifying the severity of disease.


Asunto(s)
Ventrículos Cardíacos/patología , Sarcómeros/genética , Adolescente , Adulto , Pueblo Asiatico/genética , Señalización del Calcio , Niño , Preescolar , Femenino , Ventrículos Cardíacos/metabolismo , Humanos , Lactante , Recién Nacido , Japón , Masculino , Mutación , Pronóstico , Sarcómeros/metabolismo , Adulto Joven
3.
Circ J ; 82(10): 2609-2618, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30122738

RESUMEN

BACKGROUND: Left ventricular non-compaction (LVNC) is a cardiomyopathy morphologically characterized by 2-layered myocardium and numerous prominent trabeculations, and is often associated with dilated cardiomyopathy (DCM). Variants in the gene encoding tafazzin (TAZ) may change mitochondrial function and cause dysfunction of many organs, but they also contribute to the DCM phenotype in LVNC, and the clinical and echocardiographic features of children with this phenotype are poorly understood. Methods and Results: We enrolled 92 DCM phenotype LVNC patients and performed next-generation sequencing to identify the genetic etiology. Ten TAZ variants were identified in 15 male patients (16.3%) of the 92 patients, including 3 novel missense substitutions. The patients with TAZ variants had a higher frequency of early onset of disease (92.3% vs. 62.3%, P=0.0182), positive family history (73.3% vs. 20.8%, P=0.0001), and higher LV posterior wall thickness Z-score (8.55±2.60 vs. 5.81±2.56, P=0.0103) than those without TAZ variants, although the mortality of both groups was similar. CONCLUSIONS: This study provides new insight into the impact of DCM phenotype LVNC and emphasizes the clinical advantages available for LVNC patients with TAZ variants.


Asunto(s)
Cardiomiopatía Dilatada/genética , No Compactación Aislada del Miocardio Ventricular/genética , Factores de Transcripción/genética , Aciltransferasas , Edad de Inicio , Cardiomiopatía Dilatada/diagnóstico por imagen , Ecocardiografía , Femenino , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , No Compactación Aislada del Miocardio Ventricular/diagnóstico por imagen , Masculino , Anamnesis , Fenotipo
4.
Cell Physiol Biochem ; 42(5): 2021-2029, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28803248

RESUMEN

BACKGROUND/AIMS: Congenital Sick Sinus Syndrome (SSS) is a disorder associated with sudden cardiac death due to severe bradycardia and prolonged pauses. Mutations in HCN4, the gene encoding inward Na+/K+ current (If), have been described as a cause of congenital SSS. The objective of this study is to develop an SSS model in embryonic zebrafish, and use zebrafish as a moderate-throughput assay to functionally characterize HCN4 variants. METHODS: To determine the function of hcn4 in zebrafish, embryos were either bathed in the If -specific blocker (ZD-7288), or endogenous hcn4 expression was knocked down using splice-blocking morpholinos. To assess whether the zebrafish model discriminates benign from pathogenic variants, we tested four HCN4 mutations known to cause human SSS and four variants of unknown significance (VUS). RESULTS: Pharmacological blockade and knockdown of hcn4 in zebrafish phenocopied human SSS, displaying bradycardia and cardiac pauses in intact embryos and explanted hearts. The zebrafish assay correctly identified all disease-causing variants. Of the VUS, the assay predicted 2 as benign and 2 as hypomorphic variants. CONCLUSIONS: We conclude that our embryonic zebrafish assay is a novel and effective tool to functionally characterize human HCN4 variants, which can be translated into important clinical prognostic information.


Asunto(s)
Variación Genética , Síndrome del Seno Enfermo/patología , Animales , Animales Modificados Genéticamente , Bradicardia/etiología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Genotipo , Corazón/efectos de los fármacos , Corazón/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Hibridación in Situ , Morfolinos/metabolismo , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutación , Técnicas de Placa-Clamp , Fenotipo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Pirimidinas/farmacología , Síndrome del Seno Enfermo/genética , Pez Cebra/metabolismo
5.
Am J Med Genet A ; 173(8): 2176-2188, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28653806

RESUMEN

Congenital left-sided cardiac lesions (LSLs) are a significant contributor to the mortality and morbidity of congenital heart disease (CHD). Structural copy number variants (CNVs) have been implicated in LSL without extra-cardiac features; however, non-penetrance and variable expressivity have created uncertainty over the use of CNV analyses in such patients. High-density SNP microarray genotyping data were used to infer large, likely-pathogenic, autosomal CNVs in a cohort of 1,139 probands with LSL and their families. CNVs were molecularly confirmed and the medical records of individual carriers reviewed. The gene content of novel CNVs was then compared with public CNV data from CHD patients. Large CNVs (>1 MB) were observed in 33 probands (∼3%). Six of these were de novo and 14 were not observed in the only available parent sample. Associated cardiac phenotypes spanned a broad spectrum without clear predilection. Candidate CNVs were largely non-recurrent, associated with heterozygous loss of copy number, and overlapped known CHD genomic regions. Novel CNV regions were enriched for cardiac development genes, including seven that have not been previously associated with human CHD. CNV analysis can be a clinically useful and molecularly informative tool in LSLs without obvious extra-cardiac defects, and may identify a clinically relevant genomic disorder in a small but important proportion of these individuals.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Cardiopatías Congénitas/genética , Corazón/fisiopatología , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Genómica , Genotipo , Cardiopatías Congénitas/fisiopatología , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Adulto Joven
6.
Pediatr Res ; 80(3): 425-32, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27089500

RESUMEN

BACKGROUND: Kawasaki disease (KD) is the most common systemic vasculitis syndrome primarily affecting medium-sized arteries, particularly the coronary arteries. Though KD may be associated with immunological problems, the involvement of microRNAs (miRs) has not been fully described. METHODS: We enrolled 23 KD patients and 12 controls. We performed miR and mRNA microarray analysis of peripheral blood mononuclear cells (PBMCs) isolated from acute KD patients and controls. Continuously, we measured specific miRs, mRNA and the expression of proteins by using reverse-transcriptase PCR (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). RESULTS: We identified strikingly high levels of miR-182 and miR-296-5p during the acute febrile phase, and of miR-93, miR-145-5p, miR-145-3p, and miR-150-3p in the defervescence stage, especially in refractory KD patients. The expression of vascular endothelial growth factor A (VEGFA) mRNA, previously reported to be controlled by miR-93, was significantly elevated during the febrile phase and normalized upon treatment, negatively correlating with the expression of miR-93. Further, plasma levels of VEGF-A correlated with PBMC VEGFA mRNA expression. CONCLUSION: Several miRs are highly specific to the acute phase of KD, and may participate in regulating the expression of genes in pathways associated with KD. In particular, miR-93 may participate in regulating expression of VEGF-A and contribute to the pathogenesis of arteritis in acute KD.


Asunto(s)
Leucocitos Mononucleares/metabolismo , MicroARNs/sangre , Síndrome Mucocutáneo Linfonodular/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Arteritis/patología , Niño , Preescolar , Femenino , Fiebre , Regulación de la Expresión Génica , Humanos , Lactante , Masculino , Transducción de Señal
7.
Am J Med Genet A ; 167A(12): 2975-84, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26284702

RESUMEN

Wolff-Parkinson-White (WPW) syndrome is a common cause of supraventricular tachycardia that carries a risk of sudden cardiac death. To date, mutations in only one gene, PRKAG2, which encodes the 5'-AMP-activated protein kinase subunit γ-2, have been identified as causative for WPW. DNA samples from five members of a family with WPW were analyzed by exome sequencing. We applied recently designed prioritization strategies (VAAST/pedigree VAAST) coupled with an ontology-based algorithm (Phevor) that reduced the number of potentially damaging variants to 10: a variant in KCNE2 previously associated with Long QT syndrome was also identified. Of these 11 variants, only MYH6 p.E1885K segregated with the WPW phenotype in all affected individuals and was absent in 10 unaffected family members. This variant was predicted to be damaging by in silico methods and is not present in the 1,000 genome and NHLBI exome sequencing project databases. Screening of a replication cohort of 47 unrelated WPW patients did not identify other likely causative variants in PRKAG2 or MYH6. MYH6 variants have been identified in patients with atrial septal defects, cardiomyopathies, and sick sinus syndrome. Our data highlight the pleiotropic nature of phenotypes associated with defects in this gene.


Asunto(s)
Exoma , Síndrome de Wolff-Parkinson-White/genética , Proteínas Quinasas Activadas por AMP/genética , Adulto , Miosinas Cardíacas/genética , Femenino , Sitios Genéticos , Humanos , Masculino , Cadenas Pesadas de Miosina/genética , Linaje , Canales de Potasio con Entrada de Voltaje/genética , Síndrome de Wolff-Parkinson-White/etiología
8.
Am J Med Genet A ; 158A(12): 3137-47, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23165927

RESUMEN

Congenital diaphragmatic hernia (CDH) is a developmental defect of the diaphragm that causes high newborn mortality. Isolated or non-syndromic CDH is considered a multifactorial disease, with strong evidence implicating genetic factors. As low heritability has been reported in isolated CDH, family-based genetic methods have yet to identify the genetic factors associated with the defect. Using the Utah Population Database, we identified distantly related patients from several extended families with a high incidence of isolated CDH. Using high-density genotyping, seven patients were analyzed by homozygosity exclusion rare allele mapping (HERAM) and phased haplotype sharing (HapShare), two methods we developed to map shared chromosome regions. Our patient cohort shared three regions not previously associated with CDH, that is, 2q11.2-q12.1, 4p13 and 7q11.2, and two regions previously involved in CDH, that is, 8p23.1 and 15q26.2. The latter regions contain GATA4 and NR2F2, two genes implicated in diaphragm formation in mice. Interestingly, three patients shared the 8p23.1 locus and one of them also harbored the 15q26.2 segment. No coding variants were identified in GATA4 or NR2F2, but a rare shared variant was found in intron 1 of GATA4. This work shows the role of heritability in isolated CDH. Our family-based strategy uncovers new chromosomal regions possibly associated with disease, and suggests that non-coding variants of GATA4 and NR2F2 may contribute to the development of isolated CDH. This approach could speed up the discovery of the genes and regulatory elements causing multifactorial diseases, such as isolated CDH.


Asunto(s)
Cromosomas Humanos , Hernias Diafragmáticas Congénitas , Adulto , Factor de Transcripción COUP II/genética , Estudios de Casos y Controles , Niño , Estudios de Cohortes , ADN/sangre , ADN/genética , Diafragma/anomalías , Salud de la Familia , Femenino , Factor de Transcripción GATA4/genética , Dosificación de Gen , Predisposición Genética a la Enfermedad , Genotipo , Hernia Diafragmática/sangre , Hernia Diafragmática/genética , Humanos , Masculino , Linaje , Polimorfismo de Nucleótido Simple
9.
Birth Defects Res A Clin Mol Teratol ; 94(9): 738-42, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22821744

RESUMEN

BACKGROUND: The AEBP1 (adipocyte enhancer binding protein) gene has two isoforms: AEBP1, the shorter of the two isoforms, and Aclp (aortic carboxypeptidase-like protein). Aclp(-/-) mice demonstrate a ventral wall defect that is similar to gastroschisis in humans. Aclp is a potential candidate gene because it is expressed in numerous tissues during early development in mice; it associates with the extracellular matrix; and is essential for abdominal wall development and wound healing. In contrast, AEBP1 encodes an intracellular protein involved in proinflammatory responses, and may play a critical role in apoptosis and cell survival. Gastroschisis is a severe abdominal wall defect more common in young women and recently associated with a genitourinary infection early in pregnancy. METHODS: We screened AEBP1 in 40 cases of gastroschisis and compared identified variants in a control population. RESULTS: We identified several novel variants in AEBP1, including synonymous and nonsynonymous single nucleotide substitutions and intronic indels. However, the frequency of these variants was not significantly different from that of the control group, and the associated amino acid changes were predicted to be benign by two prediction software programs. CONCLUSIONS: Gastroschisis remains an intriguing defect that, for an unknown reason, occurs more commonly in young women and after a genitourinary infection. Although we found many alterations in AEBP1 among the gastroschisis cases, they were predicted to be benign. However, this gene requires further understanding of its interaction with other genes involved in the immune response pathway.


Asunto(s)
Carboxipeptidasas/genética , Gastrosquisis/genética , Complicaciones del Embarazo/genética , Proteínas Represoras/genética , Infecciones Urinarias/genética , Adulto , Alelos , Exones , Femenino , Gastrosquisis/complicaciones , Gastrosquisis/epidemiología , Frecuencia de los Genes , Heterocigoto , Homocigoto , Humanos , Lactante , Intrones , Polimorfismo de Nucleótido Simple , Embarazo , Complicaciones del Embarazo/epidemiología , Estudios Prospectivos , Infecciones Urinarias/complicaciones , Infecciones Urinarias/epidemiología , Utah/epidemiología , Población Blanca
10.
Sci Adv ; 8(46): eabq3925, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36383648

RESUMEN

Direct links between carbonaceous chondrites and their parent bodies in the solar system are rare. The Winchcombe meteorite is the most accurately recorded carbonaceous chondrite fall. Its pre-atmospheric orbit and cosmic-ray exposure age confirm that it arrived on Earth shortly after ejection from a primitive asteroid. Recovered only hours after falling, the composition of the Winchcombe meteorite is largely unmodified by the terrestrial environment. It contains abundant hydrated silicates formed during fluid-rock reactions, and carbon- and nitrogen-bearing organic matter including soluble protein amino acids. The near-pristine hydrogen isotopic composition of the Winchcombe meteorite is comparable to the terrestrial hydrosphere, providing further evidence that volatile-rich carbonaceous asteroids played an important role in the origin of Earth's water.

11.
J Biol Chem ; 285(30): 23208-23, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20472559

RESUMEN

The innate antiviral response is mediated, at least in part, by Toll-like receptors (TLRs). TLR3 signaling is activated in response to viral infection, and the absence of TLR3 in mice significantly increases mortality after infection with enteroviruses that cause myocarditis and/or dilated cardiomyopathy. We screened TLR3 in patients diagnosed with enteroviral myocarditis/cardiomyopathy and identified a rare variant in one patient as well as a significantly increased occurrence of a common polymorphism compared with controls. Expression of either variant resulted in significantly reduced TLR3-mediated signaling after stimulation with synthetic double-stranded RNA. Furthermore, Coxsackievirus B3 infection of cell lines expressing mutated TLR3 abrogated activation of the type I interferon pathway, leading to increased viral replication. TLR3-mediated type I interferon signaling required cellular autophagy and was suppressed by 3-methyladenine and bafilomycin A1, by inhibitors of lysosomal proteolysis, and by reduced expression of Beclin 1, Atg5, or microtubule-associated protein 1 light chain 3beta (MAP1LC3beta). However, TLR3-mediated signaling was restored upon exogenous expression of Beclin 1 or a variant MAP1LC3beta fusion protein refractory to RNA interference. These data suggest that individuals harboring these variants may have a blunted innate immune response to enteroviral infection, leading to reduced viral clearance and an increased risk of cardiac pathology.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/virología , Enterovirus/fisiología , Mutación , Miocarditis/genética , Miocarditis/virología , Receptor Toll-Like 3/genética , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Secuencia de Bases , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Línea Celular , Cloroquina/farmacología , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Análisis Mutacional de ADN , Endosomas/metabolismo , Femenino , Humanos , Interferones/metabolismo , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Miocarditis/metabolismo , Miocarditis/patología , Fenotipo , Transporte de Proteínas , Receptor Toll-Like 3/química , Receptor Toll-Like 3/metabolismo , Replicación Viral
12.
Basic Res Cardiol ; 106(6): 1159-71, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21956162

RESUMEN

TIR-domain-containing adaptor-inducing interferon-ß (TRIF) plays a major role in Toll-like receptor 3 (TLR3) mediated signaling. Mice deficient in TLR3 and TRIF have been shown to be highly susceptible to enterovirus-induced myocardial injury. These mice have decreased production of antiviral cytokines and increased viral replication in the heart. Therefore, we hypothesized that conditional overexpression of TRIF would change cardiac myocyte susceptibility to virus infection by augmenting the antiviral response. We generated double-transgenic MHC-tTA/MHC(tetO)-TRIF mice (DT), with conditional cardiac-specific overexpression of TRIF. Naive DT mice had increased cardiac expression of antiviral cytokines and increased cellular infiltration but no alterations in cardiac function. DT mice were less susceptible to encephalomyocarditis virus (EMCV) infection and had a significantly lower viral load in the heart when compared to littermate (LM) and MHC(tetO)-TRIF (ST) mice. Histopathological examination showed that the severity of myocarditis was also attenuated in DT mice. Furthermore, the decreased virus titers in the DT mouse hearts led to less cardiac damage and better cardiac function when compared to LM and ST mice. Administration of doxycycline to DT mice suppressed the protective effects of TRIF overexpression in the heart. The findings of the present study establish the importance of cardiac-specific TRIF-mediated signaling in the heart in acute viral myocarditis and identify potentially important targets for diagnostic and therapeutic strategies.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/inmunología , Miocarditis/genética , Miocarditis/virología , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Western Blotting , Citocinas/biosíntesis , Ecocardiografía , Virus de la Encefalomiocarditis , Ensayo de Inmunoadsorción Enzimática , Femenino , Corazón , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Miocarditis/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/inmunología , Carga Viral
13.
Mol Genet Metab ; 102(2): 200-6, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20965760

RESUMEN

Left ventricular noncompaction (LVNC) is a cardiomyopathy morphologically characterized by 2-layered myocardium, numerous prominent trabeculations, and deep intertrabecular recesses communicating with the left ventricular cavity. The purpose of this study was to investigate patients with LVNC for possible disease causing mutations. We screened 4 genes (TAZ, LDB3, DTNA and TPM1) in 51 patients with LVNC for mutations by polymerase chain reaction and direct DNA sequencing. A novel missense substitution in exon 1 of TPM1 (c.109A>G: p.Lys37Glu) was identified in three affected members of a family with isolated LVNC. The substitution brings about a change in amino acid charge at a highly conserved residue and could result in aberrant mRNA splicing. This variant was not identified in 200 normal control samples. Pathologic analysis of a right ventricular myocardial specimen from the proband's maternal aunt revealed endocardial and subendocardial fibrosis with prominent elastin deposition, as well as the presence of adipose tissue between muscle layers, pathologic changes that are distinct from those seen in patients with HCM or DCM. Screening of the proband and her mother for variants in other sarcomeric protein-encoding candidate genes, MYH7, MYBPC3, TNNT2, TNNI3, ACTC, MYL2, and MYL3, did not identify any other non-synonymous variants or variants in splice donor-acceptor sequences that were potentially disease causing. We conclude TPM1 is a potential candidate disease-causing gene for isolated LVNC, especially in patients experiencing sudden death.


Asunto(s)
Muerte Súbita Cardíaca , Ventrículos Cardíacos/patología , No Compactación Aislada del Miocardio Ventricular/genética , Mutación , Tropomiosina/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Pueblo Asiatico/genética , Niño , Proteínas Asociadas a la Distrofina/genética , Electrocardiografía , Femenino , Genotipo , Ventrículos Cardíacos/diagnóstico por imagen , Humanos , No Compactación Aislada del Miocardio Ventricular/patología , Proteínas con Dominio LIM , Masculino , Persona de Mediana Edad , Neuropéptidos/genética , Linaje , Polimorfismo de Nucleótido Simple , Ultrasonografía , Adulto Joven
14.
Am J Med Genet A ; 155A(10): 2578-83, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21910241

RESUMEN

Timothy syndrome type 1 (TS-1) is a rare disorder that affects multiple organ systems and has a high incidence of sudden death due to profound QT prolongation and resultant ventricular arrhythmias. All previously described cases of TS-1 are the result of a missense mutation in exon 8A (p.G406R), an alternatively spliced variant of the L-type calcium channel gene (Ca(v)1.2, CACNA1C). Most patients reported in the literature represent highly affected individuals who present early in life with severe cardiac and neurological manifestations. Here, we describe somatic mosaicism in TS-1 patients with less severe manifestations than the typical TS-1 patient. These findings suggest that the TS prognosis may not be as dismal as previously reported. Moreover, our findings have implications for genetic counseling in that previously described de novo TS mutations may represent cases of parental mosaicism and warrant careful genotyping of parental tissue other than peripheral blood lymphocytes.


Asunto(s)
Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/patología , Mosaicismo , Fenotipo , Sindactilia/genética , Sindactilia/patología , Adolescente , Trastorno Autístico , Secuencia de Bases , Canales de Calcio Tipo L/genética , Electrocardiografía , Exones/genética , Femenino , Genotipo , Humanos , Recién Nacido , Recien Nacido Prematuro , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
15.
Int J Cardiol ; 328: 122-129, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33309763

RESUMEN

BACKGROUND: Left ventricular noncompaction (LVNC) is a hereditary cardiomyopathy, associated with high morbidity and mortality, but the role of genetics in cases of fetal-onset has not been fully evaluated. The goal of this study was to identify the genetic background in LVNC fetal-onset patients using next-generation sequencing (NGS). METHODS: Thirty-three fetal-onset Japanese probands with LVNC (20 males and 13 females) were enrolled. In the enrolled patients, 81 genes associated with cardiomyopathy were screened using next-generation sequencing (NGS) retrospectively. RESULTS: Twenty-three patients had congestive heart failure (CHF), and six patients had arrhythmias. Prominent trabeculations were mostly observed in lateral LV, posterior LV, and apex of LV in patients with LVNC. Twelve died; three patients experienced intrauterine death or termination of pregnancy. Overall, 15 variants were found among eight genes in 16 patients. Seven variants were detected in MYH7 and two in TPM1. Sarcomere gene variants accounted for 75.0%. A multivariable proportional hazards model revealed that CHF at diagnosis and a higher ratio of the noncompacted layer/compacted layer in the LV posterior wall were independent risk factors for death in LVNC fetal-onset patients (odds ratio = 4.26 × 106 and 1.36 × 108, p = 0.0075 and 0.0005, respectively). CONCLUSIONS: The present study is the first report focusing on genetic background combined with clinical features in LVNC fetal-onset patients using NGS. Sarcomere variants were most commonly identified in fetal-onset patients, and greater attention should be paid to fetal-onset patients with LVNC having prominent trabeculations in the LV because they are more likely to develop CHF.


Asunto(s)
Cardiopatías Congénitas , No Compactación Aislada del Miocardio Ventricular , Femenino , Humanos , No Compactación Aislada del Miocardio Ventricular/diagnóstico por imagen , No Compactación Aislada del Miocardio Ventricular/genética , Masculino , Embarazo , Estudios Retrospectivos , Sarcómeros/genética , Función Ventricular Izquierda
16.
Nat Commun ; 12(1): 6442, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34750360

RESUMEN

The genetic architecture of atrial fibrillation (AF) encompasses low impact, common genetic variants and high impact, rare variants. Here, we characterize a high impact AF-susceptibility allele, KCNQ1 R231H, and describe its transcontinental geographic distribution and history. Induced pluripotent stem cell-derived cardiomyocytes procured from risk allele carriers exhibit abbreviated action potential duration, consistent with a gain-of-function effect. Using identity-by-descent (IBD) networks, we estimate the broad- and fine-scale population ancestry of risk allele carriers and their relatives. Analysis of ancestral migration routes reveals ancestors who inhabited Denmark in the 1700s, migrated to the Northeastern United States in the early 1800s, and traveled across the Midwest to arrive in Utah in the late 1800s. IBD/coalescent-based allele dating analysis reveals a relatively recent origin of the AF risk allele (~5000 years). Thus, our approach broadens the scope of study for disease susceptibility alleles to the context of human migration and ancestral origins.


Asunto(s)
Fibrilación Atrial/genética , Predisposición Genética a la Enfermedad/genética , Canal de Potasio KCNQ1/genética , Mutación Missense , Polimorfismo de Nucleótido Simple , Potenciales de Acción , Alelos , Dinamarca , Emigrantes e Inmigrantes , Femenino , Genotipo , Geografía , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Linaje , Factores de Riesgo , Utah
17.
Mol Genet Metab ; 100(2): 198-203, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20303308

RESUMEN

TAZ (G4.5) was initially identified as the gene associated with Barth syndrome and left ventricular noncompaction (LVNC). The purpose of this study was to investigate patients with LVNC for disease-causing mutations in TAZ. In 124 Japanese patients, including 50 families, mutation analysis of TAZ was performed using DNA sequencing. A splice donor mutation was identified in two brothers with Barth syndrome and LVNC, and a sister who was asymptomatic. However, the variant was not identified in either parent or the maternal grandparents, all of whom were asymptomatic. Due to the recurrent inheritance of this variant by each of the children we concluded that this was evidence of gonadal mosaicism in the obligate carrier mother, the first reported occurrence of this in Barth syndrome.


Asunto(s)
Síndrome de Barth/genética , No Compactación Aislada del Miocardio Ventricular/genética , Mosaicismo , Factores de Transcripción/genética , Aciltransferasas , Pueblo Asiatico/genética , Resultado Fatal , Femenino , Disgenesia Gonadal/genética , Humanos , Lactante , Masculino , Mutación , Linaje
18.
Am J Cardiol ; 102(5): 629-31, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18721526

RESUMEN

Mutations in the genes encoding transforming growth factor-beta receptor types I and II (TGFBR1 and TGFBR2, respectively) are commonly identified in patients with Loeys-Dietz syndrome, as well as some patients with Marfan's syndrome or familial thoracic aortic aneurysms and dissections. This suggests that there is considerable phenotypic heterogeneity associated with mutations in these genes. Because bicuspid aortic valve (BAV) is a congenital heart defect in patients with Loeys-Dietz syndrome, this study was conducted to investigate whether variants in TGFBR1 or TGFBR2 are responsible for sporadic BAV. Analysis of these genes in 35 patients with BAVs identified only known single-nucleotide polymorphisms or novel synonymous or intronic substitutions. In conclusion, mutations in TGFBR1 and TGFBR2 rarely cause sporadic BAV.


Asunto(s)
Aorta Torácica , Válvula Aórtica/anomalías , ADN/genética , Cardiopatías Congénitas/genética , Mutación , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Adolescente , Adulto , Niño , Preescolar , Dilatación Patológica/etiología , Femenino , Humanos , Lactante , Masculino , Reacción en Cadena de la Polimerasa , Pronóstico , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Factor de Crecimiento Transformador beta
19.
Mol Genet Metab ; 93(4): 468-74, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18368697

RESUMEN

Left ventricular noncompaction (LVNC) is a genetically heterogenous disorder. Mutations in the human cardiac sodium channel alpha-subunit gene (SCN5A) are involved in the pathophysiology of cardiac arrhythmias and cardiomyopathies. This study was performed to compare the frequency of SCN5A variants in LVNC patients with or without arrhythmias, and to investigate the relationship between variants and disease severity. DNA was isolated from the peripheral blood of 62 Japanese probands with LVNC, comprising 17 familial cases and 45 sporadic cases. Blood samples were screened for variants in SCN5A using single-strand conformational polymorphism analysis (SSCP) and DNA sequencing. Seven variants, rs6599230:G > A, c.453C > T, c.1141-3C > A, rs1805124:A > G (p.H558R), rs1805125:C > T (p.P1090L), c.3996C > T, and rs1805126:T > C were identified in 7 familial and 12 sporadic cases. The frequency of SCN5A variants was significantly higher in the patients with arrhythmias than those without (50% vs 7%: P = 0.0003), suggesting these variants represent a risk factor for arrhythmia and supporting the hypothesis that genes encoding ion channels are involved in LVNC pathophysiology. The LVNC patients with heart failure also had high occurrence of SCN5A variants, suggesting the presence of SCN5A variants and/or arrhythmias increase the severity of LVNC.


Asunto(s)
Arritmias Cardíacas/genética , Hipertrofia Ventricular Izquierda/genética , Proteínas Musculares/genética , Canales de Sodio/genética , Adolescente , Adulto , Anciano , Pueblo Asiatico/genética , Niño , Preescolar , Femenino , Insuficiencia Cardíaca/genética , Humanos , Lactante , Recién Nacido , Japón , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5
20.
Circ Res ; 99(6): 646-55, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16917092

RESUMEN

Arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C) is characterized by progressive degeneration of the right ventricular myocardium, ventricular arrhythmias, fibrous-fatty replacement, and increased risk of sudden death. Mutations in 6 genes, including 4 encoding desmosomal proteins (Junctional plakoglobin (JUP), Desmoplakin (DSP), Plakophilin 2, and Desmoglein 2), have been identified in patients with ARVD/C. Mutation analysis of 66 probands identified 4 variants in DSP; V30M, Q90R, W233X, and R2834H. To establish a cause and effect relationship between those DSP missense mutations and ARVD/C, we performed in vitro and in vivo analyses of the mutated proteins. Unlike wild-type (WT) DSP, the N-terminal mutants (V30M and Q90R) failed to localize to the cell membrane in desomosome-forming cell line and failed to bind to and coimmunoprecipitate JUP. Multiple attempts to generate N-terminal DSP (V30M and Q90R) cardiac-specific transgenes have failed: analysis of embryos revealed evidence of profound ventricular dilation, which likely resulted in embryonic lethality. We were able to develop transgenic (Tg) mice with cardiac-restricted overexpression of the C-terminal mutant (R2834H) or WT DSP. Whereas mice overexpressing WT DSP had no detectable histologic, morphological, or functional cardiac changes, the R2834H-Tg mice had increased cardiomyocyte apoptosis, cardiac fibrosis, and lipid accumulation, along with ventricular enlargement and cardiac dysfunction in both ventricles. These mice also displayed interruption of DSP-desmin interaction at intercalated discs (IDs) and marked ultra-structural changes of IDs. These data suggest DSP expression in cardiomyocytes is crucial for maintaining cardiac tissue integrity, and DSP abnormalities result in ARVD/C by cardiomyocyte death, changes in lipid metabolism, and defects in cardiac development.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/etiología , Cardiomiopatías/etiología , Desmoplaquinas/genética , Desmosomas/fisiología , Mutación Missense , Miocitos Cardíacos/ultraestructura , Animales , Apoptosis , Comunicación Celular/genética , Comunicación Celular/fisiología , Línea Celular Tumoral , Análisis Mutacional de ADN , Desmosomas/genética , Embrión de Mamíferos , Corazón/crecimiento & desarrollo , Humanos , Uniones Intercelulares/patología , Metabolismo de los Lípidos , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA