Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Eur Heart J Cardiovasc Imaging ; 25(1): 86-94, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37523765

RESUMEN

AIMS: Anterior mitral valve leaflet (AMVL) elongation is detectable in overt and subclinical hypertrophic cardiomyopathy (HCM). We sought to investigate the dynamic motion of the aorto-mitral apparatus to understand the behaviour of the AMVL and the mechanisms of left ventricular outflow tract obstruction (LVOTO) predisposition in HCM. METHODS AND RESULTS: Cardiovascular magnetic resonance imaging using a 1.5 Tesla scanner was performed on 36 HCM sarcomere gene mutation carriers without left ventricular hypertrophy (G+LVH-), 31 HCM patients with preserved ejection fraction carrying a pathogenic sarcomere gene mutation (G+LVH+), and 53 age-, sex-, and body surface area-matched healthy volunteers. Dynamic excursion of the aorto-mitral apparatus was assessed semi-automatically on breath-held three-chamber cine steady-state free precession images. Four pre-defined regions of interest (ROIs) were tracked: ROIPMVL: hinge point of the posterior mitral valve leaflet; ROITRIG: intertrigonal mitral annulus; ROIAMVL: AMVL tip; and ROIAAO: anterior aortic annulus. Compared with controls, normalized two-dimensional displacement-vs.-time plots in G+LVH- revealed subtle but significant systolic anterior motion (SAM) of the AMVL (P < 0.0001) and reduced longitudinal excursion of ROIAAO (P = 0.014) and ROIPMVL (P = 0.048). In overt and subclinical HCM, excursion of the ROITRIG/AMVL/PMVL was positively associated with the burden of left ventricular fibrosis (P < 0.028). As expected, SAM was observed in G+LVH+ together with reduced longitudinal excursion of ROITRIG (P = 0.049) and ROIAAO (P = 0.008). CONCLUSION: Dyskinesia of the aorto-mitral apparatus, including SAM of the elongated AMVL, is detectable in subclinical HCM before the development of LVH or left atrial enlargement. These data have the potential to improve our understanding of early phenotype development and LVOTO predisposition in HCM.


Asunto(s)
Cardiomiopatía Hipertrófica , Obstrucción del Flujo Ventricular Externo , Humanos , Válvula Mitral/patología , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/complicaciones , Hipertrofia Ventricular Izquierda , Imagen por Resonancia Magnética , Fenotipo , Obstrucción del Flujo Ventricular Externo/etiología , Obstrucción del Flujo Ventricular Externo/genética
2.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35055131

RESUMEN

Hypertrophic Cardiomyopathy (HCM) is a common inherited disorder characterized by unexplained left ventricular hypertrophy with or without left ventricular outflow tract (LVOT) obstruction. Single-nuclei RNA-sequencing (snRNA-seq) of both obstructive and nonobstructive HCM patient samples has revealed alterations in communication between various cell types, but no direct and integrated comparison between the two HCM phenotypes has been reported. We performed a bioinformatic analysis of HCM snRNA-seq datasets from obstructive and nonobstructive patient samples to identify differentially expressed genes and distinctive patterns of intercellular communication. Differential gene expression analysis revealed 37 differentially expressed genes, predominantly in cardiomyocytes but also in other cell types, relevant to aging, muscle contraction, cell motility, and the extracellular matrix. Intercellular communication was generally reduced in HCM, affecting the extracellular matrix, growth factor binding, integrin binding, PDGF binding, and SMAD binding, but with increases in adenylate cyclase binding, calcium channel inhibitor activity, and serine-threonine kinase activity in nonobstructive HCM. Increases in neuron to leukocyte and dendritic cell communication, in fibroblast to leukocyte and dendritic cell communication, and in endothelial cell communication to other cell types, largely through changes in the expression of integrin-ß1 and its cognate ligands, were also noted. These findings indicate both common and distinct physiological mechanisms affecting the pathogenesis of obstructive and nonobstructive HCM and provide opportunities for the personalized management of different HCM phenotypes.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Redes Reguladoras de Genes , Hipertrofia Ventricular Izquierda/genética , Análisis de Secuencia de ARN/métodos , Obstrucción del Flujo Ventricular Externo/genética , Comunicación Celular , Biología Computacional , Bases de Datos Genéticas , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Masculino , Análisis de la Célula Individual
3.
Sci Rep ; 11(1): 13163, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162896

RESUMEN

Hypertrophic cardiomyopathy (HCM) is characterized by phenotypic heterogeneity. We investigated the molecular basis of the cardiac phenotype in two mouse models at established disease stage (mouse-HCM), and human myectomy tissue (human-HCM). We analyzed the transcriptome in 2 mouse models with non-obstructive HCM (R403Q-MyHC, R92W-TnT)/littermate-control hearts at 24 weeks of age, and in myectomy tissue of patients with obstructive HCM/control hearts (GSE36961, GSE36946). Additionally, we examined myocyte redox, cardiac mitochondrial DNA copy number (mtDNA-CN), mt-respiration, mt-ROS generation/scavenging and mt-Ca2+ handling in mice. We identified distinct allele-specific gene expression in mouse-HCM, and marked differences between mouse-HCM and human-HCM. Only two genes (CASQ1, GPT1) were similarly dysregulated in both mutant mice and human-HCM. No signaling pathway or transcription factor was predicted to be similarly dysregulated (by Ingenuity Pathway Analysis) in both mutant mice and human-HCM. Losartan was a predicted therapy only in TnT-mutant mice. KEGG pathway analysis revealed enrichment for several metabolic pathways, but only pyruvate metabolism was enriched in both mutant mice and human-HCM. Both mutant mouse myocytes demonstrated evidence of an oxidized redox environment. Mitochondrial complex I RCR was lower in both mutant mice compared to controls. MyHC-mutant mice had similar mtDNA-CN and mt-Ca2+ handling, but TnT-mutant mice exhibited lower mtDNA-CN and impaired mt-Ca2+ handling, compared to littermate-controls. Molecular profiling reveals differences in gene expression, transcriptional regulation, intracellular signaling and mt-number/function in 2 mouse models at established disease stage. Further studies are needed to confirm differences in gene expression between mouse and human-HCM, and to examine whether cardiac phenotype, genotype and/or species differences underlie the divergence in molecular profiles.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Transcriptoma , Animales , Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica/complicaciones , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/metabolismo , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Ecocardiografía , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/metabolismo , Mutación Missense , Miocardio/metabolismo , Cadenas Pesadas de Miosina/genética , Fenotipo , Mutación Puntual , ARN Mensajero/genética , Especificidad de la Especie , Troponina T/genética , Obstrucción del Flujo Ventricular Externo/etiología , Obstrucción del Flujo Ventricular Externo/genética
4.
Circ Heart Fail ; 14(1): e007022, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33430602

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is the most common genetic heart disease. While ≈50% of patients with HCM carry a sarcomere gene mutation (sarcomere mutation-positive, HCMSMP), the genetic background is unknown in the other half of the patients (sarcomere mutation-negative, HCMSMN). Genotype-specific differences have been reported in cardiac function. Moreover, HCMSMN patients have later disease onset and a better prognosis than HCMSMP patients. To define if genotype-specific derailments at the protein level may explain the heterogeneity in disease development, we performed a proteomic analysis in cardiac tissue from a clinically well-phenotyped HCM patient group. METHODS: A proteomics screen was performed in cardiac tissue from 39 HCMSMP patients, 11HCMSMN patients, and 8 nonfailing controls. Patients with HCM had obstructive cardiomyopathy with left ventricular outflow tract obstruction and diastolic dysfunction. A novel MYBPC32373insG mouse model was used to confirm functional relevance of our proteomic findings. RESULTS: In all HCM patient samples, we found lower levels of metabolic pathway proteins and higher levels of extracellular matrix proteins. Levels of total and detyrosinated α-tubulin were markedly higher in HCMSMP than in HCMSMN and controls. Higher tubulin detyrosination was also found in 2 unrelated MYBPC3 mouse models and its inhibition with parthenolide normalized contraction and relaxation time of isolated cardiomyocytes. CONCLUSIONS: Our findings indicate that microtubules and especially its detyrosination contribute to the pathomechanism of patients with HCMSMP. This is of clinical importance since it represents a potential treatment target to improve cardiac function in patients with HCMSMP, whereas a beneficial effect may be limited in patients with HCMSMN.


Asunto(s)
Cardiomiopatía Hipertrófica/metabolismo , Tubulina (Proteína)/metabolismo , Tirosina/metabolismo , Obstrucción del Flujo Ventricular Externo/metabolismo , Adulto , Anciano , Animales , Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Proteínas Portadoras/genética , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Haploinsuficiencia , Humanos , Masculino , Persona de Mediana Edad , Cadenas Pesadas de Miosina/genética , Proteómica , Sarcómeros/genética , Troponina I/genética , Troponina T/genética , Obstrucción del Flujo Ventricular Externo/genética , Obstrucción del Flujo Ventricular Externo/fisiopatología , Tabique Interventricular/metabolismo
5.
Sci Rep ; 10(1): 14167, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843646

RESUMEN

BVES is a transmembrane protein, our previous work demonstrated that single nucleotide mutations of BVES in tetralogy of fallot (TOF) patients cause a downregulation of BVES transcription. However, the relationship between BVES and the pathogenesis of TOF has not been determined. Here we reported our research results about the relationship between BVES and the right ventricular outflow tract (RVOT) stenosis. BVES expression was significantly downregulated in most TOF samples compared with controls. The expression of the second heart field (SHF) regulatory network genes, including NKX2.5, GATA4 and HAND2, was also decreased in the TOF samples. In zebrafish, bves knockdown resulted in looping defects and ventricular outflow tract (VOT) stenosis, which was mostly rescued by injecting bves mRNA. bves knockdown in zebrafish also decreased the expression of SHF genes, such as nkx2.5, gata4 and hand2, consistent with the TOF samples` results. The dual-fluorescence reporter system analysis showed that BVES positively regulated the transcriptional activity of GATA4, NKX2.5 and HAND2 promoters. In zebrafish, nkx2.5 mRNA partially rescued VOT stenosis caused by bves knockdown. These results indicate that BVES downregulation may be associated with RVOT stenosis of non-syndromic TOF, and bves is probably involved in the development of VOT in zebrafish.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Proteínas Musculares/biosíntesis , Tetralogía de Fallot/genética , Obstrucción del Flujo Ventricular Externo/genética , Anomalías Múltiples , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Niño , Preescolar , Anomalías de los Vasos Coronarios , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Foramen Oval Permeable , Regulación de la Expresión Génica , Corazón/embriología , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/fisiología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Proteínas Musculares/genética , Proteínas Musculares/fisiología , ARN Mensajero/genética , Tetralogía de Fallot/complicaciones , Tetralogía de Fallot/metabolismo , Tetralogía de Fallot/patología , Obstrucción del Flujo Ventricular Externo/embriología , Obstrucción del Flujo Ventricular Externo/etiología , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
6.
PLoS Genet ; 15(5): e1007711, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31120883

RESUMEN

Dominant mutations of Gata4, an essential cardiogenic transcription factor (TF), were known to cause outflow tract (OFT) defects in both human and mouse, but the underlying molecular mechanism was not clear. In this study, Gata4 haploinsufficiency in mice was found to result in OFT defects including double outlet right ventricle (DORV) and ventricular septum defects (VSDs). Gata4 was shown to be required for Hedgehog (Hh)-receiving progenitors within the second heart field (SHF) for normal OFT alignment. Restored cell proliferation in the SHF by knocking-down Pten failed to rescue OFT defects, suggesting that additional cell events under Gata4 regulation is important. SHF Hh-receiving cells failed to migrate properly into the proximal OFT cushion, which is associated with abnormal EMT and cell proliferation in Gata4 haploinsufficiency. The genetic interaction of Hh signaling and Gata4 is further demonstrated to be important for OFT development. Gata4 and Smo double heterozygotes displayed more severe OFT abnormalities including persistent truncus arteriosus (PTA). Restoration of Hedgehog signaling renormalized SHF cell proliferation and migration, and rescued OFT defects in Gata4 haploinsufficiency. In addition, there was enhanced Gata6 expression in the SHF of the Gata4 heterozygotes. The Gata4-responsive repressive sites were identified within 1kbp upstream of the transcription start site of Gata6 by both ChIP-qPCR and luciferase reporter assay. These results suggested a SHF regulatory network comprising of Gata4, Gata6 and Hh-signaling for OFT development.


Asunto(s)
Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/genética , Proteínas Hedgehog/genética , Receptor Smoothened/genética , Obstrucción del Flujo Ventricular Externo/genética , Tabique Interventricular/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Embrión de Mamíferos , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/metabolismo , Regulación de la Expresión Génica , Haploinsuficiencia , Proteínas Hedgehog/metabolismo , Heterocigoto , Humanos , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Transducción de Señal , Receptor Smoothened/metabolismo , Tronco Arterial/anomalías , Tronco Arterial/metabolismo , Obstrucción del Flujo Ventricular Externo/metabolismo , Obstrucción del Flujo Ventricular Externo/patología , Tabique Interventricular/patología
7.
J Genet Couns ; 28(4): 779-789, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30907979

RESUMEN

Left ventricular outflow tract obstruction (LVOTO) malformations exhibit higher heritability than other cardiac lesions and cardiac screening is encouraged for first-degree relatives. This study sought to determine the uptake of familial cardiac screening in families with an infant with an LVOTO and assess parental knowledge regarding genetics and heritability of LVOTO. A chart review of the period 2010-2015 identified 69 families who received genetic counseling regarding a diagnosis of LVOTO in an infant. Surveys assessing familial cardiac screening and parental knowledge were completed by a parent in 24 families (completion rate of 35%). Forty percent (36/89) of all at-risk first-degree family members completed cardiac screening. The presence of additional congenital malformations in the affected infant was the only significant factor reducing the uptake of familial cardiac screening (p = 0.003). The reported uptake of screening for subsequent at-risk pregnancies was 11/12 (92%) compared to 25/77 (32%) of living at-risk relatives. Survey respondents answered seven knowledge questions with an average score of 5.2 and all correctly identified that LVOTO can run in families. Uptake of familial cardiac screening is occurring in less than half of at-risk individuals, despite parents demonstrating basic knowledge and receiving genetic counseling. Follow-up counseling in the outpatient setting to review familial screening recommendations should be considered to increase uptake and optimize outcomes.


Asunto(s)
Familia , Cardiopatías Congénitas/genética , Padres , Centros de Atención Terciaria , Obstrucción del Flujo Ventricular Externo/genética , Adulto , Ecocardiografía , Femenino , Asesoramiento Genético , Pruebas Genéticas , Cardiopatías Congénitas/fisiopatología , Humanos , Lactante , Masculino , Riesgo , Obstrucción del Flujo Ventricular Externo/fisiopatología
8.
Genet Epidemiol ; 43(2): 215-226, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30511478

RESUMEN

Loss of function variants in NOTCH1 cause left ventricular outflow tract obstructive defects (LVOTO). However, the risk conferred by rare and noncoding variants in NOTCH1 for LVOTO remains largely uncharacterized. In a cohort of 49 families affected by hypoplastic left heart syndrome, a severe form of LVOTO, we discovered predicted loss of function NOTCH1 variants in 6% of individuals. Rare or low-frequency missense variants were found in 16% of families. To make a quantitative estimate of the genetic risk posed by variants in NOTCH1 for LVOTO, we studied associations of 400 coding and noncoding variants in NOTCH1 in 1,085 cases and 332,788 controls from the UK Biobank. Two rare intronic variants in strong linkage disequilibrium displayed significant association with risk for LVOTO amongst European-ancestry individuals. This result was replicated in an independent analysis of 210 cases and 68,762 controls of non-European and mixed ancestry. In conclusion, carrying rare predicted loss of function variants in NOTCH1 confer significant risk for LVOTO. In addition, the two intronic variants seem to be associated with an increased risk for these defects. Our approach demonstrates the utility of population-based data sets in quantifying the specific risk of individual variants for disease-related phenotypes.


Asunto(s)
Predisposición Genética a la Enfermedad , Cardiopatías Congénitas/genética , Intrones/genética , Mutación con Pérdida de Función/genética , Mutación Missense/genética , Receptor Notch1/genética , Obstrucción del Flujo Ventricular Externo/genética , Estudios de Cohortes , Femenino , Humanos , Masculino , Linaje , Factores de Riesgo , Población Blanca/genética , Secuenciación del Exoma
9.
Cardiol Clin ; 37(1): 11-26, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30447711

RESUMEN

Hypertrophic cardiomyopathy (HCM) is an inherited condition present in about 1/500 individuals with more than 1500 causative mutations identified in primarily 10 sarcomeric proteins. Although HCM is inherited in an autosomal dominant way, there is often incomplete penetrance and variable phenotype even with the same genotype. It is characterized by a degree of hypertrophy (usually asymmetric), that is, not due to another identifiable cause, as well as variable degrees of myocardial fibrosis and microvascular abnormalities.


Asunto(s)
Cardiomiopatía Hipertrófica/diagnóstico por imagen , Ecocardiografía/métodos , Técnicas de Ablación/métodos , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/terapia , Diagnóstico Precoz , Humanos , Angiografía por Resonancia Magnética , Insuficiencia de la Válvula Mitral/diagnóstico por imagen , Insuficiencia de la Válvula Mitral/genética , Insuficiencia de la Válvula Mitral/terapia , Mutación/genética , Linaje , Fenotipo , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/terapia , Obstrucción del Flujo Ventricular Externo/diagnóstico por imagen , Obstrucción del Flujo Ventricular Externo/genética , Obstrucción del Flujo Ventricular Externo/terapia
10.
Curr Opin Pediatr ; 30(5): 609-615, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30045083

RESUMEN

PURPOSE OF REVIEW: Williams syndrome is a multisystem disorder seen with some regularity at most pediatric centers and usually fairly often at larger centers. Cardiovascular abnormalities, because of elastin deficiency, are the leading cause of morbidity and mortality in patients with Williams syndrome. The present article presents a review of the most recent developments regarding the cardiovascular issues in Williams syndrome. RECENT FINDINGS: Cardiovascular abnormalities occur in 80% of patients with Williams syndrome, the majority of which are arterial stenoses. The stenoses seen in Williams syndrome now appear to arise from deficient circumferential arterial growth. Pharmacological therapies aimed at improving the vascular stenoses have shown some promise in animal models. Surgical outcomes for supravalvar aortic stenosis are good at most centers. Transcatheter interventions are largely ineffective in Williams syndrome. Multilevel surgical pulmonary artery reconstruction has excellent results for peripheral pulmonary artery stenosis. Periprocedural risk stratification and management algorithms may decrease the risk of cardiovascular complications. SUMMARY: Cardiovascular abnormalities are a major determining factor in the clinical picture and trajectory of patients with Williams syndrome. Advances in surgical techniques, medical therapeutic options, and periprocedural management hold promise for significant improvements in the cardiovascular outcomes of these patients.


Asunto(s)
Estenosis Aórtica Supravalvular/fisiopatología , Obstrucción del Flujo Ventricular Externo/fisiopatología , Síndrome de Williams/fisiopatología , Estenosis Aórtica Supravalvular/etiología , Estenosis Aórtica Supravalvular/genética , Contraindicaciones , Humanos , Medición de Riesgo , Tomografía Computarizada por Rayos X , Obstrucción del Flujo Ventricular Externo/etiología , Obstrucción del Flujo Ventricular Externo/genética , Síndrome de Williams/complicaciones , Síndrome de Williams/diagnóstico por imagen , Síndrome de Williams/genética
11.
Mol Diagn Ther ; 21(6): 653-661, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28744745

RESUMEN

OBJECTIVE: Maternal plasma is a source of circulating placental nucleic acids. This study was designed to detect aberrantly expressed placental mRNA genes circulating in the maternal plasma of pregnancies affected with fetal conotruncal anomalies (CNTRA) and left-ventricular outflow tract (LVOT) obstruction in the second trimester of pregnancy. METHODS: This was a retrospective monocentric study conducted from 1 Jan 2016 to 31 Dec 2016. NanoString technology was used to identify aberrantly expressed genes, comparing 36 women carrying a fetus with CNTRA or LVOT obstruction to 42 controls at 19-24 weeks of gestation. The genes with differential expression were subsequently tested using real-time polymerase chain reaction. Linear discriminant analysis was used to combine all the mRNA species with discriminant ability for CNTRA and LVOT obstruction. A multivariable receiver operating characteristic (ROC) curve having the estimated discriminant score as an explanatory variable was generated for the two affected groups versus controls. RESULTS: Three genes with differential expression, namely MAPK1, IQGAP1 and Visfatin were found. The ROC curves yielded detection rates of 60 and 62.5% at a false-positive rate of 5% for CNTRA and LVOT, respectively. CONCLUSIONS: These data suggested that molecular screening of CNTRA and LVOT obstruction in the second trimester is feasible. Prospective studies are needed to test the discriminant ability of these genes and to calculate the predictive positive value in the general population.


Asunto(s)
Cardiopatías Congénitas/diagnóstico , Ventrículos Cardíacos/anomalías , ARN Mensajero/sangre , Obstrucción del Flujo Ventricular Externo/diagnóstico , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Citocinas/genética , Electrocardiografía , Femenino , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Humanos , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/genética , Nicotinamida Fosforribosiltransferasa/genética , Embarazo , Segundo Trimestre del Embarazo , Curva ROC , Estudios Retrospectivos , Obstrucción del Flujo Ventricular Externo/genética , Proteínas Activadoras de ras GTPasa/genética
12.
Nat Genet ; 49(7): 1152-1159, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28530678

RESUMEN

Congenital heart disease (CHD) affects up to 1% of live births. Although a genetic etiology is indicated by an increased recurrence risk, sporadic occurrence suggests that CHD genetics is complex. Here, we show that hypoplastic left heart syndrome (HLHS), a severe CHD, is multigenic and genetically heterogeneous. Using mouse forward genetics, we report what is, to our knowledge, the first isolation of HLHS mutant mice and identification of genes causing HLHS. Mutations from seven HLHS mouse lines showed multigenic enrichment in ten human chromosome regions linked to HLHS. Mutations in Sap130 and Pcdha9, genes not previously associated with CHD, were validated by CRISPR-Cas9 genome editing in mice as being digenic causes of HLHS. We also identified one subject with HLHS with SAP130 and PCDHA13 mutations. Mouse and zebrafish modeling showed that Sap130 mediates left ventricular hypoplasia, whereas Pcdha9 increases penetrance of aortic valve abnormalities, both signature HLHS defects. These findings show that HLHS can arise genetically in a combinatorial fashion, thus providing a new paradigm for the complex genetics of CHD.


Asunto(s)
Heterogeneidad Genética , Síndrome del Corazón Izquierdo Hipoplásico/genética , Secuencia de Aminoácidos , Animales , Aorta/embriología , Sistemas CRISPR-Cas , Mapeo Cromosómico , Cromosomas Humanos/genética , Modelos Animales de Enfermedad , Exoma , Femenino , Edición Génica , Técnicas de Inactivación de Genes , Ventrículos Cardíacos/embriología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Mutación Missense , Miocitos Cardíacos/patología , Penetrancia , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Obstrucción del Flujo Ventricular Externo/genética , Pez Cebra/genética
13.
PLoS Genet ; 12(10): e1006335, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27760138

RESUMEN

Left-ventricular outflow tract obstructions (LVOTO) encompass a wide spectrum of phenotypically heterogeneous heart malformations which frequently cluster in families. We performed family based whole-exome and targeted re-sequencing on 182 individuals from 51 families with multiple affected members. Central to our approach is the family unit which serves as a reference to identify causal genotype-phenotype correlations. Screening a multitude of 10 overlapping phenotypes revealed disease associated and co-segregating variants in 12 families. These rare or novel protein altering mutations cluster predominantly in genes (NOTCH1, ARHGAP31, MAML1, SMARCA4, JARID2, JAG1) along the Notch signaling cascade. This is in line with a significant enrichment (Wilcoxon, p< 0.05) of variants with a higher pathogenicity in the Notch signaling pathway in patients compared to controls. The significant enrichment of novel protein truncating and missense mutations in NOTCH1 highlights the allelic and phenotypic heterogeneity in our pediatric cohort. We identified novel co-segregating pathogenic mutations in NOTCH1 associated with left and right-sided cardiac malformations in three independent families with a total of 15 affected individuals. In summary, our results suggest that a small but highly pathogenic fraction of family specific mutations along the Notch cascade are a common cause of LVOTO.


Asunto(s)
Constricción Patológica/genética , Cardiopatías Congénitas/genética , Receptor Notch1/genética , Obstrucción del Flujo Ventricular Externo/genética , Válvula Aórtica/fisiopatología , Codón sin Sentido , Constricción Patológica/fisiopatología , Exoma/genética , Femenino , Estudios de Asociación Genética , Ligamiento Genético , Genoma Humano , Cardiopatías Congénitas/fisiopatología , Humanos , Masculino , Linaje , Receptores Notch/genética , Eliminación de Secuencia , Transducción de Señal/genética , Obstrucción del Flujo Ventricular Externo/fisiopatología
14.
Eur J Clin Invest ; 45(12): 1252-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26444145

RESUMEN

BACKGROUND: Coronary microvascular dysfunction (CMD) is an important feature of hypertrophic cardiomyopathy (HCM), which contributes negatively to symptoms and long-term outcome. Previous in vivo imaging studies in HCM suggest that left ventricular outflow tract (LVOT) gradient and genetic status are important contributors to CMD. CMD may be caused by reduced capillary density. Here, we investigated whether a reduction in capillary density is related to genetic status or LVOT gradient severity in an in vitro study of HCM cardiac samples. METHODS: Using immunofluorescence microscopy, we analysed capillaries (Cap) and cardiomyocytes (CM) in myectomy specimens from 18 HCM patients with maximum left ventricular (LV) wall thickness ≥15 mm. All subjects exhibited significant LVOT obstruction, necessitating septal myectomy. In addition, control myocardium from the LV septal wall was collected at autopsy of 6 individuals that suffered a noncardiac death. RESULTS: CM area was higher in patients with HCM compared to controls. Capillary density was significantly lower in patients with HCM compared with controls (1425 ± 262 vs. 2543 ± 509 Cap/mm(2) , P < 0·001), as was the number of Cap per CM corrected for CM area (2·2 ± 0·5 vs. 4·2 ± 0·9 Cap/CM area, P < 0·001). Capillary density did not differ between genotype-negative and genotype-positive HCM patients at similar resting LVOT gradients. A significant correlation was present between resting LVOT gradient and CM area (r = 0·73, P < 0·001), capillary density (r = -0·74, P < 0·001) and the number of Cap per CM corrected for CM area (r = -0·82, P < 0·001). CONCLUSIONS: Our data indicate that LVOT gradient, rather than genetic status, is associated with reduced capillary density in HCM.


Asunto(s)
Capilares/patología , Cardiomiopatía Hipertrófica/patología , Obstrucción del Flujo Ventricular Externo/patología , Adulto , Anciano , Anciano de 80 o más Años , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Miocardio/patología , Miocitos Cardíacos/patología , Obstrucción del Flujo Ventricular Externo/genética , Obstrucción del Flujo Ventricular Externo/fisiopatología , Adulto Joven
15.
J Am Coll Cardiol ; 64(24): 2589-2600, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25524337

RESUMEN

BACKGROUND: Mild hypertrophy but increased arrhythmic risk characterizes the stereotypic phenotype proposed for hypertrophic cardiomyopathy (HCM) caused by thin-filament mutations. However, whether such clinical profile is different from more prevalent thick-filament-associated disease is unresolved. OBJECTIVES: This study aimed to assess clinical features and outcomes in a large cohort of patients with HCM associated with thin-filament mutations compared with thick-filament HCM. METHODS: Adult HCM patients (age >18 years), 80 with thin-filament and 150 with thick-filament mutations, were followed for an average of 4.5 years. RESULTS: Compared with thick-filament HCM, patients with thin-filament mutations showed: 1) milder and atypically distributed left ventricular (LV) hypertrophy (maximal wall thickness 18 ± 5 mm vs. 24 ± 6 mm; p < 0.001) and less prevalent outflow tract obstruction (19% vs. 34%; p = 0.015); 2) higher rate of progression to New York Heart Association functional class III or IV (15% vs. 5%; p = 0.013); 3) higher prevalence of systolic dysfunction or restrictive LV filling at last evaluation (20% vs. 9%; p = 0.038); 4) 2.4-fold increase in prevalence of triphasic LV filling pattern (26% vs. 11%; p = 0.002); and 5) similar rates of malignant ventricular arrhythmias and sudden cardiac death (p = 0.593). CONCLUSIONS: In adult HCM patients, thin-filament mutations are associated with increased likelihood of advanced LV dysfunction and heart failure compared with thick-filament disease, whereas arrhythmic risk in both subsets is comparable. Triphasic LV filling is particularly common in thin-filament HCM, reflecting profound diastolic dysfunction.


Asunto(s)
Actinas/genética , Cardiomiopatía Hipertrófica , Quinasas Quinasa Quinasa PAM/genética , Troponina T/genética , Citoesqueleto de Actina/genética , Adulto , Cardiomiopatía Hipertrófica/complicaciones , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Muerte Súbita Cardíaca/etiología , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Pruebas de Función Cardíaca , Humanos , Italia , Masculino , Persona de Mediana Edad , Mutación , Evaluación del Resultado de la Atención al Paciente , Proteínas Serina-Treonina Quinasas , Índice de Severidad de la Enfermedad , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/genética , Fibrilación Ventricular/etiología , Fibrilación Ventricular/genética , Obstrucción del Flujo Ventricular Externo/etiología , Obstrucción del Flujo Ventricular Externo/genética
17.
Eur Heart J ; 35(39): 2706-13, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-24810389

RESUMEN

AIMS: Cardiovascular magnetic resonance (CMR) has improved diagnostic and management strategies in hypertrophic cardiomyopathy (HCM) by expanding our appreciation for the diverse phenotypic expression. We sought to characterize the prevalence and clinical significance of a recently identified accessory left ventricular (LV) muscle bundle extending from the apex to the basal septum or anterior wall (i.e. apical-basal). METHODS AND RESULTS: CMR was performed in 230 genotyped HCM patients (48 ± 15 years, 69% male), 30 genotype-positive/phenotype-negative (G+/P-) family members (32 ± 15 years, 30% male), and 126 controls. Left ventricular apical-basal muscle bundle was identified in 145 of 230 (63%) HCM patients, 18 of 30 (60%) G+/P- family members, and 12 of 126 (10%) controls (G+/P- vs. controls; P < 0.01). In HCM patients, the prevalence of an apical-basal muscle bundle was similar among those with disease-causing sarcomere mutations compared with patients without mutation (64 vs. 62%; P = 0.88). The presence of an LV apical-basal muscle bundle was not associated with LV outflow tract obstruction (P = 0.61). In follow-up, 33 patients underwent surgical myectomy of whom 22 (67%) were identified to have an accessory LV apical-basal muscle bundle, which was resected in all patients. CONCLUSION: Apical-basal muscle bundles are a unique myocardial structure commonly present in HCM patients as well as in G+/P- family members and may represent an additional morphologic marker for HCM diagnosis in genotype-positive status.


Asunto(s)
Cardiomiopatía Hipertrófica/patología , Miocardio/patología , Adulto , Análisis de Varianza , Cardiomiopatía Hipertrófica/genética , Estudios de Casos y Controles , Análisis Mutacional de ADN , Genotipo , Ventrículos Cardíacos , Humanos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Angiografía por Resonancia Magnética/métodos , Imagen por Resonancia Cinemagnética/métodos , Masculino , Persona de Mediana Edad , Mutación/genética , Linaje , Fenotipo , Obstrucción del Flujo Ventricular Externo/genética , Obstrucción del Flujo Ventricular Externo/patología
18.
J Cardiol ; 64(6): 463-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24768408

RESUMEN

BACKGROUND: Within the diverse phenotypic spectrum of hypertrophic cardiomyopathy (HCM), subgroups of patients with apical hypertrophy (APH), midventricular obstruction (MVO), and apical aneurysm (APA) have emerged. While previous studies have suggested the existence of considerable overlap between APH, MVO, and APA, there are still many unanswered questions. Therefore, we attempted to clarify the relationship of the above three phenotypes of HCM with respect to prevalence, overlap, and outcomes. METHODS: Among the 544 study HCM patients (mean follow-up period: 11.6±7.4 years), 170 with APH (31.3%), 51 with MVO (9.4%), and 24 with APA (4.4%) were examined. RESULTS: There was phenotypic overlap between APH and MVO in 17 patients, APH and APA in 14 patients, and MVO and APA in 14 patients. Furthermore, a combination of APH, MVO, and APA was observed in eight patients. Detailed analysis of the relationship between overlapping phenotypes and the prognosis showed that APA patients without a history of APH had an extremely poor outcome (probability of the combined endpoint of sudden death and potentially lethal arrhythmic events ≥50%). Conversely, APH patients without MVO had a strikingly good outcome (probability of the combined endpoint <5%). Other patients had an intermediate outcome (probability of the combined endpoint 10-40%). CONCLUSIONS: Our results suggest that overlap between these three forms of HCM is substantial, and that detailed classification of the overlapping phenotypes is clinically meaningful.


Asunto(s)
Aneurisma/genética , Cardiomiopatía Hipertrófica/genética , Hipertrofia Ventricular Izquierda/genética , Obstrucción del Flujo Ventricular Externo/genética , Aneurisma/fisiopatología , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/fisiopatología , Muerte Súbita Cardíaca , Ecocardiografía , Femenino , Humanos , Hipertrofia Ventricular Izquierda/fisiopatología , Japón , Masculino , Persona de Mediana Edad , Fenotipo , Estudios Retrospectivos , Obstrucción del Flujo Ventricular Externo/fisiopatología
19.
Herz ; 39(2): 258-63, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23880944

RESUMEN

BACKGROUND: Variations of angiotensinogen (AGT) gene have been associated with cardiac hypertrophy. We hypothesized that AGT gene polymorphism may play a modifier role in the diversity of left ventricular outflow obstruction. METHODS: The polymorphisms of the AGT gene were genotyped in 225 patients with hypertrophic cardiomyopathy (HCM) and 243 age-and sex-matched healthy controls. The effect of the A and G alleles on the expression of the reporter gene were evaluated in vitro using dual-luciferase reporter assays. RESULTS: Our results showed that the frequency of the A allele was higher in patients than in controls (50.2 % vs. 35.8 %, p < 0.05). Patients carrying the AA and AG genotypes had a higher proportion of left ventricular outflow obstruction (30.1 % vs. 17.0 %, p < 0.05) and heart failure (NYHA functional class III ~ IV, 35.4 % vs. 18.8 %, p < 0.05) than those carrying the GG genotype had. After adjusted for age, sex, the thickness of the interventricular septum, family history of HCM, and sudden death, the A allele conferred a 2.4-fold risk for left ventricular outflow obstruction than the GG genotype did (adjusted OR = 2.4, 95 %CI 1.2-4.8). The G allele suppressed the expression of the reporter gene significantly compared with the A allele (p < 0.05). CONCLUSION: AGT gene variations may be genetic modifiers for the development of HCM.


Asunto(s)
Angiotensinógeno/genética , Cardiomiopatía Hipertrófica/epidemiología , Cardiomiopatía Hipertrófica/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , Obstrucción del Flujo Ventricular Externo/epidemiología , Obstrucción del Flujo Ventricular Externo/genética , Adulto , China/epidemiología , Comorbilidad , Femenino , Marcadores Genéticos/genética , Variación Genética/genética , Humanos , Masculino , Prevalencia , Factores de Riesgo
20.
Can J Cardiol ; 29(12): 1695-703, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24140236

RESUMEN

BACKGROUND: Tetralogy of Fallot (TOF) is 1 of the most common heart defects in children, and the underlying mechanisms remain largely elusive. MicroRNAs (miRNAs) are a class of regulators of gene expression and are increasingly recognized for their roles in heart development. METHODS: To identify miRNAs abnormally expressed in TOF, microarrays were used to analyze the miRNA expression profiles of 5 samples of myectomy tissues from right ventricular outflow tract (RVOT) obstruction of infants with nonsyndromic TOF and 3 age-matched normal RVOT tissues. RESULTS: In total, 41 candidate miRNAs were identified. To further validate the microarray results, the 41 miRNAs were detected using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) in a larger independent population of tissue samples, including 21 from patients with TOF and 6 from normal controls; it was found that 18 miRNAs were expressed at significantly different levels. Bioinformatic analysis revealed that these miRNAs targeted a network of genes involved in heart development and human congenital heart diseases. Further in vitro studies indicated that upregulation of miR-424/424* promoted proliferation and inhibited migration of primary embryonic mouse cardiomyocytes, whereas miR-222 promoted cardiomyocyte proliferation and reduced the cardiomyogenic differentiation of P19 cells. The 3'UTR (3' untranslated region) luciferase assay revealed that miR-424/424* suppressed the expression of HAS2 and NF1, and their mRNAs were underexpressed in the RVOT myocardial tissues of TOF. CONCLUSIONS: Eighteen miRNAs were identified as being deregulated in RVOT myocardial tissues from infants with nonsyndromic TOF, and in vitro experiments indicated that miR-424/424* and miR-222 are involved in cardiomyocyte proliferation and migration and the cardiomyogenic differentiation of P19 cells.


Asunto(s)
Regulación de la Expresión Génica/genética , MicroARNs/genética , Miocardio/metabolismo , Tetralogía de Fallot/genética , Obstrucción del Flujo Ventricular Externo/genética , Regiones no Traducidas 3'/genética , Animales , Diferenciación Celular/genética , Línea Celular , Proliferación Celular , Estudios de Asociación Genética , Humanos , Lactante , Ratones , Modelos Genéticos , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Tetralogía de Fallot/patología , Tetralogía de Fallot/cirugía , Análisis de Matrices Tisulares , Regulación hacia Arriba/genética , Obstrucción del Flujo Ventricular Externo/patología , Obstrucción del Flujo Ventricular Externo/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...