Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
1.
J Am Heart Assoc ; 10(15): e020227, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34310159

RESUMEN

Background Impaired myocardial blood flow (MBF) in the absence of epicardial coronary disease is a feature of hypertrophic cardiomyopathy (HCM). Although most evident in hypertrophied or scarred segments, reduced MBF can occur in apparently normal segments. We hypothesized that impaired MBF and myocardial perfusion reserve, quantified using perfusion mapping cardiac magnetic resonance, might occur in the absence of overt left ventricular hypertrophy (LVH) and late gadolinium enhancement, in mutation carriers without LVH criteria for HCM (genotype-positive, left ventricular hypertrophy-negative). Methods and Results A single center, case-control study investigated MBF and myocardial perfusion reserve (the ratio of MBF at stress:rest), along with other pre-phenotypic features of HCM. Individuals with genotype-positive, left ventricular hypertrophy-negative (n=50) with likely pathogenic/pathogenic variants and no evidence of LVH, and matched controls (n=28) underwent cardiac magnetic resonance. Cardiac magnetic resonance identified LVH-fulfilling criteria for HCM in 5 patients who were excluded. Individuals with genotype-positive, left ventricular hypertrophy-negative had longer indexed anterior mitral valve leaflet length (12.52±2.1 versus 11.55±1.6 mm/m2, P=0.03), lower left ventricular end-systolic volume (21.0±6.9 versus 26.7±6.2 mm/m2, P≤0.005) and higher left ventricular ejection fraction (71.9±5.5 versus 65.8±4.4%, P≤0.005). Maximum wall thickness was not significantly different (9.03±1.95 versus 8.37±1.2 mm, P=0.075), and no subject had significant late gadolinium enhancement (minor right ventricle‒insertion point late gadolinium enhancement only). Perfusion mapping demonstrated visual perfusion defects in 9 (20%) carriers versus 0 controls (P=0.011). These were almost all septal or near right ventricle insertion points. Globally, myocardial perfusion reserve was lower in carriers (2.77±0.83 versus 3.24±0.63, P=0.009), with a subendocardial:subepicardial myocardial perfusion reserve gradient (2.55±0.75 versus 3.2±0.65, P=<0.005; 3.01±0.96 versus 3.47±0.75, P=0.026) but equivalent MBF (2.75±0.82 versus 2.65±0.69 mL/g per min, P=0.826). Conclusions Regional and global impaired myocardial perfusion can occur in HCM mutation carriers, in the absence of significant hypertrophy or scarring.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar , Hipertrofia Ventricular Izquierda , Imagen por Resonancia Cinemagnética/métodos , Imagen de Perfusión Miocárdica/métodos , Adulto , Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Circulación Coronaria/fisiología , Electrocardiografía/métodos , Femenino , Pruebas Genéticas/métodos , Ventrículos Cardíacos/diagnóstico por imagen , Heterocigoto , Humanos , Hipertrofia Ventricular Izquierda/diagnóstico , Hipertrofia Ventricular Izquierda/etiología , Angiografía por Resonancia Magnética/métodos , Masculino , Microcirculación , Mutación , Sarcómeros/genética , Sarcómeros/patología
2.
Cardiol Clin ; 39(1): 143-150, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33222809

RESUMEN

Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiac condition and highly heterogeneous. Echocardiography and genetic and clinical screening have led to detection in women of childbearing age. Maternal and fetal outcomes among women with HCM are favorable. Genetic counseling is recommended. Prepregnancy clinical evaluation and risk assessment are paramount in ensuring optimal outcomes. Most women carry moderate risk of morbidity, have clinical evaluations and echocardiography each trimester, and deliver vaginally. Those who are symptomatic or have significant left ventricular outflow obstruction or recurrent arrhythmias prior to pregnancy are at higher risk and should be monitored at least monthly.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar , Atención Preconceptiva/métodos , Complicaciones Cardiovasculares del Embarazo , Cardiomiopatía Hipertrófica Familiar/diagnóstico , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Cardiomiopatía Hipertrófica Familiar/terapia , Femenino , Humanos , Manejo de Atención al Paciente/métodos , Embarazo , Complicaciones Cardiovasculares del Embarazo/diagnóstico , Complicaciones Cardiovasculares del Embarazo/genética , Complicaciones Cardiovasculares del Embarazo/fisiopatología , Complicaciones Cardiovasculares del Embarazo/terapia , Pronóstico , Ajuste de Riesgo/métodos
3.
BMC Cardiovasc Disord ; 20(1): 516, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33297970

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disease that affects approximately one in 500 people. HCM is a recognized genetic disorder most often caused by mutations involving myosin-binding protein C (MYBPC3) and ß-myosin heavy chain (MYH7) which are responsible for approximately three-quarters of the identified mutations. METHODS: As a part of the international multidisciplinary SILICOFCM project ( www.silicofcm.eu ) the present study evaluated the association between underlying genetic mutations and clinical phenotype in patients with HCM. Only patients with confirmed single pathogenic mutations in either MYBPC3 or MYH7 genes were included in the study and divided into two groups accordingly. The MYBPC3 group was comprised of 48 patients (76%), while the MYH7 group included 15 patients (24%). Each patient underwent clinical examination and echocardiography. RESULTS: The most prevalent symptom in patients with MYBPC3 was dyspnea (44%), whereas in patients with MYH7 it was palpitations (33%). The MYBPC3 group had a significantly higher number of patients with a positive family history of HCM (46% vs. 7%; p = 0.014). There was a numerically higher prevalence of atrial fibrillation in the MYH7 group (60% vs. 35%, p = 0.085). Laboratory analyses revealed normal levels of creatinine (85.5 ± 18.3 vs. 81.3 ± 16.4 µmol/l; p = 0.487) and blood urea nitrogen (10.2 ± 15.6 vs. 6.9 ± 3.9 mmol/l; p = 0.472) which were similar in both groups. The systolic anterior motion presence was significantly more frequent in patients carrying MYH7 mutation (33% vs. 10%; p = 0.025), as well as mitral leaflet abnormalities (40% vs. 19%; p = 0.039). Calcifications of mitral annulus were registered only in MYH7 patients (20% vs. 0%; p = 0.001). The difference in diastolic function, i.e. E/e' ratio between the two groups was also noted (MYBPC3 8.8 ± 3.3, MYH7 13.9 ± 6.9, p = 0.079). CONCLUSIONS: Major findings of the present study corroborate the notion that MYH7 gene mutation patients are presented with more pronounced disease severity than those with MYBPC3.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar/genética , Proteínas Portadoras/genética , Mutación , Cadenas Pesadas de Miosina/genética , Adulto , Anciano , Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Cardiomiopatía Hipertrófica Familiar/epidemiología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Estudios Transversales , Análisis Mutacional de ADN , Ecocardiografía , Electrocardiografía , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Prevalencia , Pronóstico , Índice de Severidad de la Enfermedad
4.
Circ J ; 84(8): 1218-1226, 2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32669480

RESUMEN

Hypertrophic cardiomyopathy (HCM) is the most frequent hereditary cardiomyopathy, showing an autosomal-dominant f inheritance. A great deal of attention has been paid to genetics, left ventricular tract obstruction and the prediction and prevention of sudden cardiac death in HCM. Needless to say, these are very important, but we should recognize the heterogeneity in etiology, morphology, clinical course and management of this unique cardiomyopathy. Another important perspective is that HCM causes left ventricular remodeling over time and is a disease that requires lifelong management in the real world.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/fisiopatología , Función Ventricular Izquierda , Remodelación Ventricular , Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/terapia , Predisposición Genética a la Enfermedad , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Mutación , Fenotipo , Pronóstico , Función Ventricular Izquierda/genética , Remodelación Ventricular/genética
5.
Curr Probl Cardiol ; 45(6): 100398, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30497690

RESUMEN

Hypertrophic cardiomyopathy (HCM) is the most prevalent hereditary cardiac disease characterized by the presence of left ventricular and/or septal hypertrophy in the absence of other underlying cardiac disorders. Patients of HCM have a broad range of clinical presentation from being asymptomatic to severely ill condition requires hospitalization and urgent management. Broadly, HCM is classified in two variants: obstructive and nonobstructive. The mainstay of diagnosis is through echocardiography. As HCM chiefly affect the left heart, pulmonary hypertension (PH) is an expected complication of this disease. Though the existence of PH in HCM is known for a long time, its clinical significance, underlying mechanism, and prognostic impact in HCM have been revealed by few recent studies. Specifically, studies have shown increased events of thromboembolism, atrial fibrillation, and heart failure in patients with HCM and PH. These studies elucidated the underlying mechanism of PH in HCM--a rise of pressure in the precapillary and postcapillary pulmonary vasculature. In addition to left ventricular involvement, studies have shown right ventricular involvement and the association of left and right ventricular dysfunction in these patients. Further, it has been shown that surgical intervention to reduce septal thickness improves survival in pharmacotherapy nonresponders and the presence of PH does not increase mortality in these patients. We present a comprehensive review exploring the prevalence, underlying mechanisms, and impact of PH on HCM.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar , Manejo de la Enfermedad , Hipertensión Pulmonar , Cardiomiopatía Hipertrófica Familiar/complicaciones , Cardiomiopatía Hipertrófica Familiar/diagnóstico , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Cardiomiopatía Hipertrófica Familiar/terapia , Humanos , Hipertensión Pulmonar/epidemiología , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/terapia , Prevalencia , Pronóstico
6.
J Mol Med (Berl) ; 97(7): 1033-1047, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31101927

RESUMEN

Familial hypertrophic cardiomyopathy (HCM) is an autosomal dominant disorder characterized by ventricular hypertrophy, myofibrillar disarray, and fibrosis, and is primarily caused by mutations in sarcomeric genes. With no definitive cure for HCM, there is an urgent need for the development of novel preventive and reparative therapies. This study is focused on aspartic acid-to-valine (D166V) mutation in the myosin regulatory light chain, RLC (MYL2 gene), associated with a malignant form of HCM. Since myosin RLC phosphorylation is critical for normal cardiac function, we aimed to exploit this post-translational modification via phosphomimetic-RLC gene therapy. We hypothesized that mimicking/modulating cardiac RLC phosphorylation in non-phosphorylatable D166V myocardium would improve heart function of HCM-D166V mice. Adeno-associated virus, serotype-9 (AAV9) was used to deliver phosphomimetic human RLC variant with serine-to-aspartic acid substitution at Ser15-RLC phosphorylation site (S15D-RLC) into the hearts of humanized HCM-D166V mice. Improvement of heart function was monitored by echocardiography, invasive hemodynamics (PV-loops) and muscle contractile mechanics. A significant increase in cardiac output and stroke work and a decrease in relaxation constant, Tau, shown to be prolonged in HCM mice, were observed in AAV- vs. PBS-injected HCM mice. Strain analysis showed enhanced myocardial longitudinal shortening in AAV-treated vs. control mice. In addition, increased maximal contractile force was observed in skinned papillary muscles from AAV-injected HCM hearts. Our data suggest that myosin RLC phosphorylation may have important translational implications for the treatment of RLC mutations-induced HCM and possibly play a role in other disease settings accompanied by depressed Ser15-RLC phosphorylation. KEY MESSAGES: HCM-D166V mice show decreased RLC phosphorylation and decompensated function. AAV9-S15D-RLC gene therapy in HCM-D166V mice, but not in WT-RLC, results in improved heart performance. Global longitudinal strain analysis shows enhanced contractility in AAV vs controls. Increased systolic and diastolic function is paralleled by higher contractile force. Phosphomimic S15D-RLC has a therapeutic potential for HCM.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/terapia , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/metabolismo , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/uso terapéutico , Animales , Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Fibrosis , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Hemodinámica , Humanos , Hidroxiprolina/metabolismo , Masculino , Ratones , Contracción Muscular , Músculos/metabolismo
7.
Int Heart J ; 60(2): 477-481, 2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30745532

RESUMEN

This case report demonstrates a pair of monozygotic twins with hypertrophic cardiomyopathy (HCM) carrying the same pathogenic mutation of MYH7 (p.G768R; c.2302G>A), detected by whole exome and Sanger genetic sequencing methods. On multi-modality imaging, they were reported to have similar, but not identical, morphologic expression. Particularly, the clinical presentation and tissue characteristics were not the same. Late gadolinium enhancement (LGE) and T1 mapping of cardiac magnetic resonance showed different extents of myocardial fibrotic characteristics in the twins (twin A: 16.3% LGE and 32.6% extracellular volume [ECV] of the whole left ventricle; twin B: 5.4% LGE and 28.1% ECV of the whole left ventricle). This extraordinary case of HCM provides evidence on the complex pathophysiological mechanisms of HCM and suggests the likely impact of epigenetics and environmental factors on HCM phenotype.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar , Ecocardiografía/métodos , Electrocardiografía/métodos , Ventrículos Cardíacos , Imagen por Resonancia Cinemagnética/métodos , Cadenas Pesadas de Miosina/genética , Cardiomiopatía Hipertrófica Familiar/diagnóstico , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Medios de Contraste/farmacología , Manejo de la Enfermedad , Epigénesis Genética , Femenino , Gadolinio/farmacología , Interacción Gen-Ambiente , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/patología , Humanos , Persona de Mediana Edad , Imagen Multimodal/métodos , Mutación , Tamaño de los Órganos , Gemelos Monocigóticos
9.
Cardiol Young ; 28(9): 1106-1114, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29978770

RESUMEN

BACKGROUND: We previously reported four heterozygous missense mutations of MYH7, KCNQ1, MYLK2, and TMEM70 in a single three-generation Chinese family with dual Long QT and hypertrophic cardiomyopathy phenotypes for the first time. However, the clinical course among the family members was various, and the potential myocardial dysfunction has not been investigated. OBJECTIVES: The objective of this study was to investigate the echocardiographic and electrocardiographic characteristics in a genetic positive Chinese family with hypertrophic cardiomyopathy and further to explore the association between myocardial dysfunction and electric activity, and the identified mutations. METHODS: A comprehensive echocardiogram - standard two-dimensional Doppler echocardiography and three-dimensional speckle tracking echocardiography - and electrocardiogram were obtained for members in this family. RESULTS: As previously reported, four missense mutations - MYH7-H1717Q, KCNQ1-R190W, MYLK2-K324E, and TMEM70-I147T - were identified in this family. The MYH7-H1717Q mutation carriers had significantly increased left ventricular mass indices, elevated E/e' ratio, deteriorated global longitudinal stain, but enhanced global circumferential and radial strain compared with those in non-mutation patients (all p<0.05). The KCNQ1-R190W carriers showed significantly prolonged QTc intervals, and the MYLK2-K324E mutation carriers showed inverted T-waves (both p<0.05). However, the TMEM70-I147T mutation carriers had similar echocardiography and electrocardiographic data as non-mutation patients. CONCLUSIONS: Three of the identified four mutations had potential pathogenic effects in this family: MYH7-H1717Q was associated with increased left ventricular thickness, elevated left ventricular filling pressure, and altered myocardial deformation; KCNQ1-R190W and MYLK2-K324E mutations were correlated with electrocardiographic abnormalities reflected in long QT phenotype and inverted T-waves, respectively.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/diagnóstico , Ecocardiografía Doppler/métodos , Ecocardiografía Tridimensional/métodos , Predisposición Genética a la Enfermedad , Ventrículos Cardíacos/fisiopatología , Contracción Miocárdica/fisiología , Volumen Sistólico/fisiología , Adolescente , Adulto , Anciano , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Niño , China/epidemiología , Electrocardiografía , Femenino , Ventrículos Cardíacos/diagnóstico por imagen , Humanos , Masculino , Persona de Mediana Edad , Morbilidad/tendencias , Mutación Missense , Linaje , Fenotipo , Adulto Joven
10.
Biochem Biophys Res Commun ; 496(4): 1250-1256, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29402414

RESUMEN

AIMS: Cardiac Troponin T (TnT) mutation-linked familial hypertrophic cardiomyopathy (FHC) is known to cause sudden cardiac death at a young age. Here, we investigated the role of the Ca2+ release channel of the cardiac sarcoplasmic reticulum (SR), ryanodine receptor (RyR2), in the pathogenic mechanism of lethal arrhythmia in FHC-related TnT-mutated transgenic mice (TG; TnT-delta160E). METHODS AND RESULTS: In TG cardiomyocytes, the Ca2+ spark frequency (SpF) was much higher than that in non-TG cardiomyocytes. These differences were more pronounced in the presence of isoproterenol (ISO; 10 nM). This increase in SpF was largely reversed by a CaMKII inhibitor (KN-93), but not by a protein kinase A inhibitor (H89). CaMKII phosphorylation at Ser2814 in RyR2 was increased significantly in TG. Spontaneous Ca2+ transients (sCaTs) after cessation of a 1-5 Hz pacing, frequently observed in ISO-treated TG cardiomyocytes, were also attenuated by KN-93, but not by H89. The RyR2 stabilizer dantrolene attenuated Ca2+ sparks and sCaTs in ISO-treated TG cardiomyocytes, indicating that the mutation-linked aberrant Ca2+ release is mediated by destabilized RyR2. CONCLUSIONS: In FHC-linked TnT-mutated hearts, RyR2 is susceptible to CaMKII-mediated phosphorylation, presumably because of a mutation-linked increase in diastolic [Ca2+]i, causing aberrant Ca2+ release leading to lethal arrhythmia.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Miocitos Cardíacos/metabolismo , Troponina T/metabolismo , Animales , Arritmias Cardíacas/etiología , Cardiomiopatía Hipertrófica Familiar/complicaciones , Células Cultivadas , Ratones , Ratones Transgénicos , Fosforilación , Retículo Sarcoplasmático/metabolismo
11.
Circ Cardiovasc Imaging ; 10(2)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28193612

RESUMEN

BACKGROUND: The 2 most commonly affected genes in hypertrophic cardiomyopathy (HCM) are MYH7 (ß-myosin heavy chain) and MYBPC3 (ß-myosin-binding protein C). Phenotypic differences between patients with mutations in these 2 genes have been inconsistent. Scarce data exist on the genotype-phenotype association as assessed by tomographic imaging using cardiac magnetic resonance imaging. METHODS AND RESULTS: Cardiac magnetic resonance imaging was performed on 358 consecutive genotyped hypertrophic cardiomyopathy probands at 5 tertiary hypertrophic cardiomyopathy centers. Genetic testing revealed a pathogenic mutation in 159 patients (44.4%). The most common genes identified were MYH7 (n=53) and MYBPC3 (n=75); 33.1% and 47% of genopositive patients, respectively. Phenotypic characteristics by cardiac magnetic resonance imaging of these 2 groups were similar, including left ventricular volumes, mass, maximal wall thickness, morphology, left atrial volume, and mitral valve leaflet lengths (all P=non-significant). The presence of late gadolinium enhancement (65% versus 64%; P=0.99) and the proportion of total left ventricular mass (%late gadolinium enhancement; 10.4±13.2% versus 8.5±8.5%; P=0.44) were also similar. CONCLUSIONS: This multicenter multinational study shows lack of phenotypic differences between MYH7- and MYBPC3-associated hypertrophic cardiomyopathy when assessed by cardiac magnetic resonance imaging. Postmutational mechanisms appear more relevant to thick-filament disease expression and outcome than the disease-causing variant per se.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Cardiomiopatía Hipertrófica Familiar/genética , Proteínas Portadoras/genética , Imagen por Resonancia Cinemagnética , Mutación , Cadenas Pesadas de Miosina/genética , Adulto , Canadá , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Medios de Contraste/administración & dosificación , Europa (Continente) , Femenino , Gadolinio DTPA/administración & dosificación , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Interpretación de Imagen Asistida por Computador , Imagenología Tridimensional , Masculino , Persona de Mediana Edad , Fenotipo , Valor Predictivo de las Pruebas , Sistema de Registros , Factores de Riesgo , Volumen Sistólico , Centros de Atención Terciaria , Estados Unidos , Función Ventricular Izquierda , Remodelación Ventricular
12.
Int. j. cardiovasc. sci. (Impr.) ; 29(6): 504-511, nov.-dez.2016.
Artículo en Portugués | LILACS | ID: biblio-832441

RESUMEN

A cardiomiopatia hipertrófica é uma doença genética do músculo cardíaco, autossômica dominante, caracterizada por hipertrofia ventricular na ausência de qualquer outra condição clínica que leve à sobrecarga do coração. Estima-se prevalência de 1:500, sendo importante causa de morte súbita, especialmente em jovens, com incidência anual em torno de 1%. Entre os marcadores de risco para a ocorrência de arritmias ventriculares malignas e morte súbita neste cenário, enfatizam-se, além de um evento fatal já ocorrido e abortado, história familiar de morte súbita; espessura de parede maior ou igual a 30mm; síncope inexplicada; presença de taquicardia ventricular não sustentada ao Holter; resposta pressórica anormal no teste ergométrico; e presença de realce tardio na ressonância magnética do coração. A presença ou ausência destes marcadores pode definir a necessidade ou não do implante de cardiodesfibrilador implantável como forma de prevenir a morte súbita nestes pacientes. Entretanto, ainda existe muita controvérsia sobre a forma pela qual estes pacientes devam ser estratificados. Sabe-se que estes marcadores não têm o mesmo peso em predizer quem tem mais chance de sofrer um evento fatal. Este fato torna-se particularmente importante quando se constata que o procedimento de implante de cardiodesfibrilador implantável não é isento de complicações, além do impacto econômico, em termos do custo para o sistema de saúde. A proposta deste artigo é a realização de uma revisão sobre os principais aspectos envolvidos na morte súbita destes pacientes, desde a fisiopatologia, a avaliação de risco, a prevenção e as perspectivas futuras.


Asunto(s)
Humanos , Cardiomiopatía Hipertrófica Familiar/mortalidad , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Muerte Súbita Cardíaca/patología , Ecocardiografía , Imagen por Resonancia Magnética
13.
Cardiovasc J Afr ; 27(3): 152-158, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27841901

RESUMEN

BACKGROUND: Little is known about the clinical characteristics, spectrum of causal genetic mutations and outcome of hypertrophic cardiomyopathy (HCM) in Africans. The objective of this study was to delineate the clinical and genetic features and outcome of HCM in African patients. METHODS: Information on clinical presentation, electrocardiographic and echocardiographic findings, and outcome of cases with HCM was collected from the Cardiac Clinic at Groote Schuur Hospital over a mean duration of follow up of 9.1 ± 3.4 years. Genomic DNA was screened for mutations in 15 genes that cause HCM, i.e. cardiac myosin-binding protein C (MYBPC3), cardiac ß-myosin heavy chain (MYH7), cardiac troponin T2 (TNNT2), cardiac troponin I (TNNI3), regulatory light chain of myosin (MYL2), essential light chain of myosin (MYL3), tropomyosin 1 (TPM1), phospholamban (PLN), α-actin (ACTC1), cysteine and glycine-rich protein 3 (CSRP3), AMP-activated protein kinase (PRKAG2), α-galactosidase (GLA), four-and-a-half LIM domains 1 (FHL1), lamin A/C (LMNA) and lysosome-associated membrane protein 2 (LAMP2). Survival and its predictors were analysed using the Kaplan-Meier and Cox proportional hazards regression methods, respectively. RESULTS: Forty-three consecutive patients [mean age 38.5 ± 14.3 years; 25 (58.1%) male; and 13 (30.2%) black African] were prospectively enrolled in the study from January 1996 to December 2012. Clinical presentation was similar to that reported in other studies. The South African founder mutations that cause HCM were not found in the 42 probands. Ten of 35 index cases (28.6%) tested for mutations in 15 genes had disease-causing mutations in MYH7 (six cases or 60%) and MYBPC3 (four cases or 40%). No disease-causing mutation was found in the other 13 genes screened. The annual mortality rate was 2.9% per annum and overall survival was 74% at 10 years, which was similar to the general South African population. Cox's proportional hazards regression showed that survival was predicted by New York Heart Association (NYHA) functional class at last visit (p equals; 0.026), but not by the presence of a disease-causing mutation (p = 0.474). CONCLUSIONS: Comprehensive genetic screening was associated with a 29% yield of causal genetic mutations in South African HCM cases, all in MYH7 and MBPC3 genes. A quarter of the patients had died after a decade of follow up, with NYHA functional class serving as a predictor of survival.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar/genética , Proteínas Portadoras/genética , Mutación , Cadenas Pesadas de Miosina/genética , Adulto , Anciano , Cardiomiopatía Hipertrófica Familiar/diagnóstico , Cardiomiopatía Hipertrófica Familiar/mortalidad , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Análisis Mutacional de ADN , Ecocardiografía , Electrocardiografía , Femenino , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Estimación de Kaplan-Meier , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Fenotipo , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Factores de Riesgo , Sudáfrica/epidemiología , Factores de Tiempo
14.
Int J Cardiovasc Imaging ; 32(9): 1379-1389, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27324645

RESUMEN

To evaluate if morphological or functional abnormalities could be detected with echocardiography in hypertrophic myocardiopathy (HCM) mutation carriers without left ventricle (LV) hypertrophy has developed. HCM is caused by extensive genes mutations found in two-third of patients. Because screening for carriership of a large population is unreasonable, identification of asymptomatic subjects is confined to the use of imaging such as echocardiography, by which subtle abnormalities can be detected. Comprehensive echocardiographic studies including morphological and functional assessment were performed. Asymptomatic HCM mutation carriers without hypertrophy (Phe-/Gen+, n = 14), and HCM patients (Phe+/Gen+, n = 17) were compared with healthy control subjects (n = 32) in a prospective design. Compared to controls, septum thickness was significantly higher with an elongated mitral valve in both groups. Thickened LV muscular band (LVMB) are more likely found in Phe-/Gen+ and Phe+/Gen+. The thickness of LVMB was higher in the Phe-/Gen+ versus controls. A LVMB thickness ≥3.6 mm was associated with HCM mutation carriership (sensitivity: 76.9 %, specificity: 94.1 %). The regional strain was significantly impaired in the basal segments of the septum in the Phe-/Gen+. The GLS was significantly impaired in the Phe+/Gen+ (-16.4 % ± 2.9 vs. -21.4 % ± 2.3 in control subjects, p = 0.01). Mitral A wave velocity, septal E/e', averaged E/e' were increased in both groups. E/A ratio was significantly lower in Phe+/Gen+. Morphological and functional abnormalities in hypertrophy-free HCM mutation carriers could be detected with echocardiography. Anomalous thickened LVMB could be representing a morphological marker for the HCM disease without overt hypertrophy has developed or in patients with an ambiguous diagnosis.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Ecocardiografía Doppler , Ventrículos Cardíacos/diagnóstico por imagen , Mutación , Contracción Miocárdica , Función Ventricular Izquierda , Adolescente , Adulto , Anciano , Área Bajo la Curva , Enfermedades Asintomáticas , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Estudios de Casos y Controles , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Válvula Mitral/diagnóstico por imagen , Válvula Mitral/fisiopatología , Fenotipo , Valor Predictivo de las Pruebas , Estudios Prospectivos , Curva ROC , Adulto Joven
15.
BMC Cardiovasc Disord ; 16: 83, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27160240

RESUMEN

BACKGROUND: Fabry disease is caused by mutations in the α-galactosidase A (GLA) gene, which is located in X-chromosome coding for the lysosomal enzyme of GLA. Among many gene mutations, E66Q mutation is under discussion for its pathogenicity because there is no clinical report showing pathological evidence of Fabry disease with E66Q mutation. CASE PRESENTATION: A 65-year-old Japanese female was referred to our hospital for chest discomfort on effort. Transthoracic echocardiography showed severe left ventricular (LV) hypertrophy with LV outflow obstruction. Maximum LV outflow pressure gradient was 87 mmHg, and Valsalva maneuver increased the pressure gradient up to 98 mmHg. According to medical interview, one of her younger sister and a nephew died suddenly at age 42 and 36, respectively. Another younger sister also presented LV hypertrophy with outflow obstruction. Maximum LV outflow pressure gradient was 100 mmHg, and the E66Q mutation was detected similar to the case. Endomyocardial biopsy specimens presented vacuolation of cardiomyocytes, in which zebra bodies were detected by electron microscopic examination. Although the enzymatic activity of GLA was within normal range, the c. 196G>C nucleotide change, which lead to the E66Q mutation of GLA gene, was detected. We initially diagnosed her as cardiac Fabry disease based on the findings of zebra body. However, immunostaining showed few deposition of globotriaosylceramide in left ventricular myocardium, and gene mutations in the disease genes for hypertrophic cardiomyopathy (HCM), MYBPC3 and MYH6, were detected. Although the pathogenicity of the E66Q mutation cannot be ruled out, hypertrophic obstructive cardiomyopathy (HOCM) was more reasonable to explain the pathophysiology in the case. CONCLUSIONS: This is the confusable case of HOCM with Fabry disease with the GLA E66Q mutation. We have to take into consideration the possibility that some patients with the E66Q mutation may have similar histological findings of Fabry disease, and should be examed the possibility for harboring gene mutations associated with HCM.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/genética , Enfermedad de Fabry/genética , Mutación , Miocitos Cardíacos/ultraestructura , alfa-Galactosidasa/genética , Anciano , Biopsia , Cardiomiopatía Hipertrófica Familiar/diagnóstico , Cardiomiopatía Hipertrófica Familiar/enzimología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Análisis Mutacional de ADN , Ecocardiografía Doppler en Color , Enfermedad de Fabry/diagnóstico , Enfermedad de Fabry/enzimología , Enfermedad de Fabry/fisiopatología , Femenino , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Herencia , Humanos , Microscopía Electrónica , Miocitos Cardíacos/química , Linaje , Fenotipo , Valor Predictivo de las Pruebas , Trihexosilceramidas/análisis
16.
Am J Physiol Heart Circ Physiol ; 311(1): H125-36, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27199124

RESUMEN

Familial hypertrophic cardiomyopathy (HCM) is a disease of the sarcomere and may lead to hypertrophic, dilated, restrictive, and/or arrhythmogenic cardiomyopathy, congestive heart failure, or sudden cardiac death. We hypothesized that hearts from transgenic HCM mice harboring a mutant myosin heavy chain increase the energetic cost of contraction in a sex-specific manner. To do this, we assessed Ca(2+) sensitivity of tension and crossbridge kinetics in demembranated cardiac trabeculas from male and female wild-type (WT) and HCM hearts at an early time point (2 mo of age). We found a significant effect of sex on Ca(2+) sensitivity such that male, but not female, HCM mice displayed a decrease in Ca(2+) sensitivity compared with WT counterparts. The HCM transgene and sex significantly impacted the rate of force redevelopment by a rapid release-restretch protocol and tension cost by the ATPase-tension relationship. In each of these measures, HCM male trabeculas displayed a gain-of-function when compared with WT counterparts. In addition, cardiac remodeling measured by echocardiography, histology, morphometry, and posttranslational modifications demonstrated sex- and HCM-specific effects. In conclusion, female and male HCM mice display sex dimorphic crossbridge kinetics accompanied by sex- and HCM-dependent cardiac remodeling at the morphometric, histological, and cellular level.


Asunto(s)
Señalización del Calcio , Cardiomiopatía Hipertrófica Familiar/enzimología , Contracción Miocárdica , Miocardio/enzimología , Cadenas Pesadas de Miosina/metabolismo , Sarcómeros/enzimología , Adenosina Trifosfato/metabolismo , Animales , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/patología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Predisposición Genética a la Enfermedad , Hidrólisis , Cinética , Masculino , Ratones Transgénicos , Mutación , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Fenotipo , Fosforilación , Caracteres Sexuales , Factores Sexuales , Remodelación Ventricular
17.
Circ J ; 80(4): 938-49, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26960954

RESUMEN

BACKGROUND: Familial hypertrophic cardiomyopathy (HCM) is an autosomal dominant inherited disorder; mutations in at least 20 genes have been associated. Brugada syndrome (BrS) is an autosomal dominant inherited disorder caused by mutations mainly in theSCN5Agene. A new clinical entity that consists of HCM, typical electrical instability of BrS and sudden death (SD), is described. METHODS AND RESULTS: The family was constituted by 7 members, 4 of who presented clinical features of HCM and electrical instability of BrS. The clinical presentation of proband was ventricular fibrillation. All members were clinically evaluated by physical examination, 12-lead electrocardiography, 2-dimensional echocardiography, stress test, electrocardiogram Holter, flecainide test, and electrophysiological study. An integrated linkage analysis and next generation sequencing (NGS) approach was used to identify the causative mutation. Linkage with the α-tropomyosin (TPM1) gene on chromosome 15q22 was identified. The NGS study identified a missense mutation within theTPM1gene (c.574G>A; p.E192K), exactly located in a binding domain with polycystin-2 protein. No other pathogenic mutations were identified. CONCLUSIONS: This is the first report of an association between HCM and BrS, and the first to use a combined approach of linkage and NGS to identify a causative mutation in SD. The present study expands the clinical spectrum of disorders associated with theTPM1gene and may be useful to report novel mechanisms of electrical instability in HCM and BrS.


Asunto(s)
Síndrome de Brugada/genética , Cardiomiopatía Hipertrófica Familiar/genética , Cromosomas Humanos Par 15/genética , Ligamiento Genético , Tropomiosina/genética , Adulto , Síndrome de Brugada/fisiopatología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Electrocardiografía , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5/genética
18.
Radiographics ; 36(2): 335-54, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963450

RESUMEN

Hypertrophic cardiomyopathy (HCM) is a heterogeneous group of diseases related to sarcomere gene mutations exhibiting heterogeneous phenotypes with an autosomal dominant mendelian pattern of inheritance. The disorder is characterized by diverse phenotypic expressions and variable natural progression, which may range from dyspnea and/or syncope to sudden cardiac death. It is found across all racial groups and is associated with left ventricular hypertrophy in the absence of another systemic or cardiac disease. The management of HCM is based on a thorough understanding of the underlying morphology, pathophysiology, and clinical course. Imaging findings of HCM mirror the variable expressivity and penetrance heterogeneity, with the added advantage of diagnosis even in cases where a specific mutation may not yet be found. The diagnostic information obtained from imaging varies depending on the specific stage of HCM-phenotype manifestation, including the prehypertrophic, hypertrophic, and later stages of adverse remodeling into the burned-out phase of overt heart failure. However, subtle or obvious, these imaging findings become critical components in diagnosis, management, and follow-up of HCM patients. Although diagnosis of HCM traditionally relies on clinical assessment and transthoracic echocardiography, recent studies have demonstrated increased utility of multidetector computed tomography (CT) and particularly cardiac magnetic resonance (MR) imaging in diagnosis, phenotype differentiation, therapeutic planning, and prognostication. In this article, we provide an overview of the genetics, pathophysiology, and clinical manifestations of HCM, with the spectrum of imaging findings at MR imaging and CT and their contribution in diagnosis, risk stratification, and therapy.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Imagen por Resonancia Cinemagnética , Tomografía Computarizada Multidetector , Cardiomiopatías/diagnóstico , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Cardiomiopatía Hipertrófica Familiar/terapia , Diagnóstico Diferencial , Manejo de la Enfermedad , Progresión de la Enfermedad , Genes Dominantes , Estudios de Asociación Genética , Pruebas Genéticas , Insuficiencia Cardíaca/etiología , Tabiques Cardíacos/diagnóstico por imagen , Tabiques Cardíacos/patología , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/patología , Humanos , Hipertensión Pulmonar/etiología , Proteínas Musculares/genética , Penetrancia , Sarcómeros/ultraestructura , Obstrucción del Flujo Ventricular Externo/diagnóstico por imagen , Obstrucción del Flujo Ventricular Externo/etiología
19.
Science ; 351(6273): 617-21, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26912705

RESUMEN

Hypertrophic cardiomyopathy (HCM) is an inherited disease of heart muscle that can be caused by mutations in sarcomere proteins. Clinical diagnosis depends on an abnormal thickening of the heart, but the earliest signs of disease are hyperdynamic contraction and impaired relaxation. Whereas some in vitro studies of power generation by mutant and wild-type sarcomere proteins are consistent with mutant sarcomeres exhibiting enhanced contractile power, others are not. We identified a small molecule, MYK-461, that reduces contractility by decreasing the adenosine triphosphatase activity of the cardiac myosin heavy chain. Here we demonstrate that early, chronic administration of MYK-461 suppresses the development of ventricular hypertrophy, cardiomyocyte disarray, and myocardial fibrosis and attenuates hypertrophic and profibrotic gene expression in mice harboring heterozygous human mutations in the myosin heavy chain. These data indicate that hyperdynamic contraction is essential for HCM pathobiology and that inhibitors of sarcomere contraction may be a valuable therapeutic approach for HCM.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Bencilaminas/administración & dosificación , Miosinas Cardíacas/antagonistas & inhibidores , Cardiomiopatía Hipertrófica Familiar/tratamiento farmacológico , Contracción Miocárdica/efectos de los fármacos , Cadenas Pesadas de Miosina/antagonistas & inhibidores , Sarcómeros/efectos de los fármacos , Uracilo/análogos & derivados , Animales , Bencilaminas/química , Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar/patología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Heterocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos , Mutación , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Ratas , Uracilo/administración & dosificación , Uracilo/química
20.
Europace ; 18(4): 602-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25825456

RESUMEN

AIMS: Hypertrophic cardiomyopathy (HCM) mainly results from autosomal-dominant inherited single heterozygous mutations in cardiac sarcomere genes. Contributions of multiple gene mutations to disease heterogeneity in a three-generation family were investigated. METHODS: Clinical, electrocardiographic (ECG), and echocardiographic examination in members of a three-generation Chinese family was followed by exon and boarding intron analysis of 96 genes in the proband using second-generation sequencing. The identified mutations were confirmed by bi-directional Sanger sequencing in all family members and 300 healthy controls. RESULTS: Four missense mutations were detected in the family. These were two novel MYH7-H1717Q and MYLK2-K324E mutations accompanied by the KCNQ1-R190W and TMEM70-I147T mutations. The proband carried all four mutations and showed overlapping HCM and LQT1 phenotypes. Five family members each carried two mutations. Subject II-2 only carried TMEM70-I147T. MYH7-H1717Q and TMEM70-I147T came from the paternal side, whereas KCNQ1-R190W and MYLK2-K324E came from the maternal side. Left ventricle mass indices in MYH7-H1717Q carriers were significantly higher than in non-H1717Q carriers (90.05 ± 7.33 g/m(2), 63.20 ± 4.53 g/m(2), respectively, P < 0.01). Four KCNQ1-R190W carriers showed QTc intervals that were significantly more prolonged than those in non-R190W carriers (472.25 ± 16.18 and 408.50 ± 7.66 ms, respectively, P < 0.05). All MYLK2-K324E carriers showed inverted ECG T waves. The subject with only a TMEM70-I147T mutation showed normal ECG and echocardiographs, suggesting that this had less pathological effects at least in this family. CONCLUSIONS: We demonstrate dual LQT1 and HCM phenotypes in this multiple LQT1- and HCM-related gene mutation carrier family for the first time and suggest that LQT-related gene mutations associate with QT interval prolongation and/or arrhythmia in HCM patients.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar/genética , Heterocigoto , Canal de Potasio KCNQ1/genética , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Mutación Missense , Cadenas Pesadas de Miosina/genética , Quinasa de Cadena Ligera de Miosina/genética , Síndrome de Romano-Ward/genética , Adulto , Anciano , Pueblo Asiatico/genética , Cardiomiopatía Hipertrófica Familiar/diagnóstico , Cardiomiopatía Hipertrófica Familiar/etnología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Estudios de Casos y Controles , Niño , China , Análisis Mutacional de ADN , Ecocardiografía , Electrocardiografía , Femenino , Estudios de Asociación Genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Herencia , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Síndrome de Romano-Ward/diagnóstico , Síndrome de Romano-Ward/etnología , Síndrome de Romano-Ward/fisiopatología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA