Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Int Heart J ; 62(2): 359-366, 2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33678800

RESUMEN

Dilated cardiomyopathy (DCM) is a common cause of heart failure. TTN, which encodes titin protein, is a representative causative gene of DCM, and is presented mainly as a truncation variant. However, TTN truncation variants are also found in healthy individuals, and it is therefore important to evaluate the pathogenicity of each variant. In this study, we analyzed 67 cardiomyopathy-associated genes in a male Japanese patient who was hospitalized for recurrent severe heart failure and identified a novel truncation variant, TTN Ser17456Arg fs*14. This TTN truncation variant was located in the A-band region. Moreover, the patient's mother with heart failure harbored the same variant, whereas the father and brother without heart failure did not harbor the variant. To examine the functional changes associated with the truncation variant, H9c2 cells were subjected to genome editing to generate cells with a homologous truncation variant. The cells were differentiated using all-trans-retinoic acid, and the mRNA expression of skeletal actin and cardiac actin were found to be increased and decreased, respectively, consistent with known changes in patients with DCM or heart failure. In contrast, another cell with the titin truncation variant used as a control showed no changes in heart failure-related genes. In summary, we found a novel TTN truncation variant in familial DCM patients and confirmed its functional changes using a relatively simple cell model. The novel truncation variant was identified as a pathogenic and disease-causing mutation.


Asunto(s)
Cardiomiopatía Dilatada/genética , Conectina/genética , ADN/genética , Mutación , Miocitos Cardíacos/metabolismo , Función Ventricular Izquierda/fisiología , Biopsia , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Conectina/metabolismo , ADN/metabolismo , Análisis Mutacional de ADN , Femenino , Humanos , Japón , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/patología , Linaje
2.
Am J Physiol Heart Circ Physiol ; 314(6): H1192-H1202, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29451818

RESUMEN

Mutations in genes encoding components of the sarcomere cause cardiomyopathy, which is often associated with abnormal Ca2+ sensitivity of muscle contraction. We have previously shown that a heart-specific myosin light chain phosphatase small subunit (hHS-M21) increases the Ca2+ sensitivity of muscle contraction. The aim of the present study was to investigate the function of hHS-M21 in vivo and the causative role of abnormal Ca2+ sensitivity in cardiomyopathy. We generated transgenic mice with cardiac-specific overexpression of hHS-M21. We confirmed that hHS-M21 increased the Ca2+ sensitivity of cardiac muscle contraction in vivo, which was not followed by an increased phosphorylation of myosin light chain 2 isoforms. hHS-M21 transgenic mice developed severe systolic dysfunction with myocardial fibrosis and degeneration of cardiomyocytes in association with sinus bradycardia and atrioventricular conduction defect. The contractile dysfunction and cardiac fibrosis were improved by treatment with the Rho kinase inhibitor fasudil. Our findings suggested that the overexpression of hHS-M21 results in cardiac dysfunction and conduction disturbance via non-myosin light chain 2 phosphorylation-dependent regulation. NEW & NOTEWORTHY The present study is the first to develop mice with transgenic overexpression of a heart-specific myosin light chain phosphatase small subunit (hHS-M21) and to examine the effects of hHS-M21 on cardiac function. Elevation of hHS-M21 induced heart failure with myocardial fibrosis and degeneration of cardiomyocytes accompanied by supraventricular arrhythmias.


Asunto(s)
Arritmias Cardíacas/enzimología , Insuficiencia Cardíaca/enzimología , Miocitos Cardíacos/enzimología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Señalización del Calcio , Miosinas Cardíacas/metabolismo , Cardiomiopatías/enzimología , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Modelos Animales de Enfermedad , Fibrosis , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Frecuencia Cardíaca , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Miocárdica , Miocitos Cardíacos/patología , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/genética , Fenotipo , Fosforilación , Subunidades de Proteína , Regulación hacia Arriba , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda , Remodelación Ventricular , Quinasas Asociadas a rho/metabolismo
3.
J Hum Genet ; 63(12): 1273-1276, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30206291

RESUMEN

Hypertrophic cardiomyopathy (HCM) is characterized by unexplained left ventricular hypertrophy. This study aimed to reveal the clinical and genetic backgrounds of the unique HCM with mid-ventricular obstruction (HCM-MVO) subtype. We identified 34 patients with HCM-MVO in our cohort, and about half (47%) of these patients experienced adverse events. We analyzed 67 cardiomyopathy-associated genes in the patients. In total, 44% of patients with HCM-MVO carried the cardiomyopathy-associated genetic variant (CAGV) in 14 genes. Only 21% of patients carried HCM-associated CAGVs in major sarcomere-encoding genes, while 18% of patients carried CAGVs in dilated cardiomyopathy/arrhythmogenic right ventricular cardiomyopathy-associated genes. CAGVs were more frequent in patients with asymmetric septal hypertrophy (ASH) than in those without ASH. These findings suggest that HCM-MVO is a high-risk group and may have different etiologies from typical HCM.


Asunto(s)
Variación Genética , Hipertrofia Ventricular Izquierda/genética , Sarcómeros/genética , Anciano , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad
4.
J Hum Genet ; 63(2): 249-254, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29167554

RESUMEN

Hypertrophic cardiomyopathy is a heterogeneous disease caused by gene mutations. Most of the disease-causing mutations were found in the genes for sarcomeric proteins, but there are several cases carrying mutations in genes for extra-sarcomeric cytoskeletons. Desmin is a member of extra-sarcomeric cytoskeletons and plays an important role in muscle contraction. Mutations in the desmin gene cause various type of general myopathy and/or cardiomyopathy, known as desmin-related myopathies. We identified a novel desmin missense mutation, Thr219Pro, in the homozygous state in a patient, who first manifested with hypertrophic cardiomyopathy and later progressed to general myopathy. His parents were heterozygous for the mutation, but showed no clinical abnormality, suggesting the recessive inheritance of the mutation. We here report a severe phenotype of hypertrophic cardiomyopathy preceded the onset of general myopathy caused by a novel homozygous missense mutation in the 1B α-helix domain of desmin.


Asunto(s)
Cardiomiopatías/genética , Cardiomiopatía Hipertrófica/genética , Desmina/genética , Distrofias Musculares/genética , Mutación Missense , Adolescente , Adulto , Cardiomiopatías/patología , Cardiomiopatía Hipertrófica/patología , Femenino , Humanos , Masculino , Distrofias Musculares/patología , Dominios Proteicos
5.
J Hum Genet ; 63(9): 989-996, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29907873

RESUMEN

Hypertrophic cardiomyopathy (HCM) and restrictive cardiomyopathy (RCM) present a high risk for sudden cardiac death in pediatric patients. The aim of this study was to identify disease-associated genetic variants in Japanese patients with pediatric HCM and RCM. We analyzed 67 cardiomyopathy-associated genes in 46 HCM and 7 RCM patients diagnosed before 16 years of age using a next-generation sequencing system. We found that 78% of HCM and 71% of RCM patients carried disease-associated genetic variants. Disease-associated genetic variants were identified in 80% of HCM patients with a family history and in 77% of HCM patients with no apparent family history (NFH). MYH7 and/or MYBPC3 variants comprised 76% of HCM-associated variants, whereas troponin complex-encoding genes comprised 75% of the RCM-associated variants. In addition, 91% of HCM patients with implantable cardioverter-defibrillators and infant cases had NFH, and the 88% of HCM patients carrying disease-associated genetic variants were males who carried MYH7 or MYBPC3 variants. Moreover, two disease-associated LAMP2, one DES and one FHOD3 variants, were identified in HCM patients. In this study, pediatric HCM and RCM patients were found to carry disease-associated genetic variants at a high rate. Most of the variants were in MYH7 or MYPBC3 for HCM and TNNT2 or TNNI3 for RCM.


Asunto(s)
Cardiomegalia/genética , Cardiomiopatía Restrictiva/genética , Variación Genética , Proteínas Musculares/genética , Adolescente , Pueblo Asiatico , Niño , Preescolar , Femenino , Humanos , Japón , Masculino
6.
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
7.
J Hum Genet ; 60(10): 641-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26178432

RESUMEN

There is an overlap between the physiological cardiac remodeling associated with training in athletes, the so-called athlete's heart, and mild forms of hypertrophic cardiomyopathy (HCM), the most common hereditary cardiac disease. HCM is often accompanied by unfavorable outcomes including a sudden cardiac death in the adolescents. Because one of the initial signs of HCM is abnormality in electrocardiogram (ECG), athletes may need to monitor for ECG findings to prevent any unfavorable outcomes. HCM is caused by mutations in genes for sarcomere proteins, but there is no report on the systematic screening of gene mutations in athletes. One hundred and two genetically unrelated young Japanese athletes with abnormal ECG findings were the subjects for the analysis of four sarcomere genes, MYH7, MYBPC3, TNNT2 and TNNI3. We found that 5 out of 102 (4.9%) athletes carried mutations: a heterozygous MYH7 Glu935Lys mutation, a heterozygous MYBPC3 Arg160Trp mutation and another heterozygous MYBPC3 Thr1046Met mutation, all of which had been reported as HCM-associated mutations, in 1, 2 and 2 subjects, respectively. This is the first study of systematic screening of sarcomere gene mutations in a cohort of athletes with abnormal ECG, demonstrating the presence of sarcomere gene mutations in the athlete's heart.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomegalia , Proteínas Portadoras/genética , Electrocardiografía , Heterocigoto , Mutación Missense , Cadenas Pesadas de Miosina/genética , Sarcómeros/genética , Adolescente , Adulto , Sustitución de Aminoácidos , Atletas , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Femenino , Humanos , Masculino
8.
Hum Genome Var ; 11(1): 14, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38548731

RESUMEN

TNNI3 is a gene that causes hypertrophic cardiomyopathy (HCM). A 14-year-old girl who was diagnosed with nonobstructive HCM presented with cardiopulmonary arrest due to ventricular fibrillation. Genetic testing revealed a novel de novo heterozygous missense variant in TNNI3, NM_000363.5:c.583A>T (p.Ile195Phe), which was determined to be the pathogenic variant. The patient exhibited progressive myocardial fibrosis, left ventricular remodeling, and life-threatening arrhythmias. Genetic testing within families is useful for risk stratification in pediatric HCM patients.

10.
Hum Genome Var ; 9(1): 6, 2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35181673

RESUMEN

RBM20 is a disease-causing gene associated with dilated cardiomyopathy (DCM). The proband presented with the dilated phase of hypertrophic cardiomyopathy (HCM), and the mother also suffered from HCM. A missense variant of RBM20, p.Arg636His, previously reported as pathogenic in several families with DCM, was found in both the proband and the mother. Therefore, RBM20 p.Arg636His could be the causative variant for this familial HCM, and RBM20 might be a novel causative gene for HCM.

11.
Keio J Med ; 69(4): 77-87, 2020 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-32224552

RESUMEN

Hypertrophic cardiomyopathy (HCM) is an intractable disease that causes heart failure mainly due to unexplained severe cardiac hypertrophy and diastolic dysfunction. HCM, which occurs in 0.2% of the general population, is the most common cause of sudden cardiac death in young people. HCM has been studied extensively using molecular genetic approaches. Genes encoding cardiac ß-myosin heavy chain, cardiac myosin-binding protein C, and troponin complex, which were originally identified as causative genes, were subsequently reported to be frequently implicated in HCM. Indeed, HCM has been considered a disease of sarcomere gene mutations. However, fewer than half of patients with HCM have mutations in sarcomere genes. The others have been documented to have mutations in cardiac proteins in various other locations, including the Z disc, sarcoplasmic reticulum, plasma membrane, nucleus, and mitochondria. Next-generation sequencing makes it possible to detect mutations at high throughput, and it has become increasingly common to identify multiple cardiomyopathy-causing gene mutations in a single HCM patient. Elucidating how mutations in different genes contribute to the disease pathophysiology will be a challenge. In studies using animal models, sarcomere mutations generally tend to increase myocardial Ca2+ sensitivity, and some mutations increase the activity of myosin ATPase. Clinical trials of drugs to treat HCM are ongoing, and further new therapies based on pathophysiological analyses of the causative genes are eagerly anticipated.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Cadenas Pesadas de Miosina/metabolismo , Troponina/metabolismo , Miosinas Ventriculares/metabolismo , Animales , Calcio/metabolismo , Miosinas Cardíacas/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Estudios de Asociación Genética , Genoma , Humanos , Ratones , Mitocondrias/metabolismo , Mutación , Miocardio/metabolismo , Sarcómeros/metabolismo , Retículo Sarcoplasmático/metabolismo
12.
J Hum Genet ; 54(2): 115-21, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19165230

RESUMEN

Bradycardia is a trigger of ventricular arrhythmias in patients with arrhythmia including Brugada syndrome and long QT syndrome. The HCN4 channel controls the heart rate, and its mutations predispose to inherited sick sinus syndrome and long QT syndrome associated with bradycardia. We found a 4 base-insertion at the splice donor site of the HCN4 gene in a patient with idiopathic ventricular tachycardia, which was supposed to generate a truncated channel. To investigate the role of the HCN4 channel in ventricular arrhythmia, we introduced a ventricular action potential of I(f) channel produced by HCN4 in a computer simulation model and found that the I(f) channel generated a leaky outward current during the plateau phase of ventricular action potential. Currents through the I(f) channel were suggested to contribute to the shortening of the action potential duration and the prevention of early after-depolarization in bradycardia. These observations suggested that the HCN4 channel played a preventive role in triggering bradycardia-induced ventricular arrhythmias.


Asunto(s)
Arritmias Cardíacas/prevención & control , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Ventrículos Cardíacos/patología , Proteínas Musculares/genética , Potenciales de Acción , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Secuencia de Bases , Simulación por Computador , Ventrículos Cardíacos/fisiopatología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Activación del Canal Iónico , Modelos Biológicos , Datos de Secuencia Molecular , Mutación/genética , Canales de Potasio , Empalme del ARN/genética
13.
J Struct Biol ; 164(3): 304-13, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18835449

RESUMEN

A general framework of image-based geometric processing is presented to bridge the gap between three-dimensional (3D) imaging that provides structural details of a biological system and mathematical simulation where high-quality surface or volumetric meshes are required. A 3D density map is processed in the order of image pre-processing (contrast enhancement and anisotropic filtering), feature extraction (boundary segmentation and skeletonization), and high-quality and realistic surface (triangular) and volumetric (tetrahedral) mesh generation. While the tool-chain described is applicable to general types of 3D imaging data, the performance is demonstrated specifically on membrane-bound organelles in ventricular myocytes that are imaged and reconstructed with electron microscopic (EM) tomography and two-photon microscopy (T-PM). Of particular interest in this study are two types of membrane-bound Ca(2+)-handling organelles, namely, transverse tubules (T-tubules) and junctional sarcoplasmic reticulum (jSR), both of which play an important role in regulating the excitation-contraction (E-C) coupling through dynamic Ca(2+) mobilization in cardiomyocytes.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional/métodos , Miocitos Cardíacos/ultraestructura , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/ultraestructura , Animales , Diagnóstico por Imagen , Ventrículos Cardíacos/metabolismo , Matemática , Células Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Orgánulos/metabolismo , Sarcolema/metabolismo
14.
Circulation ; 116(5): 515-25, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-17646580

RESUMEN

BACKGROUND: Extracellular matrix proteins, such as laminins, and endothelial cells are known to influence cardiomyocyte performance; however, the underlying molecular mechanisms remain poorly understood. METHODS AND RESULTS: We used a forward genetic screen in zebrafish to identify novel genes required for myocardial function and were able to identify the lost-contact (loc) mutant, which encodes a nonsense mutation in the integrin-linked kinase (ilk) gene. This loc/ilk mutant is associated with a severe defect in cardiomyocytes and endothelial cells that leads to severe myocardial dysfunction. Additional experiments revealed the epistatic regulation between laminin-alpha4 (Lama4), integrin, and Ilk, which led us to screen for mutations in the human ILK and LAMA4 genes in patients with severe dilated cardiomyopathy. We identified 2 novel amino acid residue-altering mutations (2828C>T [Pro943Leu] and 3217C>T [Arg1073X]) in the integrin-interacting domain of the LAMA4 gene and 1 mutation (785C>T [Ala262Val]) in the ILK gene. Biacore quantitative protein/protein interaction data, which have been used to determine the equilibrium dissociation constants, point to the loss of integrin-binding capacity in case of the Pro943Leu (Kd=5+/-3 micromol/L) and Arg1073X LAMA4 (Kd=1+/-0.2 micromol/L) mutants compared with the wild-type LAMA4 protein (Kd=440+/-20 nmol/L). Additional functional data point to the loss of endothelial cells in affected patients as a direct consequence of the mutant genes, which ultimately leads to heart failure. CONCLUSIONS: This is the first report on mutations in the laminin, integrin, and ILK system in human cardiomyopathy, which has consequences for endothelial cells as well as for cardiomyocytes, thus providing a new genetic basis for dilated cardiomyopathy in humans.


Asunto(s)
Cardiomiopatía Dilatada/genética , Células Endoteliales/patología , Laminina/genética , Mutación Missense , Miocitos Cardíacos/patología , Mutación Puntual , Proteínas Serina-Treonina Quinasas/genética , Adulto , Sustitución de Aminoácidos , Animales , Células COS , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Adhesión Celular , Chlorocebus aethiops , Mapeo Cromosómico , Codón sin Sentido , Análisis Mutacional de ADN , Embrión no Mamífero/patología , Epigénesis Genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Corazón/embriología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Humanos , Integrinas/metabolismo , Laminina/fisiología , Masculino , Persona de Mediana Edad , Modelos Moleculares , Miocardio/patología , Oligonucleótidos Antisentido/toxicidad , Linaje , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/fisiología , Estructura Terciaria de Proteína , Transfección , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
15.
Sci Rep ; 8(1): 8970, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29895960

RESUMEN

RBM20 is a major regulator of heart-specific alternative pre-mRNA splicing of TTN encoding a giant sarcomeric protein titin. Mutation in RBM20 is linked to autosomal-dominant familial dilated cardiomyopathy (DCM), yet most of the RBM20 missense mutations in familial and sporadic cases were mapped to an RSRSP stretch in an arginine/serine-rich region of which function remains unknown. In the present study, we identified an R634W missense mutation within the stretch and a G1031X nonsense mutation in cohorts of DCM patients. We demonstrate that the two serine residues in the RSRSP stretch are constitutively phosphorylated and mutations in the stretch disturb nuclear localization of RBM20. Rbm20 S637A knock-in mouse mimicking an S635A mutation reported in a familial case showed a remarkable effect on titin isoform expression like in a patient carrying the mutation. These results revealed the function of the RSRSP stretch as a critical part of a nuclear localization signal and offer the Rbm20 S637A mouse as a good model for in vivo study.


Asunto(s)
Cardiomiopatía Dilatada , Mutación Missense , Señales de Localización Nuclear , Empalme del ARN , Proteínas de Unión al ARN , Adolescente , Adulto , Sustitución de Aminoácidos , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Fosforilación/genética , Dominios Proteicos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
17.
J Am Coll Cardiol ; 44(11): 2192-201, 2004 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-15582318

RESUMEN

OBJECTIVES: We sought to explore the relationship between a Tcap gene (TCAP) abnormality and cardiomyopathy. BACKGROUND: Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) cause severe heart failure and sudden death. Recent genetic investigations have revealed that mutations of genes encoding Z-disc components, including titin and muscle LIM protein (MLP), are the primary cause of both HCM and DCM. The Z-disc plays a role in establishing the mechanical coupling of sarcomeric contraction and stretching, with the titin/Tcap/MLP complex serving as a mechanical stretch sensor. Tcap interacts with the calsarcin, which tethers the calcineurin to the Z-disc. METHODS: The TCAP was analyzed in 346 patients with HCM (236 familial and 110 sporadic cases) and 136 patients with DCM (34 familial and 102 sporadic cases). Two different in vitro qualitative assays-yeast two-hybrid and glutathion S-transferase pull-down competition-were performed in order to investigate functional changes in Tcap's interaction with MLP, titin, and calsarcin-1 caused by the identified mutations and a reported DCM-associated mutation, R87Q. RESULTS: Two TCAP mutations, T137I and R153H, were found in patients with HCM, and another TCAP mutation, E132Q, was identified in a patient with DCM. It was demonstrated by the qualitative assays that the HCM-associated mutations augment the ability of Tcap to interact with titin and calsarcin-1, whereas the DCM-associated mutations impair the interaction of Tcap with MLP, titin, and calsarcin-1. CONCLUSIONS: These observations suggest that the difference in clinical phenotype (HCM or DCM) may be correlated with the property of altered binding among the Z-disc components.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica/genética , Proteínas Musculares/genética , Pueblo Asiatico/genética , Proteínas Portadoras/genética , Conectina , Cartilla de ADN , Glutatión , Humanos , Corea (Geográfico) , Proteínas con Dominio LIM , Penetrancia , Unión Proteica , Proteínas Quinasas/genética , Alineación de Secuencia
18.
Int J Cardiol ; 102(1): 39-45, 2005 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-15939097

RESUMEN

BACKGROUND: Acute aortic dissection (AAD) is sometimes complicated by respiratory failure due to severe lung oxygenation impairment. Systemic activation of inflammatory system after aortic injury may play some roles in the development of this complication. The aim of this study was to determine the significance of serum C-reactive protein (CRP) elevation in the development of oxygenation impairment and clinical outcome with distal type AAD. METHODS AND RESULTS: A total of 61 patients, who were admitted with distal type AAD within 24 h from the onset, were examined. Serum CRP levels, white blood cell (WBC) counts and body temperature were measured serially for at least 4 days. Oxygenation impairment, defined as the lowest PaO2/FIO2 ratio < or = 200 mmHg, was noted in 31 patients (51%). In patients with oxygenation impairment, peak CRP levels (20.7+/-7.9 vs. 12.7+/-3.8 mg/dl, P < 0.001), peak WBC counts (14,600+/-3600 vs. 11,800+/-4300/mm3, P = 0.008) and body temperature (38.4+/-0.5 vs. 38.0+/-0.6 degrees C, P = 0.004) were significantly higher than those without. Peak CRP level was inversely correlated with the lowest PaO2/FIO2 (P < 0.001). Patients who underwent urgent surgical treatment and/or died in the hospital had higher peak CRP levels (25.1+/-12.3 vs. 16.1+/-7.4 mg/dl, P = 0.010) than those who did not. Multivariate analysis revealed that a peak CRP level > or = 15 mg/dl (relative risk = 12.6, P < 0.001) was an independent determinant of the development of oxygenation impairment. CONCLUSION: The greater serum CRP elevation after distal type AAD was associated with a higher incidence of oxygenation impairment and poor clinical outcome. Systemic activation of the inflammatory system after aortic injury may play an important role in the development of oxygenation impairment.


Asunto(s)
Aneurisma de la Aorta Torácica/sangre , Disección Aórtica/sangre , Proteína C-Reactiva/metabolismo , Oxígeno/sangre , Síndrome de Dificultad Respiratoria/etiología , Enfermedad Aguda , Adulto , Anciano , Anciano de 80 o más Años , Disección Aórtica/complicaciones , Disección Aórtica/diagnóstico por imagen , Aneurisma de la Aorta Torácica/complicaciones , Aneurisma de la Aorta Torácica/diagnóstico por imagen , Biomarcadores/sangre , Análisis de los Gases de la Sangre , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Síndrome de Dificultad Respiratoria/sangre , Síndrome de Dificultad Respiratoria/epidemiología , Factores de Riesgo , Tasa de Supervivencia , Tomografía Computarizada por Rayos X
19.
Int J Cardiol ; 99(2): 343-5, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15749201

RESUMEN

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an autosomal dominant inherited disorder characterized by adrenergic induced polymorphic ventricular tachycardias and associated with sudden cardiac death. The human cardiac ryanodine receptor gene (RyR2) was linked to CPVT. A 20-year-old male was referred to our hospital because of recurrent syncope after physical and emotional stress. Routine cardiac examinations including catheterization revealed no structural abnormality. Exercise on treadmill induced premature ventricular contraction in bigeminy and bidirectional ventricular tachycardia was induced during isoproterenol infusion. Beta-blocking drug was effective in suppressing the arrhythmias. We performed genetic screening by PCR-SSCP method followed by DNA sequencing, and a novel missense mutation R2401H in RyR2 located in FKBP12.6 binding region was identified. This mutation was not detected in 190 healthy controls. Since FKBP12.6 plays a critical role in Ca channel gating, the R2401H mutation can be expected to alter Ca-induced Ca release and E-C coupling resulting in CPVT. This is the first report of RyR2 mutation in CPVT patient from Asia including Japan.


Asunto(s)
Agonistas Adrenérgicos beta/efectos adversos , Isoproterenol/efectos adversos , Mutación Missense , Canal Liberador de Calcio Receptor de Rianodina/genética , Taquicardia Ventricular/genética , Proteínas de Unión a Tacrolimus/genética , Adulto , Prueba de Esfuerzo/efectos adversos , Prueba de Esfuerzo/métodos , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Canal Liberador de Calcio Receptor de Rianodina/sangre , Taquicardia Ventricular/sangre , Taquicardia Ventricular/inducido químicamente
20.
FEBS Lett ; 574(1-3): 145-50, 2004 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-15358555

RESUMEN

We identified a novel mutation Ala178fs/105 missing S3-S6 and C-terminus portions of KCNQ1 channel. Ala178fs/105-KCNQ1 expressed in COS-7 cells demonstrated no current expression. Co-expression with wild-type (WT) revealed a dominant-negative effect, which suggests the formation of hetero-multimer by mutant and WT. Confocal laser microscopy displayed intracellular retention of Ala178fs/105-KCNQ1 protein. Co-expression of the mutant and WT also increased intracellular retention of channel protein compared to WT alone. Our findings suggest a novel mechanism for LQT1 that the truncated S1-S2 KCNQ1 mutant forms hetero-multimer and cause a dominant-negative effect due to trafficking defect.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Adolescente , Animales , Secuencia de Bases , Células COS , Cartilla de ADN , Femenino , Genes Dominantes , Humanos , Canales de Potasio KCNQ , Canal de Potasio KCNQ1 , Microscopía Confocal , Canales de Potasio/genética , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA