Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
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.

2.
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.

3.
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
4.
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
5.
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
6.
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
7.
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
9.
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
10.
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
11.
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
12.
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
13.
Circ Heart Fail ; 4(1): 89-97, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21036890

RESUMEN

BACKGROUND: The BIO14.6 hamster provides a useful model of hereditary cardiomyopathies and muscular dystrophy. Previous δ-sarcoglycan (δSG) gene therapy (GT) studies were limited to neonatal and young adult animals and prevented the development of cardiac and skeletal muscle dysfunction. GT of a pseudophosphorylated mutant of phospholamban (S16EPLN) moderately alleviated the progression of cardiomyopathy. METHODS AND RESULTS: We treated 4-month-old BIO14.6 hamsters with established cardiac and skeletal muscle diseases intravenously with a serotype-9 adeno-associated viral vector carrying δSG alone or in combination with S16EPLN. Before treatment at age 14 weeks, the left ventricular fractional shortening by echocardiography was 31.3% versus 45.8% in normal hamsters. In a randomized trial, GT halted progression of left ventricular dilation and left ventricular dysfunction. Also, respiratory function improved. Addition of S16EPLN had no significant additional effects. δSG-GT prevented severe degeneration of the transverse tubular system in cardiomyocytes (electron tomography) and restored distribution of dystrophin and caveolin-3. All placebo-treated hamsters, except animals removed for the hemodynamic study, died with heart failure between 34 and 67 weeks of age. In the GT group, signs of cardiac and respiratory failure did not develop, and animals lived for 92 weeks or longer, an age comparable to that reported in normal hamsters. CONCLUSION: GT was highly effective in BIO14.6 hamsters even when given in late-stage disease, a finding that may carry implications for the future treatment of hereditary cardiac and muscle diseases in humans.


Asunto(s)
Fármacos Cardiovasculares/uso terapéutico , Terapia Genética , Insuficiencia Cardíaca/prevención & control , Enfermedades Musculares/prevención & control , Insuficiencia Respiratoria/prevención & control , Sarcoglicanos/uso terapéutico , Adenoviridae/genética , Animales , Cricetinae , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Insuficiencia Cardíaca/genética , Longevidad/genética , Masculino , Mesocricetus , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Miocardio/patología , Insuficiencia Respiratoria/genética , Músculos Respiratorios/patología , Sarcoglicanos/genética
15.
IEEE Eng Med Biol Mag ; 28(2): 46-57, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19349251

RESUMEN

There is a growing body of experimental evidence suggesting that the Ca(2+) signaling in ventricular myocytes is characterized by a high gradient near the cell membrane and a more uniform Ca(2+) distribution in the cell interior [1]--[7]. An important reason for this phenomenon might be that in these cells the t-tubular system forms a network of extracellular space, extending deep into the cell interior. This allows the electrical signal, that propagates rapidly along the cell membrane, to reach the vicinity of the sarcoplasmic reticulum (SR), where intracellular Ca(2+) required for myofilament activation is stored [1], [8]--[11]. Early studies of cardiac muscle showed that the t-tubules are found at intervals of about 2 lm along the longitudinal cell axis in close proximity to the Z-disks of the sarcomeres [12]. Subsequent studies have demonstrated that the t-tubular system has also longitudinal extensions [9]--[11], [13].


Asunto(s)
Canales de Calcio Tipo L/fisiología , Señalización del Calcio/fisiología , Ventrículos Cardíacos/citología , Modelos Cardiovasculares , Miocitos Cardíacos/fisiología , Algoritmos , Compuestos de Anilina/metabolismo , Animales , Calcio/metabolismo , Simulación por Computador , Análisis de Elementos Finitos , Colorantes Fluorescentes/metabolismo , Ratas , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/fisiología , Programas Informáticos , Xantenos/metabolismo
16.
Cardiovasc Res ; 83(1): 80-8, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19377068

RESUMEN

AIMS: Z-band alternatively spliced PDZ-motif protein (ZASP)/Cypher is a Z-disc component of which several dilated cardiomyopathy (DCM)-associated mutations have been reported. Most of the mutations were found in exons 4 and 10 of ZASP/Cypher gene LDB3 and both exons were expressed preferentially in the heart. The aim of this study was to investigate the functional alteration of ZASP/Cypher caused by the DCM-associated mutations. METHODS AND RESULTS: The yeast-two-hybrid method was used to identify the protein bound to a domain encoded by exon 4 of LDB3. Interaction of ZASP/Cypher with the binding protein was investigated in relation to the functional alterations caused by LDB3 mutations. Localization of the ZASP/Cypher-binding protein was examined at the cellular level in rat cardiomyocytes. Phosphoglucomutase 1 (PGM1), a metabolic enzyme involved in glycolysis and gluconeogenesis, was identified as a protein interacting with ZASP/Cypher. PGM1 bound to ZASP/Cypher at the domains encoded by exons 4 and 10. Two LDB3 mutations in exon 4 (Ser189Leu and Thr206Ile) and another mutation in exon 10 (Ile345Met) reduced the binding to PGM1. PGM1 showed diffuse localization in the cytoplasm of rat cardiomyocytes under standard culture conditions, and distribution at the Z-discs was observed under stressed culture conditions. Binding of endogenous PGM1 and ZASP/Cypher was found to be enhanced by stress in rat cardiomyocytes. CONCLUSION: ZASP/Cypher anchors PGM1 to Z-disc under conditions of stress. The impaired binding of PGM1 to ZASP/Cypher might be involved in the pathogenesis of DCM.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cardiomiopatía Dilatada/metabolismo , Miocitos Cardíacos/metabolismo , Fosfoglucomutasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células COS , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Células Cultivadas , Chlorocebus aethiops , Modelos Animales de Enfermedad , Exones/genética , Células HeLa , Humanos , Proteínas con Dominio LIM , Mutación/genética , Miocitos Cardíacos/patología , Fosfoglucomutasa/genética , Plásmidos/genética , Unión Proteica/genética , Ratas , Ratas Sprague-Dawley , Transfección
17.
J Cell Sci ; 122(Pt 7): 1005-13, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19295127

RESUMEN

In the current study, the three-dimensional (3D) topologies of dyadic clefts and associated membrane organelles were mapped in mouse ventricular myocardium using electron tomography. The morphological details and the distribution of membrane systems, including transverse tubules (T-tubules), junctional sarcoplasmic reticulum (SR) and vicinal mitochondria, were determined and presumed to be crucial for controlling cardiac Ca(2+) dynamics. The geometric complexity of T-tubules that varied in diameter with frequent branching was clarified. Dyadic clefts were intricately shaped and remarkably small (average 4.39x10(5) nm(3), median 2.81x10(5) nm(3)). Although a dyadic cleft of average size could hold maximum 43 ryanodine receptor (RyR) tetramers, more than one-third of clefts were smaller than the size that is able to package as many as 15 RyR tetramers. The dyadic clefts were also adjacent to one another (average end-to-end distance to the nearest dyadic cleft, 19.9 nm) and were distributed irregularly along T-tubule branches. Electron-dense structures that linked membrane organelles were frequently observed between mitochondrial outer membranes and SR or T-tubules. We, thus, propose that the topology of dyadic clefts and the neighboring cellular micro-architecture are the major determinants of the local control of Ca(2+) in the heart, including the establishment of the quantal nature of SR Ca(2+) releases (e.g. Ca(2+) sparks).


Asunto(s)
Señalización del Calcio , Microscopía Electrónica/métodos , Miocardio/metabolismo , Miocardio/ultraestructura , Orgánulos/ultraestructura , Animales , Separación Celular , Electrones , Ratones , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/ultraestructura , Modelos Biológicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/ultraestructura
18.
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
19.
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
20.
Biochem Biophys Res Commun ; 362(2): 281-7, 2007 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17716621

RESUMEN

Hypertrophic cardiomyopathy (HCM) is a cardiac disease characterized by left ventricular hypertrophy with diastolic dysfunction. Molecular genetic studies have revealed that HCM is caused by mutations in genes for sarcomere/Z-band components including titin/connectin and its associate proteins. However, disease-causing mutations can be found in about half of the patients, suggesting that other disease-causing genes remain to be identified. To explore a novel disease gene, we searched for obscurin gene (OBSCN) mutations in HCM patients, because obscurin interacts with titin/connectin. Two linked variants, Arg4344Gln and Ala4484Thr, were identified in a patient and functional analyses demonstrated that Arg4344Gln affected binding of obscurin to Z9-Z10 domains of titin/connectin, whereas Ala4484Thr did not. Myc-tagged obscurin showed that Arg4344Gln impaired obscurin localization to Z-band. These observations suggest that the obscurin abnormality may be involved in the pathogenesis of HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas Musculares/genética , Mutación , Adulto , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Cardiomiopatía Hipertrófica/patología , Células Cultivadas , Análisis Mutacional de ADN , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Inmunoprecipitación , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Factores de Intercambio de Guanina Nucleótido Rho , Sarcómeros/metabolismo , Homología de Secuencia de Aminoácido , Transfección , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA