Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Eur Heart J ; 37(18): 1469-75, 2016 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-26429810

RESUMEN

AIM: Ventricular fibrillation (VF), the main cause of sudden cardiac death (SCD), occurs most frequently in the acute phase of myocardial infarction: a certain fraction of VF, however, develops in an apparently healthy heart, referred as idiopathic VF. The contribution of perturbation in the fast conduction system in the ventricle, the His-Purkinje system, for idiopathic VF has been implicated, but the underlying mechanism remains unknown. Irx3/IRX3 encodes a transcription factor specifically expressed in the His-Purkinje system in the heart. Genetic deletion of Irx3 provides a mouse model of ventricular fast conduction disturbance without anatomical or contraction abnormalities. The aim of this study was to examine the link between perturbed His-Purkinje system and idiopathic VF in Irx3-null mice, and to search for IRX3 genetic defects in idiopathic VF patients in human. METHODS AND RESULTS: Telemetry electrocardiogram recording showed that Irx3-deleted mice developed frequent ventricular tachyarrhythmias mostly at night. Ventricular tachyarrhythmias were enhanced by exercise and sympathetic nerve activation. In human, the sequence analysis of IRX3 exons in 130 probands of idiopathic VF without SCN5A mutations revealed two novel IRX3 mutations, 1262G>C (R421P) and 1453C>A (P485T). Ventricular fibrillation associated with physical activities in both probands with IRX3 mutations. In HL-1 cells and neonatal mouse ventricular myocytes, IRX3 transfection up-regulated SCN5A and connexin-40 mRNA, which was attenuated by IRX3 mutations. CONCLUSION: IRX3 genetic defects and resultant functional perturbation in the His-Purkinje system are novel genetic risk factors of idiopathic VF, and would improve risk stratification and preventive therapy for SCD in otherwise healthy hearts.


Asunto(s)
Arritmias Cardíacas , Animales , Muerte Súbita Cardíaca , Sistema de Conducción Cardíaco , Proteínas de Homeodominio , Humanos , Ratones , Factores de Transcripción , Fibrilación Ventricular
3.
Int Heart J ; 57(4): 507-10, 2016 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-27385602

RESUMEN

We report the case of a 66 year-old woman with chronic atrial fibrillation, hypertrophic cardiomyopathy (HCM), and spinocerebellar atrophy (SCA). Her mother and first-born son had died of heart disease at the ages of 65 and 16 years, respectively. Four of her 8 siblings had died suddenly of unknown cause or of heart disease, and 2 others of cerebral infarction by the 7th decade. Genetic testing revealed that she had a novel mutation (c. 482C > A, p. Ala161Asp) in the troponin I gene (TNNI3), and no abnormality of the GAA repeat in the frataxin gene. Her older brother with SCA but without HCM was also analyzed, with no abnormality noted in either gene. The Ala161Asp mutation in TNNI3 was implicated in the pathogenesis of her HCM, though an association between HCM and SCA was not revealed.


Asunto(s)
Fibrilación Atrial/genética , Cardiomiopatía Hipertrófica/genética , Mutación , Ataxias Espinocerebelosas/genética , Troponina I/genética , Anciano , Fibrilación Atrial/complicaciones , Cardiomiopatía Hipertrófica/complicaciones , Femenino , Humanos , Linaje , Ataxias Espinocerebelosas/complicaciones
4.
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
5.
Hum Mol Genet ; 21(5): 1037-48, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22090424

RESUMEN

The LMNA gene encodes lamin A/C intermediate filaments that polymerize beneath the nuclear membrane, and are also found in the nucleoplasm in an uncharacterized assembly state. They are thought to have structural functions and regulatory roles in signaling pathways via interaction with transcription factors. Mutations in LMNA have been involved in numerous inherited human diseases, including severe congenital muscular dystrophy (L-CMD). We created the Lmna(ΔK32) knock-in mouse harboring a L-CMD mutation. Lmna(ΔK32/ΔK32) mice exhibited striated muscle maturation delay and metabolic defects, including reduced adipose tissue and hypoglycemia leading to premature death. The level of mutant proteins was markedly lower in Lmna(ΔK32/ΔK32), and while wild-type lamin A/C proteins were progressively relocated from nucleoplasmic foci to the nuclear rim during embryonic development, mutant proteins were maintained in nucleoplasmic foci. In the liver and during adipocyte differentiation, expression of ΔK32-lamin A/C altered sterol regulatory element binding protein 1 (SREBP-1) transcriptional activities. Taken together, our results suggest that lamin A/C relocation at the nuclear lamina seems important for tissue maturation potentially by releasing its inhibitory function on transcriptional factors, including but not restricted to SREBP-1. And importantly, L-CMD patients should be investigated for putative metabolic disorders.


Asunto(s)
Núcleo Celular/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Enfermedades Metabólicas/genética , Músculo Esquelético/crecimiento & desarrollo , Lámina Nuclear/metabolismo , Adipocitos/citología , Adipogénesis , Animales , Animales Recién Nacidos , Embrión de Mamíferos , Técnicas de Sustitución del Gen , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/metabolismo , Corazón/crecimiento & desarrollo , Lamina Tipo B/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Ratones , Mortalidad Prematura , Músculo Esquelético/anatomía & histología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Miocitos Cardíacos/citología , Tamaño de los Órganos , Fenotipo , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transcripción Genética
6.
Hum Mol Genet ; 21(9): 2039-53, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22286171

RESUMEN

Abnormalities in Z-disc proteins cause hypertrophic (HCM), dilated (DCM) and/or restrictive cardiomyopathy (RCM), but disease-causing mechanisms are not fully understood. Myopalladin (MYPN) is a Z-disc protein expressed in striated muscle and functions as a structural, signaling and gene expression regulating molecule in response to muscle stress. MYPN was genetically screened in 900 patients with HCM, DCM and RCM, and disease-causing mechanisms were investigated using comparative immunohistochemical analysis of the patient myocardium and neonatal rat cardiomyocytes expressing mutant MYPN. Cardiac-restricted transgenic (Tg) mice were generated and protein-protein interactions were evaluated. Two nonsense and 13 missense MYPN variants were identified in subjects with DCM, HCM and RCM with the average cardiomyopathy prevalence of 1.66%. Functional studies were performed on two variants (Q529X and Y20C) associated with variable clinical phenotypes. Humans carrying the Y20C-MYPN variant developed HCM or DCM, whereas Q529X-MYPN was found in familial RCM. Disturbed myofibrillogenesis with disruption of α-actinin2, desmin and cardiac ankyrin repeat protein (CARP) was evident in rat cardiomyocytes expressing MYPN(Q529X). Cardiac-restricted MYPN(Y20C) Tg mice developed HCM and disrupted intercalated discs, with disturbed expression of desmin, desmoplakin, connexin43 and vinculin being evident. Failed nuclear translocation and reduced binding of Y20C-MYPN to CARP were demonstrated using in vitro and in vivo systems. MYPN mutations cause various forms of cardiomyopathy via different protein-protein interactions. Q529X-MYPN causes RCM via disturbed myofibrillogenesis, whereas Y20C-MYPN perturbs MYPN nuclear shuttling and leads to abnormal assembly of terminal Z-disc within the cardiac transitional junction and intercalated disc.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica Familiar/genética , Proteínas Musculares/genética , Mutación , Animales , Animales Recién Nacidos , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Hipertrófica Familiar/patología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Estudios de Casos y Controles , Codón sin Sentido , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Proteínas Musculares/fisiología , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Mutación Missense , Miocardio/patología , Miocitos Cardíacos/ultraestructura , Proteínas Nucleares/metabolismo , Linaje , Fenotipo , Unión Proteica , Ratas , Ratas Mutantes , Ratas Sprague-Dawley , Proteínas Represoras/metabolismo
7.
Basic Res Cardiol ; 108(3): 349, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23572067

RESUMEN

Hypertrophic cardiomyopathy (HCM) is a myocardial disease associated with mutations in sarcomeric genes. Three mutations were found in ANKRD1, encoding ankyrin repeat domain 1 (ANKRD1), a transcriptional co-factor located in the sarcomere. In the present study, we investigated whether expression of HCM-associated ANKRD1 mutations affects contraction parameters after gene transfer in engineered heart tissues (EHTs). EHTs were generated from neonatal rat heart cells and were transduced with adeno-associated virus encoding GFP or myc-tagged wild-type (WT) or mutant (P52A, T123M, or I280V) ANKRD1. Contraction parameters were analyzed from day 8 to day 16 of culture, and evaluated in the absence or presence of the proteasome inhibitor epoxomicin for 24 h. Under standard conditions, only WT- and T123M-ANKRD1 were correctly incorporated in the sarcomere. T123M-ANKRD1-transduced EHTs exhibited higher force and velocities of contraction and relaxation than WT- P52A- and I280V-ANKRD1 were highly unstable, not incorporated into the sarcomere, and did not induce contractile alterations. After epoxomicin treatment, P52A and I280V were both stabilized and incorporated into the sarcomere. I280V-transduced EHTs showed prolonged relaxation. These data suggest different impacts of ANKRD1 mutations on cardiomyocyte function: gain-of-function for T123M mutation under all conditions and dominant-negative effect for the I280V mutation which may come into play only when the proteasome is impaired.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Proteínas Musculares/genética , Mutación , Contracción Miocárdica/genética , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Proteínas Represoras/genética , Ingeniería de Tejidos , Animales , Animales Recién Nacidos , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/fisiopatología , Células Cultivadas , Dependovirus/genética , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Vectores Genéticos , Genotipo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Musculares/metabolismo , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Proteínas Nucleares/metabolismo , Oligopéptidos/farmacología , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Ratas , Proteínas Represoras/metabolismo , Factores de Tiempo , Ingeniería de Tejidos/métodos , Transducción Genética , Transfección
8.
J Autoimmun ; 47: 25-33, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23953137

RESUMEN

HLA locus contains immune-related genes and genetically regulates immune responses against both foreign- and self-antigens in humans. Inhibitor of κB-like protein (IκBL), encoded by HLA-linked NFKBIL1, is a protein of unknown function, while genetic variations in NFKBIL1 are known to associate with the susceptibility to inflammatory and/or autoimmune diseases. In this study, we found that IκBL suppressed exon exclusion in alternative splicing of human immune-related genes such as CD45. Yeast-two-hybrid screening and immunoprecipitation assay revealed molecular association of IκBL with CLK1, a serine/threonine and tyrosine kinase, which plays a role in the alternative splicing. Unexpectedly, we found that the regulation of alternative splicing in CD45 by IκBL was independent from the kinase activity of CLK1. On the other hand, it was demonstrated that an SR protein, ASF/SF2, bound both IκBL and CLK1 at the RNA-recognition motifs of ASF/SF2, implying a competition of IκBL and CLK1 on SR protein. In addition, IκBL was found to regulate the CLK1-dependent synthesis of M2 RNA, a splice variant of influenza A virus M gene. These observations suggest a functional involvement of IκBL in the regulation of alternative splicing in both human and viral genes, which is a novel link of HLA locus to the regulation of immunity and infection in humans.


Asunto(s)
Empalme Alternativo , Antígenos HLA/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Virus de la Influenza A/genética , Antígenos Comunes de Leucocito/genética , Proteínas de la Matriz Viral/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Células COS , Chlorocebus aethiops , Células HEK293 , Células HeLa , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Empalme Serina-Arginina
9.
Circ J ; 77(12): 2990-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24088304

RESUMEN

BACKGROUND: Dilated cardiomyopathy (DCM) is characterized by a dilated left ventricular cavity with systolic dysfunction manifested by heart failure. It has been revealed that mutations in genes for cytoskeleton or sarcomere proteins cause DCM. However, the disease-causing mutations can be found only in far less than half of patients with a family history, indicating that there should be other disease genes for DCM. Formin homology 2 domain containing 3 (FHOD3) is a sarcomeric protein expressed in the heart that plays an essential role in sarcomere organization during myofibrillogenesis. The purpose of this study was to explore a possible novel disease gene for DCM. METHODS AND RESULTS: We analyzed 48 Japanese familial DCM patients for mutations in FHOD3, and a missense variant, Tyr1249Asn, which was predicted to modify the 3D structure and damage protein function, was found in a case with adult-onset DCM. Functional studies revealed that the DCM-associated mutation significantly reduced the ability to induce actin dynamics-dependent activation of serum response factor, although no remarkable change in the cellular localization was induced in neonatal rat cardiomyocytes transfected with a mutant construct of FHOD3. CONCLUSIONS: The DCM-associated FHOD3 variant may cause DCM by interfering with actin filament assembly.


Asunto(s)
Cardiomiopatía Dilatada , Proteínas de Microfilamentos , Proteínas Musculares , Mutación Missense , Miocitos Cardíacos/metabolismo , Factor de Respuesta Sérica , Adulto , Sustitución de Aminoácidos , Animales , Pueblo Asiatico , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Células Cultivadas , Forminas , Humanos , Japón , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Ratas , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo
10.
Circ J ; 77(4): 959-67, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23257389

RESUMEN

BACKGROUND: Brugada syndrome (BrS) is characterized by specific alterations on ECG in the right precordial leads and associated with ventricular arrhythmia that may manifest as syncope or sudden cardiac death. The major causes of BrS are mutations in SCN5A for a large subunit of the sodium channel, Nav1.5, but a mutation in SCN3B for a small subunit of sodium channel, Navß3, has been recently reported in an American patient. METHODS AND RESULTS: A total of 181 unrelated BrS patients, 178 Japanese and 3 Koreans, who had no mutations in SCN5A, were examined for mutations in SCN3B by direct sequencing of all exons and adjacent introns. A mutation, Val110Ile, was identified in 3 of 178 (1.7%) Japanese patients, but was not found in 480 Japanese controls. The SCN3B mutation impaired the cytoplasmic trafficking of Nav1.5, the cell surface expression of which was decreased in transfected cells. Whole-cell patch clamp recordings of the transfected cells revealed that the sodium currents were significantly reduced by the SCN3B mutation. CONCLUSIONS: The Val110Ile mutation of SCN3B is a relatively common cause of SCN5A-negative BrS in Japan, which has a reduced sodium current because of the loss of cell surface expression of Nav1.5.


Asunto(s)
Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Subunidad beta-3 de Canal de Sodio Activado por Voltaje/genética , Adolescente , Adulto , Anciano , Sustitución de Aminoácidos , Animales , Pueblo Asiatico , Línea Celular , Niño , Femenino , Humanos , Japón , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5/genética , Transporte de Proteínas/genética , Subunidad beta-3 de Canal de Sodio Activado por Voltaje/metabolismo
11.
Circ J ; 76(2): 453-61, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22112859

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy (HCM), which is inherited as an autosomal dominant trait, is the most prevalent hereditary cardiac disease. Although there are several reports on the systematic screening of mutations in the disease-causing genes in European and American populations, only limited information is available for Asian populations, including Japanese. METHODS AND RESULTS: Genetic screening of disease-associated mutations in 8 genes for sarcomeric proteins, MYH7, MYBPC3, MYL2, MYL3, TNNT2, TNNI3, TPM1, and ACTC, was performed by direct sequencing in 112 unrelated Japanese proband patients with familial HCM; 37 different mutations, including 13 novel ones in 5 genes, MYH7, MYBPC3, TNNT2, TNNI3, and TPM1, were identified in 49 (43.8%) patients. Among them, 3 carried compound heterozygous mutations in MYBPC3 or TNNT2. The frequency of patients carrying the MYBPC3, MYH7, and TNNT2 mutations were 19.6%, 10.7%, and 8.9%, respectively, and the most frequently affected genes in the northeastern and southwestern parts of Japan were MYBPC3 and MYH7, respectively. Several mutations were found in multiple unrelated proband patients, for which the geographic distribution suggested founder effects of the mutations. CONCLUSIONS: This study demonstrated the frequency and distribution of mutations in a large cohort of familial HCM in Japan.


Asunto(s)
Pueblo Asiatico/genética , Pueblo Asiatico/estadística & datos numéricos , Cardiomiopatía Hipertrófica Familiar/etnología , Cardiomiopatía Hipertrófica Familiar/genética , Sarcómeros/genética , Actinas/genética , Adulto , Anciano , Miosinas Cardíacas/genética , Proteínas Portadoras/genética , Femenino , Geografía , Humanos , Japón/epidemiología , Quinasas Quinasa Quinasa PAM/genética , Masculino , Persona de Mediana Edad , Cadenas Pesadas de Miosina/genética , Cadenas Ligeras de Miosina/genética , Linaje , Prevalencia , Proteínas Serina-Treonina Quinasas , Tropomiosina/genética , Troponina T/genética , Adulto Joven
12.
Hum Mutat ; 32(12): 1481-91, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21898660

RESUMEN

Dilated cardiomyopathy (DCM) is characterized by dilation of left ventricular cavity with systolic dysfunction. Clinical symptom of DCM is heart failure, often associated with cardiac sudden death. About 20-35% of DCM patients have apparent family histories and it has been revealed that mutations in genes for sarcomere proteins cause DCM. However, the disease-causing mutations can be found only in about 17% of Japanese patients with familial DCM. Bcl-2-associated athanogene 3 (BAG3) is a co-chaperone protein with antiapoptotic function, which localizes at Z-disc in the striated muscles. Recently, BAG3 gene mutations in DCM patients were reported, but the functional abnormalities caused by the mutations are not fully unraveled. In this study, we analyzed 72 Japanese familial DCM patients for mutations in BAG3 and found two mutations, p.Arg218Trp and p.Leu462Pro, in two cases of adult-onset DCM without skeletal myopathy, which were absent from 400 control subjects. Functional studies at the cellular level revealed that the DCM-associated BAG3 mutations impaired the Z-disc assembly and increased the sensitivities to stress-induced apoptosis. These observations suggested that BAG3 mutations present in 2.8% of Japanese familial DCM patients caused DCM possibly by interfering with Z-disc assembly and inducing apoptotic cell death under the metabolic stress.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis/fisiología , Cardiomiopatía Dilatada/genética , Mutación Missense , Miocitos Cardíacos/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Animales , Proteínas Reguladoras de la Apoptosis , Pueblo Asiatico/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Bovinos , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Miocitos Cardíacos/citología , Linaje , Ratas , Ratas Sprague-Dawley
13.
J Biol Chem ; 285(44): 33680-90, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20801872

RESUMEN

Phosphorylation of myosin regulatory light chain (MLC) plays a regulatory role in muscle contraction, and the level of MLC phosphorylation is balanced by MLC kinase and MLC phosphatase (MLCP). MLCP consists of a catalytic subunit, a large subunit (MYPT1 or MYPT2), and a small subunit. MLCP activity is regulated by phosphorylation of MYPTs, whereas the role of small subunit in the regulation remains unknown. We previously characterized a human heart-specific small subunit (hHS-M(21)) that increased the sensitivity to Ca(2+) in muscle contraction. In this study, we investigated the role of hHS-M(21) in the regulation of MLCP phosphorylation. Two isoforms of hHS-M(21), hHS-M(21)A and hHS-M(21)B, preferentially bound the C-terminal one-third region of MYPT1 and MYPT2, respectively. Amino acid substitutions at a phosphorylation site of MYPT1, Ser-852, impaired the binding of MYPT1 and hHS-M(21). The hHS-M(21) increased the phosphorylation level of MYPT1 at Thr-696, which was attenuated by Rho-associated kinase (ROCK) inhibitors and small interfering RNAs for ROCK. In addition, hHS-M(21) bound ROCK and enhanced the ROCK activity. These findings suggest that hHS-M(21) is a heart-specific effector of ROCK and plays a regulatory role in the MYPT1 phosphorylation at Thr-696 by ROCK.


Asunto(s)
Miocardio/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Células COS , Chlorocebus aethiops , Proteínas del Citoesqueleto/química , Calor , Humanos , Proteínas de la Membrana/química , Proteínas de Microfilamentos/química , Datos de Secuencia Molecular , Miosinas , Fosforilación , Técnicas del Sistema de Dos Híbridos
14.
Biochim Biophys Acta ; 1800(1): 31-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19819302

RESUMEN

BACKGROUND: It is not known if the dihydrolipoamide succinyltransferase (DLST) gene, a mitochondrial protein, undergoes alternative splicing. We identified an uncharacterized protein reacting with an anti-DLST antibody in the I bands of myofibrils in rat skeletal muscle. METHODS: Immunocytochemical staining with an anti-DLST antibody, the purification and amino acid sequence analysis of the protein, and the isolation and sequencing of the protein's cDNA were carried out to clarify the properties of the protein and its relationship to the DLST gene. RESULTS: A pyrophosphate concentration >10 mM was necessary to extract the protein from myofibrils in the presence of salt with a higher concentration than 0.6 M, at an alkaline pH of 7.5-8.0. The protein corresponded to the amino acid sequence of the C-terminal side of DLST. The cDNAs for this protein were splicing variants of the DLST gene, with deletions of both exons 2 and 3, or only exon 2 or 3. These variants possessed an open reading frame from an initiation codon in exon 8 of the DLST gene to a termination codon in exon 15, generating a protein with a molecular weight of 30 kDa. CONCLUSIONS: The DLST gene undergoes alternative splicing, generating the protein isolated from the I bands of myofibrils. GENERAL SIGNIFICANCE: The DLST gene produces two different proteins with quite different functions via alternative splicing.


Asunto(s)
Aciltransferasas/genética , Empalme Alternativo , Miofibrillas/metabolismo , Sarcómeros/metabolismo , Aciltransferasas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , ADN Complementario/química , ADN Complementario/genética , Electroforesis en Gel de Poliacrilamida , Regulación Enzimológica de la Expresión Génica , Humanos , Inmunohistoquímica , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Miofibrillas/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcómeros/enzimología , Análisis de Secuencia de ADN
15.
Circ J ; 75(11): 2654-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21799269

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a primary myocardial disorder with an autosomal-dominant pattern of inheritance mainly caused by single heterozygous mutations in sarcomere genes. Although multiple gene mutations have recently been reported in Western countries, clinical implications of multiple mutations in Japanese subjects are not clear. METHODS AND RESULTS: A comprehensive genetic analysis of 5 sarcomere genes (cardiac ß-myosin heavy chain gene [MYH7], cardiac myosin-binding protein C gene [MYBPC3], cardiac troponin T gene [TNNT2], α-tropomyosin gene [TPM1] and cardiac troponin I gene [TNNI3]) was performed in 93 unrelated patients and 14 mutations were identified in 28 patients. Twenty-six patients had single heterozygosity (20 in MYBPC3, 4 in MYH7, 1 in TNNT2, 1 in TNNI3), whereas 2 proband patients with familial HCM had double heterozygosity: 1 with P106fs in MYBPC3 and R869C in MYH7 and 1 with R945fs in MYBPC3 and E1049D in MYH7. From the results of the family survey and the previous literature on HCM mutations, P106fs, R945fs and R869C seemed to be pathological mutations and E1049D might be a rare polymorphism. The proband patient with P106fs and R869C double mutation was diagnosed as having HCM at an earlier age (28 years of age) than her relatives with single mutation, and had greater wall thickness with left ventricular outflow obstruction. CONCLUSIONS: One double mutation was identified in a Japanese cohort of HCM patients. Further studies are needed to clarify the clinical significance of multiple mutations including phenotypic severity.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/genética , Proteínas Musculares/genética , Mutación , Polimorfismo Genético , Adulto , Anciano , Pueblo Asiatico , Cardiomiopatía Hipertrófica Familiar/patología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Estudios de Cohortes , Femenino , Pruebas Genéticas , Humanos , Japón , Masculino , Persona de Mediana Edad , Linaje
16.
Cardiovasc Res ; 117(4): 1118-1131, 2021 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-32520982

RESUMEN

AIMS: During heart failure, the levels of circulatory heat shock protein family D member 1 (HSP60) increase. However, its underlying mechanism is still unknown. The apical domain of heat shock protein family D member 1 (HSPD1) is conserved throughout evolution. We found a point mutation in HSPD1 in a familial dilated cardiomyopathy (DCM) patient. A similar point mutation in HSPD1 in the zebrafish mutant, nbl, led to loss of its regenerative capacity and development of pericardial oedema under heat stress condition. In this study, we aimed to determine the direct involvement of HSPD1 in the development of DCM. METHODS AND RESULTS: By Sanger method, we found a point mutation (Thr320Ala) in the apical domain of HSPD1, in one familial DCM patient, which was four amino acids away from the point mutation (Val324Glu) in the nbl mutant zebrafish. The nbl mutants showed atrio-ventricular block and sudden death at 8-month post-fertilization. Histological and microscopic analysis of the nbl mutant hearts showed decreased ventricular wall thickness, elevated level of reactive oxygen species (ROS), increased fibrosis, mitochondrial damage, and increased autophagosomes. mRNA and protein expression of autophagy-related genes significantly increased in nbl mutants. We established HEK293 stable cell lines of wild-type, nbl-type, and DCM-type HSPD1, with tetracycline-dependent expression. Compared to wild-type, both nbl- and DCM-type cells showed decreased cell growth, increased expression of ROS and autophagy-related genes, inhibition of the activity of mitochondrial electron transport chain complexes III and IV, and decreased mitochondrial fission and fusion. CONCLUSION: Mutations in HSPD1 caused mitochondrial dysfunction and induced mitophagy. Mitochondrial dysfunction caused increased ROS and cardiac atrophy.


Asunto(s)
Autofagia , Cardiomiopatía Dilatada/genética , Chaperonina 60/genética , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/genética , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/genética , Estrés Oxidativo , Mutación Puntual , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Autofagosomas/genética , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Cardiomiopatía Dilatada/diagnóstico , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Chaperonina 60/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Masculino , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/ultraestructura , Dinámicas Mitocondriales , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/ultraestructura , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
17.
J Biol Chem ; 284(50): 35122-33, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19825999

RESUMEN

Long QT syndrome (LQTS) is a hereditary arrhythmia caused by mutations in genes for cardiac ion channels, including a potassium channel, KvLQT1. Inheritance of LQTS is usually autosomal-dominant, but autosomal-recessive inheritance can be observed in patients with LQTS accompanied by hearing loss. In this study, we investigated the functional alterations caused by KCNQ1 mutations, a deletion (delV595) and a frameshift (P631fs/19), which were identified in compound heterozygous state in two patients with autosomal-recessive LQTS not accompanied by hearing loss. Functional analyses showed that both mutations impaired cell surface expression due to trafficking defects. The mutations severely affected outward potassium currents without apparent dominant negative effects. It was found that delV595 impaired subunit binding, whereas P631fs/19 was retained in endoplasmic reticulum due to the newly added 19-amino acid sequence containing two retention motifs (R(633)GR and R(646)LR). This is the first report of novel mechanisms for trafficking abnormality of cardiac ion channels, providing us new insights into the molecular mechanisms of LQTS.


Asunto(s)
Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Mutación , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Transporte Biológico/fisiología , Línea Celular , Niño , Análisis Mutacional de ADN , Femenino , Humanos , Canal de Potasio KCNQ1/metabolismo , Masculino , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Linaje , Alineación de Secuencia
18.
J Clin Invest ; 117(5): 1282-93, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17446932

RESUMEN

Mutations in LMNA, which encodes nuclear Lamins A and C cause diseases affecting various organs, including the heart. We have determined the effects of an Lmna H222P mutation on signaling pathways involved in the development of cardiomyopathy in a knockin mouse model of autosomal dominant Emery-Dreifuss muscular dystrophy. Analysis of genome-wide expression profiles in hearts using Affymetrix GeneChips showed statistically significant differences in expression of genes in the MAPK pathways at the incipience of the development of clinical disease. Using real-time PCR, we showed that activation of MAPK pathways preceded clinical signs or detectable molecular markers of cardiomyopathy. In heart tissue and isolated cardiomyocytes, there was activation of MAPK cascades and downstream targets, implicated previously in the pathogenesis of cardiomyopathy. Expression of H222P Lamin A in cultured cells activated MAPKs and downstream target genes. Activation of MAPK signaling by mutant A-type lamins could be a cornerstone in the development of heart disease in autosomal dominant Emery-Dreifuss muscular dystrophy.


Asunto(s)
Cardiomiopatías/genética , Lamina Tipo A/genética , Sistema de Señalización de MAP Quinasas/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Distrofia Muscular de Emery-Dreifuss/enzimología , Distrofia Muscular de Emery-Dreifuss/genética , Sustitución de Aminoácidos/genética , Animales , Células COS , Cardiomiopatías/enzimología , Células Cultivadas , Chlorocebus aethiops , Activación Enzimática/genética , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Transgénicos
19.
Hum Genet ; 126(4): 539-47, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19513752

RESUMEN

Coronary artery disease (CAD) is based on the atherosclerosis of coronary artery and may manifest with myocardial infarction or angina pectoris. Although it is widely accepted that genetic factors are linked to CAD and several disease-related genes have been reported, only a few could be replicated suggesting that there might be some other CAD-related genes. To identify novel susceptibility loci for CAD, we used microsatellite markers in the screening and found six different candidate CAD loci. Subsequent single nucleotide polymorphism (SNP) association studies revealed an association between CAD and megakaryoblastic leukemia factor-1 gene (MKL1). The association with a promoter SNP of MKL1, -184C > T, was found in a Japanese population and the association was replicated in another Japanese population and a Korean population. Functional analysis of the MKL1 promoter SNP suggested that the higher MKL1 expression was associated with CAD. These findings suggest that MKL1 is involved in the pathogenesis of CAD.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Proteínas de Unión al ADN/genética , Proteínas de Fusión Oncogénica/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Anciano , Linfocitos B/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Enfermedad de la Arteria Coronaria/patología , Cartilla de ADN/química , Cartilla de ADN/genética , Proteínas de Unión al ADN/metabolismo , Susceptibilidad a Enfermedades , Femenino , Pruebas Genéticas , Genotipo , Humanos , Japón , Corea (Geográfico) , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/metabolismo , Fenotipo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Riesgo , Transactivadores
20.
Dis Model Mech ; 12(11)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31628103

RESUMEN

Hypertrophic cardiomyopathy (HCM) is a hereditary disease characterized by cardiac hypertrophy with diastolic dysfunction. Gene mutations causing HCM have been found in about half of HCM patients, while the genetic etiology and pathogenesis remain unknown for many cases of HCM. To identify novel mechanisms underlying HCM pathogenesis, we generated a cardiovascular-mutant medaka fish, non-spring heart (nsh), which showed diastolic dysfunction and hypertrophic myocardium. The nsh homozygotes had fewer myofibrils, disrupted sarcomeres and expressed pathologically stiffer titin isoforms. In addition, the nsh heterozygotes showed M-line disassembly that is similar to the pathological changes found in HCM. Positional cloning revealed a missense mutation in an immunoglobulin (Ig) domain located in the M-line-A-band transition zone of titin. Screening of mutations in 96 unrelated patients with familial HCM, who had no previously implicated mutations in known sarcomeric gene candidates, identified two mutations in Ig domains close to the M-line region of titin. In vitro studies revealed that the mutations found both in medaka fish and in familial HCM increased binding of titin to muscle-specific ring finger protein 1 (MURF1) and enhanced titin degradation by ubiquitination. These findings implicate an impaired interaction between titin and MURF1 as a novel mechanism underlying the pathogenesis of HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/etiología , Conectina/genética , Modelos Animales de Enfermedad , Proteínas Musculares/fisiología , Mutación , Proteínas de Motivos Tripartitos/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Conectina/fisiología , Humanos , Proteínas Musculares/genética , Oryzias , Transducción de Señal/fisiología , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA