Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Cardiovasc Res ; 117(4): 1118-1131, 2021 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-32520982

RESUMO

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.


Assuntos
Autofagia , Cardiomiopatia Dilatada/genética , Chaperonina 60/genética , Mitocôndrias Cardíacas/metabolismo , Proteínas Mitocondriais/genética , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/genética , Estresse Oxidativo , Mutação Puntual , Espécies Reativas de Oxigênio/metabolismo , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Autofagossomos/genética , Autofagossomos/metabolismo , Autofagossomos/ultraestrutura , Cardiomiopatia Dilatada/diagnóstico , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Chaperonina 60/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Células HEK293 , Humanos , Masculino , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/ultraestrutura , Dinâmica Mitocondrial , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/ultraestrutura , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
2.
Dis Model Mech ; 12(11)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31628103

RESUMO

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.


Assuntos
Cardiomiopatia Hipertrófica/etiologia , Conectina/genética , Modelos Animais de Doenças , Proteínas Musculares/fisiologia , Mutação , Proteínas com Motivo Tripartido/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Conectina/fisiologia , Humanos , Proteínas Musculares/genética , Oryzias , Transdução de Sinais/fisiologia , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética
3.
Am J Physiol Heart Circ Physiol ; 314(6): H1192-H1202, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29451818

RESUMO

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.


Assuntos
Arritmias Cardíacas/enzimologia , Insuficiência Cardíaca/enzimologia , Miócitos Cardíacos/enzimologia , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Arritmias Cardíacas/fisiopatologia , Sinalização do Cálcio , Miosinas Cardíacas/metabolismo , Cardiomiopatias/enzimologia , Cardiomiopatias/genética , Cardiomiopatias/fisiopatologia , Modelos Animais de Doenças , Fibrose , Predisposição Genética para Doença , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Contração Miocárdica , Miócitos Cardíacos/patologia , Cadeias Leves de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/genética , Fenótipo , Fosforilação , Subunidades Proteicas , Regulação para Cima , Disfunção Ventricular Esquerda/enzimologia , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda , Remodelação Ventricular , Quinases Associadas a rho/metabolismo
4.
Int Heart J ; 57(4): 507-10, 2016 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-27385602

RESUMO

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.


Assuntos
Fibrilação Atrial/genética , Cardiomiopatia Hipertrófica/genética , Mutação , Ataxias Espinocerebelares/genética , Troponina I/genética , Idoso , Fibrilação Atrial/complicações , Cardiomiopatia Hipertrófica/complicações , Feminino , Humanos , Linhagem , Ataxias Espinocerebelares/complicações
5.
Eur Heart J ; 37(18): 1469-75, 2016 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-26429810

RESUMO

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.


Assuntos
Arritmias Cardíacas , Animais , Morte Súbita Cardíaca , Sistema de Condução Cardíaco , Proteínas de Homeodomínio , Humanos , Camundongos , Fatores de Transcrição , Fibrilação Ventricular
6.
J Hum Genet ; 60(10): 641-5, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26178432

RESUMO

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.


Assuntos
Miosinas Cardíacas/genética , Cardiomegalia , Proteínas de Transporte/genética , Eletrocardiografia , Heterozigoto , Mutação de Sentido Incorreto , Cadeias Pesadas de Miosina/genética , Sarcômeros/genética , Adolescente , Adulto , Substituição de Aminoácidos , Atletas , Cardiomegalia/genética , Cardiomegalia/fisiopatologia , Feminino , Humanos , Masculino
7.
Circ Arrhythm Electrophysiol ; 8(2): 400-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25717017

RESUMO

BACKGROUND: Recent genome-wide association studies have demonstrated an association between MYH6, the gene encoding α-myosin heavy chain (α-MHC), and sinus node function in the general population. Moreover, a rare MYH6 variant, R721W, predisposing susceptibility to sick sinus syndrome has been identified. However, the existence of disease-causing MYH6 mutations for familial sick sinus syndrome and their underlying mechanisms remain unknown. METHODS AND RESULTS: We screened 9 genotype-negative probands with sick sinus syndrome families for mutations in MYH6 and identified an in-frame 3-bp deletion predicted to delete one residue (delE933) at the highly conserved coiled-coil structure within the binding motif to myosin-binding protein C in one patient. Co-immunoprecipitation analysis revealed enhanced binding of delE933 α-MHC to myosin-binding protein C. Irregular fluorescent speckles retained in the cytoplasm with substantially disrupted sarcomere striation were observed in neonatal rat cardiomyocytes transfected with α-MHC mutants carrying delE933 or R721W. In addition to the sarcomere impairments, delE933 α-MHC exhibited electrophysiological abnormalities both in vitro and in vivo. The atrial cardiomyocyte cell line HL-1 stably expressing delE933 α-MHC showed a significantly slower conduction velocity on multielectrode array than those of wild-type α-MHC or control plasmid transfected cells. Furthermore, targeted morpholino knockdown of MYH6 in zebrafish significantly reduced the heart rate, which was rescued by coexpressed wild-type human α-MHC but not by delE933 α-MHC. CONCLUSIONS: The novel MYH6 mutation delE933 causes both structural damage of the sarcomere and functional impairments on atrial action propagation. This report reinforces the relevance of MYH6 for sinus node function and identifies a novel pathophysiology underlying familial sick sinus syndrome.


Assuntos
Miosinas Cardíacas/genética , Mutação , Cadeias Pesadas de Miosina/genética , Síndrome do Nó Sinusal/genética , Potenciais de Ação , Animais , Animais Recém-Nascidos , Miosinas Cardíacas/metabolismo , Estimulação Cardíaca Artificial , Análise Mutacional de DNA , Eletrocardiografia Ambulatorial , Feminino , Técnicas de Silenciamento de Genes , Estudos de Associação Genética , Predisposição Genética para Doença , Células HeLa , Humanos , Pessoa de Meia-Idade , Morfolinos/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Cadeias Pesadas de Miosina/metabolismo , Linhagem , Fenótipo , Ratos , Ratos Sprague-Dawley , Sarcômeros/metabolismo , Sarcômeros/patologia , Síndrome do Nó Sinusal/diagnóstico , Síndrome do Nó Sinusal/metabolismo , Síndrome do Nó Sinusal/fisiopatologia , Síndrome do Nó Sinusal/terapia , Transfecção , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
J Am Heart Assoc ; 3(6): e001263, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25389285

RESUMO

BACKGROUND: Despite the accumulating genetic and molecular investigations into hypertrophic cardiomyopathy (HCM), it remains unclear how this condition develops and worsens pathologically and clinically in terms of the genetic-environmental interactions. Establishing a human disease model for HCM would help to elucidate these disease mechanisms; however, cardiomyocytes from patients are not easily obtained for basic research. Patient-specific induced pluripotent stem cells (iPSCs) potentially hold much promise for deciphering the pathogenesis of HCM. The purpose of this study is to elucidate the interactions between genetic backgrounds and environmental factors involved in the disease progression of HCM. METHODS AND RESULTS: We generated iPSCs from 3 patients with HCM and 3 healthy control subjects, and cardiomyocytes were differentiated. The HCM pathological phenotypes were characterized based on morphological properties and high-speed video imaging. The differences between control and HCM iPSC-derived cardiomyocytes were mild under baseline conditions in pathological features. To identify candidate disease-promoting environmental factors, the cardiomyocytes were stimulated by several cardiomyocyte hypertrophy-promoting factors. Interestingly, endothelin-1 strongly induced pathological phenotypes such as cardiomyocyte hypertrophy and intracellular myofibrillar disarray in the HCM iPSC-derived cardiomyocytes. We then reproduced these phenotypes in neonatal cardiomyocytes from the heterozygous Mybpc3-targeted knock in mice. High-speed video imaging with motion vector prediction depicted physiological contractile dynamics in the iPSC-derived cardiomyocytes, which revealed that self-beating HCM iPSC-derived single cardiomyocytes stimulated by endothelin-1 showed variable contractile directions. CONCLUSIONS: Interactions between the patient's genetic backgrounds and the environmental factor endothelin-1 promote the HCM pathological phenotype and contractile variability in the HCM iPSC-derived cardiomyocytes.


Assuntos
Cardiomiopatia Hipertrófica/metabolismo , Diferenciação Celular , Endotelina-1/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miofibrilas/efeitos dos fármacos , Animais , Fenômenos Biomecânicos , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/fisiopatologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Relação Dose-Resposta a Droga , Interação Gene-Ambiente , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miofibrilas/metabolismo , Miofibrilas/patologia , Fenótipo , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Disfunção Ventricular/genética , Disfunção Ventricular/metabolismo , Disfunção Ventricular/patologia , Disfunção Ventricular/fisiopatologia , Gravação em Vídeo
10.
Circ J ; 77(12): 2990-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24088304

RESUMO

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.


Assuntos
Cardiomiopatia Dilatada , Proteínas dos Microfilamentos , Proteínas Musculares , Mutação de Sentido Incorreto , Miócitos Cardíacos/metabolismo , Fator de Resposta Sérica , Adulto , Substituição de Aminoácidos , Animais , Povo Asiático , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Células Cultivadas , Forminas , Humanos , Japão , Masculino , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Pessoa de Meia-Idade , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Ratos , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo
11.
J Autoimmun ; 47: 25-33, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23953137

RESUMO

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.


Assuntos
Processamento Alternativo , Antígenos HLA/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Vírus da Influenza A/genética , Antígenos Comuns de Leucócito/genética , Proteínas da Matriz Viral/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Células COS , Chlorocebus aethiops , Células HEK293 , Células HeLa , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Inflamação/genética , Inflamação/imunologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/imunologia , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina
12.
Cardiovasc Res ; 99(3): 382-94, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23631840

RESUMO

AIMS: Dilated cardiomyopathy (DCM) is characterized by ventricular dilation associated with systolic dysfunction, which could be caused by mutations in lamina/C gene (LMNA). LMNA-linked DCM is severe in males in both human patients and a knock-in mouse model carrying a homozygous p.H222P mutation (LmnaH222P/H222P). The aim of this study was to investigate the molecular mechanisms underlying the gender difference of LMNA-linked DCM. METHODS AND RESULTS: A whole-exome analysis of a multiplex family with DCM exhibiting the gender difference revealed a DCM-linked LMNA mutation, p.R225X. Immunohistochemical analyses of neonatal rat cardiomyocytes expressing mutant LMNA constructs and heart samples from the LMNA-linked DCM patients and LmnaH222P/H222P mice demonstrated a nuclear accumulation of androgen receptor (AR) and its co-activators, serum response factor, and four-and-a-half LIM protein-2. Role of sex hormones in the gender difference was investigated in vivo using the LmnaH222P/H222P mice, where male and female mice were castrated and ovariectomized, respectively, or treated with testosterone or an antagonist of AR. Examination of the mice by echocardiography, followed by the analyses of histological changes and gene/protein expression profiles in the hearts, confirmed the involvement of testicular hormone in the disease progression and enhanced cardiac remodelling in the LmnaH222P/H222P mice. CONCLUSION: These observations indicated that nuclear accumulation of AR was associated with the gender difference in LMNA-linked DCM.


Assuntos
Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Lamina Tipo A/genética , Mutação , Receptores Androgênicos/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Cardiomiopatia Dilatada/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Mutantes , Miócitos Cardíacos/metabolismo , Orquiectomia , Ovariectomia , Linhagem , Ratos , Caracteres Sexuais , Transfecção
13.
Basic Res Cardiol ; 108(3): 349, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23572067

RESUMO

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.


Assuntos
Cardiomiopatia Hipertrófica/genética , Proteínas Musculares/genética , Mutação , Contração Miocárdica/genética , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Engenharia Tecidual , Animais , Animais Recém-Nascidos , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/fisiopatologia , Células Cultivadas , Dependovirus/genética , Imunofluorescência , Regulação da Expressão Gênica , Vetores Genéticos , Genótipo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Musculares/metabolismo , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Oligopeptídeos/farmacologia , Fenótipo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Ratos , Proteínas Repressoras/metabolismo , Fatores de Tempo , Engenharia Tecidual/métodos , Transdução Genética , Transfecção
14.
Circ J ; 77(4): 959-67, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23257389

RESUMO

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.


Assuntos
Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Subunidade beta-3 do Canal de Sódio Disparado por Voltagem/genética , Adolescente , Adulto , Idoso , Substituição de Aminoácidos , Animais , Povo Asiático , Linhagem Celular , Criança , Feminino , Humanos , Japão , Masculino , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Transporte Proteico/genética , Subunidade beta-3 do Canal de Sódio Disparado por Voltagem/metabolismo
15.
Circ Arrhythm Electrophysiol ; 5(6): 1098-107, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23064965

RESUMO

BACKGROUND: Mutations in genes including SCN5A encoding the α-subunit of the cardiac sodium channel (hNav1.5) cause Brugada syndrome via altered function of cardiac ion channels, but more than two-thirds of Brugada syndrome remains pathogenetically elusive. T-tubules and sarcoplasmic reticulum are essential in excitation of cardiomyocytes, and sarcolemmal membrane-associated protein (SLMAP) is a protein of unknown function localizing at T-tubules and sarcoplasmic reticulum. METHODS AND RESULTS: We analyzed 190 unrelated Brugada syndrome patients for mutations in SLMAP. Two missense mutations, Val269Ile and Glu710Ala, were found in heterozygous state in 2 patients but were not found in healthy individuals. Membrane surface expression of hNav1.5 in the transfected cells was affected by the mutations, and silencing of mutant SLMAP by small interfering RNA rescued the surface expression of hNav1.5. Whole-cell patch-clamp recordings of hNav1.5-expressing cells transfected with mutant SLMAP confirmed the reduced hNav1.5 current. CONCLUSIONS: The mutations in SLMAP may cause Brugada syndrome via modulating the intracellular trafficking of hNav1.5 channel.


Assuntos
Síndrome de Brugada/genética , Proteínas de Membrana/genética , Mutação de Sentido Incorreto/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Estudos de Casos e Controles , Inativação Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Técnicas de Patch-Clamp , RNA Interferente Pequeno/farmacologia , Retículo Sarcoplasmático/metabolismo , Transfecção
16.
Intern Med ; 51(18): 2559-64, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22989827

RESUMO

We herein report the case of a 61-year-old woman with dilated phase of hypertrophic cardiomyopathy (D-HCM) who had been diagnosed with HCM 17 years previously. On admission, her left ventricle (LV) had marked dilation, dyssynchrony with diffuse severe hypokinesis, and ventricular tachycardia. She had two mutations in the cardiac myosin binding protein-C gene, which were suspected to be the causes of the D-HCM. We performed LV reconstruction surgery and cardiac resynchronization therapy with a defibrillator for her drug-resistant severe heart failure. After surgery, her New York Heart Association class dramatically improved, and she has not been re-hospitalized since these treatments.


Assuntos
Terapia de Ressincronização Cardíaca , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/terapia , Proteínas de Transporte/genética , Desfibriladores Implantáveis , Ventrículos do Coração/cirurgia , Mutação/genética , Cardiomiopatia Hipertrófica/diagnóstico , Ecocardiografia , Eletrocardiografia , Feminino , Humanos , Pessoa de Meia-Idade , Sarcômeros/genética , Resultado do Tratamento
17.
Hum Mol Genet ; 21(9): 2039-53, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22286171

RESUMO

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.


Assuntos
Cardiomiopatia Dilatada/genética , Cardiomiopatia Hipertrófica Familiar/genética , Proteínas Musculares/genética , Mutação , Animais , Animais Recém-Nascidos , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Cardiomiopatia Hipertrófica Familiar/patologia , Cardiomiopatia Hipertrófica Familiar/fisiopatologia , Estudos de Casos e Controles , Códon sem Sentido , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Proteínas Musculares/fisiologia , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Mutação de Sentido Incorreto , Miocárdio/patologia , Miócitos Cardíacos/ultraestrutura , Proteínas Nucleares/metabolismo , Linhagem , Fenótipo , Ligação Proteica , Ratos , Ratos Mutantes , Ratos Sprague-Dawley , Proteínas Repressoras/metabolismo
18.
Circ J ; 76(2): 453-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22112859

RESUMO

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.


Assuntos
Povo Asiático/genética , Povo Asiático/estatística & dados numéricos , Cardiomiopatia Hipertrófica Familiar/etnologia , Cardiomiopatia Hipertrófica Familiar/genética , Sarcômeros/genética , Actinas/genética , Adulto , Idoso , Miosinas Cardíacas/genética , Proteínas de Transporte/genética , Feminino , Geografia , Humanos , Japão/epidemiologia , MAP Quinase Quinase Quinases/genética , Masculino , Pessoa de Meia-Idade , Cadeias Pesadas de Miosina/genética , Cadeias Leves de Miosina/genética , Linhagem , Prevalência , Proteínas Serina-Treonina Quinases , Tropomiosina/genética , Troponina T/genética , Adulto Jovem
19.
Hum Mol Genet ; 21(5): 1037-48, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22090424

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Doenças Metabólicas/genética , Músculo Esquelético/crescimento & desenvolvimento , Lâmina Nuclear/metabolismo , Adipócitos/citologia , Adipogenia , Animais , Animais Recém-Nascidos , Embrião de Mamíferos , Técnicas de Introdução de Genes , Transtornos do Crescimento/genética , Transtornos do Crescimento/metabolismo , Coração/crescimento & desenvolvimento , Lamina Tipo B/metabolismo , Fígado/metabolismo , Doenças Metabólicas/metabolismo , Camundongos , Mortalidade Prematura , Músculo Esquelético/anatomia & histologia , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Miócitos Cardíacos/citologia , Tamanho do Órgão , Fenótipo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Transcrição Gênica
20.
Hum Mutat ; 32(12): 1481-91, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21898660

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose/fisiologia , Cardiomiopatia Dilatada/genética , Mutação de Sentido Incorreto , Miócitos Cardíacos/patologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Idoso , Animais , Proteínas Reguladoras de Apoptose , Povo Asiático/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Bovinos , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Miócitos Cardíacos/citologia , Linhagem , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA