Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Bioinformatics ; 35(20): 3906-3912, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30903145

RESUMO

MOTIVATION: Non-coding rare variants (RVs) may contribute to Mendelian disorders but have been challenging to study due to small sample sizes, genetic heterogeneity and uncertainty about relevant non-coding features. Previous studies identified RVs associated with expression outliers, but varying outlier definitions were employed and no comprehensive open-source software was developed. RESULTS: We developed Outlier-RV Enrichment (ORE) to identify biologically-meaningful non-coding RVs. We implemented ORE combining whole-genome sequencing and cardiac RNAseq from congenital heart defect patients from the Pediatric Cardiac Genomics Consortium and deceased adults from Genotype-Tissue Expression. Use of rank-based outliers maximized sensitivity while a most extreme outlier approach maximized specificity. Rarer variants had stronger associations, suggesting they are under negative selective pressure and providing a basis for investigating their contribution to Mendelian disorders. AVAILABILITY AND IMPLEMENTATION: ORE, source code, and documentation are available at https://pypi.python.org/pypi/ore under the MIT license. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Genômica , Software , Criança , Documentação , Humanos , Incerteza , Sequenciamento Completo do Genoma
2.
Am J Physiol Heart Circ Physiol ; 293(1): H284-91, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17351073

RESUMO

Point mutations in cardiac myosin, the heart's molecular motor, produce distinct clinical phenotypes: hypertrophic (HCM) and dilated (DCM) cardiomyopathy. Do mutations alter myosin's molecular mechanics in a manner that is predictive of the clinical outcome? We have directly characterized the maximal force-generating capacity (F(max)) of two HCM (R403Q, R453C) and two DCM (S532P, F764L) mutant myosins isolated from homozygous mouse models using a novel load-clamped laser trap assay. F(max) was 50% (R403Q) and 80% (R453C) greater for the HCM mutants compared with the wild type, whereas F(max) was severely depressed for one of the DCM mutants (65% S532P). Although F(max) was normal for the F764L DCM mutant, its actin-activated ATPase activity and actin filament velocity (V(actin)) in a motility assay were significantly reduced (Schmitt JP, Debold EP, Ahmad F, Armstrong A, Frederico A, Conner DA, Mende U, Lohse MJ, Warshaw D, Seidman CE, Seidman JG. Proc Natl Acad Sci USA 103: 14525-14530, 2006.). These F(max) data combined with previous V(actin) measurements suggest that HCM and DCM result from alterations to one or more of myosin's fundamental mechanical properties, with HCM-causing mutations leading to enhanced but DCM-causing mutations leading to depressed function. These mutation-specific changes in mechanical properties must initiate distinct signaling cascades that ultimately lead to the disparate phenotypic responses observed in HCM and DCM.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Hipertrófica/metabolismo , Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/genética , Miosinas Ventriculares/química , Miosinas Ventriculares/genética , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Hipertrófica/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Contração Miocárdica , Pinças Ópticas , Estresse Mecânico , Relação Estrutura-Atividade
3.
J Med Genet ; 42(10): e59, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16199542

RESUMO

OBJECTIVE: To report the frequency of single and multiple gene mutations in an Australian cohort of patients with hypertrophic cardiomyopathy (HCM). METHODS: Genetic screening of seven HCM genes (beta-MHC, MyBP-C, cTnT, cTnI, ACTC, MYL2, and MYL3) was undertaken in 80 unrelated probands. Screening was by denaturing high performance liquid chromatography and direct DNA sequencing. Clinical data were collected on all patients and on genotyped family members. RESULTS: 26 mutations were identified in 23 families (29%). Nineteen probands (24%) had single mutations (11 beta-MHC, 4 MyBP-C, 3 cTnI, 1 cTnT). Multiple gene mutations were identified in four probands (5%): one had a double mutation and the others had compound mutations. Six of 14 affected individuals from multiple mutation families (43%) experienced a sudden cardiac death event, compared with 10 of 55 affected members (18%) from single mutation families (p = 0.05). There was an increase in septal wall thickness in patients with compound mutations (mean (SD): 30.7 (3.1) v 24.4 (7.4) mm; p<0.05). CONCLUSIONS: Multiple gene mutations occurring in HCM families may result in a more severe clinical phenotype because of a "double dose" effect. This highlights the importance of screening the entire panel of HCM genes even after a single mutation has been identified.


Assuntos
Cardiomiopatia Hipertrófica/genética , Aconselhamento Genético , Predisposição Genética para Doença , Mutação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Cromatografia Líquida de Alta Pressão , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Técnicas de Diagnóstico Molecular , Linhagem , Fenótipo , Análise de Sequência de DNA
4.
Int J Pediatr Otorhinolaryngol ; 68(8): 1063-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15236894

RESUMO

OBJECTIVE: To undertake a comprehensive investigation into the very high incidence of congenital deafness on the Macano peninsula of Margarita Island, Venezuela. METHODS: Numerous visits were made to the isolated island community over a 4-year-period. During these visits, it became apparent that a significant number of individuals complained of problems with hearing and vision. Socioeconomic assessments, family pedigrees and clinical histories were recorded on standard questionnaires. All individuals underwent thorough otolaryngologic and ophthalmologic examinations. Twenty milliliters of peripheral venous blood was obtained from each participant. A genome-wide linkage analysis study was performed. Polymorphic microsatellite markers were amplified by polymerase chain reaction and separated on polyacrylamide gels. An ABI 377XL sequencer was used to separate fragments and LOD scores were calculated by using published software. RESULTS: Twenty-four families were identified, comprising 329 individuals, age range 1-80 years, including 184 children. All families were categorized in the lower two (least affluent) socioeconomic categories. A high incidence of consanguinity was detected. Fifteen individuals (11 adults, 4 children) had profound congenital sensorineural hearing loss, vestibular areflexia and retinitis pigmentosa. A maximum LOD score of 6.76 (Linkage >3.0), between markers D11s4186 and D11s911, confirmed linkage to chromosome 11q13.5. The gene myosin VIIA (MYO7A) was confirmed in the interval. Clinical and genetic findings are consistent with a diagnosis of Usher syndrome 1B for those with hearing and vision problems. CONCLUSIONS: We report 15 Usher syndrome 1B individuals from a newly detected Latin American socio-demographic origin, with a very high prevalence of 76 per 100,000 population.


Assuntos
Anormalidades Congênitas/genética , Perda Auditiva Neurossensorial/genética , Retinose Pigmentar/genética , Doenças Vestibulares/genética , Transtornos da Visão/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Mapeamento Cromossômico , Anormalidades Congênitas/epidemiologia , Consanguinidade , Dineínas/genética , Feminino , Perda Auditiva Neurossensorial/epidemiologia , Humanos , Lactente , Escore Lod , Masculino , Pessoa de Meia-Idade , Miosina VIIa , Miosinas/genética , Linhagem , Reação em Cadeia da Polimerase , Retinose Pigmentar/epidemiologia , Fatores Socioeconômicos , Síndrome , Venezuela/epidemiologia , Doenças Vestibulares/epidemiologia , Transtornos da Visão/epidemiologia
5.
Muscle Nerve ; 28(1): 118-22, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12811783

RESUMO

Hallervorden-Spatz syndrome (HSS) is a neurodegenerative disorder characterized by progressive dementia, dystonia, ataxia, and rigidity. An atypical form of adult-onset HSS was observed in a 36-year-old man presenting with progressive dysarthria. Markedly dysarthric speech and a weak atrophic tongue associated with a neurogenic pattern of motor unit recruitment in bulbar-supplied muscles on electromyography led to an initial impression of bulbar amyotrophic lateral sclerosis (ALS). Lack of expected progression of symptoms, however, prompted reinvestigation. Repeat brain magnetic resonance imaging demonstrated an "eye-of-the-tiger" pattern in the basal ganglia, characteristic of HSS, thus requiring genetic studies. DNA analyses of the pantothenate kinase gene (PANK2) was conducted and revealed two novel, disease-causing exon 3 missense mutations (Cys231Ser and Tyr251Cys). This case broadens the genotypic and phenotypic spectrum of HSS to include a late-onset syndrome resembling bulbar-onset ALS.


Assuntos
Esclerose Lateral Amiotrófica/diagnóstico , Neurodegeneração Associada a Pantotenato-Quinase/diagnóstico , Adulto , Gânglios da Base/patologia , Encéfalo/patologia , Diagnóstico Diferencial , Eletromiografia , Humanos , Imageamento por Ressonância Magnética , Masculino , Músculo Esquelético/patologia , Mutação de Sentido Incorreto/genética , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fenótipo , Fosfotransferases (Aceptor do Grupo Álcool)/genética
10.
Circulation ; 104(22): 2734-9, 2001 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-11723028

RESUMO

BACKGROUND: Homozygous mutant mice expressing a truncated form of myosin-binding protein C (MyBP-C(t/t)) develop severe dilated cardiomyopathy, whereas the heterozygous mutation (MyBP-C(t/+)) causes mild hypertrophic cardiomyopathy. Adult male MyBP-C(t/t) and MyBP-C(t/+) mice were evaluated for arrhythmia vulnerability with an in vivo electrophysiology study. METHODS AND RESULTS: Surface ECGs were obtained for heart rate, rhythm, and conduction intervals. Atrial, atrioventricular, and ventricular conduction parameters and refractoriness were assessed in 22 MyBP-C(t/t), 10 MyBP-C(t/+), and 17 wild-type MyBP-C(+/+) mice with endocardial pacing and intracardiac electrogram recording. Arrhythmia induction was attempted with standardized programmed stimulation at baseline and with isoproterenol. Heart rate variability and ambient arrhythmia activity were assessed with telemetric ECG monitors. Quantitative histological characterization was performed on serial sections of excised hearts. MyBP-C(t/t) and MyBP-C(t/+) mice have normal ECG intervals and sinus node, atrial, and ventricular conduction and refractoriness. Ventricular tachycardia was reproducibly inducible in 14 of 22 MyBP-C(t/t) mice (64%) during programmed stimulation, compared with 2 of 10 MyBP-C(t/+) mice (20%) and 0 of 17 wild-type controls (P<0.001). Ventricular ectopy was present only in MyBP-C(t/t) mice during ambulatory ECG recordings. There were no differences in heart rate variability parameters. Interstitial fibrosis correlated with genotype but did not predict arrhythmia susceptibility within the MyBP-C(t/t) group. CONCLUSIONS: MyBP-C(t/t) mice, despite prominent histopathology and ventricular dysfunction, exhibit normal conduction and refractoriness, yet are vulnerable to ventricular arrhythmias. Somatic influences between genetically identical mutant mice most likely account for variability in arrhythmia susceptibility. A sarcomeric protein gene mutation leads to a dilated cardiomyopathy and ventricular arrhythmia vulnerability phenotype.


Assuntos
Arritmias Cardíacas/fisiopatologia , Cardiomiopatias/fisiopatologia , Proteínas de Transporte/genética , Ventrículos do Coração/fisiopatologia , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/genética , Cardiomiopatias/complicações , Cardiomiopatias/genética , Cardiomiopatias/patologia , Modelos Animais de Doenças , Eletrocardiografia , Técnicas Eletrofisiológicas Cardíacas , Predisposição Genética para Doença , Sistema de Condução Cardíaco/fisiopatologia , Frequência Cardíaca , Heterozigoto , Homozigoto , Masculino , Camundongos , Camundongos Mutantes , Mutação , Miocárdio/patologia , Fenótipo , Deleção de Sequência
11.
J Mol Cell Cardiol ; 33(11): 2055-60, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11708849

RESUMO

Familial hypertrophic cardiomyopathy (FHC), an autosomal dominant disorder caused by mutationally altered dominant-acting sarcomere proteins, exhibits significant clinical heterogeneity. To determine whether genetic background could influence the expression of this disease, we studied a murine model for this human condition. Hypertrophic responses to the Arg403Gln missense mutation in a cardiac myosin heavy chain gene were compared in 129SvEv (inbred; designated 129SvEv- alpha MHC403/+) and Black Swiss (outbred; designated BSw- alpha MHC403/+) strains. At 30-50 weeks of age all 129SvEv- alpha MHC403/+ showed left ventricular hypertrophy, while left ventricular wall thickness was increased in only half of BSw- alpha MHC403/+ mice demonstrating that a polymorphic modifier gene can determine the hypertrophic response to this dominant-acting sarcomere protein mutation. Further analysis suggests that SJL/J mice bear a recessive allele of this modifier gene that prevents a hypertrophic response to the Arg403Gln missense mutation. We conclude that genetic modifiers in mice, and presumably in man, can alter the hypertrophic response to sarcomere protein gene missense mutations.


Assuntos
Cardiomiopatia Hipertrófica Familiar/genética , Hipertrofia , Polimorfismo Genético , Alelos , Animais , Modelos Animais de Doenças , Ecocardiografia , Teste de Esforço , Genes Dominantes , Coração/fisiologia , Humanos , Hipertrofia Ventricular Esquerda/metabolismo , Camundongos , Mutação , Mutação de Sentido Incorreto , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/genética , Condicionamento Físico Animal , Sarcômeros/metabolismo , Fatores de Tempo
12.
Cell ; 106(6): 709-21, 2001 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-11572777

RESUMO

Heterozygous Tbx5(del/+) mice were generated to study the mechanisms by which TBX5 haploinsufficiency causes cardiac and forelimb abnormalities seen in Holt-Oram syndrome. Tbx5 deficiency in homozygous mice (Tbx5(del/del)) decreased expression of multiple genes and caused severe hypoplasia of posterior domains in the developing heart. Surprisingly, Tbx5 haploinsufficiency also markedly decreased atrial natriuretic factor (ANF) and connexin 40 (cx40) transcription, implicating these as Tbx5 target genes and providing a mechanism by which 50% reduction of T-box transcription factors cause disease. Direct and cooperative transactivation of the ANF and cx40 promoters by Tbx5 and the homeodomain transcription factor Nkx2-5 was also demonstrated. These studies provide one potential explanation for Holt-Oram syndrome conduction system defects, suggest mechanisms for intrafamilial phenotypic variability, and account for related cardiac malformations caused by other transcription factor mutations.


Assuntos
Anormalidades Múltiplas/genética , Fator Natriurético Atrial/genética , Desenvolvimento Ósseo/fisiologia , Cardiopatias Congênitas/genética , Proteínas com Domínio T/genética , Envelhecimento , Animais , Sequência de Bases , Sítios de Ligação , Desenvolvimento Ósseo/genética , Diferenciação Celular , Conexinas/genética , Modelos Animais de Doenças , Eletrocardiografia , Desenvolvimento Embrionário e Fetal , Membro Anterior/anormalidades , Coração/embriologia , Cardiopatias Congênitas/fisiopatologia , Heterozigoto , Homozigoto , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Miocárdio/citologia , Regiões Promotoras Genéticas , Ratos , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Ovinos , Síndrome , Proteínas com Domínio T/deficiência , Proteína alfa-5 de Junções Comunicantes
13.
Intern Med J ; 31(1): 53-9, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11478359

RESUMO

When Watson and Crick proposed the double helix model for DNA structure in a 2 page Nature article in 1953, no one could have predicted the enormous impact this finding would have on the study of human disease. Over the last decade in particular, major advances have been made in our understanding of both normal biological processes and basic molecular mechanisms underlying a variety of medical diseases. Knowledge obtained from basic cellular, molecular and genetic studies has enabled the development of strategies for the modification, prevention and potential cure of human diseases. This brief overview focuses on the enormous impact molecular studies have had on various aspects of medicine. The inherited cardiac disorder hypertrophic cardiomyopathy is used here as a model to illustrate how molecular studies have not only redefined 'gold standards' for diagnosis, but have also influenced management approaches, increased our understanding of fundamental disease-causing mechanisms and identified potential targets for therapeutic intervention. The near-completion of the Human Genome Project, which identifies the 3.2 billion base pairs that comprise the human genome (the so-called 'Book of Life'), has exponentially heightened the focus on the importance of molecular studies and how such studies will impact on various aspects of medicine in the 21st century.


Assuntos
Genética Médica/tendências , Cardiomiopatia Hipertrófica Familiar/diagnóstico , Cardiomiopatia Hipertrófica Familiar/genética , Previsões , Genética Médica/história , História do Século XX , História do Século XXI , Projeto Genoma Humano , Humanos , Mutação/genética
14.
J Am Coll Cardiol ; 38(2): 315-21, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11499718

RESUMO

OBJECTIVES: We sought to determine whether the development of left ventricular hypertrophy (LVH) can be demonstrated during adulthood in genetically affected relatives with hypertrophic cardiomyopathy (HCM). BACKGROUND: Hypertrophic cardiomyopathy is a heterogeneous cardiac disease caused by mutations in nine genes that encode proteins of the sarcomere. Mutations in cardiac myosin-binding protein C (MyBPC) gene have been associated with age-related penetrance. METHODS: To further analyze dormancy of LVH in patients with HCM, we studied, using echocardiography and 12-lead electrocardiography, the phenotypic expression caused by MyBPC mutations in seven genotyped pedigrees. RESULTS: Of 119 family members studied, 61 were identified with a MyBPC mutation, including 21 genetically affected relatives (34%) who did not express the HCM morphologic phenotype (by virtue of showing normal left ventricular wall thickness). Of these 21 phenotype-negative individuals, 9 were children, presumably in the prehypertrophic phase, and 12 were adults. Of the 12 adults with normal wall thickness < or = 12 mm (7 also with normal electrocardiograms), 5 subsequently underwent serial echocardiography prospectively over four to six years. Of note, three of these five adults showed development of LVH in mid-life, appearing for the first time at 33, 34 and 42 years of age, respectively, not associated with outflow obstruction or significant symptoms. CONCLUSIONS: In adults with HCM, disease-causing MyBPC mutations are not uncommonly associated with absence of LVH on echocardiogram. Delayed remodeling with the development of LVH appearing de novo in adulthood, demonstrated here for the first time in individual patients with prospectively obtained serial echocardiograms, substantiates the principle of age-related penetrance for MyBPC mutations in HCM. These observations alter prevailing perceptions regarding the HCM clinical spectrum and family screening strategies and further characterize the evolution of LVH in this disease.


Assuntos
Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/genética , Hipertrofia Ventricular Esquerda/genética , Adulto , Cardiomiopatia Hipertrófica/diagnóstico por imagem , Eletrocardiografia/métodos , Saúde da Família , Feminino , Humanos , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Penetrância , Fenótipo , Estudos Prospectivos , Ultrassonografia
16.
17.
Am J Hum Genet ; 69(1): 67-74, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11369996

RESUMO

Witkop syndrome, also known as tooth and nail syndrome (TNS), is a rare autosomal dominant disorder. Affected individuals have nail dysplasia and several congenitally missing teeth. To identify the gene responsible for TNS, we used candidate-gene linkage analysis in a three-generation family affected by the disorder. We found linkage between TNS and polymorphic markers surrounding the MSX1 locus. Direct sequencing and restriction-enzyme analysis revealed that a heterozygous stop mutation in the homeodomain of MSX1 cosegregated with the phenotype. In addition, histological analysis of Msx1-knockout mice, combined with a finding of Msx1 expression in mesenchyme of developing nail beds, revealed that not only was tooth development disrupted in these mice, but nail development was affected as well. Nail plates in Msx1-null mice were defective and were thinner than those of their wild-type littermates. The resemblance between the tooth and nail phenotype in the human family and that of Msx1-knockout mice strongly supports the conclusions that a nonsense mutation in MSX1 causes TNS and that Msx1 is critical for both tooth and nail development.


Assuntos
Anodontia/genética , Códon sem Sentido/genética , Ligação Genética/genética , Proteínas de Homeodomínio/genética , Unhas Malformadas/genética , Fatores de Transcrição , Adulto , Sequência de Aminoácidos , Animais , Anodontia/embriologia , Sequência de Bases , Mapeamento Cromossômico , Análise Mutacional de DNA , Feminino , Genes Dominantes/genética , Heterozigoto , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Humanos , Hibridização In Situ , Fator de Transcrição MSX1 , Masculino , Camundongos , Camundongos Knockout , Unhas Malformadas/embriologia , Linhagem , Fenótipo , Polimorfismo Genético/genética , Estrutura Terciária de Proteína , RNA Mensageiro/análise , RNA Mensageiro/genética , Síndrome
18.
Mol Cell Biol ; 21(5): 1730-6, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238910

RESUMO

To define the role of Irx4, a member of the Iroquois family of homeobox transcription factors in mammalian heart development and function, we disrupted the murine Irx4 gene. Cardiac morphology in Irx4-deficient mice (designated Irx4(Delta ex2/Delta ex2)) was normal during embryogenesis and in early postnatal life. Adult Irx4(Delta ex2/Delta ex2) mice developed a cardiomyopathy characterized by cardiac hypertrophy and impaired contractile function. Prior to the development of cardiomyopathy, Irx4(Delta ex2/Delta ex2) hearts had abnormal ventricular gene expression: Irx4-deficient embryos exhibited reduced ventricular expression of the basic helix-loop-helix transcription factor eHand (Hand1), increased Irx2 expression, and ventricular induction of an atrial chamber-specific transgene. In neonatal hearts, ventricular expression of atrial natriuretic factor and alpha-skeletal actin was markedly increased. Several weeks subsequent to these changes in embryonic and neonatal gene expression, increased expression of hypertrophic markers BNP and beta-myosin heavy chain accompanied adult-onset cardiac hypertrophy. Cardiac expression of Irx1, Irx2, and Irx5 may partially compensate for loss of Irx4 function. We conclude that Irx4 is not sufficient for ventricular chamber formation but is required for the establishment of some components of a ventricle-specific gene expression program. In the absence of genes under the control of Irx4, ventricular function deteriorates and cardiomyopathy ensues.


Assuntos
Cardiomiopatias/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Actinas/biossíntese , Alelos , Animais , Fator Natriurético Atrial/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Northern Blotting , Cardiomiopatias/metabolismo , Citocinas/biossíntese , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Ecocardiografia , Heterozigoto , Proteínas de Homeodomínio/biossíntese , Homozigoto , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Mutagênese , Miocárdio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Transgenes , Regulação para Cima
20.
Circ Res ; 88(4): 383-9, 2001 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-11230104

RESUMO

Although sarcomere protein gene mutations cause familial hypertrophic cardiomyopathy (FHC), individuals bearing a mutant cardiac myosin binding protein C (MyBP-C) gene usually have a better prognosis than individuals bearing beta-cardiac myosin heavy chain (MHC) gene mutations. Heterozygous mice bearing a cardiac MHC missense mutation (alphaMHC(403/+) or a cardiac MyBP-C mutation (MyBP-C(t/+)) were constructed as murine FHC models using homologous recombination in embryonic stem cells. We have compared cardiac structure and function of these mouse strains by several methods to further define mechanisms that determine the severity of FHC. Both strains demonstrated progressive left ventricular (LV) hypertrophy; however, by age 30 weeks, alphaMHC(403/+) mice demonstrated considerably more LV hypertrophy than MyBP-C(t/+) mice. In older heterozygous mice, hypertrophy continued to be more severe in the alphaMHC(403/+) mice than in the MyBP-C(t/+) mice. Consistent with this finding, hearts from 50-week-old alphaMHC(403/+) mice demonstrated increased expression of molecular markers of cardiac hypertrophy, but MyBP-C(t/+) hearts did not demonstrate expression of these molecular markers until the mice were >125 weeks old. Electrophysiological evaluation indicated that MyBP-C(t/+) mice are not as likely to have inducible ventricular tachycardia as alphaMHC(403/+) mice. In addition, cardiac function of alphaMHC(403/+) mice is significantly impaired before the development of LV hypertrophy, whereas cardiac function of MyBP-C(t/+) mice is not impaired even after the development of cardiac hypertrophy. Because these murine FHC models mimic their human counterparts, we propose that similar murine models will be useful for predicting the clinical consequences of other FHC-causing mutations. These data suggest that both electrophysiological and cardiac function studies may enable more definitive risk stratification in FHC patients.


Assuntos
Cardiomiopatia Hipertrófica/genética , Modelos Animais de Doenças , Actinas/genética , Alelos , Animais , Fator Natriurético Atrial/genética , Northern Blotting , Proteínas de Transporte/genética , Ecocardiografia , Eletrofisiologia , Saúde da Família , Masculino , Camundongos , Mutação , Mutação de Sentido Incorreto , Miocárdio/química , Miocárdio/patologia , Splicing de RNA , RNA Mensageiro/metabolismo , Sarcômeros/química , Fatores de Tempo , Transgenes , Disfunção Ventricular Esquerda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...