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

Tipo de documento
Intervalo de ano de publicação
1.
Genet Med ; 25(4): 100797, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36727595

RESUMO

PURPOSE: Population newborn genetic screening for hypertrophic cardiomyopathy (HCM) is feasible, however its benefits, harms, and cost-effectiveness are uncertain. METHODS: We developed a microsimulation model to simulate a US birth cohort of 3.7 million newborns. Those identified with pathogenic/likely pathogenic variants associated with increased risk of HCM underwent surveillance and recommended treatment, whereas in usual care, individuals with family histories of HCM underwent surveillance. RESULTS: In a cohort of 3.7 million newborns, newborn genetic screening would reduce HCM-related deaths through age 20 years by 44 (95% uncertainty interval [UI] = 10-103) however increase the numbers of children undergoing surveillance by 8127 (95% UI = 6308-9664). Compared with usual care, newborn genetic screening costs $267,000 per life year saved (95% UI, $106,000 to $919,000 per life year saved). CONCLUSION: Newborn genetic screening for HCM could prevent deaths but at a high cost and would require many healthy children to undergo surveillance. This study shows how modeling can provide insights into the tradeoffs between benefits and costs that will need to be considered as newborn genetic screening is more widely adopted.


Assuntos
Cardiomiopatia Hipertrófica , Testes Genéticos , Criança , Humanos , Recém-Nascido , Adulto Jovem , Adulto , Cardiomiopatia Hipertrófica/diagnóstico , Cardiomiopatia Hipertrófica/genética , Triagem Neonatal , Análise de Custo-Efetividade
2.
Int J Mol Sci ; 22(21)2021 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-34769381

RESUMO

Mutations in the gene encoding cardiac myosin-binding protein-C (MyBPC), a thick filament assembly protein that stabilizes sarcomeric structure and regulates cardiac function, are a common cause for the development of hypertrophic cardiomyopathy. About 10% of carriers of the Δ25bp variant of MYBPC3, which is common in individuals from South Asia, are also carriers of the D389V variant on the same allele. Compared with noncarriers and those with MYBPC3Δ25bp alone, indicators for the development of hypertrophic cardiomyopathy occur with increased frequency in MYBPC3Δ25bp/D389V carriers. Residue D389 lies in the IgI-like C2 domain that is part of the N-terminal region of MyBPC. To probe the effects of mutation D389V on structure, thermostability, and protein-protein interactions, we produced and characterized wild-type and mutant constructs corresponding to the isolated 10 kDa C2 domain and a 52 kDa N-terminal fragment that includes subdomains C0 to C2. Our results show marked reductions in the melting temperatures of D389V mutant constructs. Interactions of construct C0-C2 D389V with the cardiac isoforms of myosin-2 and actin remain unchanged. Molecular dynamics simulations reveal changes in the stiffness and conformer dynamics of domain C2 caused by mutation D389V. Our results suggest a pathomechanism for the development of HCM based on the toxic buildup of misfolded protein in young MYBPC3Δ25bp/D389V carriers that is supplanted and enhanced by C-zone haploinsufficiency at older ages.


Assuntos
Domínios C2 , Cardiomiopatia Hipertrófica/patologia , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Mutação , Domínios e Motivos de Interação entre Proteínas , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Proteínas de Transporte/genética , Humanos , Termodinâmica
3.
J Electrocardiol ; 53: 95-99, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30716529

RESUMO

Cardiomyopathies represent a well-known cause of heart failure and sudden death. Although cardiomyopathies are generally categorized in distinct nosographic entities, characterized by single gene-to-disease causal relationships, recently, oligogenic mutations have also been associated to relevant cardiac clinical features. We report the case of a master athlete carrying trigenic mutations in desmoglein-2 (DSG2), desmocollin-2 (DSC2) and heavy chain myosin 6 (MYH6), which determine a mild hypertrophic phenotype associated both to ventricular tachyarrhythmias and atrio-ventricular block. We discuss the differential diagnosis and prognostic approach in patient affected by complex cardiomyopathy phenotype, along with the importance of sport restriction and sudden death prevention.


Assuntos
Atletas , Cardiomiopatia Hipertrófica/genética , Morte Súbita Cardíaca/etiologia , Bloqueio Atrioventricular/complicações , Bloqueio Atrioventricular/genética , Miosinas Cardíacas/genética , Cardiomiopatia Hipertrófica/complicações , Desmocolinas/genética , Desmogleína 2/genética , Diagnóstico Diferencial , Eletrocardiografia , Humanos , Pessoa de Meia-Idade , Mutação , Cadeias Pesadas de Miosina/genética , Marca-Passo Artificial , Fenótipo , Prognóstico , Taquicardia Ventricular/complicações , Taquicardia Ventricular/genética
4.
Eur Heart J Cardiovasc Imaging ; 20(8): 932-938, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668650

RESUMO

AIMS: Myocardial oxygenation is impaired in hypertrophic cardiomyopathy (HCM) patients with left ventricular hypertrophy (LVH), and possibly also in HCM gene carriers without LVH. Whether these oxygenation changes are also associated with abnormalities in diastolic function or left ventricular (LV) strain are unknown. METHODS AND RESULTS: We evaluated 60 subjects: 20 MYBPC3 gene positive patients with LVH (G+LVH+), 18 MYBPC3 gene positive without LVH (G+LVH-), 11 gene negative siblings (G-), and 11 normal controls (NC). All subjects underwent 2D transthoracic echocardiography and cardiovascular magnetic resonance imaging for assessment of ventricular volumes, mass, and myocardial oxygenation at rest and adenosine stress using the blood oxygen level dependent (BOLD) technique. Maximal septal thickness was 20 mm in the G+LVH+ group, vs. 9 mm for the G+LVH- group. As expected, the G+LVH+ group had a more blunted myocardial oxygenation response to stress when compared with the G+LVH- group (-5% ± 3% vs. 2% ± 4%, P < 0.05), G- siblings (-5% ± 3% vs. 11% ± 4%, P < 0.0001) and NC (-5% ± 3% vs. 15% ± 4%, P < 0.0001). A blunted BOLD response to stress was also seen in G+LVH- subjects when compared with gene negative siblings (2% ± 4% vs. 11% ± 4%, P < 0.05) and NC (15% ± 4%, P < 0.050). G+LVH+ patients exhibited abnormal diastolic function including lower E', higher E to E' ratio and greater left atrial area compared with the G+LVH- subjects who all had normal values for these indices. CONCLUSION: Myocardial deoxygenation during stress is observed in MYBPC3 HCM patients, even in the presence of normal LV diastolic function, LV global longitudinal strain, and LV wall thickness.


Assuntos
Cardiomiopatia Hipertrófica/diagnóstico por imagem , Cardiomiopatia Hipertrófica/genética , Ecocardiografia/métodos , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Hipertrofia Ventricular Esquerda/genética , Imagem Cinética por Ressonância Magnética/métodos , Consumo de Oxigênio , Técnicas de Imagem de Sincronização Cardíaca , Proteínas de Transporte , Estudos de Casos e Controles , Meios de Contraste , Diástole , Teste de Esforço , Feminino , Humanos , Interpretação de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Miocárdio , Compostos Organometálicos
5.
Adv Gerontol ; 32(6): 901-907, 2019.
Artigo em Russo | MEDLINE | ID: mdl-32160427

RESUMO

Hypertrophic cardiomyopathy (HС) is a heterogeneous myocardial disease with a wide range of clinical manifestations and risk of development increasing with age along with myocardial changes characteristic of aging. The contribution of genetic component to the development of HC is obvious, however, the etiology and pathogenesis of this disease remains unclear in 50% of cases. The aim of the present study was to search for single nucleotide polymorphisms (SNPs) in mitochondria-associated genes that can contribute to the development of myocardial hypertrophy using RNA-Seq data from senescence-accelerated OXYS rats. Here we revealed SNPs with a possible negative effect on the function of the protein product in mitochondria-associated genes Fbxl4 and Slc25a32, mutations in which were not previously associated with HC. Alterations in the expression of these genes in the myocardium of OXYS rats at different stages of the development of pathological changes indicate that the revealed SNPs can contribute to the development of HC. Thus, SNPs in the Fbxl4 and Slc25a32 genes, as well as the genes themselves, can be considered promising molecular targets in the studies of the contribution of mitochondrial dysfunction to the development of myocardial hypertrophy.


Assuntos
Cardiomiopatia Hipertrófica/genética , Mitocôndrias/genética , Polimorfismo de Nucleotídeo Único , Envelhecimento , Animais , Ratos
6.
Circulation ; 138(14): 1387-1398, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30297972

RESUMO

Background: A better understanding of the factors that contribute to heterogeneous outcomes and lifetime disease burden in hypertrophic cardiomyopathy (HCM) is critically needed to improve patient management and outcomes. The Sarcomeric Human Cardiomyopathy Registry (SHaRe) was established to provide the scale of data required to address these issues, aggregating longitudinal datasets curated by eight international HCM specialty centers. Methods: Data on 4591 HCM patients (2763 genotyped), followed for a mean of 5.4±6.9 years (24,791 patient-years; median [interquartile range] 2.9 [0.3-7.9] years) were analyzed regarding cardiac arrest, cardiac transplantation, appropriate implantable cardioverter-defibrillator (ICD) therapy, all-cause death, atrial fibrillation, stroke, New York Heart Association Functional Class III/IV symptoms (all comprising the overall composite endpoint), and left ventricular ejection fraction (LVEF)<35%. Outcomes were analyzed individually and as composite endpoints. Results: Median age of diagnosis was 45.8 [30.9-58.1] years and 37% of patients were female. Age of diagnosis and sarcomere mutation status were predictive of outcomes. Patients <40 years old at diagnosis had a 77% [95% confidence interval: 72%, 80%] cumulative incidence of the overall composite outcome by age 60, compared to 32% [29%, 36%] by age 70 for patients diagnosed >60 years. Young HCM patients (20-29 years) had 4-fold higher mortality than the general United States population at a similar age. Patients with pathogenic/likely pathogenic sarcomere mutations had two-fold greater risk for adverse outcomes compared to patients without mutations; sarcomere variants of uncertain significance were associated with intermediate risk. Heart failure and atrial fibrillation were the most prevalent adverse events, although typically not emerging for several years after diagnosis. Ventricular arrhythmias occurred in 32% [23%, 40%] of patients <40 years at diagnosis, but in 1% [1%, 2%] >60 years. Conclusions: The cumulative burden of HCM is substantial and dominated by heart failure and atrial fibrillation occurring many years following diagnosis. Young age of diagnosis and the presence of a sarcomere mutation are powerful predictors of adverse outcomes. These findings highlight the need for close surveillance throughout life, and the need to develop disease-modifying therapies.


Assuntos
Fibrilação Atrial/genética , Cardiomiopatia Hipertrófica/genética , Efeitos Psicossociais da Doença , Insuficiência Cardíaca/genética , Mutação , Sarcômeros/genética , Adulto , Fatores Etários , Idoso , Fibrilação Atrial/mortalidade , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/terapia , Cardiomiopatia Hipertrófica/mortalidade , Cardiomiopatia Hipertrófica/fisiopatologia , Cardiomiopatia Hipertrófica/terapia , Causas de Morte , Bases de Dados Factuais , Progressão da Doença , Feminino , Predisposição Genética para Doença , Insuficiência Cardíaca/mortalidade , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Fenótipo , Prognóstico , Sistema de Registros , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Fatores de Tempo , Adulto Jovem
7.
Can J Vet Res ; 81(2): 137-146, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28408782

RESUMO

Myocardial dysfunction occurs in cats with hypertrophic cardiomyopathy (HCM), but little is known about the early stages of the disease. Strain imaging echocardiography is a method that enables the quantitative assessment of myocardial function and deformity, allowing the characterization of systolic dysfunction. The objective of this study was to assess systolic function using strain imaging echocardiography in Maine coon cats genetically tested for the A31P mutation in the MYBPC3 gene, with and without ventricular hypertrophy. For this purpose, 57 Maine coon cats of both genders, with an unknown status regarding the mutation at inclusion, were included prospectively and evaluated by conventional and strain imaging echocardiography. Comparisons were made among cats without hypertrophy (n = 45), suspect cats (n = 7), and cats with hypertrophic cardiomyopathy (n = 5), and also between the heterozygous for the mutation group (n = 26) and the negative for the mutation group (n = 28). Finally, in the group of phenotypically normal cats, heterozygous cats carrying the mutation were compared to cats without the mutation. Strain values were compared among the groups (blinded prospective study). While echocardiography demonstrated normal contractility, strain values (middle of the septum) were lower in HCM cats. Strain values (base of anterior wall of the left ventricle) were lower in heterozygous than in negative cats, even before hypertrophy. Negative correlation was observed between some values of myocardial strain and thickness. While strain imaging echocardiography was able to detect systolic abnormalities, despite apparent normality on conventional echocardiography, it was not able to identify cats that carry the A31P mutation in the MYBPC3 gene. Strain imaging echocardiography could be a useful tool, however, for detecting systolic alterations in HCM cats with an apparently normal systolic function or for detecting alterations in normal carriers of the MYBPC3 gene mutation.


Le dysfonctionnement myocardique se produit chez des chats avec une cardiomyopathie hypertrophique (CMH), mais peu de choses sont connues sur les stades initiaux de la maladie. L'échocardiographie par imagerie de déformation (strain imaging) est une méthode qui permet l'évaluation quantitative de la fonction myocardique et de la déformation, permettant ainsi la caractérisation de la dysfonction systolique. L'objectif de la présente étude était d'évaluer la fonction systolique en utilisant l'échographie par imagerie de déformation chez des chats de race Maine Coon testés génétiquement pour la mutation A31P dans le gène MYBPC3, avec et sans hypertrophie ventriculaire. À cette fin, 57 chats Maine Coon des deux sexes, avec un statut inconnu en regard de la mutation au moment de l'inclusion dans l'étude, ont été inclus de manière prospective et évalués par échographie conventionnelle et par imagerie de déformation. Des comparaisons ont été faites parmi les chats sans hypertrophie (n = 45), les chats suspects (n = 7), et les chats avec cardiomyopathie hypertrophique (n = 5), et également entre les hétérozygotes pour le groupe avec mutation (n = 26) et les négatifs pour le groupe avec mutation (n = 28). Finalement, dans le groupe de chats phénotypiquement normaux, les chats hétérozygotes porteurs de la mutation ont été comparés aux chats sans la mutation. Les valeurs de déformation ont été comparées parmi les groupes (étude prospective à l'aveugle). Alors que l'échographie a montré une contractilité normale, les valeurs de déformation (au milieu du septum) étaient plus faibles chez les chats avec CMH. Les valeurs de déformation (à la base de la paroi antérieure du ventricule gauche) étaient plus faibles chez les chats hétérozygotes que chez les chats négatifs, et ce même avant l'hypertrophie. Une corrélation négative fut observée entre quelques valeurs de déformation myocardique et d'épaisseur. Bien que l'échographie par imagerie de déformation était en mesure de détecter des anomalies systoliques, malgré une apparence de normalité lors de l'échographie conventionnelle, elle n'était pas en mesure d'identifier les chats porteurs de la mutation A31P dans le gène MYBPS3. L'échographie par imagerie de déformation pourrait toutefois être un outil utile pour détecter des altérations systoliques chez des chats CMH avec une fonction systolique apparemment normale ou pour détecter des altérations chez des porteurs normaux de la mutation dans le gène MYBPC3(Traduit par Docteur Serge Messier).


Assuntos
Cardiomiopatia Hipertrófica/veterinária , Proteínas de Transporte/metabolismo , Doenças do Gato/patologia , Ecocardiografia/veterinária , Função Ventricular Esquerda/fisiologia , Animais , Cardiomiopatia Hipertrófica/diagnóstico por imagem , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Proteínas de Transporte/genética , Doenças do Gato/diagnóstico por imagem , Doenças do Gato/genética , Gatos , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Masculino , Mutação
9.
Circ Heart Fail ; 9(9)2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27623770

RESUMO

BACKGROUND: The noninvasive assessment of altered myocardium in patients with genetic mutations that are associated with hypertrophic cardiomyopathy (HCM) remains challenging. In this pilot study, we evaluated whether a novel echocardiography-based assessment of myocardial microstructure, the signal intensity coefficient (SIC), could detect tissue-level alterations in HCM sarcomere mutation carriers with and without left ventricular hypertrophy. METHODS AND RESULTS: We studied 3 groups of genotyped individuals: sarcomere mutation carriers with left ventricular hypertrophy (clinical HCM; n=36), mutation carriers with normal left ventricular wall thickness (subclinical HCM; n=28), and healthy controls (n=10). We compared measurements of echocardiographic SIC with validated assessments of cardiac microstructural alteration, including cardiac magnetic resonance measures of interstitial fibrosis (extracellular volume fraction), as well as serum biomarkers (NTproBNP, hs-cTnI, and PICP). In age-, sex-, and familial relation-adjusted analyses, the SIC was quantitatively different across subjects with overt HCM, subclinical HCM, and healthy controls (P<0.001). Compared with controls, the SIC was 61% higher in overt HCM and 47% higher in subclinical HCM (P<0.001 for both). The SIC was significantly correlated with extracellular volume (r=0.72; P<0.01), with left ventricular mass and E' velocity (r=0.45, -0.60, respectively; P<0.01 for both), and with serum NTproBNP levels (r=0.36; P<0.001). CONCLUSIONS: Our findings suggest that the SIC could serve as a noninvasive quantitative tool for assessing altered myocardial tissue characteristics in patients with genetic mutations associated with HCM. Further studies are needed to determine whether the SIC could be used to identify subclinical changes in patients at risk for HCM and to evaluate the effects of interventions.


Assuntos
Cardiomiopatia Hipertrófica/diagnóstico por imagem , Ecocardiografia Doppler/métodos , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Mutação , Miocárdio/patologia , Adulto , Doenças Assintomáticas , Biomarcadores/sangue , Cardiomiopatia Hipertrófica/sangue , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Estudos de Casos e Controles , Estudos Transversais , Feminino , Predisposição Genética para Doença , Humanos , Hipertrofia Ventricular Esquerda/sangue , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/patologia , Processamento de Imagem Assistida por Computador , Imagem Cinética por Ressonância Magnética , Masculino , Fenótipo , Projetos Piloto , Valor Preditivo dos Testes , Prognóstico , Reprodutibilidade dos Testes , Adulto Jovem
10.
N Engl J Med ; 375(7): 655-65, 2016 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-27532831

RESUMO

BACKGROUND: For more than a decade, risk stratification for hypertrophic cardiomyopathy has been enhanced by targeted genetic testing. Using sequencing results, clinicians routinely assess the risk of hypertrophic cardiomyopathy in a patient's relatives and diagnose the condition in patients who have ambiguous clinical presentations. However, the benefits of genetic testing come with the risk that variants may be misclassified. METHODS: Using publicly accessible exome data, we identified variants that have previously been considered causal in hypertrophic cardiomyopathy and that are overrepresented in the general population. We studied these variants in diverse populations and reevaluated their initial ascertainments in the medical literature. We reviewed patient records at a leading genetic-testing laboratory for occurrences of these variants during the near-decade-long history of the laboratory. RESULTS: Multiple patients, all of whom were of African or unspecified ancestry, received positive reports, with variants misclassified as pathogenic on the basis of the understanding at the time of testing. Subsequently, all reported variants were recategorized as benign. The mutations that were most common in the general population were significantly more common among black Americans than among white Americans (P<0.001). Simulations showed that the inclusion of even small numbers of black Americans in control cohorts probably would have prevented these misclassifications. We identified methodologic shortcomings that contributed to these errors in the medical literature. CONCLUSIONS: The misclassification of benign variants as pathogenic that we found in our study shows the need for sequencing the genomes of diverse populations, both in asymptomatic controls and the tested patient population. These results expand on current guidelines, which recommend the use of ancestry-matched controls to interpret variants. As additional populations of different ancestry backgrounds are sequenced, we expect variant reclassifications to increase, particularly for ancestry groups that have historically been less well studied. (Funded by the National Institutes of Health.).


Assuntos
Negro ou Afro-Americano/genética , Cardiomiopatia Hipertrófica/genética , Reações Falso-Positivas , Predisposição Genética para Doença , Variação Genética , Adolescente , Adulto , Idoso , Asiático/genética , Criança , Exoma , Testes Genéticos , Genótipo , Disparidades nos Níveis de Saúde , Hispânico ou Latino/genética , Humanos , Pessoa de Meia-Idade , Mutação , Análise de Sequência de DNA , Estados Unidos , População Branca/genética , Adulto Jovem
11.
J Phys Chem B ; 120(33): 8264-75, 2016 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-27133568

RESUMO

Mutations in cardiac troponin C (D75Y, E59D, and G159D), a key regulatory protein of myofilament contraction, have been associated with dilated cardiomyopathy (DCM). Despite reports of altered myofilament function in these mutants, the underlying molecular alterations caused by these mutations remain elusive. Here we investigate in silico the intramolecular mechanisms by which these mutations affect myofilament contraction. On the basis of the location of cardiac troponin C (cTnC) mutations, we tested the hypothesis that intramolecular effects can explain the altered myofilament calcium sensitivity of force development for D75Y and E59D cTnC, whereas altered cardiac troponin C-troponin I (cTnC-cTnI) interaction contributes to the reported contractile effects of the G159D mutation. We employed a multiscale approach combining molecular dynamics (MD) and Brownian dynamics (BD) simulations to estimate cTnC calcium association and hydrophobic patch opening. We then integrated these parameters into a Markov model of myofilament activation to compute the steady-state force-pCa relationship. The analysis showed that myofilament calcium sensitivity with D75Y and E59D can be explained by changes in calcium binding affinity of cTnC and the rate of hydrophobic patch opening, if a partial cTnC interhelical opening angle (110°) is sufficient for cTnI switch peptide association to cTnC. In contrast, interactions between cTnC and cTnI within the cardiac troponin complex must also be accounted for to explain contractile alterations due to G159D. In conclusion, this is the first multiscale in silico study to elucidate how direct molecular effects of genetic mutations in cTnC translate to altered myofilament contractile function.


Assuntos
Cálcio/metabolismo , Mutação , Miofibrilas/metabolismo , Troponina C/metabolismo , Troponina I/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/fisiopatologia , Cristalografia por Raios X , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cadeias de Markov , Simulação de Dinâmica Molecular , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , Ratos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Troponina C/genética , Troponina I/genética
12.
Eur J Hum Genet ; 23(11): 1462-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25649380

RESUMO

The introduction of next-generation sequencing in everyday clinical genetics practise is increasing the number of genetic disorders that can be confirmed at DNA-level, and consequently increases the possibilities for cascade screening. This leads to a greater need for genetic counselling, whereas the number of professionals available to provide this is limited. We therefore piloted group genetic counselling for symptomatic cardiomyopathy patients at regional hospitals, to assess whether this could be an acceptable alternative to individual counselling. We performed a cohort study with pre- and post-counselling patient measurements using questionnaires, supplemented with evaluations of the group counselling format by the professionals involved. Patients from eight regional hospitals in the northern part of the Netherlands were included. Questionnaires comprised patient characteristics, psychological measures (personal perceived control (PPC), state and trait anxiety inventory (STAI)), and satisfaction with counsellors, counselling content and design. In total, 82 patients (mean age 57.5 year) attended one of 13 group sessions. Median PPC and STAI scores showed significantly higher control and lower anxiety after the counselling. Patients reported they were satisfied with the counsellors, and almost 75% of patients were satisfied with the group counselling. Regional professionals were also, overall, satisfied with the group sessions. The genetics professionals were less satisfied, mainly because of their perceived large time investment and less-than-expected group interaction. Hence, a group approach to cardiogenetic counselling is feasible, accessible, and psychologically effective, and could be one possible approach to counselling the increasing patient numbers in cardiogenetics.


Assuntos
Cardiomiopatia Dilatada/genética , Cardiomiopatia Hipertrófica/genética , Aconselhamento Genético , Adulto , Idoso , Cardiomiopatia Dilatada/epidemiologia , Cardiomiopatia Dilatada/psicologia , Cardiomiopatia Hipertrófica/epidemiologia , Cardiomiopatia Hipertrófica/psicologia , Análise Custo-Benefício , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Países Baixos , Satisfação Pessoal , Inquéritos e Questionários
13.
Genet Med ; 17(11): 880-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25611685

RESUMO

PURPOSE: Hypertrophic cardiomyopathy (HCM) is caused primarily by pathogenic variants in genes encoding sarcomere proteins. We report genetic testing results for HCM in 2,912 unrelated individuals with nonsyndromic presentations from a broad referral population over 10 years. METHODS: Genetic testing was performed by Sanger sequencing for 10 genes from 2004 to 2007, by HCM CardioChip for 11 genes from 2007 to 2011 and by next-generation sequencing for 18, 46, or 51 genes from 2011 onward. RESULTS: The detection rate is ~32% among unselected probands, with inconclusive results in an additional 15%. Detection rates were not significantly different between adult and pediatric probands but were higher in females compared with males. An expanded gene panel encompassing more than 50 genes identified only a very small number of additional pathogenic variants beyond those identifiable in our original panels, which examined 11 genes. Familial genetic testing in at-risk family members eliminated the need for longitudinal cardiac evaluations in 691 individuals. Based on the projected costs derived from Medicare fee schedules for the recommended clinical evaluations of HCM family members by the American College of Cardiology Foundation/American Heart Association, our data indicate that genetic testing resulted in a minimum cost savings of about $0.7 million. CONCLUSION: Clinical HCM genetic testing provides a definitive molecular diagnosis for many patients and provides cost savings to families. Expanded gene panels have not substantively increased the clinical sensitivity of HCM testing, suggesting major additional causes of HCM still remain to be identified.


Assuntos
Cardiomiopatia Hipertrófica/diagnóstico , Cardiomiopatia Hipertrófica/genética , Testes Genéticos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Cardiomiopatia Hipertrófica/epidemiologia , Criança , Pré-Escolar , Custos e Análise de Custo , Feminino , Predisposição Genética para Doença , Testes Genéticos/economia , Testes Genéticos/métodos , Testes Genéticos/normas , Variação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos/economia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Análise de Sequência com Séries de Oligonucleotídeos/normas , Sensibilidade e Especificidade , Adulto Jovem
15.
J Physiol ; 592(15): 3257-72, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24928957

RESUMO

The first mutation associated with hypertrophic cardiomyopathy (HCM) is the R403Q mutation in the gene encoding ß-myosin heavy chain (ß-MyHC). R403Q locates in the globular head of myosin (S1), responsible for interaction with actin, and thus motor function of myosin. Increased cross-bridge relaxation kinetics caused by the R403Q mutation might underlie increased energetic cost of tension generation; however, direct evidence is absent. Here we studied to what extent cross-bridge kinetics and energetics are related in single cardiac myofibrils and multicellular cardiac muscle strips of three HCM patients with the R403Q mutation and nine sarcomere mutation-negative HCM patients (HCMsmn). Expression of R403Q was on average 41 ± 4% of total MYH7 mRNA. Cross-bridge slow relaxation kinetics in single R403Q myofibrils was significantly higher (P < 0.0001) than in HCMsmn myofibrils (0.47 ± 0.02 and 0.30 ± 0.02 s(-1), respectively). Moreover, compared to HCMsmn, tension cost was significantly higher in the muscle strips of the three R403Q patients (2.93 ± 0.25 and 1.78 ± 0.10 µmol l(-1) s(-1) kN(-1) m(-2), respectively) which showed a positive linear correlation with relaxation kinetics in the corresponding myofibril preparations. This correlation suggests that faster cross-bridge relaxation kinetics results in an increase in energetic cost of tension generation in human HCM with the R403Q mutation compared to HCMsmn. Therefore, increased tension cost might contribute to HCM disease in patients carrying the R403Q mutation.


Assuntos
Miosinas Cardíacas/genética , Cardiomiopatia Hipertrófica/fisiopatologia , Relaxamento Muscular , Contração Miocárdica , Cadeias Pesadas de Miosina/genética , Sarcômeros/fisiologia , Trifosfato de Adenosina/metabolismo , Adulto , Idoso , Miosinas Cardíacas/metabolismo , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Cadeias Pesadas de Miosina/metabolismo , Sarcômeros/metabolismo
16.
Cardiovasc Res ; 103(2): 248-57, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24835277

RESUMO

AIMS: Disease mechanisms regarding hypertrophic cardiomyopathy (HCM) are largely unknown and disease onset varies. Sarcomere mutations might induce energy depletion for which until now there is no direct evidence at sarcomere level in human HCM. This study investigated if mutations in genes encoding myosin-binding protein C (MYBPC3) and myosin heavy chain (MYH7) underlie changes in the energetic cost of contraction in the development of human HCM disease. METHODS AND RESULTS: Energetic cost of contraction was studied in vitro by measurements of force development and ATPase activity in cardiac muscle strips from 26 manifest HCM patients (11 MYBPC3mut, 9 MYH7mut, and 6 sarcomere mutation-negative, HCMsmn). In addition, in vivo, the ratio between external work (EW) and myocardial oxygen consumption (MVO2) to obtain myocardial external efficiency (MEE) was determined in 28 pre-hypertrophic mutation carriers (14 MYBPC3mut and 14 MYH7mut) and 14 healthy controls using [(11)C]-acetate positron emission tomography and cardiovascular magnetic resonance imaging. Tension cost (TC), i.e. ATPase activity during force development, was higher in MYBPC3mut and MYH7mut compared with HCMsmn at saturating [Ca(2+)]. TC was also significantly higher in MYH7mut at submaximal, more physiological [Ca(2+)]. EW was significantly lower in both mutation carrier groups, while MVO2 did not differ. MEE was significantly lower in both mutation carrier groups compared with controls, showing the lowest efficiency in MYH7 mutation carriers. CONCLUSION: We provide direct evidence that sarcomere mutations perturb the energetic cost of cardiac contraction. Gene-specific severity of cardiac abnormalities may underlie differences in disease onset and suggests that early initiation of metabolic treatment may be beneficial, in particular, in MYH7 mutation carriers.


Assuntos
Miosinas Cardíacas/genética , Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/genética , Mutação/genética , Contração Miocárdica/genética , Cadeias Pesadas de Miosina/genética , Citoesqueleto de Actina/genética , Adulto , Idoso , Miosinas Cardíacas/metabolismo , Feminino , Regulação da Expressão Gênica , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Contração Miocárdica/fisiologia , Sarcômeros/genética , Sarcômeros/patologia
17.
Invest. clín ; 55(1): 23-31, mar. 2014. ilus, tab
Artigo em Inglês | LILACS | ID: lil-746282

RESUMO

Hypertrophic cardiomyopathy (HCM) is a cardiac disease, characterized by marked hypertrophy and genetic variability. HCM has been associated with sarcomere protein mutations, being cardiac b-myosin (coded by the MYH7 gene) and myosin binding protein C (coded by the MYBPC3 gene) the most frequently affected proteins. As in Venezuela only the clinical analysis are performed in HCM patients, we decided to search for genetic variations in the MYH7 gene. Coding regions, including the junction exon-intron of the MYH7 gene, were studied in 58 HCM patients, whose samples were collected at the ASCARDIO Hospital (Barquisimeto, Lara state, Venezuela) and 106 control subjects from the ASCARDIO Hospital and the IVIC (Barquisimeto Lara state and Miranda, Venezuela, respectively). The blood samples were analyzed by genomic DNA isolation, followed by polymerase chain reaction and sequence analysis. The screening of the MYH7 gene revealed eight already reported polymorphic variants, as well as two intronic variations in these HCM patients. Neither any missense mutations nor other pathological mutations in the MYH7 gene were found in the HCM patients.


La miocardiopatía hipertrófica (MH) es una enfermedad cardiaca primaria, caracterizada por una marcada hipertrofia y variabilidad genética. MH ha sido asociada con mutaciones en las proteínas del sarcómero, siendo la beta miosina cardiaca, codificada por el gen MYH7 y la proteína de unión a miosina C, codificada por el gen MYBPC3, las principalmente afectadas. En Venezuela únicamente se realiza el diagnóstico clínico de MH, por lo cual el objetivo principal de este trabajo fue realizar el análisis genético en los pacientes, iniciando con el gen MYH7. Para ello, se estudió la región codificante, incluyendo la región de unión exón-intron del gen MYH7 en 58 pacientes provenientes de ASCARDIO (Barquisimeto, estado Lara) y 106 controles provenientes de ASCARDIO e IVIC (estados Lara y Miranda, Venezuela). Se colectaron las muestras de sangre para el aislamiento del ADN genómico, se realizó la técnica de PCR, seguido del análisis de secuencias para la detección de mutaciones en pacientes y controles. Se encontraron 8 polimorfismos previamente reportados, y 2 variaciones intrónicas. No se encontraron mutaciones que involucraran un cambio de aminoácido en ninguno de los exones del gen MYH7 de la beta miosina cardiaca.


Assuntos
Adolescente , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Miosinas Cardíacas/genética , Cardiomiopatia Hipertrófica/genética , Variação Genética , Cadeias Pesadas de Miosina/genética , Cardiomiopatia Hipertrófica/epidemiologia , DNA , Análise Mutacional de DNA , Éxons/genética , Frequência do Gene , Testes Genéticos , Hipertrofia Ventricular Esquerda/epidemiologia , Hipertrofia Ventricular Esquerda/genética , Íntrons/genética , Polimorfismo de Nucleotídeo Único , Venezuela/epidemiologia
18.
Clin Genet ; 85(4): 365-70, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23590259

RESUMO

Mendelian cardiomyopathies and arrhythmias are characterized by an important genetic heterogeneity, rendering Sanger sequencing very laborious and expensive. As a proof of concept, we explored multiplex targeted high-throughput sequencing (HTS) as a fast and cost-efficient diagnostic method for individuals suffering from Mendelian cardiac disorders. We designed a DNA capture assay including all exons from 130 genes involved in cardiovascular Mendelian disorders and analysed simultaneously four samples by multiplexing. Two patients had familial hypertrophic cardiomyopathy (HCM) and two patients suffered from long QT syndrome (LQTS). In patient 1 with HCM, we identified two known pathogenic missense variants in the two most frequently mutated sarcomeric genes MYH7 and MYBPC. In patient 2 with HCM, a known acceptor splice site variant in MYBPC3 was found. In patient 3 with LQTS, two missense variants in the genes SCN5A and KCNQ were identified. Finally, in patient 4 with LQTS a known missense variant was found in MYBPC3, which is usually mutated in patients with cardiomyopathy. Our results showed that multiplex targeted HTS works as an efficient and cost-effective tool for molecular diagnosis of heterogeneous disorders in clinical practice and offers new insights in the pathogenesis of these complex diseases.


Assuntos
Cardiomiopatia Hipertrófica/genética , Sequenciamento de Nucleotídeos em Larga Escala , Síndrome do QT Longo/genética , Mutação , Idoso , Criança , Sequenciamento de Nucleotídeos em Larga Escala/economia , Humanos , Masculino , Pessoa de Meia-Idade
19.
JACC Cardiovasc Imaging ; 5(7): 702-11, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22789938

RESUMO

OBJECTIVES: The aim of this study was to analyze the geometric pattern of hypertrophy (HT) in patients with asymmetrical septal hypertrophic cardiomyopathy (HCM) using cardiac magnetic resonance (CMR) and to test the hypothesis that at least in some patients, the HT follows a longitudinal spiral pattern. BACKGROUND: The highly heterogeneous phenotypic expression of HCM is a well-known phenomenon. CMR has emerged as a robust 3-dimensional (3D) tomographic imaging technique that is increasingly used to explore phenotypic expression. METHODS: Short-axis cine CMR was used to study the 3D extent of HT (i.e., radial, circumferential, and longitudinal extent, as well as the relation between circumferential and longitudinal extent). Inclusion criteria were septal wall thickness (WT) ≥15 mm and septal to free wall WT ratio >1.3. RESULTS: CMR was performed in 132 patients. Maximal WT was 22 ± 5 mm, with a circumferential extent of 131 ± 51°, and a longitudinal extent of 64 ± 19%, resulting in a hypertrophied left ventricular (LV) surface of 26 ± 15%. Linear regression analysis showed in 86% of patients a consistent course of HT along the longitudinal direction. The HT invariably started at the basal anteroseptum and rotated, except in 2 patients, in a counterclockwise direction (CC-spiral patients) with a mean global rotation of 116 ± 68° (range 5° to 350°). After the CC-spiral patients were divided according to magnitude of rotation quartiles (Q1: 5° to 70°, Q2: 75° to 105°, Q3: 110° to 150°, and Q4: 155° to 350°), Q4 patients were significantly older and had more LV outflow tract obstruction and hypertension than patients without the spiraling pattern. In 11 patients, continuation of HT into an apical form of HCM was found. CONCLUSIONS: Using 3D analysis, we found that the majority of patients with asymmetrical septal HCM in fact showed a spiral pattern of HT following a counterclockwise (or "left-handed") spiral trajectory. The variation in magnitude of rotation among patients, however, was highly variable. Further research is warranted to better understand the significance of the current findings, in particular to relate them to the genetic and morphological substrate, hemodynamic consequences, and patient outcome.


Assuntos
Cardiomiopatia Hipertrófica/diagnóstico , Septos Cardíacos/patologia , Hipertrofia Ventricular Esquerda/diagnóstico , Imagem Cinética por Ressonância Magnética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Bélgica , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/fisiopatologia , Distribuição de Qui-Quadrado , Progressão da Doença , Feminino , Predisposição Genética para Doença , Humanos , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Interpretação de Imagem Assistida por Computador , Imageamento Tridimensional , Itália , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Fenótipo , Valor Preditivo dos Testes , Índice de Gravidade de Doença , Volume Sistólico , Função Ventricular Esquerda , Adulto Jovem
20.
Eur J Hum Genet ; 20(10): 1018-23, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22453290

RESUMO

Since the 1990s, many countries in Europe and the United States have enacted genetic non-discrimination legislation to prevent people from deferring genetic tests for fear that insurers or employers would discriminate against them based on that information. Although evidence for genetic discrimination exists, little is known about the origins and backgrounds of fears of discrimination and how it affects decisions for uptake of genetic testing. The aim of this article is to gain a better understanding of these fears and its possible impact on the uptake of testing by studying the case of hypertrophic cardiomyopathy (HCM). In a qualitative study, we followed six Dutch extended families involved in genetic testing for HCM for three-and-a-half years. Semi-structured interviews were conducted with 57 members of these families. Based on the narratives of the families, we suggest that fears of discrimination have to be situated in the broader social and life-course context of family and kin. We describe the processes in which families developed meaningful interpretations of genetic discrimination and how these interpretations affected family members' decisions to undergo genetic testing. Our findings show that fears of genetic discrimination do not so much stem from the opportunity of genetic testing but much more from earlier experiences of discrimination of diseased family members. These results help identify the possible limitations of genetic non-discrimination regulations and provide direction to clinicians supporting their clients as they confront issues of genetic testing and genetic discrimination.


Assuntos
Cardiomiopatia Hipertrófica/genética , Medo , Testes Genéticos/legislação & jurisprudência , Discriminação Social/legislação & jurisprudência , Cardiomiopatia Hipertrófica/psicologia , Família , Privacidade Genética/ética , Privacidade Genética/legislação & jurisprudência , Testes Genéticos/ética , Humanos , Seguro Saúde/ética , Seguro Saúde/legislação & jurisprudência , Países Baixos , Discriminação Social/ética , Discriminação Social/psicologia , Meio Social
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA