Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Dis Model Mech ; 13(12)2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33033063

RESUMO

The causal genetic underpinnings of congenital heart diseases, which are often complex and multigenic, are still far from understood. Moreover, there are also predominantly monogenic heart defects, such as cardiomyopathies, with known disease genes for the majority of cases. In this study, we identified mutations in myomesin 2 (MYOM2) in patients with Tetralogy of Fallot (TOF), the most common cyanotic heart malformation, as well as in patients with hypertrophic cardiomyopathy (HCM), who do not exhibit any mutations in the known disease genes. MYOM2 is a major component of the myofibrillar M-band of the sarcomere, and a hub gene within interactions of sarcomere genes. We show that patient-derived cardiomyocytes exhibit myofibrillar disarray and reduced passive force with increasing sarcomere lengths. Moreover, our comprehensive functional analyses in the Drosophila animal model reveal that the so far uncharacterized fly gene CG14964 [herein referred to as Drosophila myomesin and myosin binding protein (dMnM)] may be an ortholog of MYOM2, as well as other myosin binding proteins. Its partial loss of function or moderate cardiac knockdown results in cardiac dilation, whereas more severely reduced function causes a constricted phenotype and an increase in sarcomere myosin protein. Moreover, compound heterozygous combinations of CG14964 and the sarcomere gene Mhc (MYH6/7) exhibited synergistic genetic interactions. In summary, our results suggest that MYOM2 not only plays a critical role in maintaining robust heart function but may also be a candidate gene for heart diseases such as HCM and TOF, as it is clearly involved in the development of the heart.This article has an associated First Person interview with Emilie Auxerre-Plantié and Tanja Nielsen, joint first authors of the paper.


Assuntos
Cardiomiopatia Hipertrófica/genética , Conectina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Estudos de Associação Genética , Proteínas de Membrana/genética , Tetralogia de Fallot/genética , Animais , Proteínas de Drosophila/metabolismo , Feminino , Humanos , Locomoção , Masculino , Proteínas de Membrana/metabolismo , Músculos/metabolismo , Mutação/genética , Miocárdio , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miofibrilas/metabolismo , Miofibrilas/patologia , Especificidade de Órgãos , Ligação Proteica , Mapeamento de Interação de Proteínas
2.
Rev Port Cardiol (Engl Ed) ; 39(6): 317-327, 2020 Jun.
Artigo em Inglês, Português | MEDLINE | ID: mdl-32565061

RESUMO

INTRODUCTION AND OBJECTIVES: Hypertrophic cardiomyopathy (HCM) is a genetically and phenotypically heterogeneous disease; there is still a large proportion of patients with no identified disease-causing mutation. Although the majority of mutations are found in the MYH7 and MYBPC3 genes, mutations in Z-disk-associated proteins have also been linked to HCM. METHODS: We assessed a small family with HCM based on family history, physical examination, 12-lead ECG, echocardiogram and magnetic resonance imaging. After exclusion of mutations in eleven HCM disease genes, we performed direct sequencing of the TCAP gene encoding the Z-disk protein titin-cap (also known as telethonin). RESULTS: We present a novel TCAP mutation in a small family affected by HCM. The identified p.C57W mutation showed a very low population frequency, as well as high conservation across species. All of the bioinformatic prediction tools used considered this mutation to be damaging/deleterious. Family members were screened for this new mutation and a co-segregation pattern was detected. Both affected members of this family presented with late-onset HCM, moderate asymmetric left ventricular hypertrophy, atrial fibrillation and heart failure with preserved ejection fraction and low risk of sudden cardiac death. CONCLUSIONS: We present evidence supporting the classification of the TCAP p.C57W mutation, encoding the Z-disk protein titin-cap/telethonin as a new likely pathogenic variant of hypertrophic cardiomyopathy, with a specific phenotype in the family under analysis.


Assuntos
Cardiomiopatia Hipertrófica , Proteínas de Transporte , Conectina , Proteínas de Transporte/genética , Conectina/genética , Humanos , Mutação , Portugal
3.
J Interv Card Electrophysiol ; 51(3): 215-220, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29460234

RESUMO

PURPOSE: Catheter ablation is performed under fluoroscopic guidance. Reduction of radiation dose for patients and staff is emphasized by current recommendations. Previous studies have shown that lower operator experience leads to increased radiation dose. On the other hand, less experienced operators may depend even more on fluoroscopic guidance. Our study aimed to evaluate feasibility and efficacy of a non-fluoroscopic approach in different training levels. METHODS: From January 2017, a near-zero fluoroscopy approach was established in two centers. Four operators (beginner, 1st year fellow, 2nd year fellow, expert) were instructed to perform the complete procedure with the use of a 3-D mapping system without fluoroscopy. A historical cohort that underwent procedures with fluoroscopy use served as control group. Dose area product (DPA), procedure duration, acute procedural success, and complications were compared between the groups and for each operator. RESULTS: Procedures were performed in 157 patients. The first 100 patients underwent procedures with fluoroscopic guidance, the following 57 procedures were performed with the near-zero fluoroscopy approach. The results show a significant reduction in DPA for all operators immediately after implementation of the near-zero fluoroscopy protocol (control 637 ± 611 µGy/m2; beginner 44.1 ± 79.5 µGy/m2, p = 0.002; 1st year fellow 24.3 ± 46.4.5 µGy/m2, p = 0.001; 2nd year fellow 130.3 ± 233.3 µGy/m2, p = 0.003; expert 9.3 ± 37.4 µGy/m2, P < 0.001). Procedure duration, acute success, and complications were not significantly different between the groups. CONCLUSION: Our results show a 90% reduction of DPA shortly after implementation of a near-zero fluoroscopy approach in interventional electrophysiology even in operators in training.


Assuntos
Arritmias Cardíacas/cirurgia , Ablação por Cateter/métodos , Doses de Radiação , Exposição à Radiação/prevenção & controle , Monitoramento de Radiação/métodos , Idoso , Análise de Variância , Arritmias Cardíacas/diagnóstico , Mapeamento Potencial de Superfície Corporal/métodos , Eletrofisiologia Cardíaca/métodos , Estudos de Casos e Controles , Feminino , Fluoroscopia/efeitos adversos , Humanos , Masculino , Pessoa de Meia-Idade , Valores de Referência , Medição de Risco , Estatísticas não Paramétricas
4.
J Mol Cell Cardiol ; 113: 9-21, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28941705

RESUMO

The ability to generate patient-specific induced pluripotent stem cells (iPSCs) provides a unique opportunity for modeling heart disease in vitro. In this study, we generated iPSCs from a patient with dilated cardiomyopathy (DCM) caused by a missense mutation S635A in RNA-binding motif protein 20 (RBM20) and investigated the functionality and cell biology of cardiomyocytes (CMs) derived from patient-specific iPSCs (RBM20-iPSCs). The RBM20-iPSC-CMs showed abnormal distribution of sarcomeric α-actinin and defective calcium handling compared to control-iPSC-CMs, suggesting disorganized myofilament structure and altered calcium machinery in CMs of the RBM20 patient. Engineered heart muscles (EHMs) from RBM20-iPSC-CMs showed that not only active force generation was impaired in RBM20-EHMs but also passive stress of the tissue was decreased, suggesting a higher visco-elasticity of RBM20-EHMs. Furthermore, we observed a reduced titin (TTN) N2B-isoform expression in RBM20-iPSC-CMs by demonstrating a reduction of exon skipping in the PEVK region of TTN and an inhibition of TTN isoform switch. In contrast, in control-iPSC-CMs both TTN isoforms N2B and N2BA were expressed, indicating that the TTN isoform switch occurs already during early cardiogenesis. Using next generation RNA sequencing, we mapped transcriptome and splicing target profiles of RBM20-iPSC-CMs and identified different cardiac gene networks in response to the analyzed RBM20 mutation in cardiac-specific processes. These findings shed the first light on molecular mechanisms of RBM20-dependent pathological cardiac remodeling leading to DCM. Our data demonstrate that iPSC-CMs coupled with EHMs provide a powerful tool for evaluating disease-relevant functional defects and for a deeper mechanistic understanding of alternative splicing-related cardiac diseases.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Adulto , Animais , Cálcio/metabolismo , Células Cultivadas , Conectina/metabolismo , Feminino , Humanos , Camundongos , Mutação , Fenótipo , Splicing de RNA/genética , Sarcômeros/metabolismo , Transcriptoma/genética
5.
FASEB J ; 31(2): 556-568, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28148779

RESUMO

Cardiac diseases are the leading cause of death. Available treatment approaches are not sufficient to reverse persistent cardiac damage after injury; thus, the search for new therapeutic targets is essential. Our microarray-based screening in rat hearts 24 h after myocardial infarction (MI) yielded glycoprotein nonmetastatic melanoma protein B (GPNMB), which is known to be involved in inflammation and fibrosis after tissue injury. However, its role in the heart was elusive. We found increased cardiac expression levels of GPNMB in rats and mice after MI. Analysis of DBA/2J mice, which lack functional GPNMB due to a spontaneous point mutation, showed that systemic GPNMB deficiency was associated with preserved cardiac function and less left ventricular dilation after MI compared with DBA/2J mice with reconstituted GPNMB expression. These improvements were associated with decreased expression of matrix metalloproteinase 9, the cardiac stress genes for natriuretic peptides (atrial natriuretic peptide and brain natriuretic peptide), and ß-myosin heavy chain after MI. Moreover, GPNMB deficiency attenuated the dilated cardiomyopathy in muscle lim protein knockout mice but could not prevent cardiac hypertrophy induced by isoprenaline infusion. This is the first experimental study to show that GPNMB adversely influences myocardial remodeling.-Järve, A., Mühlstedt, S., Qadri, F., Nickl, B., Schulz, H., Hübner, N., Özcelik, C., Bader, M. Adverse left ventricular remodeling by glycoprotein nonmetastatic melanoma protein B in myocardial infarction.


Assuntos
Proteínas do Olho/metabolismo , Glicoproteínas de Membrana/metabolismo , Infarto do Miocárdio/metabolismo , Remodelação Ventricular/fisiologia , Animais , Proteínas do Olho/genética , Regulação da Expressão Gênica/fisiologia , Inflamação , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação Puntual , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Células-Tronco/fisiologia
6.
Cardiovasc Res ; 112(3): 637-644, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27496871

RESUMO

AIMS: B-type natriuretic peptide (BNP)-natriuretic peptide receptor A (NPR-A) receptor signalling inhibits cardiac sympathetic neurotransmission, although C-type natriuretic peptide (CNP) is the predominant neuropeptide of the nervous system with expression in the heart and vasculature. We hypothesized that CNP acts similarly to BNP, and that transgenic rats (TGRs) with neuron-specific overexpression of a dominant negative NPR-B receptor would develop heightened sympathetic drive. METHODS AND RESULTS: Mean arterial pressure and heart rate (HR) were significantly (P < 0.05) elevated in freely moving TGRs (n = 9) compared with Sprague Dawley (SD) controls (n = 10). TGR had impaired left ventricular systolic function and spectral analysis of HR variability suggested a shift towards sympathoexcitation. Immunohistochemistry demonstrated co-staining of NPR-B with tyrosine hydroxylase in stellate ganglia neurons. In SD rats, CNP (250 nM, n = 8) significantly reduced the tachycardia during right stellate ganglion stimulation (1-7 Hz) in vitro whereas the response to bath-applied norepinephrine (NE, 1 µM, n = 6) remained intact. CNP (250 nM, n = 8) significantly reduced the release of 3H-NE in isolated atria and this was prevented by the NPR-B antagonist P19 (250 nM, n = 6). The neuronal Ca2+ current (n = 6) and intracellular Ca2+ transient (n = 9, using fura-2AM) were also reduced by CNP in isolated stellate neurons. Treatment of the TGR (n = 9) with the sympatholytic clonidine (125 µg/kg per day) significantly reduced mean arterial pressure and HR to levels observed in the SD (n = 9). CONCLUSION: C-type natriuretic peptide reduces cardiac sympathetic neurotransmission via a reduction in neuronal calcium signalling and NE release through the NPR-B receptor. Situations impairing CNP-NPR-B signalling lead to hypertension, tachycardia, and impaired left ventricular systolic function secondary to sympatho-excitation.


Assuntos
Coração/inervação , Peptídeo Natriurético Tipo C/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Sistema Nervoso Simpático/metabolismo , Transmissão Sináptica , Animais , Pressão Arterial , Sinalização do Cálcio , Predisposição Genética para Doença , Frequência Cardíaca , Hipertensão/genética , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Norepinefrina/metabolismo , Fenótipo , Ratos Sprague-Dawley , Ratos Transgênicos , Receptores do Fator Natriurético Atrial/genética , Gânglio Estrelado/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Sístole , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda
7.
J Mol Med (Berl) ; 94(9): 1005-14, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27251706

RESUMO

UNLABELLED: The chemokine CXCL12/SDF-1 is crucial for heart development and affects cardiac repair processes due to its ability to attract leukocytes and stem cells to injured myocardium. However, there is a great controversy whether CXCL12 is beneficial or detrimental after myocardial infarction (MI). The divergence in the reported CXCL12 actions may be due to the cellular source and time of release of the chemokine after MI. This study was designed to evaluate the role of cardiomyocyte-derived CXCL12 for cardiogenesis and heart repair after MI. We generated two rodent models each targeting CXCL12 in only one cardiac cell type: cardiomyocyte-specific CXCL12-overexpressing transgenic (Tg) rats and CXCL12 conditional knockout (cKO) mice. Animals of both models did not show any signs of cardiac abnormalities under baseline conditions. After induction of MI, cKO mice displayed preserved cardiac function and remodeling. Moreover, fibrosis was less pronounced in the hearts of cKO mice after MI. Accordingly, CXCL12 Tg rats revealed impaired cardiac function post-MI accompanied by enhanced fibrosis. Furthermore, we observed decreased numbers of infiltrating Th1 cells in the hearts of cKO mice. Collectively, our findings demonstrate that cardiomyocyte-derived CXCL12 is not involved in cardiac development but has adverse effects on the heart after injury via promotion of inflammation and fibrosis. KEY MESSAGES: • CXCL12 in cardiomyocytes is not involved in cardiac development. • CXCL12 deficiency in cardiomyocytes improves outcome of myocardial infarction. • CXCL12 overexpression in cardiomyocytes worsens outcome of myocardial infarction. • CXCL12 increases fibrosis and invasion of Th1 cells in the heart after infarction.


Assuntos
Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Organogênese/genética , Animais , Biópsia , Modelos Animais de Doenças , Fibrose , Expressão Gênica , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Especificidade de Órgãos/genética , Prognóstico , Ratos , Ratos Transgênicos , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia
8.
Arch Med Sci ; 12(2): 263-78, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27186169

RESUMO

INTRODUCTION: Transgenic mice overexpressing mutated NEBL, encoding the cardiac-specific Z-disk protein nebulette, develop severe cardiac phenotypes. Since cardiomyopathies are commonly familial and because mutations in a single gene may result in variable phenotypes, we tested the hypothesis that NEBL mutations are associated with cardiomyopathy. MATERIAL AND METHODS: We analyzed 389 patients, including cohorts of patients with dilated cardiomyopathy (DCM), hypertrophic cardiomyopathy (HCM), and left ventricular non-compaction cardiomyopathy (LVNC). The 28 coding exons of the NEBL gene were sequenced. Further bioinformatic analysis was used to distinguish variants. RESULTS: In total, we identified six very rare heterozygous missense mutations in NEBL in 7 different patients (frequency 1.8%) in highly conserved codons. The mutations were not detectable in 320 Caucasian sex-matched unrelated individuals without cardiomyopathy and 192 Caucasian sex-matched blood donors without heart disease. Known cardiomyopathy genes were excluded in these patients. The mutations p.H171R and p.I652L were found in 2 HCM patients. Further, p.Q581R and p.S747L were detected in 2 DCM patients, while the mutation p.A175T was identified independently in two unrelated patients with DCM. One LVNC patient carried the mutation p.P916L. All HCM and DCM related mutations were located in the nebulin-like repeats, domains responsible for actin binding. Interestingly, the mutation associated with LVNC was located in the C-terminal serine-rich linker region. CONCLUSIONS: Our data suggest that NEBL mutations may cause various cardiomyopathies. We herein describe the first NEBL mutations in HCM and LVNC. Our findings underline the notion that the cardiomyopathies are true allelic diseases.

9.
Pediatr Cardiol ; 36(2): 295-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25135600

RESUMO

The genetic basis of congenital heart disease remains unknown in most of the cases. Recently, a novel mouse model shed new light on the role of CCN1/CYR61, a matricellular regulatory factor, in cardiac morphogenesis. In a candidate gene approach, we analyzed a cohort of 143 patients with atrial septal defects (ASD) by sequencing the coding exons of CCN1. In addition to three frequent polymorphisms, we identified an extremely rare novel heterozygous missense mutation (c.139C > T; p.R47W) in one patient with severe ASD. The mutation leads to an exchange of residues with quite different properties in a highly conserved position of the N-terminal insulin-like growth factor binding protein module. Further bioinformatic analysis, exclusion of known ASD disease genes as well as the exclusion of the mutation in a very high number of ethnically matched controls (more than 1,000 individuals) and in public genetic databases, indicates that the p.R47W variant is a probable disease-associated mutation. The report about ASD in mice in heterozygous Ccn 1 +/- animals strongly supports this notion. Our study is the first to suggest a relationship between a probable CCN1 mutation and ASD. Our purpose here was to draw attention to CCN1, a gene that we believe may be important for genetic analysis in patients with congenital heart disease.


Assuntos
Proteína Rica em Cisteína 61/genética , Comunicação Interatrial/genética , Adulto , Análise Mutacional de DNA , Feminino , Estudos de Associação Genética , Variação Genética , Comunicação Interatrial/diagnóstico por imagem , Humanos , Masculino , Mutação , Polimorfismo de Nucleotídeo Único/genética , Ultrassonografia
10.
Front Mol Neurosci ; 7: 95, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25520616

RESUMO

The second messenger cyclic GMP affects synaptic transmission and modulates synaptic plasticity and certain types of learning and memory processes. The impact of the natriuretic peptide receptor B (NPR-B) and its ligand C-type natriuretic peptide (CNP), one of several cGMP producing signaling systems, on hippocampal synaptic plasticity and learning is, however, less well understood. We have previously shown that the NPR-B ligand CNP increases the magnitude of long-term depression (LTD) in hippocampal area CA1, while reducing the induction of long-term potentiation (LTP). We have extended this line of research to show that bidirectional plasticity is affected in the opposite way in rats expressing a dominant-negative mutant of NPR-B (NSE-NPR-BΔKC) lacking the intracellular guanylyl cyclase domain under control of a promoter for neuron-specific enolase. The brain cells of these transgenic rats express functional dimers of the NPR-B receptor containing the dominant-negative NPR-BΔKC mutant, and therefore show decreased CNP-stimulated cGMP-production in brain membranes. The NPR-B transgenic rats display enhanced LTP but reduced LTD in hippocampal slices. When the frequency-dependence of synaptic modification to afferent stimulation in the range of 1-100 Hz was assessed in transgenic rats, the threshold for both, LTP and LTD induction, was shifted to lower frequencies. In parallel, NPR-BΔKC rats exhibited an enhancement in exploratory and learning behavior. These results indicate that bidirectional plasticity and learning and memory mechanism are affected in transgenic rats expressing a dominant-negative mutant of NPR-B. Our data substantiate the hypothesis that NPR-B-dependent cGMP signaling has a modulatory role for synaptic information storage and learning.

11.
Int J Cardiol ; 168(4): 3647-51, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23711456

RESUMO

BACKGROUND: Single nucleotide polymorphisms (SNPs) of EPHX2 alter sEH activity and are associated with increased [rs41507953 (K55R)] or reduced [rs751141 (R287Q)] cardiovascular risk via modulation of fibrosis, inflammation or cardiac ion channels. This indicates an effect on development and therapy response of AF. This study tested the hypothesis that variations in the EPHX2 gene encoding human soluble epoxide hydrolase (sEH) are associated with atrial fibrillation (AF) and recurrence of atrial fibrillation after catheter ablation. METHODS AND RESULTS: A total of 218 consecutive patients who underwent catheter ablation for drug refractory AF and 268 controls were included. Two SNPs, rs41507953 and rs751141, were genotyped by direct sequencing. In the ablation group, holter recordings 3, 12 and 24 months after ablation were used to detect AF recurrence. No significant association of the SNPs and AF at baseline was detected. In the ablation group, recurrence of AF occurred in 20% of the patients 12 months after ablation and in 35% 24 months after ablation. The presence of the rs751141 polymorphism significantly increased the risk of AF recurrence 12 months (odds ratio [OR]: 3.2, 95% confidence interval [CI]: 1.237 to 8.276, p=0.016) and 24 months (OR: 6.076, 95% CI: 2.244 to 16.451, p<0.0001) after catheter ablation. CONCLUSIONS: The presence of rs751141 polymorphism is associated with a significantly increased risk of AF recurrence after catheter ablation. These results point to stratification of catheter ablation by genotype and differential use of sEH-inhibitory drugs in the future.


Assuntos
Fibrilação Atrial/genética , Fibrilação Atrial/cirurgia , Ablação por Cateter/tendências , Epóxido Hidrolases/genética , Variação Genética/genética , Idoso , Fibrilação Atrial/enzimologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Recidiva , Fatores de Risco , Resultado do Tratamento
12.
PLoS One ; 8(5): e64243, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23717576

RESUMO

Dystrophic epidermolysis bullosa, a severely disabling hereditary skin fragility disorder, is caused by mutations in the gene coding for collagen VII, a specialized adhesion component of the dermal-epidermal junction zone. Both recessive and dominant forms are known; the latter account for about 40% of cases. Patients with dominant dystrophic epidermolysis bullosa exhibit a spectrum of symptoms ranging from mild localized to generalized skin manifestations. Individuals with the same mutation can display substantial phenotypic variance, emphasizing the role of modifying genes in this disorder. The etiology of dystrophic epidermolysis bullosa has been known for around two decades; however, important pathogenetic questions such as involvement of modifier genes remain unanswered and a causative therapy has yet to be developed. Much of the failure to make progress in these areas is due to the lack of suitable animal models that capture all aspects of this complex monogenetic disorder. Here, we report the first rat model of dominant dystrophic epidermolysis bullosa. Affected rats carry a spontaneous glycine to aspartic acid substitution, p.G1867D, within the main structural domain of collagen VII. This confers dominant-negative interference of protein folding and decreases the stability of mutant collagen VII molecules and their polymers, the anchoring fibrils. The phenotype comprises fragile and blister-prone skin, scarring and nail dystrophy. The model recapitulates all signs of the human disease with complete penetrance. Homozygous carriers of the mutation are more severely affected than heterozygous ones, demonstrating for the first time a gene-dosage effect of mutated alleles in dystrophic epidermolysis bullosa. This novel viable and workable animal model for dominant dystrophic epidermolysis bullosa will be valuable for addressing molecular disease mechanisms, effects of modifying genes, and development of novel molecular therapies for patients with dominantly transmitted skin disease.


Assuntos
Substituição de Aminoácidos , Colágeno Tipo VII/genética , Epidermólise Bolhosa Distrófica/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Colágeno Tipo VII/metabolismo , Análise Mutacional de DNA , Modelos Animais de Doenças , Epidermólise Bolhosa Distrófica/metabolismo , Dosagem de Genes , Expressão Gênica , Estudos de Associação Genética , Humanos , Dados de Sequência Molecular , Estabilidade Proteica , Ratos , Pele/patologia
13.
Circ Heart Fail ; 5(5): 610-20, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22874137

RESUMO

BACKGROUND: The aim of this study was to investigate the myocardial systolic and diastolic performance of the left ventricle (LV) in patients with heart failure with normal LV ejection fraction (HFNEF) through novel LV myocardial indices, which assess the systolic and diastolic function of the whole myocardium of the LV. METHODS AND RESULTS: LV myocardial systolic and diastolic performance were assessed as the average value of peak systolic strain and peak early-diastolic strain rate, respectively, in longitudinal, circumferential, and radial directions from all LV segments using 2-dimensional speckle-tracking echocardiography. We studied patients with HFNEF and a control group consisting of asymptomatic subjects with LV diastolic dysfunction of similar age, sex, and LV ejection fraction. A total of 322 patients were included (92 with HFNEF and 230 with asymptomatic LV diastolic dysfunction). Myocardial systolic and diastolic LV performance were significantly lower in HFNEF (20.13±6.02% and 1.14±0.27 s(-1)) than in patients with asymptomatic LV diastolic dysfunction (25.33±6.06% and 1.37±0.33 s(-1), respectively; all P<0.0001). In addition, patients with HFNEF with low systolic and diastolic LV myocardial performance had significantly higher LV filling pressures (17.1±6.6 and 17.6±6.3 versus 12.0±5.1 and 11.7±4.7, respectively; all P<0.001) and lower cardiac output (4.8±1.0 L/min and 4.9±1.1 L/min versus 5.7±1.2 L/min and 5.8±1.1 L/min, respectively; all P<0.001) than patients with normal LV myocardial performance. In relation to these findings, the symptomatic status (ie, New York Heart Association functional class) was significantly altered in those patients with low systolic and diastolic LV myocardial performance. CONCLUSIONS: In patients with HFNEF, both systolic and diastolic LV myocardial performance are impaired, which is associated with increased LV filling pressures, decreased cardiac output, and worse New York Heart Association functional class. Therefore, the measurement of these myocardial parameters could be of great importance in HFNEF because these echocardiographic indices assess the multidirectional function of the whole myocardium of the LV, thereby allowing detection of an alteration of the global function of the LV which is associated with a worse symptomatic status in these patients.


Assuntos
Ecocardiografia Doppler , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/fisiopatologia , Contração Miocárdica , Volume Sistólico , Função Ventricular Esquerda , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , Diástole , Ecocardiografia Doppler de Pulso , Feminino , Alemanha , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Reprodutibilidade dos Testes , Sístole , Pressão Ventricular
14.
Europace ; 14(9): 1369-70, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22628451

RESUMO

A 32-year-old woman was admitted with a third-degree atrioventricular block. A permanent pacemaker was implanted and the patient was discharged. One week later, the patient presented again with a sustained ventricular tachycardia. Coronary angiography and computed tomography imaging with three-dimensional reconstructions revealed the absence of the proximal part of the right coronary artery (RCA) with a fistula into the pulmonary vein. This is the first case describing an absent proximal RCA combined with a pulmonary vein fistula.


Assuntos
Fístula Arteriovenosa/diagnóstico , Anomalias dos Vasos Coronários/diagnóstico , Veias Pulmonares/anormalidades , Adulto , Arritmias Cardíacas/diagnóstico , Fístula Arteriovenosa/diagnóstico por imagem , Angiografia Coronária/métodos , Anomalias dos Vasos Coronários/diagnóstico por imagem , Eletrocardiografia , Feminino , Bloqueio Cardíaco/diagnóstico , Bloqueio Cardíaco/diagnóstico por imagem , Humanos , Veias Pulmonares/diagnóstico por imagem , Recidiva , Síncope/diagnóstico , Tomografia Computadorizada por Raios X/métodos
15.
Nat Med ; 18(5): 766-73, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22466703

RESUMO

Alternative splicing has a major role in cardiac adaptive responses, as exemplified by the isoform switch of the sarcomeric protein titin, which adjusts ventricular filling. By positional cloning using a previously characterized rat strain with altered titin mRNA splicing, we identified a loss-of-function mutation in the gene encoding RNA binding motif protein 20 (Rbm20) as the underlying cause of pathological titin isoform expression. The phenotype of Rbm20-deficient rats resembled the pathology seen in individuals with dilated cardiomyopathy caused by RBM20 mutations. Deep sequencing of the human and rat cardiac transcriptome revealed an RBM20-dependent regulation of alternative splicing. In addition to titin (TTN), we identified a set of 30 genes with conserved splicing regulation between humans and rats. This network is enriched for genes that have previously been linked to cardiomyopathy, ion homeostasis and sarcomere biology. Our studies emphasize the key role of post-transcriptional regulation in cardiac function and provide mechanistic insights into the pathogenesis of human heart failure.


Assuntos
Cardiomiopatia Dilatada/genética , Proteínas Musculares/genética , Proteínas Quinases/genética , Splicing de RNA , Proteínas de Ligação a RNA/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Conectina , Humanos , Proteínas com Domínio LIM/genética , Dados de Sequência Molecular , Mutação , Proteínas de Ligação a RNA/fisiologia , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344
16.
Eur Heart J Cardiovasc Imaging ; 13(7): 556-67, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22383372

RESUMO

AIMS: The purpose of this study was to test the hypothesis that left ventricular (LV) mechanical dyssynchrony deteriorates the longitudinal systolic and diastolic function of the left ventricle (LV) in patients with heart failure with a normal LV ejection fraction (HFNEF). METHODS AND RESULTS: In patients with HFNEF and in a control group consisting of asymptomatic patients with LV diastolic dysfunction [LVDD], matched by age, gender, and LV ejection fraction, we assessed the global longitudinal systolic (global strain), diastolic [global early-diastolic strain rate (SRe)], and synchronous (standard deviation of time-to-peak systolic strain) function of the LV by two-dimensional speckle-tracking echocardiography using a 18-segment LV model. A total of 325 patients were included (85 with HFNEF and 240 with asymptomatic LVDD). Patients with HFNEF had a significant impairment of the longitudinal systolic and diastolic function of the LV as compared with the control group. Concerning the pathophysiological mechanisms linked to these findings, we found that HFNEF patients with asynchronous LV contractions had significantly more impaired longitudinal systolic and diastolic LV function (global strain -14.76 ± 3.44%, global SRe 0.79 ± 0.24 s(-1)) than patients without asynchronous LV contractions (global strain -18.57 ± 3.10%, global SRe 1.06 ± 0.32 s(-1); all P < 0.0001). Accordingly, in HFNEF patients with LV mechanical dyssynchrony the rates of LV longitudinal systolic and diastolic dysfunction were 64 and 70%, respectively, whereas these rates were significantly lower (19.5 and 41.3%), respectively, in patients without asynchronous LV contractions. In addition, HFNEF patients with LV mechanical dyssynchrony presented higher LV filling pressures and worse NYHA functional class than those with normal LV contractions. CONCLUSION: In patients with HFNEF, LV mechanical dyssynchrony is associated with an important impairment of the longitudinal systolic and diastolic function of the LV. Therefore, the restoration of asynchronous LV contractions could help to improve and/or correct both the systolic and the diastolic longitudinal dysfunction of the LV in HFNEF and thereby improve the symptomatology of these patients.


Assuntos
Ecocardiografia/métodos , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/fisiopatologia , Volume Sistólico/fisiologia , Análise de Variância , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , Comorbidade , Diástole/fisiologia , Fibrose Endomiocárdica/fisiopatologia , Feminino , Humanos , Modelos Lineares , Masculino , Fatores de Risco , Sístole/fisiologia
17.
PLoS One ; 6(12): e28872, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22194935

RESUMO

Secundum-type atrial septal defects (ASDII) account for approximately 10% of all congenital heart defects (CHD) and are associated with a familial risk. Mutations in transcription factors represent a genetic source for ASDII. Yet, little is known about the role of mutations in sarcomeric genes in ASDII etiology. To assess the role of sarcomeric genes in patients with inherited ASDII, we analyzed 13 sarcomeric genes (MYH7, MYBPC3, TNNT2, TCAP, TNNI3, MYH6, TPM1, MYL2, CSRP3, ACTC1, MYL3, TNNC1, and TTN kinase region) in 31 patients with familial ASDII using array-based resequencing. Genotyping of family relatives and control subjects as well as structural and homology analyses were used to evaluate the pathogenic impact of novel non-synonymous gene variants. Three novel missense mutations were found in the MYH6 gene encoding alpha-myosin heavy chain (R17H, C539R, and K543R). These mutations co-segregated with CHD in the families and were absent in 370 control alleles. Interestingly, all three MYH6 mutations are located in a highly conserved region of the alpha-myosin motor domain, which is involved in myosin-actin interaction. In addition, the cardiomyopathy related MYH6-A1004S and the MYBPC3-A833T mutations were also found in one and two unrelated subjects with ASDII, respectively. No mutations were found in the 11 other sarcomeric genes analyzed. The study indicates that sarcomeric gene mutations may represent a so far underestimated genetic source for familial recurrence of ASDII. In particular, perturbations in the MYH6 head domain seem to play a major role in the genetic origin of familial ASDII.


Assuntos
Miosinas Cardíacas/genética , Doença/genética , Comunicação Interatrial/genética , Cadeias Pesadas de Miosina/genética , Sarcômeros/genética , Sarcômeros/patologia , Adulto , Criança , Pré-Escolar , Família , Feminino , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/genética , Linhagem , Fenótipo
18.
Basic Res Cardiol ; 106(6): 1041-55, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21769673

RESUMO

Familial hypertrophic cardiomyopathy (FHC) is an autosomal dominant disease, which in about 30% of the patients is caused by missense mutations in one allele of the ß-myosin heavy chain (ß-MHC) gene (MYH7). To address potential molecular mechanisms underlying the family-specific prognosis, we determined the relative expression of mutant versus wild-type MYH7-mRNA. We found a hitherto unknown mutation-dependent unequal expression of mutant to wild-type MYH7-mRNA, which is paralleled by similar unequal expression of ß-MHC at the protein level. Relative abundance of mutated versus wild-type MYH7-mRNA was determined by a specific restriction digest approach and by real-time PCR (RT-qPCR). Fourteen samples from M. soleus and myocardium of 12 genotyped and clinically well-characterized FHC patients were analyzed. The fraction of mutated MYH7-mRNA in five patients with mutation R723G averaged to 66 and 68% of total MYH7-mRNA in soleus and myocardium, respectively. For mutations I736T, R719W and V606M, fractions of mutated MYH7-mRNA in M. soleus were 39, 57 and 29%, respectively. For all mutations, unequal abundance was similar at the protein level. Importantly, fractions of mutated transcripts were comparable among siblings, in younger relatives and unrelated carriers of the same mutation. Hence, the extent of unequal expression of mutated versus wild-type transcript and protein is characteristic for each mutation, implying cis-acting regulatory mechanisms. Bioinformatics suggest mRNA stability or splicing effectors to be affected by certain mutations. Intriguingly, we observed a correlation between disease expression and fraction of mutated mRNA and protein. This strongly suggests that mutation-specific allelic imbalance represents a new pathogenic factor for FHC.


Assuntos
Cardiomiopatia Hipertrófica Familiar/genética , Miosinas Ventriculares/genética , Adulto , Alelos , Desequilíbrio Alélico , Análise Mutacional de DNA , Genótipo , Humanos , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Linhagem , Estabilidade de RNA , RNA Mensageiro/análise , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adulto Jovem
19.
Eur J Heart Fail ; 13(11): 1185-92, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21750094

RESUMO

AIMS: Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) can both be due to mutations in the genes encoding ß-myosin heavy chain (MYH7) or cardiac myosin-binding protein C (MYBPC3). The aim of the present study was to determine the prevalence and spectrum of mutations in both genes in German HCM and DCM patients and to establish novel genotype-to-phenotype correlations. METHODS AND RESULTS: Coding exons and intron flanks of the two genes MYH7 and MYBPC3 of 236 patients with HCM and 652 patients with DCM were sequenced by conventional and array-based means. Clinical records were established following standard protocols. Mutations were detected in 41 and 11% of the patients with HCM and DCM, respectively. Differences were observed in the frequency of splice site and frame-shift mutations in the gene MYBPC3, which occurred more frequently (P< 0.02, P< 0.001, respectively) in HCM than in DCM, suggesting that cardiac myosin-binding protein C haploinsufficiency predisposes to hypertrophy rather than to dilation. Additional novel genotype-to-phenotype correlations were found in HCM, among these a link between MYBPC3 mutations and a particularly large thickness of the interventricular septum (P= 0.04 vs. carriers of a mutation in MYH7). Interestingly, this correlation and a link between MYH7 mutations and a higher degree of mitral valve regurgitation held true for both HCM and DCM, indicating that the gene affected by a mutation may determine the magnitude of structural and functional alterations in both HCM and DCM. CONCLUSION: A large clinical-genetic study has unravelled novel genotype-to-phenotype correlations in HCM and DCM which warrant future investigation of both the underlying mechanisms and the prognostic use.


Assuntos
Cardiomiopatia Dilatada/genética , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Dilatada/epidemiologia , Cardiomiopatia Hipertrófica/epidemiologia , Predisposição Genética para Doença , Humanos , Mutação , Fenótipo
20.
PLoS One ; 6(6): e20711, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21673957

RESUMO

Patent foramen ovale (PFO) is associated with clinical conditions including cryptogenic stroke, migraine and varicose veins. Data from studies in humans and mouse suggest that PFO and the secundum form of atrial septal defect (ASDII) exist in an anatomical continuum of septal dysmorphogenesis with a common genetic basis. Mutations in multiple members of the evolutionarily conserved cardiac transcription factor network, including GATA4, cause or predispose to ASDII and PFO. Here, we assessed whether the most prevalent variant of the GATA4 gene, S377G, was significantly associated with PFO or ASD. Our analysis of world indigenous populations showed that GATA4 S377G was largely Caucasian-specific, and so subjects were restricted to those of Caucasian descent. To select for patients with larger PFO, we limited our analysis to those with cryptogenic stroke in which PFO was a subsequent finding. In an initial study of Australian subjects, we observed a weak association between GATA4 S377G and PFO/Stroke relative to Caucasian controls in whom ASD and PFO had been excluded (OR = 2.16; p = 0.02). However, in a follow up study of German Caucasians no association was found with either PFO or ASD. Analysis of combined Australian and German data confirmed the lack of a significant association. Thus, the common GATA4 variant S377G is likely to be relatively benign in terms of its participation in CHD and PFO/Stroke.


Assuntos
Forame Oval Patente/complicações , Forame Oval Patente/genética , Fator de Transcrição GATA4/genética , Miocárdio/metabolismo , Polimorfismo Genético , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/genética , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Forame Oval Patente/diagnóstico por imagem , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Ultrassonografia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA