Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
HGG Adv ; 2(3)2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34888534

RESUMO

Bicuspid aortic valve (BAV) with ~1%-2% prevalence is the most common congenital heart defect (CHD). It frequently results in valve disease and aorta dilation and is a major cause of adult cardiac surgery. BAV is genetically linked to rare left-heart obstructions (left ventricular outflow tract obstructions [LVOTOs]), including hypoplastic left heart syndrome (HLHS) and coarctation of the aorta (CoA). Mouse and human studies indicate LVOTO is genetically heterogeneous with a complex genetic etiology. Homozygous mutation in the Pcdha protocadherin gene cluster in mice can cause BAV, and also HLHS and other LVOTO phenotypes when accompanied by a second mutation. Here we show two common deletion copy number variants (delCNVs) within the PCDHA gene cluster are associated with LVOTO. Analysis of 1,218 white individuals with LVOTO versus 463 disease-free local control individuals yielded odds ratios (ORs) at 1.47 (95% confidence interval [CI], 1.13-1.92; p = 4.2 × 10-3) for LVOTO, 1.47 (95% CI, 1.10-1.97; p = 0.01) for BAV, 6.13 (95% CI, 2.75-13.7; p = 9.7 × 10-6) for CoA, and 1.49 (95% CI, 1.07-2.08; p = 0.019) for HLHS. Increased OR was observed for all LVOTO phenotypes in homozygous or compound heterozygous PCDHA delCNV genotype comparison versus wild type. Analysis of an independent white cohort (381 affected individuals, 1,352 control individuals) replicated the PCDHA delCNV association with LVOTO. Generalizability of these findings is suggested by similar observations in Black and Chinese individuals with LVOTO. Analysis of Pcdha mutant mice showed reduced PCDHA expression at regions of cell-cell contact in aortic smooth muscle and cushion mesenchyme, suggesting potential mechanisms for BAV pathogenesis and aortopathy. Together, these findings indicate common variants causing PCDHA deficiency play a significant role in the genetic etiology of common and rare LVOTO-CHD.

2.
Thromb Haemost ; 120(1): 94-106, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31752040

RESUMO

Thrombocytopenia and platelet dysfunction induced by extracorporeal blood circulation are thought to contribute to postsurgical bleeding complications in neonates undergoing cardiac surgery with cardiopulmonary bypass (CPB). In this study, we examined how changes in platelet function relate to changes in platelet count and to excessive bleeding in neonatal CPB surgery. Platelet counts and platelet P-selectin exposure in response to agonist stimulation were measured at four times before, during, and after CPB surgery in neonates with normal versus excessive levels of postsurgical bleeding. Relative to baseline, platelet counts were reduced in patients while on CPB, as was platelet activation by the thromboxane A2 analog U46619, thrombin receptor activating peptide (TRAP), and collagen-related peptide (CRP). Platelet activation by adenosine diphosphate (ADP) was instead reduced after platelet transfusion. We provide evidence that thrombocytopenia is a likely contributor to CPB-associated defects in platelet responsiveness to U46619 and TRAP, CPB-induced collagen receptor downregulation likely contributes to defective platelet responsiveness to CRP, and platelet transfusion may contribute to defective platelet responses to ADP. Platelet transfusion restored to baseline levels platelet counts and responsiveness to all agonists except ADP but did not prevent excessive bleeding in all patients. We conclude that platelet count and function defects are characteristic of neonatal CPB surgery and that platelet transfusion corrects these defects. However, since CPB-associated coagulopathy is multifactorial, platelet transfusion alone is insufficient to treat bleeding events in all patients. Therefore, platelet transfusion must be combined with treatment of other factors that contribute to the coagulopathy to prevent excessive bleeding.


Assuntos
Plaquetas/fisiologia , Ponte Cardiopulmonar , Circulação Extracorpórea , Cardiopatias Congênitas/cirurgia , Transfusão de Plaquetas/métodos , Hemorragia Pós-Operatória/prevenção & controle , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Difosfato de Adenosina/metabolismo , Células Cultivadas , Feminino , Humanos , Recém-Nascido , Masculino , Ativação Plaquetária , Contagem de Plaquetas , Testes de Função Plaquetária
3.
World J Pediatr Congenit Heart Surg ; 9(4): 424-433, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29945504

RESUMO

BACKGROUND: Thrombocytopenia and hypofibrinogenemia during neonatal cardiopulmonary bypass (CPB) contribute to bleeding and morbidity. Rotational thromboelastometry (ROTEM) is a viscoelastic assay with a rapid turnaround time. Data validating ROTEM during neonatal cardiac surgery remain limited. This study examined perioperative hemostatic trends in neonates treated with standardized platelet and cryoprecipitate transfusion during CPB. We hypothesized that ROTEM would predict thrombocytopenia, hypofibrinogenemia, and the correction thereof. METHODS: Forty-four neonates undergoing CPB were included in this prospective observational study. Blood samples were obtained at Baseline, On CPB, Post-CPB, and Postoperative. The ROTEM analysis included extrinsically activated (Extem) and fibrinogen-specific (Fibtem) assays. Platelet-specific thromboelastometry (Pltem) values were calculated. Platelet and cryoprecipitate transfusion was initiated prior to termination of CPB. RESULTS: Platelet count and Extem amplitude decreased significantly On CPB ( P < .0001), increased significantly Post-CPB ( P < .0001), and Postoperative values were not significantly different from Baseline. Extem amplitude at 10 minutes (A10) > 46.5 mm (AUC = 0.941) and Pltem A10 > 37.5 mm [area under curve (AUC) = 0.960] predicted platelet count > 100 × 103/µL, and they highly correlated with platelet count ( R = 0.89 and R = 0.90, respectively). Fibrinogen concentration and Fibtem amplitude decreased significantly On CPB ( P ≤ .0001) and normalized after cryoprecipitate transfusion. Fibtem A10 > 9.5 mm predicted fibrinogen >200 mg/dL (AUC = 0.817), but it correlated less well with fibrinogen concentration ( R = 0.65). CONCLUSIONS: ROTEM analysis during neonatal cardiac surgery is sensitive and specific for thrombocytopenia and hypofibrinogenemia, identifying deficits within 10 minutes. Platelet and cryoprecipitate transfusion during neonatal CPB normalizes platelet count, fibrinogen level, and ROTEM amplitudes.


Assuntos
Afibrinogenemia/diagnóstico , Ponte Cardiopulmonar , Cuidados Intraoperatórios/métodos , Complicações Intraoperatórias/diagnóstico , Tromboelastografia/métodos , Trombocitopenia/diagnóstico , Afibrinogenemia/etiologia , Feminino , Humanos , Recém-Nascido , Masculino , Estudos Prospectivos , Sensibilidade e Especificidade , Trombocitopenia/etiologia
4.
Eur J Radiol ; 84(10): 1938-42, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26210092

RESUMO

INTRODUCTION: Right ventricular (RV) size and function in Duchenne muscular dystrophy (DMD) have not been well described. Using cardiac magnetic resonance (CMR) imaging we describe the relationship of RV and left ventricular (LV) size and function in a large DMD cohort. METHODS: Latest CMR scans of 272 patients consecutively seen at a single tertiary referral center (2011-2014) with skeletal muscle biopsy confirmed DMD were included. 1.5 and 3 Tesla CMR scanners were used. Biventricular ejection fraction (EF), end-diastolic volume index (EDVI), mass and mass index were compared across categories of LVEF. RESULTS: Mean age was 13.5 ± 4.9 years. 71% had normal (≥ 55%) LVEF while mild (EF 45-54%), moderate (EF 30-44%), and severe LV dysfunction (EF <30%) was present in 20%, 6% and 3% respectively. The correlation between RVEF and LVEF was weak. Even in patients with severe LV dysfunction, RVEF (49.7% ± 12.9%) was relatively preserved. There were no significant differences in RVEDVI and RV mass index across categories of LV function. CONCLUSION: In a large DMD cohort, RVEF was relatively preserved and RV size was preserved across categories of LV dysfunction.


Assuntos
Ventrículos do Coração/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Função Ventricular Esquerda/fisiologia , Função Ventricular Direita/fisiologia , Adolescente , Adulto , Volume Cardíaco/fisiologia , Criança , Estudos de Coortes , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Distrofia Muscular de Duchenne/patologia , Tamanho do Órgão , Volume Sistólico/fisiologia , Disfunção Ventricular Esquerda/fisiopatologia , Adulto Jovem
5.
Am J Cardiol ; 115(7): 967-71, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25702278

RESUMO

Duchenne and Becker muscular dystrophies are caused by mutations in dystrophin. Cardiac manifestations vary broadly, making prognosis difficult. Current dystrophin genotype-cardiac phenotype correlations are limited. For skeletal muscle, the reading-frame rule suggests in-frame mutations tend to yield milder phenotypes. We performed dystrophin genotype-cardiac phenotype correlations using a protein-effect model and cardiac magnetic resonance imaging. A translational model was applied to patient-specific deletion, indel, and nonsense mutations to predict exons and protein domains present within truncated dystrophin protein. Patients were dichotomized into predicted present and predicted absent groups for exons and protein domains of interest. Development of myocardial fibrosis (represented by late gadolinium enhancement [LGE]) and depressed left ventricular ejection fraction (LVEF) were compared. Patients (n = 274) with predicted present cysteine-rich domain (CRD), C-terminal domain (CTD), and both the N-terminal actin-binding and cysteine-rich domains (ABD1 + CRD) had a decreased risk of LGE and trended toward greater freedom from LGE. Patients with predicted present CTD (exactly the same as those with in-frame mutations) and ABD1 + CRD trended toward decreased risk of and greater freedom from depressed LVEF. In conclusion, genotypes previously implicated in altering the dystrophinopathic cardiac phenotype were not significantly related to LGE and depressed LVEF. Patients with predicted present CRD, CTD/in-frame mutations, and ABD1 + CRD trended toward milder cardiac phenotypes, suggesting that the reading-frame rule may be applicable to the cardiac phenotype. Genotype-phenotype correlations may help predict the cardiac phenotype for dystrophinopathic patients and guide future therapies.


Assuntos
Cardiomiopatias/diagnóstico , DNA/genética , Distrofina/genética , Imagem Cinética por Ressonância Magnética/métodos , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Mutação , Adolescente , Adulto , Cardiomiopatias/etiologia , Cardiomiopatias/genética , Criança , Pré-Escolar , Análise Mutacional de DNA , Distrofina/metabolismo , Éxons , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/metabolismo , Deleção de Sequência , Adulto Jovem
6.
N Engl J Med ; 371(22): 2061-71, 2014 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-25405392

RESUMO

BACKGROUND: Aortic-root dissection is the leading cause of death in Marfan's syndrome. Studies suggest that with regard to slowing aortic-root enlargement, losartan may be more effective than beta-blockers, the current standard therapy in most centers. METHODS: We conducted a randomized trial comparing losartan with atenolol in children and young adults with Marfan's syndrome. The primary outcome was the rate of aortic-root enlargement, expressed as the change in the maximum aortic-root-diameter z score indexed to body-surface area (hereafter, aortic-root z score) over a 3-year period. Secondary outcomes included the rate of change in the absolute diameter of the aortic root; the rate of change in aortic regurgitation; the time to aortic dissection, aortic-root surgery, or death; somatic growth; and the incidence of adverse events. RESULTS: From January 2007 through February 2011, a total of 21 clinical centers enrolled 608 participants, 6 months to 25 years of age (mean [±SD] age, 11.5±6.5 years in the atenolol group and 11.0±6.2 years in the losartan group), who had an aortic-root z score greater than 3.0. The baseline-adjusted rate of change in the mean (±SE) aortic-root z score did not differ significantly between the atenolol group and the losartan group (-0.139±0.013 and -0.107±0.013 standard-deviation units per year, respectively; P=0.08). Both slopes were significantly less than zero, indicating a decrease in the aortic-root diameter relative to body-surface area with either treatment. The 3-year rates of aortic-root surgery, aortic dissection, death, and a composite of these events did not differ significantly between the two treatment groups. CONCLUSIONS: Among children and young adults with Marfan's syndrome who were randomly assigned to losartan or atenolol, we found no significant difference in the rate of aortic-root dilatation between the two treatment groups over a 3-year period. (Funded by the National Heart, Lung, and Blood Institute and others; ClinicalTrials.gov number, NCT00429364.).


Assuntos
Antagonistas Adrenérgicos beta/uso terapêutico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Aorta/efeitos dos fármacos , Aneurisma Aórtico/prevenção & controle , Atenolol/uso terapêutico , Losartan/uso terapêutico , Síndrome de Marfan/tratamento farmacológico , Antagonistas Adrenérgicos beta/efeitos adversos , Adulto , Bloqueadores do Receptor Tipo 1 de Angiotensina II/efeitos adversos , Aorta/crescimento & desenvolvimento , Aorta/cirurgia , Insuficiência da Valva Aórtica , Atenolol/efeitos adversos , Criança , Pré-Escolar , Intervalo Livre de Doença , Feminino , Humanos , Lactente , Modelos Lineares , Losartan/efeitos adversos , Masculino , Síndrome de Marfan/mortalidade , Síndrome de Marfan/fisiopatologia , Resultado do Tratamento , Adulto Jovem
7.
Circ Arrhythm Electrophysiol ; 6(5): 946-51, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23995044

RESUMO

BACKGROUND: Fetal arrhythmias characteristic of long QT syndrome (LQTS) include torsades de pointes (TdP) and/or 2° atrioventricular block, but sinus bradycardia, defined as fetal heart rate<3% for gestational age, is most common. We hypothesized that prenatal rhythm phenotype might predict LQTS genotype and facilitate improved risk stratification and management. METHOD AND RESULTS: Records of subjects exhibiting fetal LQTS arrhythmias were reviewed. Fetal echocardiograms, neonatal ECG, and genetic testing were evaluated. We studied 43 subjects exhibiting fetal LQTS arrhythmias: TdP±2° atrioventricular block (group 1, n=7), isolated 2° atrioventricular block (group 2, n=4), and sinus bradycardia (group 3, n=32). Mutations in known LQTS genes were found in 95% of subjects tested. SCN5A mutations occurred in 71% of group 1, whereas 91% of subjects with KCNQ1 mutations were in group 3. Small numbers of subjects with KCNH2 mutations (n=4) were scattered in all 3 groups. Age at presentation did not differ among groups, and most subjects (n=42) were live-born with gestational ages of 37.5±2.8 weeks (mean±SD). However, those with TdP were typically delivered earlier. Prenatal treatment in group 1 terminated (n=2) or improved (n=4) TdP. The neonatal heart rate-corrected QT interval (mean±SE) of group 1 (664.7±24.9) was longer than neonatal heart rate-corrected QT interval in both group 2 (491.2±27.6; P=0.004) and group 3 (483.1±13.7; P<0.001). Despite medical and pacemaker therapy, postnatal cardiac arrest (n=4) or sudden death (n=1) was common among subjects with fetal/neonatal TdP. CONCLUSIONS: Rhythm phenotypes of fetal LQTS have genotype-suggestive features that, along with heart rate-corrected QT interval duration, may risk stratify perinatal management.


Assuntos
Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/genética , Bradicardia/diagnóstico , Bradicardia/genética , Canais de Potássio Éter-A-Go-Go/genética , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Diagnóstico Pré-Natal , Bloqueio Atrioventricular/fisiopatologia , Bradicardia/fisiopatologia , Canal de Potássio ERG1 , Ecocardiografia , Eletrocardiografia , Feminino , Testes Genéticos , Genótipo , Frequência Cardíaca , Humanos , Recém-Nascido , Síndrome do QT Longo/fisiopatologia , Mutação , Fenótipo , Gravidez , Resultado da Gravidez , Medição de Risco
8.
Am Heart J ; 165(5): 828-835.e3, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23622922

RESUMO

BACKGROUND: The Pediatric Heart Network designed a clinical trial to compare aortic root growth and other short-term cardiovascular outcomes in children and young adults with Marfan syndrome randomized to receive atenolol or losartan. We report here the characteristics of the screened population and enrolled subjects. METHODS AND RESULTS: Between 2007 and 2011, 21 clinical sites randomized 608 subjects, aged 6 months to 25 years who met the original Ghent criteria and had a body surface area-adjusted aortic root diameter z-score >3.0. The mean age at study entry was 11.2 years, 60% were male, and 25% were older teenagers and young adults. The median aortic root diameter z-score was 4.0. Aortic root diameter z-score did not vary with age. Mitral valve prolapse and mitral regurgitation were more common in females. Among those with a positive family history, 56% had a family member with aortic surgery, and 32% had a family member with a history of aortic dissection. CONCLUSIONS: Baseline demographic, clinical, and anthropometric characteristics of the randomized cohort are representative of patients in this population with moderate to severe aortic root dilation. The high percentage of young subjects with relatives who have had aortic dissection or surgery illustrates the need for more definitive therapy; we expect that the results of the study and the wealth of systematic data collected will make an important contribution to the management of individuals with Marfan syndrome.


Assuntos
Aneurisma da Aorta Torácica/tratamento farmacológico , Atenolol/uso terapêutico , Losartan/uso terapêutico , Síndrome de Marfan/tratamento farmacológico , Adolescente , Antagonistas de Receptores Adrenérgicos beta 1/uso terapêutico , Adulto , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Aneurisma da Aorta Torácica/complicações , Criança , Pré-Escolar , Feminino , Seguimentos , Humanos , Lactente , Masculino , Síndrome de Marfan/complicações , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem
9.
J Am Coll Cardiol ; 59(1 Suppl): S1-42, 2012 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-22192720

RESUMO

In the recent era, no congenital heart defect has undergone a more dramatic change in diagnostic approach, management, and outcomes than hypoplastic left heart syndrome (HLHS). During this time, survival to the age of 5 years (including Fontan) has ranged from 50% to 69%, but current expectations are that 70% of newborns born today with HLHS may reach adulthood. Although the 3-stage treatment approach to HLHS is now well founded, there is significant variation among centers. In this white paper, we present the current state of the art in our understanding and treatment of HLHS during the stages of care: 1) pre-Stage I: fetal and neonatal assessment and management; 2) Stage I: perioperative care, interstage monitoring, and management strategies; 3) Stage II: surgeries; 4) Stage III: Fontan surgery; and 5) long-term follow-up. Issues surrounding the genetics of HLHS, developmental outcomes, and quality of life are addressed in addition to the many other considerations for caring for this group of complex patients.


Assuntos
Técnica de Fontan/métodos , Síndrome do Coração Esquerdo Hipoplásico/diagnóstico , Síndrome do Coração Esquerdo Hipoplásico/cirurgia , Diagnóstico Pré-Natal/métodos , Procedimentos Cirúrgicos Cardíacos/métodos , Procedimentos Cirúrgicos Cardíacos/mortalidade , Desenvolvimento Infantil/fisiologia , Ecocardiografia Doppler/métodos , Feminino , Técnica de Fontan/mortalidade , Humanos , Síndrome do Coração Esquerdo Hipoplásico/mortalidade , Recém-Nascido , Masculino , Monitorização Fisiológica/métodos , Assistência Perioperatória/métodos , Gravidez , Prognóstico , Medição de Risco , Análise de Sobrevida , Resultado do Tratamento , Ultrassonografia Doppler/métodos
10.
Am J Cardiol ; 107(5): 761-6, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21247531

RESUMO

Atrioventricular (AV) block and endocardial fibroelastosis associated with dilated cardiomyopathy are the most common clinical manifestations of anti-Ro/SSA-mediated fetal cardiac disease. Valvar dysfunction has not been a prominent feature of this disease; however, recent anecdotal cases have suggested an association between rupture of the AV valve tensor apparatus and maternal anti-Ro/SSA antibodies. In the present study, we have described the clinical and laboratory findings and reviewed the published data for infants of anti-Ro/SSA-positive pregnancies with AV valve insufficiency due to chordal rupture from the papillary muscles. The histopathologic features of the papillary muscle and ventricular free wall and septum biopsy specimens were examined and compared to the sections of AV leaflets from 6 autopsied fetuses with anti-Ro/SSA-mediated complete AV block without chordal disruption. Specific epitopes to the p200 region of Ro52, and Ro60 antibodies were evaluated in cases with chordal rupture. Severe AV valve insufficiency was detected prenatally (as early as 34 weeks of gestation) or postnatally (as late as 182 days) after areas of patchy echogenicity were noted in the papillary muscle at 19 to 22 weeks of gestation. Postnatally, urgent valve surgery was performed in 5 of 6 patients; 1 of 6 patients died preoperatively. All patients tested positive for Ro52. Valve leaflet tissue from the autopsy specimens was normal. The ventricular free wall and septum biopsy specimens from a patient with chordal rupture showed normal tissue; however, the papillary muscle biopsy specimens demonstrated severe atrophy with near total replacement of myocytes by fibrosis and dystrophic calcifications, and negative immunochemistry findings. In conclusion, these findings have defined an underappreciated complication of fetal antibody-mediated cardiac inflammation.


Assuntos
Anticorpos Antinucleares/imunologia , Bloqueio Atrioventricular/congênito , Ruptura Cardíaca/congênito , Troca Materno-Fetal/imunologia , Valva Mitral , Músculos Papilares , Complicações na Gravidez/imunologia , Adulto , Bloqueio Atrioventricular/diagnóstico , Bloqueio Atrioventricular/imunologia , Biópsia , Diagnóstico Diferencial , Evolução Fatal , Feminino , Coração Fetal/diagnóstico por imagem , Coração Fetal/fisiopatologia , Idade Gestacional , Ruptura Cardíaca/diagnóstico , Ruptura Cardíaca/imunologia , Humanos , Gravidez , Resultado da Gravidez , Ultrassonografia Pré-Natal
11.
Hum Mol Genet ; 19(4): 648-56, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19942620

RESUMO

Congenital heart defects comprise the most common form of major birth defects, affecting 0.7% of all newborn infants. Jacobsen syndrome (11q-) is a rare chromosomal disorder caused by deletions in distal 11q. We have previously determined that a wide spectrum of the most common congenital heart defects occur in 11q-, including an unprecedented high frequency of hypoplastic left heart syndrome (HLHS). We identified an approximately 7 Mb 'cardiac critical region' in distal 11q that contains a putative causative gene(s) for congenital heart disease. In this study, we utilized chromosomal microarray mapping to characterize three patients with 11q- and congenital heart defects that carry interstitial deletions overlapping the 7 Mb cardiac critical region. We propose that this 1.2 Mb region of overlap harbors a gene(s) that causes at least a subset of the congenital heart defects that occur in 11q-. We demonstrate that one gene in this region, ETS-1 (a member of the ETS family of transcription factors), is expressed in the endocardium and neural crest during early mouse heart development. Gene-targeted deletion of ETS-1 in mice in a C57/B6 background causes, with high penetrance, large membranous ventricular septal defects and a bifid cardiac apex, and less frequently a non-apex-forming left ventricle (one of the hallmarks of HLHS). Our results implicate an important role for the ETS-1 transcription factor in mammalian heart development and should provide important insights into some of the most common forms of congenital heart disease.


Assuntos
Deleção de Genes , Comunicação Interventricular/genética , Ventrículos do Coração/anormalidades , Síndrome da Deleção Distal 11q de Jacobsen/genética , Proteína Proto-Oncogênica c-ets-1/genética , Animais , Deleção Cromossômica , Mapeamento Cromossômico , Cromossomos Humanos Par 11/genética , Comunicação Interventricular/embriologia , Comunicação Interventricular/metabolismo , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Humanos , Síndrome da Deleção Distal 11q de Jacobsen/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Proto-Oncogênica c-ets-1/metabolismo
12.
Hum Mol Genet ; 18(10): 1813-24, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19251731

RESUMO

Mutations identified in a cohort of patients with atrioventricular septal defects as a part of Ellis van Creveld syndrome (EvC syndrome) led us to study the role of two non-homologous genes, EVC and LBN, in heart development and disease pathogenesis. To address the cause of locus heterogeneity resulting in an indistinguishable heart-hand phenotype, we carried out in situ hybridization and immunofluorescence and identified co-localization of Evc and Lbn mRNA and protein. In the heart, expression was identified to be strongest in the secondary heart field, including both the outflow tract and the dorsal mesenchymal protrusion, but was also found in mesenchymal structures of the atrial septum and the atrioventricular cushions. Finally, we studied the transcriptional hierarchy of EVC and LBN but did not find any evidence of direct transcriptional interregulation between the two. Due to the locus heterogeneity of human mutations predicted to result in a loss of protein function, a bidirectional genomic organization and overlapping expression patterns, we speculate that these proteins function coordinately in cardiac development and that loss of this coordinate function results in the characteristics of EvC syndrome.


Assuntos
Doenças Cardiovasculares/genética , Síndrome de Ellis-Van Creveld/genética , Coração/crescimento & desenvolvimento , Proteínas de Membrana/genética , Proteínas/genética , Animais , Doenças Cardiovasculares/metabolismo , Estudos de Coortes , Síndrome de Ellis-Van Creveld/metabolismo , Feminino , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Miocárdio/metabolismo , Células NIH 3T3 , Proteínas/metabolismo
13.
J Am Coll Cardiol ; 50(16): 1590-5, 2007 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-17936159

RESUMO

OBJECTIVES: This study sought to determine the size of the genetic effect (heritability) in families identified by a hypoplastic left heart syndrome (HLHS) proband. BACKGROUND: Hypoplastic left heart syndrome is a severe form of cardiovascular malformation (CVM), and it remains a leading cause of infant mortality and childhood morbidity. Familial clustering of HLHS and bicuspid aortic valve (BAV) has been observed, and pedigree analysis has suggested recessive inheritance. The genetic significance of these observations is unknown. METHODS: In 38 probands with HLHS, a 3-generation family history was obtained; using a sequential sampling strategy, echocardiograms on family members were performed. A total of 235 participants were recruited. Heritability (h2) of HLHS and associated CVM was estimated using maximum-likelihood-based variance decomposition. RESULTS: All HLHS probands had aortic valve hypoplasia and dysplasia; dysplasia of the mitral (94%), tricuspid (56%), and pulmonary (11%) valves was also noted. Overall, 21 of 38 (55%) families had more than 1 affected individual, and 36% of participants had CVM, including 11% with BAV. The heritability of HLHS alone and with associated CVM were 99% and 74% (p < 0.00001), respectively. The sibling recurrence risk for HLHS was 8%, and for CVM was 22%. CONCLUSIONS: The high heritability of HLHS suggests that it is determined largely by genetic factors. The frequent occurrence of left- and right-sided valve dysplasia in HLHS probands and the increased prevalence of BAV in family members suggests that HLHS is a severe form of valve malformation.


Assuntos
Síndrome do Coração Esquerdo Hipoplásico/genética , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Ecocardiografia Doppler , Feminino , Predisposição Genética para Doença , Cardiopatias Congênitas/genética , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Recidiva , Irmãos
14.
Hum Genet ; 121(2): 275-84, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17203300

RESUMO

The aim of this study was to identify regions of the genome that harbor genes influencing inheritance of bicuspid aortic valve (BAV) and/or associated cardiovascular malformation (CVM). Aortic valve disease is an important clinical problem, which often results in valve replacement, the second most common cardiac surgery in the United States. In every age group, a majority of cases of valve disease involves a BAV. BAV is the most common CVM with a reported prevalence of 1-2%. Heritability studies indicate that BAV determination is almost entirely genetic. We used a family-based genome-wide linkage analysis with microsatellite markers. Parametric and nonparametric analyses were performed with the software GENEHUNTER and SOLAR (Sequential Oligogenic Linkage Analysis Routines). Thirty-eight families (353 subjects) with BAV and/or associated CVM were assessed. Each participant underwent a standardized echocardiographic examination. The highest LOD score, 3.8, occurred on chromosome 18q between markers D18S68 and D18S1161. Two other chromosomal regions, 5q15-21 (between D5S644 and D5S2027) and 13q33-qter (between D13S1265 and 13qter), exhibited suggestive evidence of linkage (LOD > 2.0). Further, two previously reported linkage peaks on 9q34 and 17q24 were replicated in family specific analyses. No significant X chromosome linkage peaks were identified. In this genome-wide scan we demonstrate for the first time, that BAV and/or associated CVM exhibit linkage to chromosomes 18q, 5q and 13q. These regions likely contain genes whose mutation results in BAV and/or associated CVM indicating their important role in valvulogenesis and cardiac development.


Assuntos
Valva Aórtica/anormalidades , Anormalidades Cardiovasculares/genética , Cromossomos Humanos Par 13 , Cromossomos Humanos Par 18 , Cromossomos Humanos Par 5 , Ligação Genética , Valva Mitral/anormalidades , Estudos de Coortes , Ecocardiografia , Saúde da Família , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Repetições de Microssatélites
15.
Neuromuscul Disord ; 16(11): 745-8, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17005398

RESUMO

We report the use of continuous intravenous inotrope infusion as a palliative management strategy for the treatment of symptomatic, refractory, end stage cardiac dysfunction in patients with Duchenne muscular dystrophy. Milrinone and/or dobutamine administered by continuous intravenous infusion provided symptomatic and objective cardiovascular improvement up to 30 months in 3 individuals with Duchenne muscular dystrophy and severe dilated cardiomyopathy. Continuous inotrope infusion should be considered a practical treatment strategy for end stage cardiac dysfunction in Duchenne muscular dystrophy patients when cardiac transplantation is not a viable option.


Assuntos
Baixo Débito Cardíaco/tratamento farmacológico , Baixo Débito Cardíaco/etiologia , Cardiotônicos/uso terapêutico , Dobutamina/uso terapêutico , Terapia por Infusões no Domicílio/métodos , Milrinona/uso terapêutico , Distrofia Muscular de Duchenne/complicações , Adolescente , Baixo Débito Cardíaco/fisiopatologia , Cardiotônicos/administração & dosagem , Progressão da Doença , Dobutamina/administração & dosagem , Relação Dose-Resposta a Droga , Humanos , Infusões Intravenosas/métodos , Masculino , Milrinona/administração & dosagem , Distrofia Muscular de Duchenne/fisiopatologia , Cuidados Paliativos , Qualidade de Vida , Fatores de Tempo
16.
Dev Dyn ; 235(1): 38-49, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16245335

RESUMO

Nkx2-5 gene mutations cause cardiac abnormalities, including deficits of function in the atrioventricular conduction system (AVCS). In the chick, Nkx2-5 is elevated in Purkinje fiber AVCS cells relative to working cardiomyocytes. Here, we show that Nkx2-5 expression rises to a peak as Purkinje fibers progressively differentiate. To disrupt this pattern, we overexpressed Nkx2-5 from embryonic day 10, as Purkinje fibers are recruited within developing chick hearts. Overexpression of Nkx2-5 caused inhibition of slow tonic myosin heavy chain protein (sMHC), a late Purkinje fiber marker but did not affect Cx40 levels. Working cardiomyocytes overexpressing Nkx2-5 in these hearts ectopically up-regulated Cx40 but not sMHC. Isolated embryonic cardiomyocytes overexpressing Nkx2-5 also displayed increased Cx40 and suppressed sMHC. By contrast, overexpression of a human NKX2-5 mutant did not effect these markers in vivo or in vitro, suggesting one possible mechanism for clinical phenotypes. We conclude that a prerequisite for normal Purkinje fiber maturation is precise regulation of Nkx2-5 levels.


Assuntos
Proteínas Aviárias/biossíntese , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/biossíntese , Ramos Subendocárdicos/citologia , Fatores de Transcrição/biossíntese , Adenoviridae , Animais , Proteínas Aviárias/genética , Biomarcadores , Núcleo Celular/metabolismo , Embrião de Galinha , Conexinas/metabolismo , Vetores Genéticos , Proteínas de Homeodomínio/genética , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/genética , Ramos Subendocárdicos/metabolismo , Fatores de Transcrição/genética , Proteína alfa-5 de Junções Comunicantes
17.
J Am Coll Cardiol ; 44(6): 1283-91, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15364333

RESUMO

OBJECTIVES: The purpose of this research was to determine whether an intronic variant (T1945+6C) in KCNH2 is a disease-causing mutation, and if expanded phenotyping criteria produce improved identification of long QT syndrome (LQTS) patients. BACKGROUND: Long QT syndrome is usually caused by mutations in conserved coding regions or invariant splice sites, yet no mutation is found in 30% to 50% of families. In one such family, we identified an intronic variant in KCNH2. Long QT syndrome diagnosis is hindered by reduced penetrance, as the long QT phenotype is absent on baseline electrocardiogram (ECG) in about 30%. METHODS: Fifty-two family members were phenotyped by baseline QTc, QTc maximum on serial ECGs (Ser QTc-max), and on exercise ECGs (Ex QTc-max) and by T-wave patterns. Linkage analysis tested association of the intronic change with phenotype. The consequences of T1945+6C on splicing was studied using a minigene system and on function by heterologous expression. RESULTS: Expanded phenotype/pedigree criteria identified 23 affected and 29 unaffected. Affected versus unaffected had baseline QTc 484 +/- 48 ms versus 422 +/- 20 ms, Ser QTc-max 508 +/- 48 ms versus 448 +/- 10 ms, Ex QTc-max 513 +/- 54 ms versus 444 +/- 11 ms, and LQT2 T waves in 87% versus 0%. Linkage analysis demonstrated a logarithm of odds score of 10.22. Splicing assay showed T1945+6C caused downstream intron retention. Complementary deoxyribonucleic acid with retained intron 7 failed to produce functional channels. CONCLUSIONS: T1945+6C is a disease-causing mutation. It alters KCNH2 splicing and cosegregates with the LQT2 phenotype. Expanded ECG criteria plus pedigree analysis provided accurate clinical diagnosis of all carriers including those with reduced penetrance. Intronic mutations may be responsible for LQTS in some families with otherwise negative mutation screening.


Assuntos
Íntrons/genética , Síndrome do QT Longo/genética , Mutação/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise Mutacional de DNA , Canal de Potássio ERG1 , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go , Saúde da Família , Seguimentos , Triagem de Portadores Genéticos , Ligação Genética/genética , Predisposição Genética para Doença/genética , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Canais de Potássio/genética , RNA Complementar/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatística como Assunto
18.
Am J Cardiol ; 92(8): 991-5, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14556882

RESUMO

Based on similarity of electrocardiographic features, bidirectional ventricular tachycardia has been considered a variant of long QT syndrome. Genes causing long QT syndrome were used as candidate genes in 4 patients with bidirectional ventricular tachycardia. In 2 patients, we identified a common low penetrance HERG allele (R1047L) with an intermediate biophysical phenotype.


Assuntos
Proteínas de Transporte de Cátions , Proteínas de Ligação a DNA , Síndrome do QT Longo/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Taquicardia Ventricular/genética , Transativadores , Adolescente , Alelos , Pré-Escolar , Canal de Potássio ERG1 , Técnicas Eletrofisiológicas Cardíacas , Canais de Potássio Éter-A-Go-Go , Feminino , Humanos , Masculino , Fenótipo , Canais de Sódio/genética , Regulador Transcricional ERG
19.
Novartis Found Symp ; 250: 242-52; discussion 252-9, 276-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12956334

RESUMO

Atrioventricular (AV) conduction disturbance (block) describes impairment of the electrical continuity between the atria and ventricles. Clinical classification of AV block has utilized biophysical characteristics, usually the extent (1st, 2nd, 3rd degree) and site of block (above or below His bundle recording site). The genetic significance of this classification is not known. In some cases AV block occurrence is associated with intrauterine exposure to maternal antibody (anti-Ro, anti-La), and other cases are associated with injury (e.g. surgery). Based on familial clustering of idiopathic AV block, a genetic cause has also been suspected. Published pedigrees show autosomal dominant inheritance, and associated heart disease is common (e.g. congenital heart malformation, cardiomyopathy, etc.). The latter finding is not unexpected given the common origin of working myocytes and elements of the specialized conduction system. Using genetic models incorporating reduced penetrance (presence of disease genotype in absence of phenotype), variable expressivity (presence of a disease genotype with variable phenotypes) and genetic heterogeneity (similar phenotypes, different disease genotypes), molecular genetic causes of AV block are being identified. These findings are significant as they provide insight into the molecular basis of a clinical condition previously defined only by biophysical characteristics.


Assuntos
Arritmias Cardíacas/genética , Nó Atrioventricular/fisiopatologia , Bloqueio Cardíaco/genética , Animais , Nó Atrioventricular/patologia , Eletrocardiografia , Bloqueio Cardíaco/etiologia , Cardiopatias Congênitas/fisiopatologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Canal de Sódio Disparado por Voltagem NAV1.5 , Doenças Neuromusculares/complicações , Canais de Sódio/química , Canais de Sódio/genética , Canais de Sódio/metabolismo
20.
Pediatr Res ; 53(5): 744-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12621127

RESUMO

Previous studies have identified mutations in five ion channel genes as a cause of long QT syndrome, a heterogeneous disorder characterized by prolongation of the QT interval, multiform ventricular tachycardia (torsades de pointes), seizures, syncope, and sudden death. However, in these studies, the average age of initial symptoms is in the third decade of life or later, and few reports have described the genetic causes of long QT syndrome presenting in the prenatal or neonatal period. We used a candidate gene approach to identify the genetic cause of long QT syndrome in an infant whose initial manifestations were detected in utero. Direct bidirectional sequencing of long QT syndrome genes identified a previously unreported HERG missense mutation (R752Q). Three asymptomatic family members were heterozygous for R752Q, and the proband, who manifested ventricular tachycardia in utero, was homozygous. R752Q was not found in 100 normal unrelated chromosomes. Paternal DNA was unavailable for testing. Transient transfection of HERG generated robust IKr, but no current was observed for the mutant HERG. The HERG mutant, R752Q, is associated with a mild phenotype, inasmuch as family members with a heterozygous mutation appear unaffected. The homozygous mutation results in absence of functional IKr, causing a profound loss of HERG channel function, creating the equivalent of a "HERG knockout" and leading to a severe phenotype.


Assuntos
Proteínas de Transporte de Cátions , Proteínas de Ligação a DNA , Síndrome do QT Longo/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Transativadores , Sequência de Bases , Canal de Potássio ERG1 , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go , Saúde da Família , Feminino , Homozigoto , Humanos , Recém-Nascido , Síndrome do QT Longo/fisiopatologia , Masculino , Potenciais da Membrana , Dados de Sequência Molecular , Linhagem , Fenótipo , Índice de Gravidade de Doença , Regulador Transcricional ERG
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA