Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Ultrasound Obstet Gynecol ; 58(6): 853-863, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34096674

RESUMO

OBJECTIVES: The primary aim of this study was to evaluate the feasibility of automated measurement of fetal atrioventricular (AV) plane displacement (AVPD) over several cardiac cycles using myocardial velocity traces obtained by color tissue Doppler imaging (cTDI). The secondary objectives were to establish reference ranges for AVPD during the second half of normal pregnancy, to assess fetal AVPD in prolonged pregnancy in relation to adverse perinatal outcome and to evaluate AVPD in fetuses with a suspicion of intrauterine growth restriction (IUGR). METHODS: The population used to develop the reference ranges consisted of women with an uncomplicated singleton pregnancy at 18-42 weeks of gestation (n = 201). The prolonged-pregnancy group comprised women with an uncomplicated singleton pregnancy at ≥ 41 + 0 weeks of gestation (n = 107). The third study cohort comprised women with a singleton pregnancy and suspicion of IUGR, defined as an estimated fetal weight < 2.5th centile or an estimated fetal weight < 10th centile and umbilical artery pulsatility index > 97.5th centile (n = 35). Cineloops of the four-chamber view of the fetal heart were recorded using cTDI. Regions of interest were placed at the AV plane in the left and right ventricular walls and the interventricular septum, and myocardial velocity traces were integrated and analyzed using an automated algorithm developed in-house to obtain mitral (MAPSE), tricuspid (TAPSE) and septal (SAPSE) annular plane systolic excursion. Gestational-age specific reference ranges were constructed and normalized for cardiac size. The correlation between AVPD measurements obtained using cTDI and those obtained by anatomic M-mode were evaluated, and agreement between these two methods was assessed using Bland-Altman analysis. The mean Z-scores of fetal AVPD in the cohort of prolonged pregnancies were compared between cases with normal and those with adverse outcome using Mann-Whitney U-test. The mean Z-scores of fetal AVPD in IUGR fetuses were compared with those in the normal reference population using Mann-Whitney U-test. Inter- and intraobserver variability for acquisition of cTDI recordings and offline analysis was assessed by calculating coefficients of variation (CV) using the root mean square method. RESULTS: Fetal MAPSE, SAPSE and TAPSE increased with gestational age but did not change significantly when normalized for cardiac size. The fitted mean was highest for TAPSE throughout the second half of gestation, followed by SAPSE and MAPSE. There was a significant correlation between MAPSE (r = 0.64; P < 0.001), SAPSE (r = 0.72; P < 0.001) and TAPSE (r = 0.84; P < 0.001) measurements obtained by M-mode and those obtained by cTDI. The geometric means of ratios between AVPD measured by cTDI and by M-mode were 1.38 (95% limits of agreement (LoA), 0.84-2.25) for MAPSE, 1.00 (95% LoA, 0.72-1.40) for SAPSE and 1.20 (95% LoA, 0.92-1.57) for TAPSE. In the prolonged-pregnancy group, the mean ± SD Z-scores for MAPSE (0.14 ± 0.97), SAPSE (0.09 ± 1.02) and TAPSE (0.15 ± 0.90) did not show any significant difference compared to the reference ranges. Twenty-one of the 107 (19.6%) prolonged pregnancies had adverse perinatal outcome. The AVPD Z-scores were not significantly different between pregnancies with normal and those with adverse outcome in the prolonged-pregnancy cohort. The mean ± SD Z-scores for SAPSE (-0.62 ± 1.07; P = 0.006) and TAPSE (-0.60 ± 0.89; P = 0.002) were significantly lower in the IUGR group compared to those in the normal reference population, but the differences were not significant when the values were corrected for cardiac size. The interobserver CVs for the automated measurement of MAPSE, SAPSE and TAPSE were 28.1%, 17.7% and 15.3%, respectively, and the respective intraobserver CVs were 33.5%, 15.0% and 17.9%. CONCLUSIONS: This study showed that fetal AVPD can be measured automatically by integrating cTDI velocities over several cardiac cycles. Automated analysis of AVPD could potentially help gather larger datasets to facilitate use of machine-learning models to study fetal cardiac function. The gestational-age associated increase in AVPD is most likely a result of increasing cardiac size, as the AVPD normalized for cardiac size did not change significantly between 18 and 42 weeks. A decrease was seen in TAPSE and SAPSE in IUGR fetuses, but not after correction for cardiac size. © 2021 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.


Assuntos
Nó Atrioventricular/diagnóstico por imagem , Ecocardiografia Doppler em Cores/estatística & dados numéricos , Coração Fetal/diagnóstico por imagem , Sístole/fisiologia , Ultrassonografia Pré-Natal/estatística & dados numéricos , Nó Atrioventricular/embriologia , Velocidade do Fluxo Sanguíneo , Estudos de Viabilidade , Feminino , Retardo do Crescimento Fetal/diagnóstico por imagem , Retardo do Crescimento Fetal/fisiopatologia , Coração Fetal/embriologia , Peso Fetal , Idade Gestacional , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/embriologia , Humanos , Gravidez , Fluxo Pulsátil , Valores de Referência , Volume Sistólico , Valva Tricúspide/diagnóstico por imagem , Valva Tricúspide/embriologia , Septo Interventricular/diagnóstico por imagem , Septo Interventricular/embriologia
2.
J Gynecol Obstet Hum Reprod ; 48(2): 121-127, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30415076

RESUMO

OBJECTIVES: We propose an image scoring method to improve the quality and the reproducibility of measurement of the AV interval before establishing reference tables of the measurements and studies on the prevention and treatment of first-degree AV block especially if the first child has been diagnosed AV block. METHOD: Prospective study from May 2015 to June 2016. Sonographers were asked to measure AV interval with pulsed Doppler in a five-chamber view in standard second-trimester screening before and after having received our image scoring method. Images were scored by 2 blinded reviewers. RESULTS: The intra-class correlation coefficient (ICC) between the two reviewers for the overall score was 0.91. On average, the measurement quality increased by 2.5 points/10 (95% CI 1.0-4.0). In the second set of images, after the scoring method was given, the score stared at 6.50 for the first image, with a significant improvement of 0.18 (p = 0.016) per subsequent image comparing to a non significant improvement for the first set of image. There was a significant improvement in intra-observer reliability, ICC: 0.680 [95% CI 0.606-0.854] versus 0.458 [95% CI 0.140-0.651]. CONCLUSION: The use of this scoring method is simple, reproducible and improves image quality and reproducibility of AV interval measurement in a five-chamber view.


Assuntos
Nó Atrioventricular/diagnóstico por imagem , Nó Atrioventricular/embriologia , Ecocardiografia Doppler de Pulso/métodos , Ultrassonografia Pré-Natal/métodos , Bloqueio Atrioventricular/diagnóstico por imagem , Bloqueio Atrioventricular/embriologia , Feminino , Idade Gestacional , Humanos , Variações Dependentes do Observador , Gravidez , Estudos Prospectivos , Valores de Referência , Reprodutibilidade dos Testes
3.
Sci Rep ; 8(1): 4722, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29549339

RESUMO

Atrioventricular septal defects often result from impaired endocardial cushion development. Endothelial-to-mesenchymal transition (EndoMT) is a critical event in endocardial cushion development that initiates in the atrioventricular canal (AVC). In ex vivo EndoMT studies, mouse AVCs are flat-mounted on a collagen gel. In the explant outgrowths, the ratio of elongated spindle-like mesenchymal cells over cobblestone-shaped cells, generally considered as endothelial cells, reflects EndoMT. Using this method, several key signalling pathways have been attributed important functions during EndoMT. Using genetic lineage tracing and cell-type-specific markers, we show that monolayers of cobblestone-shaped cells are predominantly of epicardial rather than endothelial origin. Furthermore, this epicardium is competent to undergo mesenchymal transition. Contamination by epicardium is common and inherent as this tissue progressively attaches to AVC myocardium. Inhibition of TGFß signalling, previously shown to blunt EndoMT, caused an enrichment in epicardial monolayers. The presence of epicardium thus confounds interpretations of EndoMT signalling pathways in this assay. We advocate to systematically use lineage tracers and cell-type-specific markers on stage-matched AVC explants. Furthermore, a careful reconsideration of earlier studies on EndoMT using this explant assay may identify unanticipated epicardial effects and/or the presence of epicardial-to-mesenchymal transition (EpiMT), which would alter the interpretation of results on endothelial-to-mesenchymal transition.


Assuntos
Nó Atrioventricular/fisiologia , Embrião de Mamíferos/fisiologia , Endotélio Vascular/fisiologia , Transição Epitelial-Mesenquimal , Pericárdio/fisiologia , Animais , Nó Atrioventricular/embriologia , Bioensaio , Embrião de Mamíferos/citologia , Endotélio Vascular/citologia , Feminino , Masculino , Camundongos , Pericárdio/citologia , Ratos , Transdução de Sinais
6.
J Cell Mol Med ; 19(6): 1375-89, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25752780

RESUMO

The presence of distinct electrophysiological pathways within the atrioventricular node (AVN) is a prerequisite for atrioventricular nodal reentrant tachycardia to occur. In this study, the different cell contributions that may account for the anatomical and functional heterogeneity of the AVN were investigated. To study the temporal development of the AVN, the expression pattern of ISL1, expressed in cardiac progenitor cells, was studied in sequential stages performing co-staining with myocardial markers (TNNI2 and NKX2-5) and HCN4 (cardiac conduction system marker). An ISL1+/TNNI2+/HCN4+ continuity between the myocardium of the sinus venosus and atrioventricular canal was identified in the region of the putative AVN, which showed a pacemaker-like phenotype based on single cell patch-clamp experiments. Furthermore, qPCR analysis showed that even during early development, different cell populations can be identified in the region of the putative AVN. Fate mapping was performed by in ovo vital dye microinjection. Embryos were harvested and analysed 24 and 48 hrs post-injection. These experiments showed incorporation of sinus venosus myocardium in the posterior region of the atrioventricular canal. The myocardium of the sinus venosus contributes to the atrioventricular canal. It is postulated that the myocardium of the sinus venosus contributes to nodal extensions or transitional cells of the AVN since these cells are located in the posterior region of the AVN. This finding may help to understand the origin of atrioventricular nodal reentrant tachycardia.


Assuntos
Nó Atrioventricular/metabolismo , Proteínas Aviárias/genética , Miocárdio/metabolismo , Animais , Nó Atrioventricular/anatomia & histologia , Nó Atrioventricular/embriologia , Proteínas Aviárias/metabolismo , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Coração/anatomia & histologia , Coração/embriologia , Coração/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Imageamento Tridimensional , Imuno-Histoquímica , Hibridização In Situ , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Potenciais da Membrana , Microscopia de Fluorescência , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Troponina I/genética , Troponina I/metabolismo
7.
Circ Cardiovasc Genet ; 8(2): 284-93, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25613430

RESUMO

BACKGROUND: Several transcription factors regulate cardiac conduction system (CCS) development and function but the role of each in specifying distinct CCS components remains unclear. GATA-binding factor 6 (GATA6) is a zinc-finger transcription factor that is critical for patterning the cardiovascular system. However, the role of GATA6 in the embryonic heart and CCS has never been shown. METHODS AND RESULTS: We report that Gata6 is expressed abundantly in the proximal CCS during midgestation in mice. Myocardial-specific deletion of the carboxyl zinc-finger of Gata6 induces loss of hyperpolarizing cyclic nucleotide-gated channel, subtype 4 staining in the compact atrioventricular node with some retention of hyperpolarizing cyclic nucleotide-gated channel, subtype 4 staining in the atrioventricular bundle, but has no significant effect on the connexin-40-positive bundle branches. Furthermore, myocardial-specific deletion of the carboxyl zinc-finger of Gata6 alters atrioventricular conduction in postnatal life as assessed by surface and invasive electrophysiological evaluation, as well as decreasing the number of ventricular myocytes and inducing compensatory myocyte hypertrophy. Myocardial-specific deletion of the carboxyl zinc-finger of Gata6 is also associated with downregulation of the transcriptional repressor ID2 and the cardiac sodium-calcium exchanger NCX1 in the proximal CCS, where GATA6 transactivates both of these factors. Finally, carboxyl zinc-finger deletion of Gata6 reduces cell-cycle exit of TBX3+ myocytes in the developing atrioventricular bundle during the period of atrioventricular node specification, which results in fewer TBX3+ cells in the proximal CCS of mature mutant mice. CONCLUSIONS: GATA6 contributes to the development and postnatal function of the murine atrioventricular node by promoting cell-cycle exit of specified cardiomyocytes toward a conduction system lineage.


Assuntos
Nó Atrioventricular/embriologia , Fator de Transcrição GATA6/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Fator de Transcrição GATA6/genética , Camundongos , Camundongos Mutantes
8.
Methods Mol Biol ; 1092: 195-219, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24318822

RESUMO

Supraventricular tachycardias are the most prevalent group of arrhythmias observed in the fetus and infant and their incidence increases through early childhood. The molecular pathogenesis of embryonic cardiac dysfunction is poorly understood, due in part to the absence of imaging techniques that provide functional information at the cellular and molecular levels in the developing mammalian heart, particularly during early heart formation. The combination of protein engineering, genetic specification, and high-resolution optical imaging enables new insights into cardiac function and dysfunction during cardiac development. Here we describe the use of GCaMP2, a genetically encoded Ca(2+) indicator (GECI), to determine the processes of cardiac electrical activation during cardiac organogenesis. Transgenic specification of GCaMP2 in mice allows sufficient expression for Ca(2+) imaging as early as embryonic day (e.d.) 9.5, just after the heart begins to function at e.d. 8.5. Crosses with knockout lines in which lethality occurs due to cardiac dysfunction will enable precise determination of the conduction or excitation-contraction coupling phenotypes and thereby improve the understanding of the genetic basis of heart development and the consequence of gene mutations. Moreover, lineage-specific targeting of these sensors of cell signaling provides a new window on the molecular specification of the heart conduction system. We describe mouse lines and imaging methods used to examine conduction in the pre-septated heart (e.d. 10.5), which occurs through dramatically slowed atrioventricular (AV) canal conduction, producing a delay between atrial and ventricular activation prior to the development of the AV node. Genetic constructs including single and bi-allelic minimal promoter systems, and single allele BAC transgenes, enable general or lineage-specific targeting of GCaMP2. High-resolution imaging of embryonic heart conduction provides a new window on one of the most complex events in the mammalian body plan.


Assuntos
Arritmias Cardíacas/genética , Nó Atrioventricular/embriologia , Frequência Cardíaca/genética , Coração/fisiopatologia , Animais , Arritmias Cardíacas/embriologia , Arritmias Cardíacas/patologia , Nó Atrioventricular/crescimento & desenvolvimento , Desenvolvimento Embrionário/genética , Coração/embriologia , Ventrículos do Coração/embriologia , Camundongos , Engenharia de Proteínas , Transgenes
9.
Circ Res ; 111(12): 1528-38, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22982984

RESUMO

RATIONALE: The gap junctional protein connexin (Cx) 45 is strongly expressed in the early embryonic myocardium. In the adult hearts of mice and humans, the expression mainly is restricted to the cardiac conduction system. Cx45 plays an essential role for development and function of the embryonic heart because general and cardiomyocyte-directed deficiencies of Cx45 in mice lead to embryonic lethality attributable to morphological and functional cardiovascular defects. The function of Cx45 in the adult mouse has not yet been cleared. OBJECTIVE: To clarify the function of Cx45 in the adult mouse heart. METHODS AND RESULTS: To circumvent the embryonic lethality resulting from Cx45 deficiency, mice were generated in which deletion of Cx45 specifically was induced in cardiomyocytes of adult mice. These Cx45-deficient mice were viable but showed a decrease in atrioventricular nodal conductivity. In addition, the Cx30.2 protein that is coexpressed with Cx45 in the cardiac conduction system was posttranscriptionally reduced by 70% in mutant hearts. Furthermore, deletion of both Cx45 and Cx30.2 resulted in viable mice that, however, showed stronger impairment of atrioventricular nodal conduction than the single Cx45-deficient mice. CONCLUSIONS: Cx45 is required for optimal impulse propagation in the atrioventricular node and stabilizes the level of the coexpressed Cx30.2 protein in the adult mouse heart. In contrast to the embryo, Cx45 is not essential for the viability of adult mice.


Assuntos
Nó Atrioventricular/embriologia , Nó Atrioventricular/metabolismo , Conexinas/fisiologia , Coração/embriologia , Coração/fisiologia , Animais , Conexinas/deficiência , Conexinas/genética , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/metabolismo , Camundongos , Camundongos Knockout
10.
PLoS One ; 7(9): e44231, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984480

RESUMO

The endothermic state of mammals and birds requires high heart rates to accommodate the high rates of oxygen consumption. These high heart rates are driven by very similar conduction systems consisting of an atrioventricular node that slows the electrical impulse and a His-Purkinje system that efficiently activates the ventricular chambers. While ectothermic vertebrates have similar contraction patterns, they do not possess anatomical evidence for a conduction system. This lack amongst extant ectotherms is surprising because mammals and birds evolved independently from reptile-like ancestors. Using conserved genetic markers, we found that the conduction system design of lizard (Anolis carolinensis and A. sagrei), frog (Xenopus laevis) and zebrafish (Danio rerio) adults is strikingly similar to that of embryos of mammals (mouse Mus musculus, and man) and chicken (Gallus gallus). Thus, in ectothermic adults, the slow conducting atrioventricular canal muscle is present, no fibrous insulating plane is formed, and the spongy ventricle serves the dual purpose of conduction and contraction. Optical mapping showed base-to-apex activation of the ventricles of the ectothermic animals, similar to the activation pattern of mammalian and avian embryonic ventricles and to the His-Purkinje systems of the formed hearts. Mammalian and avian ventricles uniquely develop thick compact walls and septum and, hence, form a discrete ventricular conduction system from the embryonic spongy ventricle. Our study uncovers the evolutionary building plan of heart and indicates that the building blocks of the conduction system of adult ectothermic vertebrates and embryos of endotherms are similar.


Assuntos
Evolução Biológica , Sistema de Condução Cardíaco/embriologia , Animais , Nó Atrioventricular/anatomia & histologia , Nó Atrioventricular/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Sistema de Condução Cardíaco/anatomia & histologia , Sistema de Condução Cardíaco/diagnóstico por imagem , Sistema de Condução Cardíaco/metabolismo , Ventrículos do Coração/anatomia & histologia , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Humanos , Imageamento Tridimensional , Lagartos/embriologia , Lagartos/genética , Modelos Biológicos , Fenótipo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Ultrassonografia , Xenopus/embriologia , Xenopus/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética
11.
J Cardiovasc Electrophysiol ; 23(3): 309-18, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21985309

RESUMO

INTRODUCTION: Understanding sinoatrial node (SAN) development could help in developing therapies for SAN dysfunction. However, electrophysiological investigation of SAN development remains difficult because mutant mice with SAN dysfunctions are frequently embryonically lethal. Most research on SAN development is therefore limited to immunocytochemical observations without comparable functional studies. METHODS AND RESULTS: We applied a multielectrode array (MEA) recording system to study SAN development in mouse hearts acutely isolated at embryonic ages (E) 8.5-12.5 days. Physiological heart rates were routinely restored, enabling accurate functional assessment of SAN development. We found that dominant pacemaking activity originated from the left inflow tract (LIFT) region at E8.5, but switched to the right SAN by E12.5. Combining MEA recordings and pharmacological agents, we show that intracellular calcium (Ca(2+))-mediated automaticity develops early and is the major mechanism of pulse generation in the LIFT of E8.5 hearts. Later in development at E12.5, sarcolemmal ion channels develop in the SAN at a time when pacemaker channels are down-regulated in the LIFT, leading to a switch in the dominant pacemaker location. Additionally, low micromolar concentrations of tetrodotoxin (TTX), a sodium channel blocker, minimally affect pacemaker rhythm at E8.5-E12.5, but suppress atrial activation and reveal a TTX-resistant SAN-atrioventricular node (internodal) pathway that mediates internodal conduction in E12.5 hearts. CONCLUSIONS: Using a physiological mapping method, we demonstrate that differential mechanistic development of automaticity between the left and right inflow tract regions confers the pacemaker location switch. Moreover, a TTX-resistant pathway mediates preferential internodal conduction in E12.5 mouse hearts.


Assuntos
Nó Atrioventricular/fisiologia , Relógios Biológicos/fisiologia , Fenômenos Eletrofisiológicos , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/fisiologia , Coração/embriologia , Nó Sinoatrial/fisiologia , Algoritmos , Animais , Nó Atrioventricular/efeitos dos fármacos , Nó Atrioventricular/embriologia , Relógios Biológicos/efeitos dos fármacos , Compostos de Boro/farmacologia , Sinalização do Cálcio/fisiologia , Feminino , Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Hibridização In Situ , Técnicas In Vitro , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Potenciais da Membrana/fisiologia , Camundongos , Gravidez , Rianodina/farmacologia , Sarcolema/efeitos dos fármacos , Sarcolema/metabolismo , Nó Sinoatrial/efeitos dos fármacos , Nó Sinoatrial/embriologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia
12.
Clin Dev Immunol ; 2012: 432176, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23320018

RESUMO

BACKGROUND: The presence of anti-SSA/Ro and anti-SSB/La antibodies during pregnancy is associated with fetal congenital heart block (CHB), which is primarily diagnosed through fetal echocardiography. Conclusive information about the complete electrophysiology of the fetal cardiac conducting system is still lacking. In addition to echocardiography, fetal magnetocardiography (fMCG) can be used. fMCG is the magnetic analogue of the fetal electrocardiogram (ECG). PATIENTS AND METHODS: Forty-eight pregnant women were enrolled in an observational study; 16 of them tested positive for anti-SSA/Ro and anti-SSB/La antibodies. In addition to routine fetal echocardiography, fMCG was used. Fetal cardiac time intervals (fCTIs) were extracted from the magnetic recordings by predefined procedures. ECGs in the neonates of the study group were performed within the first month after delivery. RESULTS: The PQ segment of the fCTI was significantly prolonged in the study group (P = 0.007), representing a delay of the electrical impulse in the atrioventricular (AV) node. Other fCTIs were within normal range. None of the anti-SSA/Ro and/or anti-SSB/La fetuses progressed to a more advanced heart block during pregnancy or after birth. CONCLUSION: The study identified a low-risk population within antibody positive mothers, where PQ segment prolongation is associated with a lack of progression of the disease.


Assuntos
Anticorpos Antinucleares/imunologia , Nó Atrioventricular/embriologia , Nó Atrioventricular/patologia , Feto/imunologia , Feto/patologia , Adulto , Nó Atrioventricular/imunologia , Estudos de Casos e Controles , Ecocardiografia/métodos , Eletrocardiografia/métodos , Feminino , Bloqueio Cardíaco/congênito , Bloqueio Cardíaco/diagnóstico , Bloqueio Cardíaco/imunologia , Bloqueio Cardíaco/patologia , Humanos , Magnetocardiografia/métodos , Pessoa de Meia-Idade , Gravidez , Diagnóstico Pré-Natal , Adulto Jovem
13.
Birth Defects Res A Clin Mol Teratol ; 91(6): 565-77, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21630423

RESUMO

Defects originating from the atrioventricular canal region are part of a wide spectrum of congenital cardiovascular malformations that frequently affect newborns. These defects include partial or complete atrioventricular septal defects, atrioventricular valve defects, and arrhythmias, such as atrioventricular re-entry tachycardia, atrioventricular nodal block, and ventricular preexcitation. Insight into the cellular origin of the atrioventricular canal myocardium and the molecular mechanisms that control its development will aid in the understanding of the etiology of the atrioventricular defects. This review discusses current knowledge concerning the origin and fate of the atrioventricular canal myocardium, the molecular mechanisms that determine its specification and differentiation, and its role in the development of certain malformations such as those that underlie ventricular preexcitation.


Assuntos
Nó Atrioventricular/citologia , Nó Atrioventricular/embriologia , Linhagem da Célula , Animais , Nó Atrioventricular/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/patologia , Humanos
14.
Circ Arrhythm Electrophysiol ; 4(4): 532-42, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21576278

RESUMO

BACKGROUND: Recent studies in experimental animals have revealed some molecular mechanisms underlying the differentiation of the myocardium making up the conduction system. To date, lack of gene expression data for the developing human conduction system has precluded valid extrapolations from experimental studies to the human situation. METHODS AND RESULTS: We performed immunohistochemical analyses of the expression of key transcription factors, such as ISL1, TBX3, TBX18, and NKX2-5, ion channel HCN4, and connexins in the human embryonic heart. We supplemented our molecular analyses with 3-dimensional reconstructions of myocardial TBX3 expression. TBX3 is expressed in the developing conduction system and in the right venous valve, atrioventricular ring bundles, and retro-aortic nodal region. TBX3-positive myocardium, with exception of the top of the ventricular septum, is devoid of fast-conducting connexin40 and connexin43 and hence identifies slowly conducting pathways. In the early embryonic heart, we found wide expression of the pacemaker channel HCN4 at the venous pole, including the atrial chambers. HCN4 expression becomes confined during later developmental stages to the components of the conduction system. Patterns of expression of transcription factors, known from experimental studies to regulate the development of the sinus node and atrioventricular conduction system, are similar in the human and mouse developing hearts. CONCLUSIONS: Our findings point to the comparability of mechanisms governing the development of the cardiac conduction patterning in human and mouse, which provide a molecular basis for understanding the functioning of the human developing heart before formation of a discrete conduction system.


Assuntos
Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/metabolismo , Coração/embriologia , Miocárdio/metabolismo , Fatores de Transcrição/metabolismo , Nó Atrioventricular/embriologia , Nó Atrioventricular/metabolismo , Nó Atrioventricular/patologia , Conexina 43/metabolismo , Conexinas/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Sistema de Condução Cardíaco/patologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Proteínas Musculares/metabolismo , Miocárdio/patologia , Canais de Potássio , Nó Sinoatrial/embriologia , Nó Sinoatrial/metabolismo , Nó Sinoatrial/patologia , Proteínas com Domínio T/metabolismo , Proteína alfa-5 de Junções Comunicantes
15.
Pediatr Res ; 70(1): 37-43, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21399557

RESUMO

Atrioventricular reentry tachycardia (AVRT) requiring an accessory atrioventricular pathway (AP) is the most common type of arrhythmia in the perinatal period. The etiology of these arrhythmias is not fully understood as well as their capability to dissipate spontaneously in the first year of life. Temporary presence of APs during annulus fibrosus development might cause this specific type of arrhythmias. To study the presence of APs, electrophysiological recordings of ventricular activation patterns and immunohistochemical analyses with antibodies specifically against atrial myosin light chain 2 (MLC-2a), Periostin, Nkx2.5, and Connexin-43 were performed in embryonic mouse hearts ranging from 11.5 to 18.5 days post-conception (dpc). The electrophysiological recordings revealed the presence of functional APs in early (13.5-15.5 dpc) and late (16.5-18.5 dpc) postseptated stages of mouse heart development. These APs stained positive for MLC-2a and Nkx2.5 and negative for Periostin and Connexin-43. Longitudinal analyses showed that APs gradually decreased in number (p = 0.003) and size (p = 0.035) at subsequent developmental stages (13.5-18.5 dpc). Expression of periostin was observed in the developing annulus fibrosus, adjacent to APs and other locations where formation of fibrous tissue is essential. We conclude that functional APs are present during normal mouse heart development. These APs can serve as transient substrate for AVRTs in the perinatal period of development.


Assuntos
Feixe Acessório Atrioventricular/fisiopatologia , Nó Atrioventricular/fisiopatologia , Taquicardia por Reentrada no Nó Atrioventricular/fisiopatologia , Feixe Acessório Atrioventricular/embriologia , Feixe Acessório Atrioventricular/metabolismo , Potenciais de Ação , Animais , Nó Atrioventricular/embriologia , Nó Atrioventricular/metabolismo , Moléculas de Adesão Celular/metabolismo , Conexina 43/metabolismo , Idade Gestacional , Frequência Cardíaca , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Cadeias Leves de Miosina/metabolismo , Organogênese , Taquicardia por Reentrada no Nó Atrioventricular/embriologia , Taquicardia por Reentrada no Nó Atrioventricular/metabolismo , Fatores de Transcrição/metabolismo
16.
J Clin Invest ; 121(2): 513-6, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266771

RESUMO

In the normal heart, an insulating barrier separates the atria and ventricles. The only way in which electrical impulses can cross this barrier is via the atrioventricular (AV) node, which delays impulse conduction to ensure the forward flow of the blood. However, in some individuals, additional muscular bundles (accessory pathways) allow rapid conduction of electrical impulses from the atria to the ventricles, resulting in premature ventricular excitation and contraction. In this issue of the JCI, two independent research groups demonstrate that erroneous development of the embryonic AV canal, which performs a similar function to that of the adult AV node, is a novel mechanism by which accessory pathways can form.


Assuntos
Feixe Acessório Atrioventricular/embriologia , Feixe Acessório Atrioventricular/fisiopatologia , Nó Atrioventricular/embriologia , Nó Atrioventricular/fisiopatologia , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/fisiopatologia , Ventrículos do Coração/fisiopatologia , Adulto , Animais , Humanos , Camundongos , Síndrome de Wolff-Parkinson-White
17.
J Clin Invest ; 121(2): 534-44, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266775

RESUMO

Ventricular preexcitation, a feature of Wolff-Parkinson-White syndrome, is caused by accessory myocardial pathways that bypass the annulus fibrosus. This condition increases the risk of atrioventricular tachycardia and, in the presence of atrial fibrillation, sudden death. The developmental mechanisms underlying accessory pathway formation are poorly understood but are thought to primarily involve malformation of the annulus fibrosus. Before birth, slowly conducting atrioventricular myocardium causes a functional atrioventricular activation delay in the absence of the annulus fibrosus. This myocardium remains present after birth, suggesting that the disturbed development of the atrioventricular canal myocardium may mediate the formation of rapidly conducting accessory pathways. Here we show that myocardium-specific inactivation of T-box 2 (Tbx2), a transcription factor essential for atrioventricular canal patterning, leads to the formation of fast-conducting accessory pathways, malformation of the annulus fibrosus, and ventricular preexcitation in mice. The accessory pathways ectopically express proteins required for fast conduction (connexin-40 [Cx40], Cx43, and sodium channel, voltage-gated, type V, α [Scn5a]). Additional inactivation of Cx30.2, a subunit for gap junctions with low conductance expressed in the atrioventricular canal and unaffected by the loss of Tbx2, did not affect the functionality of the accessory pathways. Our results suggest that malformation of the annulus fibrosus and preexcitation arise from the disturbed development of the atrioventricular myocardium.


Assuntos
Feixe Acessório Atrioventricular , Nó Atrioventricular , Sistema de Condução Cardíaco , Morfogênese , Proteínas com Domínio T/metabolismo , Síndrome de Wolff-Parkinson-White/patologia , Síndrome de Wolff-Parkinson-White/fisiopatologia , Feixe Acessório Atrioventricular/embriologia , Feixe Acessório Atrioventricular/patologia , Feixe Acessório Atrioventricular/fisiopatologia , Animais , Nó Atrioventricular/embriologia , Nó Atrioventricular/patologia , Nó Atrioventricular/fisiopatologia , Conexina 43/genética , Conexina 43/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/patologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/metabolismo , Gravidez , Proteínas com Domínio T/genética
18.
J Clin Invest ; 121(2): 525-33, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266778

RESUMO

Ventricular preexcitation, which characterizes Wolff-Parkinson-White syndrome, is caused by the presence of accessory pathways that can rapidly conduct electrical impulses from atria to ventricles, without the intrinsic delay characteristic of the atrioventricular (AV) node. Preexcitation is associated with an increased risk of tachyarrhythmia, palpitations, syncope, and sudden death. Although the pathology and electrophysiology of preexcitation syndromes are well characterized, the developmental mechanisms are poorly understood, and few animal models that faithfully recapitulate the human disorder have been described. Here we show that activation of Notch signaling in the developing myocardium of mice can produce fully penetrant accessory pathways and ventricular preexcitation. Conversely, inhibition of Notch signaling in the developing myocardium resulted in a hypoplastic AV node, with specific loss of slow-conducting cells expressing connexin-30.2 (Cx30.2) and a resulting loss of physiologic AV conduction delay. Taken together, our results suggest that Notch regulates the functional maturation of AV canal embryonic myocardium during the development of the specialized conduction system. Our results also show that ventricular preexcitation can arise from inappropriate patterning of the AV canal-derived myocardium.


Assuntos
Feixe Acessório Atrioventricular , Nó Atrioventricular , Sistema de Condução Cardíaco , Receptores Notch/antagonistas & inibidores , Transdução de Sinais/fisiologia , Síndrome de Wolff-Parkinson-White , Feixe Acessório Atrioventricular/embriologia , Feixe Acessório Atrioventricular/patologia , Feixe Acessório Atrioventricular/fisiopatologia , Animais , Nó Atrioventricular/anatomia & histologia , Nó Atrioventricular/embriologia , Nó Atrioventricular/fisiopatologia , Ecocardiografia , Eletrocardiografia , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/patologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Camundongos , Receptores Notch/genética , Receptores Notch/metabolismo , Síndrome de Wolff-Parkinson-White/patologia , Síndrome de Wolff-Parkinson-White/fisiopatologia
19.
Circ Res ; 107(6): 728-36, 2010 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-20671237

RESUMO

RATIONALE: The clinically important atrioventricular conduction axis is structurally complex and heterogeneous, and its molecular composition and developmental origin are uncertain. OBJECTIVE: To assess the molecular composition and 3D architecture of the atrioventricular conduction axis in the postnatal mouse heart and to define the developmental origin of its component parts. METHODS AND RESULTS: We generated an interactive 3D model of the atrioventricular junctions in the mouse heart using the patterns of expression of Tbx3, Hcn4, Cx40, Cx43, Cx45, and Nav1.5, which are important for conduction system function. We found extensive figure-of-eight rings of nodal and transitional cells around the mitral and tricuspid junctions and in the base of the atrial septum. The rings included the compact node and nodal extensions. We then used genetic lineage labeling tools (Tbx2(+/Cre), Mef2c-AHF-Cre, Tbx18(+/Cre)), along with morphometric analyses, to assess the developmental origin of the specific components of the axis. The majority of the atrial components, including the atrioventricular rings and compact node, are derived from the embryonic atrioventricular canal. The atrioventricular bundle, including the lower cells of the atrioventricular node, in contrast, is derived from the ventricular myocardium. No contributions to the conduction system myocardium were identified from the sinus venosus, the epicardium, or the dorsal mesenchymal protrusion. CONCLUSIONS: The atrioventricular conduction axis comprises multiple domains with distinctive molecular signatures. The atrial part proliferates from the embryonic atrioventricular canal, along with myocytes derived from the developing atrial septum. The atrioventricular bundle and lower nodal cells are derived from ventricular myocardium.


Assuntos
Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/crescimento & desenvolvimento , Processamento de Imagem Assistida por Computador , Animais , Nó Atrioventricular/anatomia & histologia , Nó Atrioventricular/embriologia , Nó Atrioventricular/crescimento & desenvolvimento , Feminino , Coração/anatomia & histologia , Coração/embriologia , Coração/crescimento & desenvolvimento , Sistema de Condução Cardíaco/anatomia & histologia , Processamento de Imagem Assistida por Computador/métodos , Imageamento Tridimensional , Camundongos , Camundongos Transgênicos , Gravidez
20.
Mol Cell Biol ; 30(14): 3711-21, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20457809

RESUMO

One essential downstream signaling pathway of receptor tyrosine kinases (RTKs), such as vascular endothelial growth factor receptor (VEGFR) and the Tie2 receptor, is the phosphoinositide-3 kinase (PI3K)-phosphoinositide-dependent protein kinase 1 (PDK1)-Akt/protein kinase B (PKB) cascade that plays a critical role in development and tumorigenesis. However, the role of PDK1 in cardiovascular development remains unknown. Here, we deleted PDK1 specifically in endothelial cells in mice. These mice displayed hemorrhage and hydropericardium and died at approximately embryonic day 11.5 (E11.5). Histological analysis revealed defective vascular remodeling and development and disrupted integrity between the endothelium and trabeculae/myocardium in the heart. The atrioventricular canal (AVC) cushion and valves failed to form, indicating a defect in epithelial-mesenchymal transition (EMT), together with increased endothelial apoptosis. Consistently, ex vivo AVC explant culture showed impeded mesenchymal outgrowth. Snail protein was reduced and was absent from the nucleus in AVC cells. Delivery of the Snail S6A mutant to the AVC explant effectively rescued EMT defects. Furthermore, adenoviral Akt delivery rescued EMT defects in AVC explant culture, and deletion of PTEN delayed embryonic lethality of PDK1 endothelial deletion mice by 1 day and rendered normal development of the AVC cushion in the PDK1-deficient heart. Taken together, these results have revealed an essential role of PDK1 in cardiovascular development through activation of Akt and Snail.


Assuntos
Coração Fetal/embriologia , Coração Fetal/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Apoptose , Nó Atrioventricular/anormalidades , Nó Atrioventricular/embriologia , Nó Atrioventricular/enzimologia , Sequência de Bases , Primers do DNA/genética , Técnicas de Cultura Embrionária , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Epitélio/embriologia , Epitélio/enzimologia , Feminino , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/enzimologia , Cardiopatias Congênitas/genética , Valvas Cardíacas/anormalidades , Valvas Cardíacas/embriologia , Valvas Cardíacas/enzimologia , Mesoderma/embriologia , Mesoderma/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PTEN Fosfo-Hidrolase/metabolismo , Gravidez , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...