Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Mol Cell Cardiol ; 143: 26-37, 2020 06.
Article in English | MEDLINE | ID: mdl-32277975

ABSTRACT

RATIONALE: After cardiac damage, excessive neurite outgrowth (sympathetic hyperinnervation) can occur, which is related to ventricular arrhythmias/sudden cardiac death. Post-damage reactivation of epicardium causes epicardium-derived cells (EPDCs) to acquire a mesenchymal character, contributing to cardiac regeneration. Whether EPDCs also contribute to cardiac re/hyperinnervation, is unknown. AIM: To investigate whether mesenchymal EPDCs influence cardiac sympathetic innervation. METHODS AND RESULTS: Sympathetic ganglia were co-cultured with mesenchymal EPDCs and/or myocardium, and neurite outgrowth and sprouting density were assessed. Results showed a significant increase in neurite density and directional (i.e. towards myocardium) outgrowth when ganglia were co-cultured with a combination of EPDCs and myocardium, as compared to cultures with EPDCs or myocardium alone. In absence of myocardium, this outgrowth was not directional. Neurite differentiation of PC12 cells in conditioned medium confirmed these results via a paracrine effect, in accordance with expression of neurotrophic factors in myocardial explants co-cultured with EPDCs. Of interest, EPDCs increased the expression of nerve growth factor (NGF) in cultured, but not in fresh myocardium, possibly due to an "ischemic state" of cultured myocardium, supported by TUNEL and Hif1α expression. Cardiac tissues after myocardial infarction showed robust NGF expression in the infarcted, but not remote area. CONCLUSION: Neurite outgrowth and density increases significantly in the presence of EPDCs by a paracrine effect, indicating a new role for EPDCs in the occurrence of sympathetic re/hyperinnervation after cardiac damage.


Subject(s)
Heart/innervation , Myocardium/metabolism , Pericardium/metabolism , Sympathetic Fibers, Postganglionic/physiology , Animals , Apoptosis/genetics , Cell Line, Tumor , Cells, Cultured , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/metabolism , Humans , Mice , Myocardium/cytology , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Neuronal Outgrowth
2.
Dev Dyn ; 248(8): 634-656, 2019 08.
Article in English | MEDLINE | ID: mdl-31063648

ABSTRACT

The mechanisms of the evolution and development of the heart in metazoans are highlighted, starting with the evolutionary origin of the contractile cell, supposedly the precursor of cardiomyocytes. The last eukaryotic common ancestor is likely a combination of several cellular organisms containing their specific metabolic pathways and genetic signaling networks. During evolution, these tool kits diversified. Shared parts of these conserved tool kits act in the development and functioning of pumping hearts and open or closed circulations in such diverse species as arthropods, mollusks, and chordates. The genetic tool kits became more complex by gene duplications, addition of epigenetic modifications, influence of environmental factors, incorporation of viral genomes, cardiac changes necessitated by air-breathing, and many others. We evaluate mechanisms involved in mollusks in the formation of three separate hearts and in arthropods in the formation of a tubular heart. A tubular heart is also present in embryonic stages of chordates, providing the septated four-chambered heart, in birds and mammals passing through stages with first and second heart fields. The four-chambered heart permits the formation of high-pressure systemic and low-pressure pulmonary circulation in birds and mammals, allowing for high metabolic rates and maintenance of body temperature. Crocodiles also have a (nearly) separated circulation, but their resting temperature conforms with the environment. We argue that endothermic ancestors lost the capacity to elevate their body temperature during evolution, resulting in ectothermic modern crocodilians. Finally, a clinically relevant paragraph reviews the occurrence of congenital cardiac malformations in humans as derailments of signaling pathways during embryonic development.


Subject(s)
Biological Evolution , Embryonic Development , Heart , Animals , Heart/embryology , Heart Defects, Congenital/etiology , Humans
3.
Pediatr Cardiol ; 39(4): 690-694, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29340729

ABSTRACT

The aortic and pulmonary valve share a common developmental origin from the embryonic arterial trunk. Bicuspid aortic valve is the most common congenital anomaly and can occur isolated as well as in association with other congenital heart disease (CHD). Data on pulmonary valve morphology in these cases are scarce. In this study, we aimed to determine pulmonary valve morphology in hearts with BAV associated with CHD. In 83 post-mortem heart specimens with BAV and associated CHD, pulmonary valve morphology was studied and related to BAV morphology. In 14/83 (17%) hearts, the pulmonary valve was affected, bicuspid in 8/83 (10%), dome-shaped in 3/83 (4%) and atretic in 3/83 (4%). In specimens with a bicuspid pulmonary valve, 5/8 (63%) had a strictly bicuspid aortic valve (without raphe), 2/3 hearts (67%) with dome-shaped pulmonary valves and 2/3 hearts (67%) with atretic pulmonary valves had BAV without raphe. Six out of eight (75%) specimens with a bicuspid pulmonary valve had a perimembranous ventricular septal defect (VSD). 4/8 (50%) specimens with a bicuspid pulmonary valve were associated with chromosomal abnormalities: 3 (38%) had trisomy 18 and 1 (13%) had trisomy 13. In BAV with associated CHD, abnormal pulmonary valve morphology was observed in 17% of the hearts. The majority of hearts with abnormal pulmonary valve morphology had a Type B bicuspid aortic valve (without raphe). Bilateral semilunar valvular disease is associated with Type B BAVs and in many cases related to chromosomal abnormalities. As this study was performed in post-mortem specimens with high frequency of associated CHD, caution is warranted with application of these results to the general BAV population.


Subject(s)
Aortic Valve/abnormalities , Heart Defects, Congenital/epidemiology , Heart Valve Diseases/complications , Pulmonary Valve/abnormalities , Adolescent , Bicuspid Aortic Valve Disease , Cadaver , Child , Child, Preschool , Female , Humans , Incidence , Infant , Infant, Newborn , Male , Young Adult
4.
Dev Dyn ; 245(11): 1107-1123, 2016 11.
Article in English | MEDLINE | ID: mdl-27580238

ABSTRACT

BACKGROUND: 14-3-3ε plays an important role in the maturation of the compact ventricular myocardium by modulating the cardiomyocyte cell cycle via p27kip1 . However, additional cardiac defects are possible given the ubiquitous expression pattern of this protein. RESULTS: Germ line deletion of 14-3-3ε led to malalignment of both the outflow tract (OFT) and atrioventricular (AV) cushions, with resulting tricuspid stenosis and atresia, mitral valve abnormalities, and perimembranous ventricular septal defects (VSDs). We confirmed myocardial non-compaction and detected a spongy septum with muscular VSDs and blebbing of the epicardium. These defects were associated with abnormal patterning of p27kip1 expression in the subendocardial and possibly the epicardial cell populations. In addition to abnormal pharyngeal arch artery patterning, we found deep endocardial recesses and paucity of intramyocardial coronary vasculature as a result of defective coronary plexus remodeling. CONCLUSIONS: The malalignment of both endocardial cushions provides a new explanation for tricuspid and mitral valve defects, while myocardial non-compaction provides the basis for the abnormal coronary vasculature patterning. These abnormalities might arise from p27kip1 dysregulation and a resulting defect in epithelial-to-mesenchymal transformation. These data suggest that 14-3-3ε, in addition to left ventricular non-compaction (LVNC), might be linked to different forms of congenital heart disease (CHD). Developmental Dynamics 245:1107-1123, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
14-3-3 Proteins/metabolism , Endocardium/pathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , 14-3-3 Proteins/genetics , Animals , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endocardium/metabolism , Gene Expression Regulation, Developmental , Mice , Myocardium/metabolism , Myocardium/pathology
5.
Physiol Genomics ; 48(11): 771-784, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27591124

ABSTRACT

Regenerative therapies hold great potential to change the treatment paradigm for cardiac diseases. Human cardiac progenitor cells can be used for drug discovery in this area and also provide a renewable source of cardiomyocytes. However, a better understanding of their characteristics is critical for interpreting data obtained from drug screening using these cells. In the present study, we performed global transcriptional analysis of two important sources of cardiac progenitors, i.e., patient epicardium-derived cells (EPDCs) and cardiac progenitor cells (CPCs) derived from human induced pluripotent stem cells. In addition, we also compared the gene expression profiles of these cells when they were cultured under normoxic and hypoxic conditions. We identified 3,289 mRNAs that were differentially expressed between EPDCs and CPCs. Gene ontology annotation and pathway enrichment analyses further revealed possible unique functions of these two cell populations. Notably, the impact of hypoxia vs normoxia on gene expression was modest and only a few genes (e.g., AK4, ALDOC, BNIP3P1, PGK1, and SLC2A1) were upregulated in EPDCs and CPCs after the cells were exposed to low oxygen for 24 h. Finally, we also performed a focused analysis of the gene expression patterns of a predefined set of 92 paracrine factors. We identified 30 of these genes as differentially expressed, and 29 were expressed at higher levels in EPDCs compared with CPCs. Taken together, the results of the present study advance our understanding of the transcriptional programs in EPDCs and CPCs and highlights important differences and similarities between these cell populations.


Subject(s)
Gene Expression Profiling , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Pericardium/cytology , Biomarkers/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cluster Analysis , Gene Expression Regulation/drug effects , Gene Ontology , Humans , Induced Pluripotent Stem Cells/drug effects , Molecular Sequence Annotation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxygen/pharmacology , Paracrine Communication/genetics , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics
6.
J Mol Cell Cardiol ; 89(Pt B): 251-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26527381

ABSTRACT

The cardiac autonomic nervous system (cANS) modulates heart rate, contraction force and conduction velocity. The embryonic chicken heart already responds to epinephrine prior to establishment of the cANS. The aim of this study was to define the regions of the heart that might participate in modulating the early autonomic response to epinephrine. Immunofluorescence analysis reveals expression of neural markers tubulin beta-3 chain and neural cell adhesion molecule in the epicardium during early development. In addition, expression of the ß2 adrenergic receptor, the receptor for epinephrine, was found in the epicardium. Ex-ovo micro-electrode recordings in hearts with inhibition of epicardial outgrowth showed a significantly reduced response of the heart rate to epinephrine compared to control hearts. This study suggests a role for the epicardium as autonomic modulator during early cardiac development.


Subject(s)
Autonomic Nervous System/embryology , Embryonic Development , Pericardium/embryology , Animals , Autonomic Nervous System/metabolism , Biomarkers/metabolism , Chick Embryo , Epinephrine/pharmacology , Gene Expression Regulation, Developmental , Humans , Mice , Neurons/metabolism , Pericardium/metabolism , Receptors, Adrenergic, beta/metabolism , Spinal Cord/metabolism , Tubulin/metabolism , WT1 Proteins/metabolism
7.
J Cell Mol Med ; 19(6): 1375-89, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25752780

ABSTRACT

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.


Subject(s)
Atrioventricular Node/metabolism , Avian Proteins/genetics , Myocardium/metabolism , Animals , Atrioventricular Node/anatomy & histology , Atrioventricular Node/embryology , Avian Proteins/metabolism , Chick Embryo , Gene Expression Regulation, Developmental , Heart/anatomy & histology , Heart/embryology , Heart/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Imaging, Three-Dimensional , Immunohistochemistry , In Situ Hybridization , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Membrane Potentials , Microscopy, Fluorescence , Myocardium/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Troponin I/genetics , Troponin I/metabolism
8.
Eur Heart J ; 35(14): 920-31, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23048191

ABSTRACT

AIMS: Hypoplastic coronary artery disease is a rare congenital abnormality that is associated with sudden cardiac death. However, molecular mechanisms responsible for this disease are not clear. The aim of the present study was to assess the role of nitric oxide synthase-3 (NOS3) in the pathogenesis of hypoplastic coronary arteries. METHODS AND RESULTS: Wild-type (WT), NOS3(-/-), and a novel cardiac-specific NOS3 overexpression mouse model were employed. Deficiency in NOS3 resulted in coronary artery hypoplasia in foetal mice and spontaneous myocardial infarction in postnatal hearts. Coronary artery diameters, vessel density, and volume were significantly decreased in NOS3(-/-) mice at postnatal day 0. In addition, NOS3(-/-) mice showed a significant increase in the ventricular wall thickness, myocardial volume, and cardiomyocyte cell size compared with WT mice. Lack of NOS3 also down-regulated the expression of Gata4, Wilms tumour-1, vascular endothelial growth factor, basic fibroblast growth factor and erythropoietin, and inhibited migration of epicardial cells. These abnormalities and hypoplastic coronary arteries in the NOS3(-/-) mice were completely rescued by the cardiac-specific overexpression of NOS3. CONCLUSION: Nitric oxide synthase-3 is required for coronary artery development and deficiency in NOS3 leads to hypoplastic coronary arteries.


Subject(s)
Coronary Vessel Anomalies/enzymology , Myocardial Infarction/enzymology , Nitric Oxide Synthase Type III/deficiency , Animals , Animals, Newborn , Coronary Vessel Anomalies/embryology , Mice, Transgenic , Myocardial Infarction/embryology , Survival Analysis
9.
Cardiovasc Diabetol ; 13: 46, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24533448

ABSTRACT

BACKGROUND: Pregestational diabetes is a major risk factor of congenital heart defects (CHDs). Glutathione is depleted and reactive oxygen species (ROS) production is elevated in diabetes. In the present study, we aimed to examine whether treatment with N-acetylcysteine (NAC), which increases glutathione synthesis and inhibits ROS production, prevents CHDs induced by pregestational diabetes. METHODS: Female mice were treated with streptozotocin (STZ) to induce pregestational diabetes prior to breeding with normal males to produce offspring. Some diabetic mice were treated with N-acetylcysteine (NAC) in drinking water from E0.5 to the end of gestation or harvesting of the embryos. CHDs were identified by histology. ROS levels, cell proliferation and gene expression in the fetal heart were analyzed. RESULTS: Our data show that pregestational diabetes resulted in CHDs in 58% of the offspring, including ventricular septal defect (VSD), atrial septal defect (ASD), atrioventricular septal defects (AVSD), transposition of great arteries (TGA), double outlet right ventricle (DORV) and tetralogy of Fallot (TOF). Treatment with NAC in drinking water in pregestational diabetic mice completely eliminated the incidence of AVSD, TGA, TOF and significantly diminished the incidence of ASD and VSD. Furthermore, pregestational diabetes increased ROS, impaired cell proliferation, and altered Gata4, Gata5 and Vegf-a expression in the fetal heart of diabetic offspring, which were all prevented by NAC treatment. CONCLUSIONS: Treatment with NAC increases GSH levels, decreases ROS levels in the fetal heart and prevents the development of CHDs in the offspring of pregestational diabetes. Our study suggests that NAC may have therapeutic potential in the prevention of CHDs induced by pregestational diabetes.


Subject(s)
Acetylcysteine/administration & dosage , Cardiotonic Agents/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Heart Defects, Congenital/prevention & control , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Female , Heart Defects, Congenital/blood , Heart Defects, Congenital/pathology , Male , Mice , Mice, Inbred C57BL , Pregnancy , Pregnancy in Diabetics/blood , Pregnancy in Diabetics/drug therapy , Pregnancy in Diabetics/pathology
10.
ScientificWorldJournal ; 2014: 531324, 2014.
Article in English | MEDLINE | ID: mdl-24707208

ABSTRACT

BACKGROUND: Heart development is a complex process, and abnormal development may result in congenital heart disease (CHD). Currently, studies on animal models mainly focus on cardiac morphology and the availability of hemodynamic data, especially of the right heart half, is limited. Here we aimed to assess the morphological and hemodynamic parameters of normal developing mouse embryos/fetuses by using a high-frequency ultrasound system. METHODS: A timed breeding program was initiated with a WT mouse line (Swiss/129Sv background). All recordings were performed transabdominally, in isoflurane sedated pregnant mice, in hearts of sequential developmental stages: 12.5, 14.5, and 17.5 days after conception (n = 105). RESULTS: Along development the heart rate increased significantly from 125 ± 9.5 to 219 ± 8.3 beats per minute. Reliable flow measurements could be performed across the developing mitral and tricuspid valves and outflow tract. M-mode measurements could be obtained of all cardiac compartments. An overall increase of cardiac systolic and diastolic function with embryonic/fetal development was observed. CONCLUSION: High-frequency echocardiography is a promising and useful imaging modality for structural and hemodynamic analysis of embryonic/fetal mouse hearts.


Subject(s)
Echocardiography, Doppler, Pulsed/methods , Fetal Heart/growth & development , Animals , Diastole/physiology , Female , Fetal Heart/diagnostic imaging , Heart Rate , Male , Mice , Pregnancy , Ultrasonography, Prenatal
11.
Fetal Diagn Ther ; 36(1): 59-68, 2014.
Article in English | MEDLINE | ID: mdl-24903004

ABSTRACT

INTRODUCTION: Bicuspid aortic valve (BAV) is common in Turner syndrome (TS). In adult TS, 82-95% of BAVs have fusion of the right and left coronary leaflets. Data in fetal stages are scarce. The purpose of this study was to gain insight into aortic valve morphology and associated cardiovascular abnormalities in a fetal TS cohort with adverse outcome early in development. MATERIAL AND METHODS: We studied post-mortem heart specimens of 36 TS fetuses and 1 TS newborn. RESULTS: BAV was present in 28 (76%) hearts. BAVs showed fusion of the right and left coronary leaflet (type 1 BAV) in 61%, and fusion of the right coronary and non-coronary leaflet (type 2 BAV) in 39%. There were no significant differences in occurrence of additional cardiovascular abnormalities between type 1 and type 2 BAV. However, all type 2 BAV hearts showed ascending aorta hypoplasia and tubular hypoplasia of the B segment, as opposed to only 55 and 64% of type 1 BAV hearts, respectively. DISCUSSION: The proportion of type 2 BAV seems higher in TS fetuses than in adults. Fetal type 2 BAV hearts all had severe aortic pathology, possibly contributing to a worse prognosis of type 2 than type 1 BAV in TS.


Subject(s)
Aortic Valve/abnormalities , Fetal Heart/abnormalities , Fetal Heart/pathology , Heart Valve Diseases/pathology , Turner Syndrome/pathology , Aortic Valve/embryology , Aortic Valve/pathology , Bicuspid Aortic Valve Disease , Cardiovascular Abnormalities/embryology , Cardiovascular Abnormalities/pathology , Female , Fetal Heart/embryology , Heart/embryology , Heart Valve Diseases/embryology , Humans , Infant, Newborn , Turner Syndrome/embryology
12.
Differentiation ; 84(1): 41-53, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22652098

ABSTRACT

The importance of the epicardium covering the heart and the intrapericardial part of the great arteries has reached a new summit. It has evolved as a major cellular component with impact both in development, disease and more recently also repair potential. The role of the epicardium in development, its differentiation from a proepicardial organ at the venous pole (vPEO) and the differentiation capacities of the vPEO initiating cardiac epicardium (cEP) into epicardium derived cells (EPDCs) have been extensively described in recent reviews on growth and transcription factor pathways. In short, the epicardium is the source of the interstitial, the annulus fibrosus and the adventitial fibroblasts, and differentiates into the coronary arterial smooth muscle cells. Furthermore, EPDCs induce growth of the compact myocardium and differentiation of the Purkinje fibers. This review includes an arterial pole located PEO (aPEO) that provides the epicardium covering the intrapericardial great vessels. In avian and mouse models disturbance of epicardial outgrowth and maturation leads to a broad spectrum of cardiac anomalies with main focus on non-compaction of the myocardium, deficient annulus fibrosis, valve malformations and coronary artery abnormalities. The discovery that in disease both arterial and cardiac epicardium can again differentiate into EPDCs and thus reactivate its embryonic program and potential has highly broadened the scope of research interest. This reactivation is seen after myocardial infarction and also in aneurysm formation of the ascending aorta. Use of EPDCs for cell therapy show their positive function in paracrine mediated repair processes which can be additive when combined with the cardiac progenitor stem cells that probably share the same embryonic origin with EPDCs. Research into the many cell-autonomous and cell-cell-based capacities of the adult epicardium will open up new realistic therapeutic avenues.


Subject(s)
Cell Lineage , Heart Defects, Congenital/embryology , Pericardium/cytology , Pericardium/embryology , Animals , Cell Differentiation , Cell Transplantation , Chick Embryo , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Fibroblasts/cytology , Heart Defects, Congenital/therapy , Humans , Mice , Myoblasts, Cardiac/cytology , Myoblasts, Smooth Muscle/cytology , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology , Pericardium/physiopathology
13.
Differentiation ; 84(1): 131-48, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22664174

ABSTRACT

The cardiac conduction system is a specialized network that initiates and closely coordinates the heart beat. Cardiac conduction system development is intricately related to the development and maturation of the embryonic heart towards its four-chambered form, as is indicated by the fact that disturbed development of cardiac structures is often accompanied by a disturbed formation of the CCS. Electrophysiological studies have shown that selected conduction disturbances and cardiac arrhythmias do not take place randomly in the heart but rather at anatomical predilection sites. Knowledge on development of the CCS may facilitate understanding of the etiology of arrhythmogenic events. In this review we will focus on embryonic development of the CCS in relation to clinical arrhythmias, as well as on specific cardiac conduction abnormalities that are observed in patients with congenital heart disease.


Subject(s)
Arrhythmias, Cardiac/etiology , Heart Conduction System/embryology , Heart Defects, Congenital/physiopathology , Adult , Animals , Arrhythmias, Cardiac/embryology , Child , Disease Models, Animal , Fetal Heart/abnormalities , Heart Conduction System/pathology , Heart Rate , Humans , Mice
14.
Dev Dyn ; 241(9): 1413-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22826212

ABSTRACT

BACKGROUND: Understanding of cardiac outflow tract (OFT) remodeling is essential to explain repositioning of the aorta and pulmonary orifice. In wild type embryos (E9.5-14.5), second heart field contribution (SHF) to the OFT was studied using expression patterns of Islet 1, Nkx2.5, MLC-2a, WT-1, and 3D-reconstructions. Abnormal remodeling was studied in VEGF120/120 embryos. RESULTS: In wild type, Islet 1 and Nkx2.5 positive myocardial precursors formed an asymmetric elongated column almost exclusively at the pulmonary side of the OFT up to the pulmonary orifice. In VEGF120/120 embryos, the Nkx2.5-positive mesenchymal population was disorganized with a short extension along the pulmonary OFT. CONCLUSIONS: We postulate that normally the pulmonary trunk and orifice are pushed in a higher and more frontal position relative to the aortic orifice by asymmetric addition of SHF-myocardium. Deficient or disorganized right ventricular OFT expansion might explain cardiac malformations with abnormal position of the great arteries, such as double outlet right ventricle.


Subject(s)
Body Patterning , Heart/embryology , Lung/physiology , Morphogenesis , Animals , Body Patterning/genetics , Body Patterning/physiology , Computer Simulation , Embryo, Mammalian , Gestational Age , Heart/anatomy & histology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lung/embryology , Lung/metabolism , Mechanical Phenomena , Mice , Mice, Transgenic , Models, Cardiovascular , Morphogenesis/genetics , Morphogenesis/physiology , Rotation , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
J Cell Mol Med ; 16(7): 1508-21, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22003890

ABSTRACT

We previously showed that human cardiomyocyte progenitor cells (hCMPCs) injected after myocardial infarction (MI) had differentiated into cardiomyocytes in vivo 3 months after MI. Here, we investigated the short-term (2 weeks) effects of hCMPCs on the infarcted mouse myocardium. MI was induced in immunocompromised (NOD/scid) mice, immediately followed by intramyocardial injection of hCMPCs labelled with enhanced green fluorescent protein (hCMPC group) or vehicle only (control group). Sham-operated mice served as reference. Cardiac performance was measured 2 and 14 days after MI by magnetic resonance imaging at 9.4 T. Left ventricular (LV) pressure-volume measurements were performed at day 15 followed by extensive immunohistological analysis. Animals injected with hCMPCs demonstrated a higher LV ejection fraction, lower LV end-systolic volume and smaller relaxation time constant than control animals 14 days after MI. hCMPCs engrafted in the infarcted myocardium, did not differentiate into cardiomyocytes, but increased vascular density and proliferation rate in the infarcted and border zone area of the hCMPC group. Injected hCMPCs engraft into murine infarcted myocardium where they improve LV systolic function and attenuate the ventricular remodelling process 2 weeks after MI. Since no cardiac differentiation of hCMPCs was evident after 2 weeks, the observed beneficial effects were most likely mediated by paracrine factors, targeting amongst others vascular homeostasis. These results demonstrate that hCMPCs can be applied to repair infarcted myocardium without the need to undergo differentiation into cardiomyocytes.


Subject(s)
Cell Differentiation , Ischemia/therapy , Myoblasts/transplantation , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Animals , Cells, Cultured , Green Fluorescent Proteins/metabolism , Humans , Ischemia/pathology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred NOD , Mice, SCID , Myoblasts/cytology , Myocardial Infarction/pathology , Ventricular Function, Left , Ventricular Remodeling
16.
Hum Mol Genet ; 19(7): 1286-301, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20071345

ABSTRACT

Total anomalous pulmonary venous return (TAPVR) is a congenital heart defect inherited via complex genetic and/or environmental factors. We report detailed mapping in extended TAPVR kindreds and mutation analysis in TAPVR patients that implicate the PDGFRA gene in the development of TAPVR. Gene expression studies in mouse and chick embryos for both the Pdgfra receptor and its ligand Pdgf-a show temporal and spatial patterns consistent with a role in pulmonary vein (PV) development. We used an in ovo function blocking assay in chick and a conditional knockout approach in mouse to knock down Pdgfra expression in the developing venous pole during the period of PV formation. We observed that loss of PDGFRA function in both organisms causes TAPVR with low penetrance (approximately 7%) reminiscent of that observed in our human TAPVR kindreds. Intermediate inflow tract anomalies occurred in a higher percentage of embryos (approximately 30%), suggesting that TAPVR occurs at one end of a spectrum of defects. We show that the anomalous pulmonary venous connection seen in chick and mouse is highly similar to TAPVR discovered in an abnormal early stage embryo from the Kyoto human embryo collection. Whereas the embryology of the normal venous pole and PV is becoming understood, little is known about the embryogenesis or molecular pathogenesis of TAPVR. These models of TAPVR provide important insight into the pathogenesis of PV defects. Taken together, these data from human genetics and animal models support a role for PDGF-signaling in normal PV development, and in the pathogenesis of TAPVR.


Subject(s)
Heart Defects, Congenital/genetics , Pulmonary Veins/abnormalities , Receptor, Platelet-Derived Growth Factor alpha/genetics , Animals , Chick Embryo , Humans , Mice , Mice, Mutant Strains , Models, Animal , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism
17.
Dev Dyn ; 240(7): 1670-80, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21604321

ABSTRACT

Endothelial cells (EC) translate biomechanical forces into functional and phenotypic responses that play important roles in cardiac development. Specifically, EC in areas of high shear stress, i.e., in the cardiac outflow tract and atrioventricular canal, are characterized by high expression of Krüppel-like factor 2 (Klf2) and by transforming growth factor-beta (Tgfß)-driven endothelial-to-mesenchymal transition. Extraembryonic venous obstruction (venous clip model) results in congenital heart malformations, and venous clip-induced alterations in shear stress-related gene expression are suggestive for an increase in cardiac shear stress. Here, we study the effects of shear stress on Klf2 expression and Tgfß-associated signaling in embryonic EC in vivo using the venous clip model and in vitro by subjecting cultured EC to fluid flow. Cellular responses were assessed by analysis of Klf2, Tgfß ligands, and their downstream signaling targets. Results show that, in embryonic EC, shear stress activates Tgfß/Alk5 signaling and that induction of Klf2 is an Alk5 dependent process.


Subject(s)
Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Shear Strength/physiology , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Chick Embryo , Chickens , Endothelial Cells/cytology , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Middle Aged , Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type I , Umbilical Veins/cytology
18.
Ann Cardiothorac Surg ; 11(4): 380-388, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35958528

ABSTRACT

A bicuspid aortic valve (BAV) is the most prevalent congenital cardiac anomaly, in which the valve has only two leaflets, instead of the normal three. Patients with a BAV have an increased risk of aneurysm formation and the development of an aortic dissection. Vascular smooth muscle cells in both the non- and dilated aortic wall are characterized by a maturation defect in all BAV patients, as compared to patients with a tricuspid aortic valve, which can contribute to inherent developmental susceptibility. Besides structural abnormalities of the vascular wall, a turbulent blood flow, caused by bicuspid valve geometry, could expedite the pathological process in the aortic wall, leading to aortopathy. Although the risk for aortopathy is significant, not all BAV patients experience (acute) aortic complications in their lifespan, highlighting the complexity of the pathogenetic process. Recent studies have focused on the embryonic development of semilunar valves and the ascending aortic wall. Their findings highlight that a defect in the embryogenesis could not only explain the development of a malformed aortic valve but also the increased risk for ascending aorta and arch pathology. This review presents an overview of the normal and abnormal development of the aortic valve and the aortic wall: a common defect in early embryogenesis causes the development of a BAV and associated aortopathy.

19.
Eur Heart J Cardiovasc Imaging ; 23(3): 412-422, 2022 02 22.
Article in English | MEDLINE | ID: mdl-33585887

ABSTRACT

AIMS: The Leiden Convention coronary coding system structures the large variety of coronary anatomical patterns; isolated and in congenital heart disease. It is widely used by surgeons but not by cardiologists as the system uses a surgeons' cranial view. Since thoracic surgeons and cardiologists work closely together, a coronary coding system practical for both disciplines is mandatory. To this purpose, the 'surgical' coronary coding system was adapted to an 'imaging' system, extending its applicability to different cardiac imaging techniques. METHODS AND RESULTS: The physician takes place in the non-facing sinus of the aortic valve, oriented with the back towards the pulmonary valve, looking outward from the sinus. From this position, the right-hand sinus is sinus 1, and the left-hand sinus is sinus 2. Next, a clockwise rotation is adopted starting at sinus 1 and the encountered coronary branches described. Annotation of the normal anatomical pattern is 1R-2LCx, corresponding to the 'surgical' coding system. The 'imaging' coding system was made applicable for Computed Tomography (CT), Magnetic Resonance Imaging (MRI), echocardiography, and coronary angiography, thus facilitating interdisciplinary use. To assess applicability in daily clinical practice, images from different imaging modalities were annotated by cardiologists and cardiology residents and results scored. The average score upon evaluation was 87.5%, with the highest scores for CT and MRI images (average 90%). CONCLUSION: The imaging Leiden Convention is a coronary coding system that unifies the annotation of coronary anatomy for thoracic surgeons, cardiologists, and radiologists. Validation of the coding system shows it can be easily and reliably applied in clinical practice.


Subject(s)
Echocardiography , Heart Defects, Congenital , Coronary Angiography/methods , Humans , Magnetic Resonance Imaging , Tomography, X-Ray Computed
20.
J Cell Mol Med ; 15(12): 2675-83, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21251220

ABSTRACT

The myocardium of the developing heart tube is covered by epicardium. These epicardial cells undergo a process of epithelial-to-mesenchymal transformation (EMT) and develop into epicardium-derived cells (EPDCs). The ingrowing EPDCs differentiate into several celltypes of which the cardiac fibroblasts form the main group. Disturbance of EMT of the epicardium leads to serious hypoplasia of the myocardium, abnormal coronary artery differentiation and Purkinje fibre paucity. Interestingly, the electrophysiological properties of epicardial cells and whether EMT influences electrical conductivity of epicardial cells is not yet known. We studied the electrophysiological aspects of epicardial cells before and after EMT in a dedicated in vitro model, using micro-electrode arrays to investigate electrical conduction across epicardial cells. Therefore, human adult epicardial cells were placed between two neonatal rat cardiomyocyte populations. Before EMT the epicardial cells have a cobblestone (epithelium-like) phenotype that was confirmed by staining for the cell-adhesion molecule ß-catenin. After spontaneous EMT in vitro the EPDCs acquired a spindle-shaped morphology confirmed by vimentin staining. When comparing both types we observed that the electrical conduction is influenced by EMT, resulting in significantly reduced conductivity of spindle-shaped EPDCs, associated with a conduction block. Furthermore, the expression of both gap junction (connexins 40, Cx43 and Cx45) and ion channel proteins (SCN5a, CACNA1C and Kir2.1) was down-regulated after EMT. This study shows for the first time the conduction differences between epicardial cells before and after EMT. These differences may be of relevance for the role of EPDCs in cardiac development, and in EMT-related cardiac dysfunction.


Subject(s)
Cell Differentiation , Electric Conductivity , Epithelial-Mesenchymal Transition/physiology , Pericardium/cytology , Pericardium/metabolism , Animals , Blotting, Western , Cells, Cultured , Humans , Male , Microscopy, Fluorescence , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL