Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters








Publication year range
1.
Development ; 151(20)2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39012059

ABSTRACT

Regeneration is the remarkable phenomenon through which an organism can regrow lost or damaged parts with fully functional replacements, including complex anatomical structures, such as limbs. In 2019, Development launched its 'Model systems for regeneration' collection, a series of articles introducing some of the most popular model organisms for studying regeneration in vivo. To expand this topic further, this Perspective conveys the voices of five expert biologists from the field of regenerative biology, each of whom showcases some less well-known, but equally extraordinary, species for studying regeneration.


Subject(s)
Regeneration , Animals , Humans , Extremities/physiology , Models, Biological , Regeneration/physiology
2.
J Am Heart Assoc ; 11(7): e023348, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35343246

ABSTRACT

Background Binding of Slit ligands to their Robo receptors regulates signaling pathways that are important for heart development. Genetic variants in ROBO1and ROBO4 have been linked to congenital heart defects in humans. These defects are recapitulated in mouse models with ubiquitous deletions of the Slit ligands or Robo receptors and include additional heart defects not currently linked to SLIT or ROBO mutations in humans. Given the broad expression patterns of these genes, the question remains open which tissue-specific ligand-receptor interactions are important for the correct development of different cardiac structures. Methods and Results We used tissue-specific knockout mouse models of Robo1/Robo2, Robo4, Slit2 andSlit3 and scored cardiac developmental defects in perinatal mice. Knockout of Robo2 in either the whole heart, endocardium and its derivatives, or the neural crest in ubiquitous Robo1 knockout background resulted in ventricular septal defects. Neural crest-specific removal of Robo2 in Robo1 knockouts showed fully penetrant bicuspid aortic valves (BAV). Endocardial knock-out of either Slit2or Robo4 caused low penetrant BAV. In contrast, endocardial knockout of Slit3 using a newly generated line resulted in fully penetrant BAV, while removal from smooth muscle cells also resulted in BAV. Caval vein and diaphragm defects observed in ubiquitous Slit3 mutants were recapitulated in the tissue-specific knockouts. Conclusions Our data will help understand defects observed in patients with variants in ROBO1 and ROBO4. The results strongly indicate interaction between endocardial Slit3and neural crest Robo2 in the development of BAV, highlighting the need for further studies of this connection.


Subject(s)
Nerve Tissue Proteins , Receptors, Immunologic , Animals , Diaphragm/metabolism , Female , Heart , Humans , Membrane Proteins , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pregnancy , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
3.
Cells ; 11(4)2022 02 10.
Article in English | MEDLINE | ID: mdl-35203259

ABSTRACT

Advances in sequencing and assembly technology have led to the creation of genome assemblies for a wide variety of non-model organisms. The rapid production and proliferation of updated, novel assembly versions can create vexing problems for researchers when multiple-genome assembly versions are available at once, requiring researchers to work with more than one reference genome. Multiple-genome assemblies are especially problematic for researchers studying the genetic makeup of individual cells, as single-cell RNA sequencing (scRNAseq) requires sequenced reads to be mapped and aligned to a single reference genome. Using the Astyanax mexicanus, this study highlights how the interpretation of a single-cell dataset from the same sample changes when aligned to its two different available genome assemblies. We found that the number of cells and expressed genes detected were drastically different when aligning to the different assemblies. When the genome assemblies were used in isolation with their respective annotations, cell-type identification was confounded, as some classic cell-type markers were assembly-specific, whilst other genes showed differential patterns of expression between the two assemblies. To overcome the problems posed by multiple-genome assemblies, we propose that researchers align to each available assembly and then integrate the resultant datasets to produce a final dataset in which all genome alignments can be used simultaneously. We found that this approach increased the accuracy of cell-type identification and maximised the amount of data that could be extracted from our single-cell sample by capturing all possible cells and transcripts. As scRNAseq becomes more widely available, it is imperative that the single-cell community is aware of how genome assembly alignment can alter single-cell data and their interpretation, especially when reviewing studies on non-model organisms.


Subject(s)
Genome , Base Sequence , Genome/genetics , Sequence Analysis, DNA/methods , Sequence Analysis, RNA , Exome Sequencing
4.
Nat Commun ; 12(1): 1447, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664263

ABSTRACT

Identifying the genetic factors that underlie complex traits is central to understanding the mechanistic underpinnings of evolution. Cave-dwelling Astyanax mexicanus populations are well adapted to subterranean life and many populations appear to have evolved troglomorphic traits independently, while the surface-dwelling populations can be used as a proxy for the ancestral form. Here we present a high-resolution, chromosome-level surface fish genome, enabling the first genome-wide comparison between surface fish and cavefish populations. Using this resource, we performed quantitative trait locus (QTL) mapping analyses and found new candidate genes for eye loss such as dusp26. We used CRISPR gene editing in A. mexicanus to confirm the essential role of a gene within an eye size QTL, rx3, in eye formation. We also generated the first genome-wide evaluation of deletion variability across cavefish populations to gain insight into this potential source of cave adaptation. The surface fish genome reference now provides a more complete resource for comparative, functional and genetic studies of drastic trait differences within a species.


Subject(s)
Adaptation, Physiological/genetics , Characidae/embryology , Characidae/genetics , Eye/embryology , Multifactorial Inheritance/genetics , Animals , Biological Evolution , Caves , Chromosome Mapping , Evolution, Molecular , Gene Editing , Genome/genetics , Homeodomain Proteins/genetics , Mitogen-Activated Protein Kinase Phosphatases/genetics , Quantitative Trait Loci/genetics
5.
J Cardiovasc Dev Dis ; 8(1)2021 Jan 16.
Article in English | MEDLINE | ID: mdl-33467137

ABSTRACT

The adult human heart cannot repair itself after injury and, instead, forms a permanent fibrotic scar that impairs cardiac function and can lead to incurable heart failure. The zebrafish, amongst other organisms, has been extensively studied for its innate capacity to repair its heart after injury. Understanding the signals that govern successful regeneration in models such as the zebrafish will lead to the development of effective therapies that can stimulate endogenous repair in humans. To date, many studies have investigated cardiac regeneration using a reverse genetics candidate gene approach. However, this approach is limited in its ability to unbiasedly identify novel genes and signalling pathways that are essential to successful regeneration. In contrast, drawing comparisons between different models of regeneration enables unbiased screens to be performed, identifying signals that have not previously been linked to regeneration. Here, we will review in detail what has been learnt from the comparative approach, highlighting the techniques used and how these studies have influenced the field. We will also discuss what further comparisons would enhance our knowledge of successful regeneration and scarring. Finally, we focus on the Astyanax mexicanus, an intraspecies comparative fish model that holds great promise for revealing the secrets of the regenerating heart.

6.
Proc Natl Acad Sci U S A ; 117(31): 18617-18626, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32675240

ABSTRACT

Genome-wide association studies have identified noncoding variants near TBX3 that are associated with PR interval and QRS duration, suggesting that subtle changes in TBX3 expression affect atrioventricular conduction system function. To explore whether and to what extent the atrioventricular conduction system is affected by Tbx3 dose reduction, we first characterized electrophysiological properties and morphology of heterozygous Tbx3 mutant (Tbx3+/-) mouse hearts. We found PR interval shortening and prolonged QRS duration, as well as atrioventricular bundle hypoplasia after birth in heterozygous mice. The atrioventricular node size was unaffected. Transcriptomic analysis of atrioventricular nodes isolated by laser capture microdissection revealed hundreds of deregulated genes in Tbx3+/- mutants. Notably, Tbx3+/- atrioventricular nodes showed increased expression of working myocardial gene programs (mitochondrial and metabolic processes, muscle contractility) and reduced expression of pacemaker gene programs (neuronal, Wnt signaling, calcium/ion channel activity). By integrating chromatin accessibility profiles (ATAC sequencing) of atrioventricular tissue and other epigenetic data, we identified Tbx3-dependent atrioventricular regulatory DNA elements (REs) on a genome-wide scale. We used transgenic reporter assays to determine the functionality of candidate REs near Ryr2, an up-regulated chamber-enriched gene, and in Cacna1g, a down-regulated conduction system-specific gene. Using genome editing to delete candidate REs, we showed that a strong intronic bipartite RE selectively governs Cacna1g expression in the conduction system in vivo. Our data provide insights into the multifactorial Tbx3-dependent transcriptional network that regulates the structure and function of the cardiac conduction system, which may underlie the differences in PR duration and QRS interval between individuals carrying variants in the TBX3 locus.


Subject(s)
Atrioventricular Node , T-Box Domain Proteins , Transcriptome/genetics , Animals , Arrhythmias, Cardiac , Atrioventricular Node/metabolism , Atrioventricular Node/physiology , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
7.
Development ; 147(8)2020 04 27.
Article in English | MEDLINE | ID: mdl-32341028

ABSTRACT

Runx1 is a transcription factor that plays a key role in determining the proliferative and differential state of multiple cell types, during both development and adulthood. Here, we report how Runx1 is specifically upregulated at the injury site during zebrafish heart regeneration, and that absence of runx1 results in increased myocardial survival and proliferation, and overall heart regeneration, accompanied by decreased fibrosis. Using single cell sequencing, we found that the wild-type injury site consists of Runx1-positive endocardial cells and thrombocytes that induce expression of smooth muscle and collagen genes. Both these populations cannot be identified in runx1 mutant wounds that contain less collagen and fibrin. The reduction in fibrin in the mutant is further explained by reduced myofibroblast formation and upregulation of components of the fibrin degradation pathway, including plasminogen receptor annexin 2A as well as downregulation of plasminogen activator inhibitor serpine1 in myocardium and endocardium, resulting in increased levels of plasminogen. Our findings suggest that Runx1 controls the regenerative response of multiple cardiac cell types and that targeting Runx1 is a novel therapeutic strategy for inducing endogenous heart repair.


Subject(s)
Cicatrix/pathology , Core Binding Factor Alpha 2 Subunit/metabolism , Heart/physiopathology , Myocardium/pathology , Regeneration , Zebrafish Proteins/metabolism , Zebrafish/physiology , Animals , Annexin A2/metabolism , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/genetics , Endocardium/pathology , Gene Expression Regulation, Developmental , Muscle, Smooth/metabolism , Mutation/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Myosin Heavy Chains/metabolism , Up-Regulation/genetics , Zebrafish Proteins/genetics
8.
Cell Rep ; 25(8): 1997-2007.e7, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30462998

ABSTRACT

Although Astyanax mexicanus surface fish regenerate their hearts after injury, their Pachón cave-dwelling counterparts cannot and, instead, form a permanent fibrotic scar, similar to the human heart. Myocardial proliferation peaks at similar levels in both surface fish and Pachón 1 week after injury. However, in Pachón, this peak coincides with a strong scarring and immune response, and ultimately, cavefish cardiomyocytes fail to replace the scar. We identified lrrc10 to be upregulated in surface fish compared with Pachón after injury. Similar to cavefish, knockout of lrrc10 in zebrafish impairs heart regeneration without affecting wound cardiomyocyte proliferation. Furthermore, using quantitative trait locus (QTL) analysis, we have linked the degree of heart regeneration to three loci in the genome, identifying candidate genes fundamental to the difference between scarring and regeneration. Our study provides evidence that successful heart regeneration entails a delicate interplay between cardiomyocyte proliferation and scarring.


Subject(s)
Characidae/physiology , Heart/physiology , Regeneration/physiology , Animals , Cell Proliferation , Characidae/genetics , Kinetics , Mutation/genetics , Myocardium/cytology , Myocytes, Cardiac/cytology , Quantitative Trait Loci/genetics , Up-Regulation , Wound Healing , Zebrafish/physiology , Zebrafish Proteins/metabolism
9.
Development ; 145(17)2018 09 03.
Article in English | MEDLINE | ID: mdl-30042181

ABSTRACT

A small network of spontaneously active Tbx3+ cardiomyocytes forms the cardiac conduction system (CCS) in adults. Understanding the origin and mechanism of development of the CCS network are important steps towards disease modeling and the development of biological pacemakers to treat arrhythmias. We found that Tbx3 expression in the embryonic mouse heart is associated with automaticity. Genetic inducible fate mapping revealed that Tbx3+ cells in the early heart tube are fated to form the definitive CCS components, except the Purkinje fiber network. At mid-fetal stages, contribution of Tbx3+ cells was restricted to the definitive CCS. We identified a Tbx3+ population in the outflow tract of the early heart tube that formed the atrioventricular bundle. Whereas Tbx3+ cardiomyocytes also contributed to the adjacent Gja5+ atrial and ventricular chamber myocardium, embryonic Gja5+ chamber cardiomyocytes did not contribute to the Tbx3+ sinus node or to atrioventricular ring bundles. In conclusion, the CCS is established by progressive fate restriction of a Tbx3+ cell population in the early developing heart, which implicates Tbx3 as a useful tool for developing strategies to study and treat CCS diseases.


Subject(s)
Bundle of His/embryology , Myocytes, Cardiac/metabolism , T-Box Domain Proteins/metabolism , Animals , Bundle of His/metabolism , Connexins/metabolism , Embryo Culture Techniques , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/cytology , Organogenesis/physiology , T-Box Domain Proteins/genetics , Gap Junction alpha-5 Protein
10.
Dev Biol ; 441(2): 272-284, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29940142

ABSTRACT

Regulation of heart size and shape is one of the least understood processes in developmental biology. We have for the first time analysed the hearts of Astyanax mexicanus and identified several differences in heart morphology between the surface (epigean morph) and cave-dwelling (troglomorph) morphs. Examination of the adult revealed that the troglomorph possesses a smaller heart with a rounder ventricle in comparison to the epigean morph. The size differences identified appear to arise early in development, as early as 24 h post-fertilisation (hpf), while shape differences begin to appear at 2 days post-fertilisation. The heart of the first-generation cross between the cave-dwelling and river-dwelling morph shows uncoupling of different phenotypes observed in the parental populations and indicates that the cardiac differences have become embedded in the genome during evolution. The differences in heart morphology are accompanied by functional changes between the two morphs, with the cave-dwelling morph exhibiting a slower heart rate than the river-dwelling morph. The identification of morphological and functional differences in the A. mexicanus heart could allow us to gain more insight into how such parameters are regulated during cardiac development, with potential relevance to cardiac pathologies in humans.


Subject(s)
Characiformes , Crosses, Genetic , Evolution, Molecular , Genome/physiology , Heart Rate/physiology , Heart/embryology , Animals , Characiformes/embryology , Characiformes/genetics , Humans , Organ Size
11.
Cardiovasc Res ; 114(6): 794-804, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29538649

ABSTRACT

The Slit ligands and their Robo receptors are well-known for their roles during axon guidance in the central nervous system but are still relatively unknown in the cardiac field. However, data from different animal models suggest a broad involvement of the pathway in many aspects of heart development, from cardiac cell migration and alignment, lumen formation, chamber formation, to the formation of the ventricular septum, semilunar and atrioventricular valves, caval veins, and pericardium. Absence of one or more of the genes in the pathway results in defects ranging from bicuspid aortic valves to ventricular septal defects and abnormal venous connections to the heart. Congenital heart defects are the most common congenital malformations found in life new-born babies and progress in methods for large scale human genetic testing has significantly enhanced the identification of new causative genes involved in human congenital heart disease. Recently, loss of function variants in ROBO1 have also been linked to ventricular septal defects and tetralogy of Fallot in patients. Here, we will give an overview of the role of the Slit-Robo signalling pathway in Drosophila, zebrafish, and mouse heart development. The extent of these data warrant further attention on the SLIT-ROBO signalling pathway as a candidate for an array of human congenital heart defects.


Subject(s)
Glycoproteins/metabolism , Heart Defects, Congenital/metabolism , Heart/embryology , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Heart Defects, Congenital/embryology , Humans , Ligands , Morphogenesis , Roundabout Proteins
12.
J Med Genet ; 54(12): 825-829, 2017 12.
Article in English | MEDLINE | ID: mdl-28592524

ABSTRACT

BACKGROUND: Congenital heart disease (CHD) is a common birth defect affecting approximately 1% of newborns. Great progress has been made in elucidating the genetic aetiology of CHD with advances in genomic technology, which we leveraged in recovering a new pathway affecting heart development in humans previously known to affect heart development in an animal model. METHODS: Four hundred and sixteen individuals from Thailand and the USA diagnosed with CHD and/or congenital diaphragmatic hernia were evaluated with chromosomal microarray and whole exome sequencing. The DECIPHER Consortium and medical literature were searched for additional patients. Murine hearts from ENU-induced mouse mutants and transgenic mice were evaluated using both episcopic confocal histopathology and troponin I stained sections. RESULTS: Loss of function ROBO1 variants were identified in three families; each proband had a ventricular septal defect, and one proband had tetralogy of Fallot. Additionally, a microdeletion in an individual with CHD was found in the medical literature. Mouse models showed perturbation of the Slit-Robo signalling pathway, causing septation and outflow tract defects and craniofacial anomalies. Two probands had variable facial features consistent with the mouse model. CONCLUSION: Our findings identify Slit-Robo as a significant pathway in human heart development and CHD.


Subject(s)
Heart Septal Defects/diagnosis , Heart Septal Defects/genetics , Loss of Function Mutation , Nerve Tissue Proteins/genetics , Phenotype , Receptors, Immunologic/genetics , Tetralogy of Fallot/diagnosis , Tetralogy of Fallot/genetics , Animals , Child , DNA Copy Number Variations , Disease Models, Animal , Female , Genetic Association Studies , Humans , Infant , Male , Mice , Polymorphism, Single Nucleotide , Roundabout Proteins
13.
Cardiovasc Res ; 106(1): 55-66, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25691540

ABSTRACT

AIMS: The mesenchymal cushions lining the early embryonic heart undergo complex remodelling to form the membranous ventricular septum as well as the atrioventricular and semilunar valves in later life. Disruption of this process underlies the most common congenital heart defects. Here, we identified a novel role for Slit-Robo signalling in the development of the murine membranous ventricular septum and cardiac valves. METHODS AND RESULTS: Expression of Robo1 and Robo2 receptors and their ligands, Slit2 and Slit3, was present in or adjacent to all cardiac cushions/valves. Loss of Robo1 or both Robo1 and Robo2 resulted in membranous ventricular septum defects at birth, a defect also found in Slit3, but not in Slit2 mutants. Additionally, Robo1;Robo2 double mutants showed thickened immature semilunar and atrioventricular valves as well as highly penetrant bicuspid aortic valves. Slit2 mutants recapitulated the semilunar phenotype, whereas Slit3 mutants displayed thickened atrioventricular valves. Bicuspid aortic cushions were already observed at E12.5 in the Robo1;Robo2 double mutants. Expression of Notch- and downstream Hey and Hes genes was down-regulated in Robo1 mutants, suggesting that reduced Notch signalling in mice lacking Robo might underlie the defects. Luciferase assays confirmed regulation of Notch signalling by Robo. CONCLUSION: Cardiac defects in mutants for Robo or Slit range from membranous ventricular septum defects to bicuspid aortic valves. These ligands and receptors have unique functions during development of specific cardiac cushion derivatives, and the Slit-Robo signalling pathway likely enforces its role by regulating Notch signalling, making these mutants a valuable new model to study cardiac valve formation.


Subject(s)
Aortic Valve/abnormalities , Heart Defects, Congenital/genetics , Heart Valve Diseases/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, Immunologic/genetics , Signal Transduction/genetics , Animals , Aortic Valve/physiopathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Bicuspid Aortic Valve Disease , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Disease Models, Animal , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Heart Defects, Congenital/physiopathology , Heart Valve Diseases/physiopathology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/physiology , Receptors, Immunologic/physiology , Receptors, Notch/genetics , Receptors, Notch/physiology , Signal Transduction/physiology , Transcription Factor HES-1 , Ventricular Septum/pathology , Roundabout Proteins
14.
J Neurosci ; 34(16): 5717-31, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741061

ABSTRACT

The elaborate cytoarchitecture of the mammalian neocortex requires the timely production of its constituent pyramidal neurons and interneurons and their disposition in appropriate layers. Numerous chemotropic factors present in the forebrain throughout cortical development play important roles in the orchestration of these events. The Roundabout (Robo) family of receptors and their ligands, the Slit proteins, are expressed in the developing forebrain, and are known to play important roles in the generation and migration of cortical interneurons. However, few studies have investigated their function(s) in the development of pyramidal cells. Here, we observed expression of Robo1 and Slit genes (Slit1, Slit2) in cells lining the telencephalic ventricles, and found significant increases in progenitor cells (basal and apical) at embryonic day (E)12.5 and E14.5 in the developing cortex of Robo1(-/-), Slit1(-/-), and Slit1(-/-)/Slit2(-/-), but not in mice lacking the other Robo or Slit genes. Using layer-specific markers, we found that both early- and late-born pyramidal neuron populations were significantly increased in the cortices of Robo1(-/-) mice at the end of corticogenesis (E18.5). The excess number of cortical pyramidal neurons generated prenatally appears to die in early postnatal life. The observed increase in pyramidal neurons was due to prolonged proliferative activity of their progenitors and not due to changes in cell cycle events. This finding, confirmed by in utero electroporation with Robo1 short hairpin RNA (shRNA) or control constructs into progenitors along the ventricular zone as well as in dissociated cortical cell cultures, points to a novel role for Robo1 in regulating the proliferation and generation of pyramidal neurons.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Developmental/genetics , Neocortex , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurons/physiology , Receptors, Immunologic/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cerebral Ventricles/cytology , Cerebral Ventricles/embryology , Cerebral Ventricles/growth & development , Embryo, Mammalian , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/cytology , Neocortex/embryology , Neocortex/growth & development , Nerve Tissue Proteins/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/genetics , Roundabout Proteins
15.
Circ Res ; 112(3): 465-75, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23255421

ABSTRACT

RATIONALE: The Slit-Roundabout (Robo) signaling pathway has pleiotropic functions during Drosophila heart development. However, its role in mammalian heart development is largely unknown. OBJECTIVE: To analyze the role of Slit-Robo signaling in the formation of the pericardium and the systemic venous return in the murine heart. METHODS AND RESULTS: Expression of genes encoding Robo1 and Robo2 receptors and their ligands Slit2 and Slit3 was found in or around the systemic venous return and pericardium during development. Analysis of embryos lacking Robo1 revealed partial absence of the pericardium, whereas Robo1/2 double mutants additionally showed severely reduced sinus horn myocardium, hypoplastic caval veins, and a persistent left inferior caval vein. Mice lacking Slit3 recapitulated the defects in the myocardialization, alignment, and morphology of the caval veins. Ligand binding assays confirmed Slit3 as the preferred ligand for the Robo1 receptor, whereas Slit2 showed preference for Robo2. Sinus node development was mostly unaffected in all mutants. In addition, we show absence of cross-regulation with previously identified regulators Tbx18 and Wt1. We provide evidence that pericardial defects are created by abnormal localization of the caval veins combined with ectopic pericardial cavity formation. Local increase in neural crest cell death and impaired neural crest adhesive and migratory properties underlie the ectopic pericardium formation. CONCLUSIONS: A novel Slit-Robo signaling pathway is involved in the development of the pericardium, the sinus horn myocardium, and the alignment of the caval veins. Reduced Slit3 binding in the absence of Robo1, causing impaired cardiac neural crest survival, adhesion, and migration, underlies the pericardial defects.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Pericardium/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Venae Cavae/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Movement , Gene Expression Regulation, Developmental , Gestational Age , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Crest/abnormalities , Neural Crest/metabolism , Pericardium/abnormalities , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Sinoatrial Node/abnormalities , Sinoatrial Node/metabolism , T-Box Domain Proteins/metabolism , Tissue Culture Techniques , Venae Cavae/abnormalities , WT1 Proteins/metabolism , Roundabout Proteins
16.
PLoS One ; 7(12): e52685, 2012.
Article in English | MEDLINE | ID: mdl-23285148

ABSTRACT

NKX2-5 is a homeodomain-containing transcription factor implied in both heart and thyroid development. Numerous mutations in NKX2-5 have been reported in individuals with congenital heart disease (CHD), but recently a select few have been associated with thyroid dysgenesis, among which the p.A119S variation. We sequenced NKX2-5 in 303 sporadic CHD patients and 38 families with at least two individuals with CHD. The p.A119S variation was identified in two unrelated patients: one was found in the proband of a family with four affected individuals with CHD and the other in a sporadic CHD patient. Clinical evaluation of heart and thyroid showed that the mutation did not segregate with CHD in the familial case, nor did any of the seven mutation carriers have thyroid abnormalities. We tested the functional consequences of the p.A119S variation in a cellular context by performing transactivation assays with promoters relevant for both heart and thyroid development in rat heart derived H10 cells and HELA cells. There was no difference between wildtype NKX2-5 and p.A119S NKX2-5 in activation of the investigated promoters in both cell lines. Additionally, we reviewed the current literature on the topic, showing that there is no clear evidence for a major pathogenic role of NKX2-5 mutations in thyroid dysgenesis. In conclusion, our study demonstrates that p.A119S does not cause CHD or TD and that it is a rare variation that behaves equal to wildtype NKX2-5. Furthermore, given the wealth of published evidence, we suggest that NKX2-5 mutations do not play a major pathogenic role in thyroid dysgenesis, and that genetic testing of NKX2-5 in TD is not warranted.


Subject(s)
Homeodomain Proteins/genetics , Mutation , Thyroid Dysgenesis/genetics , Transcription Factors/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Child , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Humans , Male , Middle Aged , Pedigree , Phenotype , Promoter Regions, Genetic , Protein Transport , Sequence Alignment , Transcription Factors/metabolism , Transcriptional Activation , Young Adult
17.
Circ Res ; 107(6): 728-36, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20671237

ABSTRACT

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.


Subject(s)
Heart Conduction System/embryology , Heart Conduction System/growth & development , Image Processing, Computer-Assisted , Animals , Atrioventricular Node/anatomy & histology , Atrioventricular Node/embryology , Atrioventricular Node/growth & development , Female , Heart/anatomy & histology , Heart/embryology , Heart/growth & development , Heart Conduction System/anatomy & histology , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional , Mice , Mice, Transgenic , Pregnancy
18.
Circ Res ; 106(7): 1212-20, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20185795

ABSTRACT

RATIONALE: The cardiac venous pole is a common focus of congenital malformations and atrial arrhythmias, yet little is known about the cellular and molecular mechanisms that regulate its development. The systemic venous return myocardium (sinus node and sinus horns) forms only late in cardiogenesis from a pool of pericardial mesenchymal precursor cells. OBJECTIVE: To analyze the cellular and molecular mechanisms directing the formation of the fetal sinus horns. METHODS AND RESULTS: We analyzed embryos deficient for the Wt1 (Wilms tumor 1) gene and observed a failure to form myocardialized sinus horns. Instead, the cardinal veins become embedded laterally in the pleuropericardial membranes that remain tethered to the lateral body wall by the persisting subcoelomic mesenchyme, a finding that correlates with decreased apoptosis in this region. We show by expression analysis and lineage tracing studies that Wt1 is expressed in the subcoelomic mesenchyme surrounding the cardinal veins, but that this Wt1-positive mesenchyme does not contribute cells to the sinus horn myocardium. Expression of the Raldh2 (aldehyde dehydrogenase family 1, subfamily A2) gene was lost from this mesenchyme in Wt1(-/-) embryos. Phenotypic analysis of Raldh2 mutant mice rescued from early cardiac defects by retinoic acid food supply revealed defects of the venous pole and pericardium highly similar to those of Wt1(-/-) mice. CONCLUSIONS: Pericardium and sinus horn formation are coupled and depend on the expansion and correct temporal release of pleuropericardial membranes from the underlying subcoelomic mesenchyme. Wt1 and downstream Raldh2/retinoic acid signaling are crucial regulators of this process. Thus, our results provide novel insight into the genetic and cellular pathways regulating the posterior extension of the mammalian heart and the formation of its coelomic lining.


Subject(s)
Coronary Sinus/metabolism , Mesoderm/metabolism , Pericardium/metabolism , Pleura/metabolism , Signal Transduction , Sinoatrial Node/metabolism , Tretinoin/metabolism , WT1 Proteins/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Apoptosis , Cell Lineage , Coronary Sinus/embryology , Fetal Death , Gene Expression Regulation, Developmental , Genotype , Gestational Age , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Pericardium/embryology , Phenotype , Pleura/embryology , Signal Transduction/genetics , Sinoatrial Node/embryology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , WT1 Proteins/deficiency , WT1 Proteins/genetics
19.
Cardiovasc Res ; 87(1): 92-101, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20110338

ABSTRACT

AIMS: During development, the heart tube grows by differentiation of Isl1(+)/Nkx2-5(+) progenitors to the arterial and venous pole and dorsal mesocardium. However, after the establishment of the heart tube, Tbx18(+) progenitors were proposed to form the Tbx18(+)/Nkx2-5(-) sinus venosus and proepicardium. To elucidate the relationship between these contributions, we investigated the origin of the Tbx18(+) sinus venosus progenitor population in the cardiogenic mesoderm and its spatial and temporal relation to the second heart field during murine heart development. METHODS AND RESULTS: Explant culture revealed that the Tbx18(+) cell population has the potential to form Nkx2-5(-) sinus venosus myocardium. Three-dimensional reconstruction of expression patterns showed that during heart tube elongation, the Tbx18(+) progenitors remained spatially and temporally separate from the Isl1(+) second heart field, only overlapping with the Isl1(+) domain at the right lateral side of the inflow tract, where the sinus node developed. Consistently, genetic lineage analysis revealed that the Tbx18(+) descendants formed the sinus venosus myocardium, but did not contribute to the pulmonary vein myocardium that developed in the Isl1(+) second heart field. By means of DiI labelling and expression analysis, the origin of the sinus venosus progenitor population was traced to the lateral rim of splanchnic mesoderm that down-regulated Nkx2-5 expression approximately 2 days before its differentiation into sinus venosus myocardium. CONCLUSION: Our data indicate that the cardiogenic mesoderm contains an additional progenitor subpopulation that contributes to the sinus venosus myocardium. After patterning of the cardiogenic mesoderm, this progenitor population remains spatially separated and genetically distinctive from the second heart field subpopulation.


Subject(s)
Heart/embryology , Mesoderm/metabolism , Myocytes, Cardiac/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Gestational Age , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins , Lac Operon/genetics , Mesoderm/cytology , Mice , Mice, Transgenic , Morphogenesis , Proteins/genetics , Pulmonary Veins/embryology , Pulmonary Veins/metabolism , RNA, Untranslated , Recombinant Fusion Proteins/metabolism , Sinoatrial Node/embryology , Sinoatrial Node/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tissue Culture Techniques , Transcription Factors/metabolism
20.
Heart Rhythm ; 6(12): 1818-24, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19726237

ABSTRACT

The myocardial sleeves of the systemic venous tributaries and the pulmonary veins are known to be common anatomic substrates for atrial fibrillation. Rapidly evolving evidence has shown that a substantial part of the paroxysmal variant of this abnormal rhythm has a familial heritage, and the number of genes found to be involved is increasing. One of the mechanisms underlying the condition is ectopic pacemaking activity. Knowledge of the normal embryological development of the atrial myocardium, in particular the myocardial sleeves clothing the systemic venous tributaries and the pulmonary veins at their junctions with the atrial chambers, may contribute to the understanding of the origins of such ectopic pacing. In this respect, it is now well established that the myocardial sleeves of the systemic venous tributaries have a distinct origin and program of gene expression when compared with the pulmonary venous myocardium. The myocardium clothing the pulmonary veins, however, is particularly susceptible to changes in the levels of gene expression, with the changes then favoring the presence of genes responsible for pacemaking. Only recently has interest developed in the genetic and heritable bases of atrial fibrillation, and much is still to be learned. Better understanding of both the developmental and genetic factors, nonetheless, will surely be helpful in the diagnosis, prevention, and treatment of this troublesome arrhythmia. With this in mind, therefore, we have reviewed the current knowledge concerning the initial development of the pulmonary venous myocardium, emphasizing its crucial differences from the systemic venous myocardium.


Subject(s)
Atrial Fibrillation/pathology , Heart Atria/pathology , Myocardium/pathology , Atrial Fibrillation/genetics , Genetic Variation , Heart Conduction System , Humans , Mutation , Phenotype , Pulmonary Veins/pathology , Sinoatrial Node
SELECTION OF CITATIONS
SEARCH DETAIL