Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Growth Factors ; 36(1-2): 58-68, 2018 04.
Article in English | MEDLINE | ID: mdl-30035654

ABSTRACT

Ryk is a member of the receptor tyrosine kinase (RTK) family of proteins that control and regulate cellular processes. It is distinguished by binding Wnt ligands and having no detectable intrinsic protein tyrosine kinase activity suggesting Ryk is a pseudokinase. Here, we show an essential role for Ryk in directing morphogenetic events required for normal cardiac development through the examination of Ryk-deficient mice. We employed vascular corrosion casting, vascular perfusion with contrast dye, and immunohistochemistry to characterize cardiovascular and pharyngeal defects in Ryk-/- embryos. Ryk-/- mice exhibit a variety of malformations of the heart and outflow tract that resemble human congenital heart defects. This included stenosis and interruption of the aortic arch, ventriculoarterial malalignment, ventricular septal defects and abnormal pharyngeal arch artery remodelling. This study therefore defines a key intersection between a subset of growth factor receptors involved in planar cell polarity signalling, the Wnt family and mammalian cardiovascular development.


Subject(s)
Heart Defects, Congenital/etiology , Pharynx/abnormalities , Receptor Protein-Tyrosine Kinases/physiology , Wnt Proteins/metabolism , Animals , Aorta, Thoracic/abnormalities , Female , Mice , Morphogenesis , Pregnancy
2.
J Mol Cell Cardiol ; 80: 1-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25536179

ABSTRACT

In utero interventions aimed at restoring left ventricular hemodynamic forces in fetuses with prenatally diagnosed hypoplastic left heart syndrome failed to stimulate ventricular myocardial growth during gestation, suggesting chamber growth during development may not rely upon fluid forces. We therefore hypothesized that ventricular hypertrophy during development may depend upon fundamental Ca(2+)-dependent growth pathways that function independent of hemodynamic forces. To test this hypothesis, zebrafish embryos were treated with inhibitors or activators of Ca(2+) signaling in the presence or absence of contraction during the period of chamber development. Abolishment of contractile function alone in the setting of preserved Ca(2+) signaling did not impair ventricular hypertrophy. In contrast, inhibition of L-type voltage-gated Ca(2+) influx abolished contraction and led to reduced ventricular hypertrophy, whereas increasing L-type voltage-gated Ca(2+) influx led to enhanced ventricular hypertrophy in either the presence or absence of contraction. Similarly, inhibition of the downstream Ca(2+)-sensitive phosphatase calcineurin, a known regulator of adult cardiac hypertrophy, led to reduced ventricular hypertrophy in the presence or absence of contraction, whereas hypertrophy was rescued in the absence of L-type voltage-gated Ca(2+) influx and contraction by expression of a constitutively active calcineurin. These data suggest that ventricular cardiomyocyte hypertrophy during chamber formation is dependent upon Ca(2+) signaling pathways that are unaffected by heart function or hemodynamic forces. Disruption of Ca(2+)-dependent hypertrophy during heart development may therefore represent one mechanism for impaired chamber formation that is not related to impaired blood flow.


Subject(s)
Calcium Signaling , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Myocardial Contraction , Regional Blood Flow , Animals , Animals, Genetically Modified , Calcineurin/metabolism , Calcium Channels, L-Type/metabolism , Cardiomegaly/genetics , Disease Models, Animal , Hemodynamics , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Hypoplastic Left Heart Syndrome/genetics , Hypoplastic Left Heart Syndrome/metabolism , Hypoplastic Left Heart Syndrome/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Zebrafish
3.
FASEB J ; 28(6): 2525-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24577120

ABSTRACT

Point mutations in the calcium-permeable TRPV4 ion channel have been identified as the cause of autosomal-dominant human motor neuropathies, arthropathies, and skeletal malformations of varying severity. The objective of this study was to determine the mechanism by which TRPV4 channelopathy mutations cause skeletal dysplasia. The human TRPV4(V620I) channelopathy mutation was transfected into primary porcine chondrocytes and caused significant (2.6-fold) up-regulation of follistatin (FST) expression levels. Pore altering mutations that prevent calcium influx through the channel prevented significant FST up-regulation (1.1-fold). We generated a mouse model of the TRPV4(V620I) mutation, and found significant skeletal deformities (e.g., shortening of tibiae and digits, similar to the human disease brachyolmia) and increases in Fst/TRPV4 mRNA levels (2.8-fold). FST was significantly up-regulated in primary chondrocytes transfected with 3 different dysplasia-causing TRPV4 mutations (2- to 2.3-fold), but was not affected by an arthropathy mutation (1.1-fold). Furthermore, FST-loaded microbeads decreased bone ossification in developing chick femora (6%) and tibiae (11%). FST gene and protein levels were also increased 4-fold in human chondrocytes from an individual natively expressing the TRPV4(T89I) mutation. Taken together, these data strongly support that up-regulation of FST in chondrocytes by skeletal dysplasia-inducing TRPV4 mutations contributes to disease pathogenesis.


Subject(s)
Bone Diseases, Developmental/embryology , Channelopathies/physiopathology , Follistatin/physiology , TRPV Cation Channels/genetics , Animals , Bone Diseases, Developmental/genetics , Chick Embryo , Chondrocytes/metabolism , Humans , Mice , Mutation , Osteochondrodysplasias , Osteogenesis/genetics , Swine , Up-Regulation
4.
J Clin Invest ; 123(4): 1638-46, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23549079

ABSTRACT

The identification of a gain-of-function mutation in CACNA1C as the cause of Timothy Syndrome (TS), a rare disorder characterized by cardiac arrhythmias and syndactyly, highlighted unexpected roles for the L-type voltage-gated Ca2+ channel CaV1.2 in nonexcitable cells. How abnormal Ca2+ influx through CaV1.2 underlies phenotypes such as the accompanying syndactyly or craniofacial abnormalities in the majority of affected individuals is not readily explained by established CaV1.2 roles. Here, we show that CaV1.2 is expressed in the first and second pharyngeal arches within the subset of cells that give rise to jaw primordia. Gain-of-function and loss-of-function studies in mouse, in concert with knockdown/rescue and pharmacological approaches in zebrafish, demonstrated that Ca2+ influx through CaV1.2 regulates jaw development. Cranial neural crest migration was unaffected by CaV1.2 knockdown, suggesting a role for CaV1.2 later in development. Focusing on the mandible, we observed that cellular hypertrophy and hyperplasia depended upon Ca2+ signals through CaV1.2, including those that activated the calcineurin signaling pathway. Together, these results provide new insights into the role of voltage-gated Ca2+ channels in nonexcitable cells during development.


Subject(s)
Calcium Channels, L-Type/physiology , Mandible/embryology , Zebrafish Proteins/physiology , Animals , Autistic Disorder , Branchial Region/embryology , Branchial Region/metabolism , Branchial Region/pathology , Calcineurin/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Calcium Signaling , Cell Movement , Cells, Cultured , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/metabolism , Gene Expression , Gene Knockdown Techniques , Heart/embryology , Humans , Hyperplasia/embryology , Hyperplasia/genetics , Hyperplasia/metabolism , Hypertrophy/embryology , Hypertrophy/genetics , Hypertrophy/metabolism , Long QT Syndrome/genetics , Mandible/metabolism , Mandible/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Morpholinos/genetics , Mutation, Missense , Neural Crest/metabolism , Stem Cells/metabolism , Stem Cells/physiology , Syndactyly/genetics , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
PLoS One ; 7(6): e38454, 2012.
Article in English | MEDLINE | ID: mdl-22715383

ABSTRACT

The phenotype of somatic cells has recently been found to be reversible. Direct reprogramming of one cell type into another has been achieved with transduction and over expression of exogenous defined transcription factors emphasizing their role in specifying cell fate. To discover early and novel endogenous transcription factors that may have a role in adult-derived stem cell acquisition of a cardiomyocyte phenotype, mesenchymal stem cells from human and mouse bone marrow and rat liver were co-cultured with neonatal cardiomyocytes as an in vitro cardiogenic microenvironment. Cell-cell communications develop between the two cell types as early as 24 hrs in co-culture and are required for elaboration of a myocardial phenotype in the stem cells 8-16 days later. These intercellular communications are associated with novel Ca(2+) oscillations in the stem cells that are synchronous with the Ca(2+) transients in adjacent cardiomyocytes and are detected in the stem cells as early as 24-48 hrs in co-culture. Early and significant up-regulation of Ca(2+)-dependent effectors, CAMTA1 and RCAN1 ensues before a myocardial program is activated. CAMTA1 loss-of-function minimizes the activation of the cardiac gene program in the stem cells. While the expression of RCAN1 suggests involvement of the well-characterized calcineurin-NFAT pathway as a response to a Ca(2+) signal, the CAMTA1 up-regulated expression as a response to such a signal in the stem cells was unknown. Cell-cell communications between the stem cells and adjacent cardiomyocytes induce Ca(2+) signals that activate a myocardial gene program in the stem cells via a novel and early Ca(2+)-dependent intermediate, up-regulation of CAMTA1.


Subject(s)
Adult Stem Cells/metabolism , Calcium Signaling/physiology , Calcium-Binding Proteins/metabolism , Cell Communication/physiology , Cell Differentiation/physiology , Myocytes, Cardiac/metabolism , Trans-Activators/metabolism , Adult , Adult Stem Cells/cytology , Animals , Calcium-Binding Proteins/genetics , Cell Line , Coculture Techniques , Humans , Mice , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Rats , Trans-Activators/genetics , Up-Regulation/physiology
6.
Development ; 138(11): 2389-98, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21558385

ABSTRACT

The secondary heart field is a conserved developmental domain in avian and mammalian embryos that contributes myocardium and smooth muscle to the definitive cardiac arterial pole. This field is part of the overall heart field and its myocardial component has been fate mapped from the epiblast to the heart in both mammals and birds. In this study we show that the population that gives rise to the arterial pole of the zebrafish can be traced from the epiblast, is a discrete part of the mesodermal heart field, and contributes myocardium after initial heart tube formation, giving rise to both smooth muscle and myocardium. We also show that Isl1, a transcription factor associated with undifferentiated cells in the secondary heart field in other species, is active in this field. Furthermore, Bmp signaling promotes myocardial differentiation from the arterial pole progenitor population, whereas inhibiting Smad1/5/8 phosphorylation leads to reduced myocardial differentiation with subsequent increased smooth muscle differentiation. Molecular pathways required for secondary heart field development are conserved in teleosts, as we demonstrate that the transcription factor Tbx1 and the Sonic hedgehog pathway are necessary for normal development of the zebrafish arterial pole.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Heart/embryology , Heart/growth & development , Myocardium/metabolism , Zebrafish/embryology , Animals , Cell Differentiation , Germ Layers , Hedgehog Proteins/metabolism , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins , Muscle Development , Muscle, Smooth/embryology , Myocardium/cytology , Phosphorylation , Signal Transduction , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , T-Box Domain Proteins/metabolism , Transcription Factors , Zebrafish/metabolism , Zebrafish Proteins/metabolism
7.
Dev Biol ; 354(1): 18-30, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21419761

ABSTRACT

Cardiac neural crest cells migrate into the pharyngeal arches where they support development of the pharyngeal arch arteries. The pharyngeal endoderm and ectoderm both express high levels of FGF8. We hypothesized that FGF8 is chemotactic for cardiac crest cells. To begin testing this hypothesis, cardiac crest was explanted for migration assays under various conditions. Cardiac neural crest cells migrated more in response to FGF8. Single cell tracing indicated that this was not due to proliferation and subsequent transwell assays showed that the cells migrate toward an FGF8 source. The migratory response was mediated by FGF receptors (FGFR) 1 and 3 and MAPK/ERK intracellular signaling. To test whether FGF8 is chemokinetic and/or chemotactic in vivo, dominant negative FGFR1 was electroporated into the premigratory cardiac neural crest. Cells expressing the dominant negative receptor migrated slower than normal cardiac neural crest cells and were prone to remain in the vicinity of the neural tube and die. Treating with the FGFR1 inhibitor, SU5402 or an FGFR3 function-blocking antibody also slowed neural crest migration. FGF8 over-signaling enhanced neural crest migration. Neural crest cells migrated to an FGF8-soaked bead placed dorsal to the pharynx. Finally, an FGF8 producing plasmid was electroporated into an ectopic site in the ventral pharyngeal endoderm. The FGF8 producing cells attracted a thick layer of mesenchymal cells. DiI labeling of the neural crest as well as quail-to-chick neural crest chimeras showed that neural crest cells migrated to and around the ectopic site of FGF8 expression. These results showing that FGF8 is chemotactic and chemokinetic for cardiac neural crest adds another dimension to understanding the relationship of FGF8 and cardiac neural crest in cardiovascular defects.


Subject(s)
Cell Movement/drug effects , Chemotaxis/drug effects , Fibroblast Growth Factor 8/pharmacology , Neural Crest/cytology , Animals , Apoptosis/drug effects , Butadienes/pharmacology , Cell Proliferation/drug effects , Chick Embryo , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart/embryology , Immunohistochemistry , In Situ Hybridization , Mesoderm/embryology , Mesoderm/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Myocardium/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Nitriles/pharmacology , Pharynx/embryology , Pharynx/metabolism , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
8.
Circ Res ; 107(12): 1428-44, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21148449

ABSTRACT

The myocardium of the heart is composed of multiple highly specialized myocardial lineages, including those of the ventricular and atrial myocardium, and the specialized conduction system. Specification and maturation of each of these lineages during heart development is a highly ordered, ongoing process involving multiple signaling pathways and their intersection with transcriptional regulatory networks. Here, we attempt to summarize and compare much of what we know about specification and maturation of myocardial lineages from studies in several different vertebrate model systems. To date, most research has focused on early specification, and although there is still more to learn about early specification, less is known about factors that promote subsequent maturation of myocardial lineages required to build the functioning adult heart.


Subject(s)
Cell Lineage , Myocardium/cytology , Myocytes, Cardiac/cytology , Animals , Heart/growth & development , Humans
9.
Cell Adh Migr ; 4(4): 609-21, 2010.
Article in English | MEDLINE | ID: mdl-20890117

ABSTRACT

Cardiac neural crest cells originate as part of the postotic caudal rhombencephalic neural crest stream. Ectomesenchymal cells in this stream migrate to the circumpharyngeal ridge and then into the caudal pharyngeal arches where they condense to form first a sheath and then the smooth muscle tunics of the persisting pharyngeal arch arteries. A subset of the cells continue migrating into the cardiac outflow tract where they will condense to form the aorticopulmonary septum. Cell signaling, extracellular matrix and cell-cell contacts are all critical for the initial migration, pauses, continued migration, and condensation of these cells. This review elucidates what is currently known about these factors.


Subject(s)
Heart/embryology , Neural Crest/cytology , Animals , Cell Communication , Cell Movement/genetics , Epithelial-Mesenchymal Transition , Extracellular Matrix/metabolism , Ganglia, Autonomic/embryology , Heart/innervation , Humans , Intercellular Junctions/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neural Crest/metabolism , Pharynx/blood supply , Pharynx/embryology , Pharynx/innervation
10.
Dev Biol ; 348(2): 167-76, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20920499

ABSTRACT

Sonic hedgehog signaling in the secondary heart field has a clear role in cardiac arterial pole development. In the absence of hedgehog signaling, proliferation is reduced in secondary heart field progenitors, and embryos predominantly develop pulmonary atresia. While it is expected that proliferation in the secondary heart field would be increased with elevated hedgehog signaling, this idea has never been tested. We hypothesized that up-regulating hedgehog signaling would increase secondary heart field proliferation, which would lead to arterial pole defects. In culture, secondary heart field explants proliferated up to 6-fold more in response to the hedgehog signaling agonist SAG, while myocardial differentiation and migration were unaffected. Treatment of chick embryos with SAG at HH14, just before the peak in secondary heart field proliferation, resulted unexpectedly in stenosis of both the aortic and pulmonary outlets. We examined proliferation in the secondary heart field and found that SAG-treated embryos exhibited a much milder increase in proliferation than was indicated by the in vitro experiments. To determine the source of other signaling factors that could modulate increased hedgehog signaling, we co-cultured secondary heart field explants with isolated pharyngeal endoderm or outflow tract and found that outflow tract co-cultures prevented SAG-induced proliferation. BMP2 is made and secreted by the outflow tract myocardium. To determine whether BMP signaling could prevent SAG-induced proliferation, we treated explants with SAG and BMP2 and found that BMP2 inhibited SAG-induced proliferation. In vivo, SAG-treated embryos showed up-regulated BMP2 expression and signaling. Together, these results indicate that BMP signaling from the outflow tract modulates hedgehog-induced proliferation in the secondary heart field.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Heart/embryology , Hedgehog Proteins/metabolism , Signal Transduction , Animals , Bone Morphogenetic Proteins/genetics , Cell Proliferation , Chick Embryo , Cyclohexylamines/pharmacology , Hedgehog Proteins/genetics , Myocardium/metabolism , Organogenesis , Thiophenes/pharmacology , Up-Regulation
11.
Development ; 137(18): 3001-11, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20702561

ABSTRACT

During heart development, a subpopulation of cells in the heart field maintains cardiac potential over several days of development and forms the myocardium and smooth muscle of the arterial pole. Using clonal and explant culture experiments, we show that these cells are a stem cell population that can differentiate into myocardium, smooth muscle and endothelial cells. The multipotent stem cells proliferate or differentiate into different cardiovascular cell fates through activation or inhibition of FGF and BMP signaling pathways. BMP promoted myocardial differentiation but not proliferation. FGF signaling promoted proliferation and induced smooth muscle differentiation, but inhibited myocardial differentiation. Blocking the Ras/Erk intracellular pathway promoted myocardial differentiation, while the PLCgamma and PI3K pathways regulated proliferation. In vivo, inhibition of both pathways resulted in predictable arterial pole defects. These studies suggest that myocardial differentiation of arterial pole progenitors requires BMP signaling combined with downregulation of the FGF/Ras/Erk pathway. The FGF pathway maintains the pool of proliferating stem cells and later promotes smooth muscle differentiation.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Cell Proliferation , Fibroblast Growth Factor 8/metabolism , MAP Kinase Signaling System , Stem Cells/cytology , Stem Cells/metabolism , Animals , Arteries/cytology , Arteries/growth & development , Arteries/metabolism , Body Patterning , Cell Lineage , Chick Embryo , Gene Expression Regulation, Developmental , Heart/embryology , Muscle, Smooth/cytology , Muscle, Smooth/embryology , Muscle, Smooth/metabolism , Myocardium/cytology , Myocardium/metabolism , Quail , Tissue Culture Techniques
12.
Congenit Heart Dis ; 5(3): 316-20, 2010.
Article in English | MEDLINE | ID: mdl-20576054

ABSTRACT

Aortic valve atresia with interruption of the aortic arch is an extremely rare anomaly; only eleven cases of this anomaly have been reported to date. In the absence of additional sources of blood flow to the ascending aorta, aortic valve atresia with interruption of the aortic arch is fatal. We present, to the best of our knowledge, the first case of a live birth with aortic valve atresia and interrupted left aortic arch (type B) without evidence of an aorticopulmonary communication or ductal supply to the native ascending aorta. Instead, blood flow to the native aortic root was derived from a persistent right embryonic dorsal aorta.


Subject(s)
Abnormalities, Multiple/physiopathology , Aorta, Thoracic/physiopathology , Aortic Valve/physiopathology , Heart Defects, Congenital/physiopathology , Hemodynamics , Vascular Malformations/physiopathology , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/surgery , Aorta, Thoracic/abnormalities , Aorta, Thoracic/surgery , Aortic Valve/abnormalities , Aortic Valve/surgery , Cardiac Surgical Procedures/adverse effects , Fatal Outcome , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/surgery , Humans , Live Birth , Tomography, X-Ray Computed , Treatment Outcome , Vascular Malformations/diagnosis , Vascular Malformations/surgery
14.
Dev Biol ; 336(2): 137-44, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19835857

ABSTRACT

Although de la Cruz and colleagues showed as early as 1977 that the outflow tract was added after the heart tube formed, the source of these secondarily added cells was not identified for nearly 25 years. In 2001, three pivotal publications described a secondary or anterior heart field that contributed to the developing outflow tract. This review details the history of the heart field, the discovery and continuing elucidation of the secondarily adding myocardial cells, and how the different populations identified in 2001 are related to the more recent lineage tracing studies that defined the first and second myocardial heart fields/lineages. Much recent work has focused on secondary heart field progenitors that give rise to the myocardium and smooth muscle at the definitive arterial pole. These progenitors are the last to be added to the arterial pole and are particularly susceptible to abnormal development, leading to conotruncal malformations in children. The major signaling pathways (Wnt, BMP, FGF8, Notch, and Shh) that control various aspects of secondary heart field progenitor behavior are discussed.


Subject(s)
Heart/embryology , Animals , Humans , Signal Transduction
15.
Dev Biol ; 335(2): 367-73, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19765571

ABSTRACT

Cardiac neural crest cells (CNCC) migrate into the caudal pharynx and arterial pole of the heart to form the outflow septum. Ablation of the CNCC results in arterial pole malalignment and failure of outflow septation, resulting in a common trunk overriding the right ventricle. Unlike preotic cranial crest, the postotic CNCC do not normally regenerate. We applied the hedgehog signaling inhibitor, cyclopamine (Cyc), to chick embryos after CNCC ablation and found normal heart development at day 9 suggesting that the CNCC population was reconstituted. We ablated the CNCC, and labeled the remaining neural tube with DiI/CSRE and applied cyclopamine. Cells migrated from the neural tube in the CNCC-ablated, cyclopamine-treated embryos but not in untreated CNCC-ablated embryos. The newly generated cells followed the CNCC migration pathways, expressed neural crest markers and supported normal heart development. Finally, we tested whether reducing hedgehog signaling caused redeployment of the dorsal-ventral axis of the injured neural tube, allowing generation of new neural crest-like cells. The dorsal neural tube marker, Pax7, was maintained 12 h after CNCC ablation with Cyc treatment but not in the CNCC-ablated alone. This disruption of dorsal-ventral neural patterning permits a new wave of migratory cardiac neural crest-like cells.


Subject(s)
Heart/embryology , Neural Crest/embryology , Neural Tube/embryology , Signal Transduction , Animals , Chick Embryo , Immunohistochemistry
17.
Circ Res ; 105(5): 431-41, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19628790

ABSTRACT

RATIONALE: The epicardium contributes to the majority of nonmyocardial cells in the adult heart. Recent studies have reported that the epicardium is derived from Nkx2.5-positive progenitors and can differentiate into cardiomyocytes. Not much is known about the relation between the myocardial and epicardial lineage during development, whereas insights into these embryonic mechanisms could facilitate the design of future regenerative strategies. OBJECTIVE: Acquiring insight into the signaling pathways involved in the lineage separation leading to the differentiation of myocardial and (pro)epicardial cells at the inflow of the developing heart. METHODS AND RESULTS: We made 3D reconstructions of Tbx18 gene expression patterns to give insight into the developing epicardium in relation to the developing myocardium. Next, using DiI tracing, we show that the (pro)epicardium separates from the same precursor pool as the inflow myocardium. In vitro, we show that this lineage separation is regulated by a crosstalk between bone morphogenetic protein (BMP) signaling and fibroblast growth factor (FGF) signaling. BMP signaling via Smad drives differentiation toward the myocardial lineage, which is inhibited by FGF signaling via mitogen-activated protein kinase kinase (Mek)1/2. Embryos exposed to recombinant FGF2 in vivo show enhanced epicardium formation, whereas a misbalance between FGF and BMP by Mek1/2 inhibition and BMP stimulation causes a developmental arrest of the epicardium and enhances myocardium formation at the inflow of the heart. CONCLUSION: Our data show that FGF signaling via Mek1/2 is dominant over BMP signaling via Smad and is required to separate the epicardial lineage from precardiac mesoderm. Consequently, myocardial differentiation requires BMP signaling via Smad and inhibition of FGF signaling at the level of Mek1/2. These findings are of clinical interest for the development of regeneration-based therapies for heart disease.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Lineage , Fibroblast Growth Factors/metabolism , Heart/embryology , Myocardium/metabolism , Pericardium/embryology , Pericardium/metabolism , Signal Transduction , Animals , Apoptosis , Bone Morphogenetic Protein 2/metabolism , Butadienes/pharmacology , Carbocyanines , Cell Differentiation , Cell Line , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Proliferation , Chick Embryo , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/metabolism , Fluorescent Dyes , Gene Expression Regulation, Developmental , Heart/drug effects , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism , Microscopy, Fluorescence , Nitriles/pharmacology , Pericardium/drug effects , Phenotype , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smad Proteins/metabolism , T-Box Domain Proteins/genetics
18.
Dev Biol ; 330(2): 305-17, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19361493

ABSTRACT

The Sonic hedgehog (Shh)-null mouse was initially described as a phenotypic mimic of Tetralogy of Fallot with pulmonary atresia (Washington Smoak, I., Byrd, N.A., Abu-Issa, R., Goddeeris, M.M., Anderson, R., Morris, J., Yamamura, K., Klingensmith, J., and Meyers, E.N. 2005. Sonic hedgehog is required for cardiac outflow tract and neural crest cell development. Dev. Biol. 283, 357-372.); however, subsequent reports describe only a single outflow tract, leaving the phenotype and its developmental mechanism unclear. We hypothesized that the phenotype that occurs in response to Shh knockdown is pulmonary atresia and is directly related to the abnormal development of the secondary heart field. We found that Shh was expressed by the pharyngeal endoderm adjacent to the secondary heart field and that its receptor Ptc2 was expressed in a gradient in the secondary heart field, with the most robust expression in the caudal secondary heart field, closest to the Shh expression. In vitro culture of secondary heart field with the hedgehog inhibitor cyclopamine significantly reduced proliferation. In ovo, cyclopamine treatment before the secondary heart field adds to the outflow tract reduced proliferation only in the caudal secondary heart field, which coincided with the region of high Ptc2 expression. After outflow tract septation should occur, embryos treated with cyclopamine exhibited pulmonary atresia, pulmonary stenosis, and persistent truncus arteriosus. In hearts with pulmonary atresia, cardiac neural crest-derived cells, which form the outflow tract septum, migrated into the outflow tract and formed a septum. However, this septum divided the outflow tract into two unequal sized vessels and effectively closed off the pulmonary outlet. These experiments show that Shh is necessary for secondary heart field proliferation, which is required for normal pulmonary trunk formation, and that embryos with pulmonary atresia have an outflow tract septum.


Subject(s)
Cell Proliferation , Heart/embryology , Hedgehog Proteins/physiology , Myocardium/cytology , Animals , Base Sequence , DNA Primers , Heart/drug effects , Hedgehog Proteins/genetics , In Situ Hybridization , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Veratrum Alkaloids/pharmacology
19.
Circ Res ; 104(2): 179-88, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19059840

ABSTRACT

Recent studies have shown that the primary heart tube continues to grow by addition of cells from the coelomic wall. This growth occurs concomitantly with embryonic folding and formation of the coelomic cavity, making early heart formation morphologically complex. A scarcity of data on localized growth parameters further hampers the understanding of cardiac growth. Therefore, we investigated local proliferation during early heart formation. Firstly, we determined the cell cycle length of primary myocardium of the early heart tube to be 5.5 days, showing that this myocardium is nonproliferating and implying that initial heart formation occurs solely by addition of cells. In line with this, we show that the heart tube rapidly lengthens at its inflow by differentiation of recently divided precursor cells. To track the origin of these cells, we made quantitative 3D reconstructions of proliferation in the forming heart tube and the mesoderm of its flanking coelomic walls. These reconstructions show a single, albeit bilateral, center of rapid proliferation in the caudomedial pericardial back wall. This center expresses Islet1. Cell tracing showed that cells from this caudal growth center, besides feeding into the venous pole of the heart, also move cranially via the dorsal pericardial mesoderm and differentiate into myocardium at the arterial pole. Inhibition of caudal proliferation impairs the formation of both the atria and the right ventricle. These data show how a proliferating growth center in the caudal coelomic wall elongates the heart tube at both its venous and arterial pole, providing a morphological mechanism for early heart formation.


Subject(s)
Cell Differentiation , Cell Movement , Cell Proliferation , Heart/embryology , Myocardium/cytology , Animals , Bromodeoxyuridine/metabolism , Cell Cycle , Chick Embryo , Heart Ventricles/embryology , Homeodomain Proteins/metabolism , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Immunohistochemistry , LIM-Homeodomain Proteins , Mesoderm/cytology , Models, Anatomic , Models, Cardiovascular , Myocardium/metabolism , Organogenesis , Pericardium/embryology , Time Factors , Transcription Factors
20.
Article in English | MEDLINE | ID: mdl-20490374

ABSTRACT

Cardiac neural crest cells represent a unique subpopulation of cranial neural crest cells that are specified, delaminate and migrate from the developing neural tube to the caudal pharynx where they support aortic arch artery development. From the caudal pharynx, a subset of these cells migrates into the cardiac outflow tract where they are needed for outflow septation. Many signaling factors are known to be involved in specifying and triggering the migration of neural crest cells. These factors have not been specifically studied in cardiac crest but are assumed to be the same as for the other regions of crest. Signaling factors like Ephs and Semaphorins guide the cells into the caudal pharynx. Support of the cells in the pharynx is from endothelin, PDGF and the TGFbeta/BMP signaling pathways. Mutants in the TGFbeta/BMP pathway show abnormal migration or survival in the pharynx, whereas the migration of the neural crest cells into the outflow tract is orchestrated by Semaphorin/Plexin signaling. Although TGFbeta family members have been well studied and show defective neural crest function in outflow septation, their mechanism of action remains unclear.


Subject(s)
Fetal Heart/embryology , Neural Crest/embryology , Animals , Body Patterning/genetics , Body Patterning/physiology , Bone Morphogenetic Proteins/physiology , Cell Movement , Humans , Mutation , Platelet-Derived Growth Factor/physiology , Semaphorins/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Systems Biology , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL