Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 280
Filter
1.
Circ Res ; 130(3): 352-365, 2022 02 04.
Article in English | MEDLINE | ID: mdl-34995101

ABSTRACT

BACKGROUND: Unraveling how new coronary arteries develop may provide critical information for establishing novel therapeutic approaches to treating ischemic cardiac diseases. There are 2 distinct coronary vascular populations derived from different origins in the developing heart. Understanding the formation of coronary arteries may provide insights into new ways of promoting coronary artery formation after myocardial infarction. METHODS: To understand how intramyocardial coronary arteries are generated to connect these 2 coronary vascular populations, we combined genetic lineage tracing, light sheet microscopy, fluorescence micro-optical sectioning tomography, and tissue-specific gene knockout approaches to understand their cellular and molecular mechanisms. RESULTS: We show that a subset of intramyocardial coronary arteries form by angiogenic extension of endocardium-derived vascular tunnels in the neonatal heart. Three-dimensional whole-mount fluorescence imaging showed that these endocardium-derived vascular tunnels or tubes adopt an arterial fate in neonates. Mechanistically, we implicate Mettl3 (methyltransferase-like protein 3) and Notch signaling in regulating endocardium-derived intramyocardial coronary artery formation. Functionally, these intramyocardial arteries persist into adulthood and play a protective role after myocardial infarction. CONCLUSIONS: A subset of intramyocardial coronary arteries form by extension of endocardium-derived vascular tunnels in the neonatal heart.


Subject(s)
Coronary Vessels/embryology , Endocardium/embryology , Animals , Coronary Vessels/growth & development , Coronary Vessels/metabolism , Endocardium/growth & development , Endocardium/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Organogenesis
2.
Dev Biol ; 465(1): 46-57, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32628938

ABSTRACT

Endocardium is critically important for proper function of the cardiovascular system. Not only does endocardium connect the heart to blood vasculature, it also plays an important role in heart morphogenesis, valve formation, and ventricular trabeculation. The extracellular protein Fibronectin (Fn1) promotes endocardial differentiation, but the signaling pathways downstream of Fn1 that regulate endocardial development are not understood. Here, we analyzed the role of the Fibronectin receptors Integrin alpha5 (Itga5) and Integrin alpha4 (Itga4) in zebrafish heart development. We show that itga5 mRNA is expressed in both endocardium and myocardium during early stages of heart development. Through analysis of both itga5 single mutants and itga4;itga5 double mutants, we show that loss of both itga5 and itga4 results in enhanced defects in endocardial differentiation and morphogenesis compared to loss of itga5 alone. Loss of both itga5 and itga4 results in cardia bifida and severe myocardial morphology defects. Finally, we find that loss of itga5 and itga4 results in abnormally narrow anterior endodermal sheet morphology. Together, our results support a model in which Itga5 and Itga4 cooperate to promote endocardial differentiation, medial migration of endocardial and myocardial cells, and morphogenesis of anterior endoderm.


Subject(s)
Cell Differentiation , Endocardium/embryology , Integrin alpha4/metabolism , Integrin alpha5/metabolism , Models, Biological , Organogenesis , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Integrin alpha4/genetics , Integrin alpha5/genetics , Mutation , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
Development ; 145(13)2018 07 02.
Article in English | MEDLINE | ID: mdl-29853617

ABSTRACT

Signaling interactions between the myocardium and endocardium pattern embryonic cardiac regions, instructing their development to fulfill specific functions in the mature heart. We show that ectopic Bmp2 expression in the mouse chamber myocardium changes the transcriptional signature of adjacent chamber endocardial cells into valve tissue, and enables them to undergo epithelial-mesenchyme transition. This induction is independent of valve myocardium specification and requires high levels of Notch1 activity. Biochemical experiments suggest that Bmp2-mediated Notch1 induction is achieved through transcriptional activation of the Notch ligand Jag1, and physical interaction of Smad1/5 with the intracellular domain of the Notch1 receptor. Thus, widespread myocardial Bmp2 and endocardial Notch signaling drive presumptive ventricular endocardium to differentiate into valve endocardium. Understanding the molecular basis of valve development is instrumental to designing therapeutic strategies for congenital heart valve defects.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Embryo, Mammalian/embryology , Endocardium/embryology , Heart Valves/embryology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Bone Morphogenetic Protein 2/genetics , Embryo, Mammalian/cytology , Endocardium/cytology , Heart Valves/cytology , Mice , Mice, Transgenic , Myocardium/cytology , Myocardium/metabolism , Receptors, Notch/genetics , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism
4.
J Anat ; 238(2): 508-514, 2021 02.
Article in English | MEDLINE | ID: mdl-32920869

ABSTRACT

Fsp1 (a.k.a S100A4 or Metastatin) is an intracellular and secreted protein widely regarded as a fibroblast marker. Recent studies have nonetheless shown that Fsp1 is also expressed by other cell types, including small subsets of endothelial cells. Since no detailed and systematic description of Fsp1 spatio-temporal expression pattern in cardiac vascular cells is available in the literature, we have used a transgenic murine line (Fsp1-GFP) to study Fsp1 expression in the developing and postnatal cardiac vasculature and endocardium. Our work shows that Fsp1 is expressed in the endocardium and mesenchyme of atrioventricular valve primordia, as well as in some coronary venous and lymphatic endothelial cells. Fsp1 expression in cardiac venous and lymphatic endothelium is progressively restricted to the leaflets of cardiac venous and lymphatic valves. Our results suggest that Fsp1 could play a role in the development of atrioventricular valves and participate in the patterning and morphogenesis of cardiac venous and lymphatic vessel valves.


Subject(s)
Coronary Vessels/embryology , Embryo, Mammalian/metabolism , Endocardium/embryology , S100 Calcium-Binding Protein A4/metabolism , Animals , Coronary Vessels/metabolism , Endocardium/metabolism , Endothelium, Lymphatic/metabolism , Female , Mice , Mice, Transgenic , Pregnancy , Venous Valves/metabolism
5.
Clin Sci (Lond) ; 135(6): 829-846, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33720353

ABSTRACT

The endothelial-to-mesenchymal transition (EndMT) is a critical process that occurs during the development of the outflow tract (OFT). Malformations of the OFT can lead to the occurrence of conotruncal defect (CTD). SOX7 duplication has been reported in patients with congenital CTD, but its specific role in OFT development remains poorly understood. To decipher this, histological analysis showed that SRY-related HMG-box 7 (SOX7) was regionally expressed in the endocardial endothelial cells and in the mesenchymal cells of the OFT, where EndMT occurs. Experiments, using in vitro collagen gel culture system, revealed that SOX7 was a negative regulator of EndMT that inhibited endocardial cell (EC) migration and resulted in decreased number of mesenchymal cells. Forced expression of SOX7 in endothelial cells blocked further migration and improved the expression of the adhesion protein vascular endothelial (VE)-cadherin (VE-cadherin). Moreover, a VE-cadherin knockdown could partly reverse the SOX7-mediated repression of cell migration. Luciferase and electrophoretic mobility shift assay (EMSA) demonstrated that SOX7 up-regulated VE-cadherin by directly binding to the gene's promoter in endothelial cells. The coding exons and splicing regions of the SOX7 gene were also scanned in the 536 sporadic CTD patients and in 300 unaffected controls, which revealed four heterozygous SOX7 mutations. Luciferase assays revealed that two SOX7 variants weakened the transactivation of the VE-cadherin promoter. In conclusion, SOX7 inhibited EndMT during OFT development by directly up-regulating the endothelial-specific adhesion molecule VE-cadherin. SOX7 mutations can lead to impaired EndMT by regulating VE-cadherin, which may give rise to the molecular mechanisms associated with SOX7 in CTD pathogenesis.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Endocardium/embryology , Heart Defects, Congenital/embryology , SOXF Transcription Factors/metabolism , Animals , Antigens, CD/genetics , Cadherins/genetics , Cell Movement , Embryo, Mammalian , Endocardium/cytology , Endothelium/growth & development , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Developmental , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice, Inbred C57BL , Promoter Regions, Genetic , Rats , SOXF Transcription Factors/genetics
6.
Mol Cell Proteomics ; 18(9): 1782-1795, 2019 09.
Article in English | MEDLINE | ID: mdl-31249105

ABSTRACT

The endocardium is a specialized endothelium that lines the inner surface of the heart. Functional studies in mice and zebrafish have established that the endocardium is a source of instructive signals for the development of cardiac structures, including the heart valves and chambers. Here, we characterized the NOTCH-dependent endocardial secretome by manipulating NOTCH activity in mouse embryonic endocardial cells (MEEC) followed by mass spectrometry-based proteomics. We profiled different sets of soluble factors whose secretion not only responds to NOTCH activation but also shows differential ligand specificity, suggesting that ligand-specific inputs may regulate the expression of secreted proteins involved in different cardiac development processes. NOTCH signaling activation correlates with a transforming growth factor-ß2 (TGFß2)-rich secretome and the delivery of paracrine signals involved in focal adhesion and extracellular matrix (ECM) deposition and remodeling. In contrast, NOTCH inhibition is accompanied by the up-regulation of specific semaphorins that may modulate cell migration. The secretome protein expression data showed a good correlation with gene profiling of RNA expression in embryonic endocardial cells. Additional characterization by in situ hybridization in mouse embryos revealed expression of various NOTCH candidate effector genes (Tgfß2, Loxl2, Ptx3, Timp3, Fbln2, and Dcn) in heart valve endocardium and/or mesenchyme. Validating these results, mice with conditional Dll4 or Jag1 loss-of-function mutations showed gene expression alterations similar to those observed at the protein level in vitro These results provide the first description of the NOTCH-dependent endocardial secretome and validate MEEC as a tool for assaying the endocardial secretome response to a variety of stimuli and the potential use of this system for drug screening.


Subject(s)
Endocardium/embryology , Endocardium/metabolism , Heart Valves/embryology , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Benzazepines/pharmacology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Endocardium/cytology , Endocardium/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Heart Valves/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Mice, Mutant Strains , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Notch/genetics , Reproducibility of Results
7.
Development ; 143(3): 373-86, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26839341

ABSTRACT

Endocardial cells are cardiac endothelial cells that line the interior of the heart tube. Historically, their contribution to cardiac development has mainly been considered from a morphological perspective. However, recent studies have begun to define novel instructive roles of the endocardium, as a sensor and signal transducer of biophysical forces induced by blood flow, and as an angiocrine signalling centre that is involved in myocardial cellular morphogenesis, regeneration and reprogramming. In this Review, we discuss how the endocardium develops, how endocardial-myocardial interactions influence the developing embryonic heart, and how the dysregulation of blood flow-responsive endocardial signalling can result in pathophysiological changes.


Subject(s)
Endocardium/embryology , Mechanotransduction, Cellular , Morphogenesis , Animals , Hemodynamics , Humans , Regeneration , Stem Cells/cytology
8.
Development ; 143(12): 2217-27, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27302398

ABSTRACT

During cardiac valve development, the single-layered endocardial sheet at the atrioventricular canal (AVC) is remodeled into multilayered immature valve leaflets. Most of our knowledge about this process comes from examining fixed samples that do not allow a real-time appreciation of the intricacies of valve formation. Here, we exploit non-invasive in vivo imaging techniques to identify the dynamic cell behaviors that lead to the formation of the immature valve leaflets. We find that in zebrafish, the valve leaflets consist of two sets of endocardial cells at the luminal and abluminal side, which we refer to as luminal cells (LCs) and abluminal cells (ALCs), respectively. By analyzing cellular rearrangements during valve formation, we observed that the LCs and ALCs originate from the atrium and ventricle, respectively. Furthermore, we utilized Wnt/ß-catenin and Notch signaling reporter lines to distinguish between the LCs and ALCs, and also found that cardiac contractility and/or blood flow is necessary for the endocardial expression of these signaling reporters. Thus, our 3D analyses of cardiac valve formation in zebrafish provide fundamental insights into the cellular rearrangements underlying this process.


Subject(s)
Heart Valves/cytology , Heart Valves/embryology , Imaging, Three-Dimensional , Animals , Cell Movement , Coronary Circulation , Endocardium/cytology , Endocardium/embryology , Gene Expression Regulation, Developmental , Heart Atria/cytology , Heart Atria/embryology , Heart Ventricles/cytology , Heart Ventricles/embryology , Mutation/genetics , Myocardial Contraction , Organogenesis/genetics , Receptors, Notch/metabolism , Wnt Signaling Pathway , Zebrafish
9.
Development ; 143(19): 3522-3531, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27578788

ABSTRACT

Coronary vessel development is a highly coordinated process during heart formation. Abnormal development and dysfunction of the coronary network are contributory factors in the majority of heart disease. Understanding the molecular mechanisms that regulate coronary vessel formation is crucial for preventing and treating the disease. We report a zebrafish gene-trap vinculin b (vclb) mutant that displays abnormal coronary vessel development among multiple cardiac defects. The mutant shows overproliferation of epicardium-derived cells and disorganization of coronary vessels, and they eventually die off at juvenile stages. Mechanistically, Vclb deficiency results in the release of another cytoskeletal protein, paxillin, from the Vclb complex and the upregulation of ERK and FAK phosphorylation in epicardium and endocardium, causing disorganization of endothelial cells and pericytes during coronary vessel development. By contrast, cardiac muscle development is relatively normal, probably owing to redundancy with Vcla, a vinculin paralog that is expressed in the myocardium but not epicardium. Together, our results reveal a previously unappreciated function of vinculin in epicardium and endocardium and reinforce the notion that well-balanced FAK activity is essential for coronary vessel development.


Subject(s)
Coronary Vessels/metabolism , Coronary Vessels/pathology , Hyperplasia/metabolism , Pericardium/metabolism , Vinculin/metabolism , Zebrafish Proteins/metabolism , Animals , Coronary Vessels/embryology , Endocardium/embryology , Endocardium/metabolism , Endocardium/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart/embryology , Hyperplasia/pathology , Pericardium/embryology , Pericardium/pathology , Phosphorylation , Vinculin/genetics , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics
10.
Development ; 143(3): 473-82, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26674310

ABSTRACT

The elucidation of mechanisms in semilunar valve development might enable the development of new therapies for congenital heart disorders. Here, we found differences in proliferation-associated genes and genes repressed by VEGF between human semilunar valve leaflets from first and second trimester hearts. The proliferation of valve interstitial cells and ventricular valve endothelial cells (VECs) and cellular density declined from the first to the second trimester. Cytoplasmic expression of NFATC1 was detected in VECs (4 weeks) and, later, cells in the leaflet/annulus junction mesenchyme expressing inactive NFATC1 (5.5-9 weeks) were detected, indicative of endocardial-to-mesenchymal transformation (EndMT) in valvulogenesis. At this leaflet/annulus junction, CD44(+) cells clustered during elongation (11 weeks), extending toward the tip along the fibrosal layer in second trimester leaflets. Differing patterns of maturation in the fibrosa and ventricularis were detected via increased fibrosal periostin content, which tracked the presence of the CD44(+) cells in the second trimester. We revealed that spatiotemporal NFATC1 expression actively regulates EndMT during human valvulogenesis, as early as 4 weeks. Additionally, CD44(+) cells play a role in leaflet maturation toward the trilaminar structure, possibly via migration of VECs undergoing EndMT, which subsequently ascend from the leaflet/annulus junction.


Subject(s)
Endocardium/embryology , Heart Valves/cytology , Heart Valves/embryology , Mesoderm/cytology , Mesoderm/embryology , Cell Adhesion Molecules/metabolism , Cell Count , Cell Differentiation , Cell Proliferation , Endothelial Cells/metabolism , Female , Gene Expression Regulation, Developmental , Humans , Hyaluronan Receptors/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Pregnancy , Pregnancy Trimester, Second , Spatio-Temporal Analysis , Time Factors , Vascular Endothelial Growth Factor A/metabolism
11.
Circ Res ; 118(12): 1880-93, 2016 06 10.
Article in English | MEDLINE | ID: mdl-27056912

ABSTRACT

RATIONALE: There is persistent uncertainty regarding the developmental origins of coronary vessels, with 2 principal sources suggested as ventricular endocardium or sinus venosus (SV). These 2 proposed origins implicate fundamentally distinct mechanisms of vessel formation. Resolution of this controversy is critical for deciphering the programs that result in the formation of coronary vessels and has implications for research on therapeutic angiogenesis. OBJECTIVE: To resolve the controversy over the developmental origin of coronary vessels. METHODS AND RESULTS: We first generated nuclear factor of activated T cells (Nfatc1)-Cre and Nfatc1-Dre lineage tracers for endocardium labeling. We found that Nfatc1 recombinases also label a significant portion of SV endothelial cells in addition to endocardium. Therefore, restricted endocardial lineage tracing requires a specific marker that distinguishes endocardium from SV. By single-cell gene expression analysis, we identified a novel endocardial gene natriuretic peptide receptor 3 (Npr3). Npr3 is expressed in the entirety of the endocardium but not in the SV. Genetic lineage tracing based on Npr3-CreER showed that endocardium contributes to a minority of coronary vessels in the free walls of embryonic heart. Intersectional genetic lineage tracing experiments demonstrated that endocardium minimally contributes to coronary endothelium in the embryonic ventricular free walls. CONCLUSIONS: Our study suggested that SV, but not endocardium, is the major origin for coronary endothelium in the embryonic ventricular free walls. This work thus resolves the recent controversy over the developmental origin of coronary endothelium, providing the basis for studying coronary vessel formation and regeneration after injury.


Subject(s)
Cell Lineage , Coronary Vessels/embryology , Endocardium/embryology , Endothelium, Vascular/metabolism , Heart Ventricles/embryology , Animals , Coronary Vessels/cytology , Coronary Vessels/metabolism , Endocardium/cytology , Endocardium/metabolism , Endothelium, Vascular/cytology , Heart Ventricles/cytology , Heart Ventricles/metabolism , Mice , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism
12.
Dev Biol ; 412(1): 71-82, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26892463

ABSTRACT

The atrioventricular canal (AVC) connects the atrial and ventricular chambers of the heart and its formation is critical for the development of the cardiac valves, chamber septation and formation of the cardiac conduction system. Consequently, problems in AVC formation can lead to congenital defects ranging from cardiac arrhythmia to incomplete cardiac septation. While our knowledge about early heart tube formation is relatively comprehensive, much remains to be investigated about the genes that regulate AVC formation. Here we identify a new role for the basic helix-loop-helix factor Id4 in zebrafish AVC valve development and function. id4 is first expressed in the AVC endocardium and later becomes more highly expressed in the atrial chamber. TALEN induced inactivation of id4 causes retrograde blood flow at the AV canal under heat induced stress conditions, indicating defects in AV valve function. At the molecular level, we found that id4 inactivation causes misexpression of several genes important for AVC and AV valve formation including bmp4 and spp1. We further show that id4 appears to control the number of endocardial cells that contribute to the AV valves by regulating Wnt signaling in the developing AVC endocardium.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Endocardium/embryology , Inhibitor of Differentiation Proteins/physiology , Signal Transduction , Zebrafish/embryology , Animals
13.
Dev Biol ; 409(1): 272-276, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26522286

ABSTRACT

Heart outflow tract septation in mouse embryos carrying mutations in retinoic acid receptor genes fails with complete penetrance. In this mutant background, ectopic TGFß signaling in the distal outflow tract is responsible for septation failure, but it was uncertain what tissue was responsive to ectopic TGFß and why this response interfered with septation. By combining RAR gene mutation with tissue-specific Cre drivers and a conditional type II TGFß receptor (Tgfbr2) allele, we determined that ectopic activation of TGFß signaling in the endocardium is responsible for septation defects. Ectopic TGFß signaling results in ectopic mesenchymal transformation of the endocardium and thereby in improperly constituted distal OFT cushions. Our analysis highlights the interactions between myocardium, endocardium, and neural crest cells in outflow tract morphogenesis, and demonstrates the requirement for proper TGFß signaling in outflow tract cushion organization and septation.


Subject(s)
Endocardium/pathology , Heart Failure/pathology , Heart Septal Defects/pathology , Mesoderm/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Endocardium/embryology , Endocardium/metabolism , Heart Failure/embryology , Heart Failure/metabolism , Heart Septal Defects/embryology , Heart Septal Defects/metabolism , Mesoderm/embryology , Mice , Mutation/genetics , Organ Specificity , Phenotype , Receptors, Retinoic Acid/metabolism
14.
Dev Biol ; 414(2): 228-43, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27158029

ABSTRACT

Formation of the heart tube requires synchronized migration of endocardial and myocardial precursors. Our previous studies indicated that in S1pr2/Gα13-deficient embryos, impaired endoderm convergence disrupted the medial migration of myocardial precursors, resulting in the formation of two myocardial populations. Here we show that endoderm convergence also regulates endocardial migration. In embryos defective for S1pr2/Gα13 signaling, endocardial precursors failed to migrate towards the midline, and the presumptive endocardium surrounded the bilaterally-located myocardial cells rather than being encompassed by them. In vivo imaging of control embryos revealed that, like their myocardial counterparts, endocardial precursors migrated with the converging endoderm, though from a more anterior point, then moved from the dorsal to the ventral side of the endoderm (subduction), and finally migrated posteriorly towards myocardial precursors, ultimately forming the inner layer of the heart tube. In embryos defective for endoderm convergence due to an S1pr2/Gα13 deficiency, both the medial migration and the subduction of endocardial precursors were impaired, and their posterior migration towards the myocardial precursors was premature. This placed them medial to the myocardial populations, physically blocking the medial migration of the myocardial precursors. Furthermore, contact between the endocardial and myocardial precursor populations disrupted the epithelial architecture of the myocardial precursors, and thus their medial migration; in embryos depleted of endocardial cells, the myocardial migration defect was partially rescued. Our data indicate that endoderm convergence regulates the medial migration of endocardial precursors, and that premature association of the endocardial and myocardial populations contributes to myocardial migration defects observed in S1pr2/Gα13-deficient embryos. The demonstration that endoderm convergence regulates the synchronized migration of endocardial and myocardial precursors reveals a new role of the endoderm in heart development.


Subject(s)
Body Patterning/physiology , Endocardium/embryology , Endoderm/embryology , GTP-Binding Protein alpha Subunits, G12-G13/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Body Patterning/genetics , Cell Movement , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , GTP-Binding Protein alpha Subunits, G12-G13/deficiency , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Humans , Luminescent Proteins/analysis , Morpholinos/genetics , Morpholinos/pharmacology , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
15.
Biochim Biophys Acta ; 1863(7 Pt B): 1937-47, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26828773

ABSTRACT

Endocardial development involves a complex orchestration of cell fate decisions that coordinate with endoderm formation and other mesodermal cell lineages. Historically, investigations into the contribution of endocardium in the developing embryo was constrained to the heart where these cells give rise to the inner lining of the myocardium and are a major contributor to valve formation. In recent years, studies have continued to elucidate the complexities of endocardial fate commitment revealing a much broader scope of lineage potential from developing endocardium. These studies cover a wide range of species and model systems and show direct contribution or fate potential of endocardium giving rise to cardiac vasculature, blood, fibroblast, and cardiomyocyte lineages. This review focuses on the marked expansion of knowledge in the area of endocardial fate potential. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Endocardium/physiology , Endothelial Cells/physiology , Endothelium, Vascular/physiology , Animals , Endocardium/embryology , Endocardium/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/physiology , Morphogenesis , Phenotype
16.
Development ; 141(15): 2959-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25053429

ABSTRACT

A complex regulatory network of morphogens and transcription factors is essential for normal cardiac development. Nkx2-5 is among the earliest known markers of cardiac mesoderm that is central to the regulatory pathways mediating second heart field (SHF) development. Here, we have examined the specific requirements for Nkx2-5 in the SHF progenitors. We show that Nkx2-5 potentiates Wnt signaling by regulating the expression of the R-spondin3 (Rspo3) gene during cardiogenesis. R-spondins are secreted factors and potent Wnt agonists that in part regulate stem cell proliferation. Our data show that Rspo3 is markedly downregulated in Nkx2-5 mutants and that Rspo3 expression is regulated by Nkx2-5. Conditional inactivation of Rspo3 in the Isl1 lineage resulted in embryonic lethality secondary to impaired development of SHF. More importantly, we find that Wnt signaling is significantly attenuated in Nkx2-5 mutants and that enhancing Wnt/ß-catenin signaling by pharmacological treatment or by transgenic expression of Rspo3 rescues the SHF defects in the conditional Nkx2-5(+/-) mutants. We have identified a previously unrecognized genetic link between Nkx2-5 and Wnt signaling that supports continued cardiac growth and proliferation during development. Identification of Rspo3 in cardiac development provides a new paradigm in temporal regulation of Wnt signaling by cardiac-specific transcription factors.


Subject(s)
Heart/embryology , Homeodomain Proteins/physiology , Thrombospondins/physiology , Transcription Factors/physiology , Wnt Signaling Pathway , Animals , Base Sequence , Cell Lineage , Cell Proliferation , Endocardium/embryology , Female , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Sequence Homology, Nucleic Acid , Stem Cells/cytology , Thrombospondins/genetics , Transcription Factors/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism
17.
Circ Res ; 116(7): 1216-30, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25814683

ABSTRACT

Although numerous preclinical investigations have consistently demonstrated salubrious effects of c-kit(pos) cardiac cells administered after myocardial infarction, the mechanism of action remains highly controversial. We and others have found little or no evidence that these cells differentiate into mature functional cardiomyocytes, suggesting paracrine effects. In this review, we propose a new paradigm predicated on a comprehensive analysis of the literature, including studies of cardiac development; we have (facetiously) dubbed this conceptual construct "string theory" of c-kit(pos) cardiac cells because it reconciles multifarious and sometimes apparently discrepant results. There is strong evidence that, during development, the c-kit receptor is expressed in different pools of cardiac progenitors (some capable of robust cardiomyogenesis and others with little or no contribution to myocytes). Accordingly, c-kit positivity, in itself, does not define the embryonic origins, lineage capabilities, or differentiation capacities of specific cardiac progenitors. C-kit(pos) cells derived from the first heart field exhibit cardiomyogenic potential during development, but these cells are likely depleted shortly before or after birth. The residual c-kit(pos) cells found in the adult heart are probably of proepicardial origin, possess a mesenchymal phenotype (resembling bone marrow mesenchymal stem/stromal cells), and are capable of contributing significantly only to nonmyocytic lineages (fibroblasts, smooth muscle cells, and endothelial cells). If these 2 populations (first heart field and proepicardium) express different levels of c-kit, the cardiomyogenic potential of first heart field progenitors might be reconciled with recent results of c-kit(pos) cell lineage tracing studies. The concept that c-kit expression in the adult heart identifies epicardium-derived, noncardiomyogenic precursors with a mesenchymal phenotype helps to explain the beneficial effects of c-kit(pos) cell administration to ischemically damaged hearts despite the observed paucity of cardiomyogenic differentiation of these cells.


Subject(s)
Cell Lineage , Models, Cardiovascular , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Proto-Oncogene Proteins c-kit , Adventitia/cytology , Blood Vessels/cytology , Blood Vessels/embryology , Cell Differentiation , Clinical Trials, Phase I as Topic , Cytokines/physiology , Endocardium/cytology , Endocardium/embryology , Epithelial-Mesenchymal Transition , Graft Survival , Heart/embryology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Muscle, Smooth/cytology , Myocytes, Cardiac/chemistry , Paracrine Communication , Pericardium/cytology , Pericardium/embryology , Stem Cells/chemistry , Stem Cells/classification , Stem Cells/cytology , Transplantation, Autologous
18.
Dev Biol ; 407(1): 158-72, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26100917

ABSTRACT

The formation of intricately organized aortic and pulmonic valves from primitive endocardial cushions of the outflow tract is a remarkable accomplishment of embryonic development. While not always initially pathologic, developmental semilunar valve (SLV) defects, including bicuspid aortic valve, frequently progress to a disease state in adults requiring valve replacement surgery. Disrupted embryonic growth, differentiation, and patterning events that "trigger" SLV disease are coordinated by gene expression changes in endocardial, myocardial, and cushion mesenchymal cells. We explored roles of chromatin regulation in valve gene regulatory networks by conditional inactivation of the Brg1-associated factor (BAF) chromatin remodeling complex in the endocardial lineage. Endocardial Brg1-deficient mouse embryos develop thickened and disorganized SLV cusps that frequently become bicuspid and myxomatous, including in surviving adults. These SLV disease-like phenotypes originate from deficient endocardial-to-mesenchymal transformation (EMT) in the proximal outflow tract (pOFT) cushions. The missing cells are replaced by compensating neural crest or other non-EMT-derived mesenchyme. However, these cells are incompetent to fully pattern the valve interstitium into distinct regions with specialized extracellular matrices. Transcriptomics reveal genes that may promote growth and patterning of SLVs and/or serve as disease-state biomarkers. Mechanistic studies of SLV disease genes should distinguish between disease origins and progression; the latter may reflect secondary responses to a disrupted developmental system.


Subject(s)
Aortic Valve/embryology , DNA Helicases/physiology , Endocardium/embryology , Heart Valve Diseases/etiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Disease Models, Animal , Female , Mice , NFATC Transcription Factors/physiology
19.
Development ; 140(9): 1946-57, 2013 May.
Article in English | MEDLINE | ID: mdl-23571217

ABSTRACT

Trabeculation and compaction of the embryonic myocardium are morphogenetic events crucial for the formation and function of the ventricular walls. Fkbp1a (FKBP12) is a ubiquitously expressed cis-trans peptidyl-prolyl isomerase. Fkbp1a-deficient mice develop ventricular hypertrabeculation and noncompaction. To determine the physiological function of Fkbp1a in regulating the intercellular and intracellular signaling pathways involved in ventricular trabeculation and compaction, we generated a series of Fkbp1a conditional knockouts. Surprisingly, cardiomyocyte-restricted ablation of Fkbp1a did not give rise to the ventricular developmental defect, whereas endothelial cell-restricted ablation of Fkbp1a recapitulated the ventricular hypertrabeculation and noncompaction observed in Fkbp1a systemically deficient mice, suggesting an important contribution of Fkbp1a within the developing endocardia in regulating the morphogenesis of ventricular trabeculation and compaction. Further analysis demonstrated that Fkbp1a is a novel negative modulator of activated Notch1. Activated Notch1 (N1ICD) was significantly upregulated in Fkbp1a-ablated endothelial cells in vivo and in vitro. Overexpression of Fkbp1a significantly reduced the stability of N1ICD and direct inhibition of Notch signaling significantly reduced hypertrabeculation in Fkbp1a-deficient mice. Our findings suggest that Fkbp1a-mediated regulation of Notch1 plays an important role in intercellular communication between endocardium and myocardium, which is crucial in controlling the formation of the ventricular walls.


Subject(s)
Endocardium/metabolism , Heart Ventricles/pathology , Myocardium/metabolism , Receptor, Notch1/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Cell Lineage , Cells, Cultured , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Embryonic Development , Endocardium/embryology , Endocardium/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Heart Ventricles/embryology , Heart Ventricles/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout/embryology , Mice, Knockout/metabolism , Myocardium/pathology , Neural Crest/metabolism , Neural Crest/pathology , Phenotype , Receptor, Notch1/genetics , Signal Transduction , Tacrolimus Binding Proteins/genetics , Transfection
20.
Proc Natl Acad Sci U S A ; 110(42): 16898-903, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24082093

ABSTRACT

Despite their abundance and multiple functions in a variety of organ systems, the function and signaling mechanisms of adhesion G protein-coupled receptors (GPCRs) are poorly understood. Adhesion GPCRs possess large N termini containing various functional domains. In addition, many of them are autoproteolytically cleaved at their GPS sites into an N-terminal fragment (NTF) and C-terminal fragment. Here we demonstrate that Gpr126 is expressed in the endocardium during early mouse heart development. Gpr126 knockout in mice and knockdown in zebrafish caused hypotrabeculation and affected mitochondrial function. Ectopic expression of Gpr126-NTF that lacks the GPS motif (NTF(ΔGPS)) in zebrafish rescued the trabeculation but not the previously described myelination phenotype in the peripheral nervous system. These data support a model in which the NTF of Gpr126, in contrast to the C-terminal fragment, plays an important role in heart development. Collectively, our analysis provides a unique example of the versatile function and signaling properties of adhesion GPCRs in vertebrates.


Subject(s)
Endocardium/embryology , Mitochondria, Heart/metabolism , Models, Biological , Receptors, G-Protein-Coupled/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Endocardium/cytology , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Organ Specificity/physiology , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL