Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Dev Biol ; 498: 77-86, 2023 06.
Article in English | MEDLINE | ID: mdl-37037405

ABSTRACT

Outflow tract (OFT) develops from cardiac progenitor cells in the second heart field (SHF) domain. APJ, a G-Protein Coupled Receptor, is expressed by cardiac progenitors in the SHF. By lineage tracing APJ+SHF cells, we show that these cardiac progenitors contribute to the cells of OFT, which eventually give rise to aorta and pulmonary trunk/artery upon its morphogenesis. Furthermore, we show that early APJ â€‹+ â€‹cells give rise to both aorta and pulmonary cells but late APJ â€‹+ â€‹cells predominantly give rise to pulmonary cells. APJ is expressed by the outflow tract progenitors in the SHF but its role is unclear. We performed knockout studies to determine the role of APJ in SHF cell proliferation and survival. Our data suggested that APJ knockout in the SHF reduced the proliferation of SHF progenitors, while there was no significant impact on survival. In addition, we show that ectopic overexpression of WNT in these cells disrupted aorta and pulmonary morphogenesis from OFT. Overall, our study has identified APJ â€‹+ â€‹progenitor population within the SHF that give rise to aorta and pulmonary trunk/artery cells. Furthermore, we show that APJ signaling stimulates proliferation of these cells in the SHF.


Subject(s)
Heart , Signal Transduction , Stem Cells , Pulmonary Artery , Aorta , Myocardium , Gene Expression Regulation, Developmental
2.
Dev Cell ; 57(22): 2517-2532.e6, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36347256

ABSTRACT

Endocardial cells lining the heart lumen are coronary vessel progenitors during embryogenesis. Re-igniting this developmental process in adults could regenerate blood vessels lost during cardiac injury, but this requires additional knowledge of molecular mechanisms. Here, we use mouse genetics and scRNA-seq to identify regulators of endocardial angiogenesis and precisely assess the role of CXCL12/CXCR4 signaling. Time-specific lineage tracing demonstrated that endocardial cells differentiated into coronary endothelial cells primarily at mid-gestation. A new mouse line reporting CXCR4 activity-along with cell-specific gene deletions-demonstrated it was specifically required for artery morphogenesis rather than angiogenesis. Integrating scRNA-seq data of endocardial-derived coronary vessels from mid- and late-gestation identified a Bmp2-expressing transitioning population specific to mid-gestation. Bmp2 stimulated endocardial angiogenesis in vitro and in injured neonatal mouse hearts. Our data demonstrate how understanding the molecular mechanisms underlying endocardial angiogenesis can identify new potential therapeutic targets promoting revascularization of the injured heart.


Subject(s)
Coronary Vessels , Endocardium , Animals , Female , Mice , Pregnancy , Bone Morphogenetic Protein 2 , Cell Differentiation , Endothelial Cells , Heart , Organogenesis
3.
Science ; 376(6594): eabl4896, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35549404

ABSTRACT

Molecular characterization of cell types using single-cell transcriptome sequencing is revolutionizing cell biology and enabling new insights into the physiology of human organs. We created a human reference atlas comprising nearly 500,000 cells from 24 different tissues and organs, many from the same donor. This atlas enabled molecular characterization of more than 400 cell types, their distribution across tissues, and tissue-specific variation in gene expression. Using multiple tissues from a single donor enabled identification of the clonal distribution of T cells between tissues, identification of the tissue-specific mutation rate in B cells, and analysis of the cell cycle state and proliferative potential of shared cell types across tissues. Cell type-specific RNA splicing was discovered and analyzed across tissues within an individual.


Subject(s)
Atlases as Topic , Cells , Organ Specificity , RNA Splicing , Single-Cell Analysis , Transcriptome , B-Lymphocytes/metabolism , Cells/metabolism , Humans , Organ Specificity/genetics , T-Lymphocytes/metabolism
4.
Nat Cardiovasc Res ; 1(8): 775-790, 2022 Aug.
Article in English | MEDLINE | ID: mdl-37305211

ABSTRACT

Collateral arteries bridge opposing artery branches, forming a natural bypass that can deliver blood flow downstream of an occlusion. Inducing coronary collateral arteries could treat cardiac ischemia, but more knowledge on their developmental mechanisms and functional capabilities is required. Here we used whole-organ imaging and three-dimensional computational fluid dynamics modeling to define spatial architecture and predict blood flow through collaterals in neonate and adult mouse hearts. Neonate collaterals were more numerous, larger in diameter and more effective at restoring blood flow. Decreased blood flow restoration in adults arose because during postnatal growth coronary arteries expanded by adding branches rather than increasing diameters, altering pressure distributions. In humans, adult hearts with total coronary occlusions averaged 2 large collaterals, with predicted moderate function, while normal fetal hearts showed over 40 collaterals, likely too small to be functionally relevant. Thus, we quantify the functional impact of collateral arteries during heart regeneration and repair-a critical step toward realizing their therapeutic potential.

5.
Elife ; 102021 12 15.
Article in English | MEDLINE | ID: mdl-34910626

ABSTRACT

Most cell fate trajectories during development follow a diverging, tree-like branching pattern, but the opposite can occur when distinct progenitors contribute to the same cell type. During this convergent differentiation, it is unknown if cells 'remember' their origins transcriptionally or whether this influences cell behavior. Most coronary blood vessels of the heart develop from two different progenitor sources-the endocardium (Endo) and sinus venosus (SV)-but whether transcriptional or functional differences related to origin are retained is unknown. We addressed this by combining lineage tracing with single-cell RNA sequencing (scRNAseq) in embryonic and adult mouse hearts. Shortly after coronary development begins, capillary endothelial cells (ECs) transcriptionally segregated into two states that retained progenitor-specific gene expression. Later in development, when the coronary vasculature is well established but still remodeling, capillary ECs again segregated into two populations, but transcriptional differences were primarily related to tissue localization rather than lineage. Specifically, ECs in the heart septum expressed genes indicative of increased local hypoxia and decreased blood flow. Adult capillary ECs were more homogeneous with respect to both lineage and location. In agreement, SV- and Endo-derived ECs in adult hearts displayed similar responses to injury. Finally, scRNAseq of developing human coronary vessels indicated that the human heart followed similar principles. Thus, over the course of development, transcriptional heterogeneity in coronary ECs is first influenced by lineage, then by location, until heterogeneity declines in the homeostatic adult heart. These results highlight the plasticity of ECs during development, and the validity of the mouse as a model for human coronary development.


Subject(s)
Coronary Vessels/embryology , Embryo, Mammalian/metabolism , Embryonic Development , Endothelial Cells/metabolism , Animals , Humans , Mice , RNA-Seq , Single-Cell Analysis
7.
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
8.
Cell ; 176(5): 1128-1142.e18, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30686582

ABSTRACT

Collateral arteries are an uncommon vessel subtype that can provide alternate blood flow to preserve tissue following vascular occlusion. Some patients with heart disease develop collateral coronary arteries, and this correlates with increased survival. However, it is not known how these collaterals develop or how to stimulate them. We demonstrate that neonatal mouse hearts use a novel mechanism to build collateral arteries in response to injury. Arterial endothelial cells (ECs) migrated away from arteries along existing capillaries and reassembled into collateral arteries, which we termed "artery reassembly". Artery ECs expressed CXCR4, and following injury, capillary ECs induced its ligand, CXCL12. CXCL12 or CXCR4 deletion impaired collateral artery formation and neonatal heart regeneration. Artery reassembly was nearly absent in adults but was induced by exogenous CXCL12. Thus, understanding neonatal regenerative mechanisms can identify pathways that restore these processes in adults and identify potentially translatable therapeutic strategies for ischemic heart disease.


Subject(s)
Collateral Circulation/physiology , Heart/growth & development , Regeneration/physiology , Animals , Animals, Newborn/growth & development , Chemokine CXCL12/metabolism , Coronary Vessels/growth & development , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Receptors, CXCR4/metabolism , Signal Transduction
9.
Nature ; 559(7714): 356-362, 2018 07.
Article in English | MEDLINE | ID: mdl-29973725

ABSTRACT

Arteries and veins are specified by antagonistic transcriptional programs. However, during development and regeneration, new arteries can arise from pre-existing veins through a poorly understood process of cell fate conversion. Here, using single-cell RNA sequencing and mouse genetics, we show that vein cells of the developing heart undergo an early cell fate switch to create a pre-artery population that subsequently builds coronary arteries. Vein cells underwent a gradual and simultaneous switch from venous to arterial fate before a subset of cells crossed a transcriptional threshold into the pre-artery state. Before the onset of coronary blood flow, pre-artery cells appeared in the immature vessel plexus, expressed mature artery markers, and decreased cell cycling. The vein-specifying transcription factor COUP-TF2 (also known as NR2F2) prevented plexus cells from overcoming the pre-artery threshold by inducing cell cycle genes. Thus, vein-derived coronary arteries are built by pre-artery cells that can differentiate independently of blood flow upon the release of inhibition mediated by COUP-TF2 and cell cycle factors.


Subject(s)
Arteries/cytology , Coronary Vessels/cytology , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism , Veins/cytology , Animals , Arteries/metabolism , COUP Transcription Factor II/metabolism , Cell Cycle/genetics , Cell Differentiation , Cell Lineage , Coronary Vessels/metabolism , Female , Male , Mice , Sequence Analysis, RNA , Veins/metabolism
10.
Nat Commun ; 9(1): 368, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371594

ABSTRACT

During development, the formation of a mature, well-functioning heart requires transformation of the ventricular wall from a loose trabecular network into a dense compact myocardium at mid-gestation. Failure to compact is associated in humans with congenital diseases such as left ventricular non-compaction (LVNC). The mechanisms regulating myocardial compaction are however still poorly understood. Here, we show that deletion of the Ino80 chromatin remodeler in vascular endothelial cells prevents ventricular compaction in the developing mouse heart. This correlates with defective coronary vascularization, and specific deletion of Ino80 in the two major coronary progenitor tissues-sinus venosus and endocardium-causes intermediate phenotypes. In vitro, endothelial cells promote myocardial expansion independently of blood flow in an Ino80-dependent manner. Ino80 deletion increases the expression of E2F-activated genes and endothelial cell S-phase occupancy. Thus, Ino80 is essential for coronary angiogenesis and allows coronary vessels to support proper compaction of the heart wall.


Subject(s)
Adenosine Triphosphatases/metabolism , Endothelium, Vascular/metabolism , Heart Defects, Congenital/metabolism , Neovascularization, Pathologic/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Animals , Coronary Vessels/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins , Endocardium/metabolism , Endocardium/pathology , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelium, Vascular/pathology , Heart Defects, Congenital/genetics , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Neovascularization, Pathologic/genetics
11.
PLoS One ; 13(12): e0203100, 2018.
Article in English | MEDLINE | ID: mdl-30596653

ABSTRACT

During vertebrate cardiac development NOTCH signaling activity in the endocardium is essential for the crosstalk between endocardium and myocardium that initiates ventricular trabeculation and valve primordium formation. This crosstalk leads later to the maturation and compaction of the ventricular chambers and the morphogenesis of the cardiac valves, and its alteration may lead to disease. Although endocardial NOTCH signaling has been shown to be crucial for heart development, its physiological role in the myocardium has not been clearly established. Here we have used mouse genetics to evaluate the role of NOTCH in myocardial development. We have inactivated the unique and ubiquitous NOTCH effector RBPJ in early cardiomyocytes progenitors, and examined its consequences in cardiac development and function. Our results show that mice with Tnnt2-Cre-mediated myocardial-specific deletion of Rbpj develop to term, with homozygous mutant animals showing normal expression of cardiac development markers, and normal adult heart function. Similar observations have been obtained after Notch1 deletion with Tnnt2-Cre. We have also deleted Rbpj in both myocardial and endocardial progenitor cells, using the Nkx2.5-Cre driver, resulting in ventricular septal defect (VSD), double outlet right ventricle (DORV), and bicuspid aortic valve (BAV), due to NOTCH signaling abrogation in the endocardium of cardiac valves. Our data demonstrate that NOTCH-RBPJ inactivation in the myocardium does not affect heart development or adult cardiac function.


Subject(s)
Gene Deletion , Heart Defects, Congenital , Heart/embryology , Myocardium/metabolism , Receptor, Notch1/deficiency , Signal Transduction , rab GTP-Binding Proteins/deficiency , Animals , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Mice , Mice, Knockout , Myocardium/pathology
12.
Genes Dev ; 31(13): 1308-1324, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28779009

ABSTRACT

Sufficient blood flow to tissues relies on arterial blood vessels, but the mechanisms regulating their development are poorly understood. Many arteries, including coronary arteries of the heart, form through remodeling of an immature vascular plexus in a process triggered and shaped by blood flow. However, little is known about how cues from fluid shear stress are translated into responses that pattern artery development. Here, we show that mice lacking endothelial Dach1 had small coronary arteries, decreased endothelial cell polarization, and reduced expression of the chemokine Cxcl12 Under shear stress in culture, Dach1 overexpression stimulated endothelial cell polarization and migration against flow, which was reversed upon CXCL12/CXCR4 inhibition. In vivo, DACH1 was expressed during early arteriogenesis but was down in mature arteries. Mature artery-type shear stress (high, uniform laminar) specifically down-regulated DACH1, while the remodeling artery-type flow (low, variable) maintained DACH1 expression. Together, our data support a model in which DACH1 stimulates coronary artery growth by activating Cxcl12 expression and endothelial cell migration against blood flow into developing arteries. This activity is suppressed once arteries reach a mature morphology and acquire high, laminar flow that down-regulates DACH1. Thus, we identified a mechanism by which blood flow quality balances artery growth and maturation.


Subject(s)
Coronary Vessels/growth & development , Eye Proteins/genetics , Eye Proteins/metabolism , Neovascularization, Physiologic/genetics , Signal Transduction/genetics , Animals , Blood Flow Velocity/physiology , Cell Movement/genetics , Cells, Cultured , Chemokine CXCL12/genetics , Coronary Vessels/physiopathology , Endothelial Cells/cytology , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Mice, Inbred C57BL , Mutation , Organ Culture Techniques , Receptors, CXCR4/genetics , Stress, Mechanical
13.
Cardiovasc Res ; 112(2): 568-580, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27496872

ABSTRACT

AIM: To determine the role of NOTCH during the arterial injury response and the subsequent chronic arterial-wall inflammation underlying atherosclerosis. METHODS AND RESULTS: We have generated a mouse model of endothelial-specific (Cdh5-driven) depletion of the Notch effector recombination signal binding protein for immunoglobulin kappa J region (RBPJ) [(ApoE-/-); homozygous RBPJk conditional mice (RBPJflox/flox); Cadherin 5-CreERT, tamoxifen inducible driver mice (Cdh5-CreERT)]. Endothelial-specific deletion of RBPJ or systemic deletion of Notch1 in athero-susceptible ApoE-/- mice fed a high-cholesterol diet for 6 weeks resulted in reduced atherosclerosis in the aortic arch and sinus. Intravital microscopy revealed decreased leucocyte rolling on the endothelium of ApoE-/-; RBPJflox/flox; Cdh5-CreERT mice, correlating with a lowered content of leucocytes and macrophages in the vascular wall. Transcriptome analysis revealed down-regulation of proinflammatory and endothelial activation pathways in atherosclerotic tissue of RBPJ-mutant mice. During normal Notch activation, Jagged1 signalling up-regulation in endothelial cells promotes nuclear translocation of the Notch1 intracellular domain (N1ICD) and its physical interaction with nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). This N1ICD-NF-κB interaction is required for reciprocal transactivation of target genes, including vascular cell adhesion molecule-1. CONCLUSIONS: Notch signalling pathway inactivation decreases leucocyte rolling, thereby preventing endothelial dysfunction and vascular inflammation. Attenuation of Notch signalling might provide a treatment strategy for atherosclerosis.

14.
FEBS J ; 283(23): 4223-4237, 2016 12.
Article in English | MEDLINE | ID: mdl-27260948

ABSTRACT

The vertebrate heart is the first organ to form and function during embryogenesis. Primitive streak-derived cardiac progenitors located bilaterally move rostral to form the primitive heart tube that subsequently undergoes rightward looping, remodelling and septation to give rise to the mature four-chambered heart. Tightly regulated tissue interactions orchestrate the patterning, proliferation and differentiation processes that give rise to the adult ventricles. Studies in animal models have demonstrated the crucial function of the Notch signalling pathway in ventricular development and how alterations in human NOTCH signalling may lead to disease in the form of cardiomyopathies, such as left ventricular noncompaction (LVNC). In this review, we discuss how during trabecular formation and ventricular compaction, Dll4-Notch1 signals from chamber endocardium to regulate cardiomyocyte proliferation and differentiation in a noncell autonomous fashion and how, at later stages, myocardial Jag1 and Jag2 activate Notch1 in chamber endocardium to sustain chamber patterning and compaction with simultaneous coronary vessel development mediated by Dll4-Notch1. We suggest that alterations in these molecular mechanisms underlie MIB1-related familial LVNC and favour the hypothesis that this cardiomyopathy has a congenital nature.


Subject(s)
Cardiomyopathies/metabolism , Myocardium/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Cardiomyopathies/embryology , Endocardium/embryology , Endocardium/metabolism , Heart Ventricles/embryology , Heart Ventricles/metabolism , Humans , Models, Cardiovascular , Organogenesis
15.
Cell Metab ; 23(5): 881-92, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27166947

ABSTRACT

Heart muscle maintains blood circulation, while skeletal muscle powers skeletal movement. Despite having similar myofibrilar sarcomeric structures, these striated muscles differentially express specific sarcomere components to meet their distinct contractile requirements. The mechanism responsible is still unclear. We show here that preservation of the identity of the two striated muscle types depends on epigenetic repression of the alternate lineage gene program by the chromatin remodeling complex Chd4/NuRD. Loss of Chd4 in the heart triggers aberrant expression of the skeletal muscle program, causing severe cardiomyopathy and sudden death. Conversely, genetic depletion of Chd4 in skeletal muscle causes inappropriate expression of cardiac genes and myopathy. In both striated tissues, mitochondrial function was also dependent on the Chd4/NuRD complex. We conclude that an epigenetic mechanism controls cardiac and skeletal muscle structural and metabolic identities and that loss of this regulation leads to hybrid striated muscle tissues incompatible with life.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Homeostasis , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Muscle, Striated/metabolism , Aging/pathology , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Differentiation/genetics , CpG Islands/genetics , Gene Expression Regulation, Developmental , Heart/embryology , Mice, Transgenic , Mitochondria, Heart/metabolism , Muscle, Striated/embryology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Promoter Regions, Genetic/genetics , Protein Binding
16.
Circ Res ; 118(10): 1480-97, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27056911

ABSTRACT

RATIONALE: The Notch signaling pathway is crucial for primitive cardiac valve formation by epithelial-mesenchymal transition, and NOTCH1 mutations cause bicuspid aortic valve; however, the temporal requirement for the various Notch ligands and receptors during valve ontogeny is poorly understood. OBJECTIVE: The aim of this study is to determine the functional specificity of Notch in valve development. METHODS AND RESULTS: Using cardiac-specific conditional targeted mutant mice, we find that endothelial/endocardial deletion of Mib1-Dll4-Notch1 signaling, possibly favored by Manic-Fringe, is specifically required for cardiac epithelial-mesenchymal transition. Mice lacking endocardial Jag1, Notch1, or RBPJ displayed enlarged valve cusps, bicuspid aortic valve, and septal defects, indicating that endocardial Jag1 to Notch1 signaling is required for post-epithelial-mesenchymal transition valvulogenesis. Valve dysmorphology was associated with increased mesenchyme proliferation, indicating that Jag1-Notch1 signaling restricts mesenchyme cell proliferation non-cell autonomously. Gene profiling revealed upregulated Bmp signaling in Jag1-mutant valves, providing a molecular basis for the hyperproliferative phenotype. Significantly, the negative regulator of mesenchyme proliferation, Hbegf, was markedly reduced in Jag1-mutant valves. Hbegf expression in embryonic endocardial cells could be readily activated through a RBPJ-binding site, identifying Hbegf as an endocardial Notch target. Accordingly, addition of soluble heparin-binding EGF-like growth factor to Jag1-mutant outflow tract explant cultures rescued the hyperproliferative phenotype. CONCLUSIONS: During cardiac valve formation, Dll4-Notch1 signaling leads to epithelial-mesenchymal transition and cushion formation. Jag1-Notch1 signaling subsequently restrains Bmp-mediated valve mesenchyme proliferation by sustaining Hbegf-EGF receptor signaling. Our studies identify a mechanism of signaling cross talk during valve morphogenesis involved in the origin of congenital heart defects associated with reduced NOTCH function.


Subject(s)
Mitral Valve/metabolism , Morphogenesis , Receptor, Notch1/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitral Valve/abnormalities , Mitral Valve/embryology , Receptor, Notch1/metabolism , Up-Regulation
17.
Circ Res ; 118(1): e1-e18, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26635389

ABSTRACT

The Notch signaling pathway is an ancient and highly conserved signaling pathway that controls cell fate specification and tissue patterning in the embryo and in the adult. Region-specific endocardial Notch activity regulates heart morphogenesis through the interaction with multiple myocardial-, epicardial-, and neural crest-derived signals. Mutations in NOTCH signaling elements cause congenital heart disease in humans and mice, demonstrating its essential role in cardiac development. Studies in model systems have provided mechanistic understanding of Notch function in cardiac development, congenital heart disease, and heart regeneration. Notch patterns the embryonic endocardium into prospective territories for valve and chamber formation, and later regulates the signaling processes leading to outflow tract and valve morphogenesis and ventricular trabeculae compaction. Alterations in NOTCH signaling in the endocardium result in congenital structural malformations that can lead to disease in the neonate and adult heart.


Subject(s)
Endocardium/growth & development , Endocardium/metabolism , Heart Diseases/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Endocardium/pathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Diseases/pathology , Humans
18.
Nat Cell Biol ; 18(1): 7-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26641715

ABSTRACT

Ventricular chambers are essential for the rhythmic contraction and relaxation occurring in every heartbeat throughout life. Congenital abnormalities in ventricular chamber formation cause severe human heart defects. How the early trabecular meshwork of myocardial fibres forms and subsequently develops into mature chambers is poorly understood. We show that Notch signalling first connects chamber endocardium and myocardium to sustain trabeculation, and later coordinates ventricular patterning and compaction with coronary vessel development to generate the mature chamber, through a temporal sequence of ligand signalling determined by the glycosyltransferase manic fringe (MFng). Early endocardial expression of MFng promotes Dll4-Notch1 signalling, which induces trabeculation in the developing ventricle. Ventricular maturation and compaction require MFng and Dll4 downregulation in the endocardium, which allows myocardial Jag1 and Jag2 signalling to Notch1 in this tissue. Perturbation of this signalling equilibrium severely disrupts heart chamber formation. Our results open a new research avenue into the pathogenesis of cardiomyopathies.


Subject(s)
Heart Ventricles/metabolism , Organogenesis/physiology , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing , Calcium-Binding Proteins/metabolism , Cells, Cultured , Glucosyltransferases , Heart Ventricles/embryology , Hexosyltransferases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology
19.
Nat Med ; 19(2): 193-201, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23314057

ABSTRACT

Left ventricular noncompaction (LVNC) causes prominent ventricular trabeculations and reduces cardiac systolic function. The clinical presentation of LVNC ranges from asymptomatic to heart failure. We show that germline mutations in human MIB1 (mindbomb homolog 1), which encodes an E3 ubiquitin ligase that promotes endocytosis of the NOTCH ligands DELTA and JAGGED, cause LVNC in autosomal-dominant pedigrees, with affected individuals showing reduced NOTCH1 activity and reduced expression of target genes. Functional studies in cells and zebrafish embryos and in silico modeling indicate that MIB1 functions as a dimer, which is disrupted by the human mutations. Targeted inactivation of Mib1 in mouse myocardium causes LVNC, a phenotype mimicked by inactivation of myocardial Jagged1 or endocardial Notch1. Myocardial Mib1 mutants show reduced ventricular Notch1 activity, expansion of compact myocardium to proliferative, immature trabeculae and abnormal expression of cardiac development and disease genes. These results implicate NOTCH signaling in LVNC and indicate that MIB1 mutations arrest chamber myocardium development, preventing trabecular maturation and compaction.


Subject(s)
Cardiomyopathies/etiology , Heart Ventricles , Mutation , Receptors, Notch/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/genetics , Amino Acid Sequence , Animals , Cardiomyopathies/genetics , Female , HEK293 Cells , Heart/embryology , Heart Ventricles/embryology , Humans , Male , Mice , Molecular Sequence Data , Protein Multimerization , Ubiquitin-Protein Ligases/physiology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...