Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Rep ; 13(1): 183-195, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26411676

ABSTRACT

The homeobox transcription factors NKX2-5 and MEIS1 are essential for vertebrate heart development and normal physiology of the adult heart. We show that, during cardiac differentiation, the two transcription factors have partially overlapping expression patterns, with the result that as cardiac progenitors from the anterior heart field differentiate and migrate into the cardiac outflow tract, they sequentially experience high levels of MEIS1 and then increasing levels of NKX2-5. Using the Popdc2 gene as an example, we also show that a significant proportion of target genes for NKX2-5 contain a binding motif recognized by NKX2-5, which overlaps with a binding site for MEIS1. Binding of the two factors to such overlapping sites is mutually exclusive, and this provides a simple regulatory mechanism for spatial and temporal synchronization of a common pool of targets between NKX2-5 and MEIS1.


Subject(s)
Cell Adhesion Molecules/metabolism , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Neoplasm Proteins/metabolism , Organogenesis/genetics , Transcription Factors/metabolism , Animals , Binding Sites , Cell Adhesion Molecules/genetics , Embryo, Mammalian , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle Proteins/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Nucleotide Motifs , Protein Binding , Signal Transduction , Transcription Factors/genetics , Troponin/genetics , Troponin/metabolism , Troponin I/genetics , Troponin I/metabolism
2.
PLoS Biol ; 11(9): e1001666, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24086110

ABSTRACT

Cardiomyocytes are vulnerable to hypoxia in the adult, but adapted to hypoxia in utero. Current understanding of endogenous cardiac oxygen sensing pathways is limited. Myocardial oxygen consumption is determined by regulation of energy metabolism, which shifts from glycolysis to lipid oxidation soon after birth, and is reversed in failing adult hearts, accompanying re-expression of several "fetal" genes whose role in disease phenotypes remains unknown. Here we show that hypoxia-controlled expression of the transcription factor Hand1 determines oxygen consumption by inhibition of lipid metabolism in the fetal and adult cardiomyocyte, leading to downregulation of mitochondrial energy generation. Hand1 is under direct transcriptional control by HIF1α. Transgenic mice prolonging cardiac Hand1 expression die immediately following birth, failing to activate the neonatal lipid metabolising gene expression programme. Deletion of Hand1 in embryonic cardiomyocytes results in premature expression of these genes. Using metabolic flux analysis, we show that Hand1 expression controls cardiomyocyte oxygen consumption by direct transcriptional repression of lipid metabolising genes. This leads, in turn, to increased production of lactate from glucose, decreased lipid oxidation, reduced inner mitochondrial membrane potential, and mitochondrial ATP generation. We found that this pathway is active in adult cardiomyocytes. Up-regulation of Hand1 is protective in a mouse model of myocardial ischaemia. We propose that Hand1 is part of a novel regulatory pathway linking cardiac oxygen levels with oxygen consumption. Understanding hypoxia adaptation in the fetal heart may allow development of strategies to protect cardiomyocytes vulnerable to ischaemia, for example during cardiac ischaemia or surgery.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Energy Metabolism/genetics , Lipid Metabolism/genetics , Myocardium/metabolism , Oxygen Consumption/genetics , Adenosine Triphosphate/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia/genetics , Cell Line , Gene Expression Regulation, Developmental , Heart/embryology , Heart/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Transgenic , Mitochondria/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Transcriptional Activation
3.
Dev Biol ; 333(1): 121-31, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19563797

ABSTRACT

Tbx2 is a member of the T-box family of transcription factors that play important roles during heart development. In the embryonic heart tube, Tbx2 is expressed in non-chamber myocardium (outflow tract and interventricular canal) and has been shown to block chamber formation. We have developed a genetic system to conditionally misexpress Tbx2 in the embryonic mouse heart at early stages of development. We show that Tbx2 expression throughout the myocardium of the heart tube both represses proliferation and impairs secondary heart field (SHF) progenitor cell deployment into the outflow tract (OFT). Repression of proliferation is accompanied by the upregulation of Ndrg2 and downregulation of Ndrg4 expression, both genes believed to be involved in cell growth and proliferation. Impaired deployment of SHF cells from the pharyngeal mesoderm is accompanied by downregulation of the cell adhesion molecules Alcam and N-cadherin in the anterior part of the embryonic heart. Tbx2 misexpression also results in downregulation of Tbx20 within the OFT, indicating complex and region-specific transcriptional cross-regulation between the two T-box genes.


Subject(s)
Cell Differentiation/physiology , Heart/embryology , Myocardium/cytology , Stem Cells/cytology , T-Box Domain Proteins/biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Body Patterning , Cadherins/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Myocardium/metabolism , Nerve Tissue Proteins/metabolism , Proteins/metabolism , Stem Cells/physiology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
4.
BMC Dev Biol ; 8: 74, 2008 Jul 28.
Article in English | MEDLINE | ID: mdl-18662378

ABSTRACT

BACKGROUND: GATA factors 4/5/6 have been implicated in the development of the heart and endodermal derivatives in vertebrates. Work in zebrafish has indicated that GATA5 is required for normal development earlier than GATA4/6. However, the GATA5 knockout mouse has no apparent embryonic phenotype, thereby questioning the importance of the gene for vertebrate development. RESULTS: In this study we show that in Xenopus embryos GATA5 is essential for early development of heart and liver precursors. In addition, we have found that in Xenopus embryos GATA4 is important for development of heart and liver primordia following their specification, and that in this role it might interact with GATA6. CONCLUSION: Our results suggest that GATA5 acts earlier than GATA4 to regulate development of heart and liver precursors, and indicate that one early direct target of GATA5 is homeobox gene Hex.


Subject(s)
GATA4 Transcription Factor/physiology , GATA5 Transcription Factor/physiology , Heart/embryology , Liver/embryology , Xenopus Proteins/physiology , Xenopus/embryology , Animals , Zinc Fingers
5.
Dev Dyn ; 236(8): 2159-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17615576

ABSTRACT

Harnessing toxic proteins to destroy selective cells in an embryo is an attractive method for exploring details of cell fate and cell-cell interdependency. However, no existing "suicide gene" system has proved suitable for aquatic vertebrates. We use the M2(H37A) toxic ion channel of the influenza-A virus to induce cell-ablations in Xenopus laevis. M2(H37A) RNA injected into blastomeres of early stage embryos causes death of their progeny by late-blastula stages. Moreover, M2(H37A) toxicity can be controlled using the M2 inhibitor rimantadine. We have tested the ablation system using transgenesis to target M2(H37A) expression to selected cells in the embryo. Using the myocardial MLC2 promoter, M2(H37A)-mediated cell death causes dramatic loss of cardiac structure and function by stage 39. With the LURP1 promoter, we induce cell-ablations of macrophages. These experiments demonstrate the effectiveness of M2(H37A)-ablation in Xenopus and its utility in monitoring the progression of developmental abnormalities during targeted cell death experiments.


Subject(s)
Cell Death/drug effects , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Toxins, Biological/pharmacology , Viral Proteins/toxicity , Animals , Gene Transfer Techniques , Influenza A virus/chemistry , Ion Channels/toxicity , Methods , Toxins, Biological/genetics , Xenopus laevis
6.
Genesis ; 45(3): 135-44, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17334998

ABSTRACT

Mouse-lines expressing Cre recombinase in a tissue-specific manner are a powerful tool in developmental biology. Here, we report that a 3 kb fragment of the Xenopus laevis myosin light-chain 2 (XMLC2) promoter drives Cre recombinase expression in a cardiac-restricted fashion in the mouse embryo. We have isolated two XMLC2-Cre lines that express recombinase exclusively within cardiomyocytes, from the onset of their differentiation in the cardiac crescent of the early embryo. Expression is maintained throughout the myocardium of the embryonic heart tube and subsequently the mature myocardium of the chambered heart. Recombinase activity is detected in all myocardial tissue, including the pulmonary veins. One XMLC2-Cre line shows uniform expression while the other only expresses recombinase in a mosaic fashion encompassing less than 50% of the myocardial cells. Both lines cause severe cardiac malformations when crossed to a conditional Tbx5 line, resulting in embryonic death at midgestation. Optical projection tomography reveals that the spectrum of developmental abnormalities includes a shortening of the outflow tract and its abnormal alignment, along with a dramatic reduction in trabeculation of the ventricular segment of the looping heart tube.


Subject(s)
Gene Expression Regulation, Developmental , Heart/embryology , Integrases/metabolism , Myocardium/enzymology , Animals , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Heart Defects, Congenital/chemically induced , Integrases/genetics , Mice , Mice, Transgenic , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , T-Box Domain Proteins/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
7.
Dev Dyn ; 232(4): 1003-12, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15736168

ABSTRACT

Many details of cardiac chamber morphogenesis could be revealed if muscle fiber development could be visualized directly within the hearts of living vertebrate embryos. To achieve this end, we have used the active promoter of the MLC1v gene to drive expression of green fluorescent protein (GFP) in the developing tadpole heart. By using a line of Xenopus laevis frogs transgenic for the MLC1v-EGFP reporter, we have observed regionalized patterns of muscle formation within the ventricular chamber and maturation of the atrial chambers, from the onset of chamber formation through to the adult frog. In f1 generation MLC1v-EGFP animals, promoter activity is first detected within the looping heart tube and delineates the forming ventricular chamber and proximal outflow tract throughout their development. The 8-kb MLC1v promoter faithfully reproduces the embryonic expression of the endogenous MLC1v mRNA. At later larval stages, weak patches of EGFP fluorescence are found on the atrial side of the atrioventricular boundary. Subsequently, an extensive lattice of MLC1v-expressing fibers extend across the mature atrial chambers of adult frog hearts and the transgene reveals the differing arrangement of muscle fibers in chamber versus outflow myocardium. The complete activity of the promoter resides within the proximal 4.5 kb of the MLC1v DNA fragment, whereas key elements regulating chamber-specific expression are present in the proximal-most 1.5 kb. Finally, we demonstrate how cardiac and craniofacial muscle expression of the MLC1v promoter can be used to diagnose mutant phenotypes in living embryos, using the injection of RNA encoding a Tbx1-engrailed repressor-fusion protein as an example.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Heart Ventricles/embryology , Myocardium/metabolism , Myosin Light Chains/biosynthesis , Xenopus Proteins/biosynthesis , Xenopus/genetics , Animals , Animals, Genetically Modified , Genetic Markers/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Myosin Light Chains/genetics , Xenopus Proteins/genetics
8.
Development ; 131(3): 669-79, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14711876

ABSTRACT

The mechanisms by which transcription factors, which are not themselves tissue restricted, establish cardiomyocyte-specific patterns of transcription in vivo are unknown. Nor do we understand how positional cues are integrated to provide regionally distinct domains of gene expression within the developing heart. We describe regulation of the Xenopus XMLC2 gene, which encodes a regulatory myosin light chain of the contractile apparatus in cardiac muscle. This gene is expressed from the onset of cardiac differentiation in the frog embryo and is expressed throughout all the myocardium, both before and after heart chamber formation. Using transgenesis in frog embryos, we have identified an 82 bp enhancer within the proximal promoter region of the gene that is necessary and sufficient for heart-specific expression of an XMLC2 transgene. This enhancer is composed of two GATA sites and a composite YY1/CArG-like site. We show that the low-affinity SRF site is essential for transgene expression and that cardiac-specific expression also requires the presence of at least one adjacent GATA site. The overlapping YY1 site within the enhancer appears to act primarily as a repressor of ectopic expression, although it may also have a positive role. Finally, we show that the frog MLC2 promoter drives pan myocardial expression of a transgene in mice, despite the more restricted patterns of expression of murine MLC2 genes. We speculate that a common regulatory mechanism may be responsible for pan-myocardial expression of XMLC2 in both the frog and mouse, modulation of which could have given rise to more restricted patterns of expression within the heart of higher vertebrates.


Subject(s)
Cardiac Myosins/physiology , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental/physiology , Heart/embryology , Myosin Light Chains/physiology , Animals , Base Sequence , Binding Sites , Cardiac Myosins/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Heart/physiology , Molecular Sequence Data , Myocardium/metabolism , Myosin Light Chains/genetics , Promoter Regions, Genetic , Xenopus
9.
Development ; 130(16): 3865-76, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12835401

ABSTRACT

The earliest step in heart formation in vertebrates occurs during gastrulation, when cardiac tissue is specified. Dorsoanterior endoderm is thought to provide a signal that induces adjacent mesodermal cells to adopt a cardiac fate. However, the nature of this signalling and the precise role of endoderm are unknown because of the close proximity and interdependence of mesoderm and endoderm during gastrulation. To better define the molecular events that underlie cardiac induction, we have sought to develop a simple means of inducing cardiac tissue. We show that the transcription factor GATA4, which has been implicated in regulating cardiac gene expression, is sufficient to induce cardiac differentiation in Xenopus embryonic ectoderm (animal pole) explants, frequently resulting in beating tissue. Lineage labelling experiments demonstrate that GATA4 can trigger cardiac differentiation not only in cells in which it is present, but also in neighbouring cells. Surprisingly, cardiac differentiation can occur without any stable differentiation of anterior endoderm and is in fact enhanced under conditions in which endoderm formation is inhibited. Remarkably, cardiac tissue is formed even when GATA4 activity is delayed until long after explants have commenced differentiation into epidermal tissue. These findings provide a simple assay system for cardiac induction that may allow elucidation of pathways leading to cardiac differentiation. Better knowledge of the pathways governing this process may help develop procedures for efficient generation of cardiomyocytes from pluripotent stem cells.


Subject(s)
DNA-Binding Proteins/metabolism , Ectoderm/metabolism , Embryonic Induction , Heart/embryology , High Mobility Group Proteins , Myocytes, Cardiac/physiology , Transcription Factors/metabolism , Xenopus Proteins , Xenopus laevis/embryology , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/metabolism , Culture Techniques , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , Dishevelled Proteins , Ectoderm/cytology , GATA4 Transcription Factor , Gastrula/physiology , Gene Expression Regulation, Developmental , Muscle, Skeletal/physiology , Myocytes, Cardiac/cytology , Nodal Protein , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , SOXF Transcription Factors , Signal Transduction/physiology , Trans-Activators/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Wnt Proteins , beta Catenin
10.
Mech Dev ; 117(1-2): 173-86, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12204257

ABSTRACT

Phagocytic myeloid cells provide the principle line of immune defence during early embryogenesis in lower vertebrates. They may also have important functions during normal embryo morphogenesis, not least through the phagocytic clearance of cell corpses arising from apoptosis. We have identified two cDNAs that provide sensitive molecular markers of embryonic leukocytes in the early Xenopus embryo. These encode a peroxidase (XPOX2) and a Ly-6/uPAR-related protein (XLURP-1). We show that myeloid progenitors can first be detected at an antero-ventral site in early tailbud stage embryos (a region previously termed the anterior ventral blood island) and transiently express the haematopoetic transcription factors SCL and AML. Phagocytes migrate from this site along consistent routes and proliferate, becoming widely distributed throughout the tadpole long before the circulatory system is established. This migration can be followed in living embryos using a 5 kb portion of the XLURP-1 promoter to drive expression of EGFP specifically in the myeloid cells. Interestingly, whilst much of this migration occurs by movement of individual cells between embryonic germ layers, the rostral-most myeloid cells apparently migrate in an anterior direction along the ventral midline within the mesodermal layer itself. The transient presence of such cells as a strip bisecting the cardiac mesoderm immediately prior to heart tube formation suggests that embryonic myeloid cells may play a role in early cardiac morphogenesis.


Subject(s)
Mannose-Binding Lectins , Membrane Glycoproteins/genetics , Peroxidase/genetics , Receptors, Cell Surface/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Heart/embryology , In Situ Hybridization , Molecular Sequence Data , Myelopoiesis/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Transcription Factors/genetics , Xenopus laevis/metabolism
11.
Dev Biol ; 245(1): 57-70, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11969255

ABSTRACT

During vertebrate embryonic development, cardiac and skeletal muscle originates from distinct precursor populations. Despite the profound structural and functional differences in the striated muscle tissue they eventually form, such progenitors share many features such as components of contractile apparatus. In vertebrate embryos, the alpha-cardiac actin gene encodes a major component of the myofibril in both skeletal and cardiac muscle. Here, we show that expression of Xenopus cardiac alpha-actin in the myotomes and developing heart tube of the tadpole requires distinct enhancers within its proximal promoter. Using transgenic embryos, we find that mutations in the promoter-proximal CArG box and 5 bp downstream of it specifically eliminate expression of a GFP transgene within the developing heart, while high levels of expression in somitic muscle are maintained. This sequence is insufficient on its own to limit expression solely to the myocardium, such restriction requiring multiple elements within the proximal promoter. Two additional enhancers are active in skeletal muscle of the embryo, either one of which has to interact with the proximal CArG box for correct expression to be established. Transgenic reporters containing multimerised copies of CArG box 1 faithfully detect most sites of SRF expression in the developing embryo as do equivalent reporters containing the SRF binding site from the c-fos promoter. Significantly, while these motifs possess a different A/T core within the CC(A/T)(6)GG consensus and show no similarity in flanking sequence, each can interact with a myotome-specific distal enhancer of cardiac alpha-actin promoter, to confer appropriate cardiac alpha-actin-specific regulation of transgene expression. Together, these results suggest that the role of CArG box 1 in the cardiac alpha-actin gene promoter is to act solely as a high-affinity SRF binding site.


Subject(s)
Actins/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental/genetics , Heart/embryology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Animals , Base Sequence , DNA Primers , Electrophoretic Mobility Shift Assay , Genes, fos , Muscle, Skeletal/embryology , Promoter Regions, Genetic , Xenopus/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...