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










Publication year range
1.
Methods Mol Biol ; 2198: 193-208, 2021.
Article in English | MEDLINE | ID: mdl-32822033

ABSTRACT

5-methylcytosine (5mC) is an epigenetic modification to DNA which modulates transcription. 5mC can be sequentially oxidized to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC). Collectively, these marks are referred to as the oxidized derivatives of 5mC (i.e., oxi-mCs). Their formation is catalyzed by the ten-eleven translocation methylcytosine dioxygenases (TETs 1, 2 and 3). Various techniques have been developed for the detection of oxi-mCs. The following chapter describes an immunochemical protocol for the simultaneous detection of 5hmC and 5caC in embryonic zebrafish tissue sections. The embryos are fixed, permeabilized and embedded in paraffin blocks. The blocks are cut into sections that are mounted onto slides. Depurination of the DNA is performed to allow immunodetection of the oxi-mCs. The 5hmC is detected with the help of a mouse anti-5hmC monoclonal primary antibody and a goat anti-mouse Alexa Fluor 633-conjugated secondary antibody. The weak 5caC signal requires enzymatic amplification. Its detection involves a rabbit anti-5caC polyclonal primary antibody and a goat anti-rabbit secondary antibody that is conjugated to horseradish peroxidase (HRP). HRP amplifies the 5caC signal by catalyzing the deposition of large quantities of fluorescein-labeled tyramide. Sections immunostained for 5hmC and 5caC are analyzed by fluorescent light or confocal laser scanning microscopy. This immunochemical method allows for highly sensitive detection of 5hmC and 5caC in zebrafish tissues.


Subject(s)
5-Methylcytosine/analogs & derivatives , DNA/immunology , Immunohistochemistry/methods , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Animals , Antibodies/metabolism , Cell Nucleus/metabolism , Cytosine/analogs & derivatives , DNA/genetics , DNA Methylation , Dioxygenases , Embryo, Nonmammalian , Zebrafish
2.
Blood Adv ; 2(20): 2589-2606, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30309860

ABSTRACT

The transcriptional repressors Gfi1(a) and Gfi1b are epigenetic regulators with unique and overlapping roles in hematopoiesis. In different contexts, Gfi1 and Gfi1b restrict or promote cell proliferation, prevent apoptosis, influence cell fate decisions, and are essential for terminal differentiation. Here, we show in primitive red blood cells (prRBCs) that they can also set the pace for cellular differentiation. In zebrafish, prRBCs express 2 of 3 zebrafish Gfi1/1b paralogs, Gfi1aa and Gfi1b. The recently identified zebrafish gfi1aa gene trap allele qmc551 drives erythroid green fluorescent protein (GFP) instead of Gfi1aa expression, yet homozygous carriers have normal prRBCs. prRBCs display a maturation defect only after splice morpholino-mediated knockdown of Gfi1b in gfi1aa qmc551 homozygous embryos. To study the transcriptome of the Gfi1aa/1b double-depleted cells, we performed an RNA-Seq experiment on GFP-positive prRBCs sorted from 20-hour-old embryos that were heterozygous or homozygous for gfi1aa qmc551 , as well as wt or morphant for gfi1b We subsequently confirmed and extended these data in whole-mount in situ hybridization experiments on newly generated single- and double-mutant embryos. Combined, the data showed that in the absence of Gfi1aa, the synchronously developing prRBCs were delayed in activating late erythroid differentiation, as they struggled to suppress early erythroid and endothelial transcription programs. The latter highlighted the bipotent nature of the progenitors from which prRBCs arise. In the absence of Gfi1aa, Gfi1b promoted erythroid differentiation as stepwise loss of wt gfi1b copies progressively delayed Gfi1aa-depleted prRBCs even further, showing that Gfi1aa and Gfi1b together set the pace for prRBC differentiation from hemangioblasts.


Subject(s)
DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Erythroblasts/metabolism , Hemangioblasts/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Zebrafish
3.
Front Cell Dev Biol ; 6: 27, 2018.
Article in English | MEDLINE | ID: mdl-29616219

ABSTRACT

5-methylcytosine (5mC) is the best understood DNA modification and is generally believed to be associated with repression of gene expression. Over the last decade, sequentially oxidized forms of 5mC (oxi-mCs) have been discovered within the genomes of vertebrates. Their discovery was accompanied by that of the ten-eleven translocation (TET) methylcytosine dioxygenases, the enzymes that catalyze the formation of the oxi-mCs. Although a number of studies performed on different vertebrate models and embryonic stem cells demonstrated that both TET enzymes and oxi-mCs are likely to be important for several developmental processes it is currently unclear whether their developmental roles are conserved among vertebrates. Here, we summarize recent developments in this field suggesting that biological roles of TETs/oxi-mCs may significantly differ between mice and zebrafish. Thus, although the role of TET proteins in late organogenesis has been documented for both these systems; unlike in mice the enzymatic oxidation of 5mC does not seem to be involved in zygotic reprogramming or gastrulation in zebrafish. Our analysis may provide an insight into the general principles of epigenetic regulation of animal development and cellular differentiation.

4.
Curr Biol ; 27(6): 784-794, 2017 Mar 20.
Article in English | MEDLINE | ID: mdl-28262487

ABSTRACT

Axons require the axonal NAD-synthesizing enzyme NMNAT2 to survive. Injury or genetically induced depletion of NMNAT2 triggers axonal degeneration or defective axon growth. We have previously proposed that axonal NMNAT2 primarily promotes axon survival by maintaining low levels of its substrate NMN rather than generating NAD; however, this is still debated. NMN deamidase, a bacterial enzyme, shares NMN-consuming activity with NMNAT2, but not NAD-synthesizing activity, and it delays axon degeneration in primary neuronal cultures. Here we show that NMN deamidase can also delay axon degeneration in zebrafish larvae and in transgenic mice. Like overexpressed NMNATs, NMN deamidase reduces NMN accumulation in injured mouse sciatic nerves and preserves some axons for up to three weeks, even when expressed at a low level. Remarkably, NMN deamidase also rescues axonal outgrowth and perinatal lethality in a dose-dependent manner in mice lacking NMNAT2. These data further support a pro-degenerative effect of accumulating NMN in axons in vivo. The NMN deamidase mouse will be an important tool to further probe the mechanisms underlying Wallerian degeneration and its prevention.


Subject(s)
Amidohydrolases/genetics , Axons/pathology , Nerve Degeneration/genetics , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Wallerian Degeneration/genetics , Amidohydrolases/metabolism , Animals , Mice , Mice, Transgenic , Nerve Degeneration/metabolism , Wallerian Degeneration/metabolism
5.
Dev Biol ; 417(1): 25-39, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27432513

ABSTRACT

A transposon-mediated gene trap screen identified the zebrafish line qmc551 that expresses a GFP reporter in primitive erythrocytes and also in haemogenic endothelial cells, which give rise to haematopoietic stem and progenitor cells (HSPCs) that seed sites of larval and adult haematopoiesis. The transposon that mediates this GFP expression is located in intron 1 of the gfi1aa gene, one of three zebrafish paralogs that encode transcriptional repressors homologous to mammalian Gfi1 and Gfi1b proteins. In qmc551 transgenics, GFP expression is under the control of the endogenous gfi1aa promoter, recapitulates early gfi1aa expression and allows live observation of gfi1aa promoter activity. While the transposon integration interferes with the expression of gfi1aa mRNA in haematopoietic cells, homozygous qmc551 fish are viable and fertile, and display normal primitive and definitive haematopoiesis. Retained expression of Gfi1b in primitive erythrocytes and up-regulation of Gfi1ab at the onset of definitive haematopoiesis in homozygous qmc551 carriers, are sufficient to allow normal haematopoiesis. This finding contradicts previously published morpholino data that suggested an essential role for zebrafish Gfi1aa in primitive erythropoiesis.


Subject(s)
DNA Transposable Elements/genetics , DNA-Binding Proteins/biosynthesis , Erythrocytes/cytology , Erythropoiesis/genetics , Hematopoietic Stem Cells/cytology , Zebrafish Proteins/biosynthesis , Zebrafish/embryology , Animals , Animals, Genetically Modified , Base Sequence , Cell Differentiation , Core Binding Factor Alpha 2 Subunit/biosynthesis , DNA-Binding Proteins/genetics , Erythrocytes/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , Receptors, Notch/genetics , Receptors, Notch/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Cell Rep ; 13(11): 2539-2552, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26686637

ABSTRACT

Axon injury leads to rapid depletion of NAD-biosynthetic enzyme NMNAT2 and high levels of its substrate, NMN. We proposed a key role for NMN in Wallerian degeneration but downstream events and their relationship to other mediators remain unclear. Here, we show, in vitro and in vivo, that axotomy leads to a late increase in intra-axonal Ca(2+), abolished by pharmacological or genetic reduction of NMN levels. NMN requires the pro-degenerative protein SARM1 to stimulate Ca(2+) influx and axon degeneration. While inhibition of NMN synthesis and SARM1 deletion block Ca(2+) rise and preserve axonal integrity, they fail to prevent early mitochondrial dynamic changes. Furthermore, depolarizing mitochondria does not alter the rate of Wallerian degeneration. These data reveal that NMN and SARM1 act in a common pathway culminating in intra-axonal Ca(2+) increase and fragmentation and dissociate mitochondrial dysfunctions from this pathway, elucidating which steps may be most effective as targets for therapy.


Subject(s)
Armadillo Domain Proteins/genetics , Calcium/metabolism , Cytoskeletal Proteins/genetics , Mitochondria/metabolism , Nicotinamide Mononucleotide/metabolism , Acrylamides/pharmacology , Amidohydrolases/metabolism , Animals , Armadillo Domain Proteins/deficiency , Axons/drug effects , Axons/metabolism , Cytoskeletal Proteins/deficiency , Ions/chemistry , Ions/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinamide Mononucleotide/pharmacology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Piperidines/pharmacology , Wallerian Degeneration/pathology
7.
Cardiovasc Res ; 100(2): 252-61, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23812297

ABSTRACT

AIMS: The contribution of blood flow to angiogenesis is incompletely understood. We examined the effect of blood flow on Notch signalling in the vasculature of zebrafish embryos, and whether blood flow regulates angiogenesis in zebrafish with constitutively up-regulated hypoxic signalling. METHODS AND RESULTS: Developing zebrafish (Danio rerio) embryos survive via diffusion in the absence of circulation induced by knockdown of cardiac troponin T2 or chemical cardiac cessation. The absence of blood flow increased vascular Notch signalling in 48 h post-fertilization old embryos via up-regulation of the Notch ligand dll4. Despite this, patterning of the intersegmental vessels is not affected by absent blood flow. We therefore examined homozygous vhl mutant zebrafish that have constitutively up-regulated hypoxic signalling. These display excessive and aberrant angiogenesis from 72 h post-fertilization, with significantly increased endothelial number, vessel diameter, and length. The absence of blood flow abolished these effects, though normal vessel patterning was preserved. CONCLUSION: We show that blood flow suppresses vascular Notch signalling via down-regulation of dll4. We have also shown that blood flow is required for angiogenesis in response to hypoxic signalling but is not required for normal vessel patterning. These data indicate important differences in hypoxia-driven vs. developmental angiogenesis.


Subject(s)
Blood Circulation/physiology , Hypoxia/physiopathology , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Neovascularization, Physiologic , Receptors, Notch/physiology , Signal Transduction/physiology , Animals , Diacetyl/analogs & derivatives , Diacetyl/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Tumor Suppressor Proteins/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology , Zebrafish/embryology , Zebrafish Proteins/physiology
8.
PLoS One ; 8(6): e65170, 2013.
Article in English | MEDLINE | ID: mdl-23755189

ABSTRACT

The importance of microRNAs in development is now widely accepted. However, identifying the specific targets of individual microRNAs and understanding their biological significance remains a major challenge. We have used the zebrafish model system to evaluate the expression and function of microRNAs potentially involved in muscle development and study their interaction with predicted target genes. We altered expression of the miR-30 microRNA family and generated phenotypes that mimicked misregulation of the Hedgehog pathway. Inhibition of the miR-30 family increases activity of the pathway, resulting in elevated ptc1 expression and increased numbers of superficial slow-muscle fibres. We show that the transmembrane receptor smoothened is a target of this microRNA family. Our results indicate that fine coordination of smoothened activity by the miR-30 family allows the correct specification and differentiation of distinct muscle cell types during zebrafish embryonic development.


Subject(s)
Hedgehog Proteins/metabolism , MicroRNAs/genetics , Muscle Development/genetics , Receptors, G-Protein-Coupled/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , 3' Untranslated Regions , Animals , Base Sequence , Binding Sites , Body Patterning/genetics , Gene Expression Regulation, Developmental , Membrane Proteins , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Patched Receptors , Patched-1 Receptor , RNA Interference , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Smoothened Receptor , Zebrafish/genetics , Zebrafish Proteins/metabolism
9.
Arterioscler Thromb Vasc Biol ; 33(6): 1257-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23559631

ABSTRACT

OBJECTIVE: Coarctation of the aorta is rarely associated with known gene defects. Blomstrand chondrodysplasia, caused by mutations in the parathyroid hormone receptor 1 (PTHR1) is associated with coarctation of the aorta in some cases, although it is unclear whether PTHR1 deficiency causes coarctation of the aorta directly. The zebrafish allows the study of vascular development using approaches not possible in other models. We therefore examined the effect of loss of function of PTHR1 or its ligand parathyroid hormone-related peptide (PTHrP) on aortic formation in zebrafish. APPROACH AND RESULTS: Morpholino antisense oligonucleotide knockdown of either PTHR1 or PTHrP led to a localized occlusion of the mid-aorta in developing zebrafish. Confocal imaging of transgenic embryos showed that these defects were caused by loss of endothelium, rather than failure to lumenize. Using a Notch reporter transgenic ([CSL:Venus]qmc61), we found both PTHR1 and PTHrP knockdown-induced defective Notch signaling in the hypochord at the site of the aortic defect before onset of circulation, and the aortic occlusion was rescued by inducible Notch upregulation. CONCLUSIONS: Loss of function of either PTHR1 or PTHrP leads to a localized aortic defect that is Notch dependent. These findings may underlie the aortic defect seen in Blomstrand chondrodysplasia, and reveal a link between parathyroid hormone and Notch signaling during aortic development.


Subject(s)
Aorta/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Receptor, Notch1/genetics , Receptor, Parathyroid Hormone, Type 1/genetics , Signal Transduction/genetics , Zebrafish Proteins/genetics , Animals , Aortic Coarctation/genetics , Aortic Coarctation/physiopathology , Female , Male , Models, Animal , Mutation/genetics , Neovascularization, Physiologic/genetics , Reference Values , Up-Regulation , Zebrafish
10.
Epigenetics ; 7(4): 383-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22419071

ABSTRACT

5-hydroxymethyl-cytosine (5-hmC) is a cytosine modification that is relatively abundant in mammalian pre-implantation embryos and embryonic stem cells (ESC) derived from mammalian blastocysts. Recent observations imply that both 5-hmC and Tet1/2/3 proteins, catalyzing the conversion of 5-methyl-cytosine to 5-hmC, may play an important role in self renewal and differentiation of ESCs. Here we assessed the distribution of 5-hmC in zebrafish and chick embryos and found that, unlike in mammals, 5-hmC is immunochemically undetectable in these systems before the onset of organogenesis. In addition, Tet1/2/3 transcripts are either low or undetectable at corresponding stages of zebrafish development. However, 5-hmC is enriched in later zebrafish and chick embryos and exhibits tissue-specific distribution in adult zebrafish. Our findings show that 5-hmC enrichment of non-committed cells is not a universal feature of vertebrate development and give insights both into evolution of embryonic pluripotency and the potential role of 5-hmC in its regulation.


Subject(s)
Cytosine/analogs & derivatives , Gene Expression Regulation, Developmental , Vertebrates/embryology , Zebrafish/metabolism , 5-Methylcytosine/metabolism , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation , Chick Embryo , Culture Media, Conditioned , Cytosine/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryonic Development , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Immunohistochemistry , Male , Mice , Organogenesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Time Factors , Vertebrates/genetics , Vertebrates/metabolism , Zebrafish/embryology , Zebrafish/genetics
11.
Blood ; 116(12): 2046-56, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20511544

ABSTRACT

Hematopoietic stem cells (HSCs) are essential for homeostasis and injury-induced regeneration of the vertebrate blood system. Although HSC transplantations constitute the most common type of stem cell therapy applied in the clinic, we know relatively little about the molecular programming of HSCs during vertebrate embryogenesis. In vertebrate embryos, HSCs form in close association with the ventral wall of the dorsal aorta. We have shown previously that in zebrafish, HSC formation depends on the presence of a signaling cascade that involves Hedgehog, vascular endothelial growth factor, and Notch signaling. Here, we reveal that Hey2, a hairy/enhancer-of-split-related basic helix-loop-helix transcription factor often believed to act downstream of Notch, is also required for HSC formation. In dorsal aorta progenitors, Hey2 expression is induced downstream of cloche and the transcription factor Scl/Tal1, and is maintained by Hedgehog and vascular endothelial growth factor signaling. Whereas knockdown of Hey2 expression results in a loss of Notch receptor expression in dorsal aorta angioblasts, activation of Notch signaling in hey2 morphants rescues HSC formation in zebrafish embryos. These results establish an essential role for Hey2 upstream of Notch in HSC formation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Hematopoietic Stem Cells/cytology , Receptors, Notch , Zebrafish Proteins/physiology , Animals , Aorta , Cell Lineage , Embryo, Nonmammalian , Embryonic Development , Gene Expression Regulation, Developmental , Transcription Factors , Vascular Endothelial Growth Factor A , Zebrafish
12.
J Cell Physiol ; 222(1): 11-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19725072

ABSTRACT

The Notch signalling pathway is repeatedly employed during embryonic development and adult homeostasis of a variety of tissues. In particular, its frequent involvement in the regulation of stem and progenitor cell maintenance and proliferation, as well as its role in binary fate decisions in cells that are destined to differentiate, is remarkable. Here, we review its role in the development of haematopoietic stem cells during vertebrate embryogenesis and put it into the context of Notch's functions in arterial specification, angiogenic vessel sprouting and vessel maintenance. We further discuss interactions with other signalling cascades, and pinpoint open questions and some of the challenges that lie ahead.


Subject(s)
Cell Differentiation , Embryonic Development , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Core Binding Factor Alpha 2 Subunit/metabolism , Humans
13.
Dev Cell ; 16(6): 909-16, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19531361

ABSTRACT

Hematopoietic stem cells (HSCs) are first detected in the floor of the embryonic dorsal aorta (DA), and we investigate the signals that induce the HSC program there. We show that while continued Hedgehog (Hh) signaling from the overlying midline structures maintains the arterial program characteristic of the DA roof, a ventral Bmp4 signal induces the blood stem cell program in the DA floor. This patterning of the DA by Hh and Bmp is the mirror image of that in the neural tube, with Hh favoring dorsal rather than ventral cell types, and Bmp favoring ventral rather than dorsal. With the majority of current data supporting a model whereby HSCs derive from arterial endothelium, our data identify the signal driving this conversion. These findings are important for the study of the production of HSCs from embryonic stem cells and establish a paradigm for the development of adult stem cells.


Subject(s)
Aorta/cytology , Body Patterning , Bone Morphogenetic Protein 4/metabolism , Cell Polarity , Hedgehog Proteins/metabolism , Hematopoietic Stem Cells/cytology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Aorta/embryology , Arteries/cytology , Arteries/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Zebrafish Proteins/genetics
14.
Cell Stem Cell ; 2(4): 293-4, 2008 Apr 10.
Article in English | MEDLINE | ID: mdl-18397744

ABSTRACT

Notch signaling is essential for hematopoietic stem cell (HSC) formation during embryogenesis, and hitherto it was also thought to be required for HSC maintenance. However, in this issue of Cell Stem Cell, Maillard et al. (2008) demonstrate rather conclusively that inactivation of the Notch pathway in HSCs does not interfere with their self-renewal.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/cytology , Receptors, Notch/physiology , Signal Transduction/physiology , Animals , Humans
15.
Blood ; 109(6): 2389-98, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17090656

ABSTRACT

The transcription factors Scl and Lmo2 are crucial for development of all blood. An important early requirement for Scl in endothelial development has also been revealed recently in zebrafish embryos, supporting previous findings in scl(-/-) embryoid bodies. Scl depletion culminates most notably in failure of dorsal aorta formation, potentially revealing a role in the formation of hemogenic endothelium. We now present evidence that the requirements for Lmo2 in zebrafish embryos are essentially the same as for Scl. The expression of important hematopoietic regulators is lost, reduced, or delayed, panendothelial gene expression is down-regulated, and aorta-specific marker expression is lost. The close similarity of the phenotypes for Scl and Lmo2 suggest that they perform these early functions in hemangioblast development within a multiprotein complex, as shown for erythropoiesis. Consistent with this, we find that scl morphants cannot be rescued by a non-Lmo2-binding form of Scl but can be rescued by non-DNA-binding forms, suggesting tethering to target genes through DNA-binding partners linked via Lmo2. Interestingly, unlike other hematopoietic regulators, the Scl/Lmo2 complex does not appear to autoregulate, as neither gene's expression is affected by depletion of the other. Thus, expression of these critical regulators is dependent on continued expression of upstream regulators, which may include cell-extrinsic signals.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Hematopoiesis , Metalloproteins/metabolism , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/blood , Zebrafish/metabolism , Adaptor Proteins, Signal Transducing , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , DNA/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , LIM Domain Proteins , Metalloproteins/deficiency , Metalloproteins/genetics , Mice , Myeloid Cells/cytology , Myeloid Cells/metabolism , Phenotype , Protein Binding , Proto-Oncogene Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
16.
Development ; 133(24): 4913-23, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17108004

ABSTRACT

Ldb1, a ubiquitously expressed LIM domain binding protein, is essential in a number of tissues during development. It interacts with Gata1, Tal1, E2A and Lmo2 to form a transcription factor complex regulating late erythroid genes. We identify a number of novel Ldb1 interacting proteins in erythroleukaemic cells, in particular the repressor protein Eto-2 (and its family member Mtgr1), the cyclin-dependent kinase Cdk9, and the bridging factor Lmo4. MO-mediated knockdowns in zebrafish show these factors to be essential for definitive haematopoiesis. In accordance with the zebrafish results these factors are coexpressed in prehaematopoietic cells of the early mouse embryo, although we originally identified the complex in late erythroid cells. Based on the change in subcellullar localisation of Eto-2 we postulate that it plays a central role in the transition from the migration and expansion phase of the prehaematopoietic cells to the establishment of definitive haematopoietic stem cells.


Subject(s)
DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Embryo, Nonmammalian , Erythropoiesis/genetics , Hematopoietic Stem Cells/metabolism , Zebrafish Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Differentiation , Cell Line, Tumor , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase 9/metabolism , Erythroid Cells/cytology , Homeodomain Proteins/metabolism , LIM Domain Proteins , Leukemia, Erythroblastic, Acute , Mice , Nuclear Proteins/metabolism , Protein Binding , Repressor Proteins/metabolism , Transcription Factor 4 , Transcription Factors/metabolism , Transfection , Zebrafish
17.
Dev Cell ; 8(3): 389-400, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15737934

ABSTRACT

Studies with embryonic explants and embryonic stem cells have suggested a role for Hedgehog (Hh) signaling in hematopoiesis. However, targeted deletion of Hh pathway components in the mouse has so far failed to provide in vivo evidence. Here we show that zebrafish embryos mutant in the Hh pathway or treated with the Hh signaling inhibitor cyclopamine display defects in adult hematopoietic stem cell (HSC) formation but not in primitive hematopoiesis. Hh is required in the trunk at three consecutive stages during vascular development: for the medial migration of endothelial progenitors of the dorsal aorta (DA), for arterial gene expression, and for the formation of intersomitic vessel sprouts. Interference with Hh signaling during the first two stages also interferes with HSC formation. Furthermore, HSC and DA formation also share Vegf and Notch requirements, which further distinguishes them from primitive hematopoiesis and underlines their close relationship during vertebrate development.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Signal Transduction/physiology , T-Lymphocytes/cytology , Trans-Activators/metabolism , Animals , Body Patterning/drug effects , Body Patterning/physiology , Cell Movement/drug effects , Cell Movement/physiology , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins , Hematopoiesis/drug effects , Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , Mutation/genetics , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins/metabolism , Receptors, Notch , Signal Transduction/drug effects , T-Lymphocytes/metabolism , Trans-Activators/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Veratrum Alkaloids/toxicity , Zebrafish
18.
Blood ; 105(9): 3502-11, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15644413

ABSTRACT

Blood and endothelial cells arise in close association in developing embryos, possibly from a shared precursor, the hemangioblast, or as hemogenic endothelium. The transcription factor, Scl/Tal1 (stem cell leukemia protein), is essential for hematopoiesis but thought to be required only for remodeling of endothelium in mouse embryos. By contrast, it has been implicated in hemangioblast formation in embryoid bodies. To resolve the role of scl in endothelial development, we knocked down its synthesis in zebrafish embryos where early precursors and later phenotypes can be more easily monitored. With respect to blood, the zebrafish morphants phenocopied the mouse knockout and positioned scl in the genetic hierarchy. Importantly, endothelial development was also clearly disrupted. Dorsal aorta formation was substantially compromised and gene expression in the posterior cardinal vein was abnormal. We conclude that scl is especially critical for the development of arteries where adult hematopoietic stem cells emerge, implicating scl in the formation of hemogenic endothelium.


Subject(s)
Aorta/growth & development , Blood , DNA-Binding Proteins/physiology , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Aorta/embryology , Basic Helix-Loop-Helix Transcription Factors , Blood Circulation/genetics , DNA-Binding Proteins/deficiency , Embryo, Nonmammalian , Endothelium, Vascular/cytology , Endothelium, Vascular/embryology , Endothelium, Vascular/growth & development , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Mice , Neovascularization, Physiologic , Proto-Oncogene Proteins/deficiency , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/deficiency , Zebrafish , Zebrafish Proteins/deficiency
19.
Gene Expr Patterns ; 4(1): 85-92, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14678833

ABSTRACT

Basic helix-loop-helix (bHLH) transcription factors play key roles in the development of the central nervous system. Here we report the isolation of a zebrafish gene that encodes a homologue of the mammalian bHLH transcription factor, Tal2. In zebrafish embryos, tal2, like its mammalian homologue, is strongly expressed in the diencephalon and the mesencephalon, with the latter expression located in post-mitotic cells of the tectum. However, in addition to this conserved brain expression, we also detect expression in the floor plate of the spinal cord. By the location of this expression relative to other genes expressed in the floor plate and by analysing expression in a selection of midline mutants, we reveal that tal2 is expressed within the lateral floor plate as opposed to the medial floor plate, and also in more dorsal cells which are distinct from motorneurons and depend on either sonic hedgehog signalling or a signal coming from the lateral floor plate. This is to our knowledge the first report of a gene expressed specifically in lateral cells of the floor plate in the spinal cord.


Subject(s)
Spinal Cord/metabolism , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Embryo, Nonmammalian/metabolism , Embryonic Development , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genetic Markers , In Situ Hybridization , Molecular Sequence Data , Sequence Homology, Amino Acid , Spinal Cord/embryology , Time Factors , Zebrafish/embryology
20.
Development ; 130(25): 6187-99, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14602685

ABSTRACT

The LIM domain protein Lmo2 and the basic helix-loop-helix transcription factor Scl/Tal1 are expressed in early haematopoietic and endothelial progenitors and interact with each other in haematopoietic cells. While loss-of-function studies have shown that Lmo2 and Scl/Tal1 are essential for haematopoiesis and angiogenic remodelling of the vasculature, gain-of-function studies have suggested an earlier role for Scl/Tal1 in the specification of haemangioblasts, putative bipotential precursors of blood and endothelium. In zebrafish embryos, Scl/Tal1 can induce these progenitors from early mesoderm mainly at the expense of the somitic paraxial mesoderm. We show that this restriction to the somitic paraxial mesoderm correlates well with the ability of Scl/Tal1 to induce ectopic expression of its interaction partner Lmo2. Co-injection of lmo2 mRNA with scl/tal1 dramatically extends its effect to head, heart, pronephros and pronephric duct mesoderm inducing early blood and endothelial genes all along the anteroposterior axis. Erythroid development, however, is expanded only into pronephric mesoderm, remaining excluded from head, heart and somitic paraxial mesoderm territories. This restriction correlates well with activation of gata1 transcription and co-injection of gata1 mRNA along with scl/tal1 and lmo2 induces erythropoiesis more broadly without ventralising or posteriorising the embryo. While no ectopic myeloid development from the Scl/Tal1-Lmo2-induced haemangioblasts was observed, a dramatic increase in the number of endothelial cells was found. These results suggest that, in the absence of inducers of erythroid or myeloid haematopoiesis, Scl/Tal1-Lmo2-induced haemangioblasts differentiate into endothelial cells.


Subject(s)
Cell Differentiation/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Endothelium, Vascular/cytology , Hematopoiesis/physiology , Mesoderm/cytology , Metalloproteins/genetics , Neovascularization, Physiologic/physiology , Transcription Factors/deficiency , Transcription Factors/genetics , Zebrafish Proteins/genetics , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Body Patterning , Embryo, Nonmammalian/physiology , Erythroid-Specific DNA-Binding Factors , Functional Laterality , GATA1 Transcription Factor , Helix-Loop-Helix Motifs , LIM Domain Proteins , Open Reading Frames , Proto-Oncogene Proteins , RNA, Messenger/genetics , Repressor Proteins/genetics , Restriction Mapping , T-Cell Acute Lymphocytic Leukemia Protein 1 , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...