Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(36): eadf9706, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37672585

ABSTRACT

Trained immunity is a long-term memory of innate immune cells, generating an improved response upon reinfection. Shigella is an important human pathogen and inflammatory paradigm for which there is no effective vaccine. Using zebrafish larvae, we demonstrate that after Shigella training, neutrophils are more efficient at bacterial clearance. We observe that Shigella-induced protection is nonspecific and has differences with training by BCG and ß-glucan. Analysis of histone ChIP-seq on trained neutrophils revealed that Shigella training deposits the active H3K4me3 mark on promoter regions of 1612 genes, dramatically changing the epigenetic landscape of neutrophils toward enhanced microbial recognition and mitochondrial ROS production. Last, we demonstrate that mitochondrial ROS plays a key role in enhanced antimicrobial activity of trained neutrophils. It is envisioned that signals and mechanisms we discover here can be used in other vertebrates, including humans, to suggest new therapeutic strategies involving neutrophils to control bacterial infection.


Subject(s)
Enterobacteriaceae Infections , Epigenesis, Genetic , Mycobacterium bovis , Neutrophils , Trained Immunity , beta-Glucans , Enterobacteriaceae Infections/immunology , Animals , Zebrafish , Larva , Neutrophils/immunology , Neutrophils/metabolism , Shigella flexneri/physiology , Mycobacterium bovis/immunology , beta-Glucans/administration & dosage , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
2.
Front Cell Dev Biol ; 10: 982477, 2022.
Article in English | MEDLINE | ID: mdl-36133924

ABSTRACT

The T-box family transcription factor Eomesodermin (Eomes) is present in all vertebrates, with many key roles in the developing mammalian embryo and immune system. Homozygous Eomes mutant mouse embryos exhibit early lethality due to defects in both the embryonic mesendoderm and the extraembryonic trophoblast cell lineage. In contrast, zebrafish lacking the predominant Eomes homologue A (Eomesa) do not suffer complete lethality and can be maintained. This suggests fundamental differences in either the molecular function of Eomes orthologues or the molecular configuration of processes in which they participate. To explore these hypotheses we initially analysed the expression of distinct Eomes isoforms in various mouse cell types. Next we compared the functional capabilities of these murine isoforms to zebrafish Eomesa. These experiments provided no evidence for functional divergence. Next we examined the functions of zebrafish Eomesa and other T-box family members expressed in early development, as well as its paralogue Eomesb. Though Eomes is a member of the Tbr1 subfamily we found evidence for functional redundancy with the Tbx6 subfamily member Tbx16, known to be absent from eutherians. However, Tbx16 does not appear to synergise with Eomesa cofactors Mixl1 and Gata5. Finally, we analysed the ability of Eomesa and other T-box factors to induce zebrafish left-right organiser progenitors (known as dorsal forerunner cells) known to be positively regulated by vgll4l, a gene we had previously shown to be repressed by Eomesa. Here we demonstrate that Eomesa indirectly upregulates vgll4l expression via interlocking feedforward loops, suggesting a role in establishment of left-right asymmetry. Conversely, other T-box factors could not similarly induce left-right organiser progenitors. Overall these findings demonstrate conservation of Eomes molecular function and participation in similar processes, but differential requirements across evolution due to additional co-expressed T-box factors in teleosts, albeit with markedly different molecular capabilities. Our analyses also provide insights into the role of Eomesa in left-right organiser formation in zebrafish.

4.
Sci Rep ; 11(1): 22717, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34811400

ABSTRACT

Retinoic acid (RA) is a key signal for the specification of the pancreas. Still, the gene regulatory cascade triggered by RA in the endoderm remains poorly characterized. In this study, we investigated this regulatory network in zebrafish by combining RNA-seq, RAR ChIP-seq and ATAC-seq assays. By analysing the effect of RA and of the RA receptor (RAR) inverse-agonist BMS493 on the transcriptome and on the chromatin accessibility of endodermal cells, we identified a large set of genes and regulatory regions regulated by RA signalling. RAR ChIP-seq further defined the direct RAR target genes in zebrafish, including hox genes as well as several pancreatic regulators like mnx1, insm1b, hnf1ba and gata6. Comparison of zebrafish and murine RAR ChIP-seq data highlighted the conserved direct target genes and revealed that some RAR sites are under strong evolutionary constraints. Among them, a novel highly conserved RAR-induced enhancer was identified downstream of the HoxB locus and driving expression in the nervous system and in the gut in a RA-dependent manner. Finally, ATAC-seq data unveiled the role of the RAR-direct targets Hnf1ba and Gata6 in opening chromatin at many regulatory loci upon RA treatment.


Subject(s)
Genomics , Pancreas/drug effects , Receptors, Retinoic Acid/agonists , Transcriptome , Tretinoin/pharmacology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation Sequencing , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Pancreas/embryology , Pancreas/metabolism , RNA-Seq , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Development ; 147(8)2020 04 28.
Article in English | MEDLINE | ID: mdl-32345657

ABSTRACT

Skeletal muscle derives from dorsal mesoderm formed during vertebrate gastrulation. Fibroblast growth factor (Fgf) signalling cooperates with Tbx transcription factors to promote dorsal mesoderm formation, but their role in myogenesis has been unclear. Using zebrafish, we show that dorsally derived Fgf signals act through Tbx16 and Tbxta to induce slow and fast trunk muscle precursors at distinct dorsoventral positions. Tbx16 binds to and directly activates the myf5 and myod genes, which are required for commitment to myogenesis. Tbx16 activity depends on Fgf signalling from the organiser. In contrast, Tbxta is not required for myf5 expression, but binds a specific site upstream of myod that is not bound by Tbx16 and drives (dependent on Fgf signals) myod expression in adaxial slow precursors, thereby initiating trunk myogenesis. After gastrulation, when similar muscle cell populations in the post-anal tail are generated from tailbud, declining Fgf signalling is less effective at initiating adaxial myogenesis, which is instead initiated by Hedgehog signalling from the notochord. Our findings suggest a hypothesis for ancestral vertebrate trunk myogenic patterning and how it was co-opted during tail evolution to generate similar muscle by new mechanisms.This article has an associated 'The people behind the papers' interview.


Subject(s)
Fibroblast Growth Factors/metabolism , Muscle Development , MyoD Protein/metabolism , T-Box Domain Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Body Patterning/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Muscle Development/genetics , MyoD Protein/genetics , Signal Transduction , T-Box Domain Proteins/genetics , Transcription, Genetic , Up-Regulation/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics
6.
J Muscle Res Cell Motil ; 40(2): 211-226, 2019 06.
Article in English | MEDLINE | ID: mdl-31301002

ABSTRACT

MYOD is a master regulator of the skeletal myogenic program. But what regulates expression of Myod? More than 20 years ago, studies established that Myod expression is largely controlled by just two enhancer regions located within a region 24 kb upstream of the transcription start site in mammals, which regulate Myod expression in the embryo, fetus and adult. Despite this apparently simple arrangement, Myod regulation is complex, with different combinations of transcription factors acting on these enhancers in different muscle progenitor cells and phases of differentiation. A range of epigenetic modifications in the Myod upstream region also play a part in activating and repressing Myod expression during development and regeneration. Here the evidence for this binding at Myod control regions is summarized, giving an overview of our current understanding of Myod expression regulation in mammals.


Subject(s)
Cell Differentiation , Enhancer Elements, Genetic , Gene Expression Regulation , MyoD Protein/biosynthesis , Myoblasts/metabolism , Transcription, Genetic , Animals , History, 20th Century , History, 21st Century , Humans , MyoD Protein/genetics , MyoD Protein/history
7.
Cell Rep ; 19(13): 2782-2795, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28658625

ABSTRACT

T-box transcription factors T/Brachyury homolog A (Ta) and Tbx16 are essential for correct mesoderm development in zebrafish. The downstream transcriptional networks guiding their functional activities are poorly understood. Additionally, important contributions elsewhere are likely masked due to redundancy. Here, we exploit functional genomic strategies to identify Ta and Tbx16 targets in early embryogenesis. Surprisingly, we discovered they not only activate mesodermal gene expression but also redundantly regulate key endodermal determinants, leading to substantial loss of endoderm in double mutants. To further explore the gene regulatory networks (GRNs) governing endoderm formation, we identified targets of Ta/Tbx16-regulated homeodomain transcription factor Mixl1, which is absolutely required in zebrafish for endoderm formation. Interestingly, we find many endodermal determinants coordinately regulated through common genomic occupancy by Mixl1, Eomesa, Smad2, Nanog, Mxtx2, and Pou5f3. Collectively, these findings augment the endoderm GRN and reveal a panel of target genes underlying the Ta, Tbx16, and Mixl1 mutant phenotypes.


Subject(s)
Endoderm/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Zebrafish
8.
J Clin Invest ; 127(3): 874-887, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28165338

ABSTRACT

The mechanisms underlying the neurodevelopmental deficits associated with CHARGE syndrome, which include cerebellar hypoplasia, developmental delay, coordination problems, and autistic features, have not been identified. CHARGE syndrome has been associated with mutations in the gene encoding the ATP-dependent chromatin remodeler CHD7. CHD7 is expressed in neural stem and progenitor cells, but its role in neurogenesis during brain development remains unknown. Here we have shown that deletion of Chd7 from cerebellar granule cell progenitors (GCps) results in reduced GCp proliferation, cerebellar hypoplasia, developmental delay, and motor deficits in mice. Genome-wide expression profiling revealed downregulated expression of the gene encoding the glycoprotein reelin (Reln) in Chd7-deficient GCps. Recessive RELN mutations have been associated with severe cerebellar hypoplasia in humans. We found molecular and genetic evidence that reductions in Reln expression contribute to GCp proliferative defects and cerebellar hypoplasia in GCp-specific Chd7 mouse mutants. Finally, we showed that CHD7 is necessary for maintaining an open, accessible chromatin state at the Reln locus. Taken together, this study shows that Reln gene expression is regulated by chromatin remodeling, identifies CHD7 as a previously unrecognized upstream regulator of Reln, and provides direct in vivo evidence that a mammalian CHD protein can control brain development by modulating chromatin accessibility in neuronal progenitors.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cerebellum/embryology , DNA-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Developmental , Genetic Loci , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Serine Endopeptidases/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cerebellum/abnormalities , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Extracellular Matrix Proteins/genetics , Genome-Wide Association Study , Humans , Mice , Mice, Transgenic , Motor Disorders/genetics , Motor Disorders/metabolism , Nerve Tissue Proteins/genetics , Nervous System Malformations/embryology , Nervous System Malformations/genetics , Reelin Protein , Serine Endopeptidases/genetics
9.
J Mol Cell Cardiol ; 92: 105-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26827899

ABSTRACT

The adult human myocardium is incapable of regeneration; yet, the zebrafish (Danio rerio) can regenerate damaged myocardium. Similar to the zebrafish heart, hearts of neonatal, but not adult mice are capable of myocardial regeneration. We performed a proteomics analysis of adult zebrafish hearts and compared their protein expression profile to hearts from neonatal and adult mice. Using difference in-gel electrophoresis (DIGE), there was little overlap between the proteome from adult mouse (>8weeks old) and adult zebrafish (18months old) hearts. Similarly, there was a significant degree of mismatch between the protein expression in neonatal and adult mouse hearts. Enrichment analysis of the selected proteins revealed over-expression of DNA synthesis-related proteins in the cardiac proteome of the adult zebrafish heart similar to neonatal and 4days old mice, whereas in hearts of adult mice there was a mitochondria-related predominance in protein expression. Importantly, we noted pronounced differences in the myofilament composition: the adult zebrafish heart lacks many of the myofilament proteins of differentiated adult cardiomyocytes such as the ventricular isoforms of myosin light chains and nebulette. Instead, troponin I and myozenin 1 were expressed as skeletal isoforms rather than cardiac isoforms. The relative immaturity of the adult zebrafish heart was further supported by cardiac microRNA data. Our assessment of zebrafish and mammalian hearts challenges the assertions on the translational potential of cardiac regeneration in the zebrafish model. The immature myofilament composition of the fish heart may explain why adult mouse and human cardiomyocytes lack this endogenous repair mechanism.


Subject(s)
Heart/growth & development , Proteome/biosynthesis , Proteomics , Regeneration/genetics , Zebrafish/genetics , Animals , Gene Expression Regulation, Developmental , Heart Ventricles/growth & development , Heart Ventricles/metabolism , Humans , Mice , MicroRNAs/biosynthesis , Microfilament Proteins/biosynthesis , Muscle Proteins/biosynthesis , Myocytes, Cardiac/metabolism , Proteome/genetics , Transcriptome , Troponin I/biosynthesis , Zebrafish/growth & development
10.
F1000Res ; 4: 235, 2015.
Article in English | MEDLINE | ID: mdl-26594335

ABSTRACT

Chromatin immunoprecipitation (ChIP) is a technique widely used in the study of epigenetics and transcriptional regulation of gene expression. However, its antibody-centric nature exposes it to similar challenges faced by other antibody-based procedures, of which the most prominent are issues of specificity and affinity in antigen recognition. As with other techniques that make use of antibodies, recent studies have shown the need for validation of ChIP antibodies in order to be sure they recognize the advertised protein or epitope. We summarize here the issues surrounding ChIP antibody usage, and highlight the toolkit of validation methods that can be employed by investigators looking to appraise these reagents.

11.
Development ; 142(6): 1159-68, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25725067

ABSTRACT

During embryonic development, the paraxial mesoderm becomes segmented into somites, within which proliferative muscle progenitors and muscle fibers establish the skeletal musculature. Here, we demonstrate that a gene network previously implicated in somite boundary formation, involving the transcriptional regulators Tbx6, Mesp-b and Ripply1, also confers spatial and temporal regulation to skeletal myogenesis in zebrafish. We show that Tbx6 directly regulates mesp-b and ripply1 expression in vivo, and that the interactions within the regulatory network are largely conserved among vertebrates. Mesp-b is necessary and sufficient for the specification of a subpopulation of muscle progenitors, the central proportion of the Pax3(+)/Pax7(+) dermomyotome. Conditional ubiquitous expression indicates that Mesp-b acts by inhibiting myogenic differentiation and by inducing the dermomyotome marker meox1. By contrast, Ripply1 induces a negative-feedback loop by promoting Tbx6 protein degradation. Persistent Tbx6 expression in Ripply1 knockdown embryos correlates with a deficit in dermomyotome and myotome marker gene expression, suggesting that Ripply1 promotes myogenesis by terminating Tbx6-dependent inhibition of myogenic maturation. Together, our data suggest that Mesp-b is an intrinsic upstream regulator of skeletal muscle progenitors and that, in zebrafish, the genes regulating somite boundary formation also regulate the development of the dermomyotome in the anterior somite compartment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Muscle Development/physiology , Muscle, Skeletal/embryology , Nuclear Proteins/metabolism , T-Box Domain Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified , Antibodies, Monoclonal , Base Sequence , Chromatin Immunoprecipitation , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Gene Regulatory Networks/genetics , Gene Regulatory Networks/physiology , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Morpholinos/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Somites/embryology , T-Box Domain Proteins/immunology , Zebrafish Proteins/immunology
12.
BMC Biol ; 12: 81, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25277163

ABSTRACT

BACKGROUND: Nodal signalling is an absolute requirement for normal mesoderm and endoderm formation in vertebrate embryos, yet the transcriptional networks acting directly downstream of Nodal and the extent to which they are conserved is largely unexplored, particularly in vivo. Eomesodermin also plays a role in patterning mesoderm and endoderm in vertebrates, but its mechanisms of action, and how it interacts with the Nodal signalling pathway are still unclear. RESULTS: Using a combination of ChIP-seq and expression analysis we identify direct targets of Smad2, the effector of Nodal signalling in blastula stage zebrafish embryos, including many novel target genes. Through comparison of these data with published ChIP-seq data in human, mouse and Xenopus we show that the transcriptional network driven by Smad2 in mesoderm and endoderm is conserved in these vertebrate species. We also show that Smad2 and zebrafish Eomesodermin a (Eomesa) bind common genomic regions proximal to genes involved in mesoderm and endoderm formation, suggesting Eomesa forms a general component of the Smad2 signalling complex in zebrafish. Combinatorial perturbation of Eomesa and Smad2-interacting factor Foxh1 results in loss of both mesoderm and endoderm markers, confirming the role of Eomesa in endoderm formation and its functional interaction with Foxh1 for correct Nodal signalling. Finally, we uncover a novel, role for Eomesa in repressing ectodermal genes in the early blastula. CONCLUSION: Our data demonstrate that evolutionarily conserved developmental functions of Nodal signalling occur through maintenance of the transcriptional network directed by Smad2. This network is modulated by Eomesa in zebrafish which acts to promote mesoderm and endoderm formation in combination with Nodal signalling, whilst Eomesa also opposes ectoderm gene expression. Eomesa therefore regulates the formation of all three germ layers in the early zebrafish embryo.


Subject(s)
Body Patterning , Gene Expression Regulation, Developmental , Smad2 Protein/genetics , T-Box Domain Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Ectoderm/embryology , Ectoderm/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endoderm/embryology , Endoderm/metabolism , Gene Regulatory Networks , Mesoderm/embryology , Mesoderm/metabolism , Signal Transduction , Smad2 Protein/metabolism , T-Box Domain Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
14.
Development ; 140(7): 1385-95, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23482485

ABSTRACT

It is a truth (almost) universally acknowledged that conserved non-coding genomic sequences function in the cis regulation of neighbouring genes. But is this a misconception? The literature is strewn with examples of conserved non-coding sequences being able to drive reporter expression, but the extent to which such sequences are actually used endogenously in vivo is only now being rigorously explored using unbiased genome-scale approaches. Here, we review the emerging picture, examining the extent to which conserved non-coding sequences equivalently regulate gene expression in different species, or at different developmental stages, and how genomics approaches are revealing the relationship between sequence conservation and functional use of cis-regulatory elements.


Subject(s)
Conserved Sequence/genetics , Gene Expression Regulation, Developmental/genetics , Regulatory Sequences, Nucleic Acid/physiology , Animals , Base Sequence , Conserved Sequence/physiology , DNA, Intergenic/genetics , Evolution, Molecular , Humans , Models, Biological , Phylogeny , Regulatory Sequences, Nucleic Acid/genetics
15.
Dev Biol ; 371(1): 110-20, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22877946

ABSTRACT

Somites form by an iterative process from unsegmented, presomitic mesoderm (PSM). Notch pathway components, such as deltaC (dlc) have been shown to play a role in this process, while the T-box transcription factors Ntla and Tbx16 regulate somite formation upstream of this by controlling supply and movement of cells into the PSM during gastrulation and tailbud outgrowth. In this work, we report that Ntla and Tbx16 play a more explicit role in segmentation by directly regulating dlc expression. In addition we describe a cis-regulatory module (CRM) upstream of dlc that drives expression of a reporter in the tailbud, PSM and somites during somitogenesis. This CRM is bound by both Ntla and Tbx16 at a cluster of T-box binding sites, which are required in combination for activation of the CRM.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Mesoderm/metabolism , Somites/metabolism , T-Box Domain Proteins/pharmacology , Tail/metabolism , Zebrafish Proteins/pharmacology , Animals , Base Sequence , Chromatin Immunoprecipitation , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Fetal Proteins , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mesoderm/embryology , Molecular Sequence Data , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Sequence Analysis, DNA , Somites/embryology , T-Box Domain Proteins/genetics , Tail/embryology , Zebrafish , Zebrafish Proteins/genetics
16.
J Pathol ; 228(3): 274-85, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22847733

ABSTRACT

Chordoma is a rare malignant tumour of bone, the molecular marker of which is the expression of the transcription factor, brachyury. Having recently demonstrated that silencing brachyury induces growth arrest in a chordoma cell line, we now seek to identify its downstream target genes. Here we use an integrated functional genomics approach involving shRNA-mediated brachyury knockdown, gene expression microarray, ChIP-seq experiments, and bioinformatics analysis to achieve this goal. We confirm that the T-box binding motif of human brachyury is identical to that found in mouse, Xenopus, and zebrafish development, and that brachyury acts primarily as an activator of transcription. Using human chordoma samples for validation purposes, we show that brachyury binds 99 direct targets and indirectly influences the expression of 64 other genes, thereby acting as a master regulator of an elaborate oncogenic transcriptional network encompassing diverse signalling pathways including components of the cell cycle, and extracellular matrix components. Given the wide repertoire of its active binding and the relative specific localization of brachyury to the tumour cells, we propose that an RNA interference-based gene therapy approach is a plausible therapeutic avenue worthy of investigation.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/physiopathology , Chordoma/genetics , Chordoma/physiopathology , Fetal Proteins/genetics , Fetal Proteins/physiology , Genomics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/physiology , Animals , Bone Neoplasms/pathology , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation , Chordoma/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Genetic Therapy , Humans , Mice , Notochord/pathology , RNA Interference , Transcription, Genetic/physiology , Xenopus , Zebrafish
17.
Int J Dev Biol ; 56(4): 285-94, 2012.
Article in English | MEDLINE | ID: mdl-22562204

ABSTRACT

Mesp proteins play crucial roles in the formation of heart, vasculature and somites during vertebrate embryogenesis. We have used phylogenetic and genomic analysis, combined with qRT-PCR and in situ hybridization, to characterize two novel additional mesp genes in zebrafish, mesp-ab and mesp-bb, and describe their expression pattern in wild type and segmentation mutants. Both mesp-ab and mesp-bb are expressed in early mesoderm with mesp-ab expression starting during late blastula stages and mesp-bb expression initiating later, at the end of gastrulation. During somitogenesis, both mesp genes are expressed dynamically in the anterior presomitic mesoderm. mesp-ab is expressed in presumptive somites S-I and S-II, while mesp-bb is detected in S-I, S-II and S0, with expression restricted to the rostral compartment of presumptive somites. We show that the segmentation clock program regulates expression of these newly identified zebrafish mesp genes in a similar manner to their ohnologs, mesp-aa and mesp-ba. We also present evidence that zebrafish, minnow and salmon retained these additional mesp genes after the teleost whole genome duplication, while medaka, stickleback, fugu and tetraodon did not. Finally we show that although expression and regulation of zebrafish mesp genes appears highly comparable, there is no conservation in non-coding regions with other teleosts. In this study we have completed the description of the Mesp family in zebrafish, which will enable correct genome annotation and facilitate further functional studies on the role of these proteins in zebrafish.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Developmental , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/classification , Body Patterning/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Profiling , In Situ Hybridization , Mesoderm/embryology , Mesoderm/metabolism , Molecular Sequence Data , Mutation , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Somites/embryology , Somites/metabolism , Zebrafish/embryology , Zebrafish Proteins/classification
18.
PLoS One ; 7(3): e33346, 2012.
Article in English | MEDLINE | ID: mdl-22479388

ABSTRACT

BACKGROUND: The T-box transcription factor Brachyury (T) is essential for formation of the posterior mesoderm and the notochord in vertebrate embryos. Work in the frog and the zebrafish has identified some direct genomic targets of Brachyury, but little is known about Brachyury targets in the mouse. METHODOLOGY/PRINCIPAL FINDINGS: Here we use chromatin immunoprecipitation and mouse promoter microarrays to identify targets of Brachyury in embryoid bodies formed from differentiating mouse ES cells. The targets we identify are enriched for sequence-specific DNA binding proteins and include components of signal transduction pathways that direct cell fate in the primitive streak and tailbud of the early embryo. Expression of some of these targets, such as Axin2, Fgf8 and Wnt3a, is down regulated in Brachyury mutant embryos and we demonstrate that they are also Brachyury targets in the human. Surprisingly, we do not observe enrichment of the canonical T-domain DNA binding sequence 5'-TCACACCT-3' in the vicinity of most Brachyury target genes. Rather, we have identified an (AC)(n) repeat sequence, which is conserved in the rat but not in human, zebrafish or Xenopus. We do not understand the significance of this sequence, but speculate that it enhances transcription factor binding in the regulatory regions of Brachyury target genes in rodents. CONCLUSIONS/SIGNIFICANCE: Our work identifies the genomic targets of a key regulator of mesoderm formation in the early mouse embryo, thereby providing insights into the Brachyury-driven genetic regulatory network and allowing us to compare the function of Brachyury in different species.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Fetal Proteins/metabolism , Proteins/metabolism , T-Box Domain Proteins/metabolism , Animals , Axin Protein/genetics , Axin Protein/metabolism , Base Sequence , Binding Sites/genetics , Blotting, Western , Cell Line , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Fetal Proteins/genetics , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Mice , Mice, 129 Strain , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Binding , Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/genetics , Time Factors , Wnt3A Protein/genetics , Wnt3A Protein/metabolism
19.
Proc Natl Acad Sci U S A ; 106(10): 3829-34, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19225104

ABSTRACT

Using chromatin immunoprecipitation combined with genomic microarrays we have identified targets of No tail (Ntl), a zebrafish Brachyury ortholog that plays a central role in mesoderm formation. We show that Ntl regulates a downstream network of other transcription factors and identify an in vivo Ntl binding site that resembles the consensus T-box binding site (TBS) previously identified by in vitro studies. We show that the notochord-expressed gene floating head (flh) is a direct transcriptional target of Ntl and that a combination of TBSs in the flh upstream region are required for Ntl-directed expression. Using our genome-scale data we have assembled a preliminary gene regulatory network that begins to describe mesoderm formation and patterning in the early zebrafish embryo.


Subject(s)
Fetal Proteins/metabolism , Gene Regulatory Networks , Mesoderm/embryology , T-Box Domain Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Animals , Base Sequence , Binding Sites , Body Patterning/genetics , Cell Lineage , Conserved Sequence , Fetal Proteins/genetics , Gastrulation/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Molecular Sequence Data , Muscles/cytology , Protein Binding , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
Development ; 136(5): 749-60, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19158186

ABSTRACT

The zebrafish genes spadetail (spt) and no tail (ntl) encode T-box transcription factors that are important for early mesoderm development. Although much has been done to characterize these genes, the identity and location of target regulatory elements remain largely unknown. Here, we survey the genome for downstream target genes of the Spt and Ntl T-box transcription factors. We find evidence for extensive additive interactions towards gene activation and limited evidence for combinatorial and antagonistic interactions between the two factors. Using in vitro binding selection assays to define Spt- and Ntl-binding motifs, we searched for target regulatory sequence via a combination of binding motif searches and comparative genomics. We identified regulatory elements for tbx6 and deltaD, and, using chromatin immunoprecipitation, in vitro DNA binding assays and transgenic methods, we provide evidence that both are directly regulated by T-box transcription factors. We also find that deltaD is directly activated by T-box factors in the tail bud, where it has been implicated in starting the segmentation clock, suggesting that spt and ntl act upstream of this process.


Subject(s)
T-Box Domain Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Binding Sites/genetics , DNA/genetics , DNA/metabolism , Fetal Proteins , Gene Expression Regulation, Developmental , Mesoderm/embryology , Mesoderm/metabolism , Mice , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Elements, Transcriptional , Sequence Homology, Nucleic Acid , Signal Transduction , T-Box Domain Proteins/metabolism , Tail/embryology , Tail/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...