Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Methods Mol Biol ; 2198: 193-208, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32822033

RESUMO

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.


Assuntos
5-Metilcitosina/análogos & derivados , DNA/imunologia , Imuno-Histoquímica/métodos , 5-Metilcitosina/química , 5-Metilcitosina/metabolismo , Animais , Anticorpos/metabolismo , Núcleo Celular/metabolismo , Citosina/análogos & derivados , DNA/genética , Metilação de DNA , Dioxigenases , Embrião não Mamífero , Peixe-Zebra
2.
Blood Adv ; 2(20): 2589-2606, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30309860

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Eritroblastos/metabolismo , Hemangioblastos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Peixe-Zebra
3.
Front Cell Dev Biol ; 6: 27, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29616219

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-28262487

RESUMO

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.


Assuntos
Amidoidrolases/genética , Axônios/patologia , Degeneração Neural/genética , Nicotinamida-Nucleotídeo Adenililtransferase/deficiência , Degeneração Walleriana/genética , Amidoidrolases/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Degeneração Neural/metabolismo , Degeneração Walleriana/metabolismo
5.
Dev Biol ; 417(1): 25-39, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27432513

RESUMO

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.


Assuntos
Elementos de DNA Transponíveis/genética , Proteínas de Ligação a DNA/biossíntese , Eritrócitos/citologia , Eritropoese/genética , Células-Tronco Hematopoéticas/citologia , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Sequência de Bases , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core/biossíntese , Proteínas de Ligação a DNA/genética , Eritrócitos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , Receptores Notch/genética , Receptores Notch/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
6.
Cell Rep ; 13(11): 2539-2552, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26686637

RESUMO

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.


Assuntos
Proteínas do Domínio Armadillo/genética , Cálcio/metabolismo , Proteínas do Citoesqueleto/genética , Mitocôndrias/metabolismo , Mononucleotídeo de Nicotinamida/metabolismo , Acrilamidas/farmacologia , Amidoidrolases/metabolismo , Animais , Proteínas do Domínio Armadillo/deficiência , Axônios/efeitos dos fármacos , Axônios/metabolismo , Proteínas do Citoesqueleto/deficiência , Íons/química , Íons/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mononucleotídeo de Nicotinamida/farmacologia , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo , Piperidinas/farmacologia , Degeneração Walleriana/patologia
7.
Cardiovasc Res ; 100(2): 252-61, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23812297

RESUMO

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.


Assuntos
Circulação Sanguínea/fisiologia , Hipóxia/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Neovascularização Fisiológica , Receptores Notch/fisiologia , Transdução de Sinais/fisiologia , Animais , Diacetil/análogos & derivados , Diacetil/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Proteínas Supressoras de Tumor/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/fisiologia
8.
PLoS One ; 8(6): e65170, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23755189

RESUMO

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.


Assuntos
Proteínas Hedgehog/metabolismo , MicroRNAs/genética , Desenvolvimento Muscular/genética , Receptores Acoplados a Proteínas G/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Regiões 3' não Traduzidas , Animais , Sequência de Bases , Sítios de Ligação , Padronização Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Receptores Patched , Receptor Patched-1 , Interferência de RNA , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Receptor Smoothened , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 33(6): 1257-63, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23559631

RESUMO

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.


Assuntos
Aorta/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas do Tecido Nervoso/genética , Receptor Notch1/genética , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Transdução de Sinais/genética , Proteínas de Peixe-Zebra/genética , Animais , Coartação Aórtica/genética , Coartação Aórtica/fisiopatologia , Feminino , Masculino , Modelos Animais , Mutação/genética , Neovascularização Fisiológica/genética , Valores de Referência , Regulação para Cima , Peixe-Zebra
10.
Epigenetics ; 7(4): 383-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22419071

RESUMO

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.


Assuntos
Citosina/análogos & derivados , Regulação da Expressão Gênica no Desenvolvimento , Vertebrados/embriologia , Peixe-Zebra/metabolismo , 5-Metilcitosina/metabolismo , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Diferenciação Celular , Embrião de Galinha , Meios de Cultivo Condicionados , Citosina/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Organogênese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Tempo , Vertebrados/genética , Vertebrados/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética
11.
Blood ; 116(12): 2046-56, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20511544

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Células-Tronco Hematopoéticas/citologia , Receptores Notch , Proteínas de Peixe-Zebra/fisiologia , Animais , Aorta , Linhagem da Célula , Embrião não Mamífero , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição , Fator A de Crescimento do Endotélio Vascular , Peixe-Zebra
12.
J Cell Physiol ; 222(1): 11-6, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19725072

RESUMO

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.


Assuntos
Diferenciação Celular , Desenvolvimento Embrionário , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Humanos
13.
Dev Cell ; 16(6): 909-16, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19531361

RESUMO

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.


Assuntos
Aorta/citologia , Padronização Corporal , Proteína Morfogenética Óssea 4/metabolismo , Polaridade Celular , Proteínas Hedgehog/metabolismo , Células-Tronco Hematopoéticas/citologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Aorta/embriologia , Artérias/citologia , Artérias/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/metabolismo , Transdução de Sinais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Proteínas de Peixe-Zebra/genética
14.
Cell Stem Cell ; 2(4): 293-4, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18397744

RESUMO

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.


Assuntos
Proliferação de Células , Células-Tronco Hematopoéticas/citologia , Receptores Notch/fisiologia , Transdução de Sinais/fisiologia , Animais , Humanos
15.
Blood ; 109(6): 2389-98, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17090656

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Hematopoese , Metaloproteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/sangue , Peixe-Zebra/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Eritroides/citologia , Células Eritroides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas com Domínio LIM , Metaloproteínas/deficiência , Metaloproteínas/genética , Camundongos , Células Mieloides/citologia , Células Mieloides/metabolismo , Fenótipo , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Transcrição , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
16.
Development ; 133(24): 4913-23, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17108004

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Embrião não Mamífero , Eritropoese/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Quinase 9 Dependente de Ciclina/metabolismo , Células Eritroides/citologia , Proteínas de Homeodomínio/metabolismo , Proteínas com Domínio LIM , Leucemia Eritroblástica Aguda , Camundongos , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas Repressoras/metabolismo , Fator de Transcrição 4 , Fatores de Transcrição/metabolismo , Transfecção , Peixe-Zebra
17.
Dev Cell ; 8(3): 389-400, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15737934

RESUMO

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.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Transdução de Sinais/fisiologia , Linfócitos T/citologia , Transativadores/metabolismo , Animais , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core , Proteínas de Ligação a DNA/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Hedgehog , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Membrana/metabolismo , Mutação/genética , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Notch , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/metabolismo , Transativadores/genética , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Alcaloides de Veratrum/toxicidade , Peixe-Zebra
18.
Blood ; 105(9): 3502-11, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15644413

RESUMO

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.


Assuntos
Aorta/crescimento & desenvolvimento , Sangue , Proteínas de Ligação a DNA/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fatores de Transcrição/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Aorta/embriologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Circulação Sanguínea/genética , Proteínas de Ligação a DNA/deficiência , Embrião não Mamífero , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Endotélio Vascular/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/citologia , Camundongos , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas/deficiência , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Transcrição/deficiência , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência
19.
Gene Expr Patterns ; 4(1): 85-92, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14678833

RESUMO

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.


Assuntos
Medula Espinal/metabolismo , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Marcadores Genéticos , Hibridização In Situ , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Medula Espinal/embriologia , Fatores de Tempo , Peixe-Zebra/embriologia
20.
Development ; 130(25): 6187-99, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14602685

RESUMO

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.


Assuntos
Diferenciação Celular/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Endotélio Vascular/citologia , Hematopoese/fisiologia , Mesoderma/citologia , Metaloproteínas/genética , Neovascularização Fisiológica/fisiologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/genética , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Padronização Corporal , Embrião não Mamífero/fisiologia , Fatores de Ligação de DNA Eritroide Específicos , Lateralidade Funcional , Fator de Transcrição GATA1 , Sequências Hélice-Alça-Hélice , Proteínas com Domínio LIM , Fases de Leitura Aberta , Proteínas Proto-Oncogênicas , RNA Mensageiro/genética , Proteínas Repressoras/genética , Mapeamento por Restrição , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA