Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 501: 92-103, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37353106

RESUMO

During embryonic development, primitive and definitive waves of hematopoiesis take place to provide proper blood cells for each developmental stage, with the possible involvement of epigenetic factors. We previously found that lysine-specific demethylase 1 (LSD1/KDM1A) promotes primitive hematopoietic differentiation by shutting down the gene expression program of hemangioblasts in an Etv2/Etsrp-dependent manner. In the present study, we demonstrated that zebrafish LSD1 also plays important roles in definitive hematopoiesis in the development of hematopoietic stem and progenitor cells. A combination of genetic approaches and imaging analyses allowed us to show that LSD1 promotes the egress of hematopoietic stem and progenitor cells into the bloodstream during the endothelial-to-hematopoietic transition. Analysis of compound mutant lines with Etv2/Etsrp mutant zebrafish revealed that, unlike in primitive hematopoiesis, this function of LSD1 was independent of Etv2/Etsrp. The phenotype of LSD1 mutant zebrafish during the endothelial-to-hematopoietic transition was similar to that of previously reported compound knockout mice of Gfi1/Gfi1b, which forms a complex with LSD1 and represses endothelial genes. Moreover, co-knockdown of zebrafish Gfi1/Gfi1b genes inhibited the development of hematopoietic stem and progenitor cells. We therefore hypothesize that the shutdown of the Gfi1/Gfi1b-target genes during the endothelial-to-hematopoietic transition is one of the key evolutionarily conserved functions of LSD1 in definitive hematopoiesis.


Assuntos
Células-Tronco , Peixe-Zebra , Animais , Camundongos , Diferenciação Celular , Hematopoese/genética , Histona Desmetilases/genética , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Development ; 147(8)2020 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-32341028

RESUMO

Runx1 is a transcription factor that plays a key role in determining the proliferative and differential state of multiple cell types, during both development and adulthood. Here, we report how Runx1 is specifically upregulated at the injury site during zebrafish heart regeneration, and that absence of runx1 results in increased myocardial survival and proliferation, and overall heart regeneration, accompanied by decreased fibrosis. Using single cell sequencing, we found that the wild-type injury site consists of Runx1-positive endocardial cells and thrombocytes that induce expression of smooth muscle and collagen genes. Both these populations cannot be identified in runx1 mutant wounds that contain less collagen and fibrin. The reduction in fibrin in the mutant is further explained by reduced myofibroblast formation and upregulation of components of the fibrin degradation pathway, including plasminogen receptor annexin 2A as well as downregulation of plasminogen activator inhibitor serpine1 in myocardium and endocardium, resulting in increased levels of plasminogen. Our findings suggest that Runx1 controls the regenerative response of multiple cardiac cell types and that targeting Runx1 is a novel therapeutic strategy for inducing endogenous heart repair.


Assuntos
Cicatriz/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Coração/fisiopatologia , Miocárdio/patologia , Regeneração , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia , Animais , Anexina A2/metabolismo , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Endocárdio/patologia , Regulação da Expressão Gênica no Desenvolvimento , Músculo Liso/metabolismo , Mutação/genética , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Cadeias Pesadas de Miosina/metabolismo , Regulação para Cima/genética , Proteínas de Peixe-Zebra/genética
3.
Commun Biol ; 3(1): 71, 2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-32054973

RESUMO

Gata2 is a key transcription factor required to generate Haematopoietic Stem and Progenitor Cells (HSPCs) from haemogenic endothelium (HE); misexpression of Gata2 leads to haematopoietic disorders. Here we deleted a conserved enhancer (i4 enhancer) driving pan-endothelial expression of the zebrafish gata2a and showed that Gata2a is required for HE programming by regulating expression of runx1 and of the second Gata2 orthologue, gata2b. By 5 days, homozygous gata2aΔi4/Δi4 larvae showed normal numbers of HSPCs, a recovery mediated by Notch signalling driving gata2b and runx1 expression in HE. However, gata2aΔi4/Δi4 adults showed oedema, susceptibility to infections and marrow hypo-cellularity, consistent with bone marrow failure found in GATA2 deficiency syndromes. Thus, gata2a expression driven by the i4 enhancer is required for correct HE programming in embryos and maintenance of steady-state haematopoietic stem cell output in the adult. These enhancer mutants will be useful in exploring further the pathophysiology of GATA2-related deficiencies in vivo.


Assuntos
Reprogramação Celular/genética , Sequência Conservada , Endotélio/metabolismo , Elementos Facilitadores Genéticos , Fator de Transcrição GATA2/genética , Hematopoese/genética , Deleção de Sequência , Fatores Etários , Animais , Sequência de Bases , Cromatina/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Loci Gênicos , Células-Tronco Hematopoéticas/metabolismo , Peixe-Zebra
4.
Nat Commun ; 10(1): 3577, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31395869

RESUMO

Haematopoietic stem cells are generated from the haemogenic endothelium (HE) located in the floor of the dorsal aorta (DA). Despite being integral to arteries, it is controversial whether HE and arterial endothelium share a common lineage. Here, we present a transgenic zebrafish runx1 reporter line to isolate HE and aortic roof endothelium (ARE)s, excluding non-aortic endothelium. Transcriptomic analysis of these populations identifies Runx1-regulated genes and shows that HE initially expresses arterial markers at similar levels to ARE. Furthermore, runx1 expression depends on prior arterial programming by the Notch ligand dll4. Runx1-/- mutants fail to downregulate arterial genes in the HE, which remains integrated within the DA, suggesting that Runx1 represses the pre-existing arterial programme in HE to allow progression towards the haematopoietic fate. These findings strongly suggest that, in zebrafish, aortic endothelium is a precursor to HE, with potential implications for pluripotent stem cell differentiation protocols for the generation of transplantable HSCs.


Assuntos
Artérias/embriologia , Endotélio Vascular/embriologia , Hemangioblastos/fisiologia , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Artérias/citologia , Artérias/metabolismo , Linhagem da Célula , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Embrião não Mamífero , Desenvolvimento Embrionário , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Técnicas de Inativação de Genes , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
5.
J Comput Biol ; 26(7): 719-725, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31140835

RESUMO

Blood is an example of a highly regenerative tissue and its regeneration depends on the presence of stem cells residing in the bone marrow in humans. A better understanding of how these stem cells are programmed would benefit their use in clinical practice and shed light on the mechanisms by which the unique properties of stem cells are established. Our approach is to delineate the gene regulatory networks (GRNs) that specify these cells during their development in the embryo, and we use the amphibian experimental model because a wealth of evidence shows that the mechanisms used are conserved in mammals including humans. Blood stem cells are made during the intraembryonic wave of hematopoiesis during embryonic development where they emerge from endothelial precursors in the floor of the dorsal aorta (DA). These cells are derived from lateral plate mesoderm and so we have focused on the subset of cells in the lateral plate mesoderm fated to become blood and endothelium known as definitive hemangioblasts. We have found that their programming results from the activities of vascular endothelial growth factor A (VEGFA) and bone morphogenetic protein (BMP) signaling and the inhibition by miRNA of transforming growth factor beta signaling. VEGFA is first generated in the somites adjacent to the lateral plate mesoderm, and one of the responses of the lateral plate mesoderm is to activate endogenous VEGFA expression. BMP has multiple inputs into the programming of these cells via the activation of the transcription factor (TF), Gata2, and of the VEGFA receptor. These actions culminate in the expression of the leukemia-associated TF, Scl/Tal1, which is essential for blood fate specification. The activity of VEGFA in driving endothelial development resides in the small isoform, but the medium and large isoforms are required to initiate the blood stem cell program in the floor of the DA. The expression of the small isoform is dependent on the blood TF with leukemia connections, Tel1/Etv6, whereas the larger isoforms depend on another transcription-associated factor with leukemia connections, Eto2, raising the possibility that the regulation of VEGFA expression may be the mode of action of these leukemic factors. The action of Tel1/Etv6 in directly activating VEGFA expression in the somites was unexpected because this TF had only been reported to repress transcription. Using chromatin immunoprecipitation technology, we were able to show that Tel1/Etv6 does indeed work by repressing the expression of a VEGFA repressor, FoxC3, but it also acts directly to activate VEGFA expression, working together with Klf4. Finally, we have also looked at the mesodermal population that gives rise to the earlier waves of hematopoiesis, which do not generate a stem cell. We find significant differences including differential use of TFs of the E-Twenty-Six (ETS) family. In conclusion, we have elucidated the GRN responsible for preparing the lateral mesoderm for blood stem cell production.


Assuntos
Sangue/metabolismo , Redes Reguladoras de Genes , Regeneração/genética , Animais , Embrião de Mamíferos/metabolismo , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Nat Commun ; 10(1): 1083, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30842454

RESUMO

VEGFA signaling controls physiological and pathological angiogenesis and hematopoiesis. Although many context-dependent signaling pathways downstream of VEGFA have been uncovered, vegfa transcriptional regulation in vivo remains unclear. Here, we show that the ETS transcription factor, Etv6, positively regulates vegfa expression during Xenopus blood stem cell development through multiple transcriptional inputs. In agreement with its established repressive functions, Etv6 directly inhibits expression of the repressor foxo3, to prevent Foxo3 from binding to and repressing the vegfa promoter. Etv6 also directly activates expression of the activator klf4; reflecting a genome-wide paucity in ETS-binding motifs in Etv6 genomic targets, Klf4 then recruits Etv6 to the vegfa promoter to activate its expression. These two mechanisms (double negative gate and feed-forward loop) are classic features of gene regulatory networks specifying cell fates. Thus, Etv6's dual function, as a transcriptional repressor and activator, controls a major signaling pathway involved in endothelial and blood development in vivo.


Assuntos
Proteína Forkhead Box O3/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Repressoras/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/fisiologia , Animais , Embrião não Mamífero , Endotélio/embriologia , Endotélio/metabolismo , Proteína Forkhead Box O3/antagonistas & inibidores , Proteína Forkhead Box O3/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Redes Reguladoras de Genes/fisiologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Fatores de Transcrição Kruppel-Like/genética , Morfolinos/genética , Oligonucleotídeos Antissenso/genética , Proteínas Proto-Oncogênicas c-ets/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Transdução de Sinais/fisiologia , Somitos/embriologia , Somitos/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Xenopus/antagonistas & inibidores , Proteínas de Xenopus/genética , Variante 6 da Proteína do Fator de Translocação ETS
7.
Proc Natl Acad Sci U S A ; 114(23): 5814-5821, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28584091

RESUMO

Hematopoietic stem cells (HSCs) that sustain lifelong blood production are created during embryogenesis. They emerge from a specialized endothelial population, termed hemogenic endothelium (HE), located in the ventral wall of the dorsal aorta (DA). In Xenopus, we have been studying the gene regulatory networks (GRNs) required for the formation of HSCs, and critically found that the hemogenic potential is defined at an earlier time point when precursors to the DA express hematopoietic as well as endothelial genes, in the definitive hemangioblasts (DHs). The GRN for DH programming has been constructed and, here, we show that bone morphogenetic protein (BMP) signaling is essential for the initiation of this GRN. BMP2, -4, and -7 are the principal ligands expressed in the lineage forming the HE. To investigate the requirement and timing of all BMP signaling in HSC ontogeny, we have used a transgenic line, which inducibly expresses an inhibitor of BMP signaling, Noggin, as well as a chemical inhibitor of BMP receptors, DMH1, and described the inputs from BMP signaling into the DH GRN and the HE, as well as into primitive hematopoiesis. BMP signaling is required in at least three points in DH programming: first to initiate the DH GRN through gata2 expression, then for kdr expression to enable the DH to respond to vascular endothelial growth factor A (VEGFA) ligand from the somites, and finally for gata2 expression in the DA, but is dispensable for HE specification after hemangioblasts have been formed.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/genética , Linhagem da Célula , Redes Reguladoras de Genes , Células-Tronco Hematopoéticas/citologia , Animais , Animais Geneticamente Modificados , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Xenopus laevis
8.
FEBS Lett ; 590(22): 4002-4015, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27531714

RESUMO

Haematopoietic stem cells (HSCs) emerge from the haemogenic endothelium (HE) localised in the ventral wall of the embryonic dorsal aorta (DA). The HE generates HSCs through a process known as the endothelial to haematopoietic transition (EHT), which has been visualised in live embryos and is currently under intense study. However, EHT is the culmination of multiple programming events, which are as yet poorly understood, that take place before the specification of HE. A number of haematopoietic precursor cells have been described before the emergence of definitive HSCs, but only one haematovascular progenitor, the definitive haemangioblast (DH), gives rise to the DA, HE and HSCs. DHs emerge in the lateral plate mesoderm (LPM) and have a distinct origin and genetic programme compared to other, previously described haematovascular progenitors. Although DHs have so far only been established in Xenopus embryos, evidence for their existence in the LPM of mouse and chicken embryos is discussed here. We also review the current knowledge of the origins, lineage relationships, genetic programming and differentiation of the DHs that leads to the generation of HSCs. Importantly, we discuss the significance of the gene regulatory network (GRN) that controls the programming of DHs, a better understanding of which may aid in the establishment of protocols for the de novo generation of HSCs in vitro.


Assuntos
Desenvolvimento Embrionário/genética , Células-Tronco Hematopoéticas , Mesoderma/crescimento & desenvolvimento , Xenopus laevis/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Embrião de Galinha , Embrião de Mamíferos , Embrião não Mamífero , Hemangioblastos/metabolismo , Mesoderma/embriologia , Camundongos , Xenopus laevis/embriologia
9.
Dev Cell ; 38(4): 358-70, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27499523

RESUMO

Hematopoietic stem cells (HSCs) are self-renewing multipotent stem cells that generate mature blood lineages throughout life. They, together with hematopoietic progenitor cells (collectively known as HSPCs), emerge from hemogenic endothelium in the floor of the embryonic dorsal aorta by an endothelial-to-hematopoietic transition (EHT). Here we demonstrate that transforming growth factor ß (TGFß) is required for HSPC specification and that it regulates the expression of the Notch ligand Jagged1a in endothelial cells prior to EHT, in a striking parallel with the epithelial-to-mesenchymal transition (EMT). The requirement for TGFß is two fold and sequential: autocrine via Tgfß1a and Tgfß1b produced in the endothelial cells themselves, followed by a paracrine input of Tgfß3 from the notochord, suggesting that the former programs the hemogenic endothelium and the latter drives EHT. Our findings have important implications for the generation of HSPCs from pluripotent cells in vitro.


Assuntos
Endotélio Vascular/embriologia , Células-Tronco Hematopoéticas/citologia , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Transição Epitelial-Mesenquimal , Proteína Jagged-1/biossíntese , Proteína Jagged-1/genética , Morfolinos/genética , Células-Tronco Multipotentes/citologia , Notocorda/embriologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta3/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
10.
Nat Commun ; 5: 5588, 2014 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-25429520

RESUMO

Haematopoietic stem cells (HSCs) are produced during embryogenesis from the floor of the dorsal aorta. The localization of HSCs is dependent on the presence of instructive signals on the ventral side of the vessel. The nature of the extrinsic molecular signals that control the aortic haematopoietic niche is currently poorly understood. Here we demonstrate a novel requirement for FGF signalling in the specification of aortic haemogenic endothelium. Our results demonstrate that FGF signalling normally acts to repress BMP activity in the subaortic mesenchyme through transcriptional inhibition of bmp4, as well as through activation of two BMP antagonists, noggin2 and gremlin1a. Taken together, these findings demonstrate a key role for FGF signalling in establishment of the developmental HSC niche via its regulation of BMP activity in the subaortic mesenchyme. These results should help inform strategies to recapitulate the development of HSCs in vitro from pluripotent precursors.


Assuntos
Proteína Morfogenética Óssea 4/genética , Proteínas de Transporte/genética , Diferenciação Celular , Endotélio Vascular/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Peixe-Zebra/genética , Animais , Aorta/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Proteínas de Transporte/metabolismo , Mesoderma/metabolismo , Transdução de Sinais , Nicho de Células-Tronco , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
11.
Exp Hematol ; 42(8): 669-83, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24950425

RESUMO

Hematopoietic stem cells (HSCs) sustain blood production throughout life and are of pivotal importance in regenerative medicine. Although HSC generation from pluripotent stem cells would resolve their shortage for clinical applications, this has not yet been achieved mainly because of the poor mechanistic understanding of their programming. Bone marrow HSCs are first created during embryogenesis in the dorsal aorta (DA) of the midgestation conceptus, from where they migrate to the fetal liver and, eventually, the bone marrow. It is currently accepted that HSCs emerge from specialized endothelium, the hemogenic endothelium, localized in the ventral wall of the DA through an evolutionarily conserved process called the endothelial-to-hematopoietic transition. However, the endothelial-to-hematopoietic transition represents one of the last steps in HSC creation, and an understanding of earlier events in the specification of their progenitors is required if we are to create them from naïve pluripotent cells. Because of their ready availability and external development, zebrafish and Xenopus embryos have enormously facilitated our understanding of the early developmental processes leading to the programming of HSCs from nascent lateral plate mesoderm to hemogenic endothelium in the DA. The amenity of the Xenopus model to lineage tracing experiments has also contributed to the establishment of the distinct origins of embryonic (yolk sac) and adult (HSC) hematopoiesis, whereas the transparency of the zebrafish has allowed in vivo imaging of developing blood cells, particularly during and after the emergence of HSCs in the DA. Here, we discuss the key contributions of these model organisms to our understanding of developmental hematopoiesis.


Assuntos
Hematopoese , Animais , Diferenciação Celular , Células Endoteliais/citologia , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Xenopus/embriologia , Peixe-Zebra/embriologia
12.
Angiogenesis ; 17(1): 77-91, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23959107

RESUMO

Arterial and venous specification is critical for establishing and maintaining a functioning vascular system, and defects in key arteriovenous signaling pathways including VEGF (vascular endothelial growth factor) lead to congenital arteriopathies. The activities of VEGF, are in part controlled by heparan sulfate (HS) proteoglycans, significant components of the endothelial glycocalyx. The level of 6-O sulfation on HS polysaccharide chains, that mediate the interaction between HS and VEGFA, is edited at the cell surface by the enzyme SULF1. We investigated the role of sulf1 in vascular development. In zebrafish sulf1 is expressed in the head and tail vasculature, corresponding spatially and temporally with vascular development. Targeted knockdown of sulf1 by antisense morpholinos resulted in severe vascular patterning and maturation defects. 93 % of sulf1 morphants show dysmorphogenesis in arterial development leading to occlusion of the distal aorta and lack of axial and cranial circulation. Co-injection of vegfa165 mRNA rescued circulatory defects. While the genes affecting haematopoiesis are unchanged, expression of several arterial markers downstream of VegfA signalling such as notch and ephrinB2 are severely reduced in the dorsal aorta, with a concomitant increase in expression of the venous markers flt4 in the dorsal aorta of the morphants. Furthermore, in vitro, lack of SULF1 expression downregulates VEGFA-mediated arterial marker expression, confirming that Sulf1 mediates arterial specification by regulating VegfA165 activity. This study provides the first in vivo evidence for the integral role of the endothelial glycocalyx in specifying arterial-venous identity, vascular patterning and arterial integrity, and will help to better understand congenital arteriopathies.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Transdução de Sinais/fisiologia , Sulfatases/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/metabolismo , Animais , Artérias/embriologia , Artérias/metabolismo , Efrina-B2/imunologia , Efrina-B2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glicocálix/genética , Glicocálix/metabolismo , Morfolinos/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfatases/antagonistas & inibidores , Sulfatases/genética , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Veias/embriologia , Veias/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética
13.
Dev Cell ; 26(3): 237-49, 2013 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-23911199

RESUMO

Hematopoietic stem cells (HSCs) emerge during embryogenesis from hemogenic endothelium, but it remains unclear how the HSC lineage is initially established from mesoderm during ontogeny. In Xenopus, the definitive hemangioblast precursors of the HSC lineage have been identified in dorsal lateral plate (DLP) mesoderm, and a transcriptional gene regulatory network (GRN) controlling hemangioblast programming has been elucidated. Herein, we identify an essential role for microRNAs (miRNAs) in establishing the mesodermal lineage leading to both HSC emergence and vasculogenesis and determine that a single miRNA, miR-142-3p, is primarily responsible for initiation of definitive hemangioblast specification. miR-142-3p forms a double-negative gate unlocking entry into the hemangioblast program, in part by inhibiting TGFß signaling. Our results table miR-142-3p as a master regulator of HSC lineage specification, sitting at the apex of the hierarchy programming the adult hemangioblast, thus illustrating that miRNAs can act as instructive determinants of cell fate during development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Hemangioblastos/fisiologia , Células-Tronco Hematopoéticas/fisiologia , MicroRNAs/fisiologia , Neovascularização Fisiológica/genética , Animais , Diferenciação Celular , Linhagem da Célula/fisiologia , Desenvolvimento Embrionário/fisiologia , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Morfolinos , Proteínas/genética , Proteínas de Ligação a RNA , Xenopus laevis
14.
Blood Cells Mol Dis ; 51(4): 248-55, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23927967

RESUMO

Hematopoietic stem cells (HSCs) are essential for the maintenance of the hematopoietic system. However, these cells cannot be maintained or created in vitro, and very little is known about their generation during embryogenesis. Many transcription factors and signaling pathways play essential roles at various stages of HSC development. Members of the ETS ('E twenty-six') family of transcription factors are recognized as key regulators within the gene regulatory networks governing hematopoiesis, including the ontogeny of HSCs. Remarkably, although all ETS transcription factors bind the same DNA consensus sequence and overlapping tissue expression is observed, individual ETS transcription factors play unique roles in the development of HSCs. Also, these transcription factors are recurrently used throughout development and their functions are context-dependent, increasing the challenge of studying their mechanism of action. Critically, ETS factors also play roles under pathological conditions, such as leukemia and, therefore, deciphering their mechanism of action will not only enhance our knowledge of normal hematopoiesis, but also inform protocols for their creation in vitro from pluripotent stem cells and the design of new therapeutic approaches for the treatment of malignant blood cell diseases. In this review, we summarize the key findings on the roles of ETS transcription factors in HSC development and discuss novel mechanisms by which they could control hematopoiesis.


Assuntos
Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Linhagem da Célula , Transdiferenciação Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Hematopoese/fisiologia , Humanos , Mesoderma/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo
15.
Cancer Cell ; 24(2): 229-41, 2013 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-23871637

RESUMO

Limited clinical benefits derived from anti-VEGF therapy have driven the identification of new targets involved in tumor angiogenesis. Here, we report an integrative meta-analysis to define the transcriptional program underlying angiogenesis in human cancer. This approach identified ELTD1, an orphan G-protein-coupled receptor whose expression is induced by VEGF/bFGF and repressed by DLL4 signaling. Extensive analysis of multiple cancer types demonstrates significant upregulation of ELTD1 in tumor-associated endothelial cells, with a higher expression correlating with favorable prognosis. Importantly, ELTD1 silencing impairs endothelial sprouting and vessel formation in vitro and in vivo, drastically reducing tumor growth and greatly improving survival. Collectively, these results provide insight into the regulation of tumor angiogenesis and highlight ELTD1 as key player in blood vessel formation.


Assuntos
Células Endoteliais/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Células Endoteliais/patologia , Feminino , Predisposição Genética para Doença , Células HCT116 , Humanos , Camundongos , Camundongos Nus , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais
16.
Cell Rep ; 4(1): 135-47, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23831025

RESUMO

Cell fate is governed by combinatorial actions of transcriptional regulators assembling into multiprotein complexes. However, the molecular details of how these complexes form are poorly understood. One such complex, which contains the basic-helix-loop-helix heterodimer SCL:E47 and bridging proteins LMO2:LDB1, critically regulates hematopoiesis and induces T cell leukemia. Here, we report the crystal structure of (SCL:E47)bHLH:LMO2:LDB1LID bound to DNA, providing a molecular account of the network of interactions assembling this complex. This reveals an unexpected role for LMO2. Upon binding to SCL, LMO2 induces new hydrogen bonds in SCL:E47, thereby strengthening heterodimer formation. This imposes a rotation movement onto E47 that weakens the heterodimer:DNA interaction, shifting the main DNA-binding activity onto additional protein partners. Along with biochemical analyses, this illustrates, at an atomic level, how hematopoietic-specific SCL sequesters ubiquitous E47 and associated cofactors and supports SCL's reported DNA-binding-independent functions. Importantly, this work will drive the design of small molecules inhibiting leukemogenic processes.


Assuntos
DNA/química , Hematopoese/genética , Proteínas com Domínio LIM/química , Simulação de Acoplamento Molecular , Transcrição Gênica , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular Tumoral , DNA/metabolismo , Células HEK293 , Humanos , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Camundongos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mutação , Multimerização Proteica , Peixe-Zebra
17.
Development ; 140(12): 2632-42, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23637333

RESUMO

The first haematopoietic stem cells share a common origin with the dorsal aorta and derive from putative adult haemangioblasts in the dorsal lateral plate (DLP) mesoderm. Here we show that the transcription factor (TF) stem cell leukaemia (Scl/Tal1) is crucial for development of these adult haemangioblasts in Xenopus and establish the regulatory cascade controlling its expression. We show that VEGFA produced in the somites is required to initiate adult haemangioblast programming in the adjacent DLP by establishing endogenous VEGFA signalling. This response depends on expression of the VEGF receptor Flk1, driven by Fli1 and Gata2. Scl activation requires synergy between this VEGFA-controlled pathway and a VEGFA-independent pathway controlled by Fli1, Gata2 and Etv2/Etsrp/ER71, which also drives expression of the Scl partner Lmo2. Thus, the two ETS factors Fli1 and Etv6, which drives the VEGFA expression in both somites and the DLP, sit at the top of the adult haemangioblast gene regulatory network (GRN). Furthermore, Gata2 is initially activated by Fli1 but later maintained by another ETS factor, Etv2. We also establish that Flk1 and Etv2 act independently in the two pathways to Scl activation. Thus, detailed temporal, epistatic measurements of key TFs and VEGFA plus its receptor have enabled us to build a Xenopus adult haemangioblast GRN.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Hemangioblastos/citologia , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animais , Western Blotting , Linhagem da Célula , Clonagem Molecular , Proteínas de Ligação a DNA/genética , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Redes Reguladoras de Genes , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Morfolinos/administração & dosagem , Morfolinos/farmacologia , Proteína Proto-Oncogênica c-fli-1/genética , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Somitos/citologia , Somitos/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis/sangue , Variante 6 da Proteína do Fator de Translocação ETS
18.
Blood ; 122(3): 367-75, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23591790

RESUMO

Reprogramming of somatic cells to desired cell types holds great promise in regenerative medicine. However, production of transplantable hematopoietic stem cells (HSCs) in vitro by defined factors has not yet been achieved. Therefore, it is critical to fully understand the molecular mechanisms of HSC development in vivo. Here, we show that Fev, an ETS transcription factor, is a pivotal regulator of HSC development in vertebrates. In fev-deficient zebrafish embryos, the first definitive HSC population was compromised and fewer T cells were found in the thymus. Genetic and chemical analyses support a mechanism whereby Fev regulates HSC through direct regulation of ERK signaling. Blastula transplant assay demonstrates that Fev regulation of HSC development is cell autonomous. Experiments performed with purified cord blood show that fev is expressed and functions in primitive HSCs in humans, indicating its conserved role in higher vertebrates. Our data indicate that Fev-ERK signaling is essential for hemogenic endothelium-based HSC development.


Assuntos
Proteínas de Ligação a DNA/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/enzimologia , Sistema de Sinalização das MAP Quinases , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Aorta/metabolismo , Linhagem da Célula , Embrião não Mamífero/citologia , Embrião não Mamífero/enzimologia , Endotélio/metabolismo , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Humanos
19.
Dev Cell ; 24(2): 144-58, 2013 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-23318133

RESUMO

VEGFA signaling is critical for endothelial and hematopoietic stem cell (HSC) specification. However, blood defects resulting from perturbation of the VEGFA pathway are always accompanied by impaired vascular/arterial development. Because HSCs derive from arterial cells, it is unclear whether VEGFA directly contributes to HSC specification. This is an important question for our understanding of how HSCs are formed and for developing their production in vitro. Through knockdown of the regulator ETO2 in embryogenesis, we report a specific decrease in expression of medium/long Vegfa isoforms in somites. This leads to absence of Notch1 expression and failure of HSC specification in the dorsal aorta (DA), independently of vessel formation and arterial specification. Vegfa hypomorphs and isoform-specific (medium/long) morphants not only recapitulate this phenotype but also demonstrate that VEGFA short isoform is sufficient for DA development. Therefore, sequential, isoform-specific VEGFA signaling successively induces the endothelial, arterial, and HSC programs in the DA.


Assuntos
Aorta/embriologia , Artérias/embriologia , Proteínas Correpressoras/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Animais , Aorta/metabolismo , Artérias/metabolismo , Proteínas Correpressoras/genética , Desenvolvimento Embrionário , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Morfolinos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transdução de Sinais , Fatores de Transcrição , Transcrição Gênica , Proteínas de Xenopus/genética , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
20.
Biochim Biophys Acta ; 1830(2): 2370-4, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22705943

RESUMO

BACKGROUND: Hematopoietic stem cells (HSCs) are a population of multipotent cells that can self-renew and differentiate into all blood lineages. HSC development must be tightly controlled from cell fate determination to self-maintenance during adulthood. This involves a panel of important developmental signaling pathways and other factors which act synergistically within the HSC population and/or in the HSC niche. Genetically conserved processes of HSC development plus many other developmental advantages make the zebrafish an ideal model organism to elucidate the regulatory mechanisms underlying HSC programming. SCOPE OF REVIEW: This review summarizes recent progress on zebrafish HSCs with particular focus on how developmental signaling controls hemogenic endothelium-derived HSC development. We also describe the interaction of different signaling pathways during these processes. MAJOR CONCLUSIONS: The hematopoietic stem cell system is a paradigm for stem cell studies. Use of the zebrafish model to study signaling regulation of HSCs in vivo has resulted in a great deal of information concerning HSC biology in vertebrates. GENERAL SIGNIFICANCE: These new findings facilitate a better understanding of molecular mechanisms of HSC programming, and will provide possible new strategies for the treatment of HSC-related hematological diseases, such as leukemia. This article is part of a Special Issue entitled Biochemistry of Stem Cells.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Peixe-Zebra/metabolismo , Animais , Padronização Corporal , Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Peixe-Zebra/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA