Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Methods Mol Biol ; 2608: 425-450, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36653721

RESUMO

Analysis of cardiovascular development in zebrafish embryos has become a major driver of vascular research in recent years. Imaging-based analyses have allowed the discovery or verification of morphologically distinct processes and mechanisms of, e.g., endothelial cell migration, angiogenic sprouting, tip or stalk cell behavior, and vessel anastomosis. In this chapter, we describe the techniques and tools used for confocal imaging of zebrafish endothelial development in combination with general experimental approaches for molecular dissection of involved signaling pathways.


Assuntos
Transdução de Sinais , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Morfogênese , Proteínas de Peixe-Zebra/metabolismo , Movimento Celular , Neovascularização Fisiológica
2.
Nature ; 606(7914): 570-575, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35614218

RESUMO

The lineage and developmental trajectory of a cell are key determinants of cellular identity. In the vascular system, endothelial cells (ECs) of blood and lymphatic vessels differentiate and specialize to cater to the unique physiological demands of each organ1,2. Although lymphatic vessels were shown to derive from multiple cellular origins, lymphatic ECs (LECs) are not known to generate other cell types3,4. Here we use recurrent imaging and lineage-tracing of ECs in zebrafish anal fins, from early development to adulthood, to uncover a mechanism of specialized blood vessel formation through the transdifferentiation of LECs. Moreover, we demonstrate that deriving anal-fin vessels from lymphatic versus blood ECs results in functional differences in the adult organism, uncovering a link between cell ontogeny and functionality. We further use single-cell RNA-sequencing analysis to characterize the different cellular populations and transition states involved in the transdifferentiation process. Finally, we show that, similar to normal development, the vasculature is rederived from lymphatics during anal-fin regeneration, demonstrating that LECs in adult fish retain both potency and plasticity for generating blood ECs. Overall, our research highlights an innate mechanism of blood vessel formation through LEC transdifferentiation, and provides in vivo evidence for a link between cell ontogeny and functionality in ECs.


Assuntos
Vasos Sanguíneos , Transdiferenciação Celular , Vasos Linfáticos , Nadadeiras de Animais/citologia , Animais , Vasos Sanguíneos/citologia , Linhagem da Célula , Células Endoteliais/citologia , Vasos Linfáticos/citologia , Peixe-Zebra
3.
Angiogenesis ; 24(3): 695-714, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33983539

RESUMO

Sprouting angiogenesis is key to many pathophysiological conditions, and is strongly regulated by vascular endothelial growth factor (VEGF) signaling through VEGF receptor 2 (VEGFR2). Here we report that the early endosomal GTPase Rab5C and its activator RIN2 prevent lysosomal routing and degradation of VEGF-bound, internalized VEGFR2 in human endothelial cells. Stabilization of endosomal VEGFR2 levels by RIN2/Rab5C is crucial for VEGF signaling through the ERK and PI3-K pathways, the expression of immediate VEGF target genes, as well as specification of angiogenic 'tip' and 'stalk' cell phenotypes and cell sprouting. Using overexpression of Rab mutants, knockdown and CRISPR/Cas9-mediated gene editing, and live-cell imaging in zebrafish, we further show that endosomal stabilization of VEGFR2 levels is required for developmental angiogenesis in vivo. In contrast, the premature degradation of internalized VEGFR2 disrupts VEGF signaling, gene expression, and tip cell formation and migration. Thus, an endosomal feedforward mechanism maintains receptor signaling by preventing lysosomal degradation, which is directly linked to the induction of target genes and cell fate in collectively migrating cells during morphogenesis.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica , Proteólise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Proteínas de Transporte/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Peixe-Zebra/genética , Proteínas rab5 de Ligação ao GTP/genética
4.
Elife ; 92020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32955436

RESUMO

To form new blood vessels (angiogenesis), endothelial cells (ECs) must be activated and acquire highly migratory and proliferative phenotypes. However, the molecular mechanisms that govern these processes are incompletely understood. Here, we show that Apelin signaling functions to drive ECs into such an angiogenic state. Zebrafish lacking Apelin signaling exhibit defects in endothelial tip cell morphology and sprouting. Using transplantation experiments, we find that in mosaic vessels, wild-type ECs leave the dorsal aorta (DA) and form new vessels while neighboring ECs defective in Apelin signaling remain in the DA. Mechanistically, Apelin signaling enhances glycolytic activity in ECs at least in part by increasing levels of the growth-promoting transcription factor c-Myc. Moreover, APELIN expression is regulated by Notch signaling in human ECs, and its function is required for the hypersprouting phenotype in Delta-like 4 (Dll4) knockdown zebrafish embryos. These data provide new insights into fundamental principles of blood vessel formation and Apelin signaling, enabling a better understanding of vascular growth in health and disease.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Quimiocinas/genética , Morfogênese/genética , Transdução de Sinais/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Vasos Sanguíneos/metabolismo , Quimiocinas/metabolismo , Células Endoteliais/metabolismo , Proteínas de Peixe-Zebra/metabolismo
5.
Dev Biol ; 457(2): 181-190, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30862465

RESUMO

To ensure tissue homeostasis the brain needs to be protected from blood-derived fluctuations or pathogens that could affect its function. Therefore, the brain capillaries develop tissue-specific properties to form a selective blood-brain barrier (BBB), allowing the passage of essential molecules to the brain and blocking the penetration of potentially harmful compounds or cells. Previous studies reported the presence of this barrier in zebrafish. The intrinsic features of the zebrafish embryos and larvae in combination with optical techniques, make them suitable for the study of barrier establishment and maturation. In this review, we discuss the most recent contributions to the development and formation of a functional zebrafish BBB. Moreover, we compare the molecular and cellular characteristic of the zebrafish and the mammalian BBB.


Assuntos
Barreira Hematoencefálica/embriologia , Encéfalo/irrigação sanguínea , Sistema Cardiovascular/embriologia , Neovascularização Fisiológica/fisiologia , Peixe-Zebra/embriologia , Animais , Encéfalo/embriologia , Células Endoteliais/fisiologia , Endotélio Vascular/embriologia , Endotélio Vascular/fisiologia , Junções Íntimas/fisiologia
6.
Nat Commun ; 10(1): 4113, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31511517

RESUMO

Intra-organ communication guides morphogenetic processes that are essential for an organ to carry out complex physiological functions. In the heart, the growth of the myocardium is tightly coupled to that of the endocardium, a specialized endothelial tissue that lines its interior. Several molecular pathways have been implicated in the communication between these tissues including secreted factors, components of the extracellular matrix, or proteins involved in cell-cell communication. Yet, it is unknown how the growth of the endocardium is coordinated with that of the myocardium. Here, we show that an increased expansion of the myocardial atrial chamber volume generates higher junctional forces within endocardial cells. This leads to biomechanical signaling involving VE-cadherin, triggering nuclear localization of the Hippo pathway transcriptional regulator Yap1 and endocardial proliferation. Our work suggests that the growth of the endocardium results from myocardial chamber volume expansion and ends when the tension on the tissue is relaxed.


Assuntos
Endocárdio/crescimento & desenvolvimento , Miocárdio/metabolismo , Transdução de Sinais , Peixe-Zebra/embriologia , Animais , Antígenos CD/metabolismo , Fenômenos Biomecânicos , Caderinas/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células , Tamanho Celular , Proteínas do Citoesqueleto/metabolismo , Endocárdio/citologia , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Proteína Homeobox Nkx-2.5/metabolismo , Junções Intercelulares/metabolismo , Modelos Biológicos , Mutação/genética , Transativadores/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Sinalização YAP , Proteínas de Peixe-Zebra/metabolismo
7.
Nat Commun ; 10(1): 453, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692543

RESUMO

Venous endothelial cells are molecularly and functionally distinct from their arterial counterparts. Although veins are often considered the default endothelial state, genetic manipulations can modulate both acquisition and loss of venous fate, suggesting that venous identity is the result of active transcriptional regulation. However, little is known about this process. Here we show that BMP signalling controls venous identity via the ALK3/BMPR1A receptor and SMAD1/SMAD5. Perturbations to TGF-ß and BMP signalling in mice and zebrafish result in aberrant vein formation and loss of expression of the venous-specific gene Ephb4, with no effect on arterial identity. Analysis of a venous endothelium-specific enhancer for Ephb4 shows enriched binding of SMAD1/5 and a requirement for SMAD binding motifs. Further, our results demonstrate that BMP/SMAD-mediated Ephb4 expression requires the venous-enriched BMP type I receptor ALK3/BMPR1A. Together, our analysis demonstrates a requirement for BMP signalling in the establishment of Ephb4 expression and the venous vasculature.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Proteínas Morfogenéticas Ósseas/genética , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais/genética , Veias/metabolismo , Animais , Animais Geneticamente Modificados , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Células Endoteliais/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Receptor EphB4/genética , Receptor EphB4/metabolismo , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Veias/embriologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
Nat Commun ; 9(1): 4860, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30451830

RESUMO

Canonical Wnt signaling is crucial for vascularization of the central nervous system and blood-brain barrier (BBB) formation. BBB formation and modulation are not only important for development, but also relevant for vascular and neurodegenerative diseases. However, there is little understanding of how Wnt signaling contributes to brain angiogenesis and BBB formation. Here we show, using high resolution in vivo imaging and temporal and spatial manipulation of Wnt signaling, different requirements for Wnt signaling during brain angiogenesis and BBB formation. In the absence of Wnt signaling, premature Sphingosine-1-phosphate receptor (S1pr) signaling reduces VE-cadherin and Esama at cell-cell junctions. We suggest that Wnt signaling suppresses S1pr signaling during angiogenesis to enable the dynamic junction formation during anastomosis, whereas later S1pr signaling regulates BBB maturation and VE-cadherin stabilization. Our data provides a link between brain angiogenesis and BBB formation and identifies Wnt signaling as coordinator of the timing and as regulator of anastomosis.


Assuntos
Antígenos CD/genética , Encéfalo/metabolismo , Caderinas/genética , Neovascularização Fisiológica/genética , Receptores de Lisoesfingolipídeo/genética , Via de Sinalização Wnt , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , beta Catenina/genética , Animais , Animais Geneticamente Modificados , Antígenos CD/metabolismo , Barreira Hematoencefálica/crescimento & desenvolvimento , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/crescimento & desenvolvimento , Caderinas/metabolismo , Capilares/crescimento & desenvolvimento , Capilares/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Circulação Cerebrovascular/genética , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo , beta Catenina/metabolismo , Proteína Vermelha Fluorescente
9.
Dev Biol ; 430(1): 142-155, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28811218

RESUMO

During vertebrate embryogenesis, vascular endothelial cells (ECs) and primitive erythrocytes become specified within close proximity in the posterior lateral plate mesoderm (LPM) from a common progenitor. However, the signaling cascades regulating the specification into either lineage remain largely elusive. Here, we analyze the contribution of ß-catenin dependent Wnt signaling to EC and erythrocyte specification during zebrafish embryogenesis. We generated novel ß-catenin dependent Wnt signaling reporters which, by using destabilized fluorophores (Venus-Pest, dGFP), specifically allow us to detect Wnt signaling responses in narrow time windows as well as in spatially restricted domains, defined by Cre recombinase expression (Tg(axin2BAC:Venus-Pest)mu288; Tg(14TCF:loxP-STOP-loxP-dGFP)mu202). We therefore can detect ß-catenin dependent Wnt signaling activity in a subset of the Fli1a-positive progenitor population. Additionally, we show that mesodermal Wnt3a-mediated signaling via the transcription factor Lef1 positively regulates EC specification (defined by kdrl expression) at the expense of primitive erythrocyte specification (defined by gata1 expression) in zebrafish embryos. Using mesoderm derived from human embryonic stem cells, we identified the same principle of Wnt signaling dependent EC specification in conjunction with auto-upregulation of LEF1. Our data indicate a novel role of ß-catenin dependent Wnt signaling in regulating EC specification during vasculogenesis.


Assuntos
Linhagem da Célula , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Contagem de Células , Diferenciação Celular , Linhagem Celular , Eritrócitos/citologia , Eritrócitos/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Modelos Biológicos , Organogênese , Somitos/embriologia , Somitos/metabolismo , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
10.
Nat Cell Biol ; 19(6): 653-665, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28530658

RESUMO

The hierarchical organization of properly sized blood vessels ensures the correct distribution of blood to all organs of the body, and is controlled via haemodynamic cues. In current concepts, an endothelium-dependent shear stress set point causes blood vessel enlargement in response to higher flow rates, while lower flow would lead to blood vessel narrowing, thereby establishing homeostasis. We show that during zebrafish embryonic development increases in flow, after an initial expansion of blood vessel diameters, eventually lead to vessel contraction. This is mediated via endothelial cell shape changes. We identify the transforming growth factor beta co-receptor endoglin as an important player in this process. Endoglin mutant cells and blood vessels continue to enlarge in response to flow increases, thus exacerbating pre-existing embryonic arterial-venous shunts. Together, our data suggest that cell shape changes in response to biophysical cues act as an underlying principle allowing for the ordered patterning of tubular organs.


Assuntos
Forma Celular , Endoglina/metabolismo , Células Endoteliais/metabolismo , Hemodinâmica , Mecanotransdução Celular , Proteínas de Peixe-Zebra/metabolismo , Animais , Malformações Arteriovenosas/genética , Malformações Arteriovenosas/metabolismo , Malformações Arteriovenosas/fisiopatologia , Endoglina/deficiência , Endoglina/genética , Predisposição Genética para Doença , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos Knockout , Mutação , Neovascularização Fisiológica , Fenótipo , Fluxo Sanguíneo Regional , Estresse Mecânico , Fatores de Tempo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
11.
J Cell Biol ; 215(3): 415-430, 2016 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-27799363

RESUMO

During cardiovascular development, tight spatiotemporal regulation of molecular cues is essential for controlling endothelial cell (EC) migration. Secreted class III Semaphorins play an important role in guidance of neuronal cell migration and were lately linked to regulating cardiovascular development. Recently, SEMA3D gene disruptions were associated with cardiovascular defects in patients; however, the mechanisms of action were not revealed. Here we show for the first time that Sema3d regulates collective EC migration in zebrafish through two separate mechanisms. Mesenchymal Sema3d guides outgrowth of the common cardinal vein via repulsion and signals through PlexinD1. Additionally, within the same ECs, we identified a novel function of autocrine Sema3d signaling in regulating Actin network organization and EC morphology. We show that this new function requires Sema3d signaling through Neuropilin1, which then regulates Actin network organization through RhoA upstream of Rock, stabilizing the EC sheet. Our findings are highly relevant for understanding EC migration and the mechanisms of collective migration in other contexts.


Assuntos
Movimento Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fatores de Crescimento Neural/metabolismo , Neuropilina-1/metabolismo , Receptores de Superfície Celular/metabolismo , Semaforinas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Actinas/metabolismo , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Caderinas/metabolismo , Comunicação Celular , Forma Celular , Mesoderma/citologia , Mesoderma/metabolismo , Modelos Biológicos , Pseudópodes/metabolismo , Transdução de Sinais , Peixe-Zebra/embriologia , Proteína rhoA de Ligação ao GTP/metabolismo
12.
Cell Rep ; 17(6): 1595-1606, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27806298

RESUMO

All mature blood cell types in the adult animal arise from hematopoietic stem and progenitor cells (HSPCs). However, the developmental cues regulating HSPC ontogeny are incompletely understood. In particular, the details surrounding a requirement for Wnt/ß-catenin signaling in the development of mature HSPCs are controversial and difficult to consolidate. Using zebrafish, we demonstrate that Wnt signaling is required to direct an amplification of HSPCs in the aorta. Wnt9a is specifically required for this process and cannot be replaced by Wnt9b or Wnt3a. This proliferative event occurs independently of initial HSPC fate specification, and the Wnt9a input is required prior to aorta formation. HSPC arterial amplification occurs prior to seeding of secondary hematopoietic tissues and proceeds, in part, through the cell cycle regulator myca (c-myc). Our results support a general paradigm, in which early signaling events, including Wnt, direct later HSPC developmental processes.


Assuntos
Aorta/citologia , Aorta/embriologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Contagem de Células , Ciclo Celular , Proliferação de Células , Hemangioblastos/metabolismo , Via de Sinalização Wnt
13.
Elife ; 52016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27852438

RESUMO

Vascular networks surrounding individual organs are important for their development, maintenance, and function; however, how these networks are assembled remains poorly understood. Here we show that CNS progenitors, referred to as radial glia, modulate vascular patterning around the spinal cord by acting as negative regulators. We found that radial glia ablation in zebrafish embryos leads to excessive sprouting of the trunk vessels around the spinal cord, and exclusively those of venous identity. Mechanistically, we determined that radial glia control this process via the Vegf decoy receptor sFlt1: sflt1 mutants exhibit the venous over-sprouting observed in radial glia-ablated larvae, and sFlt1 overexpression rescues it. Genetic mosaic analyses show that sFlt1 function in trunk endothelial cells can limit their over-sprouting. Together, our findings identify CNS-resident progenitors as critical angiogenic regulators that determine the precise patterning of the vasculature around the spinal cord, providing novel insights into vascular network formation around developing organs.


Assuntos
Diferenciação Celular/genética , Organogênese/genética , Medula Espinal/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Proteínas de Peixe-Zebra/genética , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Mosaicismo , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Transdução de Sinais/genética , Medula Espinal/irrigação sanguínea , Medula Espinal/crescimento & desenvolvimento , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
14.
Nat Commun ; 7: 11805, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27248505

RESUMO

Endothelial cells (ECs) respond to shear stress by aligning in the direction of flow. However, how ECs respond to flow in complex in vivo environments is less clear. Here we describe an endothelial-specific transgenic zebrafish line, whereby the Golgi apparatus is labelled to allow for in vivo analysis of endothelial polarization. We find that most ECs polarize within 4.5 h after the onset of vigorous blood flow and, by manipulating cardiac function, observe that flow-induced EC polarization is a dynamic and reversible process. Based on its role in EC migration, we analyse the role of Apelin signalling in EC polarization and find that it is critical for this process. Knocking down Apelin receptor function in human primary ECs also affects their polarization. Our study provides new tools to analyse the mechanisms of EC polarization in vivo and reveals an important role in this process for a signalling pathway implicated in cardiovascular disease.


Assuntos
Receptores de Apelina/genética , Apelina/genética , Polaridade Celular , Quimiocinas/genética , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Apelina/metabolismo , Receptores de Apelina/metabolismo , Fenômenos Biomecânicos , Movimento Celular , Quimiocinas/metabolismo , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Complexo de Golgi/metabolismo , Hemorreologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/ultraestrutura , Humanos , Hibridização in Situ Fluorescente , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Transdução de Sinais , Estresse Mecânico , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
15.
Development ; 143(12): 2217-27, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27302398

RESUMO

During cardiac valve development, the single-layered endocardial sheet at the atrioventricular canal (AVC) is remodeled into multilayered immature valve leaflets. Most of our knowledge about this process comes from examining fixed samples that do not allow a real-time appreciation of the intricacies of valve formation. Here, we exploit non-invasive in vivo imaging techniques to identify the dynamic cell behaviors that lead to the formation of the immature valve leaflets. We find that in zebrafish, the valve leaflets consist of two sets of endocardial cells at the luminal and abluminal side, which we refer to as luminal cells (LCs) and abluminal cells (ALCs), respectively. By analyzing cellular rearrangements during valve formation, we observed that the LCs and ALCs originate from the atrium and ventricle, respectively. Furthermore, we utilized Wnt/ß-catenin and Notch signaling reporter lines to distinguish between the LCs and ALCs, and also found that cardiac contractility and/or blood flow is necessary for the endocardial expression of these signaling reporters. Thus, our 3D analyses of cardiac valve formation in zebrafish provide fundamental insights into the cellular rearrangements underlying this process.


Assuntos
Valvas Cardíacas/citologia , Valvas Cardíacas/embriologia , Imageamento Tridimensional , Animais , Movimento Celular , Circulação Coronária , Endocárdio/citologia , Endocárdio/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Átrios do Coração/citologia , Átrios do Coração/embriologia , Ventrículos do Coração/citologia , Ventrículos do Coração/embriologia , Mutação/genética , Contração Miocárdica , Organogênese/genética , Receptores Notch/metabolismo , Via de Sinalização Wnt , Peixe-Zebra
16.
Cell Rep ; 15(4): 787-800, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27149845

RESUMO

It has previously been reported that mouse epiblast stem cell (EpiSC) lines comprise heterogeneous cell populations that are functionally equivalent to cells of either early- or late-stage postimplantation development. So far, the establishment of the embryonic stem cell (ESC) pluripotency gene regulatory network through the widely known chemical inhibition of MEK and GSK3beta has been impractical in late-stage EpiSCs. Here, we show that chemical inhibition of casein kinase 1alpha (CK1alpha) induces the conversion of recalcitrant late-stage EpiSCs into ESC pluripotency. CK1alpha inhibition directly results in the simultaneous activation of the WNT signaling pathway, together with inhibition of the TGFbeta/SMAD2 signaling pathway, mediating the rewiring of the gene regulatory network in favor of an ESC-like state. Our findings uncover a molecular mechanism that links CK1alpha to ESC pluripotency through the direct modulation of WNT and TGFbeta signaling.

17.
Angiogenesis ; 18(4): 463-75, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26198291

RESUMO

Analysis of developmental angiogenesis can help to identify regulatory networks, which also contribute to disease-related vascular growth. Vascular endothelial growth factors (Vegf) drive angiogenic processes such as sprouting, endothelial cell (EC) migration and proliferation. However, how Vegf expression is regulated during development is not well understood. By analyzing developmental zebrafish angiogenesis, we have identified Metallothionein 2 (Mt2) as a novel regulator of vegfc expression. While Metallothioneins (Mts) have been extensively analyzed for their capability of regulating homeostasis and metal detoxification, we demonstrate that Mt2 is required for EC migration, proliferation and angiogenic sprouting upstream of vegfc expression. We further demonstrate that another Mt family member cannot compensate Mt2 deficiency and therefore postulate that Mt2 regulates angiogenesis independent of its canonical Mt function. Our data not only reveal a non-canonical function of Mt2 in angiogenesis, but also propose Mt2 as a novel regulator of vegfc expression.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Metalotioneína/metabolismo , Neovascularização Fisiológica/fisiologia , Transcrição Gênica/fisiologia , Fator C de Crescimento do Endotélio Vascular/biossíntese , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/metabolismo , Animais , Células Endoteliais/citologia , Metalotioneína/genética , Fator C de Crescimento do Endotélio Vascular/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
18.
Elife ; 42015 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-26017639

RESUMO

A key step in the de novo formation of the embryonic vasculature is the migration of endothelial precursors, the angioblasts, to the position of the future vessels. To form the first axial vessels, angioblasts migrate towards the midline and coalesce underneath the notochord. Vascular endothelial growth factor has been proposed to serve as a chemoattractant for the angioblasts and to regulate this medial migration. Here we challenge this model and instead demonstrate that angioblasts rely on their intrinsic expression of Apelin receptors (Aplr, APJ) for their migration to the midline. We further show that during this angioblast migration Apelin receptor signaling is mainly triggered by the recently discovered ligand Elabela (Ela). As neither of the ligands Ela or Apelin (Apln) nor their receptors have previously been implicated in regulating angioblast migration, we hereby provide a novel mechanism for regulating vasculogenesis, with direct relevance to physiological and pathological angiogenesis.


Assuntos
Movimento Celular/fisiologia , Quimiocinas/metabolismo , Células Endoteliais/citologia , Células Progenitoras Endoteliais/fisiologia , Modelos Biológicos , Neovascularização Fisiológica/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Clonagem Molecular , Primers do DNA/genética , Células Progenitoras Endoteliais/metabolismo , Humanos , Hibridização In Situ , Mutagênese , Peixe-Zebra
19.
Semin Cell Dev Biol ; 31: 106-14, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24813365

RESUMO

The vasculature consists of an extensively branched network of blood and lymphatic vessels that ensures the efficient circulation and thereby the supply of all tissues with oxygen and nutrients. Research within the last decade has tremendously advanced our understanding of how this complex network is formed, how angiogenic growth is controlled and how differences between individual endothelial cells contribute to achieving this complex pattern. The small size and the optical clarity of the zebrafish embryo in combination with the advancements in imaging technologies cleared the way for the zebrafish as an important in vivo model for elucidating the mechanisms of angiogenesis. In this review we discuss the recent contributions of the analysis of zebrafish vascular development on how vessels establish their characteristic morphology and become patent. We focus on the morphogenetic cellular behaviors as well as the molecular mechanisms that drive these processes in the developing zebrafish embryo.


Assuntos
Neovascularização Fisiológica , Peixe-Zebra , Animais , Endotélio Vascular/citologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Peixe-Zebra/embriologia
20.
Nat Commun ; 5: 3743, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24806444

RESUMO

The assembly of individual endothelial cells into multicellular tubes is a complex morphogenetic event in vascular development. Extracellular matrix cues and cell-cell junctional communication are fundamental to tube formation. Together they determine the shape of endothelial cells and the tubular structures that they ultimately form. Little is known regarding how mechanical signals are transmitted between cells to control cell shape changes during morphogenesis. Here we provide evidence that the scaffold protein amotL2 is needed for aortic vessel lumen expansion. Using gene inactivation strategies in zebrafish, mouse and endothelial cell culture systems, we show that amotL2 associates to the VE-cadherin adhesion complex where it couples adherens junctions to contractile actin fibres. Inactivation of amotL2 dissociates VE-cadherin from cytoskeletal tensile forces that affect endothelial cell shape. We propose that the VE-cadherin/amotL2 complex is responsible for transmitting mechanical force between endothelial cells for the coordination of cellular morphogenesis consistent with aortic lumen expansion and function.


Assuntos
Antígenos CD/metabolismo , Aorta/crescimento & desenvolvimento , Caderinas/metabolismo , Proteínas Contráteis/metabolismo , Proteínas de Membrana/metabolismo , Neovascularização Fisiológica/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Citoesqueleto de Actina/metabolismo , Junções Aderentes/metabolismo , Angiomotinas , Animais , Aorta/citologia , Comunicação Celular , Forma Celular , Células Endoteliais/citologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Matriz Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Morfolinos/genética , Interferência de RNA , RNA Interferente Pequeno , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA