Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 562(7726): 223-228, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30258231

RESUMO

The earliest blood vessels in mammalian embryos are formed when endothelial cells differentiate from angioblasts and coalesce into tubular networks. Thereafter, the endothelium is thought to expand solely by proliferation of pre-existing endothelial cells. Here we show that a complementary source of endothelial cells is recruited into pre-existing vasculature after differentiation from the earliest precursors of erythrocytes, megakaryocytes and macrophages, the erythro-myeloid progenitors (EMPs) that are born in the yolk sac. A first wave of EMPs contributes endothelial cells to the yolk sac endothelium, and a second wave of EMPs colonizes the embryo and contributes endothelial cells to intraembryonic endothelium in multiple organs, where they persist into adulthood. By demonstrating that EMPs constitute a hitherto unrecognized source of endothelial cells, we reveal that embryonic blood vascular endothelium expands in a dual mechanism that involves both the proliferation of pre-existing endothelial cells and the incorporation of endothelial cells derived from haematopoietic precursors.


Assuntos
Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Linhagem da Célula , Células Endoteliais/citologia , Eritrócitos/citologia , Células Progenitoras Mieloides/citologia , Envelhecimento , Animais , Linhagem da Célula/genética , Proliferação de Células , Células Endoteliais/metabolismo , Eritrócitos/metabolismo , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Integrases/genética , Integrases/metabolismo , Fígado/citologia , Fígado/embriologia , Camundongos , Células Progenitoras Mieloides/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Rombencéfalo/irrigação sanguínea , Rombencéfalo/citologia , Rombencéfalo/embriologia , Transcrição Gênica , Saco Vitelino/citologia , Saco Vitelino/embriologia
2.
Angiogenesis ; 25(3): 343-353, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35416527

RESUMO

Blood vessels form vast networks in all vertebrate organs to sustain tissue growth, repair and homeostatic metabolism, but they also contribute to a range of diseases with neovascularisation. It is, therefore, important to define the molecular mechanisms that underpin blood vessel growth. The receptor tyrosine kinase KIT is required for the normal expansion of hematopoietic progenitors that arise during embryogenesis from hemogenic endothelium in the yolk sac and dorsal aorta. Additionally, KIT has been reported to be expressed in endothelial cells during embryonic brain vascularisation and has been implicated in pathological angiogenesis. However, it is neither known whether KIT expression is widespread in normal organ endothelium nor whether it promotes blood vessel growth in developing organs. Here, we have used single-cell analyses to show that KIT is expressed in endothelial cell subsets of several organs, both in the adult and in the developing embryo. Knockout mouse analyses revealed that KIT is dispensable for vascularisation of growing organs in the midgestation embryo, including the lung, liver and brain. By contrast, vascular changes emerged during late-stage embryogenesis in these organs from KIT-deficient embryos, concurrent with severe erythrocyte deficiency and growth retardation. These findings suggest that KIT is not required for developmental tissue vascularisation in physiological conditions, but that KIT deficiency causes foetal anaemia at late gestation and thereby pathological vascular remodelling.


Assuntos
Células Endoteliais , Neovascularização Fisiológica , Animais , Embrião de Mamíferos , Feminino , Camundongos , Camundongos Knockout , Neovascularização Patológica , Neovascularização Fisiológica/genética , Gravidez , Saco Vitelino/irrigação sanguínea
3.
Development ; 146(21)2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31690636

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons regulate puberty onset and sexual reproduction by secreting GnRH to activate and maintain the hypothalamic-pituitary-gonadal axis. During embryonic development, GnRH neurons migrate along olfactory and vomeronasal axons through the nose into the brain, where they project to the median eminence to release GnRH. The secreted glycoprotein SEMA3A binds its receptors neuropilin (NRP) 1 or NRP2 to position these axons for correct GnRH neuron migration, with an additional role for the NRP co-receptor PLXNA1. Accordingly, mutations in SEMA3A, NRP1, NRP2 and PLXNA1 have been linked to defective GnRH neuron development in mice and inherited GnRH deficiency in humans. Here, we show that only the combined loss of PLXNA1 and PLXNA3 phenocopied the full spectrum of nasal axon and GnRH neuron defects of SEMA3A knockout mice. Together with Plxna1, the human orthologue of Plxna3 should therefore be investigated as a candidate gene for inherited GnRH deficiency.


Assuntos
Axônios/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Hormônio Liberador de Gonadotropina/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Padronização Corporal , Encéfalo/fisiologia , Movimento Celular , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Proteínas do Tecido Nervoso/genética , Neuropilina-1/fisiologia , Neuropilina-2/fisiologia , Nariz , Fenótipo , Receptores de Superfície Celular/genética , Semaforina-3A/fisiologia , Maturidade Sexual/genética , Transdução de Sinais
4.
Microsc Microanal ; : 1-10, 2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35698867

RESUMO

Three-dimensional fluorescence microscopy is a key technology for inspecting biological samples, ranging from single cells to entire organisms. We recently proposed a novel approach called spatially modulated Selective Volume Illumination Microscopy (smSVIM) to suppress illumination artifacts and to reduce the required number of measurements using an LED source. Here, we discuss a new strategy based on smSVIM for imaging large transparent specimens or voluminous chemically cleared tissues. The strategy permits steady mounting of the sample, achieving uniform resolution over a large field of view thanks to the synchronized motion of the illumination lens and the camera rolling shutter. Aided by a tailored deconvolution method for image reconstruction, we demonstrate significant improvement of the resolution at different magnification using samples of varying sizes and spatial features.

5.
Development ; 143(11): 1907-13, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27048738

RESUMO

The correct migration and axon extension of neurons in the developing nervous system is essential for the appropriate wiring and function of neural networks. Here, we report that O-sulfotransferases, a class of enzymes that modify heparan sulfate proteoglycans (HSPGs), are essential to regulate neuronal migration and axon development. We show that the 6-O-sulfotransferases HS6ST1 and HS6ST2 are essential for cranial axon patterning, whilst the 2-O-sulfotransferase HS2ST (also known as HS2ST1) is important to regulate the migration of facial branchiomotor (FBM) neurons in the hindbrain. We have also investigated how HS2ST interacts with other signals in the hindbrain and show that fibroblast growth factor (FGF) signalling regulates FBM neuron migration in an HS2ST-dependent manner.


Assuntos
Orientação de Axônios , Movimento Celular/efeitos dos fármacos , Neurônios Motores/citologia , Proteoglicanas/metabolismo , Crânio/metabolismo , Sulfatos/metabolismo , Animais , Orientação de Axônios/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Camundongos Endogâmicos C57BL , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Crânio/efeitos dos fármacos , Sulfotransferases/metabolismo , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
6.
Development ; 142(2): 314-9, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25519242

RESUMO

The vascular endothelial growth factor (VEGFA, VEGF) regulates neurovascular patterning. Alternative splicing of the Vegfa gene gives rise to three major isoforms termed VEGF121, VEGF165 and VEGF189. VEGF165 binds the transmembrane protein neuropilin 1 (NRP1) and promotes the migration, survival and axon guidance of subsets of neurons, whereas VEGF121 cannot activate NRP1-dependent neuronal responses. By contrast, the role of VEGF189 in NRP1-mediated signalling pathways has not yet been examined. Here, we have combined expression studies and in situ ligand-binding assays with the analysis of genetically altered mice and in vitro models to demonstrate that VEGF189 can bind NRP1 and promote NRP1-dependent neuronal responses.


Assuntos
Encéfalo/embriologia , Modelos Neurológicos , Neurônios/fisiologia , Neuropilina-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Encéfalo/citologia , Hibridização In Situ , Camundongos , Oligonucleotídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
7.
Development ; 141(3): 556-62, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24401374

RESUMO

Neuropilin 1 (NRP1) is a receptor for class 3 semaphorins and vascular endothelial growth factor (VEGF) A and is essential for cardiovascular development. Biochemical evidence supports a model for NRP1 function in which VEGF binding induces complex formation between NRP1 and VEGFR2 to enhance endothelial VEGF signalling. However, the relevance of VEGF binding to NRP1 for angiogenesis in vivo has not yet been examined. We therefore generated knock-in mice expressing Nrp1 with a mutation of tyrosine (Y) 297 in the VEGF binding pocket of the NRP1 b1 domain, as this residue was previously shown to be important for high affinity VEGF binding and NRP1-VEGFR2 complex formation. Unexpectedly, this targeting strategy also severely reduced NRP1 expression and therefore generated a NRP1 hypomorph. Despite the loss of VEGF binding and attenuated NRP1 expression, homozygous Nrp1(Y297A/Y297A) mice were born at normal Mendelian ratios, arguing against NRP1 functioning exclusively as a VEGF164 receptor in embryonic angiogenesis. By overcoming the mid-gestation lethality of full Nrp1-null mice, homozygous Nrp1(Y297A/Y297A) mice revealed essential roles for NRP1 in postnatal angiogenesis and arteriogenesis in the heart and retina, pathological neovascularisation of the retina and angiogenesis-dependent tumour growth.


Assuntos
Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Neuropilina-1/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Sequência de Bases , Peso Corporal/genética , Carcinogênese/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dados de Sequência Molecular , Miocárdio/metabolismo , Miocárdio/patologia , Neovascularização Patológica/embriologia , Neovascularização Patológica/genética , Neovascularização Fisiológica/genética , Neuropilina-1/metabolismo , Oxigênio , Ligação Proteica , Artéria Retiniana/patologia , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Rombencéfalo/patologia , Análise de Sobrevida
8.
Arterioscler Thromb Vasc Biol ; 36(1): 19-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26603154

RESUMO

OBJECTIVE: Ocular neovascularization (ONV) is a pathological feature of sight-threatening human diseases, such as diabetic retinopathy and age-related macular degeneration. Macrophage depletion in mouse models of ONV reduces the formation of pathological blood vessels, and myeloid cells are widely considered an important source of the vascular endothelial growth factor A (VEGF). However, the importance of VEGF or its upstream regulators hypoxia-inducible factor-1α (HIF1α) and hypoxia-inducible factor-2α (HIF2α) as myeloid-derived regulators of ONV remains to be determined. APPROACH AND RESULTS: We used 2 mouse models of ONV, choroidal neovascularization and oxygen-induced retinopathy, to show that Vegfa is highly expressed by several cell types, but not myeloid cells during ONV. Moreover, myeloid-specific VEGF ablation did not reduce total ocular VEGF during choroidal neovascularization or oxygen-induced retinopathy. In agreement, the conditional inactivation of Vegfa, Hif1a, or Epas1 in recruited and resident myeloid cells that accumulated at sites of neovascularization did not significantly reduce choroidal neovascularization or oxygen-induced retinopathy. CONCLUSIONS: The finding that myeloid cells are not a significant local source of VEGF in these rodent models of ONV suggests that myeloid function in neovascular eye disease differs from skin wound healing and other neovascular pathologies.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neovascularização de Coroide/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Mieloides/metabolismo , Neovascularização Retiniana/metabolismo , Vasos Retinianos/metabolismo , Retinopatia da Prematuridade/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Neovascularização de Coroide/genética , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos Knockout , Células Mieloides/patologia , Oxigênio , Neovascularização Retiniana/induzido quimicamente , Neovascularização Retiniana/genética , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Retinopatia da Prematuridade/induzido quimicamente , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/deficiência , Fator A de Crescimento do Endotélio Vascular/genética
9.
Blood ; 121(12): 2352-62, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23315162

RESUMO

Neuropilin (NRP) 1 is a receptor for the vascular endothelial growth factor (VEGF)-A and is essential for normal angiogenesis. Previous in vitro experiments identified NRP1 interactions with VEGF-A's main signaling receptor VEGFR2 within endothelial cells, but also between nonendothelial NRP1 and endothelial VEGFR2. Consistent with an endothelial role for NRP1 in angiogenesis, we found that VEGFR2 and NRP1 were coexpressed in endothelial tip and stalk cells in the developing brain. In addition, NRP1 was expressed on two cell types that interact with growing brain vessels-the neural progenitors that secrete VEGF-A to stimulate tip cell activity and the pro-angiogenic macrophages that promote tip cell anastomosis. Selective targeting of Nrp1 in each of these cell types demonstrated that neural progenitor- and macrophage-derived NRP1 were dispensable, whereas endothelial NRP1 was essential for normal brain vessel growth. NRP1 therefore promotes brain angiogenesis cell autonomously in endothelium, independently of heterotypic interactions with nonendothelial cells. Genetic mosaic analyses demonstrated a key role for NRP1 in endothelial tip rather than stalk cells during vessel sprouting. Thus, NRP1-expressing endothelial cells attained the tip cell position when competing with NRP1-negative endothelial cells in chimeric vessel sprouts. Taken together, these findings demonstrate that NRP1 promotes endothelial tip cell function during angiogenesis.


Assuntos
Vasos Sanguíneos/embriologia , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Neovascularização Fisiológica/genética , Neuropilina-1/fisiologia , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Polaridade Celular/genética , Embrião de Mamíferos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Feminino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Morfogênese/genética , Neuropilina-1/genética , Neuropilina-1/metabolismo , Especificidade de Órgãos/genética , Gravidez
10.
Development ; 138(19): 4185-91, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21852397

RESUMO

Neuropilin 1 (NRP1) is a transmembrane glycoprotein that is essential for blood vessel development in vertebrates. Best known for its ability to bind members of the vascular endothelial growth factor (VEGF) and class 3 semaphorin families through its extracellular domain, it also has a highly conserved cytoplasmic domain, which terminates in a SEA motif that binds the PDZ protein synectin/GIPC1/NIP. Previous studies in zebrafish embryos and tissue culture models raised the possibility that the SEA motif of NRP1 is essential for angiogenesis. Here, we describe the generation of mice that express a form of NRP1 that lacks the cytoplasmic domain and, therefore, the SEA motif (Nrp1(cyto)(Δ)(/)(Δ) mice). Our analysis of pre- and perinatal vascular development revealed that vasculogenesis and angiogenesis proceed normally in these mutants, demonstrating that the membrane-anchored extracellular domain is sufficient for vessel growth. By contrast, the NRP1 cytoplasmic domain is required for normal arteriovenous patterning, because arteries and veins crossed each other at an abnormally high frequency in the Nrp1(cyto)(Δ)(/)(Δ) retina, as previously reported for mice with haploinsufficient expression of VEGF in neural progenitors. At crossing sites, the artery was positioned anteriorly to the vein, and both vessels were embedded in a shared collagen sleeve. In human eyes, similar arteriovenous crossings are risk factors for branch retinal vein occlusion (BRVO), an eye disease in which compression of the vein by the artery disrupts retinal blood flow, causing local tissue hypoxia and impairing vision. Nrp1(cyto)(Δ)(/)(Δ) mice may therefore provide a suitable genetic model to study the aetiology of BRVO.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neovascularização Patológica , Neuropilina-1/metabolismo , Artéria Retiniana/patologia , Veia Retiniana/patologia , Motivos de Aminoácidos , Animais , Sequência de Bases , Colágeno/metabolismo , Citoplasma/metabolismo , Humanos , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Artéria Retiniana/embriologia , Veia Retiniana/embriologia , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Microcirculation ; 21(4): 315-23, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24521511

RESUMO

The formation of the cardiovasculature, consisting of both the heart and blood vessels, is a critical step in embryonic development and relies on three processes termed vasculogenesis, angiogenesis, and vascular remodeling. The transmembrane protein NRP1 is an essential modulator of embryonic angiogenesis with additional roles in vessel remodeling and arteriogenesis. NRP1 also enhances arteriogenesis in adults to alleviate pathological tissue ischemia. However, in certain circumstances, vascular NRP1 signaling can be detrimental, as it may promote cancer by enhancing tumor angiogenesis or contribute to tissue edema by increasing vascular permeability. Understanding the mechanisms of NRP1 signaling is, therefore, of profound importance for the design of therapies aiming to control vascular functions. Previous work has shown that vascular NRP1 can variably serve as a receptor for two secreted glycoproteins, the VEGF-A and SEMA3A, but it also has a poorly understood role as an adhesion receptor. Here, we review current knowledge of NRP1 function during blood vessel growth and homeostasis, with special emphasis on the vascular roles of its multiple ligands and signaling partners.


Assuntos
Permeabilidade Capilar , Proteínas de Neoplasias/metabolismo , Neoplasias/irrigação sanguínea , Neovascularização Patológica/metabolismo , Neuropilina-1/metabolismo , Remodelação Vascular , Adulto , Animais , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/patologia , Semaforina-3A/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Nat Cardiovasc Res ; 3: 460-473, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38708406

RESUMO

Transcriptomic data can be mined to understand the molecular activity of cell types. Yet, functional genes may remain undetected in RNA sequencing (RNA-seq) experiments for technical reasons, such as insufficient read depth or gene dropout. Conversely, RNA-seq experiments may detect lowly expressed mRNAs thought to be biologically irrelevant products of leaky transcription. To represent a cell type's functional transcriptome more accurately, we propose compiling many bulk RNA-seq datasets into a compendium and applying established classification models to predict whether detected transcripts are likely products of active or leaky transcription. Here, we present the BulkECexplorer (bulk RNA-seq endothelial cell explorer) compendium of 240 bulk RNA-seq datasets from five vascular endothelial cell subtypes. This resource reports transcript counts for genes of interest and predicts whether detected transcripts are likely the products of active or leaky gene expression. Beyond its usefulness for vascular biology research, this resource provides a blueprint for developing analogous tools for other cell types.

13.
J Dev Biol ; 11(2)2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-37092477

RESUMO

During prenatal life, the foetal liver is colonised by several waves of haematopoietic progenitors to act as the main haematopoietic organ. Single cell (sc) RNA-seq has been used to identify foetal liver cell types via their transcriptomic signature and to compare gene expression patterns as haematopoietic development proceeds. To obtain a refined single cell landscape of haematopoiesis in the foetal liver, we have generated a scRNA-seq dataset from a whole mouse E12.5 liver that includes a larger number of cells than prior datasets at this stage and was obtained without cell type preselection to include all liver cell populations. We combined mining of this dataset with that of previously published datasets at other developmental stages to follow transcriptional dynamics as well as the cell cycle state of developing haematopoietic lineages. Our findings corroborate several prior reports on the timing of liver colonisation by haematopoietic progenitors and the emergence of differentiated lineages and provide further molecular characterisation of each cell population. Extending these findings, we demonstrate the existence of a foetal intermediate haemoglobin profile in the mouse, similar to that previously identified in humans, and a previously unidentified population of primitive erythroid cells in the foetal liver.

14.
Nat Commun ; 14(1): 8097, 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-38062045

RESUMO

Innervation of the hypothalamic median eminence by Gonadotropin-Releasing Hormone (GnRH) neurons is vital to ensure puberty onset and successful reproduction. However, the molecular and cellular mechanisms underlying median eminence development and pubertal timing are incompletely understood. Here we show that Semaphorin-6A is strongly expressed by median eminence-resident oligodendrocytes positioned adjacent to GnRH neuron projections and fenestrated capillaries, and that Semaphorin-6A is required for GnRH neuron innervation and puberty onset. In vitro and in vivo experiments reveal an unexpected function for Semaphorin-6A, via its receptor Plexin-A2, in the control of median eminence vascular permeability to maintain neuroendocrine homeostasis. To support the significance of these findings in humans, we identify patients with delayed puberty carrying a novel pathogenic variant of SEMA6A. In all, our data reveal a role for Semaphorin-6A in regulating GnRH neuron patterning by tuning the median eminence vascular barrier and thereby controlling puberty onset.


Assuntos
Hormônio Liberador de Gonadotropina , Semaforinas , Humanos , Hormônio Liberador de Gonadotropina/metabolismo , Eminência Mediana/metabolismo , Permeabilidade Capilar , Neurônios/metabolismo , Puberdade , Semaforinas/genética , Semaforinas/metabolismo
15.
Sci Adv ; 9(32): eadg9781, 2023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37566656

RESUMO

Vascularization is driven by morphogen signals and mechanical cues that coordinately regulate cellular force generation, migration, and shape change to sculpt the developing vascular network. However, it remains unclear whether developing vasculature actively regulates its own mechanical properties to achieve effective vascularization. We engineered tissue constructs containing endothelial cells and fibroblasts to investigate the mechanics of vascularization. Tissue stiffness increases during vascular morphogenesis resulting from emergent interactions between endothelial cells, fibroblasts, and ECM and correlates with enhanced vascular function. Contractile cellular forces are key to emergent tissue stiffening and synergize with ECM mechanical properties to modulate the mechanics of vascularization. Emergent tissue stiffening and vascular function rely on mechanotransduction signaling within fibroblasts, mediated by YAP1. Mouse embryos lacking YAP1 in fibroblasts exhibit both reduced tissue stiffness and develop lethal vascular defects. Translating our findings through biology-inspired vascular tissue engineering approaches will have substantial implications in regenerative medicine.


Assuntos
Células Endoteliais , Mecanotransdução Celular , Camundongos , Animais , Mecanotransdução Celular/fisiologia , Engenharia Tecidual/métodos , Morfogênese , Diferenciação Celular , Matriz Extracelular
16.
Blood ; 116(5): 829-40, 2010 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-20404134

RESUMO

Blood vessel networks expand in a 2-step process that begins with vessel sprouting and is followed by vessel anastomosis. Vessel sprouting is induced by chemotactic gradients of the vascular endothelial growth factor (VEGF), which stimulates tip cell protrusion. Yet it is not known which factors promote the fusion of neighboring tip cells to add new circuits to the existing vessel network. By combining the analysis of mouse mutants defective in macrophage development or VEGF signaling with live imaging in zebrafish, we now show that macrophages promote tip cell fusion downstream of VEGF-mediated tip cell induction. Macrophages therefore play a hitherto unidentified and unexpected role as vascular fusion cells. Moreover, we show that there are striking molecular similarities between the pro-angiogenic tissue macrophages essential for vascular development and those that promote the angiogenic switch in cancer, including the expression of the cell-surface proteins TIE2 and NRP1. Our findings suggest that tissue macrophages are a target for antiangiogenic therapies, but that they could equally well be exploited to stimulate tissue vascularization in ischemic disease.


Assuntos
Macrófagos/fisiologia , Neovascularização Fisiológica/efeitos dos fármacos , Rombencéfalo/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Polaridade Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Células Endoteliais/ultraestrutura , Endotélio Vascular/crescimento & desenvolvimento , Feminino , Técnicas de Introdução de Genes , Fator Estimulador de Colônias de Macrófagos/deficiência , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/fisiologia , Neuropilina-1/fisiologia , Proteínas Proto-Oncogênicas/deficiência , Receptores Proteína Tirosina Quinases/fisiologia , Receptor TIE-2 , Vasos Retinianos/crescimento & desenvolvimento , Rombencéfalo/embriologia , Transativadores/deficiência , Fator A de Crescimento do Endotélio Vascular/deficiência , Fator A de Crescimento do Endotélio Vascular/genética , Saco Vitelino/citologia , Peixe-Zebra/embriologia
17.
Methods Mol Biol ; 2441: 63-73, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35099728

RESUMO

Angiogenesis refers to the expansion of blood vessels from a preexisting vascular plexus, and it is a fundamental process for organ development, the female reproductive system, and wound healing, but it is also a common denominator in several diseases such as cancer and neovascular eye disease. For these reasons, shedding light on the molecular and cellular mechanisms of angiogenesis has the potential to devise new therapeutic strategies to refrain pathological vessel growth or even promote new vessel formation in ischemic conditions and organ grafts. The mouse postnatal retina provides an excellent and widely adopted model to study physiological angiogenesis in vivo, and this chapter outlines a detailed protocol for its dissection, staining, and analysis of the vasculature.


Assuntos
Neoplasias , Neovascularização Patológica , Animais , Feminino , Camundongos , Neoplasias/patologia , Neovascularização Patológica/patologia , Neovascularização Fisiológica/fisiologia , Retina/patologia , Cicatrização/fisiologia
18.
Methods Mol Biol ; 2475: 275-287, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35451765

RESUMO

Angiogenesis, the growth of new blood vessels from pre-existing ones, is a fundamental process for organ development, exercise-induced muscle growth, and wound healing, but is also associated with different diseases such as cancer and neovascular eye disease. Accordingly, elucidating the molecular and cellular mechanisms of angiogenesis has the potential to identify new therapeutic targets to stimulate new vessel formation in ischemic tissues or inhibit pathological vessel growth in disease. This chapter describes the mouse embryo hindbrain and postnatal retina as models to study physiological angiogenesis and provides detailed protocols for tissue dissection, sample staining and analysis.


Assuntos
Neovascularização Patológica , Neovascularização Fisiológica , Animais , Camundongos , Neovascularização Fisiológica/fisiologia , Retina , Rombencéfalo/irrigação sanguínea , Coloração e Rotulagem
19.
Nat Cardiovasc Res ; 1: 872-873, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36605232

RESUMO

Late fetal liver hematopoiesis was thought to primarily rely on hematopoietic stem cells (HSCs). Using new genetic-tracing tools, a study shows that EVI1-positive HSCs mainly undergo expansion in the fetal liver, while differentiated blood cell production depends on HSC-independent intermediate hematopoietic progenitors.

20.
Methods Mol Biol ; 2475: 289-295, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35451766

RESUMO

Before the endothelial mitogenic activity of the Vascular Endothelial Growth Factor A (VEGF) was described, VEGF had already been identified for its ability to induce vascular leakage. VEGF-induced vascular leakage has been most frequently studied in vivo using the Miles assay, a simple yet invaluable technique that has allowed researchers to unravel the molecular mechanisms underpinning vascular leakage both for VEGF and other permeability inducing agents. In this protocol, a mouse is intravenously injected with Evans Blue dye before VEGF is administered locally via intradermal injection. VEGF promotes vascular leak of serum proteins in the dermis, enabling Evans Blue-labeled albumin extravasation from the circulation and subsequent accumulation in the skin. As the volume of dye extravasation is proportional to the degree of vascular leak, it can be quantified as a proxy measurement of VEGF-induced vascular leakage.


Assuntos
Permeabilidade Capilar , Fator A de Crescimento do Endotélio Vascular , Animais , Azul Evans/metabolismo , Camundongos , Pele/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA