Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 162(5): 1127-39, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26279190

RESUMO

The peripheral nervous system has remarkable regenerative capacities in that it can repair a fully cut nerve. This requires Schwann cells to migrate collectively to guide regrowing axons across a 'bridge' of new tissue, which forms to reconnect a severed nerve. Here we show that blood vessels direct the migrating cords of Schwann cells. This multicellular process is initiated by hypoxia, selectively sensed by macrophages within the bridge, which via VEGF-A secretion induce a polarized vasculature that relieves the hypoxia. Schwann cells then use the blood vessels as "tracks" to cross the bridge taking regrowing axons with them. Importantly, disrupting the organization of the newly formed blood vessels in vivo, either by inhibiting the angiogenic signal or by re-orienting them, compromises Schwann cell directionality resulting in defective nerve repair. This study provides important insights into how the choreography of multiple cell-types is required for the regeneration of an adult tissue.


Assuntos
Vasos Sanguíneos/metabolismo , Macrófagos/metabolismo , Nervos Periféricos/fisiologia , Células de Schwann/metabolismo , Animais , Axônios/metabolismo , Hipóxia Celular , Células Endoteliais/metabolismo , Inflamação/metabolismo , Masculino , Camundongos , Neovascularização Fisiológica , Ratos , Ratos Sprague-Dawley , Regeneração , Fator A de Crescimento do Endotélio Vascular/genética
2.
Development ; 151(2)2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38095299

RESUMO

Binocular vision requires the segregation of retinal ganglion cell (RGC) axons extending from the retina into the ipsilateral and contralateral optic tracts. RGC axon segregation occurs at the optic chiasm, which forms at the ventral diencephalon midline. Using expression analyses, retinal explants and genetically modified mice, we demonstrate that CXCL12 (SDF1) is required for axon segregation at the optic chiasm. CXCL12 is expressed by the meninges bordering the optic pathway, and CXCR4 by both ipsilaterally and contralaterally projecting RGCs. CXCL12 or ventral diencephalon meninges potently promoted axon outgrowth from both ipsilaterally and contralaterally projecting RGCs. Further, a higher proportion of axons projected ipsilaterally in mice lacking CXCL12 or its receptor CXCR4 compared with wild-type mice as a result of misrouting of presumptive contralaterally specified RGC axons. Although RGCs also expressed the alternative CXCL12 receptor ACKR3, the optic chiasm developed normally in mice lacking ACKR3. Our data support a model whereby meningeal-derived CXCL12 helps drive axon growth from CXCR4-expressing RGCs towards the diencephalon midline, enabling contralateral axon growth. These findings further our understanding of the molecular and cellular mechanisms controlling optic pathway development.


Assuntos
Quiasma Óptico , Células Ganglionares da Retina , Animais , Camundongos , Axônios/metabolismo , Diencéfalo , Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Vias Visuais
3.
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
4.
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
5.
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
6.
Angiogenesis ; 24(2): 199-211, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33783643

RESUMO

Hematopoiesis in vertebrate embryos occurs in temporally and spatially overlapping waves in close proximity to blood vascular endothelial cells. Initially, yolk sac hematopoiesis produces primitive erythrocytes, megakaryocytes, and macrophages. Thereafter, sequential waves of definitive hematopoiesis arise from yolk sac and intraembryonic hemogenic endothelia through an endothelial-to-hematopoietic transition (EHT). During EHT, the endothelial and hematopoietic transcriptional programs are tightly co-regulated to orchestrate a shift in cell identity. In the yolk sac, EHT generates erythro-myeloid progenitors, which upon migration to the liver differentiate into fetal blood cells, including erythrocytes and tissue-resident macrophages. In the dorsal aorta, EHT produces hematopoietic stem cells, which engraft the fetal liver and then the bone marrow to sustain adult hematopoiesis. Recent studies have defined the relationship between the developing vascular and hematopoietic systems in animal models, including molecular mechanisms that drive the hemato-endothelial transcription program for EHT. Moreover, human pluripotent stem cells have enabled modeling of fetal human hematopoiesis and have begun to generate cell types of clinical interest for regenerative medicine.


Assuntos
Diferenciação Celular/efeitos da radiação , Linhagem da Célula/fisiologia , Células Endoteliais/metabolismo , Endotélio/embriologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células Endoteliais/citologia , Endotélio/citologia , Células-Tronco Hematopoéticas/citologia , Humanos
7.
Angiogenesis ; 24(2): 271-288, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33825109

RESUMO

Lymphatic vessels have critical roles in both health and disease and their study is a rapidly evolving area of vascular biology. The consensus on how the first lymphatic vessels arise in the developing embryo has recently shifted. Originally, they were thought to solely derive by sprouting from veins. Since then, several studies have uncovered novel cellular mechanisms and a diversity of contributing cell lineages in the formation of organ lymphatic vasculature. Here, we review the key mechanisms and cell lineages contributing to lymphatic development, discuss the advantages and limitations of experimental techniques used for their study and highlight remaining knowledge gaps that require urgent attention. Emerging technologies should accelerate our understanding of how lymphatic vessels develop normally and how they contribute to disease.


Assuntos
Linhagem da Célula , Células Endoteliais/metabolismo , Linfangiogênese , Vasos Linfáticos/embriologia , Animais , Humanos
8.
Development ; 145(21)2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30237243

RESUMO

The adrenal medulla is composed of neuroendocrine chromaffin cells that secrete adrenaline into the systemic circulation to maintain physiological homeostasis and enable the autonomic stress response. How chromaffin cell precursors colonise the adrenal medulla and how they become connected to central nervous system-derived preganglionic sympathetic neurons remain largely unknown. By combining lineage tracing, gene expression studies, genetic ablation and the analysis of mouse mutants, we demonstrate that preganglionic axons direct chromaffin cell precursors into the adrenal primordia. We further show that preganglionic axons and chromaffin cell precursors require class 3 semaphorin (SEMA3) signalling through neuropilins (NRP) to target the adrenal medulla. Thus, SEMA3 proteins serve as guidance cues to control formation of the adrenal neuroendocrine system by establishing appropriate connections between preganglionic neurons and adrenal chromaffin cells that regulate the autonomic stress response.


Assuntos
Medula Suprarrenal/inervação , Axônios/metabolismo , Células Cromafins/metabolismo , Gânglios/metabolismo , Neuropilinas/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Movimento Celular , Masculino , Camundongos , Crista Neural/citologia , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo
9.
Development ; 144(13): 2504-2516, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28676569

RESUMO

Visual information is relayed from the eye to the brain via retinal ganglion cell (RGC) axons. Mice lacking NRP1 or NRP1-binding VEGF-A isoforms have defective RGC axon organisation alongside brain vascular defects. It is not known whether axonal defects are caused exclusively by defective VEGF-A signalling in RGCs or are exacerbated by abnormal vascular morphology. Targeted NRP1 ablation in RGCs with a Brn3bCre knock-in allele reduced axonal midline crossing at the optic chiasm and optic tract fasciculation. In contrast, Tie2-Cre-mediated endothelial NRP1 ablation induced axon exclusion zones in the optic tracts without impairing axon crossing. Similar defects were observed in Vegfa120/120 and Vegfa188/188 mice, which have vascular defects as a result of their expression of single VEGF-A isoforms. Ectopic midline vascularisation in endothelial Nrp1 and Vegfa188/188 mutants caused additional axonal exclusion zones within the chiasm. As in vitro and in vivo assays demonstrated that vessels do not repel axons, abnormally large or ectopically positioned vessels are likely to present physical obstacles to axon growth. We conclude that proper axonal wiring during brain development depends on the precise molecular control of neurovascular co-patterning.


Assuntos
Axônios/metabolismo , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Neuropilina-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Padronização Corporal , Diencéfalo/embriologia , Diencéfalo/metabolismo , Células Endoteliais/metabolismo , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/metabolismo , Camundongos Endogâmicos C57BL , Mutação/genética , Neovascularização Fisiológica , Quiasma Óptico/embriologia , Quiasma Óptico/metabolismo , Células Ganglionares da Retina/metabolismo , Fator de Transcrição Brn-3B/metabolismo , Vias Visuais/metabolismo
10.
Acta Neuropathol ; 139(2): 383-401, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31696318

RESUMO

The vertebrate CNS is surrounded by the meninges, a protective barrier comprised of the outer dura mater and the inner leptomeninges, which includes the arachnoid and pial layers. While the dura mater contains lymphatic vessels, no conventional lymphatics have been found within the brain or leptomeninges. However, non-lumenized cells called Brain/Mural Lymphatic Endothelial Cells or Fluorescent Granule Perithelial cells (muLECs/BLECs/FGPs) that share a developmental program and gene expression with peripheral lymphatic vessels have been described in the meninges of zebrafish. Here we identify a structurally and functionally similar cell type in the mammalian leptomeninges that we name Leptomeningeal Lymphatic Endothelial Cells (LLEC). As in zebrafish, LLECs express multiple lymphatic markers, containing very large, spherical inclusions, and develop independently from the meningeal macrophage lineage. Mouse LLECs also internalize macromolecules from the cerebrospinal fluid, including Amyloid-ß, the toxic driver of Alzheimer's disease progression. Finally, we identify morphologically similar cells co-expressing LLEC markers in human post-mortem leptomeninges. Given that LLECs share molecular, morphological, and functional characteristics with both lymphatics and macrophages, we propose they represent a novel, evolutionary conserved cell type with potential roles in homeostasis and immune organization of the meninges.


Assuntos
Encéfalo/patologia , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Sistema Linfático/patologia , Meninges/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides , Animais , Feminino , Humanos , Masculino , Camundongos , Peixe-Zebra
11.
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
12.
Cereb Cortex ; 28(7): 2577-2593, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29901792

RESUMO

Vascular endothelial growth factor (Vegfa) is essential for promoting the vascularization of the embryonic murine forebrain. In addition, it directly influences neural development, although its role in the forming forebrain is less well elucidated. It was recently suggested that Vegfa may influence the development of GABAergic interneurons, inhibitory cells with crucial signaling roles in cortical neuronal circuits. However, the mechanism by which it affects interneuron development remains unknown. Here we investigated the developmental processes by which Vegfa may influence cortical interneuron development by analyzing transgenic mice that ubiquitously express the Vegfa120 isoform to perturb its signaling gradient. We found that interneurons reach the dorsal cortex at mid phases of corticogenesis despite an aberrant vascular network. Instead, endothelial ablation of Vegfa alters cortical interneuron numbers, their intracortical distribution and spatial proximity to blood vessels. We show for the first time that vascular-secreted guidance factors promote early-migrating interneurons in the intact forebrain in vivo and identify a novel role for vascular-Vegfa in this process.


Assuntos
Vasos Sanguíneos/fisiologia , Movimento Celular/genética , Neurônios GABAérgicos/fisiologia , Prosencéfalo/citologia , Prosencéfalo/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores Etários , Animais , Vasos Sanguíneos/embriologia , Quimiotaxia , Simulação por Computador , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Neurológicos , Neuropilina-1/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Transdução de Sinais/genética , Células-Tronco/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética
13.
Proc Natl Acad Sci U S A ; 113(47): 13414-13419, 2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27821771

RESUMO

In the adult rodent brain, new neurons are born in two germinal regions that are associated with blood vessels, and blood vessels and vessel-derived factors are thought to regulate the activity of adult neural stem cells. Recently, it has been proposed that a vascular niche also regulates prenatal neurogenesis. Here we identify the mouse embryo hindbrain as a powerful model to study embryonic neurogenesis and define the relationship between neural progenitor cell (NPC) behavior and vessel growth. Using this model, we show that a subventricular vascular plexus (SVP) extends through a hindbrain germinal zone populated by NPCs whose peak mitotic activity follows a surge in SVP growth. Hindbrains genetically defective in SVP formation owing to constitutive NRP1 loss showed a premature decline in both NPC activity and hindbrain growth downstream of precocious cell cycle exit, premature neuronal differentiation, and abnormal mitosis patterns. Defective regulation of NPC activity was not observed in mice lacking NRP1 expression by NPCs, but instead in mice lacking NRP1 selectively in endothelial cells, yet was independent of vascular roles in hindbrain oxygenation. Therefore, germinal zone vascularization sustains NPC proliferation in the prenatal brain.


Assuntos
Vasos Sanguíneos/fisiologia , Neurogênese , Rombencéfalo/irrigação sanguínea , Rombencéfalo/embriologia , Animais , Proliferação de Células , Autorrenovação Celular , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitose , Neovascularização Fisiológica , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neuropilina-1/metabolismo , Oxigênio/metabolismo , Fatores de Tempo
14.
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
15.
Proc Natl Acad Sci U S A ; 112(19): 6086-91, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25922531

RESUMO

Jaw morphogenesis depends on the growth of Meckel's cartilage during embryogenesis. However, the cell types and signals that promote chondrocyte proliferation for Meckel's cartilage growth are poorly defined. Here we show that neural crest cells (NCCs) and their derivatives provide an essential source of the vascular endothelial growth factor (VEGF) to enhance jaw vascularization and stabilize the major mandibular artery. We further show in two independent mouse models that blood vessels promote Meckel's cartilage extension. Coculture experiments of arterial tissue with NCCs or chondrocytes demonstrated that NCC-derived VEGF promotes blood vessel growth and that blood vessels secrete factors to instruct chondrocyte proliferation. Computed tomography and X-ray scans of patients with hemifacial microsomia also showed that jaw hypoplasia correlates with mandibular artery dysgenesis. We conclude that cranial NCCs and their derivatives provide an essential source of VEGF to support blood vessel growth in the developing jaw, which in turn is essential for normal chondrocyte proliferation, and therefore jaw extension.


Assuntos
Síndrome de Goldenhar/fisiopatologia , Mandíbula/anormalidades , Mandíbula/embriologia , Crista Neural/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adolescente , Animais , Cartilagem/embriologia , Diferenciação Celular , Proliferação de Células , Condrócitos/metabolismo , Técnicas de Cocultura , Feminino , Síndrome de Goldenhar/diagnóstico por imagem , Humanos , Hibridização In Situ , Masculino , Mandíbula/irrigação sanguínea , Camundongos , Crista Neural/citologia , Tomografia Computadorizada por Raios X , Proteína Wnt1/genética
16.
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
18.
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
19.
Semin Cell Dev Biol ; 24(3): 172-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23319134

RESUMO

The neuropilins NRP1 and NRP2 are transmembrane proteins that regulate many different aspects of vascular and neural development. Even though they were originally identified as adhesion molecules, they are most commonly studied as co-receptors for secreted signalling molecules of the class 3 semaphorin (SEMA) and vascular endothelial growth factor (VEGF) families. During nervous system development, both classes of ligands control soma migration, axon patterning and synaptogenesis in the central nervous system, and they additionally help to guide the neural crest cell precursors of neurons and glia in the peripheral nervous system. Both classes of neuropilin ligands also control endothelial cell behaviour, with NRP1 acting as a VEGF-A isoform receptor in blood vascular endothelium and as a semaphorin receptor in lymphatic valve endothelium, and NRP2 promoting lymphatic vessel growth induced by VEGF-C. Here we provide an overview of neuropilin function in neurons and neural crest cells, discuss current knowledge of neuropilin signalling in the vasculature and conclude with a summary of neuropilin roles in cancer.


Assuntos
Neoplasias/metabolismo , Neurônios/metabolismo , Neuropilinas/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Humanos , Neoplasias/irrigação sanguínea , Semaforinas/metabolismo
20.
Dev Biol ; 385(2): 220-9, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24262984

RESUMO

The vasculature and nervous system share striking similarities in their networked, tree-like architecture and in the way they are super-imposed in mature organs. It has previously been suggested that the intestinal microvasculature network directs the migration of enteric neural crest cells (ENCC) along the gut to promote the formation of the enteric nervous system (ENS). To investigate the inter-relationship of migrating ENCC, ENS formation and gut vascular development we combined fate-mapping of ENCC with immunolabelling and intravascular dye injection to visualise nascent blood vessel networks. We found that the enteric and vascular networks initially had very distinct patterns of development. In the foregut, ENCC migrated through areas devoid of established vascular networks. In vessel-rich areas, such as the midgut and hindgut, the distribution of migrating ENCC did not support the idea that these cells followed a pre-established vascular network. Moreover, when gut vascular development was impaired, either genetically in Vegfa(120/120) or Tie2-Cre;Nrp1(fl/-) mice or using an in vitro Wnt1-Cre;Rosa26(Yfp/+) mouse model of ENS development, ENCC still colonised the entire length of the gut, including the terminal hindgut. These results demonstrate that blood vessel networks are not necessary to guide migrating ENCC during ENS development. Conversely, in miRet(51) mice, which lack ENS in the hindgut, the vascular network in this region appeared to be normal suggesting that in early development both networks form independently of each other.


Assuntos
Sistema Nervoso Entérico/fisiologia , Intestinos/citologia , Neovascularização Fisiológica , Crista Neural/citologia , Animais , Intestinos/irrigação sanguínea , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA