Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Rev Rep ; 20(4): 1135-1149, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38438768

RESUMEN

In the adult bone marrow (BM), endothelial cells (ECs) are an integral component of the hematopoietic stem cell (HSC)-supportive niche, which modulates HSC activity by producing secreted and membrane-bound paracrine signals. Within the BM, distinct vascular arteriole, transitional, and sinusoidal EC subtypes display unique paracrine expression profiles and create anatomically-discrete microenvironments. However, the relative contributions of vascular endothelial subtypes in supporting hematopoiesis is unclear. Moreover, constitutive expression and off-target activity of currently available endothelial-specific and endothelial-subtype-specific murine cre lines potentially confound data analysis and interpretation. To address this, we describe two tamoxifen-inducible cre-expressing lines, Vegfr3-creERT2 and Cx40-creERT2, that efficiently label sinusoidal/transitional and arteriole endothelium respectively in adult marrow, without off-target activity in hematopoietic or perivascular cells. Utilizing an established mouse model in which cre-dependent recombination constitutively-activates MAPK signaling within adult endothelium, we identify arteriole ECs as the driver of MAPK-mediated hematopoietic dysfunction. These results define complementary tamoxifen-inducible creERT2-expressing mouse lines that label functionally-discrete and non-overlapping sinusoidal/transitional and arteriole EC populations in the adult BM, providing a robust toolset to investigate the differential contributions of vascular subtypes in maintaining hematopoietic homeostasis.


Asunto(s)
Células Endoteliales , Integrasas , Tamoxifeno , Animales , Ratones , Células Endoteliales/metabolismo , Integrasas/metabolismo , Integrasas/genética , Tamoxifeno/farmacología , Médula Ósea/metabolismo , Ratones Transgénicos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Hematopoyesis
2.
bioRxiv ; 2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36711611

RESUMEN

Background: The blood brain barrier (BBB) preserves neuronal function in the central nervous system (CNS) by tightly controlling metabolite exchanges with the blood. In the eye, the retina is likewise protected by the blood-retina barrier (BRB) to maintain phototransduction. We showed that the secreted guidance cue Netrin-1 regulated BBB integrity, by binding to endothelial Unc5B and regulating canonical ß-catenin dependent expression of BBB gene expression. Objective: Here, we investigated if Netrin-1-binding to endothelial Unc5B also controlled BRB integrity, and if this process involved Norrin/ß-catenin signaling, which is the major known driver of BRB development and maintenance. Methods: We analyzed Tamoxifen-inducible loss- and gain- of-function alleles of Unc5B, Ntn1 and Ctnnb1 in conjunction with tracer injections and biochemical signaling studies. Results: Inducible endothelial Unc5B deletion, and inducible global Ntn1 deletion in postnatal mice reduced phosphorylation of the Norrin receptor LRP5, leading to reduced ß-catenin and LEF1 expression, conversion of retina endothelial cells from a barrier-competent Claudin-5+/PLVAP- state to a Claudin-5-/PLVAP+ leaky phenotype, and extravasation of injected low molecular weight tracers. Inducible Ctnnb1 gain of function rescued vascular leak in Unc5B mutants, and Ntn1 overexpression induced BRB tightening. Unc5B expression in pericytes contributed to BRB permeability, via regulation of endothelial Unc5B. Mechanistically, Netrin-1-Unc5B signaling promoted ß-catenin dependent BRB signaling by enhancing phosphorylation of the Norrin receptor LRP5 via the Discs large homologue 1 (Dlg1) intracellular scaffolding protein. Conclusions: The data identify Netrin1-Unc5B as novel regulators of BRB integrity, with implications for diseases associated with BRB disruption.

3.
Cell Mol Life Sci ; 79(9): 491, 2022 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-35987946

RESUMEN

Endothelial cells in veins differ in morphology, function and gene expression from those in arteries and lymphatics. Understanding how venous and arterial identities are induced during development is required to understand how arterio-venous malformations occur, and to improve the outcome of vein grafts in surgery by promoting arterialization of veins. To identify factors that promote venous endothelial cell fate in vivo, we isolated veins from quail embryos, at different developmental stages, that were grafted into the coelom of chick embryos. Endothelial cells migrated out from the grafted vein and their colonization of host veins and/or arteries was quantified. We show that venous fate is promoted by sympathetic vessel innervation at embryonic day 11. Removal of sympathetic innervation decreased vein colonization, while norepinephrine enhanced venous colonization. BMP treatment or inhibition of ERK enhanced venous fate, revealing environmental neurotransmitter and BMP signaling and intrinsic ERK inhibition as actors in venous fate acquisition. We also identify the BMP antagonist Noggin as a potent mediator of venous arterialization.


Asunto(s)
Células Endoteliales , Venas , Animales , Arterias , Diferenciación Celular/fisiología , Embrión de Pollo , Transducción de Señal , Venas/metabolismo
4.
Nat Commun ; 13(1): 1169, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35246514

RESUMEN

Blood-brain barrier (BBB) integrity is critical for proper function of the central nervous system (CNS). Here, we show that the endothelial Unc5B receptor controls BBB integrity by maintaining Wnt/ß-catenin signaling. Inducible endothelial-specific deletion of Unc5B in adult mice leads to BBB leak from brain capillaries that convert to a barrier-incompetent state with reduced Claudin-5 and increased PLVAP expression. Loss of Unc5B decreases BBB Wnt/ß-catenin signaling, and ß-catenin overexpression rescues Unc5B mutant BBB defects. Mechanistically, the Unc5B ligand Netrin-1 enhances Unc5B interaction with the Wnt co-receptor LRP6, induces its phosphorylation and activates Wnt/ß-catenin downstream signaling. Intravenous delivery of antibodies blocking Netrin-1 binding to Unc5B causes a transient BBB breakdown and disruption of Wnt signaling, followed by neurovascular barrier resealing. These data identify Netrin-1-Unc5B signaling as a ligand-receptor pathway that regulates BBB integrity, with implications for CNS diseases.


Asunto(s)
Barrera Hematoencefálica , Receptores de Netrina , Animales , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Ligandos , Ratones , Receptores de Netrina/genética , Receptores de Netrina/metabolismo , Netrina-1/genética , Netrina-1/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
5.
J Clin Invest ; 131(16)2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34181595

RESUMEN

SLIT2 is a secreted polypeptide that guides migration of cells expressing Roundabout 1 and 2 (ROBO1 and ROBO2) receptors. Herein, we investigated SLIT2/ROBO signaling effects in gliomas. In patients with glioblastoma (GBM), SLIT2 expression increased with malignant progression and correlated with poor survival and immunosuppression. Knockdown of SLIT2 in mouse glioma cells and patient-derived GBM xenografts reduced tumor growth and rendered tumors sensitive to immunotherapy. Tumor cell SLIT2 knockdown inhibited macrophage invasion and promoted a cytotoxic gene expression profile, which improved tumor vessel function and enhanced efficacy of chemotherapy and immunotherapy. Mechanistically, SLIT2 promoted microglia/macrophage chemotaxis and tumor-supportive polarization via ROBO1- and ROBO2-mediated PI3K-γ activation. Macrophage Robo1 and Robo2 deletion and systemic SLIT2 trap delivery mimicked SLIT2 knockdown effects on tumor growth and the tumor microenvironment (TME), revealing SLIT2 signaling through macrophage ROBOs as a potentially novel regulator of the GBM microenvironment and immunotherapeutic target for brain tumors.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Proteínas del Tejido Nervioso/inmunología , Receptores Inmunológicos/inmunología , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioblastoma/irrigación sanguínea , Glioblastoma/patología , Xenoinjertos , Humanos , Tolerancia Inmunológica , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Pronóstico , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Proteínas Roundabout
6.
Nat Commun ; 10(1): 2350, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31138815

RESUMEN

Endothelial cell migration, proliferation and survival are triggered by VEGF-A activation of VEGFR2. However, how these cell behaviors are regulated individually is still unknown. Here we identify Endophilin-A2 (ENDOA2), a BAR-domain protein that orchestrates CLATHRIN-independent internalization, as a critical mediator of endothelial cell migration and sprouting angiogenesis. We show that EndoA2 knockout mice exhibit postnatal angiogenesis defects and impaired front-rear polarization of sprouting tip cells. ENDOA2 deficiency reduces VEGFR2 internalization and inhibits downstream activation of the signaling effector PAK but not ERK, thereby affecting front-rear polarity and migration but not proliferation or survival. Mechanistically, VEGFR2 is directed towards ENDOA2-mediated endocytosis by the SLIT2-ROBO pathway via SLIT-ROBO-GAP1 bridging of ENDOA2 and ROBO1. Blocking ENDOA2-mediated endothelial cell migration attenuates pathological angiogenesis in oxygen-induced retinopathy models. This work identifies a specific endocytic pathway controlling a subset of VEGFR2 mediated responses that could be targeted to prevent excessive sprouting angiogenesis in pathological conditions.


Asunto(s)
Aciltransferasas/genética , Células Endoteliales/metabolismo , Neovascularización Fisiológica/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular/genética , Polaridad Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Endocitosis/genética , Células Endoteliales/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Vasos Retinianos/citología , Vasos Retinianos/crecimiento & desarrollo , Quinasas p21 Activadas/metabolismo , Proteínas Roundabout
7.
Circulation ; 138(21): 2379-2394, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-29976569

RESUMEN

BACKGROUND: Hereditary hemorrhagic telangiectasia (HHT) is an inherited vascular disorder that causes arteriovenous malformations (AVMs). Mutations in the genes encoding Endoglin ( ENG) and activin-receptor-like kinase 1 ( AVCRL1 encoding ALK1) cause HHT type 1 and 2, respectively. Mutations in the SMAD4 gene are present in families with juvenile polyposis-HHT syndrome that involves AVMs. SMAD4 is a downstream effector of transforming growth factor-ß (TGFß)/bone morphogenetic protein (BMP) family ligands that signal via activin-like kinase receptors (ALKs). Ligand-neutralizing antibodies or inducible, endothelial-specific Alk1 deletion induce AVMs in mouse models as a result of increased PI3K (phosphatidylinositol 3-kinase)/AKT (protein kinase B) signaling. Here we addressed if SMAD4 was required for BMP9-ALK1 effects on PI3K/AKT pathway activation. METHODS: The authors generated tamoxifen-inducible, postnatal, endothelial-specific Smad4 mutant mice ( Smad4iΔEC). RESULTS: We found that loss of endothelial Smad4 resulted in AVM formation and lethality. AVMs formed in regions with high blood flow in developing retinas and other tissues. Mechanistically, BMP9 signaling antagonized flow-induced AKT activation in an ALK1- and SMAD4-dependent manner. Smad4iΔEC endothelial cells in AVMs displayed increased PI3K/AKT signaling, and pharmacological PI3K inhibitors or endothelial Akt1 deletion both rescued AVM formation in Smad4iΔEC mice. BMP9-induced SMAD4 inhibited casein kinase 2 ( CK2) transcription, in turn limiting PTEN phosphorylation and AKT activation. Consequently, CK2 inhibition prevented AVM formation in Smad4iΔEC mice. CONCLUSIONS: Our study reveals SMAD4 as an essential effector of BMP9-10/ALK1 signaling that affects AVM pathogenesis via regulation of CK2 expression and PI3K/AKT1 activation.


Asunto(s)
Malformaciones Arteriovenosas/patología , Quinasa de la Caseína II/metabolismo , Proteína Smad4/genética , Receptores de Activinas Tipo I/antagonistas & inhibidores , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Animales , Quinasa de la Caseína II/antagonistas & inhibidores , Modelos Animales de Enfermedad , Factores de Diferenciación de Crecimiento/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Citoplasmático Pequeño/metabolismo , Flujo Sanguíneo Regional , Retina/fisiopatología , Transducción de Señal/efectos de los fármacos , Proteína Smad4/antagonistas & inhibidores , Proteína Smad4/metabolismo
8.
Nat Commun ; 7: 13517, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27882935

RESUMEN

Vascular permeability and neovascularization are implicated in many diseases including retinopathies and diabetic wound healing. Robo4 is an endothelial-specific transmembrane receptor that stabilizes the vasculature, as shown in Robo4-/- mice that develop hyperpermeability, but how Robo4 signals remained unclear. Here we show that Robo4 deletion enhances permeability and revascularization in oxygen-induced retinopathy (OIR) and accelerates cutaneous wound healing. To determine Robo4 signalling pathways, we generated transgenic mice expressing a truncated Robo4 lacking the cytoplasmic domain (Robo4ΔCD). Robo4ΔCD expression is sufficient to prevent permeability, and inhibits OIR revascularization and wound healing in Robo4-/- mice. Mechanistically, Robo4 does not affect Slit2 signalling, but Robo4 and Robo4ΔCD counteract Vegfr2-Y949 (Y951 in human VEGFR2) phosphorylation by signalling through the endothelial UNC5B receptor. We conclude that Robo4 inhibits angiogenesis and vessel permeability independently of its cytoplasmic domain, while activating VEGFR2-Y951 via ROBO4 inhibition might accelerate tissue revascularization in retinopathy of prematurity and in diabetic patients.


Asunto(s)
Permeabilidad Capilar/genética , Neovascularización Patológica/genética , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/genética , Enfermedades de la Retina/genética , Animales , Retinopatía Diabética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Noqueados , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Netrina/metabolismo , Terapia por Inhalación de Oxígeno/efectos adversos , Fosforilación , Receptores de Superficie Celular , Receptores Inmunológicos/metabolismo , Enfermedades de la Retina/etiología , Enfermedades de la Retina/metabolismo , Retinopatía de la Prematuridad , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/genética
9.
Circ Res ; 119(5): 607-20, 2016 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-27354211

RESUMEN

RATIONALE: Arterial endothelial cells are morphologically, functionally, and molecularly distinct from those found in veins and lymphatic vessels. How arterial fate is acquired during development and maintained in adult vessels is incompletely understood. OBJECTIVE: We set out to identify factors that promote arterial endothelial cell fate in vivo. METHODS AND RESULTS: We developed a functional assay, allowing us to monitor and manipulate arterial fate in vivo, using arteries isolated from quails that are grafted into the coelom of chick embryos. Endothelial cells migrate out from the grafted artery, and their colonization of host arteries and veins is quantified. Here we show that sympathetic innervation promotes arterial endothelial cell fate in vivo. Removal of sympathetic nerves decreases arterial fate and leads to colonization of veins, whereas exposure to sympathetic nerves or norepinephrine imposes arterial fate. Mechanistically, sympathetic nerves increase endothelial ERK (extracellular signal-regulated kinase) activity via adrenergic α1 and α2 receptors. CONCLUSIONS: These findings show that sympathetic innervation promotes arterial endothelial fate and may lead to novel approaches to improve arterialization in human disease.


Asunto(s)
Fibras Adrenérgicas/enzimología , Arterias/enzimología , Arterias/inervación , Endotelio Vascular/enzimología , Endotelio Vascular/inervación , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Animales , Arterias/crecimiento & desarrollo , Movimiento Celular/fisiología , Embrión de Pollo , Membrana Corioalantoides/enzimología , Membrana Corioalantoides/crecimiento & desarrollo , Membrana Corioalantoides/inervación , Coturnix , Endotelio Vascular/crecimiento & desarrollo , Activación Enzimática/fisiología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Técnicas de Cultivo de Órganos , Sistema Nervioso Periférico/enzimología , Sistema Nervioso Periférico/crecimiento & desarrollo , Trasplante de Tejidos/métodos , Arterias Umbilicales/enzimología , Arterias Umbilicales/crecimiento & desarrollo
10.
Circulation ; 133(4): 409-21, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26659946

RESUMEN

BACKGROUND: Sprouting angiogenesis is a key process driving blood vessel growth in ischemic tissues and an important drug target in a number of diseases, including wet macular degeneration and wound healing. Endothelial cells forming the sprout must develop front-rear polarity to allow sprout extension. The adaptor proteins Nck1 and 2 are known regulators of cytoskeletal dynamics and polarity, but their function in angiogenesis is poorly understood. Here, we show that the Nck adaptors are required for endothelial cell front-rear polarity and migration downstream of the angiogenic growth factors VEGF-A and Slit2. METHODS AND RESULTS: Mice carrying inducible, endothelial-specific Nck1/2 deletions fail to develop front-rear polarized vessel sprouts and exhibit severe angiogenesis defects in the postnatal retina and during embryonic development. Inactivation of NCK1 and 2 inhibits polarity by preventing Cdc42 and Pak2 activation by VEGF-A and Slit2. Mechanistically, NCK binding to ROBO1 is required for both Slit2- and VEGF-induced front-rear polarity. Selective inhibition of polarized endothelial cell migration by targeting Nck1/2 prevents hypersprouting induced by Notch or Bmp signaling inhibition, and pathological ocular neovascularization and wound healing, as well. CONCLUSIONS: These data reveal a novel signal integration mechanism involving NCK1/2, ROBO1/2, and VEGFR2 that controls endothelial cell front-rear polarity during sprouting angiogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Polaridad Celular/fisiología , Células Endoteliales/fisiología , Eliminación de Gen , Neovascularización Fisiológica/fisiología , Proteínas Oncogénicas/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Secuencia de Aminoácidos , Animales , Marcación de Gen/métodos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Oncogénicas/deficiencia
11.
Nat Med ; 21(5): 483-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25894826

RESUMEN

Ocular neovascular diseases are a leading cause of blindness. Vascular endothelial growth factor (VEGF) blockade improves vision, but not all individuals respond to anti-VEGF treatment, making additional means to prevent neovascularization necessary. Slit-family proteins (Slits) are ligands of Roundabout (Robo) receptors that repel developing axons in the nervous system. Robo1 expression is altered in ocular neovascular diseases, and previous in vitro studies have reported both pro- and anti-angiogenic effects of Slits. However, genetic evidence supporting a role for Slits in ocular neovascularization is lacking. Here we generated conditional knockout mice deficient in various Slit and Robo proteins and found that Slit2 potently and selectively promoted angiogenesis via Robo1 and Robo2 in mouse postnatal retina and in a model of ocular neovascular disease. Mechanistically, Slit2 acting through Robo1 and Robo2 promoted the migration of endothelial cells. These receptors are required for both Slit2- and VEGF-induced Rac1 activation and lamellipodia formation. Thus, Slit2 blockade could potentially be used therapeutically to inhibit angiogenesis in individuals with ocular neovascular disease.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Neovascularización Retiniana , Animales , Apoptosis , Movimiento Celular/genética , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica , ARN Mensajero/metabolismo , Retina/embriología , Retina/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Roundabout
12.
Cell Rep ; 10(7): 1158-72, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25704818

RESUMEN

Neural stem cells (NSCs) continuously produce new neurons within the adult mammalian hippocampus. NSCs are typically quiescent but activated to self-renew or differentiate into neural progenitor cells. The molecular mechanisms of NSC activation remain poorly understood. Here, we show that adult hippocampal NSCs express vascular endothelial growth factor receptor (VEGFR) 3 and its ligand VEGF-C, which activates quiescent NSCs to enter the cell cycle and generate progenitor cells. Hippocampal NSC activation and neurogenesis are impaired by conditional deletion of Vegfr3 in NSCs. Functionally, this is associated with compromised NSC activation in response to VEGF-C and physical activity. In NSCs derived from human embryonic stem cells (hESCs), VEGF-C/VEGFR3 mediates intracellular activation of AKT and ERK pathways that control cell fate and proliferation. These findings identify VEGF-C/VEGFR3 signaling as a specific regulator of NSC activation and neurogenesis in mammals.


Asunto(s)
Células-Madre Neurales/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transducción de Señal , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/farmacología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
13.
J Clin Invest ; 124(7): 3230-40, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24937433

RESUMEN

Autonomic sympathetic nerves innervate peripheral resistance arteries, thereby regulating vascular tone and controlling blood supply to organs. Despite the fundamental importance of blood flow control, how sympathetic arterial innervation develops remains largely unknown. Here, we identified the axon guidance cue netrin-1 as an essential factor required for development of arterial innervation in mice. Netrin-1 was produced by arterial smooth muscle cells (SMCs) at the onset of innervation, and arterial innervation required the interaction of netrin-1 with its receptor, deleted in colorectal cancer (DCC), on sympathetic growth cones. Function-blocking approaches, including cell type-specific deletion of the genes encoding Ntn1 in SMCs and Dcc in sympathetic neurons, led to severe and selective reduction of sympathetic innervation and to defective vasoconstriction in resistance arteries. These findings indicate that netrin-1 and DCC are critical for the control of arterial innervation and blood flow regulation in peripheral organs.


Asunto(s)
Arterias Mesentéricas/inervación , Factores de Crecimiento Nervioso/fisiología , Sistema Nervioso Simpático/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Animales Recién Nacidos , Receptor DCC , Femenino , Conos de Crecimiento/fisiología , Masculino , Arterias Mesentéricas/crecimiento & desarrollo , Arterias Mesentéricas/fisiología , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Modelos Neurológicos , Miocitos del Músculo Liso/fisiología , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Netrina-1 , Embarazo , Receptores de Superficie Celular/fisiología , Sistema Nervioso Simpático/crecimiento & desarrollo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Vasoconstricción/fisiología
14.
Arterioscler Thromb Vasc Biol ; 34(7): 1468-76, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24764455

RESUMEN

OBJECTIVE: The H2.0-like homeobox transcription factor (HLX) plays an essential role in visceral organogenesis in mice and has been shown to regulate angiogenic sprouting in vitro and in zebrafish embryos. We therefore examined the role of HLX in vascular development in mouse and avian embryos. APPROACH AND RESULTS: In situ hybridization showed that Hlx is expressed in a subset of sprouting blood vessels in postnatal mouse retinas and embryos. Hlx expression was conserved in quail embryos and upregulated in blood vessels at the onset of circulation. In vitro assays showed that Hlx is dynamically regulated by growth factors and shear stress alterations. Proangiogenic vascular endothelial growth factor induces Hlx expression in cultured endothelial cells, whereas signals that induce stalk cell identity lead to a reduction in Hlx expression. HLX was also downregulated in embryos in which flow was ablated, whereas injection of a starch solution, which increases blood viscosity and therefore shear stress, causes an upregulation in HLX. HLX knockdown in vitro resulted in a reduction in tip cell marker expression and in reduced angiogenic sprouting, but Hlx(-/-) embryos showed no defect in vascular sprouting at E8.5, E9.5, or E11.5 in vivo. Vascular remodeling of the capillary plexus was altered in Hlx(-/-) embryos, with a modestly enlarged venous plexus and reduction of the arterial plexus. CONCLUSIONS: Our findings indicate not only that Hlx regulates sprouting in vitro, but that its role in sprouting is nonessential in vivo. We find HLX is regulated by shear stress and a subtle defect in vascular remodeling is present in knockout embryos.


Asunto(s)
Vasos Sanguíneos/metabolismo , Proteínas de Homeodominio/metabolismo , Neovascularización Fisiológica , Factores de Transcripción/metabolismo , Saco Vitelino/irrigación sanguínea , Animales , Vasos Sanguíneos/embriología , Viscosidad Sanguínea , Células Cultivadas , Embrión de Mamíferos/irrigación sanguínea , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Proteínas de Homeodominio/genética , Humanos , Mecanotransducción Celular , Ratones , Ratones Noqueados , Codorniz , Interferencia de ARN , Flujo Sanguíneo Regional , Estrés Mecánico , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transfección
15.
Dev Cell ; 20(1): 33-46, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21238923

RESUMEN

Robo4 is an endothelial cell-specific member of the Roundabout axon guidance receptor family. To identify Robo4 binding partners, we performed a protein-protein interaction screen with the Robo4 extracellular domain. We find that Robo4 specifically binds to UNC5B, a vascular Netrin receptor, revealing unexpected interactions between two endothelial guidance receptors. We show that Robo4 maintains vessel integrity by activating UNC5B, which inhibits signaling downstream of vascular endothelial growth factor (VEGF). Function-blocking monoclonal antibodies against Robo4 and UNC5B increase angiogenesis and disrupt vessel integrity. Soluble Robo4 protein inhibits VEGF-induced vessel permeability and rescues barrier defects in Robo4(-/-) mice, but not in mice treated with anti-UNC5B. Thus, Robo4-UNC5B signaling maintains vascular integrity by counteracting VEGF signaling in endothelial cells, identifying a novel function of guidance receptor interactions in the vasculature.


Asunto(s)
Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/enzimología , Permeabilidad Capilar/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Ligandos , Ratones , Modelos Biológicos , Receptores de Netrina , Unión Proteica/efectos de los fármacos , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Transducción de Señal/efectos de los fármacos , Sus scrofa , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...