Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Elife ; 82019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31621585

RESUMEN

Elevated intraocular pressure (IOP) due to insufficient aqueous humor outflow through the trabecular meshwork and Schlemm's canal (SC) is the most important risk factor for glaucoma, a leading cause of blindness worldwide. We previously reported loss of function mutations in the receptor tyrosine kinase TEK or its ligand ANGPT1 cause primary congenital glaucoma in humans and mice due to failure of SC development. Here, we describe a novel approach to enhance canal formation in these animals by deleting a single allele of the gene encoding the phosphatase PTPRB during development. Compared to Tek haploinsufficient mice, which exhibit elevated IOP and loss of retinal ganglion cells, Tek+/-;Ptprb+/- mice have elevated TEK phosphorylation, which allows normal SC development and prevents ocular hypertension and RGC loss. These studies provide evidence that PTPRB is an important regulator of TEK signaling in the aqueous humor outflow pathway and identify a new therapeutic target for treatment of glaucoma.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Glaucoma/genética , Receptor TIE-2/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Células Ganglionares de la Retina/enzimología , Alelos , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Animales , Humor Acuoso/enzimología , Recuento de Células , Modelos Animales de Enfermedad , Eliminación de Gen , Glaucoma/enzimología , Glaucoma/patología , Heterocigoto , Humanos , Presión Intraocular/fisiología , Ratones , Ratones Noqueados , Fosforilación , Receptor TIE-2/deficiencia , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/deficiencia , Células Ganglionares de la Retina/patología , Factores de Riesgo , Transducción de Señal , Malla Trabecular/enzimología , Malla Trabecular/patología
2.
J Am Soc Nephrol ; 29(4): 1097-1107, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29237738

RESUMEN

Urinary concentrating ability is central to mammalian water balance and depends on a medullary osmotic gradient generated by a countercurrent multiplication mechanism. Medullary hyperosmolarity is protected from washout by countercurrent exchange and efficient removal of interstitial fluid resorbed from the loop of Henle and collecting ducts. In most tissues, lymphatic vessels drain excess interstitial fluid back to the venous circulation. However, the renal medulla is devoid of classic lymphatics. Studies have suggested that the fenestrated ascending vasa recta (AVRs) drain the interstitial fluid in this location, but this function has not been conclusively shown. We report that late gestational deletion of the angiopoietin receptor endothelial tyrosine kinase 2 (Tie2) or both angiopoietin-1 and angiopoietin-2 prevents AVR formation in mice. The absence of AVR associated with rapid accumulation of fluid and cysts in the medullary interstitium, loss of medullary vascular bundles, and decreased urine concentrating ability. In transgenic reporter mice with normal angiopoietin-Tie2 signaling, medullary AVR exhibited an unusual hybrid endothelial phenotype, expressing lymphatic markers (prospero homeobox protein 1 and vascular endothelial growth factor receptor 3) as well as blood endothelial markers (CD34, endomucin, platelet endothelial cell adhesion molecule 1, and plasmalemmal vesicle-associated protein). Taken together, our data redefine the AVRs as Tie2 signaling-dependent specialized hybrid vessels and provide genetic evidence of the critical role of AVR in the countercurrent exchange mechanism and the structural integrity of the renal medulla.


Asunto(s)
Angiopoyetina 1/fisiología , Angiopoyetina 2/fisiología , Líquido Extracelular/metabolismo , Capacidad de Concentración Renal/fisiología , Médula Renal/irrigación sanguínea , Receptor TIE-2/fisiología , Angiopoyetina 1/deficiencia , Angiopoyetina 1/genética , Angiopoyetina 2/deficiencia , Angiopoyetina 2/genética , Animales , Tipificación del Cuerpo , Linaje de la Célula , Endotelio Vascular , Genes Reporteros , Edad Gestacional , Proteínas de Homeodominio/análisis , Enfermedades Renales Quísticas/genética , Médula Renal/embriología , Médula Renal/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Miofibroblastos/patología , Ósmosis , Receptor TIE-2/deficiencia , Receptor TIE-2/genética , Circulación Renal , Transducción de Señal , Proteínas Supresoras de Tumor/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis
3.
Sci Rep ; 7: 42127, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28198371

RESUMEN

Human endothelial cells (ECs) are widely used to study mechanisms of angiogenesis, inflammation, and endothelial permeability. Targeted gene disruption induced by Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-CRISPR-Associated Protein 9 (Cas9) nuclease gene editing is potentially an important tool for definitively establishing the functional roles of individual genes in ECs. We showed that co-delivery of adenovirus encoding EGFP-tagged Cas9 and lentivirus encoding a single guide RNA (sgRNA) in primary human lung microvascular ECs (HLMVECs) disrupted the expression of the Tie2 gene and protein. Tie2 disruption increased basal endothelial permeability and prevented permeability recovery following injury induced by the inflammatory stimulus thrombin. Thus, gene deletion via viral co-delivery of CRISPR-Cas9 in primary human ECs provides a novel platform to investigate signaling mechanisms of normal and perturbed EC function without the need for clonal expansion.


Asunto(s)
Adenoviridae/genética , Sistemas CRISPR-Cas , Células Endoteliales/metabolismo , Edición Génica/métodos , Lentivirus/genética , Mutagénesis Sitio-Dirigida/métodos , Receptor TIE-2/genética , Adenoviridae/metabolismo , Permeabilidad de la Membrana Celular , Endonucleasas/genética , Endonucleasas/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lentivirus/metabolismo , Pulmón/citología , Pulmón/metabolismo , Mutación , Cultivo Primario de Células , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Receptor TIE-2/deficiencia , Transducción de Señal , Trombina/farmacología
4.
PLoS One ; 7(9): e43568, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23049737

RESUMEN

The angiopoietins (ANGPT) are ligands for the endothelial cell (EC) receptor tyrosine kinase, Tie2. Angpt-1 is a Tie2 agonist that promotes vascular maturation and stabilization, whereas Angpt-2 is a partial agonist/antagonist involved in the initiation of postnatal angiogenesis. Therefore, we hypothesized that overexpression of Angpt-2 would be more effective than Angpt-1 for enhancing the perfusion recovery in the ischemic hindlimb. Perfusion recovery was markedly impaired in Tie2-deficient animals at day 35 in a model of chronic hindlimb ischemia. Injections of Angpt-2 or VEGFA plasmid at 7 days post femoral artery resection enhanced recovery and improved arteriogenesis as assessed by angiographic scores, whereas Angpt-1 or null plasmid had no effect. In addition, Angpt-2 together with VEGF resulted in greater improvement in perfusion and collateral vessel formation than VEGF alone. Similarly, conditional overexpression of Angpt-2 in mice improved ischemic limb blood flow recovery, while Angpt-1 overexpression was ineffective. These data from Tie2 heterozygote deficient mice demonstrate, for the first time, the importance of the Tie2 pathway in spontaneous neovascularization in response to chronic hindlimb ischemia. Moreover, they show that overexpression of the partial agonist, Angpt-2, but not Angpt-1, enhanced ischemic hind limb perfusion recovery and collateralization, suggesting that a coordinated sequence antagonist and agonist activity is required for effective therapeutic revascularization.


Asunto(s)
Angiopoyetina 1/genética , Angiopoyetina 2/genética , Endotelio Vascular/metabolismo , Miembro Posterior/irrigación sanguínea , Isquemia/genética , Receptor TIE-2/genética , Factor A de Crecimiento Endotelial Vascular/genética , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Animales , Citomegalovirus/genética , Endotelio Vascular/patología , Regulación de la Expresión Génica , Terapia Genética , Vectores Genéticos , Miembro Posterior/metabolismo , Miembro Posterior/patología , Humanos , Inyecciones Intramusculares , Isquemia/metabolismo , Isquemia/patología , Isquemia/terapia , Masculino , Ratones , Ratones Noqueados , Neovascularización Fisiológica , Ratas , Ratas Sprague-Dawley , Receptor TIE-2/agonistas , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/deficiencia , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
J Cereb Blood Flow Metab ; 27(10): 1684-91, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17356562

RESUMEN

Bone marrow stromal cells (MSCs) increase vascular endothelial growth factor (VEGF) expression and promote angiogenesis after stroke. Angiopoietin-1 (Ang1) and its receptor Tie2 mediate vascular integrity and angiogenesis as does VEGF and its receptors. In this study, we tested whether MSC treatment of stroke increases Ang1/Tie2 expression, and whether Ang1/Tie2 with VEGF/ vascular endothelial growth factor receptor 2 (VEGFR2) (Flk1), in combination, induced by MSCs enhances angiogenesis and vascular integrity. Male Wistar rats were subjected to middle cerebral artery occlusion (MCAo) and treated with or without MSCs. Marrow stromal cell treatment significantly decreased blood-brain barrier (BBB) leakage and increased Ang1, Tie2, and occludin (a tight junction protein) expression in the ischemic border compared with MCAo control. To further test the mechanisms of MSC-induced angiogenesis and vascular stabilization, cocultures of MSCs with mouse brain endothelial cells (MBECs) or astrocytes were performed. Supernatant derived from MSCs cocultured with MBECs significantly increased MBEC expression of Ang1/Tie2 and Flk1 compared with MBEC alone. Marrow stromal cells cocultured with astrocytes also significantly increased astrocyte VEGF and Ang1/Tie2 expression compared with astrocyte culture alone. Conditioned media from MSCs alone, and media from cocultures of MSCs with astrocytes or MBECs, all significantly increased capillary tube-like formation of MBEC compared with control Dulbecco's modified Eagle's medium media. Inhibition of Flk1 and/or Ang1 significantly decreased MSC-induced MBEC tube formation. Knockdown of Tie2 expression in MBECs significantly inhibited MSC-induced tube formation. Our data indicate MSC treatment of stroke promotes angiogenesis and vascular stabilization, which is at least partially mediated by VEGF/Flk1 and Ang1/Tie2.


Asunto(s)
Angiopoyetina 1/metabolismo , Células de la Médula Ósea/metabolismo , Neovascularización Patológica/metabolismo , Receptor TIE-2/metabolismo , Accidente Cerebrovascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Ratas , Ratas Wistar , Receptor TIE-2/deficiencia , Receptor TIE-2/genética , Accidente Cerebrovascular/patología , Células del Estroma/metabolismo
6.
PLoS Med ; 3(3): e46, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16417407

RESUMEN

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a devastating complication of numerous underlying conditions, most notably sepsis. Although pathologic vascular leak has been implicated in the pathogenesis of ARDS and sepsis-associated lung injury, the mechanisms promoting leak are incompletely understood. Angiopoietin-2 (Ang-2), a known antagonist of the endothelial Tie-2 receptor, was originally described as a naturally occurring disruptor of normal embryonic vascular development otherwise mediated by the Tie-2 agonist angiopoietin-1 (Ang-1). We hypothesized that Ang-2 contributes to endothelial barrier disruption in sepsis-associated lung injury, a condition involving the mature vasculature. METHODS AND FINDINGS: We describe complementary human, murine, and in vitro investigations that implicate Ang-2 as a mediator of this process. We show that circulating Ang-2 is significantly elevated in humans with sepsis who have impaired oxygenation. We then show that serum from these patients disrupts endothelial architecture. This effect of sepsis serum from humans correlates with measured Ang-2, abates with clinical improvement, and is reversed by Ang-1. Next, we found that endothelial barrier disruption can be provoked by Ang-2 alone. This signal is transduced through myosin light chain phosphorylation. Last, we show that excess systemic Ang-2 provokes pulmonary leak and congestion in otherwise healthy adult mice. CONCLUSIONS: Our results identify a critical role for Ang-2 in disrupting normal pulmonary endothelial function.


Asunto(s)
Angiopoyetina 2/sangre , Permeabilidad Capilar , Pulmón/irrigación sanguínea , Sepsis/sangre , Sepsis/fisiopatología , Anciano , Angiopoyetina 1/farmacología , Angiopoyetina 2/administración & dosificación , Angiopoyetina 2/farmacología , Animales , Vasos Sanguíneos/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Convalecencia , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/patología , Masculino , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Intercambio Gaseoso Pulmonar/fisiología , Receptor TIE-2/deficiencia , Sepsis/complicaciones , Regulación hacia Arriba/efectos de los fármacos , Quinasas Asociadas a rho
7.
Proc Natl Acad Sci U S A ; 102(23): 8293-8, 2005 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-15928093

RESUMEN

The endothelial cell receptor-tyrosine kinases, VEGF receptor 2 (VEGF-R2) and Tie-2, and their ligands, vascular endothelial growth factor (VEGF) and angiopoietins 1 and 2, respectively, play key roles in tumor angiogenesis. Several studies suggest that the VEGF receptor pathway and the Tie-2 pathway are independent and essential mediators of angiogenesis, leading to the hypothesis that simultaneous interference with both pathways should result in additive effects on tumor growth. In this study, a human melanoma xenograft model (M21) was used to analyze the effects of simultaneous intradiabody depletion of vascular endothelial growth receptor-R2 and Tie-2 on tumor angiogenesis and tumor xenograft growth. The intradiabodies were expressed from recombinant adenovirus delivered through subtumoral injection. Blockade of both VEGF-R2 and Tie-2 pathways simultaneously or the VEGF receptor pathway alone resulted in a significant inhibition of tumor growth and tumor angiogenesis (92.2% and 74.4%, respectively). In addition, immunohistochemical staining of intradiabody-treated tumors demonstrated a decreased number of tumor-associated blood vessels versus control treatment. Previous studies with intrabodies had demonstrated that the Tie-2 receptor pathway was essential for tumor growth. The simultaneous blockade of the VEGF and Tie-2 pathways resulted in effective inhibition of tumor growth and demonstrated the potential of simultaneous targeting of multiple pathways as a therapeutic strategy.


Asunto(s)
Fragmentos Fab de Inmunoglobulinas/inmunología , Melanoma/irrigación sanguínea , Melanoma/patología , Neovascularización Patológica , Receptor TIE-2/deficiencia , Receptores de Factores de Crecimiento Endotelial Vascular/deficiencia , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/farmacología , Melanoma/inmunología , Melanoma/terapia , Ratones , Trasplante de Neoplasias , Fenotipo , Conejos , Receptor TIE-2/inmunología , Receptor TIE-2/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/inmunología , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Blood ; 105(10): 3935-8, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15687236

RESUMEN

Phosphoinositide 3-kinase (PI3K) is activated by transmembrane tyrosine kinases such as vascular endothelial growth factor (VEGF) receptors and Tie2 (tunica intima endothelial kinase 2), both of which are key regulators of vascular development. However, the in vivo role of PI3K during developmental vascularization remains to be defined. Here we demonstrate that mice deficient in the p110alpha catalytic subunit of PI3K display multiple vascular defects, including dilated vessels in the head, reduced branching morphogenesis in the endocardium, lack of hierarchical order of large and small branches in the yolk sac, and impaired development of anterior cardinal veins. These vascular defects are strikingly similar to those in mice defective in the Tie2 signaling pathway. Indeed, Tie2 protein levels were significantly lower in p110alpha-deficient mice. Furthermore, RNA interference of p110alpha in cultured endothelial cells significantly reduced Tie2 protein levels. These findings raise the possibility that PI3K may function as an upstream regulator of Tie2 expression during mouse development.


Asunto(s)
Embrión de Mamíferos/anomalías , Embrión de Mamíferos/irrigación sanguínea , Regulación del Desarrollo de la Expresión Génica , Fosfatidilinositol 3-Quinasas/deficiencia , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Células Endoteliales/metabolismo , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptor TIE-2/deficiencia
9.
Circ Res ; 94(11): 1466-73, 2004 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-15117816

RESUMEN

Vein graft failure within the first month after bypass surgery is largely because of thrombosis. However, systemic study of thrombus formation in vein grafts is still lacking, and few effective techniques are available to prevent this event. Herein, we analyzed the kinetics of thrombosis and tested the effectiveness of locally applied aspirin on prevention of the disease in a mouse model. En face analysis of vein grafts revealed that 67+/-12% and 54+/-17% of the surface areas were covered by microthrombi at 1 and 3 days, respectively. Thrombus generation was also identified by labeling of platelets and fibrin, which occurred in 35 grafts examined at 1 and 3 days and 1, 2, 4, and 8 weeks. In a fifth of grafts, the thrombus occluded the vessel lumen by > or =1/4. Furthermore, a significant loss of endothelial cells was evidenced by beta-gal staining for vein grafts in transgenic mice expressing LacZ gene controlled by TIE2-endothelial specific gene promoter. Following thrombosis, neointimal lesions were significantly increased by 4-fold 2 weeks after the operation. When vein grafts were treated locally with aspirin in pluronic gel-127, the thrombus area was significantly reduced (P<0.005) at 1, 4, and 8 weeks. Interestingly, neointimal lesions were markedly reduced in the local, but not oral, aspirin-treated group at 4 and 8 weeks by 50% to 70% (P<0.005). The mechanism of reduced lesions by locally applied aspirin involved the protection of vein graft endothelium. Thus, we provide strong evidence that thrombus formation occurs before the development of neointimal lesions in vein grafts and that local aspirin treatment successfully reduces vein graft arteriosclerosis through endothelial protection, resulting in reduction of thrombosis.


Asunto(s)
Arteriosclerosis/prevención & control , Aspirina/uso terapéutico , Implantación de Prótesis Vascular , Arterias Carótidas/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Fibrinolíticos/uso terapéutico , Oclusión de Injerto Vascular/prevención & control , Inhibidores de Agregación Plaquetaria/uso terapéutico , Complicaciones Posoperatorias/prevención & control , Trombosis/prevención & control , Túnica Íntima/patología , Venas Cavas/efectos de los fármacos , Animales , Arteriosclerosis/etiología , Aspirina/farmacología , Arterias Carótidas/patología , Arterias Carótidas/cirugía , Modelos Animales de Enfermedad , Fibrinolíticos/farmacología , Oclusión de Injerto Vascular/etiología , Hiperplasia , Ratones , Ratones Noqueados , Ratones Transgénicos , Inhibidores de Agregación Plaquetaria/farmacología , Complicaciones Posoperatorias/etiología , Receptor TIE-2/deficiencia , Receptor TIE-2/genética , Receptor TIE-2/fisiología , Trombosis/etiología , Tromboxano B2/sangre , Túnica Íntima/efectos de los fármacos , Venas Cavas/patología , Venas Cavas/cirugía
10.
Proc Natl Acad Sci U S A ; 100(22): 12753-8, 2003 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-14530387

RESUMEN

In mammals, the continuous production of hematopoietic cells (HCs) is sustained by a small number of hematopoietic stem cells (HSCs) residing in the bone marrow. Early HSC activity arises in the aorta-gonad mesonephros region, within cells localized to the ventral floor of the major blood vessels, suggesting that the first HSCs may be derived from cells capable of giving rise to the hematopoietic system and to the endothelial cells of the vasculature. TIE1 (TIE) and TIE2 (TEK) are related receptor tyrosine kinases with an embryonic expression pattern in endothelial cells, their precursors, and HCs, suggestive of a role in the divergence and function of both lineages. Indeed, gene targeting approaches have shown that TIE1, TIE2, and ligands for TIE2, the angiopoietins, are essential for vascular development and maintenance. To explore possible roles for these receptors in HCs, we have examined the ability of embryonic cells lacking both TIE1 and TIE2 to contribute to developmental and adult hematopoiesis by generating chimeric animals between normal embryonic cells and cells lacking these receptors. We show here that TIE receptors are not required for differentiation and proliferation of definitive hematopoietic lineages in the embryo and fetus; surprisingly, however, these receptors are specifically required during postnatal bone marrow hematopoiesis.


Asunto(s)
Células de la Médula Ósea/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Hígado/embriología , Receptor TIE-1/genética , Receptor TIE-2/genética , Envejecimiento/fisiología , Animales , Células Cultivadas , Quimera , Ensayo de Unidades Formadoras de Colonias , Células Madre Hematopoyéticas/citología , Ligandos , Hígado/citología , Ratones , Ratones Noqueados , Receptor TIE-1/deficiencia , Receptor TIE-2/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA