Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
2.
Cell Commun Signal ; 22(1): 225, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38605348

RESUMEN

The endothelial glycocalyx, located at the luminal surface of the endothelium, plays an important role in the regulation of leukocyte adhesion, vascular permeability, and vascular homeostasis. Endomucin (EMCN), a component of the endothelial glycocalyx, is a mucin-like transmembrane glycoprotein selectively expressed by venous and capillary endothelium. We have previously shown that knockdown of EMCN impairs retinal vascular development in vivo and vascular endothelial growth factor 165 isoform (VEGF165)-induced cell migration, proliferation, and tube formation by human retinal endothelial cells in vitro and that EMCN is essential for VEGF165-stimulated clathrin-mediated endocytosis and signaling of VEGF receptor 2 (VEGFR2). Clathrin-mediated endocytosis is an essential step in receptor signaling and is of paramount importance for a number of receptors for growth factors involved in angiogenesis. In this study, we further investigated the molecular mechanism underlying EMCN's involvement in the regulation of VEGF-induced endocytosis. In addition, we examined the specificity of EMCN's role in angiogenesis-related cell surface receptor tyrosine kinase endocytosis and signaling. We identified that EMCN interacts with AP2 complex, which is essential for clathrin-mediated endocytosis. Lack of EMCN did not affect clathrin recruitment to the AP2 complex following VEGF stimulation, but it is necessary for the interaction between VEGFR2 and the AP2 complex during endocytosis. EMCN does not inhibit VEGFR1 and FGFR1 internalization or their downstream activities since EMCN interacts with VEGFR2 but not VEGFR1 or FGFR1. Additionally, EMCN also regulates VEGF121-induced VEGFR2 phosphorylation and internalization.


Asunto(s)
Células Endoteliales , Factor A de Crecimiento Endotelial Vascular , Humanos , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Sialomucinas/metabolismo , Endocitosis , Clatrina/metabolismo
4.
Free Radic Biol Med ; 178: 360-368, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34843917

RESUMEN

Late-stage dry age-related macular degeneration (AMD) or geographic atrophy (GA) is an irreversible blinding condition characterized by degeneration of retinal pigment epithelium (RPE) and the associated photoreceptors. Clinical and genetic evidence supports a role for dysfunctional lipid processing and accumulation of harmful oxidized lipids in the pathogenesis of GA. Using an oxidized low-density lipoprotein (ox-LDL)-induced RPE death assay, we screened and identified sterically-hindered phenol compounds with potent protective activities for RPE. The phenol-containing PPARγ agonist, troglitazone, protected against ox-LDL-induced RPE cell death, whereas other more potent PPARγ agonists did not protect RPE cells. Knockdown of PPARγ did not affect the protective activity of troglitazone in RPE, confirming the protective function is not due to the thiazolidine (TZD) group of troglitazone. Prototypical hindered phenol trolox and its analogs potently protected against ox-LDL-induced RPE cell death whereas potent antioxidants without the phenol group failed to protect RPE. Hindered phenols preserved lysosomal integrity against ox-LDL-induced damage and FITC-labeled trolox was localized to the lysosomes in RPE cells. Analogs of trolox inhibited reactive oxygen species (ROS) formation induced by ox-LDL uptake in a dose-dependent fashion and were effective at sub-micromolar concentrations. Treatment with trolox analog 2,2,5,7,8-pentamethyl-6-chromanol (PMC) significantly induced the expression of the lysosomal protein NPC-1 and reduced intracellular cholesterol level upon ox-LDL uptake. Our data indicate that the lysosomal-localized hindered phenols are uniquely potent in protecting the RPE against the toxic effects of ox-LDL, and may represent a novel pharmacotherapy to preserve the vision in patients with GA.


Asunto(s)
Lipoproteínas LDL , Epitelio Pigmentado de la Retina , Células Epiteliales , Humanos , Fenoles , Pigmentos Retinianos
5.
FASEB J ; 35(12): e22036, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34793603

RESUMEN

Therapeutic angiogenesis would be clinically valuable in situations such as peripheral vascular disease in diabetic patients and tissue reperfusion following ischemia or injury, but approaches using traditional isoforms of vascular endothelial growth factor-A (VEGF) have had little success. The isoform VEGF165 is both soluble and matrix-associated, but can cause pathologic vascular changes. Freely diffusible VEGF121 is not associated with pathologic angiogenesis, but its failure to remain in the vicinity of the targeted area presents therapeutic challenges. In this study, we evaluate the cellular effects of engineered VEGF variants that tether extracellular VEGF121 to the cell membrane with the goal of activating VEGF receptor 2 (VEGFR2) in a sustained, autologous fashion in endothelial cells. When expressed by primary human retinal endothelial cells (hRECs), the engineered, membrane-tethered variants eVEGF-38 and eVEGF-53 provide a lasting VEGF signal that induces cell proliferation and survival, increases endothelial permeability, promotes the formation of a cord/tube network, and stimulates the formation of elongated filopodia on the endothelial cells. The engineered VEGF variants activate VEGFR2, MAPK/ERK, and the Rho GTPase mediators CDC42 and ROCK, activities that are required for the formation of the elongated filopodia. The sustained, pro-angiogenic activities induced by eVEGF-38 and eVEGF-53 support the potential of engineered VEGF variants-overexpressing endothelial cells as a novel combination of gene and cell-based therapeutic strategy for stimulating endothelial cell-autologous therapeutic angiogenesis.


Asunto(s)
Proliferación Celular , Células Endoteliales/citología , Regulación de la Expresión Génica , Mutación , Neovascularización Fisiológica , Seudópodos/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Movimiento Celular , Células Endoteliales/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Retina/citología , Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
6.
Front Cell Dev Biol ; 9: 734346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34616740

RESUMEN

Galectin-3 (Gal3) is a carbohydrate-binding protein reported to promote angiogenesis by influencing vascular endothelial growth factor-A receptor 2 (VEGFR2) signal transduction. Here we evaluated whether the ability of Gal3 to function as an angiogenic factor involved vascular endothelial growth factor (VEGF). To address this possibility we used human retinal microvascular endothelial cells (HRECs) to determine whether exogenous Gal3 requires VEGF to activate VEGFR2 signaling and if Gal3 is required for VEGF to activate VEGFR2. VEGFR2 phosphorylation and HREC migration assays, following either VEGF neutralization with ranibizumab or Gal3 silencing, revealed that VEGF endogenously produced by the HRECs was essential for the effect of exogenous Gal3 on VEGFR2 activation and cell migration, and that VEGF-induced VEGFR2 activation was not dependent on Gal3 in HRECs. Gal3 depletion led to no reduction in VEGF-induced cell function. Since Gal3 has been suggested to be a potential therapeutic target for VEGFR2-mediated angiogenesis, it is crucial to define the possible Gal3-mediated VEGFR2 signal transduction mechanism to aid the development of efficacious therapeutic strategies.

7.
Cells ; 9(9)2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32917003

RESUMEN

Many patients with wet age-related macular degeneration do not respond well to anti- vascular endothelial growth factor A (VEGFA) therapy for choroidal neovascularization (CNV), and the efficacy of anti-VEGFA decreases over time. We investigated the hypothesis that fibrotic changes, in particular via endothelial-to-mesenchymal transition (EndoMT), play a role in CNV and alter the therapeutic effects of VEGFA pathway blockage. Induction of EndoMT of primary human retinal endothelial cells led to a significantly reduced response to VEGFA at the level of gene expression, cellular proliferation, migration, and tube formation. Suppression of EndoMT restored cell responsiveness to VEGFA. In a mouse model of spontaneous CNV, fibrotic changes and EndoMT persisted as the CNV lesions became more established over time. VEGFA receptor-2 (VEGFR2) antagonism further induced fibrosis and EndoMT in the CNV. The combination of VEGFR2 antagonism and fibrosis/EndoMT inhibition was more effective than either individual treatment in reducing CNV. Our data indicate that fibrosis and EndoMT are involved in the progression of CNV, are exacerbated by VEGFR2 inhibition, and could provide an explanation for the reduced efficacy of anti-VEGFA treatment over time.


Asunto(s)
Neovascularización Coroidal/tratamiento farmacológico , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Humanos , Ratones
8.
Cells ; 9(6)2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32517158

RESUMEN

Endomucin (EMCN) is the type I transmembrane glycoprotein, mucin-like component of the endothelial cell glycocalyx. We have previously shown that EMCN is necessary for vascular endothelial growth factor (VEGF)-induced VEGF receptor 2 (VEGFR2) internalization and downstream signaling. To explore the structural components of EMCN that are necessary for its function and the molecular mechanism of EMCN in VEGF-induced endothelial functions, we generated a series of mouse EMCN truncation mutants and examined their ability to rescue VEGF-induced endothelial functions in human primary endothelial cells (EC) in which endogenous EMCN had been knocked down using siRNA. Expression of the mouse full-length EMCN (FL EMCN) and the extracellular domain truncation mutants ∆21-81 EMCN and ∆21-121 EMCN, but not the shortest mutant ∆21-161 EMCN, successfully rescued the VEGF-induced EC migration, tube formation, and proliferation. ∆21-161 EMCN failed to interact with VEGFR2 and did not facilitate VEGFR2 internalization. Deletion of COSMC (C1GalT1C1) revealed that the abundant mucin-type O-glycans were not required for its VEGFR2-related functions. Mutation of the two N-glycosylation sites on ∆21-121 EMCN abolished its interaction with VEGFR2 and its function in VEGFR2 internalization. These results reveal ∆21-121 EMCN as the minimal extracellular domain sufficient for VEGFR2-mediated endothelial function and demonstrate an important role for N-glycosylation in VEGFR2 interaction, internalization, and angiogenic activity.


Asunto(s)
Sialomucinas/química , Sialomucinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Secuencia de Aminoácidos , Endocitosis , Glicosilación , Humanos , Mutación/genética , Dominios Proteicos , Sialomucinas/genética , Transducción de Señal
9.
J Biol Chem ; 295(19): 6641-6651, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32193206

RESUMEN

Contact between inflammatory cells and endothelial cells (ECs) is a crucial step in vascular inflammation. Recently, we demonstrated that the cell-surface level of endomucin (EMCN), a heavily O-glycosylated single-transmembrane sialomucin, interferes with the interactions between inflammatory cells and ECs. We have also shown that, in response to an inflammatory stimulus, EMCN is cleared from the cell surface by an unknown mechanism. In this study, using adenovirus-mediated overexpression of a tagged EMCN in human umbilical vein ECs, we found that treatment with tumor necrosis factor α (TNF-α) or the strong oxidant pervanadate leads to loss of cell-surface EMCN and increases the levels of the C-terminal fragment of EMCN 3- to 4-fold. Furthermore, treatment with the broad-spectrum matrix metalloproteinase inhibitor batimastat (BB94) or inhibition of ADAM metallopeptidase domain 10 (ADAM10) and ADAM17 with two small-molecule inhibitors, GW280264X and GI254023X, or with siRNA significantly reduced basal and TNFα-induced cell-surface EMCN cleavage. Release of the C-terminal fragment of EMCN by TNF-α treatment was blocked by chemical inhibition of ADAM10 alone or in combination with ADAM17. These results indicate that cell-surface EMCN undergoes constitutive cleavage and that TNF-α treatment dramatically increases this cleavage, which is mediated predominantly by ADAM10 and ADAM17. As endothelial cell-surface EMCN attenuates leukocyte-EC interactions during inflammation, we propose that EMCN is a potential therapeutic target to manage vascular inflammation.


Asunto(s)
Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteínas de la Membrana/metabolismo , Sialoglicoproteínas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/metabolismo , Inflamación/patología
10.
FASEB J ; 33(8): 9362-9373, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31141406

RESUMEN

We have previously shown that knockdown of endomucin (EMCN), an integral membrane glycocalyx glycoprotein, prevents VEGF-induced proliferation, migration, and tube formation in vitro and angiogenesis in vivo. In the endothelium, VEGF mediates most of its angiogenic effects through VEGF receptor 2 (VEGFR2). To understand the role of EMCN, we examined the effect of EMCN depletion on VEGFR2 endocytosis and activation. Results showed that although VEGF stimulation promoted VEGFR2 internalization in control endothelial cells (ECs), loss of EMCN prevented VEGFR2 endocytosis. Cell surface analysis revealed a decrease in VEGFR2 following VEGF stimulation in control but not siRNA directed against EMCN-transfected ECs. EMCN depletion resulted in heightened phosphorylation following VEGF stimulation with an increase in total VEGFR2 protein. These results indicate that EMCN modulates VEGFR2 endocytosis and activity and point to EMCN as a potential therapeutic target.-LeBlanc, M. E., Saez-Torres, K. L., Cano, I., Hu, Z., Saint-Geniez, M., Ng, Y.-S., D'Amore, P. A. Glycocalyx regulation of vascular endothelial growth factor receptor 2 activity.


Asunto(s)
Glicocálix/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adenoviridae/genética , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Endocitosis/efectos de los fármacos , Endocitosis/genética , Endocitosis/fisiología , Humanos , Fosforilación/efectos de los fármacos , Sialomucinas/genética , Sialomucinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
11.
Cell Death Dis ; 9(10): 1018, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30282966

RESUMEN

Endogenous vascular endothelial growth factor (VEGF-A) can protect retinal ganglion cells (RGC) from stress-induced cell death in ocular hypertensive glaucoma. To exploit the neuroprotective function of VEGF-A for therapeutic application in ocular disorders such as glaucoma while minimizing unwanted vascular side effects, we engineered two novel VEGF variants, eVEGF-38 and eVEGF-53. These variants of the diffusible VEGF-A isoform VEGF121 are expressed as dimeric concatamers and remain tethered to the cell membrane, thus restricting the effects of the engineered VEGF to the cells expressing the protein. For comparison, we tested a Myc-tagged version of VEGF189, an isoform that binds tightly to the extracellular matrix and heparan sulfate proteoglycans at the cell surface, supporting only autocrine and localized juxtacrine signaling. In human retinal endothelial cells (hREC), expression of eVEGF-38, eVEGF-53, or VEGF189 increased VEGFR2 phosphorylation without increasing expression of pro-inflammatory markers, relative to VEGF165 protein and vector controls. AAV2-mediated transduction of eVEGF-38, eVEGF-53, or VEGF189 into primary mouse RGC promoted synaptogenesis and increased the average total length of neurites and axons per RGC by ~ 12-fold, an increase that was mediated by VEGFR2 and PI3K/AKT signaling. Expression of eVEGF-38 in primary RGC enhanced expression of genes associated with neuritogenesis, axon outgrowth, axon guidance, and cell survival. Transduction of primary RGC with any of the membrane-associated VEGF constructs increased survival both under normal culture conditions and in the presence of the cytotoxic chemicals H2O2 (via VEGFR2/PI3K/AKT signaling) and N-methyl-D-aspartate (via reduced Ca2+ influx). Moreover, RGC number was increased in mouse embryonic stem cell-derived retinal organoid cultures transduced with the eVEGF-53 construct. The novel, engineered VEGF variants eVEGF-38 and eVEGF-53 show promise as potential therapeutics for retinal RGC neuroprotection when delivered using a gene therapy approach.


Asunto(s)
Sustancias Protectoras/metabolismo , Células Ganglionares de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Axones/metabolismo , Línea Celular , Supervivencia Celular/fisiología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Glaucoma/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Neuritas/metabolismo , Neuroprotección/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Prueba de Estudio Conceptual , Proteínas Proto-Oncogénicas c-akt/metabolismo , Retina/metabolismo , Transducción de Señal/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
Sci Rep ; 7(1): 17138, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29215001

RESUMEN

Angiogenesis is central to both normal and pathologic processes. Endothelial cells (ECs) express O-glycoproteins that are believed to play important roles in vascular development and stability. Endomucin-1 (EMCN) is a type I O-glycosylated, sialic-rich glycoprotein, specifically expressed by venous and capillary endothelium. Evidence has pointed to a potential role for EMCN in angiogenesis but it had not been directly investigated. In this study, we examined the role of EMCN in angiogenesis by modulating EMCN levels both in vivo and in vitro. Reduction of EMCN in vivo led to the impairment of angiogenesis during normal retinal development in vivo. To determine the cellular basis of this inhibition, gain- and loss-of-function studies were performed in human retinal EC (HREC) in vitro by EMCN over-expression using adenovirus or EMCN gene knockdown by siRNA. We show that EMCN knockdown reduced migration, inhibited cell growth without compromising cell survival, and suppressed tube morphogenesis of ECs, whereas over-expression of EMCN led to increased migration, proliferation and tube formation. Furthermore, knockdown of EMCN suppressed VEGF-induced signaling as measured by decreased phospho-VEGFR2, phospho-ERK1/2 and phospho-p38-MAPK levels. These results suggest a novel role for EMCN as a potent regulator of angiogenesis and point to its potential as a new therapeutic target for angiogenesis-related diseases.


Asunto(s)
Movimiento Celular , Endotelio Vascular/citología , Neovascularización Patológica/prevención & control , Neovascularización Retiniana/prevención & control , Epitelio Pigmentado de la Retina/citología , Sialoglicoproteínas/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Morfogénesis , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fosforilación , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Epitelio Pigmentado de la Retina/irrigación sanguínea , Epitelio Pigmentado de la Retina/metabolismo , Sialoglicoproteínas/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
13.
Am J Pathol ; 187(10): 2208-2221, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28739342

RESUMEN

Current treatments for choroidal neovascularization, a major cause of blindness for patients with age-related macular degeneration, treat symptoms but not the underlying causes of the disease. Inflammation has been strongly implicated in the pathogenesis of choroidal neovascularization. We examined the inflammatory role of Toll-like receptor 2 (TLR2) in age-related macular degeneration. TLR2 was robustly expressed by the retinal pigment epithelium in mouse and human eyes, both normal and with macular degeneration/choroidal neovascularization. Nuclear localization of NF-κB, a major downstream target of TLR2 signaling, was detected in the retinal pigment epithelium of human eyes, particularly in eyes with advanced stages of age-related macular degeneration. TLR2 antagonism effectively suppressed initiation and growth of spontaneous choroidal neovascularization in a mouse model, and the combination of anti-TLR2 and antivascular endothelial growth factor receptor 2 yielded an additive therapeutic effect on both area and number of spontaneous choroidal neovascularization lesions. Finally, in primary human fetal retinal pigment epithelium cells, ligand binding to TLR2 induced robust expression of proinflammatory cytokines, and end products of lipid oxidation had a synergistic effect on TLR2 activation. Our data illustrate a functional role for TLR2 in the pathogenesis of choroidal neovascularization, likely by promoting inflammation of the retinal pigment epithelium, and validate TLR2 as a novel therapeutic target for reducing choroidal neovascularization.


Asunto(s)
Neovascularización Coroidal/patología , Inflamación/patología , Degeneración Macular/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Receptor Toll-Like 2/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Neutralizantes/farmacología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/efectos de la radiación , Chlamydia/efectos de los fármacos , Chlamydia/efectos de la radiación , Neovascularización Coroidal/complicaciones , Neovascularización Coroidal/metabolismo , Citocinas/metabolismo , Dipéptidos/farmacología , Rayos gamma , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Inflamación/complicaciones , Inflamación/genética , Lípidos/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Degeneración Macular/complicaciones , Degeneración Macular/metabolismo , Masculino , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Oxidación-Reducción , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/efectos de la radiación
14.
Vasc Cell ; 8: 2, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27175278

RESUMEN

BACKGROUND: Endothelial cells (EC) grown on collagen particles inhibit intimal hyperplasia in animal models when applied perivascularly, and this effect appears to be, at least in part, the result of EC-derived soluble factors that suppress local vascular inflammation. To elucidate the molecular basis of the therapeutic effects of EC grown on collagen particles, the anti-inflammatory activity of conditioned medium from these cells was characterized. METHODS: Human aortic EC (HAEC) and, for chromatin immunoprecipitation assays, human umbilical vein EC (HUVEC) were treated with tumor necrosis factor alpha (TNFα) in the presence of conditioned medium generated by HAEC grown on collagen particles (ECPCM), and the anti-inflammatory effects were evaluated by analysing the expression of the inflammation-related adhesion molecules E-selectin and vascular cell adhesion molecule-1 (VCAM-1). The therapeutic activity of ECPCM was studied using the mouse strain JR5558, which develops spontaneous choroidal neovascularisation (CNV) lesions driven by local inflammation. RESULTS: ECPCM significantly suppressed TNFα-induced expression of E-selectin and VCAM-1. ECPCM did not affect the mRNA stability of the two genes, but suppressed TNFα-induced binding of the p65 subunit of NF-kB transcription factor to E-selectin and VCAM-1 promoters. In vivo, systemic ECPCM treatment significantly reduced the CNV area and the recruitment of activated macrophages to the lesions. Characterization of the molecule responsible for the anti-inflammatory activity in ECPCM indicates that it is unlikely to be a protein and that it is not any of the better characterized EC-derived anti-inflammatory molecules. CONCLUSIONS: Medium conditioned by HAEC grown on collagen particles exhibits significant anti-inflammatory activity via inhibition of genes that mediate inflammatory responses in EC.

15.
Am J Pathol ; 185(9): 2534-49, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26188133

RESUMEN

Choroidal neovascularization (CNV) is a defining feature of wet age-related macular degeneration. We examined the functional role of CCR3 in the development of CNV in mice and primates. CCR3 was associated with spontaneous CNV lesions in the newly described JR5558 mice, whereas CCR3 ligands localized to CNV-associated macrophages and the retinal pigment epithelium/choroid complex. Intravitreal injection of neutralizing antibodies against vascular endothelial growth factor receptor 2, CCR3, CC chemokine ligand 11/eotaxin-1, and CC chemokine ligand 24/eotaxin-2 all reduced CNV area and lesion number in these mice. Systemic administration of the CCR3 antagonists GW766994X and GW782415X reduced spontaneous CNV in JR5558 mice and laser-induced CNV in mouse and primate models in a dose-dependent fashion. Combination treatment with antivascular endothelial growth factor receptor 2 antibody and GW766994X yielded additive reductions in CNV area and hyperpermeability in mice. Interestingly, topical GW766994X and intravitreal anti-CCR3 antibody yielded strong systemic effects, reducing CNV in the untreated, contralateral eye. Contrarily, ocular administration of GW782415X in primates failed to substantially elevate plasma drug levels or to reduce the development of grade IV CNV lesions. These findings suggest that CCR3 signaling may be an attractive therapeutic target for CNV, utilizing a pathway that is at least partly distinct from that of vascular endothelial growth factor receptor. The findings also demonstrate that systemic exposure to CCR3 antagonists may be crucial for CNV-targeted activity.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Permeabilidad Capilar/efectos de los fármacos , Neovascularización Coroidal/tratamiento farmacológico , Receptores CCR3/antagonistas & inhibidores , Degeneración Macular Húmeda/tratamiento farmacológico , Animales , Permeabilidad Capilar/inmunología , Coroides/patología , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Factor A de Crecimiento Endotelial Vascular/metabolismo , Degeneración Macular Húmeda/patología
16.
J Vis Exp ; (96)2015 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-25742031

RESUMEN

Progress in understanding the pathophysiology, and providing novel treatments for glaucoma is dependent on good animal models of the disease. We present here a protocol for elevating intraocular pressure (IOP) in the rat, by injecting magnetic microspheres into the anterior chamber of the eye. The use of magnetic particles allows the user to manipulate the beads into the iridocorneal angle, thus providing a very effective blockade of fluid outflow from the trabecular meshwork. This leads to long-lasting IOP rises, and eventually neuronal death in the ganglion cell layer (GCL) as well as optic nerve pathology, as seen in patients with the disease. This method is simple to perform, as it does not require machinery, specialist surgical skills, or many hours of practice to perfect. Furthermore, the pressure elevations are very robust, and reinjection of the magnetic microspheres is not usually required unlike in some other models using plastic beads. Additionally, we believe this method is suitable for adaptation for the mouse eye.


Asunto(s)
Modelos Animales de Enfermedad , Magnetismo , Animales , Femenino , Glaucoma , Inyecciones , Presión Intraocular/efectos de los fármacos , Ratones , Microesferas , Neuronas/efectos de los fármacos , Nervio Óptico/efectos de los fármacos , Ratas , Tonometría Ocular
17.
Invest Ophthalmol Vis Sci ; 55(6): 3709-19, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24845632

RESUMEN

PURPOSE: Characterization of a mouse model of spontaneous choroidal neovascularization (sCNV) and its effect on retinal architecture and function. METHODS: The sCNV mouse phenotype was characterized by using fundus photography, fluorescein angiography, confocal scanning laser ophthalmoscopy (SLO), optical coherence tomography (OCT), ERG, immunostaining, biochemistry, and electron microscopy. A role for VEGF-A signaling in sCNV was investigated by using neutralizing antibodies and a role for macrophages explored by cell-depletion studies. RESULTS: The sCNV starts between postnatal day 10 and 15 (P10-P15), increasing in number and severity causing RPE disruption and dysfunction. Various morphological methods confirmed the choroidal origin and subretinal position of the angiogenic vessels. At approximately P25, vessels were present in the outer retina with instances of anastomosis of some sCNV lesions with the retinal vasculature. The number of CNV lesions was significantly decreased by systemic blockade of the VEGF-A pathway. Choroidal neovascularization size also was significantly modulated by reducing the number of lesion-associated macrophages. Later stages of sCNV were associated with edema, neuronal loss, and dysfunction. CONCLUSIONS: The sCNV mouse is a new model for the study of both early and late events associated with choroidal neovascularization. Pharmacological reduction in sCNV with VEGF-A antagonists and an anti-inflammatory strategy suggests the model may be useful for investigating novel targets for treating human ocular neovascular disease.


Asunto(s)
Coroides/irrigación sanguínea , Neovascularización Coroidal/metabolismo , Edema/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Coroides/ultraestructura , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Edema/patología , Electrorretinografía , Ensayo de Inmunoadsorción Enzimática , Angiografía con Fluoresceína , Fondo de Ojo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Confocal , Microscopía Electrónica , Oftalmoscopía , Fenotipo , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/fisiopatología , Vasos Retinianos/metabolismo , Vasos Retinianos/ultraestructura , Transducción de Señal , Tomografía de Coherencia Óptica
18.
PLoS One ; 8(6): e65518, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23799019

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of legal blindness in the Western world. There are effective treatments for the vascular complications of neo-vascular AMD, but no effective therapies are available for the dry/atrophic form of the disease. A previously described transgenic CFH-gene deficient mouse model, (cfh-/-), shows hallmarks of early AMD. The ocular phenotype has been further analysed to demonstrate amyloid beta (Aß) rich basement membrane deposits associated with activated complement C3. Cfh-/- mice were treated systemically in both prophylactic and therapeutic regimes with an anti-Aß monoclonal antibody (mAb), 6F6, to determine the effect on the cfh-/- retinal phenotype. Prophylactic treatment with 6F6 demonstrated a dose dependent reduction in the accumulation of both Aß and activated C3 deposition. A similar reduction in the retinal endpoints could be seen after therapeutic treatment. Serum Aß levels after systemic administration of 6F6 show accumulation of Aß in the periphery suggestive of a peripheral sink mechanism. In summary, anti-Aß mAb treatment can partially prevent or reverse ocular phenotypes of the cfh-/- mouse. The data support this therapeutic approach in humans potentially modulating two key elements in the pathogenesis of AMD - Aß and activated, complement C3.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Complemento C3/metabolismo , Degeneración Macular/tratamiento farmacológico , Fragmentos de Péptidos/metabolismo , Retina/metabolismo , Péptidos beta-Amiloides/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Hibridomas , Degeneración Macular/inmunología , Degeneración Macular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/inmunología , Retina/patología
19.
Invest Ophthalmol Vis Sci ; 54(2): 1490-500, 2013 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-23385800

RESUMEN

PURPOSE: Two noninvasive delivery strategies for VEGF/PDGF receptor tyrosine kinase inhibitors (RTKI) were explored that exploited uveal retention as a means for establishing an ocular drug depot: a single oral "loading" dose and topical administration. METHODS: Melanin binding was confirmed by centrifugation and mass spectrometry. Ocular retention was examined in pigmented and albino rats. Ocular release kinetics were measured 3 to 28 days postdosing in pigmented rats. Microautoradiography was used to demonstrate retention of RTKI in the uveal tract. A uveal drug depot of pazopanib was created by a single oral dose prior to induction of laser choroidal neovascularization (CNV). Choroid/retinal pigmented epithelium (RPE) retention of a related RTKI with enhanced topical bioavailability, GW771806, was confirmed by bioanalytics, and its ability to regress CNV compared with pazopanib. RESULTS: Pazopanib and GW771806 directly bound melanin and were retained within the uveal tract of pigmented rats for weeks following a single oral dose. Pazopanib was undetectable systemically following a single oral administration prior to CNV induction, and reduced CNV as well as twice daily dosing. Topical ocular delivery of GW771806 at 5 mg/mL led to high choroidal/RPE exposure and significantly regressed CNV lesions; 2 mg/mL prevented lesion progression. CONCLUSIONS: Uveal retention of drugs such as pazopanib can be used to create a sustained-release depot. Topical GW771806 regressed CNV. These data indicate that topical or infrequent oral loading dose treatment with VEGF/PDGF RTKI retained in the choroid/RPE might allow noninvasive treatments for ocular neovascular disease.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Neovascularización Coroidal/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Indazoles/administración & dosificación , Pirimidinas/administración & dosificación , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Sulfonamidas/administración & dosificación , Sulfonas/administración & dosificación , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Administración Oral , Administración Tópica , Inhibidores de la Angiogénesis/farmacocinética , Animales , Autorradiografía , Neovascularización Coroidal/diagnóstico , Neovascularización Coroidal/metabolismo , Femenino , Angiografía con Fluoresceína , Semivida , Indazoles/farmacocinética , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Pirimidinas/farmacocinética , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Sulfonamidas/farmacocinética , Sulfonas/farmacocinética , Úvea/metabolismo
20.
Am J Pathol ; 182(4): 1379-90, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23416159

RESUMEN

Vascular endothelial growth factor A (VEGF-A) is a validated therapeutic target in several angiogenic- and vascular permeability-related pathological conditions, including certain cancers and potentially blinding diseases, such as age-related macular degeneration and diabetic retinopathy. We and others have shown that VEGF-A also plays an important role in neuronal development and neuroprotection, including in the neural retina. Antagonism of VEGF-A function might therefore present a risk to neuronal survival as a significant adverse effect. Herein, we demonstrate that VEGF-A acts directly on retinal ganglion cells (RGCs) to promote survival. VEGF receptor-2 signaling via the phosphoinositide-3-kinase/Akt pathway was required for the survival response in isolated RGCs. These results were confirmed in animal models of staurosporine-induced RGC death and experimental hypertensive glaucoma. Importantly, we observed that VEGF-A blockade significantly exacerbated neuronal cell death in the hypertensive glaucoma model. Our findings highlight the need to better define the risks associated with use of VEGF-A antagonists in the ocular setting.


Asunto(s)
Glaucoma/tratamiento farmacológico , Glaucoma/patología , Fármacos Neuroprotectores/uso terapéutico , Retina/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Citoprotección/efectos de los fármacos , Modelos Animales de Enfermedad , Glaucoma/enzimología , Neuropilinas/metabolismo , Fármacos Neuroprotectores/farmacología , Pruebas de Neutralización , Hipertensión Ocular/tratamiento farmacológico , Hipertensión Ocular/enzimología , Hipertensión Ocular/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Retina/enzimología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/enzimología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Transducción de Señal/efectos de los fármacos , Pruebas de Toxicidad Aguda , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA