Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Invest Ophthalmol Vis Sci ; 59(2): 1033-1044, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29450547

RESUMEN

Purpose: We establish and characterize the chronic retinal neovascularization (RNV) induced by intravitreal (IVT) injection of DL-α-aminoadipic acid (AAA) in a rabbit model and investigate the extent and duration of inhibitory actions induced by IVT aflibercept on the RNV. Methods: Rabbits received a single IVT injection of AAA, with weekly follow-up fundus photography, fluorescein angiography (FA), and optical coherence tomography (OCT). After 10 weeks, they received a single IVT aflibercept or control injection. RNV leakage was quantified from FA by image analysis with Photoshop. Some eyes were collected for histologic analysis. Results: IVT AAA produced neuronal degeneration over a large fraction of the retina. RNV formed in the damaged area and by 10 weeks exhibited stable morphology and leakage, which persisted for at least 65 weeks. Control IVT injections did not affect RNV leakage, but IVT aflibercept completely blocked RNV leakage. The inhibition was reversible (i.e., the leakage returned as the drug cleared), and the duration of antileak effects with 500 µg aflibercept was approximately 8 weeks. Partial regression of the pathologic vasculature also occurred with aflibercept, with reestablishment as the drug cleared. Conclusions: This model mimics a chronic human disease in its stability and persistence, and the antileak action of aflibercept is fully reversible with a dose-dependent duration. Therefore, this large eye model is uniquely suitable for investigations into the efficacy and duration of action of novel formulations and pharmacotherapies for retinal vascular diseases, and for studying the underlying pathobiology of retinal angiogenesis.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Modelos Animales de Enfermedad , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Neovascularización Retiniana/tratamiento farmacológico , Vasos Retinianos/efectos de los fármacos , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Barrera Hematorretinal/efectos de los fármacos , Enfermedad Crónica , Relación Dosis-Respuesta a Droga , Angiografía con Fluoresceína , Inmunohistoquímica , Inyecciones Intravítreas , Masculino , Conejos , Neovascularización Retiniana/diagnóstico por imagen , Neovascularización Retiniana/fisiopatología , Factores de Tiempo , Tomografía de Coherencia Óptica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cuerpo Vítreo/metabolismo
2.
Angiogenesis ; 19(3): 389-406, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27234973

RESUMEN

Anti-vascular endothelial growth factor (VEGF) therapies have improved clinical outcomes for patients with cancers and retinal vascular diseases. Three anti-VEGF agents, pegaptanib, ranibizumab, and aflibercept, are approved for ophthalmic indications, while bevacizumab is approved to treat colorectal, lung, and renal cancers, but is also used off-label to treat ocular vascular diseases. The efficacy of bevacizumab relative to ranibizumab in treating neovascular age-related macular degeneration has been assessed in several trials. However, questions persist regarding its safety, as bevacizumab can form large complexes with dimeric VEGF165, resulting in multimerization of the Fc domain and platelet activation. Here, we compare binding stoichiometry, Fcγ receptor affinity, platelet activation, and binding to epithelial and endothelial cells in vitro for bevacizumab and aflibercept, in the absence or presence of VEGF. In contrast to bevacizumab, aflibercept forms a homogenous 1:1 complex with each VEGF dimer. Unlike multimeric bevacizumab:VEGF complexes, the monomeric aflibercept:VEGF complex does not exhibit increased affinity for low-affinity Fcγ receptors, does not activate platelets, nor does it bind to the surface of epithelial or endothelial cells to a greater degree than unbound aflibercept or control Fc. The latter finding reflects the fact that aflibercept binds VEGF in a unique manner, distinct from antibodies not only blocking the amino acids necessary for VEGFR1/R2 binding but also occluding the heparin-binding site on VEGF165.


Asunto(s)
Bevacizumab/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/efectos adversos , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/metabolismo , Bevacizumab/efectos adversos , Bevacizumab/uso terapéutico , Línea Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Degeneración Macular/inmunología , Degeneración Macular/metabolismo , Degeneración Macular/terapia , Ratones , Ratones Transgénicos , Activación Plaquetaria , Unión Proteica , Multimerización de Proteína , Receptores de IgG/genética , Receptores de IgG/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/uso terapéutico , Trombocitopenia/etiología , Trombosis/etiología , Factor A de Crecimiento Endotelial Vascular/inmunología
3.
Nat Med ; 19(10): 1331-1337, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24037094

RESUMEN

Insulin initiates diverse hepatic metabolic responses, including gluconeogenic suppression and induction of glycogen synthesis and lipogenesis. The liver possesses a rich sinusoidal capillary network with a higher degree of hypoxia and lower gluconeogenesis in the perivenous zone as compared to the rest of the organ. Here, we show that diverse vascular endothelial growth factor (VEGF) inhibitors improved glucose tolerance in nondiabetic C57BL/6 and diabetic db/db mice, potentiating hepatic insulin signaling with lower gluconeogenic gene expression, higher glycogen storage and suppressed hepatic glucose production. VEGF inhibition induced hepatic hypoxia through sinusoidal vascular regression and sensitized liver insulin signaling through hypoxia-inducible factor-2α (Hif-2α, encoded by Epas1) stabilization. Notably, liver-specific constitutive activation of HIF-2α, but not HIF-1α, was sufficient to augment hepatic insulin signaling through direct and indirect induction of insulin receptor substrate-2 (Irs2), an essential insulin receptor adaptor protein. Further, liver Irs2 was both necessary and sufficient to mediate Hif-2α and Vegf inhibition effects on glucose tolerance and hepatic insulin signaling. These results demonstrate an unsuspected intersection between Hif-2α-mediated hypoxic signaling and hepatic insulin action through Irs2 induction, which can be co-opted by Vegf inhibitors to modulate glucose metabolism. These studies also indicate distinct roles in hepatic metabolism for Hif-1α, which promotes glycolysis, and Hif-2α, which suppresses gluconeogenesis, and suggest new treatment approaches for type 2 diabetes mellitus.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Proteínas Sustrato del Receptor de Insulina/fisiología , Insulina/metabolismo , Hígado/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Diabetes Mellitus Tipo 2/terapia , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
4.
Endocrinology ; 153(4): 1972-83, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22334711

RESUMEN

Using specific inhibitors established that angiogenesis in the ovarian follicle and corpus luteum is driven by vascular endothelial growth factor. Recently, it has been demonstrated that the Notch ligand, delta-like ligand 4 (Dll4) negatively regulates vascular endothelial growth factor-mediated vessel sprouting and branching. To investigate the role of Dll4 in regulation of the ovarian vasculature, we administered a neutralizing antibody to Dll4 to marmosets at the periovulatory period. The vasculature was examined on luteal d 3 or d 10: angiogenesis was determined by incorporation of bromodeoxyuridine, staining for CD31 and cell death by staining for activated caspase-3. Ovulatory progesterone rises were monitored to determine effects of treatment on luteal function and time to recover normal cycles in a separate group of animals. Additionally, animals were treated in the follicular or midluteal phase to determine effects of Dll4 inhibition on follicular development and luteal function. Controls were treated with human IgG (Fc). Corpora lutea from marmosets treated during the periovulatory period exhibited increased angiogenesis and increased vascular density on luteal d 3, but plasma progesterone was significantly suppressed. By luteal d 10, corpora lutea in treated ovaries were significantly reduced in size, with involution of luteal cells, increased cell death, and suppressed plasma progesterone concentrations. In contrast, initiation of anti-Dll4 treatment during the midluteal phase produced only a slight suppression of progesterone for the remainder of the cycle. Moreover, Dll4 inhibition had no appreciable effect on follicular development. These results show that Dll4 has a specific and critical role in the development of the normal luteal vasculature.


Asunto(s)
Callithrix/fisiología , Cuerpo Lúteo/irrigación sanguínea , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Luteólisis/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Neovascularización Fisiológica/fisiología , Ovario/fisiología , Animales , Anticuerpos Neutralizantes/farmacología , Apoptosis/fisiología , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/patología , Femenino , Inmunoglobulina G/farmacología , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/fisiología , Modelos Animales , Neovascularización Fisiológica/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Progesterona/sangre , Receptores Notch/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología
5.
Angiogenesis ; 15(2): 171-85, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22302382
6.
Arch Ophthalmol ; 129(8): 1042-52, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21825187

RESUMEN

OBJECTIVE: To evaluate the efficacy of systemic and intravitreous administration of VEGF Trap (aflibercept) in a nonhuman primate model of choroidal neovascularization (CNV). METHODS: VEGF Trap treatment on laser-induced CNV was evaluated in 48 adult cynomolgus monkeys. In the prevention arms of the study, VEGF Trap was administered by intravenous injection (3 or 10 mg/kg weekly) or intravitreous injection (50, 250, or 500 µg/eye every 2 weeks) beginning before laser injury. In the treatment arm, a single intravitreous injection (500 µg) was given 2 weeks following laser injury. Laser-induced lesions were scored from grade 1 (no hyperfluorescence) to grade 4 (clinically relevant leakage). Representative lesions were evaluated histologically. RESULTS: Grade 4 leakage developed at 32.4% and 45.4% of the laser sites in animals receiving intravitreous or intravenous administration of placebo at 2 weeks following laser injury, respectively. In contrast, the development of grade 4 lesions was completely or nearly completely prevented in all groups receiving intravenous or intravitreous injections of VEGF Trap. A single intravitreous injection of VEGF Trap (500 µg) administered following the development of CNV reduced the frequency of grade 4 lesions from 44.4% to 0% within 14 days of treatment. Intravitreous VEGF Trap was well tolerated with either no or only mild ocular inflammation. Histological evaluation showed decreased scores for morphologic features of tissue proliferation in the VEGF Trap prevention groups. CONCLUSIONS: VEGF Trap prevented the development of clinically relevant CNV leakage when administered at the lowest doses tested. Moreover, a single intravitreous injection induced inhibition of active CNV leakage. CLINICAL RELEVANCE: The animal model used in this study has an established track record as a predictor of pharmacologic efficacy of antineovascular drugs in humans having the neovascular, or wet, form of age-related macular degeneration.


Asunto(s)
Neovascularización Coroidal/prevención & control , Modelos Animales de Enfermedad , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Permeabilidad Capilar , Neovascularización Coroidal/diagnóstico , Angiografía con Fluoresceína , Inyecciones Intravítreas , Coagulación con Láser , Macaca fascicularis , Receptores de Factores de Crecimiento Endotelial Vascular , Retina/patología
7.
Blood ; 117(24): 6728-37, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21498671

RESUMEN

Blood vessel remodeling is crucial to the formation of the definitive vasculature, but little is known about the mechanisms controlling this process. We show that Delta-like ligand 4 (Dll4)/Notch pathway regulates vessel regression in normal pathologic conditions. Genetic and pharmacologic inhibition of Dll4/Notch prevented retinal capillary regression in the oxygen-induced retinopathy (OIR) model and during normal development. Deletion of the Notch-regulated ankyrin repeat protein, a negative regulator of the Notch pathway, produced an opposite phenotype. Inhibition of Dll4/Notch reduced vessel occlusion, maintaining blood flow that is essential for survival of microvessels. Dll4/Notch inhibition up-regulated the expression of vasodilators adrenomedullin and suppressed the expression of vasoconstrictor angiotensinogen. Angiotensin II induced rapid nonperfusion and regression of developing retinal capillaries, whereas Ace1 and AT1 inhibitors and adrenomedullin attenuated vasoobliteration in OIR, indicating that both pathways are involved in modulating vessel remodeling. In contrast, inhibition of vascular endothelial growth factor-A (VEGF-A) did not result in a pervasive loss of retinal capillaries, demonstrating that reduced expression of VEGF-A is not the proximate cause of capillary regression in OIR. Modulation of VEGF-A and Dll4/Notch signaling produced distinct changes in blood vessel morphology and gene expression, indicating that these pathways can have largely independent functions in vascular remodeling.


Asunto(s)
Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Receptor Notch1/fisiología , Flujo Sanguíneo Regional/genética , Vasoconstricción/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Animales Recién Nacidos , Atrofia/genética , Vasos Sanguíneos/metabolismo , Células CHO , Proteínas de Unión al Calcio , Células Cultivadas , Cricetinae , Cricetulus , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Receptor Notch1/genética , Receptor Notch1/metabolismo , Regeneración/genética , Regeneración/fisiología , Flujo Sanguíneo Regional/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Vasoconstricción/fisiología
8.
Invest Ophthalmol Vis Sci ; 52(7): 4039-47, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21357392

RESUMEN

Purpose. To evaluate the effects of a vascular endothelial growth factor trap (VEGF Trap) on retinal vascular development and pathologic neovascularization (NV) in the canine model of oxygen-induced retinopathy (OIR). Methods. Newborn dogs (postnatal day [P]1) were exposed to 100% O(2) and then returned to room air on P5. VEGF Trap (5, 25, or 250 µg) was injected intravitreally in one eye and human FC (hFc) injected in the fellow eye of air control and oxygen-treated dogs on P8. The retinal vasculature and NV were evaluated on P21. Other oxygen-exposed animals received 5 µg of VEGF Trap or hFc on P22 after confirmation of retinopathy of prematurity (ROP)-like pathology and were evaluated at P45. Results. In air controls, both the vascularized area of the retina and the density of superficial capillaries were reduced in 250 or 25 µg VEGF Trap-injected eyes, and deep capillaries were absent. Eyes that received the 5 µg dose were indistinguishable from controls. In oxygen-treated animals, all eyes injected with VEGF Trap exhibited markedly less intravitreal NV than that of hFc-injected fellow eyes, irrespective of dose. Retinal vascular area in OIR animals was significantly reduced in eyes injected with 250 or 25 µg of VEGF Trap, but the 5 µg dose did not inhibit retinal revascularization. Eyes with existing NV that received 5 µg VEGF Trap at P22 exhibited substantial resolution of OIR pathology at P45. Conclusions. The VEGF Trap inhibited the formation of NV, but higher doses also inhibited revascularization of retina when injected at P8. In contrast, the lowest dose tested effectively blocked NV and caused regression of existing NV, without appreciably affecting vasculogenesis or retinal revascularization. These findings suggest that dose selection is an important variable when considering the use of VEGF-targeting agents for the treatment of ROP.


Asunto(s)
Neovascularización Patológica/prevención & control , Oxígeno/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/administración & dosificación , Enfermedades de la Retina/inducido químicamente , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/crecimiento & desarrollo , Envejecimiento , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Recién Nacido/crecimiento & desarrollo , Inyecciones Intravítreas , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica , Factor de Crecimiento Placentario , Proteínas Gestacionales/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular , Retinopatía de la Prematuridad/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
9.
Contraception ; 82(6): 572-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21074022

RESUMEN

BACKGROUND: This study investigated the effects of inhibition of vascular endothelial growth factor (VEGF) during the first postpartum cycle in marmosets housed with a fertile male where a 90% fertility rate is normal. METHODS: On resumption of mating, females were treated with either 25 mg/kg aflibercept, a potent VEGF inhibitor, or control Fc protein (n=6 per group) at the time of ovulation. Effects on timing of pregnancy were monitored by measuring plasma progesterone, chorionic gonadotropin (CG) and uterine palpation. RESULTS: In five of six Fc-treated controls, the postpartum rise in progesterone was maintained and followed by a sustained rise in CG by Day 30 posttreatment indicating pregnancy. In all six aflibercept-treated animals, progesterone secretion was suppressed in the treatment cycle and a CG rise did not occur by Day 30. Pregnancy was delayed to the next cycle, significantly extending interbirth interval compared to controls. Posttreatment deliveries and infant development were normal. CONCLUSION: These results show that stringent pharmacological inhibition of VEGF suppresses luteal progesterone and prevents the successful establishment of pregnancy.


Asunto(s)
Anticonceptivos Femeninos/administración & dosificación , Fase Luteínica/efectos de los fármacos , Proteínas Recombinantes de Fusión/administración & dosificación , Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Callithrix , Gonadotropina Coriónica/sangre , Femenino , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Masculino , Palpación , Embarazo , Progesterona/sangre , Progesterona/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular
10.
Am J Pathol ; 177(6): 3233-43, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20952594

RESUMEN

Vascular remodeling is a feature of chronic inflammation during which capillaries transform into venules that expand the region of the vasculature in which leakage and leukocyte emigration both occur. Recently, we found that angiopoietin/Tie2 receptor signaling drives the transformation of capillaries into venules at an early stage of the sustained inflammatory response in the airways of mice infected with Mycoplasma pulmonis. However, the precise contributions of both angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) are not clear. In this study, we sought to determine the contribution of Ang2 to this vascular remodeling. Ang2 mRNA expression levels increased and phosphorylated Tie2 immunoreactivity in mucosal blood vessels decreased, indicative of diminished receptor signaling after infection. Selective inhibition of Ang2 throughout the infection by administration of either of two distinct function-blocking antibodies reduced the suppression of Tie2 phosphorylation and decreased the remodeling of mucosal capillaries into venules, the amount of leukocyte influx, and disease severity. These findings are consistent with Ang2 acting as an antagonist of Tie2 receptors and the reduction of Tie2 phosphorylation in endothelial cells rendering the vasculature more responsive to cytokines that promote both vascular remodeling and the consequences of inflammation after M. pulmonis infection. By blocking such changes, Ang2 inhibitors may prove beneficial in the treatment of sustained inflammation in which vascular remodeling, leakage, and leukocyte influx contribute to its pathophysiology.


Asunto(s)
Angiopoyetina 2/fisiología , Vasos Sanguíneos/fisiología , Neovascularización Fisiológica/genética , Sistema Respiratorio/irrigación sanguínea , Enfermedades Respiratorias/genética , Angiopoyetina 2/genética , Angiopoyetina 2/inmunología , Angiopoyetina 2/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Inflamación/genética , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Mycoplasma/complicaciones , Infecciones por Mycoplasma/genética , Infecciones por Mycoplasma/metabolismo , Mycoplasma pulmonis/fisiología , Neovascularización Fisiológica/fisiología , Neumonía/etiología , Neumonía/genética , Neumonía/metabolismo , Neumonía/patología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor TIE-2 , Sistema Respiratorio/metabolismo , Sistema Respiratorio/patología , Enfermedades Respiratorias/metabolismo , Enfermedades Respiratorias/patología
11.
Invest Ophthalmol Vis Sci ; 51(11): 6009-17, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20538989

RESUMEN

PURPOSE: The exudative, or the wet form of age-related macular degeneration (AMD) is characterized by choroidal neovascularization (CNV). A subretinal Matrigel (BD Biosciences, Bedford MA) model of CNV is described here, along with the effects of vascular endothelial growth factor (VEGF) neutralization on the development of CNV and associated inflammation and fibrosis. METHODS: CNV was induced in adult Sprague-Dawley rats by subretinal injection of Matrigel. CNV growth and associated leukocyte infiltration and collagen deposition were examined. VEGF Trap (Regeneron Pharmaceuticals, Tarrytown, NY), a recombinant protein that comprises portions of the extracellular domains of VEGF receptors 1 and 2 and that binds all isoforms of VEGF-A as well as placental growth factor with high affinity, was administered subcutaneously. RESULTS: Initiation of CNV was detected 4 days after Matrigel injection and then increased progressively in size. Systemic administration of VEGF Trap beginning on day 2 and 6 completely prevented development of CNV. When CNV was allowed to develop for 10 days before treatment was initiated, VEGF Trap not only prevented its further progression, but also induced substantial regression of existing lesions. In addition, VEGF Trap treatment reduced the total lesion volume and largely prevented the progressive leukocyte infiltration and fibrosis associated with CNV. CONCLUSIONS: The subretinal Matrigel CNV model provides a convenient tool for the study of the diverse components of complex CNV lesions. The data not only confirm the critical roles of VEGF in the development and maintenance of CNV, but further demonstrate that VEGF and other VEGF receptor 1 ligands promote CNV-associated inflammation and fibrosis.


Asunto(s)
Neovascularización Coroidal/inducido químicamente , Neovascularización Coroidal/prevención & control , Coroiditis/prevención & control , Colágeno/toxicidad , Modelos Animales de Enfermedad , Laminina/toxicidad , Proteoglicanos/toxicidad , Proteínas Recombinantes de Fusión/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Actinas/metabolismo , Animales , Quimiotaxis de Leucocito/efectos de los fármacos , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Colágeno/metabolismo , Combinación de Medicamentos , Fibrosis/prevención & control , Inyecciones Subcutáneas , Antígenos Comunes de Leucocito/metabolismo , Microscopía Confocal , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular , Vimentina/metabolismo
12.
Eur J Ophthalmol ; 20(1): 48-54, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19882518

RESUMEN

PURPOSE: To determine the effect of vascular endothelial growth factor (VEGF) TrapR1R2 on bFGF-induced experimental corneal neovascularization (NV). METHODS: Control pellets or pellets containing 80 ng bFGF were surgically implanted into wild-type C57BL/6 and VEGF-LacZ mouse corneas. The corneas were photographed, harvested, and the percentage of corneal NV was calculated. The harvested corneas were evaluated for VEGF expression. VEGF-LacZ mice received tail vein injections of an endothelial-specific lectin after pellet implantation to determine the temporal and spatial relationship between VEGF expression and corneal NV. Intraperitoneal injections of VEGF TrapR1R2 or a human IgG Fc domain control protein were administered, and bFGF pellet-induced corneal NV was evaluated. RESULTS: NV of the corneal stroma began on day 4 and was sustained through day 21 following bFGF pellet implantation. Progression of vascular endothelial cells correlated with increased VEGF-LacZ expression. Western blot analysis showed increased VEGF expression in the corneal NV zone. Following bFGF pellet implantation, the area of corneal NV in untreated controls was 1.05+/-0.12 mm2 and 1.53+/-0.27 mm2 at days 4 and 7, respectively. This was significantly greater than that of mice treated with VEGF Trap (0.24+/-0.11 mm2 and 0.35+/-0.16 mm2 at days 4 and 7, respectively; p<0.05). CONCLUSIONS: Corneal keratocytes express VEGF after bFGF stimulation and bFGF-induced corneal NV is blocked by intraperitoneal VEGF TrapR1R2 administration. Systemic administration of VEGF TrapR1R2 may have potential therapeutic applications in the management of corneal NV.


Asunto(s)
Neovascularización de la Córnea/prevención & control , Modelos Animales de Enfermedad , Receptores de Factores de Crecimiento/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Western Blotting , Córnea/metabolismo , Córnea/patología , Neovascularización de la Córnea/inducido químicamente , Neovascularización de la Córnea/patología , Factor 2 de Crecimiento de Fibroblastos/toxicidad , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Endocrinology ; 149(12): 6076-83, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18687775

RESUMEN

To assess whether there is a link between estrogen, vascular endothelial growth factor (VEGF), and early aspects of uterine angiogenesis, an acute temporal study was conducted in which ovariectomized baboons were pretreated with VEGF Trap, which sequesters endogenous VEGF, and administered estradiol at time 0 h. Serum estradiol levels approximated 500 pg/ml 4-6 h after estradiol administration. VEGF mRNA levels in endometrial glandular epithelial and stromal cells were increased to values 6 h after estradiol that were 3.74 +/- 0.99-fold (mean +/- se) and 5.70 +/- 1.60-fold greater (P < 0.05), respectively, than at 0 h. Microvessel interendothelial cell tight junctions, which control paracellular permeability, were present in the endometrium at time 0 h, but not evident 6 h after estradiol administration. Thus, microvessel paracellular cleft width increased (P < 0.01, ANOVA) from 5.03 +/- 0.22 nm at 0 h to 7.27 +/- 0.48 nm 6 h after estrogen. In contrast, tight junctions remained intact, and paracellular cleft widths were unaltered in estradiol/VEGF Trap and vehicle-treated animals. Endometrial microvessel endothelial cell mitosis, i.e. percent Ki67+/Ki67- immunolabeled endothelial cells, increased (P < 0.05) from 2.9 +/- 0.3% at 0 h to 21.4 +/- 7.0% 6 h after estrogen treatment but was unchanged in estradiol/VEGF Trap and vehicle-treated animals. In summary, the estrogen-induced disruption of endometrial microvessel endothelial tight junctions and increase in endothelial cell proliferation were prevented by VEGF Trap. Therefore, we propose that VEGF mediates the estrogen-induced increase in microvessel permeability and endothelial cell proliferation as early steps in angiogenesis in the primate endometrium.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Estradiol/farmacología , Uniones Estrechas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Permeabilidad Capilar/efectos de los fármacos , Endometrio/citología , Células Endoteliales/citología , Células Endoteliales/ultraestructura , Estradiol/administración & dosificación , Estradiol/sangre , Femenino , Inmunohistoquímica , Antígeno Ki-67/análisis , Microscopía Electrónica de Transmisión , Microvasos/citología , Microvasos/fisiología , Ovariectomía , Papio , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/ultraestructura , Factor de von Willebrand/análisis , Factor de von Willebrand/inmunología
14.
FASEB J ; 22(10): 3571-80, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18606863

RESUMEN

Despite extensive literature on vascular endothelial growth factor (VEGF) expression and regulation by steroid hormones, the lack of clear understanding of the mechanisms of angiogenesis in the endometrium is a major limitation for use of antiangiogenic therapy targeting endometrial vessels. In the current work, we used the rhesus macaque as a primate model and the decidualized mouse uterus as a murine model to examine angiogenesis during endometrial breakdown and regeneration. We found that blockade of VEGF action with VEGF Trap, a potent VEGF blocker, completely inhibited neovascularization during endometrial regeneration in both models but had no marked effect on preexisting or newly formed vessels, suggesting that VEGF is essential for neoangiogenesis but not survival of mature vessels in this vascular bed. Blockade of VEGF also blocked reepithelialization in both the postmenstrual endometrium and the mouse uterus after decidual breakdown, evidence that VEGF has pleiotropic effects in the endometrium. In vitro studies with a scratch wound assay showed that the migration of luminal epithelial cells during repair involved signaling through VEGF receptor 2-neuropilin 1 (VEGFR2-NP1) receptors on endometrial stromal cells. The leading front of tissue growth during endometrial repair was strongly hypoxic, and this hypoxia was the local stimulus for VEGF expression and angiogenesis in this tissue. In summary, we provide novel experimental data indicating that VEGF is essential for endometrial neoangiogenesis during postmenstrual/postpartum repair.


Asunto(s)
Endometrio/irrigación sanguínea , Endometrio/fisiología , Menstruación/fisiología , Neovascularización Fisiológica , Regeneración , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Movimiento Celular , Endometrio/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Femenino , Macaca mulatta , Menstruación/metabolismo , Ratones , Ratones Endogámicos , Neovascularización Fisiológica/efectos de los fármacos , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/farmacología , Regeneración/efectos de los fármacos , Células del Estroma/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/biosíntesis
15.
Endocrinology ; 149(9): 4413-20, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18499749

RESUMEN

The present studies explore the roles of vascular endothelial growth factor (VEGF) and estradiol on angiogenesis and stromal and epithelial cell proliferation in the marmoset endometrium during the proliferative phase of the ovulatory cycle. At the start of the proliferative phase, marmosets were 1) treated with vehicle, 2) treated with a VEGF inhibitor (VEGF Trap, aflibercept), 3) ovariectomized, 4) ovariectomized and given replacement estradiol, or 5) treated with VEGF Trap and given replacement estradiol. The uterus was examined 10 d later in the late proliferative phase. Changes in endothelial and epithelial cell proliferation were quantified using a volumetric density method after immunohistochemistry for bromodeoxyuridine to localize proliferating cells, CD31 to visualize endothelial cells, and dual staining to distinguish endothelial cell proliferation. Endothelial proliferation was elevated in late proliferative controls but virtually absent after VEGF Trap. Ovariectomy had a similar inhibitory effect, whereas angiogenesis was restored by estrogen replacement. Estradiol replacement in VEGF Trap-treated marmosets resulted in only a small increase in endothelial cell proliferation that remained significantly below control values. VEGF Trap treatment and ovariectomy also markedly reduced stromal cell proliferation but resulted in increased stromal cell density associated with a reduction in overall endometrial volume. Estrogen replacement in both ovariectomized and VEGF Trap-treated animals restored stromal proliferation rates and cell density. These results show that endometrial angiogenesis and stromal proliferation during the proliferative phase are driven by estradiol and that the effect of estrogen on angiogenesis is mediated largely by VEGF.


Asunto(s)
Proliferación Celular , Endometrio/irrigación sanguínea , Estradiol/fisiología , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Callithrix , Proliferación Celular/efectos de los fármacos , Endometrio/citología , Endometrio/fisiología , Estradiol/farmacología , Femenino , Neovascularización Fisiológica/efectos de los fármacos , Tamaño de los Órganos , Ovariectomía , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
16.
Arch Ophthalmol ; 126(1): 71-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18195221

RESUMEN

OBJECTIVE: To evaluate whether hemangiogenesis, lymphangiogenesis, and concomitant invasion of mononuclear phagocytes occurring after high-risk corneal transplantation in already vascularized high-risk recipient corneal beds increase the risk for subsequent immune rejection. METHODS: Three intrastromal sutures were left in place for 6 weeks in the corneas of BALB/c mice, causing neovascularization. Three weeks after suture removal, keratoplasty was performed (donors C57BL/6 mice). The treatment group received a vascular endothelial growth factor A (VEGF-A)-neutralizing cytokine trap at 0, 4, 7, and 14 days postoperatively (Fc protein was used as the control treatment). Morphometry was performed in corneal flat mounts using lymphatic endothelial hyaluronan receptor-1 (a specific lymphatic endothelial marker), CD31 (a panendothelial marker), and F4/80 (a marker for mononuclear phagocytes). RESULTS: After corneal transplantation, significant additional hemangiogenesis (mean area covered by vessels [SD], 68% [18%] postoperatively vs 40% [18%] preoperatively; P = .03) and lymphangiogenesis (12% [1.3%] postoperatively vs 9% [2.8%] preoperatively; P = .03) were observed. Postoperative neutralization of VEGF-A inhibited operation-induced hemangiogenesis (35% [8%]; P = .007) and lymphangiogenesis (6% [1.6%]; P = .03) and decreased the recruitment of mononuclear phagocytes into the graft (mean [SD], 501 cells/mm(2) [152] in treated mice vs 684 cells/mm(2) [35] in Fc controls; P = .03). After 8 weeks, 23% of the treated corneas were not rejected, whereas all control corneas were rejected after 21 days (P = .007). CONCLUSIONS: Neutralization of VEGF-A after high-risk corneal transplantation effectively inhibits postoperative hemangiogenesis, lymphangiogenesis, and recruitment of antigen-presenting cells and improves corneal graft survival. CLINICAL RELEVANCE: Blocking of VEGF-A after high-risk corneal transplantation may be a novel approach to improve graft survival.


Asunto(s)
Neovascularización de la Córnea/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Queratoplastia Penetrante , Proteínas Recombinantes de Fusión/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Células Presentadoras de Antígenos/fisiología , Antígenos de Diferenciación/metabolismo , Neovascularización de la Córnea/etiología , Neovascularización de la Córnea/metabolismo , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Supervivencia de Injerto/fisiología , Receptores de Hialuranos/metabolismo , Linfangiogénesis/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fagocitos/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular
17.
Proc Natl Acad Sci U S A ; 104(47): 18363-70, 2007 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-18000042

RESUMEN

VEGF is the best characterized mediator of tumor angiogenesis. Anti-VEGF agents have recently demonstrated impressive efficacy in human cancer trials, but the optimal dosing of such agents must still be determined empirically, because biomarkers to guide dosing have yet to be established. The widely accepted (but unverified) assumption that VEGF production is quite low in normal adults led to the notion that increased systemic VEGF levels might quantitatively reflect tumor mass and angiogenic activity. We describe an approach to determine host and tumor production of VEGF, using a high-affinity and long-lived VEGF antagonist now in clinical trials, the VEGF Trap. Unlike antibody complexes that are usually rapidly cleared, the VEGF Trap forms inert complexes with tissue- and tumor-derived VEGF that remain stably in the systemic circulation, where they are readily assayable, providing unprecedented capability to accurately measure VEGF production. We report that VEGF production is surprisingly high in non-tumor-bearing rodents and humans, challenging the notion that systemic VEGF levels can serve as a sensitive surrogate for tumor load; tumor VEGF contribution becomes significant only with very large tumor loads. These findings have the important corollary that anti-VEGF therapies must be sufficiently dosed to avoid diversion by host-derived VEGF. We further show that our assay can indicate when VEGF is optimally blocked; such biomarkers to guide dosing do not exist for other anti-VEGF agents. Based on this assay, VEGF Trap doses currently being assessed in clinical trials are in the efficacious range.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Factores de Crecimiento Endotelial Vascular/biosíntesis , Envejecimiento/fisiología , Inhibidores de la Angiogénesis/inmunología , Animales , Anticuerpos/inmunología , Biomarcadores , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones SCID , Unión Proteica , Factores de Crecimiento Endotelial Vascular/sangre , Factores de Crecimiento Endotelial Vascular/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Displasia del Cuello del Útero/metabolismo , Displasia del Cuello del Útero/patología
18.
J Vasc Res ; 44(4): 283-91, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17406120

RESUMEN

BACKGROUND: Infusion of exogenous vascular endothelial growth factor (VEGF) into adult brain at doses above 60 ng/day induces dramatic angiogenesis accompanied by vascular leak and inflammation. Blood vessels formed by this treatment are dilated and tortuous, exhibiting a pathological morphology. Pathological VEGF-induced angiogenesis is preceded by vascular leak and inflammation, which have been proposed to mediate subsequent angiogenesis. METHODS: To test this hypothesis, we infused VEGF into the brains of adult rats to induce pathological angiogenesis. Some of these rats were treated with dexamethasone, a potent anti-inflammatory glucocorticoid, to inhibit inflammation and edema. RESULTS: We demonstrate that inhibition of inflammation by treatment with dexamethasone significantly attenuated VEGF-induced pathological angiogenesis. To present converging evidence that inflammation may be important in this angiogenic process, we also demonstrate that mice genetically deficient in the inflammatory mediator intercellular adhesion molecule-1 have attenuated VEGF-induced angiogenesis. These same mice showed normal amounts of physiological angiogenesis in response to enriched environments, however, suggesting that a generalized reduction in vascular plasticity could not account for their poor angiogenic response to VEGF. CONCLUSIONS: Taken together, the data from these experiments suggest that the inflammation which occurs before or during VEGF-induced pathological brain angiogenesis plays a contributory role in the pathological angiogenic process.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Dexametasona/farmacología , Glucocorticoides/farmacología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Neovascularización Patológica/tratamiento farmacológico , Factores de Edad , Animales , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/inmunología , Ratas , Ratas Sprague-Dawley , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/farmacología
19.
Pain ; 130(3): 267-278, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17379411

RESUMEN

Neuromedin U (NMU) has recently been reported to have a role in nociception and inflammation. To clarify the function of the two known NMU receptors, NMU receptor 1 (NMUR1) and NMU receptor 2 (NMUR2), during nociception and inflammation in vivo, we generated mice in which the genes for each receptor were independently deleted. Compared to wild type littermates, mice deficient in NMUR2 showed a reduced thermal nociceptive response in the hot plate, but not in the tail flick, test. In addition, the NMUR2 mutant mice showed a reduced behavioral response and a marked reduction in thermal hyperalgesia following capsaicin injection. NMUR2-deficient mice also showed an impaired pain response during the chronic, but not acute, phase of the formalin test. In contrast, NMUR1-deficient mice did not show any nociceptive differences compared to their wild type littermates in any of the behavioral tests used. We observed the same magnitude of inflammation in both lines of NMU receptor mutant mice compared to their wild type littermates after injection with complete Freund's adjuvant (CFA), suggesting no requirement for either receptor in this response. Thus, the pro-nociceptive effects of NMU in mice appear to be mediated through NMUR2, not NMUR1.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Nociceptores/fisiología , Umbral del Dolor/fisiología , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/fisiología , Animales , Conducta Animal , Capsaicina , Femenino , Adyuvante de Freund , Expresión Génica , Calor , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dimensión del Dolor
20.
Reproduction ; 132(4): 589-600, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17008470

RESUMEN

The intense angiogenesis characteristic of early corpus luteum development is dependent upon vascular endothelial growth factor (VEGF) as inhibitors of VEGF administered at the peri-ovulatory period suppress endothelial cell proliferation and progesterone secretion. We now report that administration of VEGF Trap, a soluble decoy receptor-based inhibitor, at the mid- or the late luteal phase in the marmoset results in a rapid decline in plasma progesterone. Since vascularisation of the corpus luteum is largely complete by the mid-luteal phase, it suggested that this functional luteolysis involved mechanisms other than inhibition of angiogenesis. A second experiment investigated the role of VEGF in maintaining the integrity of the luteal vasculature and hormone-producing cells. VEGF Trap was administered to marmosets in the mid-luteal phase and ovaries were obtained 1, 2, 4 or 8 days later for localisation of activated caspase-3 staining in the corpus luteum and compared with those obtained 2, 4 and 8 days after administration of control protein. The number of cells with activated caspase-3 staining was significantly increased after administration of VEGF Trap. Dual staining of activated caspase-3 with the endothelial cell marker CD31 showed that at 1 day post-treatment, more than 90% caspase-3-stained cells were vascular endothelium, prior to detection of an increasing incidence in death of hormone-producing cells on days 2 and 4. Staining with CD31 showed that the endothelial cell area was decreased after treatment. By 8 days after treatment, corpora lutea had regressed to varying degrees, while all control corpora lutea remained healthy. These results show that VEGF inhibition in the mid- or the late luteal phase induces functional luteolysis in the marmoset that is associated with premature and selective death of endothelial cells.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Cuerpo Lúteo/irrigación sanguínea , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/patología , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Animales , Callithrix , Muerte Celular , Cuerpo Lúteo/citología , Cuerpo Lúteo/efectos de los fármacos , Células Endoteliales/patología , Femenino , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica/métodos , Fase Luteínica , Luteólisis , Modelos Animales , Progesterona/sangre , Receptores de Factores de Crecimiento , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...