Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Diabetologia ; 56(3): 644-53, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23192694

RESUMEN

AIMS/HYPOTHESIS: We sought to determine the impact of long-standing type 1 diabetes on haematopoietic stem/progenitor cell (HSC) number and function and to examine the impact of modulating glycoprotein (GP)130 receptor in these cells. METHODS: Wild-type, gp130(-/-) and GFP chimeric mice were treated with streptozotocin to induce type 1 diabetes. Bone marrow (BM)-derived cells were used for colony-formation assay, quantification of side population (SP) cells, examination of gene expression, nitric oxide measurement and migration studies. Endothelial progenitor cells (EPCs), a population of vascular precursors derived from HSCs, were compared in diabetic and control mice. Cytokines were measured in BM supernatant fractions by ELISA and protein array. Flow cytometry was performed on enzymatically dissociated retina from gfp(+) chimeric mice and used to assess BM cell recruitment to the retina, kidney and blood. RESULTS: BM cells from the 12-month-diabetic mice showed reduced colony-forming ability, depletion of SP-HSCs with a proportional increase in SP-HSCs residing in hypoxic regions of BM, decreased EPC numbers, and reduced eNos (also known as Nos3) but increased iNos (also known as Nos2) and oxidative stress-related genes. BM supernatant fraction showed increased cytokines, GP130 ligands and monocyte/macrophage stimulating factor. Retina, kidney and peripheral blood showed increased numbers of CD11b(+)/CD45(hi)/ CCR2(+)/Ly6C(hi) inflammatory monocytes. Diabetic gp130(-/-) mice were protected from development of diabetes-induced changes in their HSCs. CONCLUSIONS/INTERPRETATION: The BM microenvironment of type 1 diabetic mice can lead to changes in haematopoiesis, with generation of more monocytes and fewer EPCs contributing to development of microvascular complications. Inhibition of GP130 activation may serve as a therapeutic strategy to improve the key aspects of this dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Células Madre Hematopoyéticas/citología , Monocitos/citología , Animales , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/fisiopatología , Células Endoteliales/citología , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes
2.
Mol Cell Biochem ; 316(1-2): 177-86, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18612802

RESUMEN

Ubiquitous protein kinase CK2 participates in a variety of key cellular functions. We have explored CK2 involvement in angiogenesis. As shown previously, CK2 inhibition reduced endothelial cell proliferation, survival and migration, tube formation, and secondary sprouting on Matrigel. Intraperitoneally administered CK2 inhibitors significantly reduced preretinal neovascularization in a mouse model of proliferative retinopathy. In this model, CK2 inhibitors had an additive effect with somatostatin analog, octreotide, resulting in marked dose reduction for the drug to achieve the same effect. CK2 inhibitors may thus emerge as potent future drugs aimed at inhibiting pathological angiogenesis. Immunostaining of the retina revealed predominant CK2 expression in astrocytes. In human diabetic retinas, mRNA levels of all CK2 subunits decreased, consistent with increased apoptosis. Importantly, a specific CK2 inhibitor prevented recruitment of bone marrow-derived hematopoietic stem cells to areas of retinal neovascularization. This may provide a novel mechanism of action of CK2 inhibitors on newly forming vessels.


Asunto(s)
Quinasa de la Caseína II/antagonistas & inhibidores , Movimiento Celular/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Neovascularización Retiniana/enzimología , Neovascularización Retiniana/prevención & control , Animales , Animales Recién Nacidos , Bovinos , Células Cultivadas , Modelos Animales de Enfermedad , Quimioterapia Combinada , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/patología , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Octreótido/farmacología , Retina/efectos de los fármacos , Retina/enzimología , Retina/patología , Neovascularización Retiniana/patología
3.
Microvasc Res ; 74(2-3): 131-44, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17585951

RESUMEN

Pathological angiogenesis in the retina and underlying choroid is a major cause of visual impairment in all age groups. The last decade has seen an explosion in the clinical availability of antiangiogenic compounds. Emphasis has been placed on inhibitors of the VEGF signaling pathway and considerable success has been achieved with aptamers and antibodies that bind VEGF. However, regression of neovascularization is rarely permanent and the regrowth of new vessels, often within a few months, requires multiple applications of drug. A number of antiangiogenic factors such as IGFBP3, SDF-1 blockers, PEDF, gamma-secretase, Delta-like ligand 4, and integrin antagonists have been identified, which act either indirectly on the VEGF system or independent of it. The importance of other candidates such as HIF-1alpha and protein kinase CK2, which act as "master" regulators of angiogenesis, offer realistic alternative targets for pharmacological intervention. The concept of combination therapy is rapidly gaining interest in the eye field and co-administration of two angiogenic agents (e.g., a CK2 inhibitor with a somatostatin analog, octreotide) are often significantly more effective at inhibiting retinal angiogenesis than either drug alone. The following review will discuss the current therapies available for aberrant ocular angiogenesis, consider new candidate targets for development of antiangiogenic compounds and emphasize the importance of combinatorial pharmacological agents in the treatment of such a dynamic cellular event as angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Coroides/efectos de los fármacos , Neovascularización Coroidal/tratamiento farmacológico , Oftalmopatías/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Factores de Crecimiento Nervioso/uso terapéutico , Neovascularización Retiniana/tratamiento farmacológico , Vasos Retinianos/efectos de los fármacos , Animales , Coroides/irrigación sanguínea , Coroides/patología , Oftalmopatías/genética , Oftalmopatías/patología , Humanos , Modelos Biológicos , Neovascularización Patológica/fisiopatología , Factores de Crecimiento Nervioso/genética , Vasos Retinianos/patología
4.
Gene Ther ; 13(9): 752-60, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16437130

RESUMEN

Insulin-like growth factor-I (IGF-I) and its receptor (IGF-IR) are essential for normal ocular development and are expressed in numerous ocular cell types including lens epithelial cells, retinal pigment epithelial cells, Müller cells and endothelial cells. Endothelial cell proliferation is a common feature of proliferative retinopathies and involves abnormal growth of blood vessels within and on the surface of the retina. In an effort to inhibit the formation of these aberrant blood vessels, we cloned an IGF-IR ribozyme into an expression vector that limits expression of the ribozyme to proliferating endothelial cells. An endothelin enhancer and Cdc6 promoter chimera drives expression of the IGF-IR ribozyme. This promoter limited retinal expression of the reporter gene to proliferating endothelial cells in two mouse models of proliferative retinopathy. In addition, expression of the IGF-IR ribozyme by this promoter inhibited aberrant retinal angiogenesis in both models while preserving normal vessels. These results demonstrate the feasibility of IGF-IR ribozyme expression in a selective manner for safer treatment of abnormal angiogenesis associated with retinopathy.


Asunto(s)
Células Endoteliales/metabolismo , Terapia Genética/métodos , ARN Catalítico/genética , Receptor IGF Tipo 1/genética , Retina/metabolismo , Neovascularización Retiniana/terapia , Animales , Proliferación Celular , Células Cultivadas , Células Endoteliales/patología , Expresión Génica , Ingeniería Genética , Humanos , Coagulación con Láser , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Regiones Promotoras Genéticas , Retina/patología , Neovascularización Retiniana/patología , Transfección/métodos
5.
Circ Res ; 93(6): 500-6, 2003 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-12919950

RESUMEN

Adenosine modulates a variety of cellular functions by interacting with specific cell surface G protein-coupled receptors (A1, A2A, A2B, and A3) and is a potential mediator of angiogenesis through the A2B receptor. The lack of a potent, selective A2B receptor inhibitor has hampered its characterization. Our goal was to design a hammerhead ribozyme that would specifically cleave the A2B receptor mRNA and examine its effect on retinal angiogenesis. Ribozymes specific for the mouse and human A2B receptor mRNAs were designed and cloned in expression plasmids. Human embryonic kidney (HEK) 293 cells were transfected with these plasmids and A2B receptor mRNA levels were determined by quantitative real-time RT-PCR. Human retinal endothelial cells (HRECs) were also transfected and cell migration was examined. The effects of these ribozymes on the levels of preretinal neovascularization were determined using a neonatal mouse model of oxygen-induced retinopathy (OIR). We produced a ribozyme with a Vmax of 515+/-125 pmol/min and a Kcat of 36.1+/-8.3 min(-1) (P< or =1x10(-5)). Transfection of HEK293 cells with the plasmid expressing the ribozyme reduced A2B receptor mRNA levels by 45+/-4.8% (P=5.1x10(-5)). Transfection of HRECs reduced NECA-stimulated migration of cells by 47.3+/-1.2% (P=7x10(-4)). Intraocular injection of the constructs into the mouse model reduced preretinal neovascularization by 53.5+/-8.2% (P=4.5x10(-5)). Our results suggest that the A2B receptor ribozyme will provide a tool for the selective inhibition of this receptor and provide further support for the role of A2B receptor in retinal angiogenesis.


Asunto(s)
Antagonistas de Receptores Purinérgicos P1 , ARN Catalítico/metabolismo , Neovascularización Retiniana/terapia , Animales , Animales Recién Nacidos , Secuencia de Bases , Línea Celular , Movimiento Celular , Células Cultivadas , Endotelio/fisiología , Humanos , Cinética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , ARN Mensajero/química , ARN Mensajero/metabolismo , Receptor de Adenosina A2B , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Retina/citología , Retina/fisiología , Neovascularización Retiniana/patología
6.
Invest Ophthalmol Vis Sci ; 42(13): 3320-4, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11726639

RESUMEN

PURPOSE: The role of adenosine receptor (AdoR) antagonists in human retinal endothelial cell function in vitro has previously been determined. In this study, efficacy of AdoR antagonist administration in reducing retinal neovascularization was examined in a mouse pup model of oxygen-induced retinopathy. METHODS: A previously described model of oxygen-induced retinal neovascularization in newborn mouse pups was used to examine the effect of various AdoR antagonists on neovascularization. The nonselective AdoR antagonist xanthine amine congener (XAC), the A(2A)-selective antagonist ZM241385, the A(2B)-selective antagonists 3-N-propylxanthine (enprofylline) and 3-isobutyl-8-pyrrolidinoxanthine (IPDX), and the A(1)-selective antagonist cyclopentyl-1,3-dipropylxanthine (CPX) were used. After the hyperoxia exposure the animals received daily intraperitoneal injections of pharmacologically relevant doses of AdoR antagonists for 5 days. Control animals received vehicle (0.1% dimethyl sulfoxide [DMSO]) alone. The animals were then killed and perfused with fluorescein-dextran. Wholemounts of retinas from one eye were prepared and examined, whereas the retinas of the contralateral eye were embedded, sectioned, and stained for counting neovascular nuclei extending beyond the internal limiting membrane into the vitreous. RESULTS: Angiography of wholemount retinas showed reduction of neovascular tufts in animals treated with selective A(2B) AdoR antagonists. Quantification of the extraretinal neovascular nuclei showed that only animals treated with XAC, enprofylline, or IPDX showed a significant reduction in retinal neovascularization. By contrast, neither CPX nor ZM241385 had an effect on neovascularization. CONCLUSIONS: The A(2B)-selective AdoR antagonists inhibited oxygen-induced retinal neovascularization in vivo and may provide a basis for developing pharmacologic therapies for the treatment of proliferative retinopathies.


Asunto(s)
Neovascularización Patológica/prevención & control , Antagonistas de Receptores Purinérgicos P1 , Vasos Retinianos , Angiografía , Animales , Animales Recién Nacidos , Ratones , Ratones Endogámicos C57BL , Pirrolidinonas/farmacología , Retina/efectos de los fármacos , Retina/patología , Vasos Retinianos/diagnóstico por imagen , Xantinas/farmacología
7.
Growth Horm IGF Res ; 11 Suppl A: S53-9, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11527089

RESUMEN

Evidence suggests the involvement of growth hormone (GH), insulin-like growth factor I (IGF-I) and somatostatin in the pathology associated with diabetic retinopathy. We examined the effect of IGF-I on human retinal endothelial cell (HREC) survival following high glucose exposure and serum starvation, examined the signalling pathways mediating the protective effect of IGF-I on HREC, and characterized somatostatin receptor-induced retinal endothelial cell death. IGF-I (10 ng/ml) protected HREC from apoptosis induced by high glucose and serum starvation. Wortmannin, a specific inhibitor of phosphotidylinositol-3-kinase, blocks the ability of IGF-I to protect HREC from apoptosis. Incubation of HREC in serum-free medium caused a time-dependent increase in c-Jun N-terminal kinase (JNK) activity, and continuous culture of HREC in the presence of IGF-I or vascular endothelial growth factor (VEGF) prevented JNK activation and arrested apoptosis. Activation of tyrosine kinase receptors results in extracellular signal-related kinase (ERK) activation and activation of ERK is required for proliferation. Both IGF-I and VEGF produced a time- and concentration-dependent increase in the activation of ERK. Type 2 and type 3 somatostatin receptors have been implicated in cell-cycle arrest and apoptosis. Activation of the type 3 receptor in HREC resulted in cell death. These studies suggest that IGF-I is critical for HREC survival, and that somatostatin analogues acting through the type 3 receptor have direct effects on retinal endothelial cells. Furthermore, it appears that the therapeutic efficacy of somatostatin analogues lies not only in systemic inhibition of GH, but also in modulating local growth factor effects.


Asunto(s)
Endotelio Corneal/efectos de los fármacos , Endotelio Corneal/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Somatostatina/farmacología , Amidas/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Medio de Cultivo Libre de Suero , Retinopatía Diabética/fisiopatología , Relación Dosis-Respuesta a Droga , Factores de Crecimiento Endotelial/farmacología , Endotelio Corneal/citología , Glucosa/farmacología , Humanos , Indoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Linfocinas/farmacología , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Transducción de Señal , Somatostatina/agonistas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
8.
Invest Ophthalmol Vis Sci ; 42(9): 2068-73, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11481274

RESUMEN

PURPOSE: The nucleoside adenosine has been implicated in angiogenesis. A previous study demonstrated that activation of the A(2B) adenosine receptor (AdoR) increases cAMP accumulation, cell proliferation, and VEGF expression in human retinal endothelial cells (HRECs). In the present study, the role of this receptor was further characterized by examination of the effects of the selective A(2B) AdoR antagonists 3-N-propylxanthine (enprofylline) and 3-isobutyl-8-pyrrolidinoxanthine (IPDX) on AdoR-mediated HREC proliferation, capillary tube formation, and signal-transduction pathways. METHODS: HRECs were exposed to the adenosine analogue 5'-N-ethylcarboxamido-adenosine (NECA) in the absence or presence of AdoR antagonists. Migration was measured using Boyden chambers. Proliferation was assessed by counting cells. Western analysis was used to assess extracellular signal-related kinase (ERK) and cAMP response element-binding protein (CREB) in cell lysates. The effect of AdoR activation on tube formation was studied using cells grown on a synthetic basement membrane matrix. RESULTS: NECA induced proliferation in a concentration-dependent manner that was inhibited by enprofylline and IPDX. NECA stimulated chemotaxis in a concentration-dependent manner that was also blocked by both A(2B) AdoR antagonists. NECA activated ERK and CREB in HRECs. Both A(2B) AdoR antagonists diminished activation of ERK by NECA exposure. ERK activation was also blocked by the ERK-mitogen-activated protein kinase (MAPK) inhibitor PD98059, but not by the protein kinase A (PKA) inhibitor H-89. CREB activation was blocked by H-89, but not by PD98059, suggesting that ERK activation is independent of PKA. NECA enhanced tube formation on the matrix, whereas both A(2B) AdoR antagonists attenuated this effect. CONCLUSIONS: The selective A(2B) AdoR antagonists, enprofylline and IPDX, inhibited NECA-stimulated proliferation, ERK activation, cell migration, and capillary tube formation. A(2B) AdoR inhibition may offer a way to inhibit retinal angiogenesis and provide a novel therapeutic approach to treatment of diseases associated with aberrant neovascularization, such as diabetic retinopathy and retinopathy of prematurity.


Asunto(s)
División Celular/fisiología , Movimiento Celular/fisiología , Endotelio Vascular/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores Purinérgicos P1/metabolismo , Vasos Retinianos/metabolismo , Adenosina-5'-(N-etilcarboxamida)/farmacología , Western Blotting , Capilares , Recuento de Células , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Antagonistas de Receptores Purinérgicos P1 , Receptor de Adenosina A2B , Transducción de Señal , Vasodilatadores/farmacología
9.
Horm Metab Res ; 33(5): 295-9, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11440276

RESUMEN

Clinical management of proliferative diabetic retinopathy has changed very little in the last 5 decades, relying primarily on laser ablation of the retinal vasculature. Several lines of clinical and experimental evidence suggest that somatostatin analogues may be efficacious in inhibiting neovascularization associated with proliferative retinopathy but the mechanism of action for these compounds is unclear. Inhibition of growth hormone secretion and the subsequent suppression of insulin-like growth factor 1 (IGF-1) production by somatostatin has been suggested as the mechanism of action, however, in vitro studies suggest that somatostatin analogues suppress endothelial cell growth through a direct, somatostatin receptor-mediated inhibition of pro-survival signaling pathways. The advent of a new generation of modified peptide and peptidomimetic somatostatin analogues has allowed investigators to more carefully define the receptor subtypes responsible for somatostatin-induced endothelial cell death and may eventually lead to the clinical development of somatostatin analogues that can reduce endothelial cell proliferation, independent of suppression of circulating hormone levels.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Receptores de Somatostatina/efectos de los fármacos , Retinopatía Diabética/patología , Humanos , Somatostatina/análogos & derivados , Somatostatina/fisiología , Somatostatina/uso terapéutico
10.
Invest Ophthalmol Vis Sci ; 41(8): 2296-302, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10892876

RESUMEN

PURPOSE: Previous studies have suggested that disturbances in plasminogen activator inhibitor (PAI)-1 may be relevant to the development of diabetic microvascular complications. To determine whether overexpression of PAI-1 in cells of retinal microvasculature would result in a disease similar to that observed in diabetes, ocular tissue from transgenic mice that overexpress human PAI-1 were examined. METHODS: Transgenic mice were administered ZnSO4 (25 mM) in their water for up to 49 weeks to activate the metallothionein promoter and stimulate human PAI-1. Colloidal gold immunocytochemistry was used to quantify the human PAI-1 antigen at 7, 20, 34, and 49 weeks of ZnSO4 administration. Cross sections of retinal microvessels were examined by electron microscopy for changes in basement membrane (BM) thickness. Retinal digest preparations were examined by light microscopy for possible microangiopathy, including changes in endothelial cell-to-pericyte ratios. RESULTS: Human PAI-1 immunoreactivity was detected throughout the retinal capillaries of transgenic mice receiving zinc and increased significantly (P < 0.001) after 20 to 49 weeks of ZnSO4 administration compared with age-matched transgenic control mice. At 20 and 49 weeks, retinal capillaries of transgenic mice that received zinc showed significantly thickened BMs compared with control animals (P < 0.001). Moreover, wholemounts of the retinal vasculature from PAI-1 transgenic mice demonstrated an increased endothelial cell-to-pericyte ratio. CONCLUSIONS: PAI-1 overexpression in retinal microvasculature leads to retinal disease similar to that observed in diabetic retinopathy.


Asunto(s)
Ratones Transgénicos , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Vasos Retinianos/metabolismo , Inhibidores de Serina Proteinasa/biosíntesis , Animales , Membrana Basal/metabolismo , Membrana Basal/ultraestructura , Capilares , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/ultraestructura , Femenino , Masculino , Ratones , Microscopía Inmunoelectrónica , Pericitos/metabolismo , Pericitos/ultraestructura , Inhibidor 1 de Activador Plasminogénico/genética , Vasos Retinianos/ultraestructura , Inhibidores de Serina Proteinasa/genética , Sulfato de Zinc/administración & dosificación
11.
Diabetes Care ; 23(4): 504-9, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10857943

RESUMEN

OBJECTIVE: The pilot study examined the ability of octreotide to retard progression of diabetic retinopathy (DR) and delay the need for panretinal photocoagulation (PRP) in patients with advanced stages of retinal disease. RESEARCH DESIGN AND METHODS: Patients with severe nonproliferative DR (NPDR) or early non-high-risk proliferative DR (PDR) were randomly assigned to conventional diabetes management (control group, 12 patients) or to treatment with maximally tolerated doses of octreotide (200-5,000 microg/day subcutaneously; 11 patients). Ocular changes in each eye were assessed at a minimum of every 3 months for 15 months or until disease progressed to high-risk PDR requiring laser surgery. Endocrine assessments occurred at 3-month intervals during the study RESULTS: Only 1 of 22 eyes from patients treated with octreotide reached high-risk PDR requiring PRP, compared with control patients, in whom 9 of 24 eyes required PRP. The decreased incidence of progression requiring laser surgery was statistically significant if events were considered independently (P < 0.006). The incidence of ocular disease progression was only 27% in patients treated with octreotide compared with 42% in patients with conventional diabetes management. This treatment effect on whether the retina worsened approached statistical significance using repeated measures analysis (P = 0.0605). Endocrine management was similar between treatment groups. Thyroxine replacement therapy was administered to maintain a euthyroid state for all octreotide-treated patients and 7 of 12 control patients. CONCLUSIONS: Our results suggest that octreotide treatment in euthyroid patients may retard progression of advanced DR and may delay the time to laser surgery.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Octreótido/uso terapéutico , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Retinopatía Diabética/fisiopatología , Progresión de la Enfermedad , Femenino , Hemoglobina Glucada/análisis , Humanos , Infusiones Intravenosas , Inyecciones Subcutáneas , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Octreótido/administración & dosificación , Proyectos Piloto , Vasoconstrictores/administración & dosificación , Vasoconstrictores/uso terapéutico
12.
Free Radic Biol Med ; 28(1): 91-101, 2000 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10656295

RESUMEN

Hyperglycemia in diabetes induces increased levels of hydrogen peroxide (H2O2), a reactive oxygen species generated by reduced nicotinamide adenine dinucleotide (NADH) oxidase. Nontoxic levels of H2O2 increase endothelial cell permeability. Using a model of non-insulin-dependent diabetes, the BBZ/Wor rat, we investigated retinal levels of H2O2, vascular endothelial growth factor (VEGF) and its receptors, VEGF-R1 and VEGF-R2 by transmission electron microscopy at sites of the blood-retinal barrier (BRB). H2O2 localization was done by the cerium NADH oxidase method, and extravasation of endogenous serum albumin was used to document disruption of the BRB. Higher levels of H2O2 were detected in blood vessels of diabetic (78.7 +/- 4.84%) as compared with vessels from nondiabetic rats (39.0 +/- 4.47%). VEGF immunoreactivity was statistically higher in the inner BRB (24.67 +/- 0.33 colloidal gold particles/63 microm2 vs. 21.52 +/- 0.43 colloidal gold particles/63 microm2, p = .0001) and outer BRB (42.56 +/- 0.45 colloidal gold particles/63 microm2 vs. 15.51 +/- 0.51 colloidal gold particles/63 microm2, p = .0001) of diabetic rats as compared with age matched nondiabetic control rats. VEGF-R1 immunoreactivity was significantly higher in diabetic retinas in both the inner BRB (21.66 +/- 0.75 colloidal gold particles/63 microm2 vs. 12.69 +/- 0.61 colloidal gold particles/63 microm2, p = .0001) and outer BRB (22.76 +/- 2.36 colloidal gold particles/63 microm2 vs. 8.53 +/- 2.67 colloidal gold particles/63 microm2, p = .0013). VEGF-R2 was statistically higher in the inner BRB (8.97 +/- 0.57 colloidal gold particles/63 microm2 versus 7.03 +/- 0.65 colloidal gold particles/63 microm2, p = .0419) but not in the outer BRB (29.42 +/- 1.25 colloidal gold particles/63 microm2 vs. 28.07 +/- 1.42 colloidal gold particles/63 microm2, p = .4889). H2O2 levels correlated with increased VEGF (correlation coefficient = 0.82, p = .001) in this model of nonproliferative diabetic retinopathy. These results support that hyperglycemia is one factor that induces retinal endothelial cells in vivo to increase H2O2 via NADH oxidase and stimulates increases in VEGF resulting in disruption of the BRB.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/metabolismo , Proteínas del Ojo/metabolismo , Peróxido de Hidrógeno/metabolismo , Linfocinas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ratas Mutantes/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Retina/metabolismo , Animales , Glucemia/análisis , Barrera Hematorretinal , Hemoglobina Glucada/análisis , Masculino , Ratas , Ratas Endogámicas , Especies Reactivas de Oxígeno/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
Circ Res ; 85(8): 699-706, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10521243

RESUMEN

Adenosine, released in increased amounts by hypoxic tissues, is thought to be an angiogenic factor that links altered cellular metabolism caused by oxygen deprivation to compensatory angiogenesis. Adenosine interacts with 4 subtypes of G protein-coupled receptors, termed A(1), A(2A), A(2B), and A(3). We investigated whether adenosine causes proliferation of human retinal endothelial cells (HRECs) and synthesis of vascular endothelial growth factor (VEGF) and, if so, which adenosine receptor subtype mediates these effects. The nonselective adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA), in a concentration-dependent manner, increased both VEGF mRNA and protein expression by HRECs, as well as proliferation. This proliferative effect of NECA was inhibited by the addition of anti-human VEGF antibody. NECA also increased insulin-like growth factor-I and basic fibroblast growth factor mRNA expression in a time-dependent manner and cAMP accumulation in these cells. In contrast, neither the A(1) agonist N(6)-cyclopentyladenosine nor the A(2A) agonist 2-p-(2-carboxyethyl) phenethylamino-NECA caused any of the above effects of NECA. The effects of NECA were not significantly attenuated by either the A(2A) antagonist SCH58261 or the A(1) antagonist 8-cyclopentyl-1, 3-dipropylxanthine. However, the nonselective adenosine receptor antagonist xanthine amine congener completely inhibited the effects of NECA. Addition of antisense oligonucleotide complementary to A(2B) adenosine receptor mRNA inhibited VEGF protein production by HRECs after NECA stimulation. Thus, the A(2B) adenosine receptor subtype appears to mediate the actions of adenosine to increase growth factor production, cAMP content, and cell proliferation of HRECs. Adenosine activates the A(2B) adenosine receptor in HRECs, which may lead to neovascularization by a mechanism involving increased angiogenic growth factor expression.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/metabolismo , Linfocinas/metabolismo , Receptores Purinérgicos P1/metabolismo , Vasos Retinianos/metabolismo , Adenosina-5'-(N-etilcarboxamida)/farmacología , Anticuerpos/farmacología , División Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo/metabolismo , AMP Cíclico/genética , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/inmunología , Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/genética , Técnica del Anticuerpo Fluorescente , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Linfocinas/genética , Linfocinas/inmunología , Oligonucleótidos Antisentido/farmacología , ARN Mensajero/metabolismo , Vasos Retinianos/citología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
14.
Life Sci ; 65(8): 795-804, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10466745

RESUMEN

Conditioned medium from gestation day 18 rat placental cultures showed potent stimulation of the directional migration of human retinal endothelial cells. To examine the role of major secreted placental proteins in this chemotaxic activity, prolactin-like proteins (PLPs)-B and C were purified from rat placenta using immuno-affinity chromatography. In contrast to conditioned medium, native PLP-B and PLP-C preparations failed to show any significant stimulation of endothelial cell migration. This study further examined the ability of PLP-B to bind to rat receptors for growth hormone (GH-R) and prolactin (PRL-R). In competitive binding assays with [125I]-hGH, neither native nor recombinant PLP-B preparations showed significant high affinity binding to the transfected rat GH-R or PRL-R. In summary, neither PLP-B nor PLP-C exhibit the potent chemotaxis stimulatory activity of placental conditioned media, nor does PLP-B show evidence of ability to act via rat GH or PRL receptors.


Asunto(s)
Placenta/citología , Proteínas Gestacionales/fisiología , Animales , Quimiotaxis/efectos de los fármacos , Quimiotaxis/fisiología , Cromatografía de Afinidad , Medios de Cultivo Condicionados/farmacología , Endotelio Vascular/citología , Hormona del Crecimiento/metabolismo , Unión Proteica , Ratas , Receptores de Prolactina/metabolismo
15.
Regul Pept ; 79(1): 47-53, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9930582

RESUMEN

Growth factors have been implicated in the pathogenesis of restenosis (myointimal hyperplasia after coronary interventions). In this study, we examined the expression of insulin-like growth factor-I (IGF-1), IGF-1 receptor, and transforming growth factor-beta (TGF-beta) in atherosclerotic and normal rabbit iliac arteries following overstretch balloon angioplasty of the iliac arteries to create a vascular lesion. Animals were sacrificed at 0, 3, 7, 15 and 42 days post angioplasty. The iliac arteries were processed for immunocytochemical localization of IGF-1, IGF-1 receptor and TGF-beta using colloidal gold and the data were quantitatively analyzed. IGF-1, IGF-1 receptor and TGF-beta immunoreactivity were all significantly increased in atherosclerotic arteries compared to control at all of the time points examined. Following balloon angioplasty, the levels of IGF-1 and IGF-1 receptor increased significantly in both control and even further in hypercholesterolemic vessels. In control vessels, the IGF-1 levels returned to preintervention levels, while in atherosclerotic vessels, the levels of IGF-1 and IGF-1 receptor remained elevated. In addition, TGF-beta levels in control vessels showed an initial rise in the first week following injury but then returned to baseline levels. In contrast, atherosclerotic vessels demonstrated a sustained expression of TGF-beta. Thus, IGF-1 and TGF-beta expression is different in normal vs. atherosclerotic vessels following vascular injury. The intensity of expression of IGF-1 and its receptor, which is not reduced at 42 days compared to 15 days following injury, support a role for IGF-1 in smooth muscle cell proliferation and migration. The sustained increase in TGF-beta could facilitate extracellular matrix (ECM) accumulation. Local vascular therapy that is directed towards modulating the effects of IGF-1 and TGF-beta could reduce restenosis.


Asunto(s)
Angioplastia de Balón , Arteriosclerosis/metabolismo , Arteriosclerosis/terapia , Arteria Ilíaca/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptor IGF Tipo 1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Angioplastia de Balón/métodos , Animales , Arteria Ilíaca/patología , Arteria Ilíaca/ultraestructura , Conejos
16.
Diabetes ; 47(8): 1311-7, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9703333

RESUMEN

The degree of hyperglycemia correlates with the development of diabetic retinopathy. We investigated the effect of glucose on the expression of matrix metalloproteinase (MMP)-2 and MMP-9 (72-kDa and 92-kDa type IV collagenases, respectively) by human retinal microvascular endothelial cells (HRECs). Cultured HRECs from nondiabetic and diabetic donors were exposed to 5 or 30 mmol/l glucose. Using gelatin zymography, conditioned medium (CM) from all cultures revealed a gelatinolytic band migrating at 65 kDa (representing the proform of MMP-2 that runs at 72 kDa under reducing conditions). This band was unchanged by glucose exposure or the disease state of the donors. CM from nondiabetic HREC cultures demonstrated an additional proteolytic activity migrating at 90 kDa when cells were exposed to 30 mmol/l glucose, but not when they were exposed to 5 mmol/l glucose. This same activity was seen in CM from HREC cultures of diabetic origin in the presence of both 5 and 30 mmol/l glucose. Western analysis confirmed the identity of the 65-kDa band as MMP-2. The anomalous activity at 90 kDa was identified as MMP-2 associated and co-migrating with a fibronectin fragment. Competition-based reverse transcription-polymerase chain reaction revealed that nondiabetic and diabetic HRECs expressed constitutively mRNA for MMP-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-2, and fibronectin. After exposure to 5 or 30 mmol/l glucose, no changes were detected in mRNA levels in MMP-2 or MMP-9, their inhibitors TIMP-1 and TIMP-2, or fibronectin in either nondiabetic or diabetic HREC cultures. These results support the notion that modulation of MMP function by extracellular matrix components occurs in response to glucose and may be relevant to the development of diabetic retinopathy.


Asunto(s)
Colagenasas/metabolismo , Gelatinasas/metabolismo , Metaloendopeptidasas/metabolismo , Vasos Retinianos/enzimología , Western Blotting , Células Cultivadas , Colagenasas/genética , Diabetes Mellitus/enzimología , Diabetes Mellitus/patología , Gelatinasas/genética , Humanos , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Metaloendopeptidasas/genética , Microcirculación/fisiología , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Vasos Retinianos/patología , Transcripción Genética
17.
Diabetes ; 47(8): 1335-40, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9703336

RESUMEN

Capillary morphogenesis involves cell-cell and cell-matrix interactions. Proteases elaborated by capillary cells modify the extracellular matrix (ECM) to facilitate capillary tube formation. Previously, we detected the presence of fibronectin fragments (Fn-f) associated with the proform of matrix metalloprotease-2 (MMP-2) in conditioned medium of human retinal endothelial cells (HRECs). Association of this fragment to latent MMP-2 prevented autocatalytic activation of MMP-2, suggesting a modulatory role of Fn-f in MMP-2 activation. In this report, we examined the potential role of Fn-f on two processes involved in angiogenesis, proliferation and migration of vascular cells. The effects of Fn-f on proliferation were determined by DNA synthesis and cell counts. Their effects on migration were assessed using modified Boyden chambers. Seven Fn-f were tested on vascular cell migration and/or proliferation. Three Fn-f induced migration. Fn-f of 30-kDa and 120-kDa size positively affected proliferation of microvascular cells but not macrovascular cells. A 45-kDa gelatin binding fragment of Fn inhibited HREC proliferation but stimulated pericyte and smooth muscle cell proliferation. The potency of these fragments exceeded that of the known angiogenic growth factor, basic fibroblast growth factor (bFGF), on HREC migration. ECM components such as fibronectin may influence capillary morphogenesis by the generation of fragments that can modulate proliferation, migration, and protease activation. In the setting of diabetes, excess Fn is generated and is available for degradation. Thus, the production of Fn-f may be specifically relevant to the angiogenesis observed in proliferative diabetic retinopathy.


Asunto(s)
Fibronectinas/farmacología , Fragmentos de Péptidos/farmacología , Vasos Retinianos/citología , Vasos Retinianos/efectos de los fármacos , Capilares/citología , Capilares/efectos de los fármacos , Recuento de Células/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Concentración Osmolar
18.
Free Radic Biol Med ; 24(1): 111-20, 1998 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9436620

RESUMEN

This morphological study demonstrates a role for endothelial cells in generating reactive oxygen species in early stages of retinopathy in the BBZ/Wor rat, an obese, noninsulin dependent model of diabetes. Hyperglycemia induced pseudohypoxia results in an imbalance in cytosolic NADH/NAD+. In the oxygen-rich environment of the retina, NADH oxidase generates superoxide radical which is dismutated to hydrogen peroxide. Localization of hydrogen peroxide by the cerium NADH oxidase enzyme activity cytochemical localization technique shows a statistically significant increase of peroxide localization in the central retina of diabetic rats as compared to age-matched, nondiabetic controls. Endothelial cell dysfunction, indicated by leakage of endogenous serum albumin, coincided with areas of NADH oxidase activity localization. In diabetic rats there are increased levels of fibronectin in areas of hydrogen peroxide localization. This in vivo, morphological study is the first demonstration of oxidative injury and endothelial cell dysfunction in the retina of a spontaneous, noninsulin dependent model of diabetes.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus/fisiopatología , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Obesidad , Estrés Oxidativo/fisiología , Retina/enzimología , Animales , Diabetes Mellitus/enzimología , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Tipo 2/enzimología , Endotelio Vascular/fisiología , Radicales Libres , Histocitoquímica , Inmunohistoquímica , Masculino , Ratas , Ratas Endogámicas
19.
Growth Horm IGF Res ; 8(2): 125-32, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10987679

RESUMEN

Human retinal endothelial cell (HREC) cultures of diabetic and non-diabetic origin were examined for the production of insulin-like growth factor I (IGF-I), IGF-I receptor and IGF-binding proteins (IGFBPs) using colloidal gold quantitative immunocytochemistry and quantitative reverse transcriptase-polymerase chain reaction (QRT-PCR). The levels of immunoreactivity for IGF-I receptor and for four IGFBPs (IGFBP-1, -2, -3 and -5) were significantly increased in diabetic HREC cultures. Moreover, diabetic HREC cultures showed significantly less immunoreactivity for IGF-I and for IGFBP-4 as compared to non-diabetic HREC cultures. Message levels for IGF-I decreased two-fold in diabetic HREC and correlated with protein levels. Message levels for IGFBP-1, -2 and -5 increased 1.5-, 1.7- and 1.6-fold, respectively, in diabetic HREC and correlated with protein levels. However, the protein levels for IGF-I receptor and IGFBP-3 and -4 did not correlate with mRNA levels. There were no differences in mRNA levels for IGF-I receptor and IGFBP-3 and -4 between diabetic and non-diabetic HREC cultures, suggesting a post-transcriptional regulation of IGF-I receptor and the two IGFBPs. The net effect, however, supports enhanced IGF-I action in HREC cultures of diabetic origin which is an important cellular event in diabetic retinopathy.


Asunto(s)
Diabetes Mellitus/metabolismo , Endotelio/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptor IGF Tipo 1/metabolismo , Retina/metabolismo , Células Cultivadas , Diabetes Mellitus/patología , Retinopatía Diabética/metabolismo , Endotelio/citología , Endotelio/ultraestructura , Femenino , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Hibridación in Situ , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Microscopía Inmunoelectrónica , Persona de Mediana Edad , ARN Mensajero/metabolismo , Receptor IGF Tipo 1/genética , Retina/citología , Retina/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Int J Radiat Oncol Biol Phys ; 39(2): 437-44, 1997 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9308948

RESUMEN

PURPOSE: To review outcome and treatment sequelae in patients treated with external beam radiotherapy for pituitary adenomas. METHODS AND MATERIALS: One hundred forty-one patients with pituitary adenomas received radiotherapy at the University of Florida and had 2-year minimum potential follow-up. One hundred twenty-one had newly diagnosed adenomas, and 20 had recurrent tumors. Newly diagnosed tumors were treated with surgery and radiotherapy (n = 98) or radiotherapy alone (n = 23). Patients with recurrent tumors received salvage treatment with surgery and radiotherapy (n = 10) or radiotherapy alone (n = 10). The impact of age, sex, presenting symptoms, tumor extent, surgery type, degree of resection, hormonal activity, primary or salvage therapy, and radiotherapy dose on tumor control was analyzed. Tumor control is defined by the absence of radiographic progression and stable or decreased hormone level (in hormonally active tumors) after treatment. Effect of therapy on vision, hormonal function, neurocognitive function, life satisfaction, and affective symptoms were examined. A Likert categorical scale survey was used for assessment of neurocognitive, life satisfaction, and affective symptom status. Survey results from the radiotherapy patients were compared with a control group treated with transsphenoidal surgery alone. Multivariate analysis used the forward step-wise sequence of chi squares for the log rank test. RESULTS: At 10 years, tumor control for the surgery and radiotherapy group (S + RT) was 95% and not statistically different (p = 0.58) than for patients treated with radiotherapy alone (RT) (90%). Patients with prolactin- and ACTH-secreting tumors had significantly worse tumor control, as did patients treated for recurrent tumors. Multivariate analysis for tumor control revealed that only young age was predictive of worse outcome (p = 0.0354). Visual function was either unaffected or improved in most patients, although four patients developed visual loss due to treatment. Hormonal function was affected adversely in 46 of the 93 patients for whom detailed hormonal information was available. Neurocognitive function evaluation revealed that patients in the S + RT group were more likely (p = 0.005) to report difficulty with memory than those in the RT-alone or S-alone groups. No significant difference in life satisfaction or affective symptoms was evident. CONCLUSIONS: Pituitary adenomas are well controlled by external beam radiotherapy, either alone or in combination with surgery. Visual symptoms often improve after treatment. Hormonal sequelae require medical intervention in many patients. Neurocognitive sequelae may be different among treatment groups.


Asunto(s)
Adenoma/radioterapia , Neoplasias Hipofisarias/radioterapia , Adenoma/metabolismo , Adenoma/cirugía , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Cognición , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/radioterapia , Recurrencia Local de Neoplasia/cirugía , Satisfacción del Paciente , Hormonas Hipofisarias/metabolismo , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/cirugía , Terapia Recuperativa , Trastornos de la Visión/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...