Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
2.
Arch Pediatr ; 18 Suppl 2: S79-85, 2011 Jul.
Artículo en Francés | MEDLINE | ID: mdl-21763979

RESUMEN

Retinopathy of prematurity (ROP) is a major cause of visual impairment in premature infants. It is characterized by an arrest in normal retinal vascular development associated with microvascular degeneration, followed by an abnormal hypoxiainduced neovascularization. Recent studies point out that ROP is a multifactorial disease, implicating both oxygen-dependent and oxygen-independent mechanisms. Oxygen-dependent factors leading to microvascular degeneration include generation of reactive oxygen species and suppression of specific oxygen-regulated vascular survival factors, such as vascular endothelial growth factor (VEGF) and erythropoietin. The other major mechanism for the initial capillary loss is oxygen-independent and implicates a deficit in growth factor IGF-1/IGFBP3. The proliferative, second phase of ROP is triggered by increases in vascular growth factors concentrations, in an attempt to compensate for the hypoxic retina. Novel signaling pathways for vascular repair, implicating both metabolite signaling and inflammatory lipids signaling, represent new therapeutic avenues for ROP.


Asunto(s)
Retinopatía de la Prematuridad/epidemiología , Retinopatía de la Prematuridad/fisiopatología , Humanos , Recién Nacido , Factor I del Crecimiento Similar a la Insulina/metabolismo , Estrés Oxidativo/fisiología , Terapia por Inhalación de Oxígeno/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Neovascularización Retiniana , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Doc Ophthalmol ; 120(1): 61-6, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20012155

RESUMEN

Although a great deal of emphasis has been placed on the vasculopathy that is associated with oxygen-induced retinopathy (OIR), our studies also revealed significant and irreversible structural (retinal histology) and functional (scotopic and photopic electroretinograms) impairments that were significantly more severe in pigmented Long-Evans rats compared to the more commonly used albino Sprague Dawley rats. In the following pages, we will highlight what we have learned about the retinal pathophysiological processes of OIR taking place in strains of both rats with the hope that this will trigger investigations into new therapeutic strategies to complement those geared at preventing the vasculopathy.


Asunto(s)
Modelos Animales de Enfermedad , Hiperoxia/fisiopatología , Retina/fisiopatología , Neovascularización Retiniana/fisiopatología , Retinopatía de la Prematuridad/fisiopatología , Animales , Animales Recién Nacidos , Electrorretinografía , Humanos , Recién Nacido , Oxígeno/toxicidad , Ratas , Vasos Retinianos/efectos de los fármacos
4.
Am J Physiol Regul Integr Comp Physiol ; 297(4): R1009-18, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19641130

RESUMEN

Vasopressin type 2 receptor (V2R) exhibits mostly important properties for hydroosmotic equilibrium and, to a lesser extent, on vasomotricity. Drugs currently acting on this receptor are analogs of the natural neuropeptide, arginine vasopressin (AVP), and hence are competitive ligands. Peptides that reproduce specific sequences of a given receptor have lately been reported to interfere with its action, and if such molecules arise from regions remote from the binding site they would be anticipated to exhibit noncompetitive antagonism, but this has yet to be shown for V2R. Six peptides reproducing juxtamembranous regions of V2R were designed and screened; the most effective peptide, cravky (labeled VRQ397), was characterized. VRQ397 was potent (IC(50) = 0.69 +/- 0.25 nM) and fully effective in inhibiting V2R-dependent physiological function, specifically desmopressin-L-desamino-8-arginine-vasopressin (DDAVP)-induced cremasteric vasorelaxation; this physiological functional assay was utilized to avoid overlooking interference of specific signaling events. A dose-response profile revealed a noncompetitive property of VRQ397; correspondingly, VRQ397 bound specifically to V2R-expressing cells could not displace its natural ligand, AVP, but modulated AVP binding kinetics (dissociation rate). Specificity of VRQ397 was further confirmed by its inability to bind to homologous V1 and oxytocin receptors and its inefficacy to alter responses to stimulation of these receptors. VRQ397 exhibited pharmacological permissiveness on V2R-induced signals, as it inhibited DDAVP-induced PGI(2) generation but not that of cAMP or recruitment of beta-arrestin2. Consistent with in vitro and ex vivo effects as a V2R antagonist, VRQ397 displayed anticipated in vivo aquaretic efficacy. We hereby describe the discovery of a first potent noncompetitive antagonist of V2R, which exhibits functional selectivity, in line with properties of a negative allosteric modulator.


Asunto(s)
Antagonistas de los Receptores de Hormonas Antidiuréticas , Antagonistas de Hormonas/farmacología , Músculo Liso/efectos de los fármacos , Miometrio/efectos de los fármacos , Oligopéptidos/farmacología , Vejiga Urinaria/efectos de los fármacos , 6-Cetoprostaglandina F1 alfa/metabolismo , Regulación Alostérica , Animales , Arginina Vasopresina/metabolismo , Línea Celular , AMP Cíclico/metabolismo , Desamino Arginina Vasopresina/metabolismo , Diuresis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Antagonistas de Hormonas/metabolismo , Humanos , Técnicas In Vitro , Ligandos , Masculino , Ratones , Relajación Muscular/efectos de los fármacos , Músculo Liso/metabolismo , Miometrio/metabolismo , Oligopéptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Vejiga Urinaria/metabolismo
6.
Doc Ophthalmol ; 118(1): 37-54, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18483821

RESUMEN

The neonatal rat is born with its eyes closed and an immature visual system, that some say is equivalent to that of a human fetus at 26 weeks of gestation. From birth, the visual system of the newborn rat will gradually mature, the first manifestation of that being the opening of the eye which usually take place at postnatal day 14. Complete maturation of the retina and visual pathways is normally reached at the end of the first month of life. The neonatal rat model thus represents a unique paradigm to study the normal and abnormal maturation of the primary visual pathways that normally occurs in utero in human subjects. Our laboratory has, over the past decade, developed two animal models of postnatally induced retinopathy, namely the Oxygen-Induced Retinopathy (OIR) that share several common features with the human Retinopathy of Prematurity (ROP) and the Light-Induced Retinopathy that is viewed by some as a valid model of some forms of Retinitis Pigmentosa (RP). The following pages review what is known of the pathophysiological processes taking place and suggest possible therapeutic avenues that could be explored in order to halt the degenerative process.


Asunto(s)
Luz , Oxígeno , Retina/efectos de los fármacos , Retina/embriología , Vías Visuales/embriología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Desarrollo Fetal , Humanos , Recién Nacido , Isoprostanos/fisiología , Factor de Activación Plaquetaria/fisiología , Ratas , Retinitis Pigmentosa/fisiopatología , Retinopatía de la Prematuridad/fisiopatología , Tromboxano A2/fisiología
7.
Artículo en Inglés | MEDLINE | ID: mdl-19075779

RESUMEN

Neovascularization (NV) of the normally avascular cornea arises from various causes including inflammation, infection, trauma, and contact lens wear. Corneal NV, whatever the cause, impairs vision and threatens the survival of corneal allografts, thus representing a serious clinical problem for which treatment is limited. Recent interest has focused on vascular endothelial growth factor (VEGF), a key angiogenic factor whose role in corneal NV is amply documented. While experimental studies underscore the efficacy of anti-VEGF targeted agents, there exists no clear consensus on the ideal treatment for this multifaceted pathology. This review discusses the therapeutic potential of CD36, a well established anti-angiogenic receptor. We present evidence that CD36 contributes significantly to the maintenance of corneal avascularity wherein its deficiency leads to age-related corneal NV. Data further reveal that activation of CD36 substantially attenuates and induces regression of inflammatory corneal NV via concerted inhibition of VEGFA, c-Jun N terminal kinase-1, and c-Jun. In parallel studies, we demonstrate that hypoxia, a fundamental stimulus of NV, markedly elevates CD36 corneal expression in a hypoxia-inducible factor-1 and reactive oxygen species dependent manner. Collectively, our findings unveil interesting avenues for future research on the involvement of CD36 in neovascular eye disease and suggest CD36 agonists as potential therapeutic agents for the management of corneal NV, possibly in combination with anti-VEGF therapies.


Asunto(s)
Antígenos CD36/efectos de los fármacos , Antígenos CD36/fisiología , Neovascularización de la Córnea/tratamiento farmacológico , Proteínas Angiogénicas/fisiología , Animales , Córnea/irrigación sanguínea , Córnea/patología , Neovascularización de la Córnea/patología , Humanos , Flujo Sanguíneo Regional/fisiología
8.
Cell Death Differ ; 14(3): 511-23, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16888648

RESUMEN

By comparing mRNA profiles in cultured fibroblasts from patients affected with lysosomal storage diseases, we identified differentially expressed genes common to these conditions. These studies, confirmed by biochemical experiments, demonstrated that lysosomal storage is associated with downregulation of ubiquitin C-terminal hydrolase, UCH-L1 in the cells of eight different lysosomal disorders, as well as in the brain of a mouse model of Sandhoff disease. Induction of lysosomal storage by the cysteine protease inhibitor E-64 also reduced UCH-L1 mRNA, protein level and activity. All cells exhibiting lysosomal storage contained ubiquitinated protein aggregates and showed reduced levels of free ubiquitin and decreased proteasome activity. The caspase-mediated apoptosis in E-64-treated fibroblasts was reversed by transfection with a UCH-L1 plasmid, and increased after downregulation of UCH-L1 by siRNA, suggesting that UCH-L1 deficiency and impairment of the ubiquitin-dependent protein degradation pathway can contribute to the increased cell death observed in many lysosomal storage disorders.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Enfermedades por Almacenamiento Lisosomal/metabolismo , ARN/metabolismo , Transducción de Señal , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina/metabolismo , Animales , Apoptosis , Inhibidores de Cisteína Proteinasa/farmacología , Fibroblastos/enzimología , Fibroblastos/metabolismo , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Enfermedades por Almacenamiento Lisosomal/enzimología , Enfermedades por Almacenamiento Lisosomal/genética , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , ARN Interferente Pequeño , Piel/citología , Piel/enzimología , Piel/metabolismo , Ubiquitina Tiolesterasa/genética
10.
Am J Physiol Regul Integr Comp Physiol ; 289(6): R1580-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16037123

RESUMEN

In hypertension, increased peripheral vascular resistance results from vascular dysfunction with or without structural changes (vessel wall remodeling and/or microvascular rarefaction). Humans with lower birth weight exhibit evidence of vascular dysfunction. The current studies were undertaken to investigate whether in utero programming of hypertension is associated with in vivo altered response and/or abnormal vascular structure. Offspring of Wistar dams fed a normal (CTRL) or low (LP)-protein diet during gestation were studied. Mean arterial blood pressure response to ANG II was significantly increased, and depressor response to sodium nitroprusside (SNP) infusions significantly decreased in male LP adult offspring relative to CTRL. No arterial remodeling was observed in male LP compared with CTRL offspring. Capillary and arteriolar density was significantly decreased in striated muscles from LP offspring at 7 and 28 days of life but was not different in late fetal life [day 21 of gestation (E21)]. Angiogenic potential of aortic rings from LP newborn (day of birth, P0) was significantly decreased. Striated muscle expressions (Western blots) of ANG II AT(1) receptor subtype, endothelial nitric oxide synthase, angiopoietin 1 and 2, Tie 2 receptor, vascular endothelial growth factor and receptor, and platelet-derived growth factor C at E21 and P7 were unaltered by antenatal diet exposure. In conclusion, blood pressure responses to ANG II and SNP are altered, and microvascular structural changes prevail in this model of fetal programming of hypertension. The capillary rarefaction is absent in the fetus and appears in the neonatal period, in association with decreased angiogenic potential. The study suggests that intrauterine protein restriction increases susceptibility to postnatal factors resulting in microvascular rarefaction, which could represent a primary event in the genesis of hypertension.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Hipertensión/patología , Hipertensión/fisiopatología , Microcirculación/patología , Microcirculación/fisiopatología , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Susceptibilidad a Enfermedades/embriología , Susceptibilidad a Enfermedades/patología , Susceptibilidad a Enfermedades/fisiopatología , Femenino , Hipertensión/embriología , Hipertensión/etiología , Masculino , Neovascularización Patológica/embriología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar , Enfermedades Vasculares/embriología , Enfermedades Vasculares/etiología , Enfermedades Vasculares/patología , Enfermedades Vasculares/fisiopatología
11.
J Thromb Haemost ; 3(5): 1056-63, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15869604

RESUMEN

In addition to its hemostatic functions, factor (F)VIIa exhibits cell proliferative properties as seen in angiogenesis and tumor growth. A role for tissue factor (TF) and protease-activated receptors (PAR)-1 and -2 in cell proliferation remain to be clarified. We tested the hypothesis that FVIIa induces cell proliferation by a mechanism involving TF and PAR-2. Human recombinant FVIIa induced cell proliferation of human BOSC23 cells transfected with plasmid containing human TF DNA sequence. Because DNA primase 1 (PRIM1) plays an essential role in cell proliferation, we used the cloned PRIM1 promoter upstream of the reporter gene chloramphenicol acetyl transferase (CAT) to elucidate the mode of action of FVIIa. FVIIa evoked a dose-dependent increase in cell proliferation and PRIM1 induction, which were markedly potentiated (4-5-fold) by the presence of TF and abrogated by TF antisense oligonucleotide. PRIM1 induction by FVIIa was also abolished by PAR-2 but not by PAR-1 antisense. In contrast, thrombin induced a small increase in CAT activity which was unaffected by TF, but was prevented only by PAR-1 antisense as well as the thrombin inhibitor hirudin. Proliferative properties of FVIIa were associated with a TF-dependent increase in intracellular calcium and were mediated by a concordant phosphorylation of p44/42 MAP kinase. In conclusion, data reveal that FVIIa induces PRIM1 and ensuing cellular proliferation via a TF- and of the PARs entirely PAR-2-dependent pathway, in distinction to that of thrombin which is PAR-1-dependent and TF-independent. We speculate that FVIIa-TF-PAR-2 inhibitors may be effective in suppressing cell proliferation.


Asunto(s)
Factor VIIa/metabolismo , Receptor PAR-2/metabolismo , Tromboplastina/fisiología , Northern Blotting , Western Blotting , Línea Celular , Proliferación Celular , Cloranfenicol O-Acetiltransferasa/metabolismo , Clonación Molecular , ADN/química , ADN Primasa/metabolismo , Relación Dosis-Respuesta a Droga , Genes Reporteros , Hirudinas/metabolismo , Humanos , Oligonucleótidos Antisentido/química , Fosforilación , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Receptor PAR-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Trombina/metabolismo , Tromboplastina/metabolismo , Timidina/química , Factores de Tiempo , Transfección
12.
Circulation ; 108(2): 198-204, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12821538

RESUMEN

BACKGROUND: Intravitreal neovascular diseases, as in ischemic retinopathies, are a major cause of blindness. Because inflammatory mechanisms influence vitreal neovascularization and cyclooxygenase (COX)-2 promotes tumor angiogenesis, we investigated the role of COX-2 in ischemic proliferative retinopathy. METHODS AND RESULTS: We describe here that COX-2 is induced in retinal astrocytes in human diabetic retinopathy, in the murine and rat model of ischemic proliferative retinopathy in vivo, and in hypoxic astrocytes in vitro. Specific COX-2 but not COX-1 inhibitors prevented intravitreal neovascularization, whereas prostaglandin E2, mainly via its prostaglandin E receptor 3 (EP3), exacerbated neovascularization. COX-2 inhibition induced an upregulation of thrombospondin-1 and its CD36 receptor, consistent with the observed antiangiogenic effects of COX-2 inhibition; EP3 stimulation reversed effects of COX-2 inhibitors on thrombospondin-1 and CD36. CONCLUSIONS: These findings point to an important role for COX-2 in ischemic proliferative retinopathy, as in diabetes.


Asunto(s)
Retinopatía Diabética/enzimología , Isquemia/enzimología , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Receptores Inmunológicos , Vitreorretinopatía Proliferativa/enzimología , Adulto , Anciano , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Astrocitos/patología , Antígenos CD36/metabolismo , División Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2 , Retinopatía Diabética/complicaciones , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/patología , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Isquemia/complicaciones , Isquemia/patología , Isoenzimas/antagonistas & inhibidores , Linfocinas/metabolismo , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Ratas , Ratas Sprague-Dawley , Receptores de Lipoproteína/metabolismo , Receptores de Prostaglandina E/efectos de los fármacos , Receptores de Prostaglandina E/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E , Subtipo EP4 de Receptores de Prostaglandina E , Receptores Depuradores , Retina/efectos de los fármacos , Retina/enzimología , Retina/patología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/patología , Trombospondina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular , Vitreorretinopatía Proliferativa/complicaciones , Vitreorretinopatía Proliferativa/tratamiento farmacológico
13.
Am J Physiol Regul Integr Comp Physiol ; 281(5): R1343-60, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11641101

RESUMEN

Prostanoids exert significant effects on circulatory beds. They play a role in the response of the vasculature to adjustments in perfusion pressure and oxygen and carbon dioxide tension, and they mediate the actions of numerous factors. The role of prostanoids in governing circulation of the perinate is suggested to surpass that in the adult. Prostanoids are abundantly generated in the perinate. They have been implicated in autoregulation of blood flow as studied in brain and eyes. Prostaglandins are also dominant regulators of ductus arteriosus tone. The effects of these autacoids are mediated through specific G protein-coupled receptors. In addition to the pharmacological characterization of the prostanoid receptors, important advances in understanding the biology of these receptors have been made in the last decade. Their cloning and the development of animals with disrupted genes of these receptors have been very informative. The involvement of prostanoid receptors in the developing subject, especially on brain and ocular vasculature and on ductus arteriosus, has also begun to be investigated; the expression of these receptors changes with development. Some but not all of the ontogenic changes in these receptors are attributed to homologous regulation. Interestingly, in the process of elucidating their effects, functional perinuclear prostaglandin E2 receptors have been uncovered. This article reviews prostanoid receptors and addresses implications on the developing subject with attention to vascular physiology.


Asunto(s)
Vasos Sanguíneos/metabolismo , Prostaglandinas/metabolismo , Receptores de Prostaglandina/fisiología , Animales , Animales Recién Nacidos , Circulación Cerebrovascular/fisiología , Conducto Arterial/fisiología , Ecocardiografía , Ojo/anatomía & histología , Ojo/irrigación sanguínea , Ojo/metabolismo , Humanos , Modelos Biológicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Flujo Sanguíneo Regional , Transducción de Señal/fisiología
14.
J Appl Physiol (1985) ; 91(4): 1655-62, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11568147

RESUMEN

We examined whether nitric oxide (NO) generated from neuronal NO synthase (nNOS) contributes to the reduced ability of the newborn to autoregulate retinal blood flow (RBF) and choroidal blood flow (ChBF) during acute rises in perfusion pressure. In newborn pigs (1-2 days old), RBF (measured by microsphere) is autoregulated over a narrow range of perfusion pressure, whereas ChBF is not autoregulated. N(G)-nitro-L-arginine methyl ester (L-NAME) or specific nNOS inhibitors 7-nitroindazole, 3-bromo-7-nitroindazole, and 1-(2-trifluoromethyl-phenyl)imidazole as well as ganglionic blocker hexamethonium, unveiled a ChBF autoregulation as observed in juvenile (4- to 6-wk old) animals, whereas autoregulation of RBF in the newborn was only enhanced by L-NAME. All NOS inhibitors and hexamethonium prevented the hypertension-induced increase in NO mediator cGMP in the choroid. nNOS mRNA expression and activity were three- to fourfold higher in the choroid of newborn pigs than in tissues of juvenile pigs. It is concluded that increased production of NO from nNOS curtails ChBF autoregulation in the newborn and suggests a role for the autonomic nervous system in this important hemodynamic function, whereas, for RBF autoregulation, endothelial NOS seems to exert a more important contribution in limiting autoregulation.


Asunto(s)
Animales Recién Nacidos/fisiología , Plexo Coroideo/irrigación sanguínea , Homeostasis/fisiología , Óxido Nítrico Sintasa/fisiología , Animales , Análisis de los Gases de la Sangre , GMP Cíclico/metabolismo , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo III , Ensayos de Protección de Nucleasas , ARN Mensajero/biosíntesis , Flujo Sanguíneo Regional/fisiología , Vasos Retinianos/efectos de los fármacos , Porcinos
15.
Am J Physiol Regul Integr Comp Physiol ; 281(2): R391-400, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11448840

RESUMEN

15-F(2t)-isoprostane (15-F(2t)-IsoP), also termed 8-isoprostaglandin F(2alpha), is one of a series of prostanoids formed by free radical-mediated peroxidation of arachidonic acid and exerts potent biological actions such as vasoconstriction. We recently demonstrated that 15-F(2t)-IsoP is metabolized in humans to a major metabolite, 2,3-dinor-5,6-dihydro-15-F(2t)-IsoP (15-F(2t)-IsoP-M). 15-F(2t)-IsoP-M can also potentially be formed as a product of free radical-induced oxidation of the low abundance fatty acid gamma-linolenic acid. We confirmed that 15-F(2t)-IsoP-M is generated during oxidation of gamma-linolenic acid and explored whether it may exhibit biological activity. 15-F(2t)-IsoP-M caused marked constriction of porcine surface retinal and intraparenchymal brain microvessels, comparable to that observed with 15-F(2t)-IsoP. These effects were associated with increased thromboxane A(2) (TXA(2)) formation and were virtually abolished by TXA(2)-synthase and -receptor inhibitors (CGS-12970 and L-670596). Vasoconstriction induced by either 15-F(2t)-IsoP or 15-F(2t)-IsoP-M on perfused ocular choroid was also abrogated by TXA(2)-synthase inhibition as well as by removal of endothelium. Similar to 15-F(2t)-IsoP, 15-F(2t)-IsoP-M evoked vasoconstriction and TXA(2) generation by activating Ca(2+) influx from nonvoltage-gated channels (SK&F96365 sensitive) in the retina and from both nonvoltage- and N-type voltage-gated Ca(2+) channels (omega-conotoxin MVIIA sensitive), respectively, in brain endothelial and astroglial cells; smooth muscle cells were unresponsive to both agents. Cross-desensitization experiments further suggest that 15-F(2t)-IsoP and 15-F(2t)-IsoP-M act on the same receptor mechanism. Findings reveal a novel concept by which a beta-oxidation metabolite of 15-F(2t)-IsoP that can also be formed by nonenzymatic oxidation of gamma-linolenic acid is equivalently bioactive to 15-F(2t)-IsoP and may prolong the vascular actions of F(2)-IsoPs.


Asunto(s)
Encéfalo/irrigación sanguínea , Dinoprost/farmacología , Vasos Retinianos/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Carbazoles/farmacología , Dinoprost/análogos & derivados , Dinoprost/química , Dinoprost/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , F2-Isoprostanos , Humanos , Técnicas In Vitro , Microcirculación/efectos de los fármacos , Microcirculación/metabolismo , Antagonistas de Prostaglandina/farmacología , Piridinas/farmacología , Vasos Retinianos/metabolismo , Porcinos , Tromboxano A2/antagonistas & inhibidores , Tromboxano A2/metabolismo , Vasoconstrictores/química , Vasoconstrictores/metabolismo , Ácido gammalinolénico/química , Ácido gammalinolénico/metabolismo
16.
Am J Physiol Regul Integr Comp Physiol ; 281(1): R291-301, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11404305

RESUMEN

Permanent closure of the full-term newborn ductus arteriosus (DA) occurs only if profound hypoxia develops within the vessel wall during luminal obliteration. We used fetal and newborn baboons and lambs to determine why the immature DA fails to remodel after birth. When preterm newborns were kept in a normoxic range (Pa(O(2)): 50-90 mmHg), 86% still had a small patent DA on the sixth day after birth; in addition, the preterm DA wall was only mildly hypoxic and had only minimal remodeling. The postnatal increase in Pa(O(2)) normally induces isometric contractile responses in rings of DA; however, the excessive inhibitory effects of endogenous prostaglandins and nitric oxide, coupled with a weaker intrinsic DA tone, make the preterm DA appear to have a smaller increment in tension in response to oxygen than the DA near term. We found that oxygen concentrations, beyond the normoxic range, produce an additional increase in tension in the preterm DA that is similar to the contractile response normally seen at term. We predicted that preterm newborns, kept at a higher Pa(O(2)), would have increased DA tone and would be more likely to obliterate their lumen. We found that preterm newborns, maintained at a Pa(O(2)) >200 mmHg, had only a 14% incidence of patent DA. Even though DA constriction was due to elevated Pa(O(2)), obliteration of the lumen produced profound hypoxia of the DA wall and the same features of remodeling that were observed at term. DA wall hypoxia appears to be both necessary and sufficient to produce anatomic remodeling in preterm newborns.


Asunto(s)
Conducto Arterial/metabolismo , Conducto Arterial/fisiología , Hipoxia/metabolismo , Hipoxia/fisiopatología , 6-Cetoprostaglandina F1 alfa/metabolismo , Animales , Animales Recién Nacidos , Inhibidores de la Ciclooxigenasa/farmacología , Dinoprostona/metabolismo , Inhibidores Enzimáticos/farmacología , Técnicas In Vitro , Indometacina/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Oxígeno/farmacología , Papio , Ovinos
17.
J Appl Physiol (1985) ; 90(6): 2279-88, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11356793

RESUMEN

Microvascular degeneration is an important event in oxygen-induced retinopathy (OIR), a model of retinopathy of prematurity. Because oxidant stress abundantly generates thromboxane A2 (TxA2), we tested whether TxA2 plays a role in retinal vasoobliteration of OIR and contributes to such vascular degeneration by direct endothelial cytotoxicity. Hyperoxia-induced retinal vasoobliteration in rat pups (80% O2 exposure from postnatal days 5-14) was associated with increased TxB2 generation and was significantly prevented by TxA2 synthase inhibitor CGS-12970 (10 mg x kg(-1) x day(-1)) or TxA2-receptor antagonist CGS-22652 (10 mg x kg(-1) x day(-1)). TxA2 mimetics U-46619 (EC50 50 nM) and I-BOP (EC50 5 nM) caused a time- and concentration-dependent cell death of neuroretinovascular endothelial cells from rats as well as newborn pigs but not of smooth muscle and astroglial cells; other prostanoids did not cause cell death. The peroxidation product 8-iso-PGF2, which is generated in OIR, stimulated TxA2 formation by endothelial cells and triggered cell death; these effects were markedly diminished by CGS-12970. TxA2-dependent neuroretinovascular endothelial cell death was mostly by necrosis and to a lesser extent by apoptosis. The data identify an important role for TxA2 in vasoobliteration of OIR and unveil a so far unknown function for TxA2 in directly triggering neuroretinal microvascular endothelial cell death. These effects of TxA2 might participate in other ischemic neurovascular injuries.


Asunto(s)
Oxígeno/toxicidad , Enfermedades de la Retina/fisiopatología , Vasos Retinianos/fisiología , Tromboxano A2/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Animales Recién Nacidos , Capilares/patología , Capilares/fisiopatología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fragmentación del ADN/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Ratas , Ratas Sprague-Dawley , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Vasos Retinianos/efectos de los fármacos , Sales de Tetrazolio , Tiazoles
18.
Semin Perinatol ; 25(2): 70-5, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11339668

RESUMEN

We compared the total density and the relative expression of EP receptor (EP) subtypes in ductus arteriosus (DA) of the newborn with that of the fetal piglet. Saturation binding experiments showed 3-fold less PGE2 receptors in the newborn than in the fetus because of loss of EP3 and EP4 receptors thus explaining, at least partly, the reduced responsiveness to PGE2 of the newborn DA. Displacement experiments showed that the relative proportions of EP2, EP3, and EP4 were similar in the fetal DA but only EP2 was detected in the DA of the newborn pig. Hence, PGE2 effects in the newborn DA seem to be exclusively mediated by EP2 receptors both in vitro and in vivo. These findings may help to propose more specific therapies for regulation of DA's tone in certain newborns for whom conventional therapy is contraindicated.


Asunto(s)
Animales Recién Nacidos/metabolismo , Conducto Arterial/química , Conducto Arterial/fisiología , Feto/metabolismo , Receptores de Prostaglandina E/fisiología , Animales , AMP Cíclico/biosíntesis , Dinoprostona/metabolismo , Dinoprostona/farmacología , Conducto Arterial/efectos de los fármacos , Receptores de Prostaglandina E/análisis , Receptores de Prostaglandina E/efectos de los fármacos , Porcinos , Tritio
19.
Circulation ; 103(13): 1806-12, 2001 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-11282914

RESUMEN

BACKGROUND: The ductus arteriosus (DA) of newborn infants exposed in utero to indomethacin is resistant to postnatal indomethacin; we hypothesized that this is due to ductus constriction in utero, with subsequent remodeling of the vessel. METHODS AND RESULTS: Infusion of fetal lambs with indomethacin for 48 hours constricted the DA and increased the thickness of the avascular zone of the DA, which in turn induced the expression of vascular endothelial growth factor, endothelial nitric oxide synthase (due to ingrowth of vasa vasorum), neointima formation, and loss of smooth muscle cells; moderate degrees of DA constriction in utero increased NO production, which inhibited DA contractility. Marked degrees of DA constriction decreased tissue distensibility and contractile capacity. CONCLUSIONS: DA patency is no longer controlled primarily by prostaglandins once it has been exposed to indomethacin in utero.


Asunto(s)
Conducto Arterial/anomalías , Conducto Arterial/efectos de los fármacos , Feto/anomalías , Indometacina/farmacología , Ovinos/embriología , 6-Cetoprostaglandina F1 alfa/biosíntesis , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Circulación Coronaria/efectos de los fármacos , Dinoprostona/biosíntesis , Dinoprostona/farmacología , Conducto Arterial/embriología , Conducto Arterial/metabolismo , Conducto Arterioso Permeable/inducido químicamente , Conducto Arterioso Permeable/enzimología , Conducto Arterioso Permeable/metabolismo , Conducto Arterioso Permeable/patología , Factores de Crecimiento Endotelial/biosíntesis , Feto/irrigación sanguínea , Feto/efectos de los fármacos , Feto/enzimología , Técnicas In Vitro , Linfocinas/biosíntesis , Modelos Biológicos , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Nitroprusiato/farmacología , Presión , Ovinos/anomalías , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
Am J Physiol Heart Circ Physiol ; 280(5): H2342-9, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11299240

RESUMEN

Although the role of PGE2 in maintaining ductus arteriosus (DA) patency is well established, the specific PGE2 receptor subtype(s) (EP) involved have not been clearly identified. We used late gestation fetal and neonatal lambs to study developmental regulation of EP receptors. In the fetal DA, radioligand binding and RT-PCR assays virtually failed to detect EP1 but detected EP2, EP3D, and EP4 receptors in equivalent proportions. In the newborn lamb, DA total density was one-third of that found in the fetus and only EP2 was detected. Stimulation of EP2 and EP4 increased cAMP formation and was associated with DA relaxation. Though stimulation of EP3 inhibited cAMP formation, it surprisingly relaxed the fetal DA both in vitro and in vivo. This EP3-induced relaxation was specifically diminished by the ATP-sensitive K(+) (K(ATP)) channel blocker glibenclamide. In conclusion, PGE2 dilates the late gestation fetal DA through pathways that involve either cAMP (EP2 and EP4) or K(ATP) channels (EP3). The loss of EP3 and EP4 receptors in the newborn DA is consistent with its decreased responsiveness to PGE2.


Asunto(s)
Alprostadil/análogos & derivados , Conducto Arterial/metabolismo , Receptores de Prostaglandina E/metabolismo , Xantonas , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Adenosina Trifosfato/metabolismo , Alprostadil/farmacología , Animales , Animales Recién Nacidos , Antiarrítmicos/farmacología , Antiulcerosos/farmacología , Unión Competitiva , Compuestos de Bifenilo/farmacología , Colforsina/farmacología , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Dinoprost/farmacología , Femenino , Feto/química , Feto/metabolismo , Reacción en Cadena de la Polimerasa , Canales de Potasio/metabolismo , Embarazo , Antagonistas de Prostaglandina/farmacología , Prostaglandinas E Sintéticas/farmacología , Ensayo de Unión Radioligante , Receptores de Prostaglandina E/análisis , Receptores de Prostaglandina E/genética , Ovinos , Tritio , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Xantenos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA