Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Am J Ophthalmol ; 158(5): 1071-78, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25089351

RESUMEN

PURPOSE: To determine the influence of omega-3 supplementation on vitreous vascular endothelial growth factor A (VEGF-A) levels in patients with exudative age-related macular degeneration (wet AMD) receiving intravitreal anti-VEGF treatment. DESIGN: Prospective, randomized, open-label, single-center, clinical trial, consecutive interventional case series. METHODS: The study included 3 cohorts with wet AMD and a control group with epiretinal membrane or macular hole. Twenty wet AMD patients being treated with anti-VEGF were randomized to daily supplementation of antioxidants, zinc, and carotenoids with omega-3 fatty acids (docosahexaenoic acid and eicosapentaenoic acid; group 1, n = 10) or without omega-3 fatty acids (group 2, n = 10). They were compared with an anti-VEGF treatment-naïve wet AMD group (group 3, n = 10) and an epiretinal membrane or macular hole group (group 4, n = 10). Primary outcome was vitreal VEGF-A levels (at the time of anti-VEGF injection). Secondary outcomes were plasma VEGF-A and central foveal thickness. Patients with new submacular hemorrhage or any other treatment within 3 months were excluded. Final analyses included 9, 6, 7, and 8 patients in groups 1 through 4, respectively. RESULTS: Patients receiving omega-3s (group 1) had significantly lower levels of vitreal VEGF-A (141.11 ± 61.89 pg/mL) when compared with group 2 (626.09 ± 279.27 pg/mL; P = .036) and group 3 (735.48 ± 216.43 pg/mL; P = .013), but similar levels to group 4 (235.81 ± 33.99 pg/mL; P = .215). All groups showed similar values for plasma VEGF-A and central foveal thickness measurements. CONCLUSIONS: This study demonstrated that omega-3 supplementation combined with anti-VEGF treatment is associated with decreased vitreal VEGF-A levels in wet AMD patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Suplementos Dietéticos , Ácidos Grasos Omega-3/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cuerpo Vítreo/metabolismo , Degeneración Macular Húmeda/terapia , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/administración & dosificación , Bevacizumab , Relación Dosis-Respuesta a Droga , Femenino , Angiografía con Fluoresceína , Estudios de Seguimiento , Fondo de Ojo , Humanos , Inyecciones Intravítreas , Masculino , Estudios Prospectivos , Tomografía de Coherencia Óptica , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Agudeza Visual , Degeneración Macular Húmeda/diagnóstico , Degeneración Macular Húmeda/etiología
2.
Arterioscler Thromb Vasc Biol ; 34(3): 581-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24458713

RESUMEN

OBJECTIVE: Regulation of angiogenesis is critical for many diseases. Specifically, pathological retinal neovascularization, a major cause of blindness, is suppressed with dietary ω3-long-chain polyunsaturated fatty acids (ω3LCPUFAs) through antiangiogenic metabolites of cyclooxygenase and lipoxygenase. Cytochrome P450 epoxygenases (CYP2C8) also metabolize LCPUFAs, producing bioactive epoxides, which are inactivated by soluble epoxide hydrolase (sEH) to transdihydrodiols. The effect of these enzymes and their metabolites on neovascularization is unknown. APPROACH AND RESULTS: The mouse model of oxygen-induced retinopathy was used to investigate retinal neovascularization. We found that CYP2C (localized in wild-type monocytes/macrophages) is upregulated in oxygen-induced retinopathy, whereas sEH is suppressed, resulting in an increased retinal epoxide:diol ratio. With a ω3LCPUFA-enriched diet, retinal neovascularization increases in Tie2-driven human-CYP2C8-overexpressing mice (Tie2-CYP2C8-Tg), associated with increased plasma 19,20-epoxydocosapentaenoic acid and retinal epoxide:diol ratio. 19,20-Epoxydocosapentaenoic acids and the epoxide:diol ratio are decreased with overexpression of sEH (Tie2-sEH-Tg). Overexpression of CYP2C8 or sEH in mice does not change normal retinal vascular development compared with their wild-type littermate controls. The proangiogenic role in retina of CYP2C8 with both ω3LCPUFA and ω6LCPUFA and antiangiogenic role of sEH in ω3LCPUFA metabolism were corroborated in aortic ring assays. CONCLUSIONS: Our results suggest that CYP2C ω3LCPUFA metabolites promote retinal pathological angiogenesis. CYP2C8 is part of a novel lipid metabolic pathway influencing retinal neovascularization.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Ácidos Grasos Omega-3/toxicidad , Macrófagos/enzimología , Monocitos/enzimología , Neovascularización Retiniana/inducido químicamente , Animales , Ácido Araquidónico/metabolismo , Hidrocarburo de Aril Hidroxilasas/genética , Biotransformación , Hipoxia de la Célula , Citocromo P-450 CYP2C8 , Grasas de la Dieta/farmacocinética , Ácidos Docosahexaenoicos/metabolismo , Ácido Eicosapentaenoico/metabolismo , Epóxido Hidrolasas/deficiencia , Epóxido Hidrolasas/genética , Epóxido Hidrolasas/fisiología , Proteínas del Ojo/metabolismo , Ácidos Grasos Omega-3/administración & dosificación , Ácidos Grasos Omega-3/clasificación , Ácidos Grasos Omega-3/farmacocinética , Ácidos Grasos Insaturados/administración & dosificación , Ácidos Grasos Insaturados/farmacocinética , Humanos , Lipooxigenasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxígeno/toxicidad , Prostaglandina-Endoperóxido Sintasas/metabolismo , ARN Mensajero/biosíntesis , Receptor TIE-2/genética , Proteínas Recombinantes de Fusión/metabolismo , Neovascularización Retiniana/prevención & control
3.
PLoS One ; 8(1): e53155, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23335958

RESUMEN

BACKGROUND: Increased intake of ω-3 long-chain polyunsaturated fatty acids (LCPUFAs) and use of peroxisome proliferator activator receptor (PPAR)-activating drugs are associated with attenuation of pathologic retinal angiogenesis. ω-3 LCPUFAs are endogenous agonists of PPARs. We postulated that DNA sequence variation in PPAR gamma (PPARG) co-activator 1 alpha (PPARGC1A), a gene encoding a co-activator of the LCPUFA-sensing PPARG-retinoid X receptor (RXR) transcription complex, may influence neovascularization (NV) in age-related macular degeneration (AMD). METHODS: We applied exact testing methods to examine distributions of DNA sequence variants in PPARGC1A for association with NV AMD and interaction of AMD-associated loci in genes of complement, lipid metabolism, and VEGF signaling systems. Our sample contained 1858 people from 3 elderly cohorts of western European ancestry. We concurrently investigated retinal gene expression profiles in 17-day-old neonatal mice on a 2% LCPUFA feeding paradigm to identify LCPUFA-regulated genes both associated with pathologic retinal angiogenesis and known to interact with PPARs or PPARGC1A. RESULTS: A DNA coding variant (rs3736265) and a 3'UTR-resident regulatory variant (rs3774923) in PPARGC1A were independently associated with NV AMD (exact P = 0.003, both SNPs). SNP-SNP interactions existed for NV AMD (P<0.005) with rs3736265 and a AMD-associated variant in complement factor B (CFB, rs512559). PPARGC1A influences activation of the AMD-associated complement component 3 (C3) promoter fragment and CFB influences activation and proteolysis of C3. We observed interaction (P ≤ 0.003) of rs3736265 with a variant in vascular endothelial growth factor A (VEGFA, rs3025033), a key molecule in retinal angiogenesis. Another PPARGC1A coding variant (rs8192678) showed statistical interaction with a SNP in the VEGFA receptor fms-related tyrosine kinase 1 (FLT1, rs10507386; P ≤ 0.003). C3 expression was down-regulated 2-fold in retinas of ω-3 LCPUFA-fed mice - these animals also showed 70% reduction in retinal NV (P ≤ 0.001). CONCLUSION: Ligands and co-activators of the ω-3 LCPUFA sensing PPAR-RXR axis may influence retinal angiogenesis in NV AMD via the complement and VEGF signaling systems. We have linked the co-activator of a lipid-sensing transcription factor (PPARG co-activator 1 alpha, PPARGC1A) to age-related macular degeneration (AMD) and AMD-associated genes.


Asunto(s)
Proteínas del Sistema Complemento/genética , Proteínas de Choque Térmico/genética , Degeneración Macular/genética , Degeneración Macular/metabolismo , Neovascularización Patológica/genética , Transducción de Señal , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Estudios de Cohortes , Ácidos Grasos Omega-3/metabolismo , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Proteínas de Choque Térmico/química , Humanos , Degeneración Macular/patología , Masculino , Ratones , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Polimorfismo de Nucleótido Simple , Retina/metabolismo , Retina/patología , Factores de Transcripción/química , Factor A de Crecimiento Endotelial Vascular/genética
4.
Neonatology ; 100(4): 343-53, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21968165

RESUMEN

Retinopathy of prematurity (ROP), an ocular disease characterized by the onset of vascular abnormalities in the developing retina, is the major cause of visual impairment and blindness in premature neonates. ROP is a complex condition in which various factors participate at different stages of the disease leading to microvascular degeneration followed by neovascularization, which in turn predisposes to retinal detachment. Current ablative therapies (cryotherapy and laser photocoagulation) used in the clinic for the treatment of ROP have limitations and patients can still have long-term effects even after successful treatment. New treatment modalities are still emerging. The most promising are the therapies directed against VEGF; more recently the use of preventive dietary supplementation with ω-3 polyunsaturated fatty acid may also be promising. Other than pharmacologic and nutritional approaches, cell-based strategies for vascular repair are likely to arise from advances in regenerative medicine using stem cells. In addition to all of these, a greater understanding of other factors involved in regulating pathologic retinal angiogenesis continues to emerge, suggesting potential targets for therapeutic approaches. This review summarizes an update on the current state of knowledge on ROP from our and other laboratories, with particular focus on the role of nitro-oxidative stress and notably trans-arachidonic acids in microvascular degeneration, semaphorin 3 operating as vasorepulsive molecules in the avascular hypoxic retina and in turn impairing revascularization, succinate and its receptor GPR91 in neuron-mediated retinal neovascularization, and ω-3 lipids as modulators of preretinal neovascularization.


Asunto(s)
Recien Nacido Prematuro , Retinopatía de la Prematuridad/etiología , Edad Gestacional , Humanos , Recién Nacido , Peroxidación de Lípido , Neovascularización Patológica , Estrés Oxidativo , Oxígeno/fisiología , Oxígeno/uso terapéutico , Receptores Acoplados a Proteínas G , Retina/embriología , Vasos Retinianos/embriología , Retinopatía de la Prematuridad/prevención & control , Retinopatía de la Prematuridad/terapia , Factores de Riesgo , Semaforinas , Ácido Succínico , Factor A de Crecimiento Endotelial Vascular
5.
Sci Transl Med ; 3(69): 69ra12, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21307302

RESUMEN

Lipid signaling is dysregulated in many diseases with vascular pathology, including cancer, diabetic retinopathy, retinopathy of prematurity, and age-related macular degeneration. We have previously demonstrated that diets enriched in ω-3 polyunsaturated fatty acids (PUFAs) effectively reduce pathological retinal neovascularization in a mouse model of oxygen-induced retinopathy, in part through metabolic products that suppress microglial-derived tumor necrosis factor-α. To better understand the protective effects of ω-3 PUFAs, we examined the relative importance of major lipid metabolic pathways and their products in contributing to this effect. ω-3 PUFA diets were fed to four lines of mice deficient in each key lipid-processing enzyme (cyclooxygenase 1 or 2, or lipoxygenase 5 or 12/15), retinopathy was induced by oxygen exposure; only loss of 5-lipoxygenase (5-LOX) abrogated the protection against retinopathy of dietary ω-3 PUFAs. This protective effect was due to 5-LOX oxidation of the ω-3 PUFA lipid docosahexaenoic acid to 4-hydroxy-docosahexaenoic acid (4-HDHA). 4-HDHA directly inhibited endothelial cell proliferation and sprouting angiogenesis via peroxisome proliferator-activated receptor γ (PPARγ), independent of 4-HDHA's anti-inflammatory effects. Our study suggests that ω-3 PUFAs may be profitably used as an alternative or supplement to current anti-vascular endothelial growth factor (VEGF) treatment for proliferative retinopathy and points to the therapeutic potential of ω-3 PUFAs and metabolites in other diseases of vasoproliferation. It also suggests that cyclooxygenase inhibitors such as aspirin and ibuprofen (but not lipoxygenase inhibitors such as zileuton) might be used without losing the beneficial effect of dietary ω-3 PUFA.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Araquidonato 5-Lipooxigenasa/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Ácidos Grasos Omega-3/uso terapéutico , Animales , Araquidonato 5-Lipooxigenasa/genética , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Ácidos Grasos Omega-6/uso terapéutico , Humanos , Inmunohistoquímica , Masculino , Ratones , Oxígeno/toxicidad , PPAR gamma/metabolismo , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Circ Res ; 107(4): 495-500, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20634487

RESUMEN

RATIONALE: Omega3 long-chain polyunsaturated fatty acids (omega3-PUFAs) are powerful modulators of angiogenesis. However, little is known about the mechanisms governing omega3-PUFA-dependent attenuation of angiogenesis. OBJECTIVE: This study aims to identify a major mechanism by which omega3-PUFAs attenuate retinal neovascularization. METHODS AND RESULTS: Administering omega3-PUFAs exclusively during the neovascular stage of the mouse model of oxygen-induced retinopathy induces a direct neovascularization reduction of more than 40% without altering vasoobliteration or the regrowth of normal vessels. Cotreatment with an inhibitor of peroxisome proliferator-activated receptor (PPAR)gamma almost completely abrogates this effect. Inhibition of PPARgamma also reverses the omega3-PUFA-induced reduction of retinal tumor necrosis factor-alpha, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, endothelial selectin, and angiopoietin 2 but not vascular endothelial growth factor. CONCLUSIONS: These results identify a direct, PPARgamma-mediated effect of omega3-PUFAs on retinal neovascularization formation and retinal angiogenic activation that is independent of vascular endothelial growth factor.


Asunto(s)
Inhibidores de la Angiogénesis/fisiología , Ácidos Grasos Omega-3/administración & dosificación , Neovascularización Patológica/metabolismo , PPAR gamma/fisiología , Enfermedades de la Retina/metabolismo , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/dietoterapia , Neovascularización Patológica/prevención & control , Enfermedades de la Retina/dietoterapia , Enfermedades de la Retina/prevención & control , Factor A de Crecimiento Endotelial Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA