Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
JAMA Netw Open ; 2(4): e193193, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-31026037

RESUMO

Importance: Adverse childhood experiences are common and are associated with changes in early development and learning, but training early childhood educators in trauma-informed approaches to care has not been evaluated with randomized clinical trials. Objective: To determine whether a 6-session (12-week) professional development course, "Enhancing Trauma Awareness," improved the quality of teachers' relationships with the children in their classrooms. Design, Setting, and Participants: This cluster randomized clinical trial conducted from September 2017 to May 2018 allocated classrooms by a computer-generated random sequence to intervention (attend Enhancing Trauma Awareness course) and control (no course) groups. Outcomes were reported by participants via survey and analyzed by group allocation. Classrooms under the auspice of the School District of Philadelphia, Pennsylvania, serving 3- and 4-year-old children living in low-income households were invited to participate. Lead and/or assistant teachers from 63 of 348 eligible classrooms (18.1%) agreed to participate, and none were excluded. Of 96 enrolled teachers, 93 (96.9%) were assessed at follow-up (61 of 63 classrooms [96.8%]). Exposures: In September 2017, 32 classrooms (48 teachers) were assigned to receive a professional development course that taught about the effects of trauma using a group-based relational process, and 31 classrooms (48 teachers) received no intervention. Teachers completed online surveys immediately before and after the course. Exploratory focus groups with intervention teachers (n = 15) were conducted 5 months after the course ended. Main Outcomes and Measures: The primary outcome was teacher-children relationship quality, with a hypothesized decrease in teacher-children conflict scores. Secondary outcomes included relational capacities (eg, empathy, emotion regulation, and dispositional mindfulness). Focus group themes described teachers' experience of the course. Results: Of 96 teachers enrolled, 93 (96.9%) were women, and 58 (60.4%) were 40 years and older. Follow-up surveys were completed by 46 teachers (95.8%) in the control group and 47 (97.9%) in the intervention group, of whom 38 (79.2%) attended 4 or more course sessions. Adjusting for baseline values, mean (SE) conflict scores after the course were not significantly different between course participants (15.8 [0.6]) and controls (15.0 [0.6]) (effect size = 0.16; 95% CI, -0.19 to 0.52). There were no significant between-group differences in secondary outcomes. However, in focus groups, the teachers reported improvements in teacher-children relationship quality and several related relational capacities. Conclusions and Relevance: A course to enhance trauma awareness among preschool teachers did not reduce teacher-children conflict scores, yet qualitative assessments suggested the potential for improved teacher-children relationship quality. Trial Registration: ClinicalTrials.gov identifier: NCT03303482.


Assuntos
Experiências Adversas da Infância , Intervenção Educacional Precoce/métodos , Professores Escolares/psicologia , Estudantes/psicologia , Capacitação de Professores/métodos , Adulto , Pré-Escolar , Análise por Conglomerados , Feminino , Humanos , Relações Interpessoais , Masculino , Pobreza/psicologia , População Urbana
2.
Diabetes ; 67(5): 974-985, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29487115

RESUMO

Retinal neuronal abnormalities occur before vascular changes in diabetic retinopathy. Accumulating experimental evidence suggests that neurons control vascular pathology in diabetic and other neovascular retinal diseases. Therefore, normalizing neuronal activity in diabetes may prevent vascular pathology. We investigated whether fibroblast growth factor 21 (FGF21) prevented retinal neuronal dysfunction in insulin-deficient diabetic mice. We found that in diabetic neural retina, photoreceptor rather than inner retinal function was most affected and administration of the long-acting FGF21 analog PF-05231023 restored the retinal neuronal functional deficits detected by electroretinography. PF-05231023 administration protected against diabetes-induced disorganization of photoreceptor segments seen in retinal cross section with immunohistochemistry and attenuated the reduction in the thickness of photoreceptor segments measured by optical coherence tomography. PF-05231023, independent of its downstream metabolic modulator adiponectin, reduced inflammatory marker interleukin-1ß (IL-1ß) mRNA levels. PF-05231023 activated the AKT-nuclear factor erythroid 2-related factor 2 pathway and reduced IL-1ß expression in stressed photoreceptors. PF-05231023 administration did not change retinal expression of vascular endothelial growth factor A, suggesting a novel therapeutic approach for the prevention of early diabetic retinopathy by protecting photoreceptor function in diabetes.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Retinopatia Diabética/metabolismo , Fatores de Crescimento de Fibroblastos/farmacologia , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Animais , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/patologia , Retinopatia Diabética/etiologia , Retinopatia Diabética/patologia , Modelos Animais de Doenças , Eletrorretinografia , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Camundongos , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neurônios Retinianos/efeitos dos fármacos , Neurônios Retinianos/metabolismo , Neurônios Retinianos/patologia , Tomografia de Coerência Óptica , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
EMBO Mol Med ; 10(1): 76-90, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29180355

RESUMO

The neural cells and factors determining normal vascular growth are not well defined even though vision-threatening neovessel growth, a major cause of blindness in retinopathy of prematurity (ROP) (and diabetic retinopathy), is driven by delayed normal vascular growth. We here examined whether hyperglycemia and low adiponectin (APN) levels delayed normal retinal vascularization, driven primarily by dysregulated photoreceptor metabolism. In premature infants, low APN levels correlated with hyperglycemia and delayed retinal vascular formation. Experimentally in a neonatal mouse model of postnatal hyperglycemia modeling early ROP, hyperglycemia caused photoreceptor dysfunction and delayed neurovascular maturation associated with changes in the APN pathway; recombinant mouse APN or APN receptor agonist AdipoRon treatment normalized vascular growth. APN deficiency decreased retinal mitochondrial metabolic enzyme levels particularly in photoreceptors, suppressed retinal vascular development, and decreased photoreceptor platelet-derived growth factor (Pdgfb). APN pathway activation reversed these effects. Blockade of mitochondrial respiration abolished AdipoRon-induced Pdgfb increase in photoreceptors. Photoreceptor knockdown of Pdgfb delayed retinal vascular formation. Stimulation of the APN pathway might prevent hyperglycemia-associated retinal abnormalities and suppress phase I ROP in premature infants.


Assuntos
Adiponectina/deficiência , Glucose/metabolismo , Hiperglicemia/complicações , Erros Inatos do Metabolismo/complicações , Células Fotorreceptoras de Vertebrados/metabolismo , Vasos Retinianos/crescimento & desenvolvimento , Retinopatia da Prematuridade/etiologia , Adiponectina/metabolismo , Animais , Linhagem Celular , Feminino , Humanos , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Camundongos Endogâmicos C57BL , Células Fotorreceptoras de Vertebrados/patologia , Neovascularização Retiniana , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/patologia
4.
Invest Ophthalmol Vis Sci ; 58(10): 3862-3870, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28763559

RESUMO

Purpose: Neovascular age-related macular degeneration (AMD) is a major cause of legal blindness in the elderly. Diets with omega3-long-chain-polyunsaturated-fatty-acid (ω3-LCPUFA) correlate with a decreased risk of AMD. Dietary ω3-LCPUFA versus ω6-LCPUFA inhibits mouse ocular neovascularization, but the underlying mechanism needs further exploration. The aim of this study was to investigate if adiponectin (APN) mediated ω3-LCPUFA suppression of neovessels in AMD. Methods: The mouse laser-induced choroidal neovascularization (CNV) model was used to mimic some of the inflammatory aspect of AMD. CNV was compared between wild-type (WT) and Apn-/- mice fed either otherwise matched diets with 2% ω3 or 2% ω6-LCPUFAs. Vldlr-/- mice were used to mimic some of the metabolic aspects of AMD. Choroid assay ex vivo and human retinal microvascular endothelial cell (HRMEC) proliferation assay in vitro was used to investigate the APN pathway in angiogenesis. Western blot for p-AMPKα/AMPKα and qPCR for Apn, Mmps, and IL-10 were used to define mechanism. Results: ω3-LCPUFA intake suppressed laser-induced CNV in WT mice; suppression was abolished with APN deficiency. ω3-LCPUFA, mediated by APN, decreased mouse Mmps expression. APN deficiency decreased AMPKα phosphorylation in vivo and exacerbated choroid-sprouting ex vivo. APN pathway activation inhibited HRMEC proliferation and decreased Mmps. In Vldlr-/- mice, ω3-LCPUFA increased retinal AdipoR1 and inhibited NV. ω3-LCPUFA decreased IL-10 but did not affect Mmps in Vldlr-/- retinas. Conclusions: APN in part mediated ω3-LCPUFA inhibition of neovascularization in two mouse models of AMD. Modulating the APN pathway in conjunction with a ω3-LCPUFA-enriched-diet may augment the beneficial effects of ω3-LCPUFA in AMD patients.


Assuntos
Adiponectina/fisiologia , Neovascularização de Coroide/prevenção & controle , Ácidos Graxos Ômega-3/farmacologia , Degeneração Macular/complicações , Animais , Biomarcadores/metabolismo , Western Blotting , Proliferação de Células/efeitos dos fármacos , Neovascularização de Coroide/metabolismo , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Metaloproteinases da Matriz/metabolismo , Camundongos , Receptores de Adiponectina/metabolismo
5.
FASEB J ; 31(10): 4492-4502, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28646017

RESUMO

Pathological proliferation of retinal blood vessels commonly causes vision impairment in proliferative retinopathies, including retinopathy of prematurity. Dysregulated crosstalk between the vasculature and retinal neurons is increasingly recognized as a major factor contributing to the pathogenesis of vascular diseases. Class 3 semaphorins (SEMA3s), a group of neuron-secreted axonal and vascular guidance factors, suppress pathological vascular growth in retinopathy. However, the upstream transcriptional regulators that mediate the function of SEMA3s in vascular growth are poorly understood. Here we showed that retinoic acid receptor-related orphan receptor α (RORα), a nuclear receptor and transcription factor, is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in a mouse model of oxygen-induced proliferative retinopathy. We found that genetic deficiency of RORα substantially induced Sema3e expression in retinopathy. Both RORα and SEMA3E were expressed in retinal ganglion cells. RORα directly bound to a specific ROR response element on the promoter of Sema3e and negatively regulated Sema3e promoter-driven luciferase expression. Suppression of Sema3e using adeno-associated virus 2 carrying short hairpin RNA targeting Sema3e promoted disoriented pathological neovascularization and partially abolished the inhibitory vascular effects of RORα deficiency in retinopathy. Our findings suggest that RORα is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in pathological retinal angiogenesis.-Sun, Y., Liu, C.-H., Wang, Z., Meng, S. S., Burnim, S. B., SanGiovanni, J. P., Kamenecka, T. M., Solt, L. A., Chen, J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis.


Assuntos
Glicoproteínas/genética , Proteínas de Membrana/genética , Neovascularização Patológica/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Neovascularização Retiniana/metabolismo , Vasos Retinianos/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas do Citoesqueleto , Células Endoteliais/metabolismo , Glicoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Camundongos Transgênicos , Neovascularização Patológica/genética , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Células Ganglionares da Retina , Neovascularização Retiniana/genética , Semaforinas
6.
J Exp Med ; 214(6): 1753-1767, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28465464

RESUMO

Pathological neovessels growing into the normally avascular photoreceptors cause vision loss in many eye diseases, such as age-related macular degeneration and macular telangiectasia. Ocular neovascularization is strongly associated with inflammation, but the source of inflammatory signals and the mechanisms by which these signals regulate the disruption of avascular privilege in photoreceptors are unknown. In this study, we found that c-Fos, a master inflammatory regulator, was increased in photoreceptors in a model of pathological blood vessels invading photoreceptors: the very low-density lipoprotein receptor-deficient (Vldlr-/- ) mouse. Increased c-Fos induced inflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor (TNF), leading to activation of signal transducer and activator of transcription 3 (STAT3) and increased TNFα-induced protein 3 (TNFAIP3) in Vldlr-/- photoreceptors. IL-6 activated the STAT3/vascular endothelial growth factor A (VEGFA) pathway directly, and elevated TNFAIP3 suppressed SOCS3 (suppressor of cytokine signaling 3)-activated STAT3/VEGFA indirectly. Inhibition of c-Fos using photoreceptor-specific AAV (adeno-associated virus)-hRK (human rhodopsin kinase)-sh_c-fos or a chemical inhibitor substantially reduced the pathological neovascularization and rescued visual function in Vldlr-/- mice. These findings suggested that the photoreceptor c-Fos controls blood vessel growth into the normally avascular photoreceptor layer through the inflammatory signal-induced STAT3/VEGFA pathway.


Assuntos
Inflamação/metabolismo , Inflamação/patologia , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Transdução de Sinais , Animais , Dependovirus/metabolismo , Interleucina-6/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/patologia , RNA Interferente Pequeno/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/metabolismo , Retina/efeitos dos fármacos , Retina/patologia , Retina/fisiopatologia , Retinoides/farmacologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
EBioMedicine ; 18: 281-287, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28373097

RESUMO

Pathological neovascularization of the outer retina is the hallmark of neovascular age-related macular degeneration (nAMD). Building on our previous observations that semaphorin 3F (Sema3f) is expressed in the outer retina and demonstrates anti-angiogenic potential, we have investigated whether Sema3f can be used to protect against subretinal neovascularization in two mouse models. Both in the very low-density lipid-receptor knockout (Vldlr-/-) model of spontaneous subretinal neovascularization as well as in the mouse model of laser-induced choroidal neovascularization (CNV), we found protective effects of Sema3f against the formation of pathologic neovascularization. In the Vldlr-/- model, AAV-induced overexpression of Sema3f reduced the size of pathologic neovascularization by 56%. In the laser-induced CNV model, intravitreally injected Sema3f reduced pathologic neovascularization by 30%. Combined, these results provide the first evidence from two distinct in vivo models for a use of Sema3f in protecting the outer retina against subretinal neovascularization.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neovascularização Retiniana/prevenção & controle , Animais , Neovascularização de Coroide/etiologia , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Angiofluoresceinografia , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Lasers , Degeneração Macular/diagnóstico , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Receptores de LDL/deficiência , Receptores de LDL/genética , Retina/metabolismo , Retina/patologia , Neovascularização Retiniana/patologia
8.
Cell Rep ; 18(7): 1606-1613, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28199833

RESUMO

Pathological neovascularization, a leading cause of blindness, is seen in retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration. Using a mouse model of hypoxia-driven retinal neovascularization, we find that fibroblast growth factor 21 (FGF21) administration suppresses, and FGF21 deficiency worsens, retinal neovessel growth. The protective effect of FGF21 against neovessel growth was abolished in adiponectin (APN)-deficient mice. FGF21 administration also decreased neovascular lesions in two models of neovascular age-related macular degeneration: very-low-density lipoprotein-receptor-deficient mice with retinal angiomatous proliferation and laser-induced choroidal neovascularization. FGF21 inhibited tumor necrosis α (TNF-α) expression but did not alter Vegfa expression in neovascular eyes. These data suggest that FGF21 may be a therapeutic target for pathologic vessel growth in patients with neovascular eye diseases, including retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration.


Assuntos
Neovascularização de Coroide/tratamento farmacológico , Fatores de Crescimento de Fibroblastos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Retina/efeitos dos fármacos , Neovascularização Retiniana/tratamento farmacológico , Animais , Neovascularização de Coroide/metabolismo , Modelos Animais de Doenças , Degeneração Macular/tratamento farmacológico , Degeneração Macular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Retina/metabolismo , Neovascularização Retiniana/metabolismo , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Clin Exp Ophthalmol ; 45(5): 529-538, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28002872

RESUMO

BACKGROUND: Retinopathy of prematurity is one of the leading causes of childhood blindness worldwide, with vessel growth cessation and vessel loss in phase I followed by neovascularization in phase II. Ischaemia contributes to its pathogenesis, and lutein protects against ischaemia-induced retinal damages. We aimed to investigate the effects of lutein on a murine model of oxygen-induced retinopathy. METHODS: Mouse pups were exposed to 75% oxygen for 5 days and returned to room air for another 5 days. Vascular obliteration, neovascularization and blood vessel leakage were examined. Immunohistochemistry for glial cells and microglia were performed. RESULTS: Compared with vehicle controls, mouse pups receiving lutein treatment displayed smaller central vaso-obliterated area and reduced blood vessel leakage. No significant difference in neovascular area was found between lutein and vehicle controls. Lutein promoted endothelial tip cell formation and maintained the astrocytic template in the avascular area in oxygen-induced retinopathy. No significant changes in Müller cell gliosis and microglial activation in the central avascular area were found in lutein-treated pups. CONCLUSIONS: Our observations indicated that lutein significantly promoted normal retinal vascular regrowth in the central avascular area, possibly through promoting endothelial tip cell formation and preserving astrocytic template. Our results indicated that lutein might be considered as a supplement for the treatment of proliferative retinopathy of prematurity because of its role in facilitating the revascularization of normal vasculature.


Assuntos
Luteína/farmacologia , Neovascularização Retiniana/prevenção & controle , Vasos Retinianos/efeitos dos fármacos , Retinopatia da Prematuridade/tratamento farmacológico , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica , Neuroglia/efeitos dos fármacos , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Retinopatia da Prematuridade/patologia
10.
EBioMedicine ; 13: 201-211, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27720395

RESUMO

Neovascular eye diseases including retinopathy of prematurity, diabetic retinopathy and age-related-macular-degeneration are major causes of blindness. Fenofibrate treatment in type 2 diabetes patients reduces progression of diabetic retinopathy independent of its peroxisome proliferator-activated receptor (PPAR)α agonist lipid lowering effect. The mechanism is unknown. Fenofibrate binds to and inhibits cytochrome P450 epoxygenase (CYP)2C with higher affinity than to PPARα. CYP2C metabolizes ω-3 long-chain polyunsaturated fatty acids (LCPUFAs). While ω-3 LCPUFA products from other metabolizing pathways decrease retinal and choroidal neovascularization, CYP2C products of both ω-3 and ω-6 LCPUFAs promote angiogenesis. We hypothesized that fenofibrate inhibits retinopathy by reducing CYP2C ω-3 LCPUFA (and ω-6 LCPUFA) pro-angiogenic metabolites. Fenofibrate reduced retinal and choroidal neovascularization in PPARα-/-mice and augmented ω-3 LCPUFA protection via CYP2C inhibition. Fenofibrate suppressed retinal and choroidal neovascularization in mice overexpressing human CYP2C8 in endothelial cells and reduced plasma levels of the pro-angiogenic ω-3 LCPUFA CYP2C8 product, 19,20-epoxydocosapentaenoic acid. 19,20-epoxydocosapentaenoic acid reversed fenofibrate-induced suppression of angiogenesis ex vivo and suppression of endothelial cell functions in vitro. In summary fenofibrate suppressed retinal and choroidal neovascularization via CYP2C inhibition as well as by acting as an agonist of PPARα. Fenofibrate augmented the overall protective effects of ω-3 LCPUFAs on neovascular eye diseases.


Assuntos
Inibidores da Angiogênese/farmacologia , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Fenofibrato/farmacologia , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Animais , Neovascularização de Coroide/tratamento farmacológico , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , PPAR alfa/metabolismo , Doenças Retinianas/tratamento farmacológico , Doenças Retinianas/etiologia , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Neovascularização Retiniana/tratamento farmacológico , Transdução de Sinais
11.
Arterioscler Thromb Vasc Biol ; 36(9): 1919-27, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27417579

RESUMO

OBJECTIVE: Pathological ocular neovascularization is a major cause of blindness. Increased dietary intake of ω-3 long-chain polyunsaturated fatty acids (LCPUFA) reduces retinal neovascularization and choroidal neovascularization (CNV), but ω-3 LCPUFA metabolites of a major metabolizing pathway, cytochrome P450 oxidase (CYP) 2C, promote ocular pathological angiogenesis. We hypothesized that inhibition of CYP2C activity will add to the protective effects of ω-3 LCPUFA on neovascular eye diseases. APPROACH AND RESULTS: The mouse models of oxygen-induced retinopathy and laser-induced CNV were used to investigate pathological angiogenesis in the retina and choroid, respectively. The plasma levels of ω-3 LCPUFA metabolites of CYP2C were determined by mass spectroscopy. Aortic ring and choroidal explant sprouting assays were used to investigate the effects of CYP2C inhibition and ω-3 LCPUFA-derived CYP2C metabolic products on angiogenesis ex vivo. We found that inhibition of CYP2C activity by montelukast added to the protective effects of ω-3 LCPUFA on retinal neovascularization and CNV by 30% and 20%, respectively. In CYP2C8-overexpressing mice fed a ω-3 LCPUFA diet, montelukast suppressed retinal neovascularization and CNV by 36% and 39% and reduced the plasma levels of CYP2C8 products. Soluble epoxide hydrolase inhibition, which blocks breakdown and inactivation of CYP2C ω-3 LCPUFA-derived active metabolites, increased oxygen-induced retinopathy and CNV in vivo. Exposure to selected ω-3 LCPUFA metabolites of CYP2C significantly reversed the suppression of both angiogenesis ex vivo and endothelial cell functions in vitro by the CYP2C inhibitor montelukast. CONCLUSIONS: Inhibition of CYP2C activity adds to the protective effects of ω-3 LCPUFA on pathological retinal neovascularization and CNV.


Assuntos
Acetatos/farmacologia , Inibidores da Angiogênese/farmacologia , Neovascularização de Coroide/prevenção & controle , Inibidores do Citocromo P-450 CYP2C8/farmacologia , Citocromo P-450 CYP2C8/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Quinolinas/farmacologia , Neovascularização Retiniana/prevenção & controle , Retinopatia da Prematuridade/prevenção & controle , Animais , Aorta/efeitos dos fármacos , Aorta/enzimologia , Células Cultivadas , Neovascularização de Coroide/enzimologia , Neovascularização de Coroide/genética , Neovascularização de Coroide/fisiopatologia , Ciclopropanos , Citocromo P-450 CYP2C8/genética , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Ácidos Graxos Ômega-3/metabolismo , Genótipo , Humanos , Hiperóxia/complicações , Lasers , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Fisiológica/efeitos dos fármacos , Fenótipo , Neovascularização Retiniana/enzimologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/fisiopatologia , Retinopatia da Prematuridade/enzimologia , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/fisiopatologia , Sulfetos , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA