Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 311: 1-14, 2019 01.
Article in English | MEDLINE | ID: mdl-30217406

ABSTRACT

Inflammation contributes to neonatal brain injury. Pro-inflammatory cytokines represent key inflammatory meditators in neonatal hypoxic-ischemic (HI) brain injury. The high mobility group box-1 (HMGB1) protein is a nuclear protein with pro-inflammatory cytokine properties when it is translocated from the nucleus and released extracellularly after stroke in adult rodents. We have previously shown that HMGB1 is translocated from the nucleus to cytosolic compartment after ischemic brain injury in fetal sheep. In the current study, we utilized the Rice-Vannucci model to investigate the time course of HMGB1 translocation and release after HI injury in neonatal rats. HMGB1 was located in cellular nuclei of brains from sham control rats. Nuclear to cytoplasmic translocation of HMGB1 was detected in the ipsilateral-HI hemisphere as early as zero h after HI, and released extracellularly as early as 6 h after HI. Immunohistochemical double staining detected HMGB1 translocation mainly in neurons along with release from apoptotic cells after HI. Serum HMGB1 increased at 3 h and decreased by 24 h after HI. In addition, rat brains exposed to hypoxic injury alone also exhibited time dependent HMGB1 translocation at 3, 12 and 48 h after hypoxia. Consequently, HMGB1 responds similarly after HI injury in the brains of neonatal and adult subjects. We conclude that HMGB1 is sensitive early indicator of neonatal HI and hypoxic brain injury.


Subject(s)
Brain/metabolism , HMGB1 Protein/metabolism , Hypoxia-Ischemia, Brain/metabolism , Neurons/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/pathology , Female , HMGB1 Protein/analysis , Hypoxia-Ischemia, Brain/pathology , Neurons/chemistry , Pregnancy , Rats , Rats, Wistar
2.
Diabetes ; 67(5): 974-985, 2018 05.
Article in English | MEDLINE | ID: mdl-29487115

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Retinopathy/metabolism , Fibroblast Growth Factors/pharmacology , Photoreceptor Cells, Vertebrate/drug effects , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/pathology , Diabetic Retinopathy/etiology , Diabetic Retinopathy/pathology , Disease Models, Animal , Electroretinography , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Retinal Neurons/drug effects , Retinal Neurons/metabolism , Retinal Neurons/pathology , Tomography, Optical Coherence , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
3.
EMBO Mol Med ; 10(1): 76-90, 2018 01.
Article in English | MEDLINE | ID: mdl-29180355

ABSTRACT

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.


Subject(s)
Adiponectin/deficiency , Glucose/metabolism , Hyperglycemia/complications , Metabolism, Inborn Errors/complications , Photoreceptor Cells, Vertebrate/metabolism , Retinal Vessels/growth & development , Retinopathy of Prematurity/etiology , Adiponectin/metabolism , Animals , Cell Line , Female , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Infant, Newborn , Infant, Premature , Male , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/pathology , Retinal Neovascularization , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology
4.
Invest Ophthalmol Vis Sci ; 58(10): 3862-3870, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28763559

ABSTRACT

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.


Subject(s)
Adiponectin/physiology , Choroidal Neovascularization/prevention & control , Fatty Acids, Omega-3/pharmacology , Macular Degeneration/complications , Animals , Biomarkers/metabolism , Blotting, Western , Cell Proliferation/drug effects , Choroidal Neovascularization/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Matrix Metalloproteinases/metabolism , Mice , Receptors, Adiponectin/metabolism
5.
FASEB J ; 31(10): 4492-4502, 2017 10.
Article in English | MEDLINE | ID: mdl-28646017

ABSTRACT

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.


Subject(s)
Glycoproteins/genetics , Membrane Proteins/genetics , Neovascularization, Pathologic/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cytoskeletal Proteins , Endothelial Cells/metabolism , Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice, Transgenic , Neovascularization, Pathologic/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Retinal Ganglion Cells , Retinal Neovascularization/genetics , Semaphorins
6.
J Exp Med ; 214(6): 1753-1767, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28465464

ABSTRACT

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.


Subject(s)
Inflammation/metabolism , Inflammation/pathology , Photoreceptor Cells, Vertebrate/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Signal Transduction , Animals , Dependovirus/metabolism , Interleukin-6/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , RNA, Small Interfering/metabolism , Receptors, LDL/deficiency , Receptors, LDL/metabolism , Retina/drug effects , Retina/pathology , Retina/physiopathology , Retinoids/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
7.
EBioMedicine ; 18: 281-287, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28373097

ABSTRACT

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.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Retinal Neovascularization/prevention & control , Animals , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Disease Models, Animal , Fluorescein Angiography , Genetic Vectors/genetics , Genetic Vectors/metabolism , Lasers , Macular Degeneration/diagnosis , Macular Degeneration/metabolism , Macular Degeneration/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Nerve Tissue Proteins/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Retina/metabolism , Retina/pathology , Retinal Neovascularization/pathology
8.
Cell Rep ; 18(7): 1606-1613, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28199833

ABSTRACT

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.


Subject(s)
Choroidal Neovascularization/drug therapy , Fibroblast Growth Factors/pharmacology , Neovascularization, Pathologic/drug therapy , Retina/drug effects , Retinal Neovascularization/drug therapy , Animals , Choroidal Neovascularization/metabolism , Disease Models, Animal , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Retina/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
9.
Clin Exp Ophthalmol ; 45(5): 529-538, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28002872

ABSTRACT

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.


Subject(s)
Lutein/pharmacology , Retinal Neovascularization/prevention & control , Retinal Vessels/drug effects , Retinopathy of Prematurity/drug therapy , Animals , Animals, Newborn , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Neuroglia/drug effects , Retinal Neovascularization/pathology , Retinal Vessels/pathology , Retinopathy of Prematurity/pathology
10.
EBioMedicine ; 13: 201-211, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27720395

ABSTRACT

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.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Fenofibrate/pharmacology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Animals , Choroidal Neovascularization/drug therapy , Disease Models, Animal , Endothelial Cells/metabolism , Fatty Acids, Omega-3/metabolism , Humans , Mice , Mice, Transgenic , PPAR alpha/metabolism , Retinal Diseases/drug therapy , Retinal Diseases/etiology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Neovascularization/drug therapy , Signal Transduction
11.
Arterioscler Thromb Vasc Biol ; 36(9): 1919-27, 2016 09.
Article in English | MEDLINE | ID: mdl-27417579

ABSTRACT

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.


Subject(s)
Acetates/pharmacology , Angiogenesis Inhibitors/pharmacology , Choroidal Neovascularization/prevention & control , Cytochrome P-450 CYP2C8 Inhibitors/pharmacology , Cytochrome P-450 CYP2C8/metabolism , Fatty Acids, Omega-3/pharmacology , Quinolines/pharmacology , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/prevention & control , Animals , Aorta/drug effects , Aorta/enzymology , Cells, Cultured , Choroidal Neovascularization/enzymology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/physiopathology , Cyclopropanes , Cytochrome P-450 CYP2C8/genetics , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Fatty Acids, Omega-3/metabolism , Genotype , Humans , Hyperoxia/complications , Lasers , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Physiologic/drug effects , Phenotype , Retinal Neovascularization/enzymology , Retinal Neovascularization/genetics , Retinal Neovascularization/physiopathology , Retinopathy of Prematurity/enzymology , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/physiopathology , Sulfides , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...