Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
PLoS One ; 10(7): e0132643, 2015.
Article in English | MEDLINE | ID: mdl-26161975

ABSTRACT

The mouse model of laser-induced choroidal neovascularization (CNV) has been used in studies of the exudative form of age-related macular degeneration using both the conventional slit lamp and a new image-guided laser system. A standardized protocol is needed for consistent results using this model, which has been lacking. We optimized details of laser-induced CNV using the image-guided laser photocoagulation system. Four lesions with similar size were consistently applied per eye at approximately double the disc diameter away from the optic nerve, using different laser power levels, and mice of various ages and genders. After 7 days, the mice were sacrificed and retinal pigment epithelium/choroid/sclera was flat-mounted, stained with Isolectin B4, and imaged. Quantification of the area of the laser-induced lesions was performed using an established and constant threshold. Exclusion criteria are described that were necessary for reliable data analysis of the laser-induced CNV lesions. The CNV lesion area was proportional to the laser power levels. Mice at 12-16 weeks of age developed more severe CNV than those at 6-8 weeks of age, and the gender difference was only significant in mice at 12-16 weeks of age, but not in those at 6-8 weeks of age. Dietary intake of omega-3 long-chain polyunsaturated fatty acid reduced laser-induced CNV in mice. Taken together, laser-induced CNV lesions can be easily and consistently applied using the image-guided laser platform. Mice at 6-8 weeks of age are ideal for the laser-induced CNV model.


Subject(s)
Choroidal Neovascularization/drug therapy , Imaging, Three-Dimensional , Laser Coagulation , Animals , Bruch Membrane/pathology , Choroidal Neovascularization/pathology , Diet , Disease Models, Animal , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Female , Male , Mice, Inbred C57BL , Volatilization
2.
Am J Clin Nutr ; 101(4): 879-88, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25833984

ABSTRACT

BACKGROUND: Retinopathy of prematurity (ROP) is a vision-threatening disease in premature infants. Serum adiponectin (APN) concentrations positively correlate with postnatal growth and gestational age, important risk factors for ROP development. Dietary ω-3 (n-3) long-chain polyunsaturated fatty acids (ω-3 LCPUFAs) suppress ROP and oxygen-induced retinopathy (OIR) in a mouse model of human ROP, but the mechanism is not fully understood. OBJECTIVE: We examined the role of APN in ROP development and whether circulating APN concentrations are increased by dietary ω-3 LCPUFAs to mediate the protective effect in ROP. DESIGN: Serum APN concentrations were correlated with ROP development and serum ω-3 LCPUFA concentrations in preterm infants. Mouse OIR was then used to determine whether ω-3 LCPUFA supplementation increases serum APN concentrations, which then suppress retinopathy. RESULTS: We found that in preterm infants, low serum APN concentrations positively correlate with ROP, and serum APN concentrations positively correlate with serum ω-3 LCPUFA concentrations. In mouse OIR, serum total APN and bioactive high-molecular-weight APN concentrations are increased by ω-3 LCPUFA feed. White adipose tissue, where APN is produced and assembled in the endoplasmic reticulum, is the major source of serum APN. In mouse OIR, adipose endoplasmic reticulum stress is increased, and APN production is suppressed. ω-3 LCPUFA feed in mice increases APN production by reducing adipose endoplasmic reticulum stress markers. Dietary ω-3 LCPUFA suppression of neovascularization is reduced from 70% to 10% with APN deficiency. APN receptors localize in the retina, particularly to pathologic neovessels. CONCLUSION: Our findings suggest that increasing APN by ω-3 LCPUFA supplementation in total parental nutrition for preterm infants may suppress ROP.


Subject(s)
Adiponectin/blood , Adiposity/drug effects , Endoplasmic Reticulum Stress/drug effects , Fatty Acids, Omega-3/administration & dosage , Retinal Neovascularization/drug therapy , 3T3-L1 Cells , Adiponectin/deficiency , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Animals, Newborn/blood , Disease Models, Animal , Fatty Acids, Omega-3/blood , Female , Humans , Infant, Newborn , Infant, Premature/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prospective Studies , Retina/drug effects , Retina/metabolism , Retinal Neovascularization/blood , Retinopathy of Prematurity/blood , Retinopathy of Prematurity/drug therapy
3.
Arterioscler Thromb Vasc Biol ; 34(3): 581-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24458713

ABSTRACT

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.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Fatty Acids, Omega-3/toxicity , Macrophages/enzymology , Monocytes/enzymology , Retinal Neovascularization/chemically induced , Animals , Arachidonic Acid/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Biotransformation , Cell Hypoxia , Cytochrome P-450 CYP2C8 , Dietary Fats/pharmacokinetics , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/metabolism , Epoxide Hydrolases/deficiency , Epoxide Hydrolases/genetics , Epoxide Hydrolases/physiology , Eye Proteins/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/classification , Fatty Acids, Omega-3/pharmacokinetics , Fatty Acids, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/pharmacokinetics , Humans , Lipoxygenase/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen/toxicity , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/biosynthesis , Receptor, TIE-2/genetics , Recombinant Fusion Proteins/metabolism , Retinal Neovascularization/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL