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










Database
Language
Publication year range
1.
J Biol Chem ; 291(14): 7242-55, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26846851

ABSTRACT

Retinal angiogenesis is tightly regulated to meet oxygenation and nutritional requirements. In diseases such as proliferative diabetic retinopathy and neovascular age-related macular degeneration, uncontrolled angiogenesis can lead to blindness. Our goal is to better understand the molecular processes controlling retinal angiogenesis and discover novel drugs that inhibit retinal neovascularization. Phenotype-based chemical screens were performed using the ChemBridge Diverset(TM)library and inhibition of hyaloid vessel angiogenesis in Tg(fli1:EGFP) zebrafish. 2-[(E)-2-(Quinolin-2-yl)vinyl]phenol, (quininib) robustly inhibits developmental angiogenesis at 4-10 µmin zebrafish and significantly inhibits angiogenic tubule formation in HMEC-1 cells, angiogenic sprouting in aortic ring explants, and retinal revascularization in oxygen-induced retinopathy mice. Quininib is well tolerated in zebrafish, human cell lines, and murine eyes. Profiling screens of 153 angiogenic and inflammatory targets revealed that quininib does not directly target VEGF receptors but antagonizes cysteinyl leukotriene receptors 1 and 2 (CysLT1-2) at micromolar IC50values. In summary, quininib is a novel anti-angiogenic small-molecule CysLT receptor antagonist. Quininib inhibits angiogenesis in a range of cell and tissue systems, revealing novel physiological roles for CysLT signaling. Quininib has potential as a novel therapeutic agent to treat ocular neovascular pathologies and may complement current anti-VEGF biological agents.


Subject(s)
Angiogenesis Inhibitors , Drug Discovery , Phenols , Quinolines , Retinal Neovascularization/drug therapy , Signal Transduction/drug effects , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Cell Line , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Humans , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Mice , Phenols/chemistry , Phenols/pharmacokinetics , Phenols/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Zebrafish
2.
Dis Model Mech ; 3(3-4): 236-45, 2010.
Article in English | MEDLINE | ID: mdl-20142328

ABSTRACT

Approximately 2.5 million people worldwide are clinically blind because of diabetic retinopathy. In the non-proliferative stage, the pathophysiology of this ocular manifestation of diabetes presents as morphological and functional disruption of the retinal vasculature, and dysfunction of retinal neurons. However, it is uncertain whether the vascular and neuronal changes are interdependent or independent events. In addition, the identity of the retinal neurons that are most susceptible to the hyperglycaemia associated with diabetes is unclear. Here, we characterise a novel model of non-proliferative diabetic retinopathy in adult zebrafish, in which the zebrafish were subjected to oscillating hyperglycaemia for 30 days. Visual function is diminished in hyperglycaemic fish. Significantly, hyperglycaemia disrupts cone photoreceptor neurons the most, as evidenced by prominent morphological degeneration and dysfunctional cone-mediated electroretinograms. Disturbances in the morphological integrity of the blood-retinal barrier were also evident. However, we demonstrate that these early vascular changes are not sufficient to induce cone photoreceptor dysfunction, suggesting that the vascular and neuronal complications in diabetic retinopathy can arise independently. Current treatments for diabetic retinopathy target the vascular complications. Our data suggest that cone photoreceptor dysfunction is a clinical hallmark of diabetic retinopathy and that the debilitating blindness associated with diabetic retinopathy may be halted by neuroprotection of cones.


Subject(s)
Diabetic Retinopathy/complications , Diabetic Retinopathy/physiopathology , Hyperglycemia/complications , Hyperglycemia/physiopathology , Retinal Cone Photoreceptor Cells/pathology , Aging/drug effects , Aging/pathology , Animals , Basement Membrane/drug effects , Basement Membrane/pathology , Basement Membrane/ultrastructure , Blood-Retinal Barrier/drug effects , Blood-Retinal Barrier/physiopathology , Disease Models, Animal , Electroretinography , Glucose/pharmacology , Retinal Cone Photoreceptor Cells/drug effects , Retinal Degeneration/complications , Retinal Degeneration/physiopathology , Retinal Vessels/drug effects , Retinal Vessels/pathology , Retinal Vessels/ultrastructure , Zebrafish
3.
PLoS One ; 4(11): e7867, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19924235

ABSTRACT

Ocular neovascularisation is a pathological hallmark of some forms of debilitating blindness including diabetic retinopathy, age related macular degeneration and retinopathy of prematurity. Current therapies for delaying unwanted ocular angiogenesis include laser surgery or molecular inhibition of the pro-angiogenic factor VEGF. However, targeting of angiogenic pathways other than, or in combination to VEGF, may lead to more effective and safer inhibitors of intraocular angiogenesis. In a small chemical screen using zebrafish, we identify LY294002 as an effective and selective inhibitor of both developmental and ectopic hyaloid angiogenesis in the eye. LY294002, a PI3 kinase inhibitor, exerts its anti-angiogenic effect in a dose-dependent manner, without perturbing existing vessels. Significantly, LY294002 delivered by intraocular injection, significantly inhibits ocular angiogenesis without systemic side-effects and without diminishing visual function. Thus, targeting of PI3 kinase pathways has the potential to effectively and safely treat neovascularisation in eye disease.


Subject(s)
Gene Expression Regulation, Enzymologic , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinases/metabolism , Retina/pathology , Retinal Vessels/pathology , Animals , Chromones/pharmacology , Electroretinography/methods , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental , Hypoxia , Inhibitory Concentration 50 , Morpholines/pharmacology , Retina/embryology , Time Factors , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...