Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 17654, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30518776

ABSTRACT

Angiogenesis is crucial for tissue development and homeostasis; however, excessive angiogenesis can lead to diseases, including arthritis and cancer metastasis. Some antiangiogenic drugs are available, but side effects remain problematic. Thus, alternative angiogenesis inhibition strategies are needed. Fibulin-7 (Fbln7) is a newly discovered member of the fibulin protein family, a group of cell-secreted glycoproteins, that functions as a cell adhesion molecule and interacts with other extracellular matrix (ECM) proteins as well as cell receptors. We previously showed that a recombinant C-terminal Fbln7 fragment (Fbln7-C) inhibits tube formation by human umbilical vein endothelial cells (HUVECs) in vitro. In the present study, we examined the in vivo antiangiogenic activity of recombinant full-length Fbln7 (Fbln7-FL) and Fbln7-C proteins using a rat corneal angiogenesis model. We found that both Fbln7-FL and Fbln7-C inhibited neovascularization. Fbln7-C bound to vascular endothelial growth factor receptor 2 (VEGFR2), inhibiting VEGFR2 and ERK phosphorylation and resulting in reduced HUVEC motility. HUVEC attachment to Fbln7-C occurred through an interaction with integrin α5ß1 and regulated changes in cellular morphology. These results suggest that Fbln7-C action may target neovascularization by altering cell/ECM associations. Therefore, Fbln7-C could have potential as a therapeutic agent for diseases associated with angiogenesis.


Subject(s)
Angiogenesis Inhibitors/metabolism , Calcium-Binding Proteins/metabolism , Neovascularization, Physiologic , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/pharmacology , Female , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha5beta1/metabolism , Mice , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Sci Rep ; 5: 9144, 2015 Mar 16.
Article in English | MEDLINE | ID: mdl-25775051

ABSTRACT

Age-related macular degeneration (AMD) has been associated with both accumulation of lipid and lipid oxidative products, as well as increased neuroinflammatory changes and microglial activation in the outer retina. However, the relationships between these factors are incompletely understood. 7-Ketocholesterol (7KCh) is a cholesterol oxidation product localized to the outer retina with prominent pro-inflammatory effects. To explore the potential relationship between 7KCh and microglial activation, we localized 7KCh and microglia to the outer retina of aged mice and investigated 7KCh effects on retinal microglia in both in vitro and in vivo systems. We found that retinal microglia demonstrated a prominent chemotropism to 7KCh and readily internalized 7KCh. Sublethal concentrations of 7KCh resulted in microglial activation and polarization to a pro-inflammatory M1 state via NLRP3 inflammasome activation. Microglia exposed to 7KCh reduced expression of neurotrophic growth factors but increased expression of angiogenic factors, transitioning to a more neurotoxic and pro-angiogenic phenotype. Finally, subretinal transplantation of 7KCh-exposed microglia promoted choroidal neovascularization (CNV) relative to control microglia in a Matrigel-CNV model. The interaction of retinal microglia with 7KCh in the aged retina may thus underlie how outer retinal lipid accumulation in intermediate AMD results in neuroinflammation that ultimately drives progression towards advanced AMD.


Subject(s)
Ketocholesterols/metabolism , Macular Degeneration/etiology , Macular Degeneration/pathology , Microglia/metabolism , Neovascularization, Pathologic , Retina/metabolism , Retina/pathology , Animals , CX3C Chemokine Receptor 1 , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Ketocholesterols/pharmacology , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/immunology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Retina/drug effects
3.
Invest Ophthalmol Vis Sci ; 56(3): 1658-64, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25678688

ABSTRACT

PURPOSE: The inflammatory process plays a major role in the pathogenesis of AMD, and recent data indicate the involvement of inflammasomes. Inflammasomes are intracellular structures that trigger inflammation by producing mature interleukin-(IL)-1ß and IL-18. This study examined the capacity of 7-ketocholesterol (7KCh), an oxysterol that accumulates in the retinal pigmented epithelium (RPE) and choroid, to initiate inflammasome formation in RPE and bone marrow-derived cells. METHODS: Tested cells included fetal human RPE (fhRPE), human ARPE-19 cells, primary human brain microglia cells, and human THP-1 monocyte cells. 7-Ketocholesterol and other compounds were added to the cell cultures, and their stimulatory effects were determined by quantitative PCR and release of cytokines, measured by ELISA and Western blotting. RESULTS: 7-Ketocholesterol efficiently induced inflammasome formation by all primed cell populations, but secreted cytokine levels were higher in cultures of bone marrow-derived cells (microglia and THP-1 cells) than in RPE cultures. Interestingly, inflammasomes formed in cells of the two populations differed strikingly in their preferential production of the two cytokines. Thus, whereas bone marrow-derived cells produced levels of IL-1ß that were higher than those of IL-18, the opposite was found with RPE cells, which secreted higher levels of IL-18. Importantly, Western blot analysis showed that IL-18, but not IL-1ß, was expressed constitutively by RPE cells. CONCLUSIONS: 7-Ketocholesterol efficiently stimulates inflammasome formation and is conceivably involved in the pathogenesis of AMD. In contrast to bone marrow-derived cells, RPE cells produced higher levels of IL-18 than IL-1ß. Further, IL-18, a multifunctional cytokine, was expressed constitutively by RPE cells. These observations provide new information about stimuli and cells and their products assumed to be involved in the pathogenesis of AMD.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Inflammasomes/drug effects , Inflammasomes/physiology , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Ketocholesterols/pharmacology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/physiology , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans
4.
Biochim Biophys Acta ; 1851(5): 605-19, 2015 May.
Article in English | MEDLINE | ID: mdl-25617738

ABSTRACT

Accumulation of 7-ketocholesterol (7KCh) in tissues has been previously associated with various chronic aging diseases. Orally ingested 7KCh is readily metabolized by the liver and does not pose a toxicity threat. However, 7KCh formed in situ, usually associated with lipoprotein deposits, can adversely affect surrounding tissues by causing inflammation and cytotoxicity. In this study we have investigated various mechanisms for extra-hepatic metabolism of 7KCh (e.g. hydroxylation, sulfation) and found only esterification to fatty acids. The esterification of 7KCh to fatty acids involves the combined action of cytosolic phospholipase A2 alpha (cPLA2α) and sterol O-acyltransferase (SOAT1). Inhibition of either one of these enzymes ablates 7KCh-fatty acid ester (7KFAE) formation. The 7KFAEs are not toxic and do not induce inflammatory responses. However, they can be unstable and re-release 7KCh. The higher the degree of unsaturation, the more unstable the 7KFAE (e.g. 18:0>18:1>18:2>18:3≫20:4). Biochemical inhibition and siRNA knockdown of SOAT1 and cPLA2α ablated the 7KFAE synthesis in cultured ARPE19 cells, but had little effect on the 7KCh-induced inflammatory response. Overexpression of SOAT1 reduced the 7KCh-induced inflammatory response and provided some protection from cell death. This effect is likely due to the increased conversion of 7KCh to 7KFAEs, which reduced the intracellular 7KCh levels. Addition of HDL selectively increased the efflux of 7KFAEs and enhanced the effect of SOAT1 overexpression. Our data suggests an additional function for HDL in aiding extra-hepatic tissues to eliminate 7KCh by returning 7KFAEs to the liver for bile acid formation.


Subject(s)
Cholesterol, HDL/metabolism , Fatty Acids/metabolism , Group IV Phospholipases A2/metabolism , Ketocholesterols/metabolism , Retinal Pigment Epithelium/enzymology , Sterol O-Acyltransferase/metabolism , Animals , Biological Transport , Biotransformation , Cell Death , Cell Line , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Esterification , Group IV Phospholipases A2/antagonists & inhibitors , Group IV Phospholipases A2/genetics , Humans , Macaca mulatta , Male , Mass Spectrometry , RNA Interference , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Sterol O-Acyltransferase/antagonists & inhibitors , Sterol O-Acyltransferase/genetics , Time Factors , Transfection
5.
Exp Eye Res ; 128: 151-5, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25261634

ABSTRACT

We analyzed by LCMS lipid extracts of lens, retina (MNR) and RPE/Choroid (MPEC) from macaque monkeys 2-25 yr in age to determine their content of 7-ketocholesterol (7KCh) as function of age. In addition we also analyzed drusen capped with retinal pigment epithelium (RPE), RPE, and neural retina from human donors age 72-95 yr. The lowest 7KCh levels were found in monkey lens (<0.5-3.5 pmol 7KCh per nmol Ch), the second highest in MNR (1-15 pmol/nmol), and the highest in MPEC (1 to >60 pmol/nmol). Despite individual variability all three tissues demonstrated a strong age-related increase. In older human donors 7KCh levels were significantly higher. The levels in human neural retina ranged from 8 to 20 pmol/nmol, similar to the oldest monkeys, but 7-KCh levels in RPE ranged from 200 to 17,000 pmol/nmol, and in RPE-capped drusen from 200 to 2000 pmol/nmol, levels that would be lethal in most cultured cell systems. Most of the 7KCh is sequestered and not readily available to the surrounding tissue, based on published histochemical evidence that extracellular cholesterol (Ch) and cholesteryl fatty acid esters (CEs) are highly concentrated in Bruch's membrane and drusen. However, adjacent tissues, especially RPE but also choriocapillaris endothelium, could be chronically inflamed and in peril of receiving a lethal exposure. Implications for initiation and progression of age-related macular degeneration are discussed.


Subject(s)
Aging/physiology , Choroid/metabolism , Ketocholesterols/metabolism , Lens, Crystalline/metabolism , Retina/metabolism , Retinal Drusen/metabolism , Aged , Aged, 80 and over , Animals , Chromatography, Liquid , Female , Humans , Macaca mulatta , Male , Mass Spectrometry
6.
PLoS One ; 9(7): e100985, 2014.
Article in English | MEDLINE | ID: mdl-25036103

ABSTRACT

The cholesterol oxide 7-ketocholesterol (7KCh) has been implicated in numerous age-related diseases such as atherosclerosis, Alzheimer's disease, Parkinson's disease, cancer and age-related macular degeneration. It is formed by the autooxidation of cholesterol and especially cholesterol-fatty acid esters found in lipoprotein deposits. This molecule causes complex and potent inflammatory responses in vitro and in vivo. It is suspected of causing chronic inflammation in tissues exposed to oxidized lipoprotein deposits. In this study we have examined the inflammatory pathways activated by 7KCh both in cultured ARPE19 cells and in vivo using 7KCh-containing implants inserted into the anterior chamber of the rat eye. Our results indicate that 7KCh-induced inflammation is mediated mostly though the TLR4 receptor with some cross-activation of EGFR-related pathways. The majority of the cytokine inductions seem to signal via the TRIF/TRAM side of the TLR4 receptor. The MyD88/TIRAP side only significantly effects IL-1ß inductions. The 7KCh-induced inflammation also seems to involve a robust ER stress response. However, this response does not seem to involve a calcium efflux-mediated UPR. Instead the ER stress response seems to be mediated by yet identified kinases activated through the TLR4 receptor. Some of the kinases identified are the RSKs which seem to mediate the cytokine inductions and the cell death pathway but do not seem to be involved in the ER stress response.


Subject(s)
Ketocholesterols/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Cell Death/drug effects , Cell Line , Chromones/pharmacology , Cyclopropanes/metabolism , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Fatty Acids, Monounsaturated/metabolism , Humans , Inflammasomes/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Morpholines/pharmacology , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism , Rats , Suppressor of Cytokine Signaling Proteins/metabolism
7.
J Neurosci ; 34(10): 3793-806, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24599476

ABSTRACT

Chronic retinal inflammation in the form of activated microglia and macrophages are implicated in the etiology of neurodegenerative diseases of the retina, including age-related macular degeneration, diabetic retinopathy, and glaucoma. However, molecular biomarkers and targeted therapies for immune cell activation in these disorders are currently lacking. To address this, we investigated the involvement and role of translocator protein (TSPO), a biomarker of microglial and astrocyte gliosis in brain degeneration, in the context of retinal inflammation. Here, we find that TSPO is acutely and specifically upregulated in retinal microglia in separate mouse models of retinal inflammation and injury. Concomitantly, its endogenous ligand, diazepam-binding inhibitor (DBI), is upregulated in the macroglia of the mouse retina such as astrocytes and Müller cells. In addition, we discover that TSPO-mediated signaling in microglia via DBI-derived ligands negatively regulates features of microglial activation, including reactive oxygen species production, TNF-α expression and secretion, and microglial proliferation. The inducibility and effects of DBI-TSPO signaling in the retina reveal a mechanism of coordinated macroglia-microglia interactions, the function of which is to limit the magnitude of inflammatory responses after their initiation, facilitating a return to baseline quiescence. Our results indicate that TSPO is a promising molecular marker for imaging inflammatory cell activation in the retina and highlight DBI-TSPO signaling as a potential target for immodulatory therapies.


Subject(s)
Cell Communication/physiology , Microglia/metabolism , Receptors, GABA/physiology , Retina/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Astrocytes/metabolism , Cells, Cultured , Female , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Receptors, GABA/genetics
8.
PLoS One ; 8(2): e56099, 2013.
Article in English | MEDLINE | ID: mdl-23409131

ABSTRACT

Accumulation of 7-Ketocholesterol (7KCh) in lipid deposits has been implicated in a variety of chronic diseases including atherosclerosis, Alzheimer's disease and age-related macular degeneration. 7KCh is known to be pro-inflammatory and cytotoxic to various types of cultured cells but little is known about its effects in vivo. In this study we have investigated the effects of 7KCh in vivo by implanting biodegradable wafers into the anterior chamber of the rat eye. The wafers were prepared using a mixture of two biodegradable polymers with different amounts of 7KCh. The 7KCh-containing implants induced massive angiogenesis and inflammation. By contrast, no angiogenesis and very little inflammation were observed with cholesterol-containing implants. The neovessel growth was monitored by fluorescein angiography. Neovessels were observed 4 days post implantation and peaked between 7 to 10 days. The angiography and isolectin IB(4) labeling demonstrated that the neovessels originated from the limbus and grew through the cornea. Immunolabeling with anti-CD68 suggested that the 7KCh-containing implants had extensive macrophage infiltration as well as other cell types. A significant increase in VEGF was also observed in 7KCh-containing implants by fluorescent immunolabeling and by immunoblot of the aqueous humor (AH). Direct measurement of VEGF, IL-1ß and GRO/KC demonstrated a marked elevation of these factors in the AH of the 7KCh-implants. In summary this study demonstrates two important things: 1) 7KCh is pro-angiogenic and pro-inflammatory in vivo and 2) implants containing 7KCh may be used to create a novel angiogenesis model in rats.


Subject(s)
Corneal Neovascularization/chemically induced , Disease Models, Animal , Ketocholesterols/pharmacology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aqueous Humor/drug effects , Aqueous Humor/metabolism , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Dose-Response Relationship, Drug , Immunohistochemistry , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Prostheses and Implants , Protein Transport/drug effects , Rats , Vascular Endothelial Growth Factor A/metabolism
9.
Biochim Biophys Acta ; 1821(4): 637-46, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22342272

ABSTRACT

Sterculic acid is a cyclopropene fatty acid with numerous biological activities. In this study we demonstrate that sterculic acid is a potent inhibitor of endoplasmic reticulum (ER) stress and related inflammation caused by 7-ketocholesterol (7KCh). 7KCh is a highly toxic oxysterol suspected in the pathogenesis of various age-related diseases such as atherosclerosis, Alzheimer's disease and age-related macular degeneration. Sterculic acid demonstrated to be 5-10 times more effective than other anti-inflammatory fatty acids at inhibiting 7KCh-mediated inflammatory responses in cultured cells. In vivo, sterculic acid was effective at inhibiting the formation of choroidal neovascularization (CNV) in the laser-injury rat model. Our data suggests that sterculic acid may be useful in treating CNV in certain forms of age-related macular degeneration.


Subject(s)
Choroidal Neovascularization/prevention & control , Cyclopropanes/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Inflammation/prevention & control , Ketocholesterols/pharmacology , Animals , Cells, Cultured , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Cytokines/genetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Drug Antagonism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Enzyme Inhibitors/pharmacology , Fatty Acids/pharmacology , Gene Expression/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Lasers/adverse effects , Male , Rats , Rats, Inbred BN , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
10.
Proc Natl Acad Sci U S A ; 107(35): 15523-8, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713727

ABSTRACT

Retinal degenerative diseases, such as retinitis pigmentosa and Leber congenital amaurosis, are a leading cause of untreatable blindness with substantive impact on the quality of life of affected individuals and their families. Mouse mutants with retinal dystrophies have provided a valuable resource to discover human disease genes and helped uncover pathways critical for photoreceptor function. Here we show that the rd11 mouse mutant and its allelic strain, B6-JR2845, exhibit rapid photoreceptor dysfunction, followed by degeneration of both rods and cones. Using linkage analysis, we mapped the rd11 locus to mouse chromosome 13. We then identified a one-nucleotide insertion (c.420-421insG) in exon 3 of the Lpcat1 gene. Subsequent screening of this gene in the B6-JR2845 strain revealed a seven-nucleotide deletion (c.14-20delGCCGCGG) in exon 1. Both sequence changes are predicted to result in a frame-shift, leading to premature truncation of the lysophosphatidylcholine acyltransferase-1 (LPCAT1) protein. LPCAT1 (also called AYTL2) is a phospholipid biosynthesis/remodeling enzyme that facilitates the conversion of palmitoyl-lysophosphatidylcholine to dipalmitoylphosphatidylcholine (DPPC). The analysis of retinal lipids from rd11 and B6-JR2845 mice showed substantially reduced DPPC levels compared with C57BL/6J control mice, suggesting a causal link to photoreceptor dysfunction. A follow-up screening of LPCAT1 in retinitis pigmentosa and Leber congenital amaurosis patients did not reveal any obvious disease-causing mutations. Previously, LPCAT1 has been suggested to be critical for the production of lung surfactant phospholipids and biosynthesis of platelet-activating factor in noninflammatory remodeling pathway. Our studies add another dimension to an essential role for LPCAT1 in retinal photoreceptor homeostasis.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/genetics , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/genetics , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Animals , Base Sequence , Blotting, Northern , Chromatography, High Pressure Liquid , Chromosome Mapping , DNA Mutational Analysis , Humans , Immunoblotting , Leber Congenital Amaurosis/genetics , Lipids/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Mice, Mutant Strains , Microscopy, Electron, Transmission , Phosphatidylcholines/analysis , Photoreceptor Cells, Vertebrate/chemistry , Photoreceptor Cells, Vertebrate/ultrastructure , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinitis Pigmentosa/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
J Lipid Res ; 51(10): 2847-62, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20567027

ABSTRACT

This review will discuss the formation and potential implications of 7-ketocholesterol (7KCh) in the retina. 7KCh is a proinflammatory oxysterol known to be present in high amounts in oxidized LDL deposits associated with atheromatous plaques. 7KCh is generated in situ in these lipoprotein deposits where it can accumulate and reach very high concentrations. In normal primate retina, 7KCh has been found associated with lipoprotein deposits in the choriocapillaris, Bruch's membrane, and the retinal pigment epithelium (RPE). In photodamaged rats, 7KCh has been found in the neural retina in areas of high mitochondrial content, ganglion cells, photoreceptor inner segments and synapses, and the RPE. Intermediates found by LCMS indicate 7KCh is formed via a free radical-mediated mechanism catalyzed by iron. 7KCh seems to activate several kinase signaling pathways that work via nuclear factor κB and cause the induction of vascular endothelial growth factor, interleukin (IL)-6, and IL-8. There seems to be little evidence of 7KCh metabolism in the retina, although some form of efflux mechanism may be active. The chronic mode of formation and the potent inflammatory properties of 7KCh indicate it may be an "age-related" risk factor in aging diseases such as atherosclerosis, Alzheimer's, and age-related macular degeneration.


Subject(s)
Cholesterol/metabolism , Inflammation/metabolism , Ketocholesterols/metabolism , Macular Degeneration/metabolism , Retina/metabolism , Animals , Cell Death , Humans , Oxidation-Reduction
12.
Invest Ophthalmol Vis Sci ; 51(10): 4942-55, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20554621

ABSTRACT

PURPOSE: 7-Ketocholesterol (7KCh) accumulates in oxidized lipoprotein deposits and is known to be involved in macrophage foam cell formation and atherosclerosis. 7-KCh is present in the primate retina and is associated with oxidized lipoprotein deposits located in the choriocapillaris, Bruch's membrane, and retinal pigment epithelium (RPE). 7-KCh can also be formed in the retina as a consequence of light-induced iron release. The purpose of this study was to examine the signaling pathways involved in the 7KCh-mediated inflammatory response focusing on three cytokines, VEGF, IL-6, and IL-8. METHODS: ARPE-19 cells were treated with 7KCh solubilized in hydroxypropyl-ß-cyclodextrin. Cytokines were quantified by qRT-PCR (mRNA) and ELISA (protein) using commercially available products. NFκB activation was determined by IκBα mRNA induction. RESULTS: Treatment of ARPE-19 cells with 15 µM 7KCh markedly induced the expression of VEGF, IL-6, and IL-8. No increase in NOX-4 expression or ROS formation was detected. 7KCh induced the phosphorylation of ERK1/2 and p38MAPK, and inhibitors to these kinases markedly reduced the cytokine expression but did not affect the IκBα mRNA expression. By contrast, inhibition of PI3K and PKCζ significantly decreased the cytokine and IκBα mRNA expression. Inhibition of the IκB kinase complex essentially ablated all cytokine induction. CONCLUSIONS: 7KCh induces cytokines via three kinase signaling pathways, AKT-PKCζ-NFκB, p38 MAPK, and ERK. The MAPK/ERK pathways seem to preferentially enhance cytokine induction downstream from NFκB activation. The results of this study suggest that 7KCh activates these pathways through interactions in the plasma membrane, but the mechanism(s) remains unknown.


Subject(s)
Ketocholesterols/pharmacology , NF-kappa B/metabolism , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/drug effects , Signal Transduction/drug effects , Cell Line , Cell Survival , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunoblotting , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Retinal Pigment Epithelium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Exp Eye Res ; 90(3): 420-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20026324

ABSTRACT

Methionine sulfoxide reductase B2 (MSRB2) is a mitochondrial enzyme that converts methionine sulfoxide (R) enantiomer back to methionine. This enzyme is suspected of functioning to protect mitochondrial proteins from oxidative damage. In this study we report that the retina is one of the human tissues with highest levels of MSRB2 mRNA expression. Other tissues with high expression were heart, kidney and skeletal muscle. Overexpression of an MSRB2-GFP fusion protein increased the MSR enzymatic activity three-fold in stably transfected cultured RPE cells. This overexpression augmented the resistance of these cells to the toxicity induced by 7-ketocholesterol, tert-butyl hydroperoxide and all-trans retinoic acid. By contrast, knockdown of MSRB2 by a miRNA in stably transfected cells did not convey increased sensitivity to the oxidative stress. In the monkey retina MSRB2 localized to the ganglion cell layer (GLC), the outer plexiform layer (OPL) and the retinal pigment epithelium (RPE). MSRB2 expression is most pronounced in the OPL of the macula and foveal regions suggesting an association with the cone synaptic mitochondria. Our data suggests that MSRB2 plays an important function in protecting cones from multiple type of oxidative stress and may be critical in preserving central vision.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Oxidative Stress , Oxidoreductases/genetics , Retina/enzymology , Retinal Pigment Epithelium/drug effects , Transcription Factors/genetics , Animals , Cell Survival , Cells, Cultured , Chromatography, High Pressure Liquid , Cytoprotection , Enzyme Inhibitors/toxicity , Female , Gene Silencing , Green Fluorescent Proteins/metabolism , Humans , Ketocholesterols/toxicity , Kidney/enzymology , Macaca mulatta , Methionine Sulfoxide Reductases , Microfilament Proteins , Muscle, Skeletal/enzymology , Myocardium/enzymology , RNA, Messenger/metabolism , Recombinant Fusion Proteins , Retinal Cone Photoreceptor Cells/enzymology , Retinal Pigment Epithelium/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tretinoin/toxicity , tert-Butylhydroperoxide/toxicity
14.
Photochem Photobiol ; 85(5): 1116-25, 2009.
Article in English | MEDLINE | ID: mdl-19500292

ABSTRACT

Albino Sprague-Dawley rats are known to undergo photoreceptor degeneration after exposure to constant light, but the molecular mechanism(s) by which the photoreceptors degenerate is not fully understood. We hypothesized that cytotoxic oxysterols are generated in situ in the retina under such conditions and may be involved in the degenerative mechanism. Thus, photodamaged and control rat retinas were analyzed for oxysterols by liquid chromatography mass spectroscopy. Elevated levels of two known cytotoxic oxysterols, 7-ketocholesterol (7KCh) and 7alphabeta-hydroxycholesterol (7HCh), were found in the photodamaged retinas, at levels six-fold and 50-fold greater, respectively, than those found in non photodamaged controls. Notably, two key intermediates, 5,6alpha,beta-epoxycholesterol (5,6-epoxyCh) and 7alphabeta-hydroperoxy-cholesterol, were also identified, indicating that the formation of 7KCh and 7HCh is mediated by a free radical mechanism. By immunohistochemistry, 7KCh was localized to the ganglion cell layer, photoreceptor inner segments and retinal pigment epithelium (RPE), which coincides with the localization of ferritin in the retina. Exposure of a mixture of ferritin and low-density lipoprotein to intense white light in vitro produced similar oxysterol species as seen in vivo. We propose that the increased levels of 7KCh and 7HCh, especially in photoreceptor inner segments and RPE, may arise due to ferritin-catalyzed reactions and may be important contributors to the photoreceptor degeneration observed in photodamaged rats.


Subject(s)
Hydroxycholesterols/metabolism , Ketocholesterols/biosynthesis , Light , Retina/radiation effects , Animals , Chromatography, High Pressure Liquid , Female , Free Radicals/metabolism , Immunohistochemistry , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Retina/metabolism
15.
Genomics ; 93(1): 62-71, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18845237

ABSTRACT

MSRAs (methionine sulfoxide reductases A) are enzymes that reverse the effects of oxidative damage by reducing methionine sulfoxide back to methionine and recovering protein function. In this study we demonstrate that the transcriptional regulation of the human MSRA gene is complex and driven by two distinct promoters. Both promoters demonstrate high expression in human brain and kidney tissues. The upstream (promoter 1) regulates the msrA1 transcript that codes for the mitochondrial form of MSRA and is highly active in a broad range of cell lines. The downstream promoter (promoter 2) regulates the msrA2/3 transcripts that code for the cytosolic/nuclear forms of MSRA and is generally less active. Promoter 2 contains a 65 bp putative enhancer region that is very active in the retinal pigment epithelium-derived D407 cell line. Both promoters are partially regulated by all-trans retinoic acid via RARA and other RARs.


Subject(s)
Gene Expression Regulation/drug effects , Oxidoreductases/genetics , Oxidoreductases/metabolism , Promoter Regions, Genetic , Tretinoin/pharmacology , Brain/metabolism , Cell Line , Enhancer Elements, Genetic , Humans , Kidney/metabolism , Methionine Sulfoxide Reductases , Receptors, Retinoic Acid/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Invest Ophthalmol Vis Sci ; 50(2): 523-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18936140

ABSTRACT

PURPOSE: 7-Ketocholesterol is a highly toxic oxysterol found in abundance in atherosclerotic plaques and is believed to play a critical role in atherosclerosis. The purpose of this study was to identify and localize 7-ketocholesterol (7kCh) in the primate retina and to examine the potential consequences of its presence in oxidized lipid deposits in the retina. METHODS: Unsterified 7kCh was identified and quantified by high-performance liquid chromatography-mass spectrometry. Localization of 7kCh was performed by immunohistochemistry. VEGF induction was determined by qRT-PCR. Cell viability was determined by measuring cellular dehydrogenase activity. Analyses were performed using ARPE19 and human vascular endothelial cells (HMVECs). RESULTS: 7-Ketocholesterol is localized mainly to deposits in the choriocapillaris and Bruch's membrane and on the surfaces of vascular endothelial cells of the neural retina. RPE/choriocapillaris regions contained approximately four times more 7kCh than the neural retina. In ARPE19 cells and HMVECs, oxidized LDL and 7kCh induced VEGF 8- to 10-fold above controls. Hypoxia inducible factor (HIF)-1alpha levels did not increase as a result of 7kCh treatment, suggesting an HIF-independent induction pathway. Cholesterol sulfate, a liver X receptor (LXR) antagonist, had marked attenuation of the 7kCh-mediated VEGF induction. LXR-specific siRNAs also reduced VEGF induction. Inhibition of NF-kappaB with BAY 11-7082 reduced IL-8 but not VEGF induction. CONCLUSIONS: The location of 7-kCh in the retina and its induction of VEGF in cultured RPE cells and HMVECs suggest it may play a critical role in choroidal neovascularization. The pathway for VEGF induction seems to be independent of HIF-1alpha and NF-kappaB but seems to be partially regulated by LXRs.


Subject(s)
Bruch Membrane/metabolism , Choroidal Neovascularization/metabolism , Ketocholesterols/metabolism , Lipid Metabolism , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Cell Line , Cell Survival , Cholesterol Esters/metabolism , Chromatography, High Pressure Liquid , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Ketocholesterols/pharmacology , Lipoproteins, LDL/pharmacology , Liver X Receptors , Macaca mulatta , Mass Spectrometry , NF-kappa B/metabolism , Orphan Nuclear Receptors , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Retina/pathology , Retinal Pigment Epithelium/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
17.
Exp Eye Res ; 86(2): 452-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18078932

ABSTRACT

Oxidative stress and loss of mitochondrial function have been implicated in age-related ocular diseases and thus studying enzymes involved in these processes may be of particular importance in these diseases. Peroxiredoxin III (PRDX3) is one of a family of six known peroxiredoxins which are known to protect cells against oxidative damage. PRDX3 is localized to the mitochondria and has been reported to be induced by hydrogen peroxide in aortic endothelial and lens epithelial cells. Using a highly specific commercially available antibody, PRDX3 was readily detected by immunoblot in monkey neural retina. Immunohistochemical analysis of human and monkey retina localized PRDX3 mainly to the photoreceptor inner segments, the outer and inner plexiform layers, and the ganglion cells. These are areas of known high mitochondrial content. In the monkey retina some of the cone inner segments were much more strongly labeled than others. Dual labeling experiments using specific anti-cone opsin antibodies determined that the high expressing cones were of the blue subtype. By contrast, in the human retina all of the cone inner segments were immunoreactive. This difference may be due to a postmortem induction of PRDX3 in the human sample. These results suggest that PRDX3 may be important in protecting photoreceptor mitochondria especially in blue cones.


Subject(s)
Macaca mulatta/metabolism , Peroxiredoxins/metabolism , Retina/enzymology , Aged, 80 and over , Animals , Female , Humans , Oxidative Stress , Retinal Cone Photoreceptor Cells/enzymology
18.
Mol Vis ; 12: 1306-18, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-17110914

ABSTRACT

PURPOSE: In this study we examined the uptake of circulating lipoproteins into the retina, using a naturally fluorescent cholesterol analog for imaging and deuterated cholesterol for quantification by mass spectroscopy. The purpose of this study was to better understand cholesterol uptake, transport and homeostasis in the retina. METHODS: Human low density lipoprotein (LDL) and high density lipoprotein (HDL) were labeled with the fluorescent cholesterol analog cholesta-5,7,9(11)-trien-3beta-ol (CTL) and deuterated cholesterol (25,26,26,26,27,27,27-[2H]cholesterol, D7Ch). Rats were injected intravenously with CTL-LDL, CTL-HDL and D7Ch-LDL. Fluorescent confocal microscopy was used to image the uptake of CTL and mass spectroscopy was used to quantify D7Ch. Immunohistochemistry and fluorescent confocal microscopy were used to localize apoB (an LDL marker protein) and LDL receptor (LDLR) protein in rat and monkey retinas. RESULTS: CTL-specific fluorescence was imaged by confocal microscopy in the retinal pigment epithelium (RPE), choriocapillaris and parts of the neural retina within 2 h post-injection and was visualized in the photoreceptor outer segments by 4 h. Replacing LDL with HDL as the CTL carrier gave a less robust and more delayed labeling of retinal layers. Human apolipoprotein B (apoB) was also localized in the rat choriocapillaris and RPE by 4 h post-injection. Human apoB was detected by immunoblot analysis in the rat retina primarily as a about 70 kDa protein, suggesting proteolytic degradation. LDL-mediated uptake of cholesterol was quantified by mass spectroscopy using deuterated cholesterol in place of CTL. In addition, apoB and LDLR were localized in monkey retina by immunohistochemistry. CONCLUSIONS: The retina is capable of rapid uptake of circulating LDL via an LDLR-mediated process primarily occurring in the RPE and also possibly Müller cells. Despite the dominance of HDL over LDL in rat serum, LDL appears to be the preferred carrier for cholesterol transport to and uptake by the retina. The results also suggest that blood-borne LDL represents a significant contributor to the steady-state levels of cholesterol and possibly other lipids in the retina.


Subject(s)
Cholesterol/metabolism , Receptors, LDL/metabolism , Retina/metabolism , Animals , Apolipoproteins B/pharmacokinetics , Cell Line , Cholestenes/pharmacokinetics , Cholesterol/pharmacokinetics , Humans , Immunohistochemistry , Injections, Intravenous , Lipoproteins, LDL/administration & dosage , Lipoproteins, LDL/pharmacokinetics , Macaca mulatta , Rats , Rats, Sprague-Dawley , Tissue Distribution
19.
Mol Vis ; 12: 1319-33, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-17110915

ABSTRACT

PURPOSE: In our companion paper we demonstrated that circulating lipoproteins enter the retina via the retinal pigment epithelium (RPE) and possibly Müller cells. In order to understand how these lipids are transported within the retina, expression and localization of the main proteins known to be involved in systemic lipid transport was determined. METHODS: Expression of ABCA1, apoA1 (the major HDL protein), SR-BI, SR-BII, CD36, lecithin:cholesterol acyltransferase (LCAT), and cholesteryl ester transfer protein (CETP) was determined by reverse transcriptase polymerase chain reaction (RT-PCR) and immunoblots. Localization was determined by immunohistochemistry using fresh monkey vibrotome sections and imaged by confocal microscopy. RESULTS: ABCA1 and apoA1 were localized to the ganglion cell layer, retinal pigment epithelium (RPE), and rod photoreceptor inner segments. ApoA1 was also observed associated with rod photoreceptor outer segments, presumably localized to the interphotoreceptor matrix (IPM). The scavenger receptors SR-BI and SR-BII localized mainly to the ganglion cell layer and photoreceptor outer segments; in the latter they appear to be associated with microtubules. LCAT and CETP localized mainly to the IPM. CONCLUSIONS: The presence and specific localization of these well-known lipid transport proteins suggest that the retina employs an internal lipid transport mechanism that involves processing and maturation of HDL-like particles.


Subject(s)
CD36 Antigens/metabolism , Lipid Metabolism/physiology , Lipoproteins, HDL/metabolism , Lysosomal Membrane Proteins/metabolism , Retina/metabolism , Sialoglycoproteins/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/metabolism , Biological Transport , Carrier Proteins/metabolism , Cholesterol Ester Transfer Proteins/metabolism , Haplorhini , Humans , Lipoproteins/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Receptors, Lipoprotein/metabolism , Receptors, Scavenger , Tissue Distribution
20.
Exp Eye Res ; 83(2): 465-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16549062

ABSTRACT

Sterol 27-hydroxylase (CYP27A1) is a mitochondrial P-450 enzyme with broad substrate specificity for C27 sterols including 7-ketocholesterol (7kCh). CYP27A1 is widely expressed in human tissues but has not been previously demonstrated in the retina. In this study, we examined the expression and localization of CYP27A1 in the monkey retina where it localized mainly to the photoreceptor inner segments. CYP27A1 was also observed in Müller cells with faint immuno staining detected in the RPE and choriocapillaris. We also determined that the 27-hydroxylation of 7-ketocholesterol (27OH7kCh) rendered it non-toxic to cultured RPE cells. Moreover, the 27OH7kCh when mixed with 7-ketocholesterol significantly reduced the toxicity of 7-ketocholesterol. These data, when taken in context of the known functions of CYP27A1 imply that expression in the retina serves to modify the biological activity of oxidized sterols that are either transported or generated locally by photo-oxidation.


Subject(s)
Retina/chemistry , Steroid Hydroxylases/analysis , Animals , Cells, Cultured , Cholestanetriol 26-Monooxygenase , Enzyme Inhibitors/metabolism , Hydroxylation , Immunohistochemistry/methods , Ketocholesterols/metabolism , Macaca mulatta , Photoreceptor Cells, Vertebrate/chemistry , Pigment Epithelium of Eye/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...