Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Vis ; 18: 1325-38, 2012.
Article in English | MEDLINE | ID: mdl-22690111

ABSTRACT

PURPOSE: Caffeic acid phenethyl ester (CAPE), an active component of honeybee propolis, has a wide range of beneficial properties. The purpose of this study was to test the protective role of CAPE in 661W cells (in vitro) against H(2)O(2)-mediated cell death and in albino rats (in vivo) against various light conditions. METHODS: The 661W cells were pretreated with CAPE and then stressed with H(2)O(2). Cell death was measured with lactate dehydrogenase (LDH) release assay, and mRNA and proteins were analyzed. Sprague Dawley rats were raised on either a control or CAPE (0.02%) diet and exposed to various light conditions for short or long periods. Retinal histology, mRNA, protein, lipid composition, and retinal function by electroretinography (ERG) were measured at the end of feeding. RESULTS: Pretreatment of 661W cells with CAPE reduced H(2)O(2)-mediated cell death in a dose-dependent manner and induced expression of heme oxygenase-1 (Ho1). Albino rats fed with CAPE had greater expression of Ho1 and intercellular adhesion molecule 1 (Icam1), less expression of FOS-like antigen (Fosl) and lipoxygenase 12 (Lox12) genes in the retina, less translocation of nuclear factor kappaB protein to the nucleus, and a lower molar ratio of n-3 polyunsaturated fatty acids. Further, the ERGs of the retinas of CAPE-fed rats were significantly higher than those of the control-fed rats when raised in dim light. CONCLUSIONS: CAPE can activate the antioxidative gene expression pathway in retinal cells in vitro and in vivo. Feeding CAPE to albino rats can enhance ERG responses and change the lipid profile in the rats' retinas.


Subject(s)
Caffeic Acids/pharmacology , Eye Proteins/genetics , Gene Expression/drug effects , Phenylethyl Alcohol/analogs & derivatives , Photoreceptor Cells, Vertebrate/drug effects , Protective Agents/pharmacology , Albinism , Animals , Cell Death/drug effects , Cell Death/genetics , Electroretinography , Eye Proteins/metabolism , Fatty Acids, Omega-3/analysis , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hydrogen Peroxide/pharmacology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , L-Lactate Dehydrogenase/analysis , L-Lactate Dehydrogenase/metabolism , Light , Lipoxygenases/genetics , Lipoxygenases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phenylethyl Alcohol/pharmacology , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/metabolism , Propolis/chemistry , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley
2.
PLoS One ; 7(6): e38616, 2012.
Article in English | MEDLINE | ID: mdl-22715395

ABSTRACT

Damage to the retinal pigment epithelium (RPE) is an early event in the pathogenesis of age-related macular degeneration (AMD). X-box binding protein 1 (XBP1) is a key transcription factor that regulates endoplasmic reticulum (ER) homeostasis and cell survival. This study aimed to delineate the role of endogenous XBP1 in the RPE. Our results show that in a rat model of light-induced retinal degeneration, XBP1 activation was suppressed in the RPE/choroid complex, accompanied by decreased anti-oxidant genes and increased oxidative stress. Knockdown of XBP1 by siRNA resulted in reduced expression of SOD1, SOD2, catalase, and glutathione synthase and sensitized RPE cells to oxidative damage. Using Cre/LoxP system, we generated a mouse line that lacks XBP1 only in RPE cells. Compared to wildtype littermates, RPE-XBP1 KO mice expressed less SOD1, SOD2, and catalase in the RPE, and had increased oxidative stress. At age 3 months and older, these mice exhibited apoptosis of RPE cells, decreased number of cone photoreceptors, shortened photoreceptor outer segment, reduced ONL thickness, and deficit in retinal function. Electron microscopy showed abnormal ultrastructure, Bruch's membrane thickening, and disrupted basal membrane infolding in XBP1-deficient RPE. These results indicate that XBP1 is an important gene involved in regulation of the anti-oxidant defense in the RPE, and that impaired activation of XBP1 may contribute to RPE dysfunction and cell death during retinal degeneration and AMD.


Subject(s)
Antioxidants/metabolism , DNA-Binding Proteins/metabolism , Eye Proteins/metabolism , Macular Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Transcription Factors/metabolism , Animals , Catalase/biosynthesis , Catalase/genetics , Cell Survival , DNA-Binding Proteins/immunology , Eye Proteins/genetics , Glutathione Synthase/biosynthesis , Glutathione Synthase/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Regulatory Factor X Transcription Factors , Retinal Pigment Epithelium/ultrastructure , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Transcription Factors/immunology , X-Box Binding Protein 1
4.
J Lipid Res ; 53(3): 494-504, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22158834

ABSTRACT

We hypothesized that reduction/loss of very long chain PUFAs (VLC-PUFAs) due to mutations in the ELOngase of very long chain fatty acid-4 (ELOVL4) protein contributes to retinal degeneration in autosomal dominant Stargardt-like macular dystrophy (STGD3) and age-related macular degeneration; hence, increasing VLC-PUFA in the retina of these patients could provide some therapeutic benefits. Thus, we tested the efficiency of elongation of C20-C22 PUFA by the ELOVL4 protein to determine which substrates are the best precursors for biosynthesis of VLC-PUFA. The ELOVL4 protein was expressed in pheochromocytoma cells, while green fluorescent protein-expressing and nontransduced cells served as controls. The cells were treated with 20:5n3, 22:6n3, and 20:4n6, either individually or in equal combinations. Both transduced and control cells internalized and elongated the supplemented FAs to C22-C26 precursors. Only ELOVL4-expressing cells synthesized C28-C38 VLC-PUFA from these precursors. In general, 20:5n3 was more efficiently elongated to VLC-PUFA in the ELOVL4-expressing cells, regardless of whether it was in combination with 22:6n3 or with 20:4n6. In each FA treatment group, C34 and C36 VLC-PUFAs were the predominant VLC-PUFAs in the ELOVL4-expressing cells. In summary, 20:5n3, followed by 20:4n6, seems to be the best precursor for boosting the synthesis of VLC-PUFA by ELOVL4 protein.


Subject(s)
Acetyltransferases/metabolism , Fatty Acids, Unsaturated/metabolism , Acetyltransferases/genetics , Animals , Arachidonic Acid/metabolism , Blotting, Western , Eye Proteins/genetics , Eye Proteins/metabolism , Fatty Acid Elongases , Gas Chromatography-Mass Spectrometry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , PC12 Cells , Rats
5.
J Biol Chem ; 286(37): 32491-501, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21785167

ABSTRACT

α-Phenyl-N-tert-butylnitrone (PBN), a free radical spin trap, has been shown previously to protect retinas against light-induced neurodegeneration, but the mechanism of protection is not known. Here we report that PBN-mediated retinal protection probably occurs by slowing down the rate of rhodopsin regeneration by inhibiting RPE65 activity. PBN (50 mg/kg) protected albino Sprague-Dawley rat retinas when injected 0.5-12 h before exposure to damaging light at 2,700 lux intensity for 6 h but had no effect when administered after the exposure. PBN injection significantly inhibited in vivo recovery of rod photoresponses and the rate of recovery of functional rhodopsin photopigment. Assays for visual cycle enzyme activities indicated that PBN inhibited one of the key enzymes of the visual cycle, RPE65, with an IC(50) = 0.1 mm. The inhibition type for RPE65 was found to be uncompetitive with K(i) = 53 µm. PBN had no effect on the activity of other visual cycle enzymes, lecithin retinol acyltransferase and retinol dehydrogenases. Interestingly, a more soluble form of PBN, N-tert-butyl-α-(2-sulfophenyl) nitrone, which has similar free radical trapping activity, did not protect the retina or inhibit RPE65 activity, providing some insight into the mechanism of PBN specificity and action. Slowing down the visual cycle is considered a treatment strategy for retinal diseases, such as Stargardt disease and dry age-related macular degeneration, in which toxic byproducts of the visual cycle accumulate in retinal cells. Thus, PBN inhibition of RPE65 catalytic action may provide therapeutic benefit for such retinal diseases.


Subject(s)
Carrier Proteins/metabolism , Cyclic N-Oxides/pharmacology , Eye Proteins/metabolism , Light/adverse effects , Neuroprotective Agents/pharmacology , Retinal Degeneration , Retinal Rod Photoreceptor Cells/enzymology , Rhodopsin/metabolism , cis-trans-Isomerases/metabolism , Acyltransferases/metabolism , Alcohol Oxidoreductases/metabolism , Animals , Rats , Rats, Sprague-Dawley , Retinal Degeneration/enzymology , Retinal Degeneration/prevention & control
6.
Invest Ophthalmol Vis Sci ; 51(9): 4422-31, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20393115

ABSTRACT

PURPOSE: Recent evidence suggests that ceramide metabolism plays an important role in retinal photoreceptor cell survival and apoptosis. The purpose of this study was to characterize sphingolipids in the retina with special emphasis on the very-long-chain-containing saturated (VLC-FA) and polyunsaturated (VLC-PUFA) fatty acid-containing species. The VLC-FAs and VLC-PUFAs are synthesized by the ELOVL4 protein, which is involved in human Stargardt's macular dystrophy type 3 (STGD3). METHODS: Total lipids were extracted from retina and other tissues, and different sphingolipid classes were isolated and purified using various combinations of liquid- and solid-phase separation. Purified sphingolipids were analyzed by high-performance thin layer chromatography (HPTLC), gas chromatography (GC), and GC-MS (GC-mass spectrometry). RESULTS: Nonsialylated sphingolipids (NSLs) comprised approximately 3.5% of total retinal lipids of which 70% was sphingomyelin. Ceramide and glycosylceramides (GCs) constituted

Subject(s)
Fatty Acids, Unsaturated/metabolism , Fatty Acids/metabolism , Retina/metabolism , Sphingolipids/metabolism , Animals , Brain/metabolism , Cattle , Cell Membrane/metabolism , Chromatography, Gas , Chromatography, Thin Layer , Fatty Acids/isolation & purification , Fatty Acids, Unsaturated/isolation & purification , Gas Chromatography-Mass Spectrometry , Liver/metabolism , Male , Organ Specificity , Rats , Reactive Oxygen Species/metabolism , Skin/metabolism , Sphingolipids/isolation & purification , Testis/metabolism
7.
J Lipid Res ; 51(7): 1624-42, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20299492

ABSTRACT

Compared with other mammalian tissues, retina is highly enriched in PUFA. Long-chain PUFA (LC-PUFA; C18-C24) are essential FAs that are enriched in the retina and are necessary for maintenance of normal retinal development and function. The retina, brain, and sperm also contain very LC-PUFA (VLC-PUFA; >C24). Although VLC-PUFA were discovered more than two decades ago, very little is known about their biosynthesis and functional roles in the retina. This is due mainly to intrinsic difficulties associated with working on these unusually long polyunsaturated hydrocarbon chains and their existence in small amounts. Recent studies on the FA elongase elongation of very long chain fatty acids-4 (ELOVL4) protein, however, suggest that VLC-PUFA probably play some uniquely important roles in the retina as well as the other tissues. Mutations in the ELOVL4 gene are found in patients with autosomal dominant Stargardt disease. Here, we review the recent literature on VLC-PUFA with special emphasis on the elongases responsible for their synthesis. We focus on a novel elongase, ELOVL4, involved in the synthesis of VLC-PUFA, and the importance of these FAs in maintaining the structural and functional integrity of retinal photoreceptors.


Subject(s)
Eye Proteins/metabolism , Fatty Acids, Unsaturated , Membrane Proteins/metabolism , Retinaldehyde/chemistry , Acetyltransferases/metabolism , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , DNA Mutational Analysis , Eye Proteins/genetics , Fatty Acid Elongases , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/metabolism , Humans , Membrane Proteins/genetics , Molecular Structure , Retina/chemistry , Retina/metabolism , Retinaldehyde/metabolism , Substrate Specificity , Tissue Distribution
8.
Adv Exp Med Biol ; 664: 233-42, 2010.
Article in English | MEDLINE | ID: mdl-20238022

ABSTRACT

The disk membranes of retinal photoreceptor outer segments and other neuronal and reproductive tissues are enriched in docosahexaenoic acid (DHA, 22:6n3), which is essential for their normal function and development. The fatty acid condensing enzyme Elongation of Very Long chain fatty acids-4 (ELOVL4) is highly expressed in retina photoreceptors as well as other tissues with high 22:6n3 content. Mutations in the ELOVL4 gene are associated with autosomal dominant Stargardt-like macular dystrophy (STGD3) and results in synthesis of a truncated protein that cannot be targeted to the endoplasmic reticulum (ER), the site of fatty acid biosynthesis. Considering the abundance and essential roles of 22:6n3 in ELOVL4-expressing tissues (except the skin), it was proposed that the ELOVL4 protein may be involved in 22:6n3 biosynthesis. We tested the hypothesis that the ELOVL4 protein is involved in 22:6n3 biosynthesis by selectively silencing expression of the protein in the cone photoreceptors derived cell line 661 w and showed that the ELOVL4 protein is not involved in DHA biosynthesis from the short chain fatty acid precursors 18:3n3 and 22:5n3.


Subject(s)
Docosahexaenoic Acids/biosynthesis , Eye Proteins/metabolism , Membrane Proteins/metabolism , Animals , Cell Line , Eye Proteins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Membrane Proteins/genetics , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
9.
Free Radic Biol Med ; 48(1): 16-25, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19686838

ABSTRACT

Mutations of the photoreceptor retinol dehydrogenase 12 (RDH12) gene cause the early onset retinal dystrophy Leber congenital amaurosis (LCA) by mechanisms not completely resolved. Determining the physiological role of RDH12 in photoreceptors is the focus of this study. Previous studies showed that RDH12, and the closely related retinol dehydrogenase RDH11, can enzymatically reduce toxic lipid peroxidation products such as 4-hydroxynonenal (4-HNE), in vitro. To explore the significance of this activity, we investigated the ability of RDH11 and RDH12 to protect stably transfected HEK-293 cells against the toxicity of 4-HNE. Both enzymes protected against 4-HNE modification of proteins and 4-HNE-induced apoptosis in HEK-293 cells. In the retina, exposure to bright light induced lipid peroxidation, 4-HNE production, and 4-HNE modification of proteins in photoreceptor inner segments, where RDH11 and RDH12 are located. In mouse retina, RDH12-but not RDH11-protected against adduct formation, suggesting that 4-HNE is a physiological substrate of RDH12. RDH12-but not RDH11-also protected against light-induced apoptosis of photoreceptors. We conclude that in mouse retina RDH12 reduces 4-HNE to a nontoxic alcohol, protecting cellular macromolecules against oxidative modification and protecting photoreceptors from light-induced apoptosis. This activity is of particular significance to the understanding of the molecular mechanisms of RDH12-induced LCA.


Subject(s)
Alcohol Oxidoreductases/metabolism , Aldehydes/metabolism , Photoreceptor Cells/metabolism , Alcohol Oxidoreductases/genetics , Aldehydes/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Light , Lipid Peroxidation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phenotype , Photoreceptor Cells/drug effects , Photoreceptor Cells/pathology
10.
Invest Ophthalmol Vis Sci ; 50(7): 3456-63, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19369235

ABSTRACT

PURPOSE: A strong association between retinal degeneration and obesity has been shown in humans. However, the molecular basis of increased risk for retinal degeneration in obesity is unknown. Thus, an animal model with obesity and retinal degeneration would greatly aid the understanding of obesity-associated retinal degeneration. The retinal abnormalities in a novel rat model (WNIN-Ob) with spontaneously developed obesity are described. METHODS: Histologic and immunohistochemical examination were performed on retinal sections of 2- to 12-month-old WNIN-Ob rats, and findings were compared with those of lean littermate controls. RNA from retinas of 12-month-old WNIN-Ob and lean littermate rats was used for microarray and qRT-PCR analysis. RESULTS: The WNIN-Ob rats developed severe obesity, with an onset at approximately 35 days. Evaluation of retinal morphology in 2- to 12-month-old WNIN-Ob and age-matched lean littermate controls revealed progressive retinal degeneration, with an onset between 4 to 6 months of age. Immunohistochemical analysis with anti-rhodopsin, anti-cone opsin, and PSD-95 antibodies further confirmed retinal degeneration, particularly rod cell loss and thinner outer plexiform layer, in the obese rat retina. Gene expression by microarray analysis and qRT-PCR established activation of stress response, tissue remodeling, impaired phototransduction, and photoreceptor degeneration in WNIN-Ob rat retina. CONCLUSIONS: WNIN-Ob rats develop increased stress in retinal tissue and progressive retinal degeneration after the onset of severe obesity. The WNIN-Ob rat is the first rat model to develop retinal degeneration after the onset of obesity. This novel rat model may be a valuable tool for investigating retinal degeneration associated with obesity in humans.


Subject(s)
Disease Models, Animal , Obesity/physiopathology , Retina/physiopathology , Retinal Degeneration/physiopathology , Animals , Disks Large Homolog 4 Protein , Female , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Microscopy, Fluorescence , Obesity/genetics , Obesity/metabolism , Oligonucleotide Array Sequence Analysis , Opsins/metabolism , Rats , Rats, Wistar , Retina/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rhodopsin/metabolism
11.
Free Radic Biol Med ; 46(5): 672-9, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19121385

ABSTRACT

Age-related macular degeneration (AMD) is a complex disease that has potential involvement of inflammatory and oxidative stress-related pathways in its pathogenesis. In search of effective therapeutic agents, we tested curcumin, a naturally occurring compound with known anti-inflammatory and antioxidative properties, in a rat model of light-induced retinal degeneration (LIRD) and in retina-derived cell lines. We hypothesized that any compound effective against LIRD, which involves significant oxidative stress and inflammation, would be a candidate for further characterization for its potential application in AMD. We observed significant retinal neuroprotection in rats fed diets supplemented with curcumin (0.2% in diet) for 2 weeks. The mechanism of retinal protection from LIRD by curcumin involves inhibition of NF-kappaB activation and down-regulation of cellular inflammatory genes. When tested on retina-derived cell lines (661W and ARPE-19), pretreatment of curcumin protected these cells from H(2)O(2)-induced cell death by up-regulating cellular protective enzymes, such as HO-1, thioredoxin. Since, curcumin with its pleiotropic activities can modulate the expression and activation of many cellular regulatory proteins such as NF-kappaB, AKT, NRF2, and growth factors, which in turn inhibit cellular inflammatory responses and protect cells; we speculate that curcumin would be an effective nutraceutical compound for preventive and augmentative therapy of AMD.


Subject(s)
Apoptosis/drug effects , Curcumin/administration & dosage , Macular Degeneration/drug therapy , Phytotherapy , Retinal Cone Photoreceptor Cells/drug effects , Animals , Apoptosis/physiology , Cells, Cultured , Curcumin/pharmacology , Cytoprotection/physiology , Dietary Supplements , Disease Models, Animal , Electroretinography , Gene Expression Profiling , Humans , Light/adverse effects , Macular Degeneration/enzymology , Macular Degeneration/etiology , Macular Degeneration/pathology , Mice , NF-kappa B/antagonists & inhibitors , Oxidative Stress/physiology , Protein Processing, Post-Translational/drug effects , Rats , Rats, Wistar , Retinal Cone Photoreceptor Cells/pathology , Retinal Cone Photoreceptor Cells/physiology , Transcriptional Activation/drug effects
12.
Hum Mol Genet ; 17(24): 3929-41, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18805803

ABSTRACT

We observed that a naturally occurring mouse strain developed age-related retinal degeneration (arrd2). These mice had normal fundi, electroretinograms (ERGs) and retinal histology at 6 months of age; vessel attenuation, RPE atrophy and pigmentary abnormalities at 14 months, which progressed to complete loss of photoreceptors and extinguished ERG by 22 months. Genetic analysis revealed that the retinal degeneration in arrd2 segregates in an autosomal recessive manner and the disease gene localizes to mouse chromosome 10. A positional candidate cloning approach detected a nonsense mutation in the mouse double minute-1 gene (Mdm1), which results in the truncation of the putative protein from 718 amino acids to 398. We have identified a novel transcript of the Mdm1 gene, which is the predominant transcript in the retina. The Mdm1 transcript is localized to the nuclear layers of neural retina. Expression of Mdm1 in the retina increases steadily from post-natal day 30 to 1 year, and a high level of Mdm1 are subsequently maintained. The Mdm1 transcript was found to be significantly depleted in the retina of arrd2 mice and the transcript was observed to degrade by nonsense-mediated decay. These results indicate that the depletion of the Mdm1 transcript may underlie the mechanism leading to late-onset progressive retinal degeneration in arrd2 mice. Analysis of a cohort of patients with age-related macular degeneration (AMD) wherein the susceptibility locus maps to chromosome 12q, a region bearing the human ortholog to MDM1, did not reveal association between human MDM1 and AMD.


Subject(s)
Aging/genetics , Codon, Nonsense/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Animals , Electroretinography , Female , Gene Deletion , Humans , Male , Mice , Mice, Inbred C57BL , Sequence Analysis, DNA
13.
Proc Natl Acad Sci U S A ; 105(35): 12843-8, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18728184

ABSTRACT

Stargardt-like macular dystrophy (STGD3) is a dominantly inherited juvenile macular degeneration that eventually leads to loss of vision. Three independent mutations causing STGD3 have been identified in exon six of a gene named Elongation of very long chain fatty acids 4 (ELOVL4). The ELOVL4 protein was predicted to be involved in fatty acid elongation, although evidence for this and the specific step(s) it may catalyze have remained elusive. Here, using a gain-of-function approach, we provide direct and compelling evidence that ELOVL4 is required for the synthesis of C28 and C30 saturated fatty acids (VLC-FA) and of C28-C38 very long chain polyunsaturated fatty acids (VLC-PUFA), the latter being uniquely expressed in retina, sperm, and brain. Rat neonatal cardiomyocytes and a human retinal epithelium cell line (ARPE-19) were transduced with recombinant adenovirus type 5 carrying mouse Elovl4 and supplemented with 24:0, 20:5n3, or 22:5n3. The 24:0 was elongated to 28:0 and 30:0; 20:5n3 and 22:5n3 were elongated to a series of C28-C38 PUFA. Because retinal degeneration is the only known phenotype in STGD3 disease, we propose that reduced VLC-PUFA in the retinas of these patients may be the cause of photoreceptor cell death.


Subject(s)
Eye Proteins/metabolism , Fatty Acids, Unsaturated/biosynthesis , Macular Degeneration/metabolism , Membrane Proteins/metabolism , Animals , Cell Line , Humans , Mice , Myocytes, Cardiac/metabolism , Rats , Transgenes
14.
Invest Ophthalmol Vis Sci ; 47(12): 5505-13, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17122142

ABSTRACT

PURPOSE: In a prior study, a S163R mutation in the complement-1q tumor necrosis factor-related protein 5 (CTRP5/ C1QTNF5) was reported to be associated with early-onset long anterior zonules (LAZ) and late-onset retinal degeneration (L-ORD). The ocular tissues involved in the phenotype are the retinal pigment epithelium (RPE) in the posterior segment and ciliary epithelium (CE) and lens in the anterior segment. The purpose of this study was to characterize the spatial and temporal expression of the mouse Ctrp5 gene, determine tissue and subcellular localization, and study the effect of the S163R mutation. METHODS: The expression of the Ctrp5 gene in the mouse was studied by quantitative (q)RT-PCR and in situ hybridization. CTRP5 protein expression and distribution were studied by Western blot analysis, immunohistochemistry, and immunoelectron microscopy. Cellular location of wild-type and mutant CTRP5 in MDCK and COS-7 cells was determined by immunofluorescence and immunoblot analysis. RESULTS: A significant level of Ctrp5 expression was detected in the adult mouse in the ciliary body (CB) and RPE, and expression started at a very early stage of embryogenesis. Immunohistochemical analysis showed CTRP5 protein in the apical processes of the RPE and forming a hexagonal lattice associated with the RPE lateral membranes. In the ciliary body, CTRP5 was localized to the apical aspects of the CE, the region between the bilayered ciliary epithelial cells. The membrane association of CTRP5 in the RPE and CE was further confirmed by immunoelectron microscopy. Furthermore, cultured cells were used to show that the CTRP5 is a secretory protein and that its secretion is impaired by the S163R mutation. CONCLUSIONS: CTRP5, a secretory and membrane-associated protein, is localized to the lateral and apical membranes of the RPE and CB. Impaired secretion of the mutant protein may underlie the pathophysiology of L-ORD and LAZ.


Subject(s)
Ciliary Body/metabolism , Membrane Proteins/genetics , Pigment Epithelium of Eye/metabolism , Point Mutation , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , Fluorescent Antibody Technique, Indirect , Gene Expression/physiology , In Situ Hybridization , Lens Diseases/genetics , Ligaments/abnormalities , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Immunoelectron , Mutagenesis, Site-Directed , Retinal Degeneration/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
15.
Invest Ophthalmol Vis Sci ; 47(12): 5514-21, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17122143

ABSTRACT

PURPOSE: Mutations in the membrane frizzled-related protein (MFRP) gene cause nanophthalmos in humans, and a splice site mutation causes recessive retinal degeneration in the rd6 mouse. In human and mouse genomes, the MFRP gene lies adjoining to the complement 1q tumor necrosis factor-related protein 5 (CTRP5/C1QTNF5) gene involved in causing retinal degeneration and abnormal lens zonules in human. The purpose of this study was to characterize the spatial and temporal expression of the mouse Mfrp gene, determine tissue and subcellular localization of MFRP protein, and study its interaction with CTRP5. METHODS: Expression of the Mfrp gene in the mouse was studied by quantitative (q)RT-PCR. MFRP protein expression and distribution were studied by Western blot analysis, immunohistochemistry, and immunoelectron microscopy. Interaction with CTRP5 was studied by immunoprecipitation and immunoblot analysis, using mouse eye and human retinal pigmented epithelium (RPE) choroid extracts and by expressing full-length CTRP5 and MFRP in a heterologous system. RESULTS: The Mfrp gene is specifically expressed in RPE and ciliary body (CB), and its expression starts during early stages of embryogenesis. In the albino mouse eye, MFRP is localized to the apical and basal membranes of RPE and ciliary epithelium (CE). In addition, MFRP and CTRP5 were found to colocalize in RPE, CE, and MDCK cells, a general model of polarized epithelia. These proteins interact with each other in ocular tissues and also in a heterologous system. CONCLUSIONS: MFRP is localized to the plasma membrane of CE and RPE, and colocalizes and interacts with CTRP5 indicating a functional relationship between these two proteins.


Subject(s)
Ciliary Body/metabolism , Eye Proteins/genetics , Gene Expression/physiology , Membrane Proteins/genetics , Pigment Epithelium of Eye/metabolism , Animals , Blotting, Western , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Ciliary Body/embryology , Eye Proteins/metabolism , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Immunoelectron , Pigment Epithelium of Eye/embryology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
Invest Ophthalmol Vis Sci ; 47(10): 4558-68, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17003453

ABSTRACT

PURPOSE: To develop and characterize a heterozygous knock-in mouse model carrying the 5-bp deletion in Elovl4 (E_mut+/-) and to study the pathology underlying Stargardt-like macular degeneration (STGD3). METHODS: E_mut+/- mice were generated by targeting a 5-bp deletion (AACTT) in the Elovl4 gene by homologous recombination. E_mut+/- mice of age 2 to 18 months and age-matched wild-type (Wt) littermate control animals were analyzed for the expression of Elovl4 transcript, ELOVL4 protein, photoreceptor-specific genes, and retinal fatty acid composition. Functional retinal changes were evaluated by electroretinography (ERG) and by morphologic and ultrastructural criteria. RESULTS: E_mut+/- mice retinas showed the presence of both Wt and mutant Elovl4 transcripts and proteins. Morphologic evaluation revealed cone photoreceptor ultrastructural abnormalities as early as 2 months of age, accumulation of lipofuscin in retinal pigment epithelium (RPE), and subretinal deposits at later ages. Shortening of rod outer segments (OS) was observed at approximately 10 months of age. Both cone and rod changes progressed with age. Unlike rod-specific genes, expression of selected cone specific genes was significantly reduced by 7 months of age. Mixed rod-cone and light-adapted b-waves were higher than normal at both 8 and 15 months. Levels of the fatty acids 20:5 (P = 0.027), 22:5 (P = 0.040) and 24:6 (P = 0.005) were found to be significantly lower in the retinas of E_mut+/- mice than in retinas of control subjects. CONCLUSIONS: E_mut+/- animals display characteristic features associated with Stargardt-like macular degeneration and serve as a model for the study of the mechanism underlying STGD3.


Subject(s)
Disease Models, Animal , Eye Proteins/genetics , Membrane Proteins/genetics , Mutation , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Animals , Base Sequence , Blotting, Western , Chromatography, Gas , Disease Progression , Electroretinography , Fatty Acids/metabolism , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells, Vertebrate/metabolism , Polymerase Chain Reaction , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Sequence Deletion
17.
Invest Ophthalmol Vis Sci ; 47(8): 3603-11, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16877435

ABSTRACT

PURPOSE: Autosomal dominant Stargardt-like (STGD3) disease results from mutations in the ELOVL4 gene (elongation of very-long-chain fatty acids). This study was undertaken to characterize a mouse model with a targeted deletion of Elovl4 and to explore the role of this gene in retinal/macular degeneration. METHODS: A construct targeted to exon 2 of the Elovl4 gene was used to suppress expression of the gene. Elovl4 homozygous pups were nonviable and were not available for study. Hence, the analysis was performed on heterozygous Elovl4(+/-) mice 16 to 22 month of age and littermate wild-type (WT) control mice of the same age. Characterization included examining gene message and protein levels, electroretinogram (ERG), retinal morphology and ultrastructure, and plasma and retinal fatty acid composition. RESULTS: Although the level of Elovl4 mRNA was reduced in Elovl4(+/-) retinas, only minimal morphologic abnormalities were found, and the retinal (ERG) function was essentially normal in Elovl4(+/-) retinas compared with the WT control retinas. Systemic fatty acid profiles of Elovl4(+/-) mice were unremarkable, although the concentration of several fatty acids was significantly lower in Elovl4(+/-) retinas, particularly the monounsaturated fatty acids. CONCLUSIONS: The detailed characterization of this animal model provides the first in vivo evidence that Elovl4 haploinsufficiency is not the underlying key disease mechanism in STGD3. The results are consistent with a dominant negative mechanism for the deletion mutation. The Elovl4 knockout mouse is one of three complementary animal models that will help elucidate the disease mechanism.


Subject(s)
Disease Models, Animal , Eye Proteins/genetics , Gene Expression Regulation/physiology , Macular Degeneration/genetics , Membrane Proteins/genetics , Animals , Blotting, Western , Electroretinography , Fatty Acids/metabolism , Female , Gene Deletion , Genotype , Haplotypes , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Retina/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Rhodopsin/genetics
18.
Invest Ophthalmol Vis Sci ; 47(9): 4091-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936129

ABSTRACT

PURPOSE: Complement factor H (CFH) is a component of the mammalian complement system, which regulates the alternative pathway of complement activation and protects the host cell from inappropriate complement activation. CFH is a key regulator of innate immunity, and CFH deficiency leads to membranoproliferative glomerulonephritis type II. A variation in human CFH, Y402H, has been shown to be associated with an increased risk for age-related macular degeneration. The authors describe studies on the spatial and temporal expression of the CFH gene and localization of this protein in ocular tissues to gain insight into its role in the eye. METHODS: CFH expression in human and mouse tissues was studied by quantitative RT-PCR and Western blot analysis, and localization of CFH was studied by immunohistochemical analysis followed by fluorescence microscopy. RESULTS: In human and mouse, CFH expression was found to be similar to the highest level of expression in the liver. In ocular tissue, CFH was detected in the distalmost optic nerve (3 mm) cut from the scleral surface of the eyeball, sclera, RPE-choroid, retina, lens, and ciliary body. In mouse, Cfh expression was observed from early embryonic stages, and in the eye its expression increased with age. CONCLUSIONS: A significant level of CFH expression is maintained in different ocular tissues during development and aging. Sustained high levels of CFH expression in eye tissues suggest that this protein may play a role in protecting these tissues from indiscriminate complement activation and inflammatory insult.


Subject(s)
Complement Factor H/genetics , Eye/metabolism , Gene Expression/physiology , Aged , Aging/physiology , Amino Acid Sequence , Animals , Blotting, Western , Complement Factor H/metabolism , Eye/embryology , Humans , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Fluorescence , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
19.
Invest Ophthalmol Vis Sci ; 46(9): 3355-62, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123440

ABSTRACT

PURPOSE: To develop and apply microarray-based resequencing technology to detect sequence alterations in multiple autosomal recessive retinal disease genes on a single high-throughput platform. METHODS: Oligonucleotides corresponding to both strands of the target exons and the flanking intron sequences of 29,214 bp from 11 genes associated with autosomal recessive retinitis pigmentosa (arRP) were tiled on 20 x 25-microm microarrays (arRP-I arrays). A total of 155 exons were amplified from 35 arRP patient DNA samples, with each sample being sequenced on an arRP-I chip by hybridization. RESULTS: With the arRP-I arrays, 97.6% of the tiled sequence were determined with more than 99% accuracy and reproducibility. Of the 2.4% unread sequence, 89.5% involved stretches of G or C. In analyzing the 903,140-bp sequence from the 35 patient samples, 506 sequence changes have been detected in which 386 are previously reported alterations, and 120 are novel. In addition to four known causative mutations, six novel sequence changes that are potentially pathogenic were observed. Additional analysis is needed to determine whether these changes are responsible for arRP in these patients. CONCLUSIONS: The use of microarray for sequencing is a novel approach, and the arRP-I chip is the first successful application of this technology for determining sequence alteration in multiple disease-related genes. These arrays can be used for high-throughput genotyping of patients with relevant retinal conditions. In addition, these arrays offer a unique opportunity to interrogate complex patterns of inheritance due to the involvement of more than one gene by screening multiple genes on a single platform.


Subject(s)
Gene Expression Profiling , Genetic Testing/methods , Oligonucleotide Array Sequence Analysis , Retinitis Pigmentosa/genetics , Base Sequence , DNA Fragmentation , DNA Mutational Analysis , Eye Proteins/genetics , Genes, Recessive , Genotype , Humans , Microtubule-Associated Proteins , Molecular Sequence Data , Nucleic Acid Hybridization , Polymerase Chain Reaction
20.
Invest Ophthalmol Vis Sci ; 46(9): 3363-71, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123441

ABSTRACT

PURPOSE: To identify the gene responsible for a complex ocular phenotype of late-onset macular degeneration, long anterior zonules (LAZ), and elevated intraocular pressure (IOP) and to study its expression. METHODS: Ocular examination, visual field, fluorescein angiography, and electrophysiology testing were performed. One affected individual was treated with vitamin A. DNA from 55 family members (UM:H389) was used for linkage, mapping, and mutation analysis. Linkage analysis of macular degeneration and LAZ phenotypes was performed independently. Mutations in candidate genes were screened by sequencing. mRNA expression of CTRP5 and MFRP, which are bicistronic genes, was studied by semiquantitative RT-PCR (qRT-PCR) in various human tissues. CTRP5 expression was also evaluated by in situ hybridization. RESULTS: Affected members had LAZ detectable by the third decade and/or macular degeneration by the fourth to fifth decade. A six-month treatment with vitamin A shortened dark adaptation considerably in one affected member. Both conditions mapped independently with zero recombination to 11q23, with maximum lod scores of 3.31 for macular degeneration and 5.41 for LAZ. The same CTRP5 missense mutation was identified in all affected individuals. Retinal pigment epithelium (RPE) and ciliary epithelium (CE) showed highest CTRP5 transcript expression, which was also true for MFRP. CTRP5 tissue expression was confirmed by in situ hybridization. CONCLUSIONS: A single locus at 11q23 is implicated in a complex ocular phenotype involving RPE and CE, tissues of neuroectodermal origin. All individuals with either LAZ and/or macular degeneration carry the same CTRP5 S163R mutation, which is transmitted in autosomal dominant manner.


Subject(s)
Collagen/genetics , Lens Diseases/genetics , Lens, Crystalline/pathology , Ligaments/pathology , Macular Degeneration/genetics , Adult , Aged , Aged, 80 and over , Chromosomes, Human, Pair 11/genetics , Collagen/metabolism , DNA Mutational Analysis , Female , Genetic Linkage , Humans , Iris/metabolism , Lens Diseases/metabolism , Macular Degeneration/metabolism , Male , Membrane Proteins/genetics , Middle Aged , Mutation, Missense , Pedigree , Phenotype , Pigment Epithelium of Eye/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vitamin A/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...