Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(9): e0138958, 2015.
Article in English | MEDLINE | ID: mdl-26402857

ABSTRACT

Dry eye is a common disorder caused by inadequate hydration of the ocular surface that results in disruption of barrier function. The homeostatic protein clusterin (CLU) is prominent at fluid-tissue interfaces throughout the body. CLU levels are reduced at the ocular surface in human inflammatory disorders that manifest as severe dry eye, as well as in a preclinical mouse model for desiccating stress that mimics dry eye. Using this mouse model, we show here that CLU prevents and ameliorates ocular surface barrier disruption by a remarkable sealing mechanism dependent on attainment of a critical all-or-none concentration. When the CLU level drops below the critical all-or-none threshold, the barrier becomes vulnerable to desiccating stress. CLU binds selectively to the ocular surface subjected to desiccating stress in vivo, and in vitro to the galectin LGALS3, a key barrier component. Positioned in this way, CLU not only physically seals the ocular surface barrier, but it also protects the barrier cells and prevents further damage to barrier structure. These findings define a fundamentally new mechanism for ocular surface protection and suggest CLU as a biotherapeutic for dry eye.


Subject(s)
Clusterin/therapeutic use , Dry Eye Syndromes/drug therapy , Eye/pathology , Administration, Topical , Animals , Clusterin/pharmacology , Cytoprotection/drug effects , Desiccation , Dry Eye Syndromes/pathology , Eye/drug effects , Female , Galectin 3/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/drug effects , Stress, Physiological/drug effects , Tears/metabolism
2.
Invest Ophthalmol Vis Sci ; 55(1): 492-501, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24370827

ABSTRACT

PURPOSE: Age-related macular degeneration (AMD) is a leading cause of visual impairment worldwide. Genetics and diet contribute to the relative risk for developing AMD, but their interactions are poorly understood. Genetic variations in Complement Factor H (CFH), and dietary glycemic index (GI) are major risk factors for AMD. We explored the effects of GI on development of early AMD-like features and changes to central nervous system (CNS) inflammation in Cfh-null mice. METHODS: Aged 11-week-old wild type (WT) C57Bl/6J or Cfh-null mice were group pair-fed high or low GI diets for 33 weeks. At 10 months of age, mice were evaluated for early AMD-like features in the neural retina and RPE by light and electron microscopy. Brains were analyzed for Iba1 macrophage/microglia immunostaining, an indicator of inflammation. RESULTS: The 10-month-old WT mice showed no retinal abnormalities on either diet. The Cfh-null mice, however, showed distinct early AMD-like features in the RPE when fed a low GI diet, including vacuolation, disruption of basal infoldings, and increased basal laminar deposits. The Cfh-null mice also showed thinning of the RPE, hypopigmentation, and increased numbers of Iba1-expressing macrophages in the brain, irrespective of diet. CONCLUSIONS: The presence of early AMD-like features by 10 months of age in Cfh-null mice fed a low GI diet is surprising, given the apparent protection from the development of such features in aged WT mice or humans consuming lower GI diets. Our findings highlight the need to consider gene-diet interactions when developing animal models and therapeutic approaches to treat AMD.


Subject(s)
Complement Factor H/genetics , Dietary Carbohydrates/pharmacology , Glycemic Index/genetics , Macular Degeneration/genetics , Animals , Blood Glucose/metabolism , Complement Factor H/metabolism , Disease Models, Animal , Genotype , Immunohistochemistry , Macular Degeneration/diet therapy , Macular Degeneration/pathology , Mice , Mice, Inbred C57BL , Microscopy, Electron , Retinal Pigment Epithelium/ultrastructure
3.
Pediatr Res ; 61(3): 273-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17314682

ABSTRACT

The Smith-Lemli-Opitz syndrome (SLOS) is the first-described in a growing family of hereditary defects in cholesterol biosynthesis, and presents with a spectrum of serious abnormalities, including multiple dysmorphologies, failure to thrive, cognitive and behavioral impairments, and retinopathy. Using a pharmacologically induced rat model of SLOS that exhibits key hallmarks of the disease, including progressive retinal degeneration and dysfunction, we show that a high-cholesterol diet can substantially correct abnormalities in retinal sterol composition, with concomitant improvement of visual function, particularly within the cone pathway. Although histologic degeneration still occurred, a high-cholesterol diet reduced the number of pyknotic photoreceptor nuclei, relative to animals on a cholesterol-free diet. These findings demonstrate that cholesterol readily crosses the blood-retina barrier (unlike the blood-brain barrier) and suggest that cholesterol supplementation may be efficacious in treating SLOS-associated retinopathy.


Subject(s)
Cholesterol, Dietary/therapeutic use , Retina/physiopathology , Smith-Lemli-Opitz Syndrome/diet therapy , Smith-Lemli-Opitz Syndrome/physiopathology , Sterols/metabolism , Animals , Cholesterol, Dietary/administration & dosage , Disease Models, Animal , Female , Humans , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/physiology , Pregnancy , Rats , Retina/pathology , Smith-Lemli-Opitz Syndrome/chemically induced , Smith-Lemli-Opitz Syndrome/metabolism , trans-1,4-Bis(2-chlorobenzaminomethyl)cyclohexane Dihydrochloride/toxicity
4.
J Cell Biol ; 173(1): 59-68, 2006 Apr 10.
Article in English | MEDLINE | ID: mdl-16585269

ABSTRACT

It is commonly assumed that photoreceptor (PR) outer segment (OS) morphogenesis is reliant upon the presence of peripherin/rds, hereafter termed Rds. In this study, we demonstrate a differential requirement of Rds during rod and cone OS morphogenesis. In the absence of this PR-specific protein, rods do not form OSs and enter apoptosis, whereas cone PRs develop atypical OSs and are viable. Such OSs consist of dysmorphic membranous structures devoid of lamellae. These tubular OSs lack any stacked lamellae and have reduced phototransduction efficiency. The loss of Rds only appears to affect the shape of the OS, as the inner segment and connecting cilium remain intact. Furthermore, these structures fail to associate with the specialized extracellular matrix that surrounds cones, suggesting that Rds itself or normal OS formation is required for this interaction. This study provides novel insight into the distinct role of Rds in the OS development of rods and cones.


Subject(s)
Cell Differentiation/genetics , Intermediate Filament Proteins/genetics , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Retinal Cone Photoreceptor Cells/abnormalities , Retinal Rod Photoreceptor Cells/abnormalities , Animals , Apoptosis/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Cell Survival/genetics , Electroretinography , Eye Proteins/genetics , Immunohistochemistry , Intracellular Membranes/metabolism , Intracellular Membranes/pathology , Intracellular Membranes/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Peripherins , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/ultrastructure , Vision, Ocular/genetics
5.
Exp Eye Res ; 82(3): 538-41, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16199034

ABSTRACT

The Smith-Lemli-Opitz syndrome (SLOS) is an autosomal recessive disease presenting with multiple congenital anomalies, caused by a defect in cholesterol biosynthesis that results in abnormally elevated levels of 7-dehydrocholesterol (7DHC). Progressive retinal degeneration has been demonstrated in a rat model of SLOS, which is markedly exacerbated by intense light, far more so than occurs in normal albino rats under the same conditions. Herein, we demonstrate that, by six postnatal weeks, retinas in the SLOS rat model contain levels of lipid hydroperoxides (LPOs) comparable to those found in light-damaged albino rats (twice the normal steady-state levels), and that intense light exposure results in a three-fold elevation of LPOs with concomitant severe retinal degeneration. These results suggest a correlation between retinal degeneration and LPO levels. We propose that the presence of 7DHC in the SLOS rat retina potentiates LPO formation, and promotes the observed hypersensitivity to light-induced retinal degeneration.


Subject(s)
Lipid Peroxides/biosynthesis , Retina/metabolism , Retinal Degeneration/metabolism , Smith-Lemli-Opitz Syndrome/metabolism , Animals , Light/adverse effects , Models, Animal , Rats , Rats, Sprague-Dawley , Retina/pathology , Retinal Degeneration/pathology , Smith-Lemli-Opitz Syndrome/pathology
6.
Arch Ophthalmol ; 122(8): 1190-200, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15302661

ABSTRACT

OBJECTIVE: To assess the electrophysiologic, histologic, and biochemical features of an animal model of Smith-Lemli-Opitz syndrome (SLOS). METHODS: Sprague-Dawley rats were treated with AY9944, a selective inhibitor of 3beta-hydroxysterol-Delta(7)-reductase (the affected enzyme in SLOS). Dark- and light-adapted electroretinograms were obtained from treated and control animals. From each animal, 1 retina was analyzed by microscopy, and the contralateral retina plus serum samples were analyzed for sterol composition. The main outcome measures were rod and cone electroretinographic amplitudes and implicit times, outer nuclear layer (ONL) thickness, rod outer segment length, pyknotic ONL nucleus counts, and the 7-dehydrocholesterol/cholesterol mole ratio in the retina and serum. RESULTS: By 10 weeks' postnatal age, rod and cone electroretinographic wave amplitudes in AY9944-treated animals were significantly reduced and implicit times were significantly increased relative to controls. Maximal rod photoresponse and gain values were reduced approximately 2-fold in treated animals relative to controls. The ONL thickness and average rod outer segment length were reduced by approximately 18% and 33%, respectively, and ONL pyknotic nucleus counts were approximately 4.5-fold greater in treated animals relative to controls. The retinal pigment epithelium of treated animals contained massive amounts of membranous/lipid inclusions not routinely observed in controls. The 7-dehydrocholesterol/cholesterol mole ratios in treated retinas and serum samples were approximately 5:1 and 9:1, respectively, whereas the ratios in control tissues were essentially zero. CONCLUSIONS: This rodent model exhibits the key biochemical hallmarks associated with SLOS and displays electrophysiologic deficits comparable to or greater than those observed in the human disease. Clinical Relevance These results predict retinal degeneration in patients with SLOS, particularly those with the more severe (type II) form of the disease, and may be more broadly relevant to other inborn errors of cholesterol biosynthesis. This animal model may also be of use in evaluating therapeutic treatments for SLOS and in understanding the slow phototransduction kinetics observed in patients with SLOS.


Subject(s)
Disease Models, Animal , Retina/physiopathology , Retinal Degeneration/physiopathology , Smith-Lemli-Opitz Syndrome/physiopathology , Animals , Anticholesteremic Agents/toxicity , Cholesterol/blood , Dark Adaptation , Dehydrocholesterols/blood , Electrophysiology , Electroretinography , Female , Pigment Epithelium of Eye , Pregnancy , Rats , Rats, Sprague-Dawley , Retina/metabolism , Retina/ultrastructure , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Smith-Lemli-Opitz Syndrome/chemically induced , Smith-Lemli-Opitz Syndrome/metabolism , trans-1,4-Bis(2-chlorobenzaminomethyl)cyclohexane Dihydrochloride/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...